WO2011103382A2 - Compositions and methods for inducing angiogenesis - Google Patents

Compositions and methods for inducing angiogenesis Download PDF

Info

Publication number
WO2011103382A2
WO2011103382A2 PCT/US2011/025374 US2011025374W WO2011103382A2 WO 2011103382 A2 WO2011103382 A2 WO 2011103382A2 US 2011025374 W US2011025374 W US 2011025374W WO 2011103382 A2 WO2011103382 A2 WO 2011103382A2
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
ischemia
protein
effective amount
angiogenesis
Prior art date
Application number
PCT/US2011/025374
Other languages
French (fr)
Other versions
WO2011103382A3 (en
Inventor
Shiladitya Sengupta
Ermanno Gherardi
Rituparana Sinha Roy
Shivani Soni
Rania Harfouche
Pooja R. Vasudevan
Original Assignee
The Brigham And Women's Hospital
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 The Brigham And Women's Hospital filed Critical The Brigham And Women's Hospital
Priority to CN2011800203513A priority Critical patent/CN103037892A/en
Publication of WO2011103382A2 publication Critical patent/WO2011103382A2/en
Publication of WO2011103382A3 publication Critical patent/WO2011103382A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1833Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Optics & Photonics (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention relates to pharmaceutical compositions and to methods for inducing angiogenesis in conditions associated with insufficient vascularization, e.g. ischemia and diabetic conditions, such as diabetic foot ulcers. In particular, pharmaceutical compositions comprising sustained release nanoparticles that contain 1K1 fragment of Hepatocyte Growth Factor/Scatter Factor are provided.

Description

COMPOSITIONS AND METHODS FOR INDUCING ANGIOGENESIS
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims benefit under 35 U.S.C. § 119(e) of the U.S. Provisional Application No. 61/378,968 filed September 01, 2010 and U.S. Provisional Application No. 61/306,768 filed February 22, 2010, the content of which are incorporated herein by reference in their entirety.
FIELD OF THE INVENTION
[0002] Embodiments of the invention relate to pharmaceutical compositions and to methods for inducing angio genesis in conditions associated with insufficient vascularization, e.g. ischemia and diabetic conditions, such as diabetic foot ulcers.
BACKGROUND OF THE INVENTION
[0003] Neovascularization in the adult holds the potential for ameliorating ischemic disease, which is the leading cause of morbidity and mortality in the US (1). Ischemic diseases include cardiovascular conditions, including angina, peripheral arterial diseases, tissue sores. Ferrara and Kerbel recently articulated that 'therapeutic angiogenesis' is an emerging and exciting frontier of cardiovascular medicine (2). The promotion of
neovascularization is critical for wound healing, for example in the management of diabetic sores. One of the major underlying problem facing diabetics is impaired wound healing. Fifteen percent of all people with diabetes (2.6 million) are expected to develop foot ulcers during their lifetime. These ulcers tend to be chronic in nature, as they do not heal or heal extremely slowly. There are more than 750,000 patients with diabetic foot ulcers in the United States, 980,000 in Europe and 1.1 million in the rest of the world, totaling 2.8 million patients. Diabetic foot ulcers are a serious problem, as up to 25% of diabetic foot ulcers will eventually require amputation. The medical importance of diabetic wound healing cannot be overstated. The capacity to heal is central to human well being, as wound healing enables a patient to overcome traumatic injury, surgery, and wounds due to metabolic disorders such as diabetes, microbial or other physical or chemical agents. Thus, agents that promote neovascularization are needed. [0004] However, with respect to 'therapeutic angiogenesis', while early preclinical studies and phase I clinical trials exhibited promising results with recombinant proteins or gene therapy, more rigorous phase II and III studies have reported conflicting outcomes (3-6). A recent review of these clinical trials highlighted vector 'washout' leading to inadequate duration of exposure to the angiogenic agent as a key factor that may have impaired the effectiveness of therapy (7). A second challenge is the stability of angiogenc factors, which are large proteins and are often unstable in normal physiological conditions. Furthermore, the glycosylation of these proteins pose significant challenge in clinical development. It would be desirable to have compositions and methods that overcome these challenges.
SUMMARY OF THE INVENTION
[0005] Herein, we have coupled protein engineering with nanotechnology to overcome challenges observed using angiogenic factors in therapy. Embodiments of the invention are based upon the observation that fragment 1K1 of the Hepatocyte Growth Factor/Scatter Factor (HGF/SF) enhanced angiogenesis when formulated as a nanoparticle as compared to administration of free 1K1. In particular, delivery of the 1K1 loaded nanoparticles resulted in sustained activation of MAPK signaling via ERK phosphorylation and induced significantly greater tubulogenesis in vitro and in vivo, as compared to administration 1K1 fragment alone.
[0006] Accordingly, pharmaceutical compositions comprising nanoparticles of biocompatible polymer and an effective amount of 1K1 protein fragment of Hepatocyte Growth Factor/Scatter Factor are provided, wherein the biodegradable polymer encapsulates the 1K1 protein. In one embodiment, the 1K1 protein comprises SEQ ID. NO:l.
[0007] In one embodiment, the nanoparticles of the pharmaceutical compositions have an average particle size of about 50 nm to about 500 nm, or of about 60 nm to about 150 nm.
[0008] Any biocompatible polymers can be used in formulation of the nanoparticles, In one embodiment, the biocompatible polymer is a biodegradable polymer and the
biodegradable polymer is selected from the group consisting of: polyesters, hydroxyaliphatic carboxylic acids, poly(lactic acid), poly(glycolic acid), poly(dl-lactide/glycolide,
poly(ethylene glycol), polysaccharides, lectins, glycosaminoglycans, chitosan, celluloses and acrylate polymers.
[0009] In one embodiment, the biocompatible polymer is a polyester, e.g. a polyester comprising poly-lactic acid-glycolic acid copolymer (PLGA).
[0010] In some embodiments, the 1K1 protein is released from the nanoparticles of the pharmaceutical composition in a therapeutically effective amount over a defined period of time of days, weeks or months, i.e. sustained release over a defined period of time. In one embodiment, the 1K1 protein is released from the nanoparticle in a therapeutically effective amount over a defined period of time of about 2 days, about 3 days, about 4 days, about 5 days, about 7 days, or about 14 days.
[0011] In some embodiments, the therapeutically effective amount that is released from the nanoparticles over the defined period of time is sufficient to result in sustained phoshorylation of cellular ERK over the defined period of time of release.
[0012] In some embodiments, the therapeutically effective amount that is released from the nanoparticles over the defined period of time is sufficient to increase angiogenesis in a subject as compared to angiogenesis in the absence of the compound by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, or about 60%.
[0013] In some embodiments, the therapeutically effective amount is a dose of about
O.lmg/kg to about 1000 mg/kg.
[0014] The pharmaceutical compositions may formulated for administration by topical administration, enteral administration, and parenteral administration. In one embodiment, the composition is formulated for topical administration and is an ointment, lotion, spray, cream, or gel.
[0015] Methods for treatment of a conditions associated with insufficient vascularization are also provided. The methods comprise administration to a subject a therapeutically effective amount of the pharmaceutical compositions comprising nanoparticles of
biocompatible polymer and an effective amount of 1K1 protein fragment of Hepatocyte Growth Factor/Scatter Factor, wherein the biodegradable polymer encapsulates the 1K1 protein. In one embodiment, the therapeutically effective amount is a dose of about O.lmg/kg to about 1000 mg/kg of 1K1 protein.
[0016] In another aspect of the invention, methods for treatment of a conditions associated with insufficient vascularization are provided that comprise administrating to a subject an effective amount of 1K1 protein, wherein the 1K1 protein is administered in a manner to result in a sustained phosphorylation of cellular ERK. For example, administration may be performed using sustained release formulations, such as capsules, gels, films, or other matrices that release 1K1 over a defined period of time of days, weeks, or months.
Administration using formulations for sustained release of 1K1 protein results in sustained phosphorylation of ERK and enhanced tubulogenesis, thereby significantly increasing angiogenesis in the subject.
[0017] Conditions contemplated for said treatment include: myocardial ischemic conditions (e.g., myocardial infarction, revascularization of necrotic tissue, for example of the myocardium after an infarction or an angioplasty, angina, heart transplants, vascular grafts, and reopening vessels to improve vascularization, perfusion, coUagenization and organization of said lesions), wound healing, and tissue and organ transplantations (e.g., enhancement of autologous or heterologous microvascular transplantation). Promotion of wound healing includes healing of incisions, bone repair, burn healing, post-infarction repair in myocardial injury, healing of gastric ulcers and other ulcers of the gastrointestinal tract and generally in promoting the formation, healing of diabetic ulcers, maintenance and repair of tissue.
Neovascularization of grafted or transplanted tissue is also contemplated, especially in subjects suffering from vascular insufficiency, such as diabetic.
[0018] In one embodiment, the condition associated with insufficient vascularization, is selected from the group consisting of cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, ischemia of tissues, coronary ischemia, peripheral arterial disease, limb ischemia, diabetic ulcers, gangrenes, wounds requiring neovascularization to facilitate healing, and Buerger's syndrome. In one embodiment, the condition associated with insufficient vascularization is a diabetic foot ulcer.
[0019] The pharmaceutical compositions of the invention may be administered by topical administration, enteral administration, or parenteral administration. In one embodiment, the pharmaceutical composition is administered multiple times, e.g. using varied treatment regimes.
BRIEF DESCRIPTION OF THE DRAWINGS
[0020] Figures 1A to 1H show structural features of the lKl-heparin complex and interaction with the MET receptor. Figure 1A shows the domain structure of full length multidomain HGF/SF. The a-chain consists of N-terminal domain (amino acids 32-121) and four kringle domains (Kl, K2, K3, and K4) and the β-chain contains serine protinease domain (spdh domain). Figure IB shows the crystal structure of NKl-heparin complex (16) (PDB accession 1GMO). Two NK1 dimers are shown bridged by heparin. Figure 1C shows the crystal structure of lKl-heparin complex (PDB accession 3MPK). Two 1K1 dimers are shown bridged by heparin. Figures IB and 1C have been drawn with Pymol. Reverse mutations K132E and R134E were introduced into Kl domain of 1K1 to inactivate the low- affinity heparin-binding sites. Figure ID is the amino acid sequence of 1K1 demonstrating the mutated sites. The underlined amino acids show two 1K1 dimers bridged by heparin. Figure IE and Figure IF shows binding of NK1 and 1K1 to MET567, respectively, in the presence of 12mer heparin using surface plasmon resonance. Twofold dilutions of each protein were used from a concentration of top concentration of 200 nM. The concentration of 12mer heparin in the sample and in the reaction buffer was 10 μΜ. Figure 1G and Figure 1H show velocity sedimentation analysis of ternary complexes of NKl-heparin-MET567 and lKl-heparin-MET567, respectively. Data show plots of c(s) against s*20,w. In the presence of lOmer heparin, the amount of ternary complex is significantly higher for 1K1 than for NK1.
[0021] Figures 2A to 2D show both HGF/SF and 1K1 induce endothelia cell proliferation. Figure 2A shows the human umbilical vein endothelial cells (HUVECs) proliferation in presence of HGF/SF and 1K1 at two different concentrations (10~10 M and 10"7 M). Figure 2B shows the effect of PI3 kinase inhibitor, LY294002, on lKl-induced cell proliferation at 24 h. Figure 2C shows the effect of Met inhibitor, PHA 665752, on 1K1- induced HUVECs proliferation at 48 h. Figure 2D shows the effect of MAP kinase inhibitor, PD98059, on 1K1 -induced HUVECs proliferation at 72 h. The data represent mean ± SEM from n = 3. The symbol indicates a p-value of less than 0.001 vs vehicle-treated control; the symbol "*" indicates a p-value of less than 0.05 vs vehicle-treated control; the symbol "###" indicates a p-value of less than 0.001 vs 1K1[10"7 M].; the symbol "#" indicates a p-value of less than 0.05 vs 1K1 [10 M].
[0022] Figures 3A to 3C show that 1K1 induces endothelial tubulogenesis by binding to MET kinase. Figures 3 A and 3B show the quantification of the images, for effect of pharmacological inhibitors on the 1K1 -induced on HUVECs tube formation on growth- factor-reduced matrigel (images not shown), using three morphometric analyses of average length of tubes and average number of nodes, respectively. The data represent mean ± SEM pixels from n = 3. The symbol "**" indicates a p-value of less than 0.01 vs. vehicle-treated control; the symbol " #" indicates a p-value of less than 0.05 vs. 1K1([10 M]). Figure 3C is a representative Western blot showing phospho Erk and total Erk and also phospho Akt and total Akt expression in HUVECs treated with Met inhibitor PHA 665752 (10~6 M),
LY294002 (50 μΜ) and PD98059 (50 μΜ) for 2 h, followed by 1K1 for 10 mins. The numbers indicate 1: vehicle, 2: HGF/SF (10~8 M), 3: 1K1 (10~7 M), 4: 1K1 + PHA 665752 (10~6 M) 5: 1K1 + LY294002 (50 μΜ), 6: 1K1 + PD98059 (50 μΜ). For sample no. 4, 5, and 6, 1K1 (10"' M) were used along with the inhibitors.
[0023] Figures 4A to 4F show lKl-nanoparticles induces angiogenesis in vitro.
Figure 4A shows the transmission electron microscopy (TEM) image of 1K1 -encapsulated nanoparticles (1K1-NP) (Bar =500 nm). Figure 4B shows the size distribution (in nm) of the 1K1 -encapsulated nanoparticles by dynamic light scattering experiment. Figure 4C shows release kinetics of IKl from the nanoparticle when incubated in PBS at room temperature. The values on the Y-axis represent the amount of IKl released from the 1K1-NP in μg unit. Figure 4D shows the effect of Met inhibitor, PHA 665752, on 1K1-NP induced HUVECs proliferation at 48 h. Figure 4E shows the effect of PI3 kinase inhibitor, LY294002, on 1K1- NP induced HUVECs proliferation at 48 h. Figure 4F shows the effect of MAP kinase inhibitor, PD98059, on 1K1-NP induced HUVECs proliferation at 48 h. The data represent mean ± SEM from n = 3. The symbol indicates a p-value of less than 0.001 vs. vehicle- treated control; the symbol "**" indicates a p-value of less than 0.01 vs. vehicle-treated control; the symbol "###" indicates a p-value of less than 0.001 vs. 1K1(10 M); the symbol "##" indicates a p-value of less than 0.01 vs. 1K1(10"7 M); the symbol "#" indicates a p- value of less than 0.05 vs. 1K1(10"7 M).
[0024] Figures 5A to 5C show that sustain release of the angiogenic factor by 1K1- NP markedly induces angiogenic response. Figure 5A shows the quantification of the images, for effect of pharmacological inhibitors on the 1K1-NP induced HUVEC tube formation on matrigel, using average length of tubes. The data represent mean ± SEM pixels from n = 3. The symbol "*" indicates a p-value of less than 0.05 vs. vehicle-treated control; the symbol "##" indicates a p-value of less than 0.01 vs. IKl (10~7 M); the symbol "#" indicates a p-value of less than 0.05 vs. IKl (10 M). Figure 5B is a representative Western blot showing phospho Erk and total Erk and also phospho Akt and total Akt in HUVECs treated with Met inhibitor PHA 665752 (10~6 M), LY294002 (50 μΜ) and PD98059 (50 μΜ) for 2 h, followed by 10 min 1K1-NP and IKl. The numbers indicate 1: vehicle (PBS), 2: vehicle (empty PLGA), 3: freshly prepared IKl (0.5 x 10"7 M), 4: IKl incubated at 37 °C for 24 h (0.5 x 10"7 M), 5: 1K1-NP incubated at 37 °C for 24 h (0.5 x 10"7 M), 6: 1K1-NP + PHA665752 (10~6 M), 7: 1K1-NP + LY294002 (50 μΜ), 8:1K1-NP + PD98059 (50 μΜ). For sample no. 6, 7, and 8, (0.5 x 10 M) 1K1-NP were used along with the inhibitors. Figure 5C is a representative Western blot showing phospho Erk and total Erk in HUVECs treated with (0.5x 10"7 M) IKl and (0.5 x 10"7 M) 1K1-NP for 8.5 h.
[0025] Figures 6A to 6F show the effect of IKl and 1K1-NP mediated angiogenesis in vivo using zebrafish model. IKl or 1K1-NP were injected with growth-factor-reduced matrigel (Mgel) near the subintestinal vessel (SIV). SIVs at indicated conditions were then stained with alkaline phosphate and visualized in bright field, as shown in Figures 6A to 6E. The indicated conditions are: Figure 6A: vehicle (Mgel); Figure 6B: empty PLGA in Mgel; Figure 6C: IKl in Mgel; Figure 6D: 1K1-NP in Mgel; and Figure 6E: 1K1-NP. Figure 6F shows morphometric quantification of the effect of treatment on angiogenesis. Data shown mean SEM ± from n = 3.
[0026] Figures 7A to 7D show the effect of 1K1 (200 ng/plug) and 1K1-NP (200 ng/plug) induced angiogenesis in growth-factor-reduced matrigel implants in vivo.
Figures 7A to 7D show cross-sections of the implant immunolabled for von Willebrand factor and counterstained with DAPI for nuclei. Figures 7A (vehicle) and 7B (empty NP) shows nuclei DAPI staining, but negative for von Willebrand factor staining. Figure 7C (1K1) and 7D (1K1-NP) shows images of blood vessels delineated with von Willebrand factor immunolabeling. Images were captured at 512 x 512 pixels resolution.
DETAILED DESCRIPTION OF THE INVENTION
[0027] Embodiments of the invention are based upon the observation that the Hepatocyte Growth Factor/Scatter Factor (HGF/SF) 1K1 fragment enhanced angiogenesis when formulated as a nanoparticle as compared to administration of free 1K1. In particular, delivery of the 1K1 nanoparticle resulted in sustained activation of MAPK signaling and induced significantly greater tubulogenesis in vitro and in vivo, as compared to administration 1K1 fragment alone.
[0028] Hepatocyte Growth Factor/Scatter Factor (HGF/SF) (SEQ ID NO: 3) is a pleiotropic growth factor which promotes proliferation and migration of endothelial cells through the Met receptor (9). In a previous study, we have demonstrated that HGF/SF induces angiogenesis independent of vascular endothelial growth factor (10) by activating the mitogen activated protein kinase (MAPK) and the phosphatidylinositol-3-kinase pathways (P13K) (11-12). Moreover, Met receptor has shown to be upregulated in hypoxia, which indicates that the ischemic tissue will be primed for activation by HGF/SF (13). These results indicate that HGF/SF could play an important role in pro-angiogenic therapy. However, HGF/SF is a heparin-binding protein with a complex multidomain structure consisting of an N-terminal (N) domain, four copies of the kringle domain (Kl to K4) and a C-terminal domain homologous to that of serine proteineases (SP) (Figure 1A) (14). Biologically active HGF/SF is produced by proteolytic cleavage of the linker connecting the fourth kringle and the SP domain yielding a two-chain (α/β) heterdimer in which both the a and the β chain display considerable heterogeneity due to five putative glycolsylation sites: three in the a- chain (residues 289,397 and 471) and two in the β-chain (residues 561 and 648). NK1 fragment is an alternatively spliced variant of the HGF/SF transcript encoding the N and Kl domains, that lacks glycosylation sites and behaves as a partial receptor agonist in the presence of heparin sulphate co-receptor or its structural analogue heparin (15). NK1 contains two heparin binding sites: one in the N domain and one in the Kl domain comprising amino acids K132, R134 and R181 (14, 16). Deletion of the heparin-binding site of the Kl domain, through reverse charge mutations of K132 and R134 yielded the protein 1K1 (K132E:R134E), a stable and non-glycosylated agonist of the Met receptor with potent activity in vitro and in vivo (17).
[0029] As used herein, the "1K1" fragment of Hepatocyte Growth Factor/Scatter Factor (HGF/SF) refers to an NK1 fragment of Hepatocyte Growth Factor/Scatter Factor (HGF/SF) (SEQ ID NO: 2) that contains at least part of the N-terminal domain and at least part the first Kringle (Kl) domain of HGF/SF where the heparin binding site of the Kl domain has been eliminated, e.g. by insertion of a reverse charge mutation, see U.S. patent No. 7,795,214. In one embodiment the 1K1 fragment is SEQ ID NO: 1, In some embodiments the 1K1 fragment has at least 70% similarity, at least 80% similarity, at least 90% similarity, or at least 95% similarity to the polypeptide of SEQ ID NO: 1. As known in the art "similarity" between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. In one embodiment the 1K1 fragment has at least 50% amino acid sequence identity, at least 60% identity, at least 70% identity, at least 80% identity, at least 90% identity, or at least 95% identity to the amino acid sequence of SEQ ID NO: 1. Amino acid sequence additions to the 1K1 fragment used herein are also contemplated.
[0030] The 1K1 fragment useful herein has angiogenic activity, i.e. promotes
angiogenesis, for example by stimulating growth and proliferation of vascular endothelial cells. Angiogenic activity can be determined by using routine assays in the art, including any of those described in the accompanying Examples. Additional assays include, but are not limited to, the cornea pocket assay that involves the use of a non- vascularized mouse eye (e.g. Kenyon et al., Invest Opthalmol. Vis. Sci. 37:625, 1996) or the rabbit eye (e.g., see Gaudric et al. Ophthal. Res. 24:181, 1992), This assay has the advantage that new blood vessels are easily detected and essentially must be newly formed blood vessels in the normally avascular cornea. Another assay involves the use of chicken chorioallantoic membrane (the CAM assay; see Wilting et al., Anat. Embryol. 183:259, 1991). Other assays in the rat, such as the rat aortic ring model, provide reproducible assays that are often utilized to identify angiogenic agonists (e.g. see Lichtenberg et al., Pharmacol Toxicol. 84:34, 1999). Activity can also be assessed by monitoring the ability to act as a MET receptor antagonist, for example as described in U.S. Patent No. 7,795,214. [0031] As used herein, the term "angiogenesis," indicates the growth or formation of blood vessels. Angiogenesis includes the growth of new blood vessels from pre-existing vessels, as well as vasculogenesis, which refers to spontaneous blood-vessel formation, and intussusception, which refers to new blood vessel formation by splitting off existing ones. Angiogenesis encompasses "neovascularization", "regeneration of blood vessels," "generation of new blood vessels", "revascularization," and "increased collateral circulation."
[0032] The terms "angiogenesis agent" and "angiogenic agent" refers to any compound or substance that stimulates, accelerates, promotes, or increases angiogenesis, whether alone or in combination with another substance.
[0033] Embodiments of the invention provide for pharmaceutical compositions of nanoparticles that comprise a biocompatible polymer and an effective amount of 1K1 protein fragment of HGF/SF.
[0034] 1K1 protein can be produced by methods well known to those of skill in the art. Sequences encoding all or part of the polypeptides of the invention and/or its regulatory elements can be readily prepared by the skilled person using the information and references contained herein and techniques known in the art (for example, see Sambrook, Fritsch and Maniatis, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1989, and Ausubel et al, Short Protocols in Molecular Biology, John Wiley and Sons, 1992). These techniques include the use of site directed mutagenesis of nucleic acid encoding NK1 (SEQ ID NO: 2). The nucleic acid encoding the 1K1 fragment of HGF/SF (e.g. SEQ ID NO:l) can be incorporated into a recombinant expression vector e.g. operably linked to a control sequence which is capable of providing for the expression of the coding sequence by the host cell.
[0035] The term "operably linked" refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under condition compatible with the control sequences.
[0036] Suitable vectors can be chosen or constructed, containing appropriate regulatory sequences, including promoter sequences, terminator fragments, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate. Vectors may be plasmids, viral e.g. phage phagemid or baculoviral, cosmids, YACs, BACs, or PACs as appropriate. The vectors may be provided with an origin of replication, optionally a promoter for the expression of the said polynucleotide and optionally a regulator of the promoter. The vectors may contain one or more selectable marker genes, for example an ampicillin resistance gene in the case of a bacterial plasmid or a neomycin resistance gene for a mammalian vector. Vectors may be used in vitro, for example for the production of RNA or used to transfect or transform a host cell. Systems for cloning and expression of a polypeptide in a variety of different host cells are well known.
[0037] Suitable host cells include bacteria, eukaryotic cells such as mammalian and yeast, and baculo virus systems. Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary cells, HeLa cells, baby hamster kidney cells, COS cells and many others.
[0038] Promoters and other expression regulation signals may be selected to be compatible with the host cell for which the expression vector is designed. For example, yeast promoters include S. cerevisiae GAL4 and ADH promoters, S. pombe nmtl and adh promoter. Mammalian promoters include the metallothionein promoter which responds to heavy metals such as cadmium. Viral promoters such as the SV40 large T antigen promoter or adenovirus promoters may also be used. All these promoters are readily available in the art.
[0039] The vectors may include other sequences so that the polypeptide is produced as a fusion and/or nucleic acid encoding secretion signals so that the polypeptide produced in the host cell is secreted from the cell. For example, in one embodiment the polypeptide is modified, for example by the addition of histidine residues, to assist their purification.
[0040] Vectors may be introduced into a suitable host cell as described above to provide for expression of the 1K1 polypeptide. The cells will be chosen to be compatible with the said vector and may for example be bacterial, yeast (e.g. Pichia pastoris), insect or mammalian.
[0041] The introduction of vectors into a host cell is followed by causing or allowing expression from the nucleic acid, e.g. by culturing host cells under conditions for expression of the gene, so that the encoded polypeptide is produced. If the polypeptide is expressed coupled to an appropriate signal leader peptide it may be secreted from the cell into the culture medium. Following production by expression, a polypeptide may be isolated and/or purified from the host cell and/or culture medium. In one embodiment the 1K1 polypeptide is in a substantially purified form, in which case it will generally comprise the polypeptide in a preparation in which more than 90%, e.g. 95%, 98% or 99% of the polypeptide in the preparation. The IK1 polypeptide can then be assayed for angiogenic activity. [0042] As used herein, "efffective amount" includes the amount of an agent, e.g., 1K1 polypeptide, which allow it to perform its intended function, e.g., stimulating angiogenesis in conditions associated with insufficient neovascularization as described herein. The effective amount will depend upon a number of factors, including mode of delivery, biological activity, age, body weight, sex, general health, severity of the condition to be treated, as well as appropriate pharmacokinetic properties. It is understood that an effective amount of an agent, such as 1K1 fragment, may be a different amount when the agent is used alone as compared to when it is used in combination with another angiogenic agent.
[0043] The term "pharmaceutical composition" refers to a formulation of a compound into a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefore.
[0044] As used herein the term "nanoparticle" refers to a particulate of not greater thanl,000 nm in size, e.g. as dertermined by a light scattering method. In one embodiment, the nanoparticles have an average particle size of about 2.5 to about 999 nm. In one embodiment, the nanoparticles have an average particle size of about.5 to about 500 nm. In some embodiments the size range is from about 25 to about 300 nm, about 50 to about 200 nm. In some embodiments, the nanoparticle size is no greater than 500 nm, no greater than 300 nm, no greater than 200 nm, no greater than 160 nm, or no greater than 150nm, or no greater than 140 nm.
[0045] The nanoparticles of the pharmaceutical composition described herein comprise a biocompatible (e.g. biodegradable) polymer that encapsulates the protein fragment 1K1. As used herein the term "encapsulate" or "entrapped" refers to the biocompatible polymer covering, or alternatively encasing 1K1 protein. In one embodiment, the biodegradable polymer fully covers, i.e. encases, 1K1 protein, e.g. with an outer spherical polymer layer. In one embodiment, the 1K1 protein is only partially covered by the biocompatible polymer. In one embodiment, the 1K1 protein is on the surface of the biocompatible polymer. In one embodiment, the 1K1 protein is found within the biocompatible polymer.
[0046] In some embodiments, the ratio of polymer to 1K1 in the composition can range from about 1:20 to about 20:1. The ratio can be in the range of from about 1:120 to about 10:1, from about 1:30 to about 5:1, or from about 1:20 to about 1:1. In some embodiments, the ratio is in the range of about 1:20 to about 1:5. In some embodiments, the ratio is around 1:13. In one embodiment, the ratio is in the range from about 1:1 to about 3:1. It is to be understood that ratio can be based on weight, volume and/or moles. [0047] In some embodiments, at least about 5% of 1K1, at least about 10% of 1K1, at least about 15% of 1K1, at least about 20% of 1K1, at least about 30% of 1K1, at least about 40% of 1K1, at least about 50% of 1K1, at least about 60% oflKl, at least about 70% of 1K1, at least about 80% oflKl, at least about 90% of 1K1, at least about 95% of 1K1, or at least about 97%, about 98%, or about 99% of 1K1, or 100% of 1K1 that is associated with the nanoparticle, can be released from the nanoparticle over a pre-defined period of time. In such embodiments, the desired amount of 1K1 can be released from the nanoparticle over a period of at least about 6 hours, at least about 12 hours, at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 1 week, at least about 2 weeks, at least about 1 month or at least about 2 months. In one embodiment, the 1K1 is released from the nanoparticle over a period of at least about 1 week.
[0048] In one embodiment, the therapeutically effective amount of 1K1 that is released from the nanoparticles over the defined period of time is sufficient to result in sustained phoshorylation of cellular ERK over the defined period of time of release. Means for measuring ERK phosphorylation are well known to those of skill in the art. In one
embodiment, ERK phosphorylation is measured by Western Blot analysis, see for e.g.
Example 1. In one embodiment, phosphorylation of cellular ERK is increased by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, as compared to ERK phosphorylation observed using an equivalent concentration of IK1 not associated with nanoparticles.
[0049] In one embodiment, the therapeutically effective amount of 1K1 that is released over the defined period of time is sufficient to increase angiogenesis in a subject as compared to the angiogenesis in the absence of the compound by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%.
[0050] Suitable biocompatible polymers, a number of which are biodegradable include, for example, poly(lactides), poly(glycolides), poly(lactide-co-glycolides), poly(lactic acids), poly(glycolic acids), poly(lactic acid-co-glycolic acids), polycaprolactone, polycarbonates, polyesteramides, polyanhydrides, poly( amides), poly( amino acids), polyethylene glycol and derivatives thereof, polyorthoesters, polyacetals, polycyanoacrylates, polyetheresters, poly(dioxanone)s, poly(alkylene alkylates), copolymers of polyethylene glycol and polyorthoesters, biodegradable polyurethanes. Other polymers include poly(ethers) such as poly)ethylene oxide), poly(ethylene glycol), and poly(tetramethylene oxide); vinyl polymers- poly(acrylates) and poly(methacrylates) such as methyl, ethyl, other alkyl, hydroxyethyl methacrylate, acrylic and methacrylic acids, and others such as poly(vinyl alcohol), poly( vinyl pyrolidone), and poly( vinyl acetate); poly(urethanes); cellulose and its derivatives such as alkyl, hydroxyalkyl, ethers, esters, nitrocellulose, and various cellulose acetates; poly(siloxanes), etc. Other polymeric materials include those based on naturally occurring materials such as polysaccharides (e.g., alginate), chitosan, agarose, hyaluronic acid, gelatin, collagen, and/or other proteins, and mixtures and/or modified forms thereof. Chemical derivatives of any of the polymers disclosed herein (e.g., substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art) are also encompassed. Polymers may have various mean chain lengths, resulting in a variety of intrinsic viscosities and polymeric properties, for example, 5,000 to 200,000, preferably 15,000 to 25,000 of molecular weight. Furthermore, blends, graft polymers, and copolymers, including block copolymers of any of these polymers can be used. It will be appreciated that a vast number of different polymer variations are available. It will be understood that certain of these polymers require use of appropriate initiators or cross-linking agents in order to polymerize.
[0051] In one embodiment, the biocompatible polymer used in the compositions described herein is biodegradable such that after administration to a subject the matrix is degraded and metabolized thereby releasing 1K1 protein over a prolonged period of time. Some examples of biodegradable polymers useful in the present invention include hydroxyaliphatic carboxylic acids, either homo- or copolymers, such as poly(lactic acid), poly(glycolic acid), Poly(dl-lactide/glycolide, poly(ethylene glycol); polysaccharides, e.g. lectins, glycosaminoglycans, e.g. chitosan; celluloses, acrylate polymers, and the like. The selection of polymer may be determined by the desired rate of degradation after
administration to a desired tissue.
[0052] In general, the following criteria are important for selection of a material to be used for delivery of the active agent(s): (1) minimal or no cytotoxicity, (2) minimal or no elicitation of immune responses and inflammation, (3) compatibility with aqueous solutions and physiological conditions, (4) compatibility of the material and its processing methods with the stability of the 1K1 protein or other agents to be incorporated. In the methods described herein it is desirable to use a material with a controlled rate of biodegradation. Features such as cross-linking and monomer concentration may be selected to provide a desired rate of degradation and release of the agent. Release of agent can be monitored by methods well known to those of skill in the art or by methods described herein, see e.g.
Example 1.
[0053] In one embodiment, the polymer is a biocompatible polyester. The term
"biocompatible polyester" includes any polyester prepared by polymerizing one or more monomers, not limited to, but including D,L-lactide, D-lactide, L-lactide, D,L-lactic acid, D- lactic acid, L-lactic acid, glycolide, glycolic acid, ε-caprolactone, ε-hydroxy hexanoic acid, γ- butyrolactone, γ-hydroxybutylic acid, δ-valerolactone, δ-hydroxy valeric acid,
hydroxybutylic acid, malic acid, etc.
[0054] In one embodiment, the biocompatible polymer is poly-lactic acid, poly glycolic acid, lactic acid-glycolic acid copolymer or lactic acid- asp aragine acid copolymer, e.g. PLGA or PEG/CS-PLGA (polyethylene glycol/chitosan derivative of PLGA). The term "PLGA", used herein, means copolymer of lactic acid or lactide and glycolic acid or glycolide in any ratio of, for example, 1:99 to 99:1, preferably 3:1. They are known also as polylactide- glycolide copolymers. PLGA may be prepared synthetically from any monomer by using conventional methods or commercially available. PLGA which may be commercially available, includes, for example, PLGA 7520 (lactic acid:glycolic acid=75:25, mean molecular weight: 20,000, Wako Junyaku, Japan). In one embodiment, the PLGA contains 25 to 65% by weight of lactic acid and glycolic acid.
[0055] In one embodiment, the surface of the biocompatible polymer is modified, e.g. by polyethylene glycol (PEG), to result in increased affinity of water-soluble protein to the polymers, which affords easier encapsulation. Other means of modification are know to those of skill in the art.
[0056] In some embodiments, the nanoparticles further comprise a target specific tag (target moiety) for targeting the nanoparticles to a site of interest, e.g. tissue, cell etc. A "targeting moiety", as used herein, refers to all molecules capable of specifically binding to a particular target molecule and forming a bound complex. Thus, the ligand and its
corresponding target molecule form a specific binding pair.
[0057] The term "specific binding" refers to that binding which occurs between such paired species as enzyme/substrate, receptor/agonist, antibody/antigen, and
lectin/carbohydrate which may be mediated by covalent or non-covalent interactions or a combination of covalent and non-covalent interactions. When the interaction of the two species produces a non-covalently bound complex, the binding which occurs is typically electrostatic, hydrogen-bonding, or the result of lipophilic interactions. Accordingly, "specific binding" occurs between a paired species where there is interaction between the two which produces a bound complex having the characteristics of an antibody/antigen or enzyme/substrate interaction. In particular, the specific binding is characterized by the binding of one member of a pair to a particular species and to no other species within the family of compounds to which the corresponding member of the binding member belongs. Thus, for example, an antibody preferably binds to a single epitope and to no other epitope within the family of proteins.
[0058] Examples of targeting moieties include, but are not limited to antibodies, lymphokines, cytokines, receptor proteins such as CD4 and CD8, solubilized receptor proteins such as soluble CD4, hormones, growth factors, peptidomimetics, synthetic ligands, and the like which specifically bind desired target cells, and nucleic acids which bind corresponding nucleic acids through base pair complementarity. Targeting moieties of particular interest include peptidomimetics, peptides, antibodies and antibody fragments (e.g. the Fab' fragment). For example, β-D-lactose has been attached on the surface to target the aloglysoprotein (ASG) found in liver cells which are in contact with the circulating blood pool.
[0059] Cellular targets include tissue specific cell surface molecules, for targeting to specific sites of interest, e.g. neural tissue, liver tissue, muscle cells, and the like. Endothelial cells are a target of particular interest, in particular endothelial cells found in blood vessels, or in the proximity of ischemic tissues, or near wounds. Among the markers present on endothelial cells are integrins, of which a number of different subtypes have been
characterized. Integrins can be specific for endothelial cells involved in particular
physiological processes, for example certain integrins are associated with inflammation and leukocyte trafficking (see Alon & Feigelson (2002) Semin Immunol. 14(2):93-104; and Johnston & Butcher (2002) Semin Immunol 14(2):83-92, herein incorporated by reference). Targeting moieties specific for molecules such as ICAM-1, VCAM-1, etc. may be used to target vessels in inflamed tissues. Targeting nanoparticles are described in, for example, U.S. patent application 2004/0126900.
Preparation of Nanoparticles
[0060] The nanopartcles described herein, may be prepared using a variety of methods that are well known and practiced by those of skill in the art. Example methods for formation of nanoparticles that encapsulate bioactive materials, include, but are not limited to, emulsion polymerization of a monomer, interfacial polymerization, interfacial polycondensation, emulsification/solvent evaporation, phase separation, solvent displacement and interfacial deposition, emulsification/dolvent diffusion (ESD), salting out methods, spray-drying, and freeze-drying, Suitable methods and guidance are provided in, e.g. Reis et al. Nano capsulation I. "Methods for the preparation of drug-loaded polymeric nanoparticles", Nanomedicine: Nanotechnology, Biology, and Medicine 2 (2006) 8-21; and Muthu,
"Nanoparticles based on PLGA and its co-polymer: an overview" Asian J. Pharmaceutics October-December 2009, p.266-273. See also U.S. patent Application Ser. No. 20080095856 and 20080131513. Engineering of specific properties into nanoparticles, such as flowability, dissolution rate, dispersability, chemical reactivity, bioefficacy, and hydrophilicity are available for a range of applications, (see Davies et al. Adv. Mater. 1998, 10, 1264-1270; Wang et al. J. Controlled Release, 1999, 57, 9-18 and Zambaux M, et al. Influence of experimental parameters on the characteristics of poly(lactic acid) nanoparticles prepared by double emulsion method. J. Control. Release 1998; 50: 31-40.).
[0061] One method for forming nanoparticles is the solvent evaporation method. In this method, the polymer is dissolved in an organic solvent such as dichloromethane, chloroform or ethyl acetate which is also used as the solvent for dissolving the hydrophobic drug. The mixture of polymer and drug solution is then emulsified in an aqueous solution containing a surfactant or emulsifying agent to form an oil in water (o/w) emulsion. After the formation of stable emulsion, the organic solvent is evaporated either by reducing the pressure or by continuous stirring. Particle size can be influenced by the type and concentrations of stabilizer, homogenizer speed and polymer concentration. In order to produce small particle size, often a high-speed homogenization or ultrasonication may be employed.
[0062] The spontaneous emulsification or solvent diffusion method is a modified version of solvent evaporation method. In this method, a watermiscible solvent along with a small amount of the water immiscible organic solvent is used as an oil phase. Due to the spontaneous diffusion of solvents an interfacial turbulence is created between the two phases leading to the formation of small particles. As the concentration of water miscible solvent increases, a decrease in the size of particle can be achieved. Both solvent evaporation and solvent diffusion methods can be used for hydrophobic or hydrophilic drugs. In the case of hydrophilic drug, a multiple w/o/w emulsion needs to be formed with the drug dissolved in the internal aqueous phase.
[0063] In one embodiment, the nanoparticles comprising 1K1 protein and biocompatible polymer are prepared by a double emulsion-solvent diffusion method, which is well practiced in the art (See for example, Rocha et al, (2008) Biomaterials, 29:2884-2890). For example, a water- in-oil-in- water (w/o/w) solvent evaporation system, is used to form the nanoparticles. The biocompatible polymer is combined with an organic solvent, such as ethyl acetate, dimethylchloride (also called methylene chloride and dichloromethane), acetonitrile, acetone, chloroform, and the like. The polymer may be provided in about a 2-15% solution, in organic solvent. The more viscous the polymer, the slower release of active agent, e.g. IKl, from the nanoparticle. An approximately equal amount of an drug/agent solution is added and the polymer solution emulsified using e.g. sonication.
[0064] The concentration of active agent/s (e.g. IKl and optionally an additional active agent) and the polymer, as well as the active agent/polymer ratio, allows for control of particle size and encapsulation yield. The concentrations are selected to provide for the desired end product by optimization, as is known in the art. In general, a lower concentration of active agent is selected for smaller particle sizes, and a higher concentration for larger particle sizes. A higher ratio of polymer to active agent will provide for a thicker polymer encapsulation, while a lower ratio of polymer to active agent will provide for a thinner coating, which in turn effects release. The concentration of IKl will usually be at least about 0.001 mg/ml, more usually at least about 0.01 mg/ml, at least about 0.1 mg/ml, or 1 mg/ml., and not more than about 100 mg/ml, usually not more than about 10 mg/ml. The
concentration of polymer will usually be at least about 0.01 mg/ml, more usually at least about 0.1 mg/ml, at least about 1 mg/ml, and not more than about 100 mg/ml, usually not more than about 50 mg/ml. The ratio of compound to polymer as a weight percent will vary, from around about 1:1000; about 1:500; about 1:100, about 1:50; about 1:10; about 1:20, about 1:5, and the like.
[0065] The emulsion is then combined with a larger volume of an aqueous solution of an emulsion stabilizer such as polyvinyl alcohol (PVA) or polyvinyl pyrrolidone. The emulsion stabilizer is typically provided in about a 2-15% solution, more typically about a 4-10% solution. The mixture is then homogenized to produce a stable w/o/w double emulsion.
Organic solvents are then evaporated; for example, see Example 1. Nanoparticles can be collected by centrifugation.
[0066] The formulation parameters can be manipulated to adjust particle size and rate of release of IKl. See Zambaux M, et al. Influence of experimental parameters on the characteristics of poly(lactic acid) nanoparticles prepared by double emulsion method. J. Control. Release 1998; 50: 31-40, herein incorporated by reference in its entirety. Small particles are produced by low aqueous phase volumes with high concentrations of PVA.
[0067] The nanoparticles may be converted into a redispersible aggregate powder (nanocomposit) by powderization through lyophilization, etc. In one embodiment, the nanoparticles are redispersiably complexed with an organic or inorganic substance and dried. For example, by using a sugar alcohol or sucrose, variation of the loading rate may be effectively controlled. Easiness of handling would be improved due to the sugar alcohol functioning as a filler. The sugar alcohols include, but not limited to, mannitol, trehalose, sorbitol, erythritol, maltitol, xylitol, etc., preferably trehalose. Conversion of the
nanoparticles to aggregates simplify handling and they may be reconstituted into highly reactive nanoparticles prior to use by contacting them with water. Alternatively, instead of lyophilization, the nanoparticles may converted into a complex by the fluid bed drying granulation method (using, for example, Aggromaster AGM, Hosokawamicron, Japan) to form redispersible aggregates.
[0068] In one embodiment, the nanoparticles contain the bioactive substance (e.g. 1K1 protein) at a ratio from 0.1 to 99% (w/v), preferably from 0.1 to 30% (w/v), and more preferably from 1 to 10% (w/v). In one embodiment, the bioactive substance includes 1K1 protein and one or more additional therapeutic agents. Examples of additional therapeutic agents that can be used in the pharmaceutical compositions include, but are not limited to, neovascular agents (e.g. other agents known to increase vascularization), antibiotics, antiinflammatory agents, and vitamins. Alternatively the pharmaceutical compositions can be formulated with an additional therapeutic agent, formulated as nanoparticles or not. In general, the additional agent should not contradict the angiogenic promoting effect of 1K1.
[0069] As used herein, the term "therapeutic agent" refers to a substance used in the diagnosis, treatment, or prevention of a disease. Any therapeutic agent known to those of ordinary skill in the art to be of benefit in the diagnosis, treatment or prevention of a disease is contemplated as a therapeutic agent in the context of the present invention. Therapeutic agents include pharmaceutically active compounds, hormones, growth factors, enzymes, DNA, plasmid DNA, RNA, siRNA, viruses, proteins, lipids, pro-inflammatory molecules, antibodies, antibiotics, anti-inflammatory agents, anti-sense nucleotides and transforming nucleic acids or combinations thereof. Any of the therapeutic agents may be combined to the extent such combination is biologically compatible.
Pharmaceutical Compositions
[0070] In one embodiment, 1K1 protein is delivered to a subject using a pharmaceutical composition that is a sustained release formulation, e.g. sustained release formulations other than nano-particles. Carriers suitable for sustained-release formulations include, but are not limited to, capsules, microspheres, particles, gels, coatings, matrices, wafers, pills or other pharmaceutical delivery compositions. Examples of such sustained-release formulations have been described previously, for example, in U.S. Pat. Nos. 6,953,593, 6,946,146, 6,656,508, 6,541,033, 6,451,346, the contents of which are incorporated herein by reference. Many methods for preparation of a sustained-release formulation are known in the art, and are disclosed in Remington's Pharmaceutical Sciences (18th ed.; Mack Publishing Company, Eaton, Pa., 1990), incorporated herein by reference, are well known to those of skill in the art.
[0071] For example, 1K1 can be entrapped in semipermeable matrices of solid
hydrophobic polymers. The matrices can be shaped into films or microcapsules. Examples of such matrices include, but are not limited to, polyesters, copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al., Biopolymers 22:547-556 (1983)), polylactides (U.S. Pat. No. 3,773,919 and EP 58,481), polylactate polyglycolate (PLGA) such as polylactide-co-glycolide (see, for example, U.S. Pat. Nos. 4,767,628 and 5,654,008), hydrogels (see, for example, Langer et al. (1981) J. Biomed. Mater. Res. 15:167-277; Langer, Chem. Tech. 12:98-105 (1982)), non-degradable ethylene-vinyl acetate (e.g. ethylene vinyl acetate disks and poly(ethylene-co -vinyl acetate)), degradable lactic acid-glycolic acid copolyers such as the Lupron Depot™, poly-D-(−)-3-hydroxybutyric acid (EP
133,988), hyaluronic acid gels (see, for example, U.S. Pat. No. 4,636,524), alginic acid suspensions, polyorthoesters (POE), and the like.
[0072] Suitable microcapsules capable of encapsulating 1K1 may also include
hydroxymethylcellulose or gelatin-microcapsules and polymethyl methacrylate
microcapsules prepared by coacervation techniques or by interfacial polymerization. In addition, microemulsions or colloidal drug delivery systems such as liposomes and albumin microspheres, may also be used. See Remington's Pharmaceutical Sciences (18(th )ed.; Mack Publishing Company Co., Eaton, Pa., 1990). Other preferred sustained-release compositions employ a bioadhesive to retain 1K1 at the site of administration.
[0073] The sustained-release formulation may comprise a biodegradable polymer into which the 1K1 agent is disposed, which may provide for non-immediate release. Non- limiting examples of biodegradable polymers suitable for the sustained-release formulations include poly(alpha-hydroxy acids), poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide (PG), polyethylene glycol (PEG) conjugates of poly(alpha-hydroxy acids), polyorthoesters, polyaspirins, polyphosphagenes, collagen, starch, chitosans, gelatin, alginates, dextrans, vinylpyrrolidone, polyvinyl alcohol (PVA), PVA-g-PLGA, PEGT-PBT copolymer (polyactive), methacrylates, poly(N-isopropylacrylamide), PEO-PPO-PEO
(pluronics), PEO-PPO-PAA copolymers, PLGA-PEO-PLGA, polyorthoesters (POE), or any combinations thereof, as described, for example, in the U.S. Pat. No. 6,991,654 and U.S. Pat. Appl. No. 20050187631, each of which is incorporated herein by reference in its entirety.
[0074] A person of ordinary skill will appreciate that different combinations of the sustained-release formulations are also suitable for this invention. For example, the practitioner may formulate an 1K1 protein as a combination of a gel and microspheres loaded with the at least one lkl, wherein the combination of gel and microspheres are placed in the target site. Depending on the carrier, the sustained-release formulations, and the total amount of 1K1, the 1K1 can be released over a period ranging between about one day to about six months is possible.
[0075] In one embodiment, the amount of 1K1 released over a defined period from the sustained release formulation is sufficient to result in sustained phosphorylation of cellular ERK over a defined period of time. Methods for assessing ERK phosphorylation are well known to those of skill in the art, and dosages can be easily determined. In one embodiment, using the sustained release formulation, tubulogenesis in the subject is increased by at least 5%, at least 10%, at least 30%, at least 50%, at least 60%, at least 70%, at least 80% , at least 90%, as compared to tubulogenesis observed using an equivalent dose of 1K1 that is not released in a sustained manner over a defined period of time. The effective amount of 1K1 released over a defined period of time will range from about 0.01 mg/kg to about 1000 mg/kg, from about .1 mg/kg to about lOOmg/kg, from about 0.1 mg/kg to about 200 mg/kg, from about 0.2 mg/kg to about 20 mg/kg.
[0076] The nanoparticles of the invention may be formulated into a pharmaceutical composition for administration to a subject. Pharmaceutical compositions can include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers of diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, buffered water, physiological saline, PBS, Ringer's solution, dextrose solution, and Hank's solution. In addition, the pharmaceutical composition or formulation can include other carriers, or non-toxic, nontherapeutic, nonimmunogenic stabilizers, excipients and the like. The compositions can also include additional substances to approximate physiological conditions, such as pH adjusting and buffering agents, toxicity adjusting agents, wetting agents and detergents.
[0077] The pharmaceutical composition can be formulated using methods well known to those of skill in the art. The pharmaceutical composition may comprise 0.1% to 99% by weight of the active ingredient, e.g. nanoparticles comprising 1K1 fragment. In one embodiment, the composition comprises from about 1% to about 80%, about 1% to about 70%, or about 1% to about 50%, or about 1% to about 20% by weight of active ingredient. Further guidance regarding formulations that are suitable for various types of administration can be found in Remington's Pharmaceutical Sciences, Mace Publishing Company,
Philadelphia, Pa., 17th ed. (1985). For a brief review of methods for drug delivery, see, Langer, Science 249:1527-1533 (1990).
[0078] The pharmaceutical compositions described herein can be administered in a variety of different ways. The pharmaceutical compositions of the present invention can be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), lozenges, dragees, capsules, pills, tablets (e.g., those targeted for buccal, sublingual, and systemic absorption), boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; (8) transmucosally; or (9) nasally.
[0079] For oral administration, the active ingredient can be administered in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions. The active component(s) can be encapsulated in gelatin capsules together with inactive ingredients and powdered carriers, such as glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium saccharin, talcum, magnesium carbonate. Examples of additional inactive ingredients that may be added to provide desirable color, taste, stability, buffering capacity, dispersion or other known desirable features are red iron oxide, silica gel, sodium lauryl sulfate, titanium dioxide, and edible white ink. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric-coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance. [0080] The active ingredient, alone or in combination with other suitable components, can be made into aerosol formulations (i.e., they can be "nebulized") to be administered via inhalation. Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen.
[0081] Formulations suitable for parenteral administration, such as, for example, by intraarticular (in the joints), intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
[0082] The components used to formulate the pharmaceutical compositions are preferably of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food (NF) grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Moreover, compositions intended for in vivo use are sterile. To the extent that a given compound must be synthesized prior to use, the resulting product is typically substantially free of any potentially toxic agents, particularly any endotoxins, which may be present during the synthesis or purification process. Compositions for parental administration are also sterile, substantially isotonic and made under GMP conditions.
Methods for promoting angiogenesis in subjects with insufficient vascualrization.
[0083] The pharmaceutical compositions may be used for treatment or prevention of conditions characterized by insufficient vascularization of the affected tissues (or predisposition thereto), i.e., conditions in which neovascularization is needed to achieve sufficient vascularization in the affected tissue.
[0084] In certain embodiments, methods and compositions of the present invention are used for treatment or prevention of any of a variety of diseases and disorders that benefit from stimulation of angiogenesis or an increase in angiogenesis in biological matter. For example, compositions and methods of the present invention may be used to promote, enhance, or increase angiogenesis in biological matter in vitro or ex vivo, e.g., in the culture, storage, or generation of tissue or organs suitable for transplant into an organism such as a mammal.
[0085] Compositions and methods of the present invention may also be used to promote, enhance, or increase angiogenesis in vivo, e.g., at a wound site or a site within an organism subject to or at risk of ischemia or hypoxia, thereby increasing blood flow and oxygenation to the tissue subject to or at risk of ischemia and reducing or preventing tissue injury at the site.
[0086] In some embodiments, the present invention includes compositions and methods for treating or preventing pathological conditions, diseases, and disorders that would benefit from enhanced blood flow. Examples of such conditions include ischemia associated diseases. Examples of ischemia associated diseases include myocardial ischemia, peripheral ischemia, cerebral ischemia, and deep vein thrombosis.
[0087] Furthermore, in related embodiments, the present invention includes compositions and methods of treatment for wound healing, diabetes (e.g., diabetic foot ulcers), ocular disease or eye disorder, cardiac disease, congestive heart failure, myocardial ischemia, peripheral ischemia, lymphatic vascular disorders, coronary artery disease, stroke, angina and peripheral vascular disease. In one embodiment, the compositions and methods of the invention are used in wound healing or reconstructive surgery.
[0088] In one embodiment, the present invention includes a method for treatment of a condition associated with insufficient neovascularization by administering to a subject in need thereof, or cells, tissue, or an organ obtained from said subject, a pharmaceutical composition comprising the lKl-nanoparticles described herein. The administration is in an amount effective for stimulating or increasing angiogenesis. In particular embodiments, the subject is a mammal. In certain embodiments, the composition is administered locally, e.g., to a site within the subject that is in need of angiogenesis. Examples of such sites within a subject include wounds and tissue or organs subjected to or at risk of ischemia or hypoxia. In other embodiments, the lKl-nanoparticles are administered systemically. In further embodiments, the 1K1 nanoparticles are administered ex vivo to cells, tissue, or an organ obtained from the subject, and the cells, tissue or organ contacted with the nanoparticles and then transplanted back into the subject.
[0089] As used herein, "promoting angiogenesis", "enhancing angiogenesis" and "increasing angiogenesis" refers to an increase in number and formation of new blood vessels.
[0090] By "treatment", "prevention" or "amelioration" of a disease or disorder is meant delaying or preventing the onset of such a disease or disorder, reversing, alleviating, ameliorating, inhibiting, slowing down or stopping the progression, aggravation or deterioration the progression or severity of a condition associated with such a disease or disorder. In one embodiment, the symptoms of a disease or disorder are alleviated by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, or at least 50%. [0091] The phrase a "therapeutically-effective amount" as used herein means that amount 1K1 protein, or composition comprising 1K1 protein of the present invention, which is effective for producing some desired therapeutic effect in at least a sub-population of cells in a subject at a reasonable benefit/risk ratio applicable to any medical treatment. For example, an amount of a compound administered to a subject that is sufficient to produce a statistically significant, measurable change in at least one symptom associated with a condition of reduced vascularization. Symptoms will vary dependent upon the tissue affected. In general, ischemia of a tissue is associated with decreased oxygen, discomfort and pain in affected tissue and tissue death; cerebral ischemia, a condition in which there is insufficient blood flow to the brain, may be associated with impairments in vision, body movement, and speaking; myocardial ischemia may be associated with decreased pumping efficiency, heart attacks, and abnormal heart rhythms; mesenteric ischemic syndromes, which occur when blood flow to the bowel or gastrointestinal system (intestines) is reduce, are associated with abdominal pain, nausea and/or vomiting and bloody stools; and so on. A skilled artisan can readily determine an appropriate symptom for measurement of a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capability of those skilled in the art. Generally, a therapeutically effective amount can vary with the subject's history, age, condition, sex, as well as the severity and type of the medical condition in the subject, and administration of other pharmaceutically active agents.
[0092] In one embodiment, the therapeutically effective amount is sufficient to increase angiogenesis in a subject (as measured in the subject, or as measured by an established angiogenesis assay), by at least about 5%, by at least about 10%, by at least about 15%, by at least about 20%, by at least about 25%, by at least about 30%, by at least about 35%, by at least about 40%, by at least about 50%, by at least about 55%, by at least about 60%, by at least about 65%, by at least about 70%, by at least about 75%, by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95%, or by about 100% as compared to angiogenesis in the absence of the compound. The % increase in angiogenesis can be assessed in vitro (e.g. proliferation assay, in vitro tube assay, or in vivo (e.g. CAM assay, cornea assay, zebrafish model.) One example method for measuring angiogenesis in humans is by transient oxygen assays.
[0093] A therapeutically effective amount will typically range from about 0.01 mg/kg to about 1000 mg/kg, from about .1 mg/kg to about lOOmg/kg, from about 0.1 mg/kg to about 200 mg/kg, from about 0.2 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or more days. In some embodiments, the therapeutically effective amount is released from the composition over a period of time, e.g. hours, days or weeks. In one embodiment a therapeutically effective amount is release in about 2 days, about 3 days, about 4 days, about 5 days, or about 7 days.
[0094] In one embodiment, the present invention includes methods of promoting, enhancing, or increasing angiogenesis in biological matter, comprising contacting the biological matter with an effective amount of nanoparticles comprising 1K1. In particular embodiments, the biological matter is mammalian, e.g., mammalian cells, tissue, organ or animal (e.g. a human subject). In particular embodiments, the biological matter is an animal such as a mammal. In particular embodiments, the amount of angiogenesis is increased by at least 5%, at least 10%, at least 25%, at least 50%, at least 100%, at least 200%, at least 500% or at least 1000% as compared to in the absence of treatment with the nanoparticles comprising 1K1. Similarly, the amount of angiogenesis may be increased at least two-fold, at least three-fold, at least four-fold, at least five-fold, or at least 10-fold, as compared to in the absence of treatment with the nanoparticles comprising 1K1. The amount of angiogenesis may be readily determined using routine assays in the art, and as described herein.
[0095] Additional applications for in vivo use the compositions comprising 1K1- nanoparticles include vascularization of ischemic tissue such as ischemic heart tissue and ischemic peripheral tissue, and vascularization of chronic wounds, burns and transplanted tissue.
[0096] As used herein, the term "ischemia" refers to any condition associated with an inadequate flow of oxygenated blood to a part of the body. Ischemia occurs any time that blood flow to a tissue is reduced below a critical level. This reduction in blood flow can result from the following non-limiting conditions: (i) the blockage of a vessel by an embolus (blood clot); (ii) the blockage of a vessel due to atherosclerosis; (iii) the breakage of a blood vessel (a bleeding stroke); (iv) the blockage of a blood vessel due to vasoconstriction such as occurs during vasospasms and possibly, during transient ischemic attacks (TIA) and following subarachnoid hemorrhage. Further conditions in which ischemia occurs, include (i) during myocardial infarction (when the heart stops, the flow of blood to organs is reduced and ischemia results); (ii) trauma; (iii) during cardiac and neurosurgery (blood flow needs to be reduced or stopped to achieve the aims of surgery and iv) diabetic related ischaemia.
[0097] The pharmaceutical compositions can be administered for prophylactic and/or therapeutic treatments. Toxicity and therapeutic efficacy of the active ingredient can be determined according to standard pharmaceutical procedures in cell cultures and/or experimental animals, including, for example, determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred.
[0098] The data obtained from cell culture and/or animal studies can be used in formulating a range of dosages for humans, e.g. the cornea pocket assay that involves the use of a non-vascularized mouse eye (e.g. Kenyon et al., Invest Opthalmol. Vis. Sci. 37:625, 1996) or the rabbit eye (e.g., see Gaudric et al. Ophthal. Res. 24:181, 1992), This assay has the advantage that new blood vessels are easily detected and essentially must be newly formed blood vessels in the normally avascular cornea. Another assay involves the use of chicken chorioallantoic membrane (the CAM assay; see Wilting et al., Anat. Embryol.
183:259, 1991). Other assays in the rat, such as the rat aortic ring model, provide
reproducible assays that are often utilized to identify angiogenic agonists (e.g. see
Lichtenberg et al., Pharmacol Toxicol. 84:34, 1999). The dosage of the active ingredient typically lines within a range of circulating concentrations that include the ED50 with low toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. In some embodiments, the 1K1 nanoparticles are delivered locally, e.g. topical or local injection.
[0099] The effective amount of a therapeutic composition to be given to a particular subject will depend on a variety of factors, several of which will be different from patient to patient. A competent clinician will be able to determine an effective amount of a therapeutic agent. The compositions can be administered to the subject multiple times, e.g. in a series of more than one administration. For therapeutic compositions, regular periodic administration (e.g., every 2-3 days) will sometimes be required, or may be desirable to reduce toxicity. Repeated-dose regimens can be used, for example administration 1 to 4 times daily at a variety of frequencies such as daily, every other day, once a week, every other week, once a month, etc. Such frequencies may depend on factors known to the person skilled in the art and be easily determined by physician. Those of skill will readily appreciate that dose levels can vary as the severity of the symptoms and the susceptibility of the subject to side effects.
[00100] In one embodiment, the pharmaceutical composition of the invention is used for treatment of diabetic foot ulcers. The pharmaceutical IKl-nanoparticle composition may be applied directly to the wound site or in a pharmaceutically acceptable wound healing medicament. The composition may be administered locally or topically and delivered via a variety of means, for example via a spray, local injection, local infusion, cream, lotion, suspension, emulsion, gel, ointment, salve, stick, soap, liquid aerosol, powder aerosol, drops, paste, endoscopically or antimicrobial dressings such as bandages.
[00101] For topical administration, which is appropriate with regard to superficial lesions (e.g. diabetic foot ulcers) standard topical formulations are employed using, for example, 10 ng/ml-100 mg/ml solutions; the preferred range is 10 ug/ml-10 mg/ml. Such solutions would be applied up to 6 times a day to the affected area. In certain applications, such as burns, a single dose would be preferred. In other applications, such as ulcers, multiple doses may be preferred. The concentration of the ointment or other formulation depends, of course, on the severity of the wound or stage of disease and the nature of the subject. In most protocols, the dose is lowered with time to lessen likelihood of scarring. For example, the most severe wounds, such as third degree burns, are typically treated with a 100 ug/ml composition, but as healing begins, the dose is progressively dropped to approximately 10 ug/ml or lower, as the wound heals.
[00102] In treatment of a wound (e.g. a diabetic foot ulcer), a therapeutically effective amount may be an amount sufficient to decrease the size of the wound by at least about 5%, by at least about 10%, by at least about 15%, by at least about 20%, by at least about 25%, by at least about 30%, by at least about 35%, by at least about 40%, by at least about 50%, by at least about 55%, by at least about 60%, by at least about 65%, by at least about 70%, by at least about 75%, by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95%
[00103] The skilled artisan will recognize that there are a plurality of ways in which a human subject may be exposed to a protein, peptide, nanoparticle, composition, small molecule and/or other agent, in a sustained manner or otherwise, to effect treatment of the subject. Exemplary methods and devices for carrying out such exposure are described in W. Mark Saltzman, Drug Delivery: Engineering Principles for Drug Therapy (Topics in
Chemical Engineering), First ed., Oxford University Press (USA), 2001; L.V. Allen et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Ninth ed., Lippincott Williams & Wilkins, 2010; B. Wang et al., Drug Delivery: Principles and Applications, First ed., Wiley-Interscience, 2007; and K.K. Jain, Drug Delivery Systems, First ed., Humana Press, 2008, each one of which is incorporated by reference herein in its entirety. These references also provide exemplary exipients that optionally may be included with the protein, peptide, nanoparticle, composition, small molecule and/or other agent of the present invention. [00104] Each reference disclosed in the present application is incorporated by reference herein in its entirety.
Definitions
[00105] Unless otherwise defined herein, scientific and technical terms used in connection with the present application shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.
[00106] As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the invention, yet open to the inclusion of unspecified elements, whether essential or not.
[00107] As used herein the term "consisting essentially of refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the invention.
[00108] The term "consisting of refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
[00109] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used in connection with percentages may mean +1%. Furthermore, the term "about" can mean within +1% of a value.
[00110] The singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. It is further to be understood that all base sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids or polypeptides are approximate, and are provided for description.
[00111] Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The term "comprises" means "includes." The abbreviation, "e.g." is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation "e.g." is synonymous with the term "for example." [00112] The terms "decrease" , "reduced", "reduction" , "decrease" or "inhibit" are all used herein generally to mean a decrease by a statistically significant amount. However, for avoidance of doubt, ""reduced", "reduction" or "decrease" or "inhibit" means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (e.g. absent level as compared to a reference sample), or any decrease between 10- 100% as compared to a reference level.
[00113] The terms "increased" /'increase" or "enhance" or "activate" are all used herein to generally mean an increase by a statically significant amount; for the avoidance of any doubt, the terms "increased", "increase" or "enhance" or "activate" means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
[00114] The term "statistically significant" or "significantly" refers to statistical significance and generally means a two standard deviation (2SD) above or below a reference level. The term refers to statistical evidence that there is a difference. It is defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true. The decision is often made using the p-value.
[00115] As used herein, the term "polymer" is intended to include both oligomeric and polymeric species, i.e., compounds which include two or more monomeric units, which may be a homopolymer or a copolymer. The term "homopolymer" is a polymer incorporating a single species of monomer units. The term "copolymer" is a polymer constructed from two or more chemically distinct species of monomer units in the same polymer chain. A "block copolymer" is a polymer which incorporates two or more segments of two or more distinct species of homopolymers or copolymers.
[00116] The terms "polypeptide", "peptide", "amino acid sequence" and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified, for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation; or any other manipulation, such as conjugation with a labeling component.
[00117] As used herein the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including but not limited to glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics. Standard single or three letter codes are used to designate amino acids. The polypeptides of the present invention may be recombinant polypeptides, natural polypeptides, or synthetic polypeptides, preferably recombinant polypeptides. The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
[00118] The term "isolated" means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
[00119] As used herein, the term "linker" means an organic moiety that connects two parts of a compound. Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NH, C(O), C(0)NH, SO, S02, S02NH or a chain of atoms, such as substituted or unsubstituted Ci-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C6-Ci2 aryl, substituted or unsubstituted C5-Ci2 heteroaryl, substituted or unsubstituted C5-Ci2
heterocyclyl, substituted or unsubstituted C3-Ci2 cycloalkyl, where one or more methylenes can be interrupted or terminated by O, S, S(O), S02, NH, C(O).
[00120] By "treatment", "prevention" or "amelioration" of a disease or disorder is meant delaying or preventing the onset of such a disease or disorder, reversing, alleviating, ameliorating, inhibiting, slowing down or stopping the progression, aggravation or deterioration the progression or severity of a condition associated with such a disease or disorder. In one embodiment, the symptoms of a disease or disorder are alleviated by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, or at least 50%.
[00121] As used here, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication,
commensurate with a reasonable benefit/risk ratio.
[00122] As used here, the term "pharmaceutically-acceptable carrier" means a
pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL and LDL; (22) C2-C12 alchols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as "excipient", "carrier", "pharmaceutically acceptable carrier" or the like are used
interchangeably herein.
[00123] As used herein, the term "administer" refers to the placement of a composition into a subject by a method or route which results in at least partial localization of the composition at a desired site such that desired effect is produced. A composition described herein can be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, and topical (including buccal and sublingual) administration.
[00124] As used herein, a "subject" means a human or animal. Examples of subjects include primates (e.g., humans, and monkeys). Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents. In certain embodiments of the aspects described herein, the subject is a mammal, e.g., a primate, e.g., a human. The terms, "patient" and "subject" are used interchangeably herein. The terms, "patient" and "subject" are used interchangeably herein. A subject can be male or female.
[00125] Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of conditions or disorders associated with decreased spine/excitatory synapse formation and/or numbers. In addition, the methods and compositions described herein can be used to treat domesticated animals and/or pets.
[00126] The present invention can be defined in any of the following numbered paragraphs:
[00127] Paragraph 1 A pharmaceutical composition comprising a nanoparticle comprising a biocompatible polymer and an effective amount of 1K1 protein fragment of Hepatocyte Growth Factor/Scatter Factor, wherein the biodegradable polymer encapsulates the 1K1 protein.
[00128] Paragraph 2 The pharmaceutical composition of paragraph 1, wherein the 1K1 protein comprises SEQ ID. NO:l.
[00129] Paragraph 3 The pharmaceutical composition of any of paragraphs 1-2, wherein the nanoparticle has an average particle size of about 50 nm to about 500 nm.
[00130] Paragraph 4 The pharmaceutical composition of any of paragraphs 1-2, wherein nanoparticle has an average particle size of about 60 nm to about 150 nm.
[00131] Paragraph 5 The pharmaceutical composition of any of paragraphs 1-4, wherein the biocompatible polymer is a biodegradable polymer and the biodegradable polymer is selected from the group consisting of: polyesters, hydroxyaliphatic carboxylic acids, poly(lactic acid), poly(glycolic acid), poly(dl-lactide/glycolide, poly(ethylene glycol), polysaccharides, lectins, glycosaminoglycans, chitosan, celluloses and acrylate polymers.
[00132] Paragraph 6 The pharmaceutical composition of any of paragraphs 1-5, wherein the biocompatible polymer is a polyester.
[00133] Paragraph 7 The pharmaceutical composition of paragraphs 6, wherein the polyester comprises poly-lactic acid-glycolic acid copolymer (PLGA).
[00134] Paragraph 8 The pharmaceutical composition of any of paragraphs 1-7, wherein the 1K1 protein is released from the nanoparticle in a therapeutically effective amount over a defined period of time of days, weeks or months.
[00135] Paragraph 9 The pharmaceutical composition of any of paragraphs 1-8, wherein the 1K1 protein is released from the nanoparticle in a therapeutically effective amount over a defined period of time of about 2 days, about 3 days, about 4 days, about 5 days, about 7 days, or about 14 days.
[00136] Paragraph 10 The pharmaceutical composition of any of paragraphs 8-9, wherein the therapeutically effective amount that is released over the defined period of time is sufficient to result in sustained phoshorylation of cellular ERK over the defined period of time of release.
[00137] Paragraph 11 The pharmaceutical composition of any of paragraphs 8-10, wherein the therapeutically effective amount is sufficient to increase angiogenesis in a subject as compared to angiogenesis in the absence of the compound by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, or about 60%.
[00138] Paragraph 12 The pharmaceutical composition of any of paragraphs 8-11, wherein the therapeutically effective amount is dose of about O.lmg/kg to about 1000 mg/kg.
[00139] Paragraph 13 The pharmaceutical composition of any of paragraphs 1-12, wherein the composition is formulated for administration by a method selected from the group consisting of: topical administration, enteral administration, and parenteral administration.
[00140] Paragraph 14 The pharmaceutical composition of paragraph 13, wherein the composition formulated for topical administration is an ointment, lotion, spray, cream, or gel.
[00141] Paragraph 15 A method for treatment of a condition associated with insufficient vascularization comprising administration to a subject a therapeutically effective amount of a pharmaceutical composition of any of paragraphs 1-14.
[00142] Paragraph 16 The paragraph of 15, wherein the therapeutically effective amount is a dose of about O.lmg/kg to about 1000 mg/kg of lkl protein. [00143] Paragraph 17 The method of any of paragraphs 15-16, wherein the condition associated with insufficient vascularization is selected from the group consisting of:
cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, ischemia of tissues, coronary ischemia, peripheral arterial disease, limb ischemia, diabetic ulcers, gangrenes, wounds requiring neovascularization to facilitate healing, and Buerger's syndrome.
[00144] Paragraph 18 The method of any of paragraphs 15-17, wherein the condition is a diabetic foot ulcer.
[00145] Paragraph 19 The method of any of paragraphs 15-18, wherein the pharmaceutical composition is administered by topical administration, enteral administration, or parenteral administration.
[00146] Paragraph 20 The method of any of paragraphs 15-19, wherein the pharmaceutical composition is administered multiple times.
[00147] Paragraph 21 Use of the pharmaceutical composition of any of paragraphs 1-14 for treatment of a condition associated with insufficient vascularization.
[00148] Paragraph 22 The use of paragraph 21, wherein the condition associated with insufficient vascularization is selected from the group consisting of: cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, ischemia of tissues, coronary ischemia, peripheral arterial disease, limb ischemia, diabetic ulcers, gangrenes, wounds requiring neovascularization to facilitate healing, and Buerger's syndrome.
[00149] Paragraph 23 The use of paragraph 21, wherein the condition is a diabetic foot ulcer.
[00150] To the extent not already indicated, it will be understood by those of ordinary skill in the art that any one of the various embodiments herein described and illustrated can be further modified to incorporate features shown in any of the other embodiments disclosed herein.
[00151] Paragraph 24 A method for treatment of a conditions associated with insufficient vascularization comprising administrating to a subject an effective amount of 1K1 protein, wherein the 1K1 protein is administered in a sustained release formulation, e.g. having a dose sufficient to result in sustained phosphorylation of ERK and enhanced tubulogenesis, thereby significantly increasing angiogenesis in the subject.
[00152] The following examples illustrate some embodiments and aspects of the invention. It will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be performed without altering the spirit or scope of the invention, and such modifications and variations are encompassed within the scope of the invention as defined in the claims which follow. The following examples do not in any way limit the invention.
EXAMPLES
[00153] The examples presented herein relate to methods and compositions of inducing angiogenesis. As demonstrated herein, protein engineering and nanotechnology were combined to maximize the therapeutic potential of HGF/SF. The protein engineering experiments targeted NKl, an alternatively spliced variant of the HGF/SF transcript to yield the protein 1K1, a stable and nonglycosylated agonist of the Met receptor, which exerted strong angiogenic activity. Furthermore, 1K1 was encapsulated in nanoparticles engineered from biodegradable D, L-lactic acid-co-glycolic acid copolymer to enable sustained release. It is demonstrated herein that the temporal release enables unique downstream signaling through the MAPK pathway resulting in an enhanced angiogenic outcome in vitro and in vivo. Our discovery opens up the possibility of combining unique approaches to protein engineering and nanovectors for modulating vascularization in impaired ischemic condition.
Example 1. Engineering 1K1, a Nonglycosylated HGF/SF Variant.
[00154] In the presence of heparan sulfate or heparin, NKl behaves as a partial receptor agonist (14). In order to develop 1K1, a rational protein engineering approach was employed based on structural and functional studies of NKl (15), NKl-heparin complexes (16), and individual N and Kl domains (17). NKl binds heparin through two distinct sites: a high- affinity site located in the N domain and formed by the side chains of R73, K60, T61, R76, K62, and K58 and main chain atoms of T61, K63, and G79 (18) and a low-affinity site in the Kl domain comprising the side chains of K132, R134, K170, and R181. The mutant 1K1 carries two reverse charge mutations at K132 and R134 (1K1: K132E:R134E) that disrupt heparin binding to the kringle domain (Figures 1A to 1H) and displays increased signaling activity on several cell types compared to wild type (16). Without wishing to be bound by theory, the higher biological activity of 1K1 over NKl can be due to two reasons: Firstly, as a result of the loss of the heparin-binding site in Kl, heparin interacts solely with the primary binding site in the N domain and causes adjacent 1K1 dimers to bind hepatocyte growth factor receptor (MNNG (N-Methyl-N'-nitro-Nnitroso-guanidine) HOS Tranforming gene) (MET) on the same plane (Figure 1C), unlike NKl, where the heparin-binding sites of the Kl domain cause heparin to align adjacent NK1 dimers on a different plane (Figure IB). Hence, 1K1 can form oligomeric ligand-receptor complexes more readily than NK1. Secondly, preformed 1K1 -heparin complexes have greater binding affinity for MET than NK1- heparin complexes as a result of the different way in which the two proteins bind heparin as shown by surface plasmon resonance (Figures IE and IF) and velocity and sedimentation experiments (Figures 1G and 1H).
Example 2. 1K1 Induces Angiogenesis in Vitro by Binding to MET Kinase.
[00155] The angiogenic phenotype is the culmination of discrete cellular steps
characterized by endothelial cell chemoinvasion, proliferation, and tubulogenesis (18). As shown in Figures 2A to 2D, both HGF/SF and 1K1 induced endothelial cell proliferation. Furthermore, 1K1 -induced endothelial cell proliferation was inhibited by the MET kinase inhibitor, PHA 665752, in a concentration-dependent manner (Figure 2C), which confirmed that 1K1 induces angiogenesis through the Met receptor. Previous studies have revealed that MET binding by HGF/SF ligand results in downstream signaling through the PI3K and the MAPK (10). However, it is not known whether the PI3K and the MAPK pathways are activated in 1K1 -mediated endothelial cell proliferation. As such, the cells were pre-treated with LY294002 or PD98059, which inhibit PI3K and MAPK respectively. As shown in Figures 2A to 2D, both LY294002 and PD98059 inhibited 1K1 -induced cell proliferation in a concentration-dependent manner, indicating that both the PI3K and MAPK pathways are implicated during 1K1 -induced endothelial cell proliferation.
[00156] It was next sought to assess whether 1K1 can induce endothelial tubulogenesis. As shown in Figures 3 A to 3C, both HGF/SF and 1K1 induced significant tubulogenesis compared with vehicle treatment when the endothelial cells were plated on a three- dimensional Matrigel matrix. 1K1 -induced tubes were phenotypically longer and had more nodes as compared with HGF/SF (Images not shown). Consistent with the cell proliferation results, pretreatment of the human umbilical vein endothelial cells (HUVECs) with 10"6 M PHA665752 completely blocked 1K1 -induced tubulogenesis (Figures 3 A to 3B;
morphological images of HUVEC tube formation are not shown herein). Additionally, both PD98059 and LY294002 inhibited 1K1 -induced tubulogenesis implicating these signaling pathways in the process (Figures 3 A to 3B; morphological images of HUVEC tube formation are not shown herein). As shown in the Western blots (Figure 3C), incubation of endothelial cells with 1K1 resulted in rapid phosphorylation of both ERK and Akt, downstream effectors of the MAPK and PI3K signaling pathways, which were blocked by LY294002 and PD98059 respectively. Western blot also revealed that pretreating the cells with 10~6 M PHA665752 blocked the IKl -induced phosphorylation of ERK and Akt. These results confirmed that IKl induces angiogenesis in vitro by signaling through the same Met-dependent pathways activated by HGF/SF (10).
Example 3. Synthesis and Characterization of 1 Kl -Nanoparticle.
[00157] A key challenge for therapeutic angiogenesis is the local and sustained delivery of the angiogenic factors at the desired site of action (6). In a previous study, Richardson et al. demonstrated that sustained release of VEGF-165 and PDGF-BB, each with distinct kinetics, from a single, structural polymer scaffold resulted in the rapid formation of a mature vascular network (19). Furthermore, the controlled intramyocardial delivery of platelet-derived growth factor was reported to improve postinfarction ventricular function with increased
vascularization (20). Similarly, in a recent study, the intramuscular injection of pivastatin- loaded nanoparticles that enabled sustained release of the drug resulted in enhanced angiogenesis (21). However, no previous studies have demonstrated a sustained release formulation of IKl. Accordingly, nanoparticles were engineered from poly-lactic acid- glycolic acid (PLGA) copolymer, which is biocompatible and biodegradable and is approved by the FDA, in order to enable a sustained release formulation of IKl. The IKl nanoparticles were made using double emulsion/solvent extraction technique (22). Transmission electron microscopy data confirmed the spherical shape and also the diameter of the nanoparticles to be 60nm-140 nm (Figure 4A), which was validated by dynamic light scattering measurement (Figure 4B). The total loading efficiency of IKl in the nanoparticles was determined to be 54.27 ± 7.12%. Sustained release of therapeutic protein in biologically active form and subsequent degradation are critical for biologic activity, and depend on desorption of surface bound molecules, diffusion through the nanoparticle matrix and erosion of the latter. Kinetics of IKl release from the nanoparticles over a 7 day period was determined by enzyme-linked immunoabsorbent assay. As shown in Figure 4C, there was an initial phase of characteristic burst release associated with nanoparticles (22, 23) followed by a steady state release over a 7 day period. This finding is in agreement with previous reports where VEGF entrapped in PLGA microsphere exhibited similar release kinetics over a 30 day period (22).
Example 4. 1K1-NP Induces Angiogenesis in Vitro. [00158] The angiogenic activity of IKl nanoparticles (IKl-NP) was examined using HUVECs proliferation as the biological read-out. As shown in Figures 4D to 4F, treatment with IKl-NP resulted in significant endothelial cell proliferation as compared with vehicle, indicating that the process of engineering the nanoparticles does not alter the biological activity of IKl. Furthermore, IKl-NP-induced cell proliferation was inhibited in presence of MET inhibitor, PHA 665752 (Figure 4D), the PI3K inhibitor, LY294002, (Figure 4E), and the MAPK inhibitor, PD98059 (Figure 4F), indicating that, following entrapment in the nanoparticle, signaling is mediated by the same pathways activated by soluble IKl.
[00159] The engineered IKl-NP was discovered to induce significantly greater tubulogenesis as compared with empty vector or free IKl, indicating the sustained release of the angiogenic factor markedly affected the angiogenic response (Figures 5 A to 5C). To mechanistically correlate the phenotypic distinctions with the temporal release, Western blotting was used to evaluate the effect of IKl-NP on downstream signaling pathways. As shown in Figure 5B, incubation of endothelial cells with both IKl-NP and IKl resulted in rapid phosphorylation within 10 min. Pretreating the cells with PHA665752 blocked the phosphorylation of ERK and Akt induced by IKl-NP. Pretreating the cells with 50 μΜ LY29400 abolished the phosphorylation of Akt while PD98059 reduced the phosphorylation of ERK consistent with IKl activity previously described herein. However, in the case of IKl-NP, a sustained activity was noted as shown by the increased phosphorylation of ERK at 8.5 h post-treatment, unlike treatment with IKl (Figure 5C). It has been previously reported that sustained activation of MAPK signaling pathway is critical for tubulogenesis, whereas transient activation is insufficient to induce such morphological alterations (8, 24, 25).
Phenotypic quantification of the vascular tubes also confirmed that pretreatment of the HUVECs with PHA665752 (10~7 M), PD98059 (50 μΜ) or LY294002 (50 μΜ) blocked IKl-NP-induced tubulogenesis (Figure 5 A; morphological images of HUVEC tube formation are not shown herein).
Example 5. In Vivo Angiogensis and Implant Assays
[00160] Zebrafish Angiogenesis Assay. The zebrafish (Danio rerio) is fast emerging as an excellent model for studying neovascularization (26, 27). To validate the angiogenic activity of IKl and IKl-NP in vivo, IKl or IKl-NP were mixed with growth-factor-reduced matrigel (Mgel) and injected into the yolk sac, next to the subintestinal vessel. As shown in Figures 6A to 6F, IKl induced significant angiogenesis compared with empty vector as quantified by nodes formed during sprouting of the subintestinal vessels. At the 48 h time points, 1K1-NP exerted greater angiogenesis than 1K1, which validates the in vitro finding described earlier herein that sustained release of angiogenic factors can result in enhanced angiogenesis.
[00161] Murine Matrigel Implant Assay. To further validate the angiogenic efficacy of 1K1 in vivo, 1K1 or 1K1-NP mixed with growth-factor-reduced matrigel was injected subcutaneously in mice at a dose of 200 nanogram (ng) of lKl/scaffold. The scaffolds were maintained for 12 d, following which the animals were killed and the skin everted to visualize the angiogenic response. Treatment with 1K1 resulted in a strong angiogenic response as shown in Figures 7A to 7D. 1K1-NP induced a significantly greater angiogenic response as compared with 1K1 (Gross morphological images of the implants after everting the murine skin are not shown). To further validate the gross morphology, the implants were cross sectioned, immunolabeled for von Willebrand factor, a marker for endothelial cells. As shown in Figures 7 A to 7D, 1K1-NP treatment (Fig. 7D) resulted in a superior angiogenic outcome to free 1K1, consistent with the observation in the zebrafish. These findings confirm that sustained release of growth factors can exert superior angiogenic outcome.
Conclusions
[00162] While significant progress has been made towards inhibition of angiogenesis in a pathological context such as in cancer, therapeutic angiogenesis in diseases characterized by insufficient vascularization, such as in coronary ischemia, peripheral arterial diseases, or in diabetic sores (4, 28-30), is still at a nascent stage. The inventors and others have previously demonstrated that HGF/SF can mount a strong angiogenic response (12), both independently of other growth factors (10) and by inducing expression of VEGF (31). Furthermore, a study to assess the safety of intramuscular injection of HGF/SF plasmid to improve limb perfusion in patients with critical limb ischemia (the HGF-STAT trial) has recently revealed that intramuscular injection of HGF plasmid was safe and well tolerated, and limb tissue perfusion as measured by transcutaneous oxygen tension in patients with critical limb ischemia was found to have significantly improved at the highest doses used (32). These observations establish a strong clinical rationale to use HGF/SF in the management of ischemic diseases. However, while sustained release of angiogenic agents has been suggested to be critical in defining the clinical outcome for therapeutic angiogenesis, no sustained release model of HGF/SF has been developed. Presented herein is a rationally engineered variant of HGF/SF integrated into a nanovector that alters the temporal presentation of the growth factor to the target cells, and thereby enhances the angiogenic outcome by modifying the downstream signaling cascade.
[00163] A key hurdle in translating growth factors to the clinics frequently lies in the heterogeneity of the polypeptide growth factor due to extensive protein glycosylation, which poses regulatory challenges for manufacturing. In the case of HGF/SF, additional challenges are the complex multi-domain structure of the native protein that results in low production yields and a propensity of the protein to aggregate in physiological buffers. As demonstrated herein, a truncated mutant of HGF/SF, which can be expressed in high yields by yeast cells (unlike native HGF/SF), lacks glycosylation sequences and retains strong signaling activity, was investigated. The biological activity and therapeutic potential of 1K1 was further improved by incorporating the protein in nanoparticles from PLGA copolymer. While free 1K1 and 1K1-NP induced similar levels of ERK phosphorylation at early time points, the nanoparticle resulted in sustained activation of the MAPK signaling in contrast to free 1K1. In a previous study, the sustained activation of MAPK was reported to be critical for tubulogenesis while a transient activation was shown to be sufficient for the induction of cell proliferation (8, 24-25). This observation can possibly explain that while 1K1 and 1K1-NP exhibited similar levels of cell proliferation, the latter resulted in enhanced neovascularization in the in vivo matrigel implant and zebrafish angiogenesis assay. The finding presented herein also highlights the potential use of nanotechnology to temporally alter signaling pathways through controlled release thereby enable the dissection of distinct regulatory controls underlying biological phenomena. These results address the emerging paradigm that the sustained focal concentration of the active agent is critical for therapeutic angiogenesis, and can offer a robust strategy for the management of ischemic diseases.
Materials and Methods
[00164] Expression of 1K1 and HGF/SF. HGF/SF was produced from a derivative of the mouse myeloma cell line NS0 transfected with a full length HGF/SF cDNA. 1K1 was produced from a derivative of the yeast Pichia pastoris transfected with the 1K1 cDNA. Both proteins were purified from culture supernatants using Heparin-Sepharose chromatography followed by cation exchange chromatography on Mono S, yielding proteins >90% pure as judged by SDS gel electrophoresis.
[00165] Surface Plasmon Resonance Experiments. A fragment of the Met ectodomain corresponding to amino acids 25-567 (MET567) was used for immobilization on single flow cells of a CM5 chip equilibrated with 20 mM phosphate (pH 7.4), 150 mM NaCl, 50 μΜ EDTA, 0.005% Surfactant P20. The chip surface was first activated using the CM5 amine coupling immobilization method in the BIACore Control software (100 μΐ^ of 0.2 M 1-Ethyl- 3-(3 dimethylamino-propyl) carbodiimide mixed with 100 μΐ^ 0.05 M N-hydroxysuccinimide to give reactive succinimide ester groups). Met567H was diluted in 10 mM sodium acetate at a final concentration of 400 nM and injected, until a ARU of - 1,500 was reached. Remaining active groups were blocked by injection of 70 μΐ^ of 1 M ethanolamine (pH 8.5). As a negative control, another flow cell of the CM5 chip was treated with the same program, but without protein. The chip was then re-equilibrated with 10 mM Hepes, 150 mM NaCl, 3 mM EDTA, 0.005% Tween 20 (HBS-EP) (10 mM Hepes (pH 7.4); 150 mM NaCl; 50 μΜ EDTA; 0.005% Surfactant P20) + 0.2 mg/ml BSA and then a range of protein concentrations (diluted in HBS-EP + BSA) were injected at 20 μΐ/min for 60 min followed by a 300 s dissociation time. The chip was regenerated using HBS-EP with 1 M NaCl.
[00166] Analytical Ultracentrifugation. Wild-type NK1 , 1 Kl , and MET567 proteins were dialyzed against 25 mM phosphate, 150 mM NaCl, pH 7.4 prior to centrifugation.
Measurements were made using a Beckman Optima XLA analytical ultracentrifuge using an intermediate speed sedimentation velocity method. Data were acquired in continuous mode analyzed using the program Sedfit in which the least-squares g(s*) sedimentation coefficient distribution is computed, where c is the concentration (in absorbance units), r is radius (in cm), t is time (in seconds), cO is the loading concentration (in absorbance units), w is the angular velocity of the rotor (in radians/second) and rm is the radius at the meniscus (in cm). Data were collected with incident light set at 278 nm. The g(s*) profiles were fitted with normal (Gaussian) distributions using ProFit, a nonlinear least-squares fitting package (Quantum Soft).
[00167] Engineering 1K1 Nanoparticle. 1K1 nanoparticles were made using the double emulsion/solvent extraction technique (22). Emulsion 1 was prepared using 50 mg PLGA (50 : 50) (MW 40-75 KDa) dissolved in 10 mL of acetonitrile. -104 μg 1K1 was
encapsulated in 50 mg PLGA by sonication. Emulsion 2 was prepared using 0.5 g PVA (polyvinyl alcohol, Sigma, MW 9,000-10,000) and was dissolved in 9.3 mL of double distilled water and 0.7 mL of acetonitrile. An organic extraction buffer was made by dissolving 0.2 g PVA in 186 mL of double distilled water and 14 mL of acetonitrile. 1 mL of emulsion 2 was added to emulsion 1 and the solution was vortexed for - 1 min and was added to that organic buffer. This solution was stirred for 12 h to evaporate acetonitrile. This solution was centrifuged at 193,190 x 4 g (Sorvall Ultra Pro 80) for 1 h to pellet the nanoparticles, which was washed again with water and then characterized for size and morphology using a Nano-zetasizer (Malvern) and transmission electron microscopy (TEM). The samples was spotted on copper grids and stained with uranyl acetate for TEM.
[00168] Cell Proliferation Assay. HUVECs were grown in 96 well plate and
synchronized in 0.1% FBS prior to treatment with growth factors for 24, 48, or 72 h. Cells were treated with signal transduction inhibitors for 2 h prior to addition of growth factors. The percentage of viable cells was quantified with 3-(4, 5-Dimethylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2 (4-sulfo-phenyl)-2H-tetrazolium (MTS) from the Cell Titer 96 Aqueous One Solution kit measured at 490 nm using a Versamax plate reader. Final absorbance, corresponding to cell proliferation, was plotted after subtracting background values from each data point.
[00169] HUVEC Tube Assay. HUVECs (25,000 cells per well) were seeded in a 96-well plate coated with growth-factor-reduced matrigel. Cells were treated with inhibitors for 2 h before adding the growth factors. After 8 h, images were taken under inverted light microscopy at lOx magnification (Nikon Eclipse).
[00170] Immunoblotting. HUVECs (70% confluent) were treated with 1K1, 1K1-NP, and HGF/SF (positive control) for 10 min and directly lysed in 3x loading buffer. The cells were pretreated with signal transduction inhibitors for at least 2 h before growth-factor treatment. Proteins were resolved on 10% SDS-PAGE gel. Membranes were probed for phosphorylated and total forms of MET, Akt, and Erk. Proteins were detected with horseradish peroxidase- conjugated anti-rabbit secondary antibodies and Lumi-LightPLUS Western Blotting Substrate (Roche Applied Science). The blots were developed using GeneSnap and optical density was quantified using Gene Tools (both from SynGene). Predetermined molecular weights standards were used as markers. Proteins were normalized against actin.
[00171] Zebrafish Angiogenesis Assay. Zebrafish (Tubingen AB) (2 d post fertilization), were anesthetized in Tricaine (MESAB-ethyl-m-aminobenzoate methanesul-phonate, 1% Na2HP04 , pH 7.0) and positioned on their side in a 1% agarose injection mold, and 9.2 nL of each sample was injected into the yolk sac next to the subintestinal vessel (SIV) using a Nanoject II injection device (Drummond Scientific). Zebrafish injected with growth-factor- reduced matrigel and 1 μΜ 1K1 as free or in nanoparticles were monitored for 24 and 48 h. Brightfield imaging was performed with a Nikon SMZ1500 stereomicro-scope and a SPOT Flex camera.
[00172] In Vivo Matrigel Angiogenesis Assay in Mouse. Growth-factor-reduced matrigel (BD), mixed with growth factors were injected subcutaneously into balb/c mice. Mice were divided in four groups: (A) vehicle control with growth-factor-reduced matrigel, (B) vehicle control with empty PLGA and growth-factor-reduced matrigel, (C) matrigel plug with 200 ng free 1K1 and (D) matrigel plug with 200 ng 1K1-NP. On day 12, gross response in the form of blood vessels formed (angiogenesis) was recorded with a high-resolution digital camera (Canon). The implants were excised and cryofrozen in optimal cutting temperature compound for immunohistochemistry.
[00173] Immunohistochemistry. The matrigel cryosections (18 μπι) were fixed in -20 °C with ice cold methanol and probed overnight with primary rabbit antibody against von Willebrand factor (Dako) at 4 °C in blocking buffer. Samples were then probed with Alexa fluor 594 conjugated anti-rabbit secondary antibody at room temperature for 2 h. Nuclei were counterstained with DAPI. Images were captured with immunofluorescence microscope (Nikon Eclipse) at lOx magnification.
[00174] Statistical Analysis. Statistical significance was tested by using 1-way ANOVA followed by Dunnett's or Friedman's post hoc test. Bonferroni's test was used to test for overall dose-response effects (Graphical Prism 3 software). A value of P < 0.05 was considered significant.
[00175] Sequences
[00176] SEQ ID NO: 1 - Accession 3mkp_b, version 3mkp_b GL303325025, Homo sapiens 1K1 fragment with mutated amino acids bolded, amino acid no. 105 was changed from a lysine to a glutamic acid, and amino acid no.107 was changed from an arginine to a glutamic acid
1 yaegqrkrrn tihefkksak ttlikidpal kiktkkvnta dqcanrctrn kglpftckaf 61 vfdkarkqcl wfpfnsmssg vkkefghefd lyenkdyirn ciigegesyk gtvsitksgi 121 kcqpwssmip hehsflpssy rgkdlqenyc rnprgeeggp wcftsnpevr yevcdipqcs 181 eve (SEQ ID NO : 1 )
[00177] SEQ ID NO:2 -NK1 Fragment Homo sapiens
1 yaegqrkrrn tihefkksak ttlikidpal kiktkkvnta dqcanrctrn kglpftckaf 61 vfdkarkqcl wfpfnsmssg vkkefghefd lyenkdyirn ciigkgrsyk gtvsitksgi 121 kcqpwssmip hehsflpssy rgkdlqenyc rnprgeeggp wcftsnpevr yevcdipqcs 181 eve (SEQ ID NO : 2 )
[00178] SEQ ID NO: 3 Full Length Hepatocyte Growth Factor Scatter Factor Accession P14210, version P14210.2 GL123116 1 mwvtkllpal llqhvllhll llpiaipyae gqrkrrntih efkksakttl ikidpalkik 61 tkkvntadqc anrctrnkgl pftckafvfd karkqclwfp fnsmssgvkk efghefdlye 121 nkdyirncii gkgrsykgtv sitksgikcq pwssmipheh sflpssyrgk dlqenycrnp 181 rgeeggpwcf tsnpevryev cdipqcseve cmtcngesyr glmdhtesgk icqrwdhqtp 241 hrhkflpery pdkgfddnyc rnpdgqprpw cytldphtrw eycaiktcad ntmndtdvpl 301 etteciqgqg egyrgtvnti wngipcqrwd sqyphehdmt penfkckdlr enycrnpdgs 361 espwcfttdp nirvgycsqi pncdmshgqd cyrgngknym gnlsqtrsgl tcsmwdknme 421 dlhrhifwep dasklnenyc rnpdddahgp wcytgnplip wdycpisrce gdttptivnl 481 dhpviscakt kqlr vngip trtnigwmvs lryrnkhicg gslikeswvl tarqcfpsrd 541 lkdyeawlgi hdvhgrgdek ckqvlnvsql vygpegsdlv lmklarpavl ddfvstidlp 601 nygctipekt scsvygwgyt glinydgllr vahlyimgne kcsqhhrgkv tlneseicag 661 aekigsgpce gdyggplvce qhkmrmvlgv ivpgrgcaip nrpgifvrva yyakwihkii 721 ltykvpqs (SEQ ID NO: 3)
References
1. Rosamond W, et al. (2007) Heart disease and stroke statistics— 2007 update: a report from the American Heart Association Statistics Committee and Stroke Statistics
Subcommittee. Circulation 115:e69-el71.
2. Ferrara N, Kerbel RS (2005) Angiogenesis as a therapeutic target. Nature 438:967-974.
3. Kutryk MJB, et al. (2001) Angiogenesis: an emerging technology for the treatment of coronary artery disease. Cardiology Rounds VI: 1-8.
4. Fortuin FD, et al. (2003) One-year follow-up of direct myocardial gene transfer of vascular endothelial growth factor-2 using naked plasmid deoxyribonucleic acid by way of thoracotomy in no-option patients. Am J Cardiol 92:436-439.
5. Simons M, et al. (2002) Pharmacological treatment of coronary artery disease with
recombinant fibroblast growth factor-2. double-blind, randomized, controlled clinical trial. Circulation 105:793.
6. Yla-Herttuala S, et al. (2007) Vascular endothelial growth factors: biology and current status of clinical applications in cardiovascular medicine. J Am Coll Cardiol 49:1015- 1026.
7. Gupta R, et al. (2009) Human studies of angiogenic gene therapy. Cir Res 105:724-736.
8. Zhou X, et al. (2008) Fibronectin fibrillogenesis regulates three dimensional neovessel formation. Gene Dev 22:1231-1243. an S (2006) Glycosylation engineering. Chem Eng News 84:13-22.
gupta S, et al. (2003) Hepatocyte growth factor/scatter factor can induce angiogenesis independently of vascular endothelial growth factor. Arterioscler Thromb Vase Biol 23:69-75.
erardi E, et al. (2006) Structural basis of hepatocyte growth factor/scatter factor and
MET signaling. Proc Nat'l Assoc Sci USA 103:4046-4051.
gupta S, et al. (2003) Targeting of mitogen- activated protein kinases and
phosphatidylinositol 3 kinase inhibits hepatocyte growth factor/scatter factor-induced angiogenesis. Circulation 107:2955-2961.
o K, et al. (1997) Enhanced expression of hepatocyte growth factor/c-Met by myocardial ischemia and reperfusion in a rat model. Circulation 95:2552-2558.
wall RH, et al. (1996) Heparin induces dimerization and confers proliferative activity onto the hepatocyte growth factor antagonists NKl and NK2. JCB 133:709-718.
irgadze DY, et al. (1999) Crystal structure of the NKl fragment of HGF/SF suggests a novel mode for growth factor dimerization and receptor binding. Nat Struct Biol 6:72- 79.
tha D, et al. (2001) Crystal structures of NKl-heparin complexes reveal the basis for
NKl activity and enable engineering of potent agonists of the MET receptor. EMBO J 20:5543-5555.
lmes O, et al. (2007) Insights into the structure/function of hepatocyte growth factor/scatter factor from studies with individual domains. J Mol Biol 367:395-408. gupta S, et al. (2004) Nitric oxide modulates hepatocyte growth factor/scatter factor- induced angiogenesis. Angiogenesis 7:285-294.
hardson TP, et al. (2001) Polymeric system for dual growth factor delivery. Nat
Biotechnol 19:1029-1034.
ieh PCH, et al. (2006) Local controlled intramycardial delivery of platelet-derived growth factor improves postinfarction ventricular function without pulmonary toxicity. Circulation 114:637-644.
bo M, et al. (2009) Therapeutic neovascularization by nanotechnology-mediated cell- selective delivery of pitavastatin into the vascular endothelium. Arterioscler Thromb Vase Biol 29:796-801.
cha FG, et al. (2008) The effect of sustained delivery of vascular endothelial growth factor on angiogenesis in tissue-engineered intestine. Biomaterials 29:2884-2890. 23. Chappell JC, et al. (2008) Targeted delivery of nanoparticles bearing fibroblast growth factor-2 by ultrasonic microbubble destruction for therapeutic arteriogenesis. Small 4:1769-1777.
24. Karihaloo A, et al. (2004) Hepatocyte growth factor-mediated renal epithelial branching morphogenesis is regulated by glypican-4 expression. Mol Cell Biol 24:8745-8752.
25. Rosario M, Birchmeier W (2003) How to make tubes: signaling by the Met receptor tyrosine kinase. Trends Cell Biol 13:328-335.
26. Stoletov K, et al. (2007) High-resolution imaging of the dynamic tumor cell-vascular interface in transparent zebrafish. Proc Natl Acad Sci USA 104:17406-17411.
27. Ny A, et al. (2006) Zebrafish and Xenops tadpoles: small animal models to study
angiogenesis and lymphangio genesis. Exp Cell Res 312:684-693.
28. Wang N, et al. (2009) Effect of hepatocyte growth-promoting factors on myocardial ischemia during exercise in patients with severe coronary artery disease. Int Heart J 50:291-299.
29. Laham RJ, et al. (2001) Gene transfer for angiogenesis in coronary artery disease. Annual
Rev Med 52:485-502.
30. Jin H, et al. (2003) Early treatment with hepatocyte growth factor improves cardiac
function in experimental heart failure induced by myocardial infarction. The Journal of Pharmacology and Experimental Therapeutics 304:654-660.
31. Van Belle E, et al. (1998) Potentiated angiogenic effect of scatter factor/hepatocyte
growth factor via induction of vascular endothelial growth factor: the case for paracrine amplification of angiogenesis. Circulation 97:381-390.
32. Powell RJ, et al. (2008) Results of a double-blind, placebo-controlled study to assess the safety of intramuscular injection of hepatocyte growth factor plasmid to improve limb perfusion in patients with critical limb ischemia. Circulation 118:58-65.
[00179] All patents and other publications identified in the specification are expressly incorporated herein by reference in their entirety for all purposes. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.

Claims

WHAT IS CLAIMED:
1. A pharmaceutical composition comprising a nanoparticle comprising a biocompatible polymer and an effective amount of 1K1 protein fragment of Hepatocyte Growth Factor/Scatter Factor, wherein the biodegradable polymer encapsulates the 1K1 protein.
2. The pharmaceutical composition of claim 1, wherein the 1K1 protein comprises SEQ ID. NO:l.
3. The pharmaceutical composition of any of claims 1-2, wherein the nanoparticle has an average particle size of about 50 nm to about 500 nm.
4. The pharmaceutical composition of any of claims 1-2, wherein nanoparticle has an average particle size of about 60 nm to about 150 nm.
5. The pharmaceutical composition of any of claims 1-4, wherein the biocompatible polymer is a biodegradable polymer and the biodegradable polymer is selected from the group consisting of: polyesters, hydroxyaliphatic carboxylic acids, poly(lactic acid), poly(glycolic acid), poly(dl-lactide/glycolide, poly(ethylene glycol),
polysaccharides, lectins, glycosaminoglycans, chitosan, celluloses and acrylate polymers.
6. The pharmaceutical composition of any of claims 1-5, wherein the biocompatible polymer is a polyester.
7. The pharmaceutical composition of claim 6, wherein the polyester comprises poly- lactic acid-glycolic acid copolymer (PLGA).
8. The pharmaceutical composition of any of claims 1-7, wherein the 1K1 protein is released from the nanoparticle in a therapeutically effective amount over a defined period of time of days, weeks or months.
9. The pharmaceutical composition of any of claims 1-8, wherein the 1K1 protein is released from the nanoparticle in a therapeutically effective amount over a defined period of time of about 2 days, about 3 days, about 4 days, about 5 days, about 7 days, or about 14 days.
10. The pharmaceutical composition of any of claims 8-9, wherein the therapeutically effective amount that is released over the defined period of time is sufficient to result in sustained phoshorylation of cellular ERK over the defined period of time of release.
11. The pharmaceutical composition of any of claims 8-10, wherein the therapeutically effective amount is sufficient to increase angiogenesis in a subject as compared to angiogenesis in the absence of the compound by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, or about 60%.
12. The pharmaceutical composition of any of claims 8-11, wherein the therapeutically effective amount is dose of about O.lmg/kg to about 1000 mg/kg.
13. The pharmaceutical composition of any of claims 1-12, wherein the composition is formulated for administration by a method selected from the group consisting of: topical administration, enteral administration, and parenteral administration.
14. The pharmaceutical composition of claim 13, wherein the composition formulated for topical administration is an ointment, lotion, spray, cream, or gel.
15. A method for treatment of a condition associated with insufficient vascularization comprising administration to a subject a therapeutically effective amount of a pharmaceutical composition of any of claims 1-14.
16. The method of 15, wherein the therapeutically effective amount is a dose of about O.lmg/kg to about 1000 mg/kg of lkl protein.
17. The method of any of claims 15-16, wherein the condition associated with insufficient vascularization is selected from the group consisting of: cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, ischemia of tissues, coronary ischemia, peripheral arterial disease, limb ischemia, diabetic ulcers, gangrenes, wounds requiring neovascularization to facilitate healing, and Buerger's syndrome.
18. The method of any of claims 15-17, wherein the condition is a diabetic foot ulcer.
19. The method of any of claims 15-18, wherein the pharmaceutical composition is
administered by topical administration, enteral administration, or parenteral administration.
20. The method of any of claims 15-19, wherein the pharmaceutical composition is
administered multiple times.
21. Use of the pharmaceutical composition of any of claims 1-14 for treatment of a
condition associated with insufficient vascularization.
22. The use of claim 21, wherein the condition associated with insufficient
vascularization is selected from the group consisting of: cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, ischemia of tissues, coronary ischemia, peripheral arterial disease, limb ischemia, diabetic ulcers, gangrenes, wounds requiring neovascularization to facilitate healing, and Buerger's syndrome.
23. The use of claim 21, wherein the condition is a diabetic foot ulcer.
PCT/US2011/025374 2010-02-22 2011-02-18 Compositions and methods for inducing angiogenesis WO2011103382A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN2011800203513A CN103037892A (en) 2010-02-22 2011-02-18 Compositions and methods for inducing angiogenesis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30676810P 2010-02-22 2010-02-22
US61/306,768 2010-02-22
US37896810P 2010-09-01 2010-09-01
US61/378,968 2010-09-01

Publications (2)

Publication Number Publication Date
WO2011103382A2 true WO2011103382A2 (en) 2011-08-25
WO2011103382A3 WO2011103382A3 (en) 2012-04-05

Family

ID=44483581

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/025374 WO2011103382A2 (en) 2010-02-22 2011-02-18 Compositions and methods for inducing angiogenesis

Country Status (2)

Country Link
CN (1) CN103037892A (en)
WO (1) WO2011103382A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013037521A1 (en) * 2011-09-16 2013-03-21 Medical Research Council Modified hgf-1k1 polypeptide
WO2016116577A1 (en) * 2015-01-21 2016-07-28 Universite Des Sciences Et Technologies De Lille-Lille 1 Met receptor agonist proteins
WO2022119563A1 (en) * 2020-12-02 2022-06-09 Shih Chun Che Composition and method for treatment of ischemic disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE049294T2 (en) * 2013-05-15 2020-09-28 Univ Leland Stanford Junior Modulation of heparin-binding epidermal growth factor activity for tympanic membrane healing

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6716450B1 (en) * 2000-05-18 2004-04-06 The United States Of America As Represented By The Secretary Of The Army Enhancing protein activity through nanoencapsulation
US20040234481A1 (en) * 2001-05-09 2004-11-25 Ryuichi Morishita Gene transfers of angiogenic factor for skin disease
US20080167224A1 (en) * 2001-04-27 2008-07-10 Medical Research Council Variants of the nk1 fragment of hepatocyte growth factor/scatter factor (hgf/sf) and their use
US20080213370A1 (en) * 1993-02-22 2008-09-04 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20090157046A1 (en) * 2007-11-09 2009-06-18 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium using cytokines and variants thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101152151A (en) * 2007-08-31 2008-04-02 深圳市第二人民医院 Vascellum esoderma growth factor sustained-release corpuscle and method for preparing the same

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080213370A1 (en) * 1993-02-22 2008-09-04 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US6716450B1 (en) * 2000-05-18 2004-04-06 The United States Of America As Represented By The Secretary Of The Army Enhancing protein activity through nanoencapsulation
US20080167224A1 (en) * 2001-04-27 2008-07-10 Medical Research Council Variants of the nk1 fragment of hepatocyte growth factor/scatter factor (hgf/sf) and their use
US20040234481A1 (en) * 2001-05-09 2004-11-25 Ryuichi Morishita Gene transfers of angiogenic factor for skin disease
US20090157046A1 (en) * 2007-11-09 2009-06-18 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium using cytokines and variants thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013037521A1 (en) * 2011-09-16 2013-03-21 Medical Research Council Modified hgf-1k1 polypeptide
WO2016116577A1 (en) * 2015-01-21 2016-07-28 Universite Des Sciences Et Technologies De Lille-Lille 1 Met receptor agonist proteins
US10435450B2 (en) 2015-01-21 2019-10-08 Centre National De La Recherche Scientifique MET receptor agonist proteins
WO2022119563A1 (en) * 2020-12-02 2022-06-09 Shih Chun Che Composition and method for treatment of ischemic disease

Also Published As

Publication number Publication date
WO2011103382A3 (en) 2012-04-05
CN103037892A (en) 2013-04-10

Similar Documents

Publication Publication Date Title
Yang et al. Inflammation-targeting polymeric nanoparticles deliver sparfloxacin and tacrolimus for combating acute lung sepsis
WO2019141266A1 (en) Hydrogel nanocarrier delivery system for targeting active cd44 molecule, preparation method therefor, and uses thereof
Garbern et al. Delivery of basic fibroblast growth factor with a pH-responsive, injectable hydrogel to improve angiogenesis in infarcted myocardium
JP6912377B2 (en) Methods and Compositions Related to the Use of Low HLB Surfactants in the Production of Synthetic NanoCarriers Containing Lapalog
Rui et al. Controlled release of vascular endothelial growth factor using poly-lactic-co-glycolic acid microspheres: in vitro characterization and application in polycaprolactone fumarate nerve conduits
CA2415414C (en) Particulate vectors for improving oral absorption of active principles
Jiang et al. Controlled release of anti-VEGF by redox-responsive polydopamine nanoparticles
Liu et al. Anti-angiogenic activity of bevacizumab-bearing dexamethasone-loaded PLGA nanoparticles for potential intravitreal applications
EP3368553B1 (en) Polymer-based therapeutics for inductive browning of fat
CN107661490A (en) Drug storage system comprising glatiramer or its pharmaceutical salts
US20120058155A1 (en) Slow-degrading polymers for undelayed and sustained drug delivery
Pascual-Gil et al. Tracking the in vivo release of bioactive NRG from PLGA and PEG–PLGA microparticles in infarcted hearts
US20190225670A1 (en) Compounds and methods for activating tie2 signaling
TW200932223A (en) Drug-containing nanoparticles
Gai et al. Delivery of benzoylaconitine using biodegradable nanoparticles to suppress inflammation via regulating NF-κB signaling
Kanemaru et al. Nanoparticle-mediated local delivery of pioglitazone attenuates bleomycin-induced skin fibrosis
TWI604854B (en) Multi-drug delivery system and use thereof
WO2011103382A2 (en) Compositions and methods for inducing angiogenesis
Qiu et al. Myocardial fibrosis reversion via rhACE2-electrospun fibrous patch for ventricular remodeling prevention
US11660345B2 (en) Method and composition for enhancing the delivery of anti-platelet drugs for the treatment of acute stroke
Lian et al. Therapeutic angiogenesis of PLGA-heparin nanoparticle in mouse ischemic limb
KR20150054543A (en) Liver-targeted nanoparticles, a preparation method thereof, and a pharmaceutical composition comprising the nanoparticles
CN109496152A (en) The method of the intramuscular inventory and its prevention and treatment of deccox composition
Qutachi et al. Spatiotemporal release of VEGF from biodegradable polylactic-co-glycolic acid microspheres induces angiogenesis in chick chorionic allantoic membrane assay
US20230110354A1 (en) Therapeutic agent delivery systems and methods of forming and uses thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180020351.3

Country of ref document: CN

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

Ref document number: 11745291

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 7712/DELNP/2012

Country of ref document: IN

122 Ep: pct application non-entry in european phase

Ref document number: 11745291

Country of ref document: EP

Kind code of ref document: A2