WO2014130648A1 - Method for treatment of pulmonary fibrosis - Google Patents

Method for treatment of pulmonary fibrosis Download PDF

Info

Publication number
WO2014130648A1
WO2014130648A1 PCT/US2014/017353 US2014017353W WO2014130648A1 WO 2014130648 A1 WO2014130648 A1 WO 2014130648A1 US 2014017353 W US2014017353 W US 2014017353W WO 2014130648 A1 WO2014130648 A1 WO 2014130648A1
Authority
WO
WIPO (PCT)
Prior art keywords
residues
linked
rhamnogalacturonate
reduction
lung
Prior art date
Application number
PCT/US2014/017353
Other languages
French (fr)
Inventor
Peter G. TRABER
Original Assignee
Galectin Therapeutics, Inc.
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 Galectin Therapeutics, Inc. filed Critical Galectin Therapeutics, Inc.
Publication of WO2014130648A1 publication Critical patent/WO2014130648A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/736Glucomannans or galactomannans, e.g. locust bean gum, guar gum

Abstract

Methods and compositions of the invention relate to the treatment of pulmonary fibrosis.

Description

METHOD FOR TREATMENT OF PULMONARY FIBROSIS
Inventor Peter G. Traber CROSS-REFERENCE TO RELATED APPLICATIONS
[001] This application claims the benefit of and priority to U.S. provisional
Application Serial Number 61/767,057, filed February 20, 2013, the entire disclosure of which is incorporated by reference in its entirety.
FIELD OF THE INVENTION
[002] Methods and compositions of the invention relate compositions and methods for treatment of pulmonary fibrosis.
BACKGROUND OF THE INVENTION
[003] Puimonary fibrosis is the formation or development of excess fibrous connective tissue (fibrosis) In the iungs. Approximately 5 million people worldwide are affected by pulmonary fibrosis. Pulmonary fibrosis Is a fatal disease In which the uncontrolled deposition of extracellular matrix leads to progressive loss of lung function. i RY OF,lHEJ ENT80,N
[004] Aspects of the invention relate to novel approaches to treat pulmonary fibrosis.
[005] Aspects of the invention relate to compositions, methods of using and methods of manufacturing compositions capable of treating pulmonary fibrosis.
[006] Other aspects of the invention relate to methods of treating a subject in need thereof. In some embodiments, the method comprises the step of obtaining a composition for parenteral administration, the composition comprising a compound in an acceptable pharmaceutical carrier and administering the composition to a subject in need thereof. [007] In some embodiments, the compound can be one of galacto- rhamnogalacturonate (GRG), galactoarabino-rhamnogalacturonate (GA-RG), galactomannan (GM), or a combination of any of the foregoing.
[008] In some embodiments, the compound can be a polysaccharide chemically defined as galacto-rhamnogalacturaonate (GRG), a selectively depolymerized branched heteropolymer whose backbone is predominantly comprised of 1 ,4-linked galacturonic acid (GalA) moieties, with a lesser backbone composition of alternating 1 ,4-linked GalA and 1 ,2-linked rhamnose (Rha), which in-turn is linked to any number of side chains, including predominantly 1 ,4-b-D-galactose (Gal) and 1 ,5-a-L-arabinose (Ara) residues. Other side chain minor constituents may include xylose (Xyl), glucose (Glu), and fucose (Fuc) or combinations thereof.
[009] In some embodiments, the GRG compound can be produced as described in U.S. Patent Application Publication No. 2008/0107622, now U.S. Patent 8,236,780, which is incorporated expressly by reference for all purposes.
[0010] In some embodiments, the GRG compound can be produced as described in U.S. Patents 8,128,966, 8,187,624, U.S. Patent Application Publication Nos 2012/0315309 and 2012/030971 1 which are incorporated expressly by reference for all purposes.
[0011] In some embodiments, the compound can be a polysaccharide chemically defined as galactoarabino-rhamnogalacturonate (GA-RG), a selectively depolymerized, branched heteropolymer whose backbone is predominantly comprised of 1 ,4-linked galacturonic acid (GalA) moieties, with a lesser backbone composition of alternating 1 ,4- linked GalA and 1 ,2-linked rhamnose (Rha), which in-turn is linked to any number of side chains, including predominantly 1 ,4-b-D-galactose (Gal) and 1 ,5-a-L-arabinose (Ara) residues. Other side chain minor constituents may include xylose (Xyl), glucose (Glu), and fucose (Fuc) or combinations thereof.
[0012] In some embodiments, the GA-RG compound can be produced as described in International Patent Application PCT/US12/5531 1 , and U.S. Patent Application Number US 2013-0261078, which is incorporated expressly by reference for all purposes. In some embodiments, the compound can be a galactomannan (GM) polysaccharide composition produced as described in U.S. Patent 8,236,780 and U.S. Patent Application Publication No. US 201 1/0077217 which are incorporated expressly by reference in their entireties for all purposes.
[0013] Some aspects of the invention relate to a method comprising obtaining a composition for parenteral or enteral administration comprising a galacto- rhamnogalacturonate in a pharmaceutical acceptable carrier, and administering to a subject in need thereof an effective dose of the composition that results in at least one of the following: at least 5% reduction of lung edema, at least 5% reduction of lung pathology severity scores associated with lung fibrosis, at least 5% reduction of lung tissue hydroxyproline accumulation, at least 5% reduction of expression of proinflammatory proteins, at least 5% reduction of expression of fibrogenic proteins. In some embodiments, the administration of the effective dose results in at least 5% improvement of pulmonary function tests including forced vital capacities or oxygen diffusion. In some embodiments, the subject in need has at least one of the following: a primary or secondary lung fibrotic disease.
[0014] In some embodiments, the effective dose of galacto-rhamnogalacturonate can be equivalent to an animal dose of about 120 mg/kg. In some embodiments, the effective dose of galacto-rhamnogalacturonate can be equivalent to an animal dose of 10 mg/kg to 180 mg/kg. In some embodiments, the effective dose can be given once, twice or three times weekly.
[0015] In some embodiments, the administration of the effective dose results in at least 5% reduction of expression of pro-inflammatory proteins. In some embodiments, the pro-inflammatory proteins can comprise TGF-beta, IL-6, IL-8, IL-13, IP-10, osteopontin, TNF-alpha, CXCL-9/10, VEGF or any combination of the foregoing.
[0016] In some embodiments, the administration of the effective dose results in at least 5% reduction of expression of fibrogenic proteins. In some embodiments, the fibrogenic proteins can comprise collagen, elastin or a combination thereof.
[0017] In some embodiments, the galacto-rhamnogalacturonate can comprise a
1 ,4-linked galacturonic acid (GalA) and methyl galacturonate (MeGalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D-galactose residues.
[0018] In some embodiments, the galacto-rhamnogalacturonate can comprise a 1 ,4-linked galacturonic acid (GalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D-galactose residues.
[0019] In some embodiments, the galacto-rhamnogalacturonate can further comprise xylose, glucose, fucose residues or combination thereof.
[0020] In some embodiments, the galacto-rhamnogalacturonate can be substantially free of 1 ,5-a-L-Ara residues.
[0021] In some embodiments, the galacto-rhamnogalacturonate is a galactoarabino-rhamnogalacturonate, comprising a 1 ,4-linked galacturonic acid (GalA) and methyl galacturonate (MeGalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D- galactose residues, 1 ,5- a -L-arabinose residues, or combinations thereof.
[0022] In some embodiments, the 1 ,4- β -D-galactose and 1 ,5- a -L-arabinose residues are present in a 2:1 or a 3:1 ratio.
[0023] In some embodiments, the 1 ,4- β -D-galactose residues, the 1 ,5- a -L- arabinose residues or combination thereof can represent at least 10 molar percent of the total molar carbohydrates.
[0024] In some embodiments, the galacto-rhamnogalacturonate can have an average molecular weight ranging from 5 kDa to 55 kDa as determined by SEC-RI and/or the SEC-MALLS methods. [0025] In some embodiments, the galacto-rhamnogalacturonate can have an average molecular weight ranging from 2 kDa to 80 kDa as determined by SEC-RI and/or the SEC-MALLS methods.
[0026] In some embodiments, the galacto-rhamnogalacturonate can an average molecular weight ranging from 20 kDa to 70 kDa as determined by SEC-RI and/or the SEC-MALLS methods.
[0027] In some aspects, the method comprises obtaining a composition for parenteral or enteral administration comprising a galactomannan polysaccharide in a pharmaceutical acceptable carrier, and administering to a subject in need thereof an effective dose of the composition that results in at least one of the following: at least 5% reduction of lung oedema, at least 5% reduction of lung pathology associated with lung fibrosis, at least at least 5% reduction of hydroxyproline accumulation, at least 5% reduction of expression of pro-inflammatory proteins, at least 5% reduction of expression of fibrogenic proteins, wherein the subject in need thereof has at least one of the following: a primary or secondary lung fibrotic disease. In some embodiments, the administration of the effective dose results in at least 5% improvement of pulmonary function tests including forced vital capacities or oxygen diffusion.
[0028] In some embodiments, the effective dose of the galactomannan polysaccharide is equivalent to an animal dose of about 60 mg/kg. In some embodiments, the effective dose of the galactomannan polysaccharide is equivalent to an animal dose of 30 mg/kg to 180 mg/kg. In some embodiments, the effective dose can be given once, twice or three times weekly.
[0029] In some embodiments, in the step of obtaining, the galactomannan polysaccharide has a mannose to galactose ratio ranging from 1 :1 to 1 :4.
[0030] In some embodiments, in the step of obtaining, the galactomannan polysaccharide has a mannose to galactose ratio of 1 .7:1 .
[0031] In some embodiments, in the step of obtaining, the galactomannan polysaccharide has a molecular weight of about 48,000 D. [0032] In some embodiments, in the step of obtaining, the galactomannan polysaccharide has average molecular weight ranging from 4,000 D to 60,000D.
[0033] In some embodiments, the administration of the effective dose results in at least 5% reduction of expression of pro-inflammatory proteins. In some embodiments, the pro-inflammatory proteins can comprise TGF-beta, IL-6, IL-8, IL-13, IP-10, osteopontin, TNF-alpha, CXCL-9/10, VEGF or any combination of the foregoing.
[0034] In some embodiments, the administration of the effective dose results in at least 5% reduction of expression of fibrogenic proteins. In some embodiments, the fibrogenic proteins can comprise collagen, elastin or a combination thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
[0035] The present invention will be further explained with reference to the attached drawings, wherein like structures are referred to by like numerals throughout the several views. The drawings shown are not necessarily to scale, with emphasis instead generally being placed upon illustrating the principles of the present invention.
[0036] FIGURE 1A and 1 B depicts the change in animal weight in the mouse model of bleomycin-induced pulmonary fibrosis with early treatment with GR-MD-02 and GM-CT-01 . FIGURE 1A shows the daily weight change of mice expressed as mean and standard error of the mean. FIGURE 1 B shows the percent change in weight for each treatment group as calculated using the area under the curve (AUC).
[0037] FIGURE 2 is a graph showing the mean lung wet weight on day 21 in treated and control groups, according to one embodiment of the invention.
[0038] FIGURE 3 is a graph showing the mean total lung hydroxyproline content (expressed as a percent of saline treated mice) in mice on day 21 in treated and control groups, according to one embodiment of the invention.
[0039] FIGURE 4 is a graph showing the modified Ashcroft lung fibrosis index scores in treated and control groups, according to one embodiment of the invention.
[0040] FIGURE 5 is a graph showing the general pulmonary fibrosis scores on treated and control groups, according to one embodiment of the invention. [0041] FIGURE 6 is a graph showing the inflammation scores on treated and control groups, according to one embodiment of the invention.
[0042] FIGURE 7 are representative photomicrographs with trichrome from treated and control groups, according to one embodiment of the invention.
[0043] FIGURE 8 are representative photomicrographs with H & E from treated and control groups, according to one embodiment of the invention.
[0044] FIGURE 9 is a graph showing the weights of the lung of the control groups, vehicle treated group, and treated groups.
[0045] FIGURE 10 is a graph showing the lung hydroxyproline (expressed as a percent of saline treated mice) of the control groups, vehicle treated group, and treated groups.
[0046] FIGURE 1 1 is a graph showing the Aschroft fibrosis index score from a histology assessment of the control groups, vehicle treated group, and treated groups.
[0047] FIGURE 12 is a graph showing the general fibrosis score from a histology assessment of the control groups, vehicle treated group, and treated groups.
[0048] FIGURE 13 is a graph showing the inflammation score from a histology assessment of the control groups, vehicle treated group, and treated groups.
[0049] FIGURE 14 are representative photomicrographs with trichrome from treated and control groups, according to one embodiment of the invention.
[0050] FIGURE 15 are representative photomicrographs with H & E from treated and control groups, according to one embodiment of the invention.
DETAILED DESCRIPTION OF THE INVENTION
[0051] Detailed embodiments of the present invention are disclosed herein; however, it is to be understood that the disclosed embodiments are merely illustrative of the invention that may be embodied in various forms. In addition, each of the examples given in connection with the various embodiments of the invention is intended to be illustrative, and not restrictive. Further, the figures are not necessarily to scale, some features may be exaggerated to show details of particular components. In addition, any measurements, specifications and the like shown in the figures are intended to be illustrative, and not restrictive. Therefore, specific structural and functional details disclosed herein are not to be interpreted as limiting, but merely as a representative basis for teaching one skilled in the art to variously employ the present invention.
[0052] Citation of documents herein is not intended as an admission that any of the documents cited herein is pertinent prior art, or an admission that the cited documents are considered material to the patentability of the claims of the present application.
[0053] Unless otherwise specified, all percentages expressed herein are weight/weight.
[0054] Aspects of the invention relate to novel approaches to treat pulmonary fibrosis using a complex carbohydrate pharmaceutical product or other agents described herein. In some embodiments, the methods disclosed herein relate to the treatment of a subject having a primary or secondary lung fibrotic disease.
Galectins
[0055] Galectins (also known as galaptins or S-lectin) are a family of lectins which bind beta-galactoside. Galectin as general name was proposed in 1994 for a family of animal lectins (Barondes, S.H., et al.: Galectins: a family of animal beta-galactoside- binding lectins. Cell 76, 597-598, 1994), The family is defined by having at least one characteristic carbohydrate recognition domain (CRD) with an affinity for beta- galactosides and sharing certain sequence elements. Within the same peptide chain, some galectins have a CRD with only a few additional amino acids, whereas others have two CRDs joined by a link peptide, and one (galectin-3) has one CRD joined to a different type of domain. The galectin carbohydrate recognition domain (CRD) is a beta-sandwich of about 135 amino acids. The two sheets are slightly bent with 6 strands forming the concave side and 5 strands forming the convex side. The concave side forms a groove in which carbohydrate is bound (Leffler H, Carlsson S, Hedlund M, Qian Y, Poirier F (2004). "Introduction to galectins". Glycoconj. J. 19 (7-9): 433-40).
[0056] A wide variety of biological phenomena have been shown to be related to galectins, e.g., development, differentiation, morphogenesis, tumor metastasis, apoptosis, RNA splicing, etc. However, relatively little is known about the mechanism by which galectins exert these functions, particularly in terms of carbohydrate recognition. [0057] Generally, the carbohydrate domain binds to galactose residues associated with glycoproteins. At least fifteen mammalian galectin proteins have been identified which have one or two carbohydrate domain in tandem.
[0058] Galectin proteins are found in the intracellular space where they have been assigned a number of functions and are secreted into the extracellular space. In the extracellular space, galectin proteins can have multiple functions including promoting interactions between glycoproteins that may lead to reduced function, or enhanced functions, or in the case of integral membrane glycoprotein receptors, modification of cellular signaling (Sato et al "Galectins as danger signals in host-pathogen and host- tumor interactions: new members of the growing group of "Alarmins." In "Galectins," (Klyosov, et al eds.), John Wiley and Sons, 1 15-145, 2008, Liu et al "Galectins in acute and chronic inflammation," Ann. N. Y. Acad. Sci. 1253: 80-91 , 2012). Galectin proteins in the extracellular space can additionally promote cell-cell and cell matrix interactions (Wang et al., "Nuclear and cytoplasmic localization of galectin-1 and galectin-3 and their roles in pre-mRNA splicing." In "Galectins" (Klyosov et al eds.), John Wiley and Sons, 87- 95, 2008).
[0059] Galectins have been shown to have domains which promote homodimerization. Thus, galectins are capable of acting as a "molecular glue" of sorts between glycoproteins. Galectins are found in multiple cellular compartments, including the nucleus and cytoplasm, and are secreted into the extracellular space where they interact with cell surface and extracellular matrix glycoproteins. The mechanism of molecular interactions can depend on the localization. While galectins can interact with glycoproteins in the extracellular space, the interactions of galectin with other proteins in the intracellular space generally occurs via protein domains. In the extracellular space the association of cell surface receptors may increase or decrease receptor signaling or the ability to interact with ligands. Galectin proteins are markedly increased in a number of animal and human disease states, including but not limited to diseases associated with inflammation, fibrosis, autoimmunity, and neoplasia. Galectins have been directly implicated in the disease pathogenesis, as described below. For example, diseases states that may be dependent on galectins include, but are not limited to, acute and chronic inflammation, allergic disorders, asthma, dermatitis, autoimmune disease, inflammatory and degenerative arthritis, immune-mediated neurological disease, fibrosis of multiple organs (including but not limited to liver, lung, kidney, pancreas, and heart), inflammatory bowel disease, atherosclerosis, heart failure, ocular inflammatory disease, a large variety of cancers.
[0060] In addition to disease states, galectins are important regulatory molecules in modulating the response of immune cells to vaccination, exogenous pathogens and cancer cells.
[0061] One of skill in the art will appreciate that compounds that can bind to galectins and/or alter galectin's affinity for glycoproteins, reduce hetero- or homo-typic interactions between galectins, or otherwise alter the function, synthesis, or metabolism of galectin proteins may have important therapeutic effects in galectin-dependent diseases.
[0062] Galectins show an affinity for galactose residues attached to other organic compounds, such as in lactose [( -D-Galactosido)-D-glucose], N-acetyl-lactosamine, poly-N-acetyllactosamine, galactomannans, fragments of pectins, as well as other galactose containing compounds. It should be noted that galactose by itself does not bind to galectins, or binds so weakly that the binding can hardly be detected.
[0063] Pectin and modified pectin have been shown to bind to galectin proteins presumably on the basis of containing galactose residues that are presented in the context of a macromolecule, in this case a complex carbohydrate rather than a glycoprotein in the case of animal cells.
[0064] Galectin proteins have been shown to be markedly increased in inflammation, fibrotic disorders, and neoplasia (Ito et al. "Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment", Cancer Metastasis Rev. PMID: 22706847 (2012), Nangia-Makker et al. Galectin-3 binding and metastasis," Methods Mol Biol. 878: 251 -266, 2012, Canesin et al. Galectin-3 expression is associated with bladder cancer progression and clinical outcome," Tumour Biol. 31 : 277-285, 2010, Wanninger et al. "Systemic and hepatic vein galectin-3 are increased in patients with alcoholic liver cirrhosis and negatively correlate with liver function," Cytokine. 55: 435-40, 201 1 . Moreover, experiments have shown that galectins, particularly galectin-1 and galectin-3, are directly involved in the pathogenesis of these classes of disease (Toussaint et al., "Galectin-1 , a gene preferentially expressed at the tumor margin, promotes glioblastoma cell invasion.", Mol Cancer. 1 1 :32, 2012, Liu et al 2012, Newlaczyl et al., "Galectin-3 - a jack-of-all-trades in cancer," Cancer Lett. 313: 123-128, 201 1 , Banh et al., "Tumor galectin-1 mediates tumor growth and metastasis through regulation of T-cell apoptosis," Cancer Res. 71 : 4423-31 , 201 1 , Lefranc et al., "Galectin-1 mediated biochemical controls of melanoma and glioma aggressive behavior," World J. Biol. Chem. 2: 193-201 , 201 1 , Forsman et al., "Galectin 3 aggravates joint inflammation and destruction in antigen- induced arthritis," Arthritis Reum. 63: 445-454, 201 1 , de Boer et al., "Galectin-3 in cardiac remodeling and heart failure," Curr. Heart Fail. Rep. 7, 1 -8, 2010, Ueland et al., "Galectin-3 in heart failure: high levels are associated with all-cause mortality," Int J Cardiol. 150: 361 -364, 201 1 , Ohshima et al., "Galectin 3 and its binding protein in rheumatoid arthritis," Arthritis Rheum. 48: 2788-2795, 2003).
[0065] The term "effective dose" means the amount of the GRG, GA-RG, GM, compounds described herein when administered as a parental dose to an animal or human result in at least 5% decrease of a chosen molecular marker.
[0066] In some embodiments, the effective dose of galacto-rhamnogalacturonate can be equivalent to an animal dose of about 120 mg/kg. In some embodiments, the effective dose of galacto-rhamnogalacturonate can be equivalent to an animal dose of 10 mg/kg to 180 mg/kg. In some embodiments, the effective dose of the galactomannan polysaccharide is equivalent to an animal dose of about 60 mg/kg. In some embodiments, the effective dose of the galactomannan polysaccharide is equivalent to an animal dose of 30 mg/kg to 180 mg/kg. In some embodiments, the effective dose can be given once, twice or three times weekly.
[0067] The term "efficacy" means demonstrating an improvement of lung fibrosis of at least 10% using treatment with the GRG, GA-RG, or GM compounds described herein as compared to treatment with vehicle-treated subject.
[0068] Other aspects of the invention relate to methods of treating a subject in need thereof. In some embodiments, the method comprises the step of obtaining a composition for intravenous or subcutaneous administration comprising a compound in an acceptable pharmaceutical carrier.
[0069] In some embodiments, the compound is a polysaccharide chemically defined as galacto-rhamnogalacturonate (GRG), a selectively depolymerized, branched heteropolymer whose backbone is predominantly comprised of 1 ,4-linked galacturonic acid (GalA) moieties, with a lesser backbone composition of alternating 1 ,4-linked GalA and 1 ,2-linked rhamnose (Rha), which in-turn is linked to any number of side chains, including predominantly 1 ,4-b-D-galactose (Gal) and 1 ,5-a-L-arabinose (Ara) residues. Other side chain minor constituents may include xylose (Xyl), glucose (Glu), and fucose (Fuc) or combinations thereof.
[0070] In some embodiments, the GRG compound is produced as described in U.S. Patent 8,236,780, which are incorporated expressly by reference for all purposes.
[0071] In some embodiments, the GRG compound can be produced as described in U.S. Patents 8,128,966, 8,187,624, U.S. Patent Application Publication Nos 2012/0315309 and 2012/030971 1 which are incorporated expressly by reference for all purposes.
[0072] In some embodiments, the compound is a polysaccharide chemically defined as galactoarabino-rhamnogalacturonate (GA-RG), a selectively depolymerized, branched heteropolymer whose backbone is predominantly comprised of 1 ,4-linked galacturonic acid (GalA) moieties, with a lesser backbone composition of alternating 1 ,4- linked GalA and 1 ,2-linked rhamnose (Rha), which in-turn is linked to any number of side chains, including predominantly 1 ,4-b-D-galactose (Gal) and 1 ,5-a-L-arabinose (Ara) residues. Other side chain minor constituents may include xylose (Xyl), glucose (Glu), and fucose (Fuc) or combinations thereof.
[0073] In some embodiments, the GA-RG compound is produced as described in International Patent Application PCT/US12/5531 1 , which is incorporated expressly by reference for all purposes.
[0074] In some embodiments, the molar percent of the 1 ,4-b-D-Gal and 1 ,5-a-L-Ara residues in the GA-RG compound of the present invention is 21 .5% with a molar ratio of 3:1 of 1 ,4-b-D-Gal to 1 ,5-a-L-Ara. [0075] In some embodiments, the compound is a polysaccharide chemically defined as galactoarabino-rhamnogalacturonate (GA-RG), with a molecular weight range of 20,000 to 70,000 Daltons as determined by SEC-RI method.
[0076] In some embodiments, the compound is a galactomannan (GM) polysaccharide composition produced as described in U.S. Patent Application US201 10077217, incorporated expressly by reference in its entirety for all purposes.
[0077] In some embodiments, the average molecular weight of the GM compound is approximately 4,000 and 60,000 Da, as determined by the SEC-MALLS method.
[0078] In some embodiments, the complex carbohydrate compounds described herein can target secreted and membrane-associated galectins, for example galectin-3.
[0079] In some embodiments, if an inducer of fibrosis, such bleomycin administered by intra-tracheal instillation, causes formation of the biochemical markers, but a concurrent administration of the inducer and a suitable complex carbohydrate compounds including but not limited to GM, GR, or GA-RG, does not lead to formation of the same marker, or leads to formation of the same marker but in a reduced amount, the polysaccharide prevents or slows down fibrosis.
[0080] Some aspects of the invention relate to a method of treating a patient at risk of pulmonary fibrosis. The method comprises administering to a patient a pharmaceutical composition comprising the carbohydrate compounds described herein in an amount sufficient to reduce the risk of developing pulmonary fibrosis. Other aspects of the invention relate to method of treating pulmonary fibrosis. The method comprises administering to a patient a pharmaceutical composition comprising the carbohydrate compounds described herein in an amount sufficient to reduce significantly one or more of the following: lung oedema, lung pathology associated with lung fibrosis, hydroxyproline accumulation, and expression of pro-inflammatory proteins, such as TGF- beta, IL-6, IL-8, IL-13, IP-10, osteopontin, TNF-alpha, CXCL-9/10, VEGF, and fibrogenic proteins, such as collagen and, elastin.
[0081] In some embodiments, the administration of the compound results in at least 5% reduction of lung oedema, at least 5% reduction of lung pathology associated with lung fibrosis, at least at least 5% reduction of hydroxyproline accumulation, at least 5% reduction of expression of pro- inflammatory proteins, at least 5% reduction of expression of fibrogenic proteins, wherein the subject in need thereof has at least one of the following: a primary or secondary lung fibrotic disease. In some embodiments, the administration of the effective dose results in at least 5% improvement of pulmonary function tests including forced vital capacities or oxygen diffusion.
EXAMPLE: Mouse Model
Example 1 : Early treatment of pulmonary fibrosis induced by bleomycin in C57BI/6 mice
[0082] Instillation of bleomycin in the trachea induces changes similar to diffuse pulmonary fibrosis and/or fibrosing alveolitis in humans. There is an increase in deposition and net synthesis of collagen in the lung with this model.
[0083] This mouse model of pulmonary fibrosis is used extensively in pre-clinical evaluation of drugs, as indicated in a review that reports on 240 individual studies (Int J of Biochem & Cell Biology 40 (2008) 362-382)
[0084] This model can be used to evaluate therapy started in the inflammatory phase of the belomycin insult (started < 7 day after bleomycin) or after the inflammatory phase and during fibrogenesis (started > 7 day after bleomycin). In this example, early therapy was evaluated.
[0085] The GM (GM-CT-01 ) and GA-RG (GR-MD-02) compounds were evaluated as preventative therapy in this mouse model.
[0086] GM-CT-01 is a galactomannan (GM) isolated from seeds of Cyamopsis tetragonoloba, or Guar gum, and subjected to a controlled partial chemical degradation. A backbone of the galactomannan is composed of (1→4)-linked β-D-mannopyranosyl units, to which single a-D-galactopyranosyl is attached by (1→6)-linkage. Chemical names of the galactomannan are 1 ,4- -D-Galactomannan, or [(1→6)-a-D-galacto-(1→4)- -D- mannan]. The average repeating unit of GM-CT-01 consists of seventeen β-D-Man residues and ten a-D-Gal residues (Man/Gal ratio is 1 .7), and an average polymeric molecule contains approximately 12 of such repeating units (for the average molecular weight ranging from 42,000 Da through 60,000 Da). [0087] GR-MD-02 is a galactoarabino-rhamnogalacturonate (GARG), a selectively depolymerized, branched heteropolymer whose backbone is predominantly comprised of 1 ,4-linked galacturonic acid (GalA) moieties, with a lesser backbone composition of alternating 1 ,4-linked GalA and 1 ,2-linked rhamnose (Rha), which in-turn is linked to any number of side chains, including predominantly 1 ,4-b-D-galactose (Gal) and 1 ,5-a-L- arabinose (Ara) residues. Other side chain minor constituents may include xylose (Xyl), glucose (Glu), and fucose (Fuc) or combinations thereof. The GR-MD-02 has an average molecular weight ranging from 20,000 Da to 70,000 Da.
[0088] Pulmonary fibrosis was induced in animals using intra-tracheal instillation of bleomycin at 2.25 U/kg. Animals were treated using a GM compound (GM-CT-01 ) or GARG compound (GR-MD-02) delivered by intravenous infusion as described in Table 1 .
Tablel
Figure imgf000016_0001
[0089] Animals were evaluated for their weight loss (Figure 1A-B). While there appeared to be a qualitative difference in weight loss between some groups, quantitative analysis of area under the curve showed no difference between groups.
[0090] Animals were euthanized on Day 21 and the lungs were weighed. Data shown in Figure 2 represent group means and SEM. Statistical comparison among all groups was performed using one-way ANOVA with Newman-Keuls multiple comparison test. Both the GM-CT-01 and the GR-MD-02 compounds were shown to significantly reduce lung weight in comparison to vehicle-treated mice.
[0091] After euthanasia, the total lung hydroxyproline content was measured. Data shown in Figure 3 represent group mean and SEM and expressed as microgram of hydroxyproline per mg of lung tissue. Statistical comparison to the "vehicle + Bleomycin" group was performed using one-way ANOVA with Holm-Sidak's multiple comparison test. Both the GM-CT-01 and the GA-MD-02 compounds were shown to reduce lung hydroxyproline content compared to vehicle-treated control mice.
[0092] The Modified Ashcroft Index score was determined for the vehicle-treated control mice and the GR-MD-02 and GM-CT-01 treated mice. Figure 4 shows the Modified Ashcroft Index score with outliers removed. There were two notable outliers. One vehicle-treated control mice was essentially not affected by bleomycin administration. In the GR treatment group, there was one animal with severe fibrosis. If these two animals are removed from the analysis, there is a significant different between vehicle and GR-MD-02 treated animals (p=0.01 12, unpaired t-test). There is also a significant difference between mice and GM-CT-01 treated animals (p=0.0199, unpaired t-test).
[0093] The general fibrosis score was determined. Figure 5 shows the general fibrosis score with outliers removed. There were two notable outliers. One vehicle- treated control mice was essentially not affected by bleomycin administration. In the GR- MD-02 treatment group, there was one animal with severe fibrosis. If these two animals are removed from the analysis, there is a significant different between vehicle and GR- MD-02 treated animals (p=0.0206, unpaired t-test) and GM-CT-01 treatment approaches significance (p=0.0979, unpaired t-test).
[0094] Inflammation score was determined. Figure 6 shows the Inflammation score with outliers removed. There were two notable outliers. One vehicle-treated control mice was essentially not affected by bleomycin administration. In the GR-MD-02 treatment group, there was one animal with severe fibrosis. If these two animals are removed from the analysis, then both treatment approach significance (GM-CT-01 vs. vehicle, p=0.0544 unpaired t-test and GR-MD-02 vs. vehicle, p=0.0508, unpaired t-test). [0095] Lung pathology with trichrome stain of saline control, vehicle + bleomycin, GM-CT-01 and GR-MD-02 treated animals was examined. All tissue samples were examined and examples are shown in Figure 7. Both GM-CT-01 and GR-MD-02 treatment were found to be effective at reducing fibrosis. Both the number of animals with fibrosis and the average degree of fibrosis were reduced with both treatments. Vehicle- treated animals tended to have contiguous areas of fibrosis affecting large areas of most lung lobes. Fibrous connective tissue stains blue with trichrome and is visualized as steaming blue fibers (asterisks, vehicle-treated animal, Figure 7). With both treatments, animals had smaller areas of fibrosis. There were often small knot-like formations (arrows, GR-MD-02 treated animal, Figure 7) or small masses (thick arrows, GM-CT-01 treated animal, Figure 7) of fibrosis, but these tended not to aggregate to the large formation seen in the vehicle-treated animals and to affect a smaller percentage of lungs.
[0096] Representative images with H & E are shown in Figure 8. H & E images show the same basic features as those stained with the trichrome stain. There are mixed mononuclear cell infiltrate associated with the fibrosis. This inflammation is more clearly evident with the HE stain (arrows, Figure 8).
[0097] The foregoing results showed that treatment with GM (GM-CT-01 ) and GA- RG (GR-MD-02) caused a significant reduction in mean lung weight and lung hydroxyproline content compared to the vehicle-treated control group.
[0098] Histologic analysis indicated that GM-CT-01 and GR-MD-02 produced a significant reduction in the Modified Ashcroft Index Score compared to the vehicle group.
[0099] Although the rest of the histological findings were not statistically significant, there were clear, notable trends suggesting a positive treatment effect for both drugs.
[00100] The results with the GM compound (GM-CT-01 ) and the GA-RG compound (GR-MD-02) are superior to the results using pirfenadone which is a small molecule comprising a modified phenyl pyridine. The results with GR-MD-02 were found to be similar to treatment with anti-TGFbeta, and the results with GM-CT-01 were found to be superior to treatment with anti-TGFbeta in the experience of the laboratory.
Example 2: Late treatment of pulmonary fibrosis induced by bleomycin in C57BI/6 mice [00101] In these experiments, treatment was initiated after the time when the inflammatory reaction is known to have subsided and the process is one of primarily fibrogenesis. Pulmonary fibrosis was induced in animals using bleomycin at 2.25 U/kg at day 0 as described above. Animals were treated using a GM compound intravenously (GM-CT-01 ) or GA-RG compound intravenously (GR-MD-02) as described in Table 2. Table 2
Figure imgf000019_0001
[00102] Animals were euthanized on Day 21 and the lungs were weighed. Data shown in Figure 9 represent group means and standard error of the means. While there was a decreased lung weight of the GR-MD-02 treated group as compared to control animals, this difference did not reach statistical significance.
[00103] The total lung hydroxyproline content was measured. Data shown in Figure 10 represent group mean and expressed as microgram of hydroxyproline per mg of lung tissue. Treatment with GR-MD-02 reduced the increase in hydroxyproline by 50%, but because of the variability in the data, this difference did not reach significance. There was less of a decrease in GM-CT-01 animals.
[00104] The Modified Ashcroft Index score was determined for the vehicle-treated control mice and the GR-MD-02 and GM-CT-0 -treated mice. Figure 1 1 shows the Aschcroft score. There was a reduction in the Aschroft score in the GR-MD-02 treated animals as compared to vehicle treated animals, but the change did not reach significance. There was little change in the GM-CT-01 treated group.
[00105] Figure 12 shows the general fibrosis score for each animal group. There was a reduction in the fibrosis score in animals treated with both GM-CT-01 and GR-MD- 02 as compared to the vehicle-treated animals, but the differences did not reach statistical significance.
[00106] Figure 13 shows the Inflammation score for each animal group. There was a reduction in inflammation score in the group treated with GR-MD-02 as compared to vehicle controls, but no change in the GM-CT-01 group as compared to vehicle control. The differences seen were not statistically significant.
[00107] Lung pathology with trichrome stain of saline control, vehicle + bleomycin, GM-CT-01 and GR-MD-02 treated animals was examined. All tissue samples were examined and examples are shown in Figure 14. GR-MD-02 treatment was found to be effective at reducing fibrosis, while GM-CT-01 had less effect. Vehicle-treated animals tended to have contiguous areas of fibrosis affecting large areas of most lung lobes. GR- MD-02 treated animals had smaller areas of fibrosis. There were often small knot-like formations or small masses of fibrosis, but these tended not to aggregate to the large formation seen in the vehicle-treated animals and to affect a smaller percentage of lungs.
[00108] Representative images with H & E are shown in Figure 15. H & E images show the same basic features as those stained with the trichrome stain. The vehicle control animals showed areas of dense fibrosis. There was some reduction in the fibrosis in the GM-CT-01 treated animals as indicated by more air space in the regions of fibrosis. In the GR-MD-02 treated animals there was a marked decrease in fibrous tissue.
[00109] Under the conditions of this study, both GM-CT-01 and GR-MD-02 treatment were effective at reducing pulmonary fibrosis in the bleomycin induced pulmonary fibrosis mouse model.
[00110] The results showed that GR-MD-02 exhibited the strongest anifibrotic effect under the conditions used. Animals in this group tended to have reduced percent of fibrotic lung, inflammation, semi quantitative general fibrosis score and fibrosis index score. While these changes were not statistically significant, they were noticeable and there were clear trends in the data supportive of a positive treatment effect.
[00111] The results showed that GM-CT-01 exhibited a more modest positive treatment effect. The percent of lung affected and semiquantitative fibrosis score were notably reduced but these findings were not statistically significant. There was less improvement in the index and inflammation scores. Regardless, there were clear trends in the data supportive of a positive treatment effect.
Conclusions
[00112] In the early treatment model, both GR-MD-02 and GM-CT-01 were shown to markedly reduce lung weight and hydroxyproline content, with reduction of histological evidence of inflammation and fibrosis when compared to vehicle-treated bleomycin mouse model.
[00113] Treatment: results showed that GR-MD-02 was more effective than GM-CT- 01 in treating pulmonary fibrosis in the mouse model.
[00114] A mouse chronic model, such as intravenous bleomycin, can be used in future studies to evaluate the effect of different dosages and/or different schedules of administration.
[00115] While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention. All publications, patents and sequence database entries mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference.

Claims

1 . A method comprising:
a. obtaining a composition for parenteral or enteral administration comprising a galacto-rhamnogalacturonate in a pharmaceutical acceptable carrier, and
b. administering to a subject in need thereof an effective dose of the composition that results in at least one of the following:
at least 5% reduction of lung edema,
at least 5% reduction of lung pathology severity scores associated with lung fibrosis, at least 5% reduction of lung tissue hydroxyproline accumulation,
at least 5% reduction of expression of pro-inflammatory proteins,
at least 5% reduction of expression of fibrogenic proteins,
wherein the subject in need thereof has at least one of the following: a primary or secondary lung fibrotic disease.
2. The method of claim 1 wherein the effective dose is equivalent to an animal dose of 10 mg/kg to 180 mg/kg given once, twice or three times weekly.
3. The method of claim 1 wherein the pro-inflammatory proteins comprise TGF-beta, IL-6, IL-8, IL-13, osteopontin, TNF-alpha, CXCL-9/10, or VEGF.
4. The method of claim 1 wherein the fibrogenic proteins comprise collagen, or elastin.
5. The method of claim 1 wherein the galacto-rhamnogalacturonate comprises a 1 ,4-linked galacturonic acid (GalA) and methyl galacturonate (MeGalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D-galactose residues.
6. The method of claim 1 wherein the galacto-rhamnogalacturonate comprises a 1 ,4-linked galacturonic acid (GalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D- galactose residues.
7. The method of claim 1 wherein the galacto-rhamnogalacturonate further comprises xylose, glucose, fucose residues or combination thereof.
8. The method of claim 1 wherein the galacto-rhamnogalacturonate is substantially free of 1 ,5-a-L-Ara residues.
9. The method of claim 1 wherein the galacto-rhamnogalacturonate is a galactoarabino-rhamnogalacturonate, comprising a 1 ,4-linked galacturonic acid (GalA) and methyl galacturonate (MeGalA) residues backbone linked to branched heteropolymers of alternating oligomers of a -1 ,2 linked rhamnose and a -1 ,4-linked GalA residues, the rhamnose residues carrying a primary branching of oligomers of 1 ,4- β -D- galactose residues, 1 ,5- a -L-arabinose residues, or combinations thereof.
10. The method of claim 9 wherein the 1 ,4- β -D-galactose and 1 ,5- a -L- arabinose residues are present in a 2:1 or a 3:1 ratio.
1 1 . The method of claim 10 wherein the 1 ,4- β -D-galactose residues, the 1 ,5- a -L-arabinose residues or combination thereof represent at least 10 molar percent of the total molar carbohydrates.
12. The method of claim 1 wherein the galacto-rhamnogalacturonate has an average molecular weight ranging from 5 kDa to 55 kDa as determined by SEC-RI and/or the SEC-MALLS methods.
13. The method of claim 1 wherein the galacto-rhamnogalacturonate has an average molecular weight ranging from 2 kDa to 80 kDa as determined by SEC-RI and/or the SEC-MALLS methods.
14. The method of claim 1 wherein the galacto-rhamnogalacturonate has an average molecular weight ranging from 20 kDa to 70 kDa as determined by SEC-RI and/or the SEC-MALLS methods.
15. A method comprising:
a. obtaining a composition for parenteral or enteral administration comprising a galactomannan polysaccharide in a pharmaceutical acceptable carrier, and
b. administering to a subject in need thereof an effective dose of the composition that results in at least one of the following:
at least 5% reduction of lung oedema,
at least 5% reduction of lung pathology associated with lung fibrosis, at least at least 5% reduction of hydroxyproline accumulation,
at least 5% reduction of expression of pro-inflammatory proteins,
at least 5% reduction of expression of fibrogenic proteins,
wherein the subject in need thereof has at least one of the following: a primary or secondary lung fibrotic disease.
16. The method of claim 15 wherein the effective dose is equivalent to an animal dose of about 60 mg/kg.
17. The method of claim 15 wherein in the step of obtaining, the galactomannan polysaccharide has a mannose to galactose ratio ranging from 1 :1 to 1 :4.
18. The method of claim 15 wherein in the step of obtaining, the galactomannan polysaccharide has a mannose to galactose ratio of 1 .7:1 .
19. The method of claim 15 wherein in the step of obtaining, the galactomannan polysaccharide has a molecular weight of about 48,000 D.
20. The method of claim 15 wherein in the step of obtaining, the galactomannan polysaccharide has an average molecular weight ranging from 4,000 D to 60,000 D.
PCT/US2014/017353 2013-02-20 2014-02-20 Method for treatment of pulmonary fibrosis WO2014130648A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361767057P 2013-02-20 2013-02-20
US61/767,057 2013-02-20

Publications (1)

Publication Number Publication Date
WO2014130648A1 true WO2014130648A1 (en) 2014-08-28

Family

ID=51351640

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/017353 WO2014130648A1 (en) 2013-02-20 2014-02-20 Method for treatment of pulmonary fibrosis

Country Status (2)

Country Link
US (2) US9339515B2 (en)
WO (1) WO2014130648A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008011216A2 (en) * 2006-05-16 2008-01-24 Pro-Pharmaceuticals, Inc. Galactose-pronged polysaccharides in a formulation for antifibrotic therapies
KR101968370B1 (en) 2011-12-28 2019-04-11 갈렉틴 테라퓨틱스, 인크. Composition of novel carbohydrate drug for treatment of human diseases
CA2885970C (en) 2012-09-17 2020-10-20 Peter G. Traber Method for enhancing specific immunotherapies in cancer treatment
CN112672765A (en) 2018-06-29 2021-04-16 格莱科斯生物医药公司 Conjugates
US20230038373A1 (en) 2019-12-18 2023-02-09 Glykos Biomedical Oy Stabile conjugate

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004091634A1 (en) * 2003-04-07 2004-10-28 Glycogenesys, Inc. Composition and uses of galectin antagonists
US20110077217A1 (en) * 2003-09-08 2011-03-31 Pro-Pharmaceuticals, Inc. Selectively depolymerized galactomannan polysaccharide
US20120282220A1 (en) * 2006-05-16 2012-11-08 Galectin Therapeutics Galactose-pronged polysaccharides in a formulation for antifibrotic therapies
WO2012162531A1 (en) * 2011-05-25 2012-11-29 Bg Medicine, Inc. Inhibitors of galectin-3 and methods of use thereof

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2132577A (en) 1934-08-28 1938-10-11 Gen Foods Corp Method of preparing pectin
US2444266A (en) 1944-03-31 1948-06-29 Us Agriculture Pectic materials and method of preparing same
US2503258A (en) 1948-01-23 1950-04-11 Us Agriculture Neutralization of pectinic acids with sodium bicarbonate
US4016351A (en) 1976-02-04 1977-04-05 Eschinasi Emile H Pectic substances with varying methoxyl content and process therefor
US4268533A (en) 1979-11-21 1981-05-19 General Foods Corporation Quick-set low methoxyl pectin composition
JPS61502124A (en) 1984-05-09 1986-09-25 ジ オ−ストラリアン ナシヨナル ユニヴア−シテイ− How to modulate the immune response
US4686106A (en) 1986-06-16 1987-08-11 General Foods Corporation Pectin food product
US5071970A (en) 1988-11-23 1991-12-10 University Of Florida Process for producing pectin with a high-to-medium methoxyl content from beet pulp
US5605938A (en) 1991-05-31 1997-02-25 Gliatech, Inc. Methods and compositions for inhibition of cell invasion and fibrosis using dextran sulfate
AU680411B2 (en) 1992-04-03 1997-07-31 Genentech Inc. Antibodies to alphavbeta3 integrin
US7491708B1 (en) 1993-03-01 2009-02-17 David Platt Modified pectin
US5498702A (en) 1993-12-16 1996-03-12 California Natural Products Treated pectinic acid process and product
US5834442A (en) 1994-07-07 1998-11-10 Barbara Ann Karmanos Cancer Institute Method for inhibiting cancer metastasis by oral administration of soluble modified citrus pectin
US5681923A (en) 1995-10-06 1997-10-28 Platt; David Tumor derived carbohydrate binding protein
US6632797B2 (en) 1996-06-24 2003-10-14 Matti Siren Method of treating angiogenesis
WO1998008528A1 (en) 1996-08-30 1998-03-05 Biomeasure Incorporated Method of inhibiting fibrosis with a somatostatin agonist
US5972995A (en) 1997-10-16 1999-10-26 Children's Hospital Medical Center Of Northern California Compositions and methods for cystic fibrosis therapy
WO1999055715A2 (en) 1998-04-28 1999-11-04 Galenica Pharmaceuticals, Inc. Polysaccharide-antigen conjugates
US7022683B1 (en) 1998-05-13 2006-04-04 Carrington Laboratories, Inc. Pharmacological compositions comprising pectins having high molecular weights and low degrees of methoxylation
KR20000011247A (en) 1998-07-23 2000-02-25 김윤 Composition and pharmaceutical dosage form for colonic drug delivery using polysaccharides
US6500807B1 (en) 1999-02-02 2002-12-31 Safescience, Inc. Modified pectin and nucleic acid composition
EP1163910B1 (en) 1999-03-25 2007-09-26 Mitsubishi Pharma Corporation Rho kinase inhibitors for the prevention or treatment of interstitial pneumonia and pulmonary fibrosis
GB9909135D0 (en) 1999-04-22 1999-06-16 Univ Wales Medicine Cystic fibrosis medicaments
CA2370814C (en) 1999-04-22 2010-07-06 Biogen, Inc. Method for the treatment of fibrosis using an antagonist of the integrin alpha-4 subunit
US6893637B1 (en) 1999-10-21 2005-05-17 Zymogenetics, Inc. Method of treating fibrosis
IT1316986B1 (en) 2000-01-25 2003-05-26 Sigma Tau Ind Farmaceuti GLYCOSAMINOGLICAN DERIVATIVES PARTIALLY DESULPHATED NONANTICOAGULANTS WITH ANTIANGIOGENIC ACTIVITY.
US6642205B2 (en) 2000-09-25 2003-11-04 Pro-Pharmaceuticals, Inc. Methods and compositions for reducing side effects in chemotherapeutic treatments
US6982255B2 (en) 2001-03-27 2006-01-03 Anatole Klyosov Delivery of a therapeutic agent in a formulation for reduced toxicity
US6645946B1 (en) 2001-03-27 2003-11-11 Pro-Pharmaceuticals, Inc. Delivery of a therapeutic agent in a formulation for reduced toxicity
DK1383516T3 (en) 2001-03-27 2012-02-20 Pro Pharmaceuticals Inc Administration of a polysaccharide concomitantly with a chemotherapeutic agent for the treatment of cancer
US6914055B2 (en) 2001-03-27 2005-07-05 Anatole Klyosov Delivery of a therapeutic agent in a formulation for reduced toxicity
US6770622B2 (en) 2001-06-08 2004-08-03 Gary A. Jarvis N-terminally truncated galectin-3 for use in treating cancer
US6680306B2 (en) 2001-06-21 2004-01-20 Glycogenesys, Inc. Method for enhancing the effectiveness of cancer therapies
US20030004132A1 (en) 2001-06-22 2003-01-02 Yan Chang Method and material for treating immune diseases
US6812230B2 (en) 2001-08-07 2004-11-02 Schering Aktiengesellschaft Non-peptide CCR1 receptor antagonists for the treatment of progressive renal fibrosis
US6890906B2 (en) 2001-11-21 2005-05-10 Glycogenesys, Inc. Method for controlling angiogenesis in animals
US20040121981A1 (en) 2001-11-21 2004-06-24 Glycogenesys, Inc. Method for controlling angiogenesis in animals
US6964761B1 (en) 2001-12-12 2005-11-15 New York University Method of treating idiopathic pulmonary fibrosis with aerosolized IFN-γ
US6986995B2 (en) 2002-02-28 2006-01-17 Prometheus Laboratories, Inc. Methods of diagnosing liver fibrosis
JP2004107295A (en) 2002-09-20 2004-04-08 National Agriculture & Bio-Oriented Research Organization Histamine release inhibitor
US20040223971A1 (en) 2003-04-07 2004-11-11 Glycogenesys, Inc. Composition and uses of galectin antagonists
RU2240123C1 (en) 2003-04-08 2004-11-20 Пятигорская государственная фармацевтическая академия Exogenous biologically active conjugating substances
WO2004096998A2 (en) 2003-04-29 2004-11-11 Vanderbilt University Nanoparticular tumor targeting and therapy
US9050352B2 (en) 2003-10-16 2015-06-09 Stephen John Ralph Immunomodulating compositions and uses therefor
US7078064B2 (en) 2003-12-03 2006-07-18 George Zabrecky Compositions and methods useful for treating and preventing chronic liver disease, chronic HCV infection and non-alcoholic steatohepatitis
EP2926818A1 (en) 2004-03-26 2015-10-07 La Jolla Pharmaceutical Company Modified pectins, compositions and methods related thereto
CN101426530B (en) 2004-05-12 2012-03-07 梅迪沃什有限公司 Wound healing polymer compositions and methods for use thereof
EP1768687A2 (en) 2004-06-29 2007-04-04 Massachusetts Institute Of Technology Methods and compositions related to the modulation of intercellular junctions
US20060074050A1 (en) 2004-07-14 2006-04-06 Glycogenesys, Inc. Composition and method for treating hyperproliferative diseases
KR101074158B1 (en) 2008-04-07 2011-10-17 재단법인 한국원자력의학원 Composition comprising polysaccharide extracted from panax ginseng preventing and treating liver diseases
KR101968370B1 (en) 2011-12-28 2019-04-11 갈렉틴 테라퓨틱스, 인크. Composition of novel carbohydrate drug for treatment of human diseases
US9763974B2 (en) 2012-06-06 2017-09-19 Galectin Therapeutics, Inc. Galacto-rhamnogalacturonate compositions for the treatment of diseases associated with elevated inducible nitric oxide synthase

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004091634A1 (en) * 2003-04-07 2004-10-28 Glycogenesys, Inc. Composition and uses of galectin antagonists
US20110077217A1 (en) * 2003-09-08 2011-03-31 Pro-Pharmaceuticals, Inc. Selectively depolymerized galactomannan polysaccharide
US20120282220A1 (en) * 2006-05-16 2012-11-08 Galectin Therapeutics Galactose-pronged polysaccharides in a formulation for antifibrotic therapies
WO2012162531A1 (en) * 2011-05-25 2012-11-29 Bg Medicine, Inc. Inhibitors of galectin-3 and methods of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MACKINNON, A ET AL.: "Regulation Of Transforming Growth Factor-Betal-Driven Lung Fibrosis By Galectin-3.", AM J RESPIR CRIT CARE MED., vol. 185, no. 5, 1 March 2012 (2012-03-01), pages 537 - 546 *

Also Published As

Publication number Publication date
US9968631B2 (en) 2018-05-15
US20140235571A1 (en) 2014-08-21
US20160213703A1 (en) 2016-07-28
US9339515B2 (en) 2016-05-17

Similar Documents

Publication Publication Date Title
US9968631B2 (en) Method for treatment of pulmonary fibrosis
Liu et al. Secreted protein acidic and rich in cysteine mediated biomimetic delivery of methotrexate by albumin-based nanomedicines for rheumatoid arthritis therapy
US8658787B2 (en) Galacto-rhamnogalacturonate compositions for the treatment of non-alcoholic steatohepatitis and non-alcoholic fatty liver disease
AU2013329148B2 (en) Galactose-pronged carbohydrate compounds for the treatment of diabetic nephropathy and associated disorders
Hadji et al. Advances in the treatment of inflammatory bowel disease: Focus on polysaccharide nanoparticulate drug delivery systems
AU2019201691B2 (en) Compound of glycosaminoglycan and its preparation method as well as application
JP6517141B2 (en) Galacto-rhamnogalacturonate compositions for treating diseases associated with highly inducible nitric oxide synthase
US20150238617A1 (en) Silk reservoirs for sustained delivery of anti-cancer agents
AU2014372566B2 (en) Uses of oligouronates in cancer treatment
CN100558368C (en) Chemical compound is used for the treatment of purposes in the medicine of immune disease in preparation
ES2458818T3 (en) Methods to evaluate the activity of a polysaccharide composition
Cai et al. Multifunctionalized brush-like glycopolymers with high affinity to P-selectin and antitumor metastasis activity
Mestechkina et al. Effect of polysaccharides on biological activity of human lactoferrin
Park et al. Molecular Targeting of VEGF with a Suramin Fragment–DOCA Conjugate by Mimicking the Action of Low Molecular Weight Heparins. Biomolecules 2021, 11, 46
Phillips CELL SIGNALLING POTENTIAL OF HYDROCOLLOIDS: POSSIBLE NEW APPLICATIONS? AO Phillips Institute of Nephrology, University of Wales College of Medicine, Heath Park, Cardiff
Head Urinary excretion of (1-3)-beta-D-glucans

Legal Events

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

Ref document number: 14753502

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14753502

Country of ref document: EP

Kind code of ref document: A1