CN116726163A - Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease - Google Patents

Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease Download PDF

Info

Publication number
CN116726163A
CN116726163A CN202310450575.2A CN202310450575A CN116726163A CN 116726163 A CN116726163 A CN 116726163A CN 202310450575 A CN202310450575 A CN 202310450575A CN 116726163 A CN116726163 A CN 116726163A
Authority
CN
China
Prior art keywords
masp
fibrosis
antibody
subject
seq
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202310450575.2A
Other languages
Chinese (zh)
Inventor
N.J.布伦斯基尔
G.A.德莫普洛斯
T.杜德勒
H-W.施维布尔
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Leicester
Omeros Corp
Original Assignee
University of Leicester
Omeros Medical Systems 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
Priority claimed from US15/399,524 external-priority patent/US10736960B2/en
Priority claimed from US15/470,647 external-priority patent/US20170253667A1/en
Application filed by University of Leicester, Omeros Medical Systems Inc filed Critical University of Leicester
Publication of CN116726163A publication Critical patent/CN116726163A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21104Mannan-binding lectin-associated serine protease-2 (3.4.21.104)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Zoology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Environmental Sciences (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Hematology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Production Of Liquid Hydrocarbon Mixture For Refining Petroleum (AREA)

Abstract

The present application relates to a method for reducing proteinuria in a human subject suffering from immunoglobulin a kidney disease. The method comprises the step of administering to a subject in need thereof an amount of a MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation.

Description

Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease
The present application is a divisional application of chinese patent application 201780060710.5, "method for reducing proteinuria in human subjects with immunoglobulin a kidney disease," having a filing date of 2017, 10, 12.
Statement regarding sequence listing
The sequence listing relevant to the present application is provided in text format instead of paper copy and is incorporated herein by reference. The text file containing the sequence list is named mp_1_0269_pct_sequence_listing_20171013_st25. The text file is 136KB and is created in 10 months and 10 days of 2017; and submitted via EFS-Web as the specification is submitted.
Technical Field
The present application relates to methods for reducing proteinuria in a human subject suffering from or at risk of developing immunoglobulin a nephropathy (IgAN).
Background
The complement system provides an early mechanism of action for initiating, enhancing and coordinating immune responses to microbial infections and other acute lesions in humans and other vertebrates (m.k.liszewski and J.P.Atkinson,1993,in Fundamental Immunology, third edition, w.e.Paul editions, raven Press, ltd., new York). Although complement activation provides an important first line defense against potential pathogens, complement activity that promotes a protective immune response may also present a potential threat to the host (K.R. Kalli, et al, springer Semin. Immunopathol.15:417-431,1994;B.P.Morgan,Eur.J.Clinical Investig.24:219-228,1994). For example, C3 and C5 protein hydrolysates recruit and activate neutrophils. Although essential for host defense, activated neutrophils indiscriminately release destructive enzymes, potentially causing organ damage. In addition, complement activation may result in the deposition of the lysed complement components on nearby host cells as well as microbial targets, resulting in host cell lysis.
The complement system has also been implicated in the pathogenesis of a number of acute and chronic disease conditions including: myocardial infarction, stroke, ARDS, reperfusion injury, septic shock, capillary leakage after thermal burn, inflammation after cardiopulmonary bypass, graft rejection, rheumatoid arthritis, multiple sclerosis, myasthenia gravis and alzheimer's disease. In almost all of these diseases, complement is not the etiology, but one of several factors involved in pathogenesis. Nonetheless, complement activation may be the primary pathological mechanism and represents a significant point in the clinical control of many such disease conditions. Increased awareness of the importance of complement-mediated tissue damage in various disease conditions has increased the need for effective complement inhibiting drugs. To date, elkulizumab Antibodies to C5 are the only complement-targeted drugs that have been approved for human use. However, C5 is one of several effector molecules located "downstream" of the complement system, and blocking of C5 does not inhibit activation of the complement system. Thus, inhibitors of the initial step of complement activation have significant advantages over "downstream" complement inhibitors.
Currently, it is widely believed that the complement system can be activated by three different pathways: classical pathway, lectin pathway and alternative pathway. The classical pathway is usually triggered by a complex of host antibodies bound to foreign particles (i.e. antigens) and thus requires prior exposure to the antigen to generate a specific antibody response. Because activation of the classical pathway depends on the host's previous adaptive immune response, the classical pathway is part of the adaptive immune system. In contrast, both the lectin pathway and the alternative pathway are independent of adaptive immunity (adaptive immunity), and are part of the innate immune system.
Activation of the complement system results in sequential activation of serine protease zymogens. The first step in classical pathway activation is the binding of the specific recognition molecule C1q to antigen-binding IgG and IgM molecules. C1q binds to C1r and C1s serine protease zymogens to a complex called C1. After binding of C1q to the immunocomplexes, the Arg-Ile site of C1r undergoes autoproteolytic cleavage, followed by C1 r-mediated cleavage and activation of C1s, thereby obtaining the ability to cleave C4 and C2. C4 cleaves into two fragments, called C4a and C4b, and similarly C2 cleaves into C2a and C2b. The C4b fragment is capable of forming a covalent bond with an adjacent hydroxyl or amino group and generates a C3 convertase (C4 b2 a) by non-covalent interaction with the C2a fragment of activated C2. The activation of C3 by proteolytic cleavage of C3 convertase (C4 b2 a) into C3a and C3b subfractions leads to the production of C5 convertase (C4 b2a3 b), which C5 convertase (C4 b2a3 b) leads to the formation of a membrane attack complex (C5 b in combination with C6, C7, C8 and C-9, also known as "MAC") by cleavage of C5, which can disrupt cell membranes leading to cell lysis. Activated forms of C3 and C4 (C3 b and C4 b) are covalently deposited on exogenous target surfaces, which are recognized by complement receptors on a variety of phagocytes.
Independently, the first step in activation of the complement system via the lectin pathway is also the binding of specific recognition molecules, followed by activation of the bound serine protease zymogen. However, the recognition molecules in the lectin pathway comprise a group of carbohydrate binding proteins (mannan binding lectin (MBL), H-fiber gelling protein (H-ficolin), M-fiber gelling protein, L-fiber gelling protein and C-lectin CL-11) (collectively referred to as lectins), rather than binding to immune complexes via C1 q. See J.Lu et al, biochim. Biophys. Acta 1572:387-400, (2002); holmskov et al, annu. Rev. Immunol.21:547-578 (2003); teh et al, immunology 101:225-232 (2000)). See also J.Luet et al Biochim Biophys Acta 1572:387-400 (2002); holmskov et al, annu Rev Immunol 21:547-578 (2003); teh et al, immunology 101:225-232 (2000); hansen et al, J.Immunol 185 (10): 6096-6104 (2010).
Ikeda et al first demonstrated that, like C1q, MBL was able to activate the complement system in a C4-dependent manner after binding to yeast mannan-coated erythrocytes (Ikeda et al J.biol. Chem.262:7451-7454, (1987)). MBL is a member of the collectin family of proteins, a calcium-dependent lectin, that binds to carbohydrates with 3-and 4-hydroxy groups located on the equatorial plane of the pyranose ring. Thus, the important ligands for MBL are D-mannose and N-acetyl-D-glucosamine, whereas carbohydrates that do not meet this steric requirement have no detectable affinity for MBL (Weis et al Nature 360:127-134, (1992)). The interaction between MBL and monovalent sugars is extremely weak, and the dissociation constant is typically in the range of single-digit millimoles (lmar). MBL achieves tight specific binding to glycan ligands by affinity, i.e., by interaction with multiple monosaccharide residues that are positioned in close proximity to each other at the same time (Lee et al, archiv. Biochem. Biophys.299:129-136, (1992)). MBL recognizes carbohydrate patterns that generally modify microorganisms such as bacteria, yeasts, parasites and certain viruses. In contrast, MBL does not recognize D-galactose and sialic acid, i.e., penultimate and penultimate sugars, which generally modify "mature" complex glycoconjugates present on mammalian plasma and cell surface glycoproteins. This binding specificity is thought to promote recognition of "foreign" surfaces and to help protect from "self-activation". MBL does, however, bind with high affinity high mannose "precursor" glycan clusters located on N-linked glycoproteins and glycolipids sequestered in the mammalian cell endoplasmic reticulum and golgi (Maynard et al, j. Biol. Chem.257:3788-3794, (1982)). Thus, damaged cells are potential targets for activation by MBL-bound lectin pathway.
The fiber gel protein (ficolin) has lectin domain of a type different from MBL, called fibrinogen-like domain. Fiber gel proteins to be Ca independent ++ In a manner that binds to the sugar residue. In humans, three types of fiber-gelled proteins have been identified (L-fiber-gelled eggsWhite, M-fiber gel protein and H-fiber gel protein). Both serum fibrinolytic proteins, L-and H-fibrinogenemic proteins, together have specificity for N-acetyl-D-glucosamine; however, H-fiber gelator also binds N-acetyl-D-galactosamine. The difference in saccharide specificity of L-fibronectin, H-fibronectin, CL-11 and MBL means that different lectins may be complementary, although overlapping, but may target different glycoconjugates. This view is supported by the recent report that in known lectins of the lectin pathway, only L-fiber gelsolin specifically binds lipoteichoic acid, a cell wall glycoconjugate present on all gram-positive bacteria (Lynch et al J.Immunol.172:1198-1202, (2004)). Collectin (i.e., MBL) and fiber-gelling proteins have no significant similarity in amino acid sequence. However, the two groups of proteins have similar domain organization, similar to C1q, assembled into an oligomeric structure, thus maximizing the likelihood of multi-site binding.
The serum concentration of MBL is highly variable in healthy humans, which is genetically controlled by polymorphisms/mutations in both the promoter and coding region of the MBL gene. As an acute phase protein, MBL expression is further up-regulated during inflammation. The L-fiber gel protein was present in serum at a concentration similar to that of MBL. Thus, the L-fiber gel protein branch of the lectin pathway may be comparable in strength to the MBL arm. MBL and fiber-gelling proteins may also function as opsonins, which allow phagocytic targeting to MBL and fiber-gelling protein-decorated surfaces (see Jack et al, JLeukoc biol.,77 (3): 328-36 (2004), matsushita and Fujita, immunobiology,205 (4-5): 490-7 (2002), aoyagi et al, JImmunol,174 (1): 418-25 (2005): this opsonin action requires these proteins to interact with phagocytic receptors (Kuhlman et al, J. Exp. Med.169:1733, (1989); matsushita et al, J. Biol. Chem.271:2448-54, (1996)), the identity of which has not been established.
Human MBL forms a specific, high affinity interaction with a unique C1r/C1 s-like serine protease, known as MBL-associated serine protease (MBL-associated serine proteases, MASP), via its collagen-like domain. Three MASP have been described so far. First, a single enzyme "MASP" was identified, which is characterized by the enzyme responsible for the initiation of the complement cascade (i.e., cleavage of C2 and C4) (Matsushita et al, J Exp Med 176 (6): 1497-1502 (1992); ji et al, J. Immunol.150:571-578 (1993)). MASP activity was then determined, in fact as a mixture of two proteases MASP-1 and MASP-2 (Thiel et al Nature 386:506-510, (1997)). However, MBL-MASP-2 complex alone proved to be sufficient for complement activation (Vorup-Jensen et al J.Immunol.165:2093-2100, (2000)). In addition, only MASP-2 rapidly cleaves C2 and C4 (Ambrus et al J.Immunol.170:1374-1382, (2003)). MASP-2 is thus a protease responsible for activating C4 and C2 to produce the C3 convertase C4b2 a. This is a significant difference from the classical pathway of the C1 complex in that the synergy of two specific serine proteases (C1 r and C1 s) in the C1 complex results in activation of the complement system. In addition, a third novel protease, MASP-3 (Dahl, M.R., et al, immunity 15:127-35,2001), has been isolated. MASP-1 and MASP-3 are alternatively spliced products of the same gene.
MASP shares the same domain organization as those of the enzyme components C1r and C1s of the C1 complex (Sim et al biochem. Soc. Trans.28:545, 2000). These domains include the N-terminal C1r/C1 s/sea urchin VEGF/bone morphogenic protein (CUB) domain, the epidermal growth factor-like domain, the second CUB domain, the tandem of complement regulatory protein domains, and serine protease domains. As in the C1 protease, activation of MASP-2 occurs by cleavage of Arg-Ile bonds near the serine protease domain, which separates the enzyme into disulfide-linked A and B chains, the latter consisting of serine protease domains.
MBL can also bind to variable slice forms of MASP-2 (referred to as the 19kDa MBL-associated protein (MAp 19) or small MBL-associated protein (sMAP), which lack the catalytic activity of MASP-2). (Stover, J.Immunol.162:3481-90, (1999); takahashi et al, int.Immunol.11:859-863, (1999)). MAp19 comprises the first two domains of MASP-2 followed by an additional sequence of 4 unique amino acids. The function of MAp19 is ambiguous (Degn et al, JImmunol. Methods, 2011). MASP-1 and MASP-2 genes are located on human chromosome 3 and chromosome 1, respectively (Schwaebe et al, immunobiology 205:455-466, (2002)).
Several lines of evidence indicate that different MBL-MASP complexes exist and that a substantial portion of MASP in serum is not complexed with MBL (Thiel, et al J.Immunol.165:878-887, (2000)). Both H-and L-fiber gelata bind to all MASP and activate the lectin complement pathway as MBL (Dahl et al, immunity 15:127-35, (2001); matsushita et al, J.Immunol.168:3502-3506, (2002)). Both the lectin and classical pathways form a common C3 convertase (C4 b2 a), where the two pathways meet.
It is widely believed that in natural hosts, the lectin pathway plays an important role in host defense against infection. Strong evidence of MBL involvement in host defense comes from analysis of patients with reduced serum levels of functional MBL (Kilpatrick, biochem. Biophys. Acta1572:401-413, (2002)). These patients show susceptibility to recurrent bacterial and fungal infections. These symptoms are often evident early in life during the vulnerable apparent window, as the antibody titer obtained from the parent is reduced but before the full antibody response has developed. This symptom is often caused by several site mutations in the collagen portion of MBL, which interfere with the correct formation of MBL oligomers. However, since MBL can function as a complement independent opsonin, it is not known how much increased susceptibility to infection is due to impaired complement activation.
All three pathways (i.e., classical, lectin, and alternative pathways) are thought to converge at C5, which is cleaved to form products with various pro-inflammatory effects. The post-convergence pathway is referred to as the terminal complement pathway. C5a is the most potent anaphylatoxin, causing smooth muscle and vascular tone and vascular permeability changes. It is also a potent chemokine and activator of both neutrophils and monocytes. C5 a-mediated cellular activation can significantly amplify inflammatory responses by inducing release of a variety of additional inflammatory mediators, including cytokines, hydrolases, arachidonic acid metabolites, and reactive oxygen species. C5 cleavage results in the formation of C5b-9, which is also known as a Membrane Attack Complex (MAC). There is strong evidence that sub-cleaved MAC deposition may play an important role in inflammation in addition to cleaving the pore-forming complex.
In addition to its fundamental role in immune defense, the complement system is responsible for tissue damage in many clinical diseases. Although there is a great deal of evidence that both the classical and alternative complement pathways exist in the pathogenesis of non-infectious human diseases, evaluation of the effects of the lectin pathway has only just begun. Recent studies provide evidence that activation of the lectin pathway may be responsible for complement activation and related inflammation in ischemia/reperfusion injury. Cold et al (2000) reported that cultured endothelial cells subjected to oxidative stress bind MBL and showed C3 deposition after exposure to human serum (Cold et al, am. J. Pathol.156:1549-1556, (2000)). In addition, treatment of human serum with blocking anti-MBL monoclonal antibodies inhibited MBL binding and complement activation. These findings were extended to myocardial ischemia-reperfusion rat models in which rats treated with blocking antibodies to rat MBL showed significantly less myocardial damage after coronary occlusion than rats treated with control antibodies (Jordan et al Circulation 104:1413-1418, (2001)). The molecular mechanism by which MBL binds to vascular endothelium following oxidative stress is not known; recent studies have shown that activation of the lectin pathway after oxidative stress is probably mediated by binding of MBL to vascular endothelial cytokeratin, rather than to glycoconjugates (Cold et al, am. J. Pathol.159:1045-1054, (2001)). Other studies have shown that the classical and alternative pathways in the pathogenesis of ischemia/reperfusion injury and the role of the lectin pathway in this disease remain controversial (Riedermann, n.c., et al, am.j. Pathl.162:363-367, 2003).
Fibrosis is excessive connective tissue formation in an organ or tissue, usually in response to injury or injury. Fibrosis is marked by the production of excessive extracellular matrix after local trauma. Normal physiological response to injury results in connective tissue deposition, but this initial beneficial repair process may persist and become ill-conditioned, altering the structure and function of the tissue. At the cellular level, epithelial cells and fibroblasts proliferate and differentiate into myofibroblasts, resulting in matrix contraction, increased strength, microvascular compression, and hypoxia. The influx of inflammatory cells (including macrophages and lymphocytes) results in cytokine release and the expansion of deposition of collagen, fibronectin and other molecular markers of fibrosis. Conventional therapeutic approaches primarily use corticosteroids and immunosuppressive drugs to target the inflammatory process of fibrosis. Unfortunately, these anti-inflammatory agents have little to no clinical effect. There is currently no effective therapy or therapeutic for fibrosis, but animal studies and realistic human reports indicate that fibrotic tissue damage is reversible (Tampe and Zeisberg, nat Rev Nephrol, vol 10:226-237, 2014).
Kidneys have limited ability to recover from injury. Various renal pathologies lead to local inflammation, which causes scarring and fibrosis of the kidney tissue. The immortalization of inflammatory stimuli drives inflammation and fibrosis of the tubular stroma and progressive impairment of renal function in chronic kidney disease. Its progression to end stage renal failure is associated with significant morbidity and mortality. Because tubular interstitial fibrosis is a common endpoint of a variety of renal pathologies, it represents a key therapeutic target aimed at preventing renal failure. Risk factors (e.g., proteinuria) that are independent of primary kidney disease promote the development of renal fibrosis and loss of renal excretion function by driving local inflammation, which in turn enhances disease progression.
In view of the role of fibrosis in many diseases and disorders, for example, tubular interstitial fibrosis leads to chronic kidney disease, there is an urgent need to develop therapeutically effective agents for treating diseases and conditions caused or exacerbated by fibrosis. Further in view of the lack of new and existing therapies targeting inflammatory pro-fibrotic pathways in renal disease, there is a need for therapeutically effective agents for developing treatments, inhibiting, preventing and/or reversing renal fibrosis, thereby preventing progressive chronic renal disease.
Disclosure of Invention
This summary is provided to introduce a selection of concepts in a simplified form that are further described below in the detailed description. This summary is not intended to identify key features of the claimed subject matter, nor is it intended to be used as an aid in determining the scope of the claimed subject matter.
In one aspect, the invention provides a method for treating, inhibiting, reducing or preventing fibrosis in a mammalian subject suffering from or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation, comprising administering to the subject an amount of a MASP-2 inhibitor effective to inhibit fibrosis. In one embodiment, the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6. In one embodiment, the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation. In one embodiment, the subject has a disease or condition caused or exacerbated by at least one of: (i) fibrosis and/or inflammation associated with ischemic reperfusion injury, (ii) renal fibrosis and/or nephritis (e.g., tubular interstitial fibrosis, chronic kidney disease, chronic renal failure, glomerular disease (e.g., focal segmental glomerulosclerosis), immune complex disorders (e.g., igA nephropathy, membranous nephropathy), lupus nephritis, nephrotic syndrome, diabetic nephropathy, tubular interstitial injury and glomerulonephritis (e.g., C3 glomerulonephropathy), (iii) pulmonary fibrosis and/or inflammation (e.g., chronic obstructive pulmonary disease, cystic fibrosis, scleroderma-associated pulmonary fibrosis, bronchiectasis and pulmonary arterial hypertension), (iv) liver fibrosis and/or inflammation (e.g., liver cirrhosis, nonalcoholic fatty liver disease (steatohepatitis)), liver fibrosis secondary to alcohol abuse, liver fibrosis secondary to acute or chronic hepatitis, biliary diseases and toxic liver injury (e.g., liver toxicity caused by drug-induced liver injury caused by acetaminophen or other drugs), (v) cardiac fibrosis and/or inflammation (e.g., cardiac fibrosis, myocardial infarction, valve fibrosis, atrial fibrosis, endocardial myocardial fibrosis, arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), (vi) vascular fibrosis (e.g., vascular disease, atherosclerotic vascular disease, vascular stenosis, restenosis, vasculitis, phlebitis, deep venous thrombosis and abdominal aortic aneurysm)), cardiac fibrosis, (vii) Skin fibrosis (e.g., excessive wound healing, scleroderma, systemic sclerosis, keloids, connective tissue disease, scarring and hypertrophic scarring), (viii) joint fibrosis (e.g., joint fibrosis), (ix) central nervous system fibrosis (e.g., stroke, traumatic brain injury and spinal cord injury), (x) digestive system fibrosis (e.g., crohn's disease, pancreatic fibrosis and ulcerative colitis), (xi) ocular fibrosis (e.g., subcapsular cataract, posterior capsular opacification, macular degeneration and retinal and vitreoretinopathy), (xii) musculoskeletal soft tissue structure fibrosis (e.g., adhesive capsulitis, dupudendum contracture and myelofibrosis), (xiii) genital organ fibrosis (e.g., endometriosis and pecies disease), (xiv) fibrosis and/or inflammatory chronic infectious diseases (e.g., alphavirus, hepatitis a, hepatitis b, hepatitis c, tuberculosis, HIV and influenza), (xv) autoimmune diseases (e.g., autoimmune diseases) causing fibrosis and/or inflammation (e.g., SLE, wherein the fibrosis and scar formation can be induced, wherein the surgical intervention (scar formation, and scar formation) can be selected from Radiation-induced fibrosis and scarring associated with burns), or (xvii) organ transplantation, breast fibrosis, muscle fibrosis, retroperitoneal fibrosis, thyroid fibrosis, lymph node fibrosis, bladder fibrosis and pleural fibrosis.
In another aspect, the invention provides a method for treating, inhibiting, reducing or preventing renal fibrosis in a mammalian subject suffering from or at risk of developing a disease or condition caused or exacerbated by renal fibrosis and/or inflammation, comprising administering to the subject an amount of a MASP-2 inhibitor effective to inhibit renal fibrosis. In one embodiment, the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6. In one embodiment, the MASP-2 antibody or fragment thereof specifically binds to a polypeptide comprising SEQ ID NO. 6 with at least 10-fold affinity for its binding to a different antigen of the complement system. In one embodiment, the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies and human antibodies. In one embodiment, the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation. In one embodiment, the MASP-2 inhibitor is administered subcutaneously, intraperitoneally, intramuscularly, intraarterially, intravenously, or as an inhalant. In one embodiment, the MASP-2 inhibitor is administered in an amount effective to inhibit tubulointerstitial fibrosis. In one embodiment, the MASP-2 inhibitor is administered in an amount effective to reduce, delay or eliminate the dialysis need in the subject. In one embodiment, the subject has a kidney disease or disorder selected from chronic kidney disease, chronic renal failure, glomerular disease (e.g., focal segmental glomerulosclerosis), immune complex disorders (e.g., igA nephropathy, membranous nephropathy), lupus nephritis, nephrotic syndrome, diabetic nephropathy, tubular interstitial injury, and glomerulonephritis (e.g., C3 glomerulopathy). In one embodiment, the subject has proteinuria and a MASP-2 inhibitor is administered in an amount effective to reduce the proteinuria in the subject. In one embodiment, the MASP-2 inhibitor is administered in an amount and for a time effective to achieve at least a 20% reduction (e.g., at least a 30% reduction or at least a 40% reduction or at least a 50% reduction) in urine protein secretion for 24 hours as compared to baseline 24 hours urine protein secretion for the subject prior to treatment. In one embodiment, the subject has a kidney disease or disorder associated with proteinuria selected from nephrotic syndrome, preeclampsia, eclampsia, kidney toxic impairment, amyloidosis, collagen vascular disease (e.g., systemic lupus erythematosus), dehydration, glomerular disease (e.g., membranous glomerulonephritis, focal segmental glomerulonephritis, C3 glomerulopathy, morbid disease, liponephrosis), intensive exercise, stress, benign orthotopic (posture) proteinuria, focal segmental glomerulosclerosis, igA nephropathy (i.e., begella disease), igM nephropathy, membranous glomerulonephritis, membranous nephropathy, morbid disease, sarcoidosis, alport syndrome, diabetes (diabetic nephropathy), drug-induced toxicity (e.g., NSAIDS, nicotine, penicillamine, lithium carbonate, gold and other heavy metals, ACE inhibitors, antibiotics (e.g., doxorubicin) or opiates (e.g., heroin) or other nephrotoxins); fabry's disease, infection (e.g., HIV, syphilis, hepatitis a, b or c, post streptococcal infection, schistosomiasis urinary); amino acid urine syndrome, van sconey syndrome, hypertensive nephrosclerosis, interstitial nephritis, sickle cell disease, hemoglobinuria, multiple myeloma, myoglobin urine, organ rejection (e.g., kidney transplant rejection), ebola hemorrhagic fever, patella nail syndrome, familial mediterranean fever, HELLP syndrome, systemic lupus erythematosus, wegener's granulomatosis, rheumatoid arthritis, glycogen storage disease type 1, goodpasture's syndrome, allergic purpura, urinary tract infections that have spread to the kidney, sjogren's syndrome, and post-infection glomerulonephritis. In one embodiment, the subject has IgA nephropathy. In one embodiment, the subject has membranous nephropathy.
In another aspect, the invention provides a method of preventing or reducing kidney damage in a subject having a proteinuria-related disease or condition, comprising administering an amount of a MASP-2 inhibitor effective to reduce or prevent proteinuria in the subject. In one embodiment, the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6. In one embodiment, the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation. In one embodiment, the disease or condition associated with proteinuria is selected from nephrotic syndrome, preeclampsia, eclampsia, toxic damage to the kidney, amyloidosis, collagen vascular disease (e.g., systemic lupus erythematosus), dehydration, glomerular disease (e.g., membranous glomerulonephritis, focal segmental glomerulonephritis, C3 glomerulopathy, minipathologic disease, liponephrosis), intensive exercise, stress, benign orthotopic (posture) proteinuria, focal segmental glomerulosclerosis, igA nephropathy (i.e., begelosis), nephropathy, igM, membranous proliferative glomerulonephritis, membranous nephropathy, minipathologic disease, sarcoidosis, alport syndrome, diabetes (diabetic nephropathy), drug-induced toxicity (e.g., NSAIDS, nicotine, penicillamine, lithium carbonate, gold and other heavy metals, ACE inhibitors, antibiotics (e.g., doxorubicin) or opiates (e.g., heroin)); fabry's disease, infection (e.g., HIV, syphilis, hepatitis a, b or c, post streptococcal infection, schistosomiasis urinary); amino acid urine syndrome, van sconey syndrome, hypertensive nephrosclerosis, interstitial nephritis, sickle cell disease, hemoglobinuria, multiple myeloma, myoglobin urine, organ rejection (e.g., kidney transplant rejection), ebola hemorrhagic fever, patella nail syndrome, familial mediterranean fever, HELLP syndrome, systemic lupus erythematosus, wegener's granulomatosis, rheumatoid arthritis, glycogen storage disease type 1, goodpasture's syndrome, allergic purpura, urinary tract infections that have spread to the kidney, sjogren's syndrome, and post-infection glomerulonephritis. In one embodiment, the MASP-2 inhibitor is administered in an amount and for a time effective to achieve at least a 20% reduction (e.g., at least a 30% reduction or at least a 40% reduction or at least a 50% reduction) in urine protein secretion for 24 hours as compared to baseline 24 hours urine protein secretion for the subject prior to treatment.
In another aspect, the invention provides a method of inhibiting the progression of a chronic kidney disease comprising administering an amount of a MASP-2 inhibitor effective to reduce or prevent renal fibrosis (e.g., tubular interstitial fibrosis) in a subject in need thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6. In one embodiment, the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation. In one embodiment, the subject in need thereof exhibits proteinuria prior to administration of the MASP-2 inhibitor, and administration of the MASP-2 inhibitor reduces proteinuria in the subject. In one embodiment, the MASP-2 inhibitor is administered in an amount and for a time effective to achieve at least a 20% reduction (e.g., at least a 30% reduction or at least a 40% reduction or at least a 50% reduction) in urine protein secretion for 24 hours as compared to baseline 24 hours urine protein secretion for the subject prior to treatment. In one embodiment, the MASP-2 inhibitor is administered in an amount effective to reduce, delay or eliminate the dialysis need in the subject.
In another aspect, the invention provides a method of protecting the kidney of a subject from kidney injury, the subject having undergone, being undergone or will undergo treatment with one or more nephrotoxic agents, comprising administering an amount of a MASP-2 inhibitor effective to prevent or ameliorate drug-induced kidney disease. In one embodiment, the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6. In one embodiment, the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation.
In another aspect, the invention provides a method of treating a human subject having immunoglobulin A kidney disease (IgAN), comprising administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody or antigen binding fragment thereof effective to inhibit MASP-2-dependent complement activation. In one embodiment, the subject has steroid dependent IgAN. In one embodiment, the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2. In one embodiment, the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies and human antibodies. In one embodiment, the MASP-2 inhibitory antibody does not significantly inhibit the classical pathway. In one embodiment, the MASP-2 inhibitory antibody is administered in an IC of 30nM or less 50 Inhibition of C3b deposition in 90% human serum. In one embodiment, the method further comprises identifying a human subject having steroid-dependent IgAN prior to the step of administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof effective to improve kidney function. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered to a pre-treatment subject at baseline 24An effective amount and for a sufficient time to achieve at least a 20% reduction in urine protein secretion at 24 hours compared to urine protein secretion at an hour. In one embodiment, the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence shown in SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
In another aspect, the invention provides a method of treating a human subject having Membranous Nephropathy (MN), comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody, or antigen-binding fragment thereof, effective to inhibit MASP-2-dependent complement activation. In one embodiment, the subject has a steroid dependent MN. In one embodiment, the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount and for a time effective to achieve at least a 20% reduction in 24 hours urine protein secretion as compared to the baseline 24 hours urine protein secretion of the subject prior to treatment. In one embodiment, the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence shown in SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
In another aspect, the invention provides a method of treating a human subject suffering from Lupus Nephritis (LN), comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody, or antigen-binding fragment thereof, effective to inhibit MASP-2-dependent complement activation. In one embodiment, the subject has steroid dependent LN. In one embodiment, the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount and for a time effective to achieve at least a 20% reduction in 24 hours urine protein secretion as compared to the baseline 24 hours urine protein secretion of the subject prior to treatment. In one embodiment, the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject. In one embodiment, the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence shown in SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
In another aspect, the invention provides a method of reducing proteinuria in a human subject suffering from IgAN, comprising administering to the subject a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence of SEQ ID NO 67 according to the following dosage regimen; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70:
a. intravenous administration of about 4mg/kg (i.e., 3.6mg/kg to 4.4 mg/kg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks; or (b)
b. Intravenous administration of about 180mg to about 725mg (i.e., 162mg to 797 mg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks,
wherein the method reduces proteinuria in the human subject.
Drawings
The foregoing aspects and many of the attendant advantages of this invention will become more readily appreciated as the same become better understood by reference to the following detailed description, when taken in conjunction with the accompanying drawings, wherein:
FIG. 1 is a diagram illustrating the structure of the human MASP-2 genome;
FIG. 2A is a schematic diagram illustrating the domain structure of human MASP-2 protein;
FIG. 2B is a schematic diagram illustrating the domain structure of human MAp19 protein;
FIG. 3 is a diagram illustrating a murine MASP-2 knockout strategy;
FIG. 4 is a diagram illustrating a human MASP-2 minigene construct;
FIG. 5A provides results demonstrating that MASP-2 deficiency results in loss of lectin pathway-mediated C4 activation, as determined by C4b deposition defects on mannans, as described in example 2;
FIG. 5B provides results demonstrating that MASP-2 deficiency results in loss of lectin pathway-mediated C4 activation, as determined by C4B deposition defects on zymosan, as described in example 2;
FIG. 5C provides results demonstrating the relative C4 activation levels of serum samples obtained from MASP-2+/-, MASP-2-/-and wild-type lines, as determined by C4b deposition on mannans and zymoglycans, as described in example 2;
FIG. 6 provides results demonstrating that the addition of murine recombinant MASP-2 to MASP-2-/-serum samples restored lectin pathway-mediated C4 activation in a protein concentration-dependent manner, as determined by C4b deposition on mannans, as described in example 2;
FIG. 7 provides results demonstrating the function of the classical pathway in MASP-2-/-lines, as described in example 8;
FIG. 8A provides results demonstrating that anti-MASP-2 Fab2 antibody #11 inhibits C3 convertase formation, as described in example 10; FIG. 8B provides results demonstrating the binding of anti-MASP-2 Fab2 antibody #11 to native rat MASP-2, as described in example 10;
FIG. 8C provides results demonstrating that anti-MASP-2 Fab2 antibody #41 inhibits C4 cleavage, as described in example 10;
FIG. 9 provides results demonstrating that all anti-MASP-2 Fab2 antibodies tested to inhibit C3 convertase formation were also found to inhibit C4 cleavage, as described in example 10;
FIG. 10 is a schematic diagram illustrating recombinant polypeptides derived from rat MASP-2 for MASP-2 blocking Fab2 antibody epitope mapping, as described in example 11;
FIG. 11 provides results demonstrating the binding of anti-MASP-2 Fab2#40 and #60 to rat MASP-2 polypeptides, as described in example 11;
FIG. 12A is a graph showing the level of MAC deposition in the presence or absence of human MASP-2 monoclonal antibody (OMS 646) under lectin pathway specific assay conditions, confirming that OMS646 was present with an IC of about 1nM 50 Values inhibit lectin-mediated MAC deposition, as described in example 12;
FIG. 12B illustrates the level of MAC deposition in the presence or absence of human MASP-2 monoclonal antibody (OMS 646) under classical pathway-specific assay conditions, confirming that OMS646 does not inhibit classical pathway-mediated MAC deposition, as described in example 12;
FIG. 12C illustrates the level of MAC deposition in the presence or absence of human MASP-2 monoclonal antibody (OMS 646) under alternative pathway-specific assay conditions, confirming that OMS646 does not inhibit alternative pathway-mediated MAC deposition, as described in example 12;
FIG. 13 illustrates Pharmacokinetic (PK) profiles of human MASP-2 monoclonal antibody (OMS 646) in mice, showing OMS646 concentration (n=3 animals/group mean) as a function of time following administration at the indicated dose, as described in example 12;
FIG. 14A illustrates the Pharmacodynamic (PD) response of human MASP-2 monoclonal antibody (OMS 646) in mice after intravenous administration as measured by a decrease in systemic lectin pathway activity, as described in example 12;
FIG. 14B illustrates the Pharmacodynamic (PD) response of human MASP-2 monoclonal antibody (OMS 646) in mice following subcutaneous administration as measured by a decrease in systemic lectin pathway activity, as described in example 12;
FIG. 15 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, wherein the tissue sections were obtained from wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), and sham-operated wild-type and MASP-2-/-mice, as described in example 14;
FIG. 16 illustrates the results of computer-based image analysis of kidney tissue sections stained with F4/80 macrophage specific antibody, wherein the tissue sections were obtained from wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), and sham operated wild-type and MASP-2-/-mice, as described in example 14.
FIG. 17 illustrates relative mRNA expression levels of collagen-4 as measured by quantitative PCR (qPCR) in kidney tissue sections of wild-type and MASP-2-/-mice and sham operated wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), as described in example 14.
FIG. 18 illustrates the relative mRNA expression levels of transforming growth factor beta-1 (TGF beta-1) as measured by qPCR in kidney tissue sections of wild-type and MASP-2-/-mice and sham operated wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), as described in example 14.
FIG. 19 illustrates relative mRNA expression levels of interleukin-6 (IL-6) measured by qPCR in kidney tissue sections of wild-type and MASP-2-/-mice and sham operated wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), as described in example 14.
FIG. 20 illustrates relative mRNA expression levels of interferon-gamma measured by qPCR in kidney tissue sections of wild-type and MASP-2-/-mice and sham-operated wild-type and MASP-2-/-mice 7 days after unilateral ureteral obstruction (UUFO), as described in example 14.
Fig. 21 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, wherein tissue sections were obtained from wild-type mice treated with MASP-2 inhibitory antibodies and isotype control antibodies 7 days after Unilateral Ureteral Obstruction (UUO), as described in example 15.
Figure 22 illustrates hydroxyproline content of kidneys harvested 7 days after Unilateral Ureteral Obstruction (UUO) from wild-type mice treated with MASP-2 inhibitory antibodies compared to the level of hydroxyproline in the tissue of obstructed kidneys from wild-type mice treated with IgG4 isotype control, as described in example 15.
Figure 23 illustrates the total amount of serum protein (mg/ml) measured on day 15 of the protein overload study in wild-type control mice (n=2) receiving saline only, wild-type mice (n=6) receiving BSA, and MASP-2-/-mice (n=6) receiving BSA, as described in example 16.
Figure 24 illustrates total secreted protein (mg) in urine collected over 24 hours on day 15 of protein overload study from wild type control mice (n=2) receiving only saline, wild type (n=6) receiving BSA, and MASP-2-/-mice (n=6) receiving BSA, as described in example 16.
Fig. 25 shows representative hematoxylin and eosin (H & E) -stained kidney tissue sections from the following groups of mice on day 15 of the protein overload study: (panel a) wild-type control mice; (Panel B) MASP-2-/-control mice; (Panel C) wild-type mice treated with BSA; and (panel D) MASP-2-/-mice treated with Bovine Serum Albumin (BSA) as described in example 16.
Fig. 26 illustrates the results of computer-based image analysis of kidney tissue sections stained with macrophage specific antibody F4/80, showing the mean area of macrophage staining (%), wherein tissue sections were obtained from wild-type control mice (n=2), BSA-treated wild-type mice (n=6), and BSA-treated MASP-2-/-mice (n=5) on day 15 of the protein overload study, as described in example 16.
Fig. 27A illustrates the presence analysis of macrophage-proteinuria correlation in each wild-type mouse treated with BSA (n=6) by plotting total secreted protein measured in urine from 24 hours samples versus macrophage infiltration (average stained area%) as described in example 16.
Figure 27B illustrates the presence analysis of macrophage-proteinuria correlation in each MASP-2-/-mouse treated with BSA (n=5) by plotting total secreted protein in urine versus macrophage infiltration (average stained area%) in 24 hour samples, as described in example 16.
Fig. 28 illustrates the results of computer-based image analysis of tissue sections stained with anti-tgfβ antibodies (as% tgfβ antibody staining area measurements) in wild-type mice treated with BSA (n=4) and MASP-2-/-mice treated with BSA (n=5), as described in example 16.
Fig. 29 illustrates the results of computer-based image analysis of tissue sections stained with anti-tnfα antibodies (measured as% tnfα antibody staining area) in wild type mice treated with BSA (n=4) and MASP-2-/-mice treated with BSA (n=5), as described in example 16.
FIG. 30 graphically depicts the results of computer-based image analysis (measured as% IL-6 antibody staining area) of tissue sections stained with anti-IL-6 antibody in wild type control mice, MASP-2-/-control mice, wild type mice treated with BSA (n=7), and MASP-2-/-mice treated with BSA (n=7), as described in example 16.
Fig. 31 illustrates the frequency of TUNEL apoptotic cells counted in 20 High Power Fields (HPF) from sequential selection of tissue sections of kidney cortex of wild type control mice (n=1), MASP-2-/-control mice (n=1), wild type mice treated with BSA (n=6) and MASP-2-/-mice treated with BSA (n=7), as described in example 16.
Fig. 32 shows representative H & E stained tissue sections from each of the following groups of mice on day 15 post treatment with BSA: (Panel A) wild-type control mice treated with saline, (Panel B) isotype antibody-treated control mice and (Panel C) wild-type mice treated with MASP-2 inhibitory antibodies as described in example 17.
Fig. 33 illustrates the frequency of TUNEL apoptotic cells counted in 20 High Power Fields (HPF) from serial selection of tissue sections of kidney cortex from wild type mice treated with saline control and BSA (n=8), wild type mice treated with isotype control antibody and BSA (n=8) and wild type mice treated with MASP-2 inhibitory antibody and BSA (n=7), as described in example 17.
Fig. 34 illustrates the results of computer-based image analysis of tissue sections stained with anti-tgfβ antibodies (measured as% tgfβ antibody staining area) in wild-type mice treated with BSA and saline (n=8), wild-type mice treated with BSA and isotype control antibodies (n=7), and wild-type mice treated with BSA and MASP-2 inhibitory antibodies (n=8), as described in example 17.
Fig. 35 illustrates the results of computer-based image analysis of tissue sections stained with anti-tnfα antibodies (measured as% tnfα antibody staining area) in wild type mice treated with BSA and saline (n=8), BSA and isotype control antibodies (n=7), and wild type mice treated with BSA and MASP-2 inhibitory antibodies (n=8), as described in example 17.
Fig. 36 illustrates the results of computer-based image analysis of tissue sections stained with anti-IL-6 antibody (measured as% IL-6 antibody staining area) in wild-type mice treated with BSA and saline (n=8), BSA and isotype control antibodies (n=7), and wild-type mice treated with BSA and MASP-2 inhibitory antibodies (n=8), as described in example 17.
Fig. 37 shows representative H & E stained tissue sections from each of the following groups of mice on day 14 after treatment with doxorubicin or saline only (control): (panels A-1, A-2, A-3) wild-type control mice treated with saline only; (panels B-1, B-2, B-3) wild-type mice treated with doxorubicin; and (FIGS. C-1, C-2, C-3) MASP-2-/-mice treated with doxorubicin, as described in example 18.
Fig. 38 illustrates the results of computer-based image analysis of kidney tissue sections stained with macrophage specific antibody F4/80, showing the average area of macrophage staining (%) from each of the following groups of mice on day 14 after treatment with doxorubicin or saline only (wild type control): wild-type control mice treated with saline only; wild-type mice treated with doxorubicin; MASP-2-/-mice treated with saline only and MASP-2-/-mice treated with doxorubicin, wherein p=0.007, as described in example 18.
Fig. 39 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, showing collagen deposition staining areas (%) from each of the following groups of mice on day 14 after treatment with doxorubicin or saline only (wild type control): wild-type control mice treated with saline only; wild-type mice treated with doxorubicin; MASP-2-/-mice treated with saline only and MASP-2-/-mice treated with doxorubicin, wherein p=0.005, as described in example 18.
FIG. 40 illustrates urinary albumin/creatinine ratio (uACR) in two IgA patients during the course of a 12 week study with MASP-2 inhibitory antibodies (OMS 646) weekly, as described in example 19.
FIG. 41 illustrates uACR (mg/g) over time for 4 IgAN patients treated with OMS646 from baseline to 120 days, as described in example 21.
Fig. 42 illustrates the change in urine protein from baseline on day 1 prior to treatment to 24 hours post treatment for 4 IgAN patients treated with OMS646, as described in example 21.
Fig. 43 illustrates the mean change in urine protein from baseline to 24 hours post-treatment for 4 IgAN patients treated with OMS646, as described in example 21.
FIG. 44 shows a flow chart of the study design of example 19.
FIG. 45 shows a flow chart of the study design of example 20.
Description of sequence Listing
SEQ ID NO. 1 human MAp19 cDNA
SEQ ID NO. 2 human MAp19 protein (with leader sequence)
SEQ ID NO. 3 human MAp19 protein (mature)
SEQ ID NO. 4 human MASP-2cDNA
SEQ ID NO. 5 human MASP-2 protein (with leader sequence)
SEQ ID NO. 6 human MASP-2 protein (mature)
SEQ ID NO. 7 human MASP-2gDNA (exons 1-6)
Antigen: (for MASP-2 mature protein)
SEQ ID NO. 8CUBI sequence (amino acids 1-121)
SEQ ID NO. 9CUBEGF sequence (amino acids 1-166)
SEQ ID NO. 10CUBEGFCUBII (amino acids 1-293)
SEQ ID NO. 11EGF region (amino acids 122-166)
SEQ ID NO. 12 serine protease domain (amino acids 429-671)
SEQ ID NO. 13 inactivated serine protease domain (amino acids 610-625, with Ser618 to Ala mutation) SEQ ID NO. 14TPLGPKWPEPVFGRL (CUBI peptide)
SEQ ID NO. 15TAPPGYRLRLYFTHFDLELSHLCEYDFVKLSSGAKVLATLCGQ (CUBI peptide) SEQ ID NO. 16TFRSDYSN (MBL binding region core)
SEQ ID NO. 17FYSLGSSLDITFRSDYSNEKPFTGF (MBL binding region)
SEQ ID NO. 18IDECQVAPG (EGF peptide)
Detailed description of SEQ ID NO. 19ANMLCAGLESGGKDSCRGDSGGALV (serine protease binding core)
Peptide inhibitors:
SEQ ID NO. 20MBL full-length cDNA
21MBL full-length protein of SEQ ID NO
SEQ ID NO. 22OGK-X-GP (consensus binding)
SEQ ID NO:23OGKLG
SEQ ID NO:24GLR GLQ GPO GKLGPO G
SEQ ID NO:25GPO GPO GLR GLQ GPO GKL GPO GPO GPO
SEQ ID NO:26GKDGRDGTKGEKGEPGQGLRGLQGPOGKLGPOG
SEQ ID NO 27 GAOGSOGEKGAOGPQGPOGGKMGPKGEOGDO (human h-fiber gel protein) SEQ ID NO 28 GCOGGOGGAGGEAGTNGKRREGGPOGGKAGPOGGAGEO (human fiber gel protein p 35)
SEQ ID NO. 29LQRALEILPNRVTIKANRPFLVFI (C4 cleavage site)
Expression inhibitor:
cDNA of 30CUBI-EGF domain (nucleotides 22-680 of SEQ ID NO: 4)
SEQ ID NO:31
5'CGGGCACACCATGAGGCTGCTGACCCTCCTGGGC 3'
Nucleotides 12-45 of SEQ ID NO. 4, including the MASP-2 translation initiation site (sense strand)
SEQ ID NO:32
5'GACATTACCTTCCGCTCCGACTCCAACGAGAAG3'
Nucleotides 361-396 (sense strand) of SEQ ID NO. 4 encoding a region containing the MASP-2MBL binding site
SEQ ID NO:33
5'AGCAGCCCTGAATACCCACGGCCGTATCCCAAA3'
Nucleotides 610 to 642 of SEQ ID NO. 4 encoding the region comprising the CUBII domain
Cloning a primer:
SEQ ID NO. 34CGGGATCCATGAGGCTGCTGACCCTC (5' PCR for CUB)
SEQ ID NO. 35GGAATTCCTAGGCTGCATA (3' PCR for CUB)
SEQ ID NO. 36GGAATTCCTACAGGGCGCT (3' PCR for CUBIEGF)
SEQ ID NO. 37GGAATTCCTAGTAGTGGAT (3' PCR for CUBIEGFCUBII)
SEQ ID NOS 38-47 is a cloning primer for a humanized antibody
SEQ ID NO. 48 is a 9 amino acid peptide bond
Expression vector:
SEQ ID NO. 49 is a MASP-2 minigene insert
SEQ ID NO. 50 is murine MASP-2cDNA
SEQ ID NO. 51 is a murine MASP-2 protein (w/leader sequence)
SEQ ID NO. 52 is a mature murine MASP-2 protein
SEQ ID NO. 53 is rat MASP-2cDNA
SEQ ID NO. 54 is the rat MASP-2 protein (w/leader)
SEQ ID NO. 55 is a mature rat MASP-2 protein
SEQ ID NOS.56-59 are oligonucleotides for site-directed mutagenesis of human MASP-2 for producing human MASP-2A
SEQ ID NO. 60-63 is an oligonucleotide for site-directed mutagenesis of murine MASP-2, which murine MASP-2 is used to produce murine MASP-2A
SEQ ID NO. 64-65 is an oligonucleotide for site-directed mutagenesis of rat MASP-2 for the production of rat MASP-2A
DNA SEQ ID NO. 66 encoding a 17D20_dc35VH21N11VL (OMS 646) heavy chain variable region (VH) (NO signal peptide) SEQ ID NO. 6717D20_dc35VH21N11VL (OMS 646) heavy chain variable region (VH) polypeptide
SEQ ID NO. 6817N16mc heavy chain variable region (VH) polypeptide
DNA of SEQ ID NO. 69 encoding 17D20_dc35VH21N11VL (OMS 646) light chain variable region (VL)
The light chain variable region (VL) polypeptide of SEQ ID NO. 7017D20_dc35VH21N11VL (OMS 646)
SEQ ID NO. 7117N16_dc17N9 light chain variable region (VL) polypeptide
SGMI-2L (full length) of SEQ ID NO 72
SEQ ID NO. 73:SGMI-2M (medium truncated form)
SEQ ID NO. 74:SGMI-2S (short truncated form)
SEQ ID NO. 75 mature polypeptide comprising VH-M2ab6-SGMI-2-N and human IgG4 constant region with hinge mutation
SEQ ID NO. 76 mature polypeptide comprising a VH-M2ab6-SGMI-2-C and a human IgG4 constant region with hinge mutation
SEQ ID NO. 77 mature polypeptide comprising VL-M2ab6-SGMI-2-N and human Ig lambda constant region
SEQ ID NO. 78 mature polypeptide comprising VL-M2ab6-SGMI-2-C and human Ig lambda constant region
SEQ ID NO. 79 peptide linker (10 aa)
SEQ ID NO. 80. Peptide linker (6 aa)
SEQ ID NO. 81 peptide linker (4 aa)
SEQ ID NO. 82A polynucleotide encoding a polypeptide having a hinge mutation comprising a VH-M2ab6-SGMI-2-N and a human IgG4 constant region
SEQ ID NO. 83 Polynucleotide encoding a polypeptide having a hinge mutation comprising a VH-M2ab6-SGMI-2-C and a human IgG4 constant region
SEQ ID NO. 84A polynucleotide encoding a polypeptide comprising a VL-M2ab6-SGMI-2-N and a human Ig lambda constant region
SEQ ID NO. 85 Polynucleotide encoding a polypeptide comprising a VL-M2ab6-SGMI-2-C and a human Ig lambda constant region
Detailed Description
The present invention is based on the surprising discovery by the inventors that lectin-associated serine protease-2 (MASP-2), a key regulator of the lectin pathway of the complement system, inhibits mannan binding significantly reduces inflammation and fibrosis in various animal models of fibrotic disease, including Unilateral Ureteral Obstruction (UUO) models of renal fibrosis, protein overload models, and doxorubicin-induced renal pathology models. Thus, the inventors have demonstrated that inhibition of MASP-2 mediated lectin pathway activation provides an effective therapeutic approach to improve, treat or prevent renal fibrosis, such as tubular interstitial inflammation and fibrosis, regardless of the root cause. As further described herein, the use of MASP-2 inhibitory antibodies (OMS 646) is effective in improving kidney function and reducing corticosteroid requirements in human subjects with immunoglobulin a kidney disease (IgAN) and membranous kidney disease (MN).
I. Definition of the definition
Unless defined otherwise herein, all terms used herein have the same meaning as understood by one of ordinary skill in the art of the present invention. To clarify the terminology used in this description and the appended claims in connection with the description of the invention, the following definitions are provided.
As used herein, the term "MASP-2" is used in accordance withDependent complement activation "includes MASP-2-dependent activation of the lectin pathway, which occurs under physiological conditions (i.e., at Ca ++ Where present), leading to the formation of the lectin pathway C3 convertase C4b2a and, after aggregation of the C3 cleavage product C3b, the formation of the subsequent C5 convertase C4b2a (C3 b) n, which C5 convertase C4b2a (C3 b) n has been determined to cause mainly opsonization.
The term "alternative pathway" as used herein refers to complement activation triggered by, for example, zymosan from fungal and yeast cell walls, lipopolysaccharide (LPS) and rabbit erythrocytes of gram-negative outer membranes, and from a variety of pure polysaccharides, rabbit erythrocytes, viruses, bacteria, animal tumor cells, parasites and damaged cells, which have traditionally been thought to be caused by spontaneous hydrolysis of complement factor C3 to produce C3 b.
The term "lectin pathway" as used herein refers to complement activation by specific binding of serum and non-serum carbohydrate binding proteins, including mannan-binding lectin (MBL), CL-11 and fiber-gelling proteins (H-fiber-gelling protein, M-fiber-gelling protein or L-fiber-gelling protein).
The term "classical pathway" as used herein refers to complement activation triggered by binding of an antibody to an exogenous particle and requiring binding to the recognition molecule C1 q.
As used herein, the term "MASP-2 inhibitor" refers to any agent that binds to MASP-2 or interacts directly with MASP-2 and is effective in inhibiting MASP-2 dependent complement activation, including anti-MASP-2 antibodies and MASP-2 binding fragments thereof, natural and synthetic peptides, small molecules, soluble MASP-2 receptors, expression inhibitors and isolated natural inhibitors, and also includes peptides that compete in the lectin pathway for binding to other recognition molecules (e.g., MBL, H-fiber-gelling protein, M-fiber-gelling protein or L-fiber-gelling protein), but does not include antibodies that bind to these other recognition molecules. MASP-2 inhibitors useful in the methods of the invention may reduce MASP-2 dependent complement activation by greater than 20%, such as greater than 50%, such as greater than 90%. In one embodiment, the MASP-2 inhibitor reduces MASP-2 dependent complement activation by greater than 90% (i.e., results in only 10% or less of MASP-2 complement activation).
The term "fibrosis" as used herein refers to the formation or presence of excessive connective tissue in an organ or tissue. Fibrosis may occur as a repair or replacement response to stimuli such as tissue injury or inflammation. The hallmark of fibrosis is the production of excessive extracellular matrix. Normal physiological responses to injury result in connective tissue deposition as part of the healing process, but such connective tissue deposition can persist and become pathological, altering the structure and function of the tissue. At the cellular level, epithelial cells and fibroblasts proliferate and differentiate into myofibroblasts, resulting in matrix contraction, increased rigidity, microvascular compression, and hypoxia.
The term "treating fibrosis in a mammalian subject suffering from or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation" as used herein refers to reversing, reducing, ameliorating or inhibiting fibrosis in the mammalian subject.
The term "proteinuria" as used herein refers to the presence of abnormal amounts of urine protein, for example, amounts of more than 0.3g protein in 24 hours urine collected from a human subject, or concentrations of more than 1 g/liter in a human subject. In some embodiments, a subject with proteinuria refers to the presence of urine proteins in an amount of greater than 1.0g protein in 24 hours urine collected from a human subject, such as a subject with immunoglobulin a (IgA) kidney disease.
The term "ameliorating proteinuria" or "reducing proteinuria" as used herein refers to reducing 24-hour urinary protein secretion by at least 20%, such as at least 30%, such as at least 40%, such as at least 50% or more in a subject suffering from proteinuria as compared to the baseline 24-hour urinary protein secretion in a subject prior to treatment with a MASP-2 inhibitor. In one embodiment, treatment with a MASP-2 inhibitor according to the methods of the invention is effective to reduce proteinuria in a human subject, e.g., to achieve a more than 20% reduction in urine protein secretion at 24 hours, or e.g., a more than 30% reduction in urine protein secretion at 24 hours, or e.g., a more than 40% reduction in urine protein secretion at 24 hours, or e.g., a more than 50% reduction in urine protein secretion at 24 hours.
The term "antibody" as used herein includes antibodies and antibody fragments thereof derived from any mammal (e.g., mice, rats, rabbits, and primates including humans) that produces antibodies, or derived from hybridomas, phage selection, recombinant expression, or transgenic animals (or other methods of producing antibodies or antibody fragments) and which specifically bind to a polypeptide of interest such as, for example, a MASP-2 polypeptide or portion thereof. The term "antibody" is not intended to be limiting with respect to the source of the antibody or the manner in which it is prepared (e.g., by hybridoma, phage selection, recombinant expression, transgenic animals, peptide synthesis, etc.). Exemplary antibodies include polyclonal antibodies, monoclonal antibodies, and recombinant antibodies; full-specific, multispecific antibodies (e.g., bispecific, trispecific antibodies); a humanized antibody; a murine antibody; chimeric, mouse-human, mouse-primate, primate-human monoclonal antibodies; and anti-idiotype antibodies, and may be any intact antibody or fragment thereof. The term "antibody" as used herein includes not only whole polyclonal or monoclonal antibodies, but also fragments thereof (such as dAb, fab, fab ', F (ab') 2 Fv), single chain (ScFv), synthetic variants thereof, naturally occurring variants, fusion proteins comprising an antibody moiety and an antigen-binding fragment of a desired specificity, humanized antibodies, chimeric antibodies, and any other modified configuration of an immunoglobulin molecule comprising an antigen-binding site or fragment (epitope recognition site) of a desired specificity.
"monoclonal antibody" refers to a homogeneous population of antibodies, wherein the monoclonal antibodies are composed of amino acids (naturally occurring amino acids and non-naturally occurring amino acids) that are involved in selectively binding to an epitope. Monoclonal antibodies are highly specific for the antigen of interest. The term "monoclonal antibody" includes not only whole monoclonal antibodies and full length monoclonal antibodies, but also fragments thereof (such as Fab, fab ', F (ab') 2 Fv), single chain (ScFv), variants thereof, fusion proteins comprising an antigen-binding portion, humanized monoclonal antibodies, chimeric monoclonal antibodies, and any immunoglobulin molecule comprising an antigen-binding fragment (epitope recognition site) having the desired specificity and binding capacity to an epitopeOther modified configurations. It is not intended to be limiting as to the source of the antibody or the manner in which it is prepared (e.g., by hybridoma, phage selection, recombinant expression, transgenic animals, etc.). The term includes the whole immunoglobulins and fragments etc. described above under the definition of "antibodies".
The term "antibody fragment" as used herein refers to a portion derived from or related to a full length antibody, such as, for example, an anti-MASP-2 antibody, generally comprising an antigen binding region or variable region thereof. Illustrative examples of antibody fragments include Fab, fab', F (ab) 2 、F(ab') 2 And Fv fragments, scFv fragments, diabodies, linear antibodies, single chain antibody molecules, and multispecific antibodies formed from antibody fragments.
As used herein, a "single chain Fv" or "scFv" antibody fragment includes V of an antibody H And V L Domains, wherein these domains are present on a single polypeptide chain. Fv polypeptides generally also include V H And V is equal to L Polypeptide linkers between domains, which enable the scFv to form the desired antigen binding structure.
As used herein, a "chimeric antibody" is a recombinant protein containing variable domains and complementarity determining regions derived from antibodies of a non-human species (e.g., rodent), while the remainder of the antibody molecule is derived from a human antibody.
As used herein, a "humanized antibody" is a chimeric antibody that comprises minimal sequences that conform to specific complementarity determining regions derived from a non-human immunoglobulin, which are implanted into a human antibody framework. Humanized antibodies are typically recombinant proteins in which only the complementarity determining regions of the antibody are of non-human origin.
The term "mannan-binding lectin" ("MBL") as used herein is equivalent to a mannan-binding protein ("MBP").
As used herein, "membrane attack complex" ("MAC") refers to a complex (also referred to as C5 b-9) that intercalates and disrupts the 5 terminal complement components of the membrane (C5 b combining C6, C7, C8 and C-9).
As used herein, "subject" includes all mammals including, but not limited to, humans, non-human primates, dogs, cats, horses, sheep, goats, cows, rabbits, pigs, and rodents.
The abbreviations for amino acid residues used herein are as follows: alanine (Ala; A), asparagine (Asn; N), aspartic acid (Asp; D), arginine (Arg; R), cysteine (Cys; C), glutamic acid (Glu; E), glutamine (Gln; Q), glycine (Gly; G), histidine (His; H), isoleucine (Ile; I), leucine (Leu; L), lysine (Lys; K), methionine (Met; M), phenylalanine (Phe; F), proline (Pro; P), serine (Ser; S), threonine (Thr; T), tryptophan (Trp; W), tyrosine (Tyr; Y) and alanine (Val; V).
Naturally occurring amino acids can be grouped according to the chemical nature of the individual amino acid side chains in the broadest sense. "hydrophobic" amino acid refers to Ile, leu, met, phe, trp, tyr, val, ala, cys or Pro. "hydrophilic" amino acid refers to Gly, asn, gln, ser, thr, asp, glu, lys, arg or His. Such groupings of amino acids may be further subdivided as follows. "uncharged hydrophilic" amino acids refer to Ser, thr, asn or Gln. "acidic" amino acid refers to Glu or Asp. "basic" amino acid refers to Lys, arg or His.
The term "conservative amino acid substitutions" as used herein is illustrated by substitutions between amino acids in each of the following groups: (1) Glycine, alanine, valine, leucine and isoleucine; (2) phenylalanine, tyrosine, and tryptophan; (3) serine and threonine; (4) aspartic acid and glutamic acid; (5) glutamine and asparagine; and (6) lysine, arginine, and histidine.
The term "oligonucleotide" as used herein refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or a mimetic thereof. The term also encompasses those oligonucleobases consisting of naturally occurring nucleotides, sugars and internucleoside (backbone) covalent bonds and oligonucleotides with non-naturally occurring modifications.
As used herein, an "epitope" refers to a site on a protein (e.g., human MASP-2 protein) that is bound by an antibody. "overlapping epitopes" include at least one (e.g., two, three, four, five, or six) common amino acid residue, including linear and non-linear epitopes.
The terms "polypeptide," "peptide," and "protein" are used interchangeably herein and refer to any peptide-linked chain of amino acids, whether length or post-translational modification. The MASP-2 proteins described herein may contain or may be wild-type proteins, or may be variants having no more than 50 (e.g., no more than one, two, three, four, five, six, seven, eight, nine, ten, 12, 15, 20, 25, 30, 35, 40, or 50) conservative amino acid substitutions. Conservative substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine and threonine; lysine, histidine and arginine; phenylalanine and tyrosine.
In some embodiments, the amino acid sequence of the human MASP-2 protein may be or greater than 70 (e.g., 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100)% identical to the human MASP-2 protein having the amino acid sequence set forth in SEQ ID NO. 5.
In some embodiments, the peptide fragment may be at least 6 (e.g., at least 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, or 600 or more) amino acid residues in length (e.g., at least 6 consecutive amino acid residues of SEQ ID NO: 5). In some embodiments, the antigenic peptide fragment of a human MASP-2 protein is less than 500 (e.g., less than 450, 400, 350, 325, 300, 275, 250, 225, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, or 6) amino acid residues in length (e.g., less than 500 consecutive amino acid residues in any of SEQ ID NOS: 5).
Percent (%) amino acid sequence identity is defined as the percentage of amino acids identical to the amino acids in the reference sequence that, after alignment and introduction of gaps, in the candidate sequence, if desired, achieve the greatest percent sequence identity. Alignment for the purpose of determining percent sequence identity may be accomplished in a variety of ways within the skill of the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN-2, or Megalign (DNASTAR) software. Suitable parameters for measuring the alignment, including any algorithms required to achieve maximum alignment over the entire length of the sequences to be compared, can be determined by known methods.
The term "about" or "approximately" as used herein when preceded by a numerical value means that the value is plus or minus a range of 10%.
Overview of the invention
As described herein, the inventors have identified a central role for the lectin pathway in initiation of renal tubular pathology and in disease progression, suggesting a critical role for lectin pathway activation in the pathophysiology of various renal diseases including IgA nephropathy, C3 glomerulopathy and other glomerulonephritis (glomeronephritide). As further described herein, the inventors found that lectin-associated serine protease-2 (MASP-2), an important regulator of the lectin pathway of the complement system, inhibited mannan binding significantly reduced inflammation and fibrosis in various animal models of fibrotic disease, including Unilateral Ureteral Obstruction (UUO) models of renal fibrosis, protein overload models, and doxorubicin-induced nephrology models. Thus, the inventors have demonstrated that inhibition of MASP-2 mediated lectin pathway activation provides an effective therapeutic approach to improve, treat or prevent renal fibrosis, e.g., tubular interstitial fibrosis, regardless of the root cause.
Lectins (MBP, M-fiber-gel protein, H-fiber-gel protein and L-fiber-gel protein and CL-11) are specific recognition molecules that trigger the congenital complement system comprising the lectin-initiating pathway and the associated terminal pathway amplification loop that amplifies lectin-initiated activation of terminal complement effector molecules. C1q is a specific recognition molecule that triggers the acquired complement system, which includes the classical initiation pathway and the associated end pathway amplification loop that amplifies activation of the C1q initiation of the end complement effector molecule. We refer to these two major complement activation systems as the lectin-dependent complement system and the C1 q-dependent complement system, respectively.
In addition to its fundamental role in immune defense, the complement system is responsible for tissue damage in many clinical diseases. Thus, there is an urgent need to develop therapeutically effective complement inhibitors to inhibit these side effects. If it were recognized that it might inhibit the lectin-mediated MASP-2 pathway leaving the classical pathway intact, it would be appreciated that what we would be highly desirable would be to specifically inhibit only the complement activation system that causes a particular pathology without completely halting the immune defenses of complement. For example, in disease states in which complement activation is mediated primarily by the lectin-dependent complement system, it would be advantageous to specifically inhibit only this system. This will preserve the integrity of the C1 q-dependent complement activation system to handle immune complex processing and aid in host defense against infection.
In the development of therapeutic agents that specifically inhibit the lectin-dependent complement system, the preferred protein component as a target is MASP-2. Of all known protein components of the lectin-dependent complement system (MBL, H-fiber gelonin, M-fiber gelonin, L-fiber gelonin, MASP-2, C2-C9, factor B, factor D and properdin), only MASP-2 is unique to the lectin-dependent complement system and is essential for this system to function. Lectins (MBL, H-fiber gelator, M-fiber gelator, L-fiber gelator and CL-11) are also unique components of the lectin-dependent complement system. However, loss of any of these lectin components does not necessarily inhibit system activation due to lectin redundancy. To ensure inhibition of the lectin-dependent complement activation system, inhibition of all 5 lectins may be necessary. Furthermore, since MBL and fiber-gelling proteins are also known to have complement-independent opsonic activity, inhibition of lectin function will result in the loss of this favorable host defense mechanism against infection. In contrast, if MASP-2 is the inhibitory target, this complement-independent lectin opsonic activity will remain intact. An additional benefit of MASP-2 as a therapeutic target for inhibition of lectin-dependent complement activation systems is that the plasma concentration of MASP-2 is the lowest of all complement proteins (about 500 ng/ml); thus, a relatively low concentration of high affinity MASP-2 inhibitor may be sufficient for complete inhibition (Moller-Kristensen, M., et al, J.Immunol Methods 282:159-167,2003).
As described in example 14 herein, mice without the MASP-2 gene (MASP-2-/-) showed significantly less kidney disease as compared to wild-type control animals as shown by inflammatory cell infiltration (75% reduction) and histological markers of fibrosis such as collagen deposition (1/3 reduction) were determined in animal models of fibrotic kidney disease (unilateral ureteral obstruction UUO). As further shown in example 15, wild-type mice treated systemically with anti-MASP-2 monoclonal antibodies that selectively block the lectin pathway while retaining the classical pathway intact were protected from renal fibrosis compared to wild-type mice treated with isotype control antibodies. These results demonstrate that the lectin pathway is a key contributor to kidney disease and further demonstrate that inhibitors of MASP-2, such as MASP-2 antibodies, that block the lectin pathway are effective as anti-fibrotic agents. As further shown in example 16, wild-type mice treated with Bovine Serum Albumin (BSA) developed proteinuria nephropathy in a protein overload model, while MASP-2-/-mice treated with the same level of BSA had reduced kidney injury. Wild-type mice that were treated systemically with anti-MASP-2 monoclonal antibodies that selectively blocked the lectin pathway while retaining the classical pathway intact were protected from kidney injury in a protein overload model, as shown in example 17. As described in example 18, MASP-2-/-mice showed less renal inflammation and tubulointerstitial damage in the doxorubicin-induced renal fibrosis nephrology model compared to wild-type mice. As described in example 19, in the phase 2 open-labeled kidney trial performed, patients with IgA nephropathy treated with anti-MASP-2 antibodies showed a clinically significant and statistically significant decrease in urine albumin to creatinine ratio (uACR) and 24 hour decrease in urine protein levels from baseline to end of treatment throughout the trial. As further described in example 19, patients with membranous nephropathy treated with anti-MASP-2 antibodies also showed a decrease in uACR during treatment in the same phase 2 kidney trial. As described in example 20, 4 of the 5 patients with Lupus Nephritis (LN) treated with anti-MASP-2 antibodies demonstrated clinically significant reductions in urine protein levels from baseline to 24 hours at the end of treatment in the 2-phase open-label kidney trial performed.
In accordance with the foregoing, the present invention relates to the use of a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody, as an anti-fibrotic agent, the use of a MASP-2 inhibitor for the manufacture of a medicament for the treatment of a fibrotic condition, and a method of preventing, treating, alleviating or reversing a fibrotic condition in a human subject in need thereof, the method comprising administering to the patient an effective amount of a MASP-2 inhibitor (e.g., an anti-MASP-2 antibody).
The methods of the invention can be used to prevent, treat, alleviate or reverse fibrotic conditions in a human subject suffering from any disease or disorder caused or exacerbated by fibrosis and/or inflammation, including diseases of the kidney (e.g., chronic kidney disease, igA nephropathy, C3 glomerulopathy and other glomerulonephritis), the lung (e.g., idiopathic pulmonary fibrosis, cystic fibrosis, bronchiectasis), liver (e.g., cirrhosis, non-alcoholic fatty liver disease), heart (e.g., myocardial infarction, atrial fibrosis, valve fibrosis, endocardial myocardial fibrosis), brain (e.g., stroke), skin (e.g., excessive wound healing, scleroderma, systemic sclerosis, keloids), blood vessels (e.g., atherosclerotic vascular disease), the intestine (e.g., crohn's disease), the eye (e.g., subcoystic cataract, posterior capsular opacification), musculoskeletal soft tissue structures (e.g., adhesive bursitis, duchenne's contracture, myelofibrosis), organs (e.g., endometriosis, pecies disease), and some infectious diseases (e.g., hepatitis a and hepatitis b).
MASP-2 action in diseases and conditions caused or exacerbated by fibrosis
Fibrosis is the formation or presence of excessive connective tissue in an organ or tissue, usually in response to injury or damage. Fibrosis is marked by the production of excessive extracellular matrix following injury. In the kidney, fibrosis is characterized by the progressive deposition of virtually harmful connective tissue in the kidney, which inevitably leads to reduced renal function, independently of the primary renal disease that causes the initial kidney injury. So-called epithelial to mesenchymal transition (EMT), a change in cellular characteristics, in which tubular epithelial cells are transformed into mesenchymal fibroblasts, constitute the primary mechanism of renal fibrosis. Fibrosis affects almost all tissue and organ systems, and can occur as a repair or replacement response to stimuli such as tissue injury or inflammation. Normal physiological responses to injury result in deposition of connective tissue, but if the response becomes pathological, scarring connective tissue replaces the highly differentiated cells to alter the structure and function of the tissue. At the cellular level, epithelial cells and fibroblasts proliferate and differentiate into myofibroblasts, resulting in matrix contraction, increased rigidity, microvascular compression, and hypoxia. There is currently no effective therapy or therapeutic for fibrosis, but animal studies and realistic human reports indicate that fibrotic tissue damage is reversible (Tampe and Zeisberg, natRev Nephrol, vol 10:226-237, 2014).
Many diseases result in fibrosis, which causes progressive organ failure, including diseases of the kidneys (e.g., chronic kidney disease, igA nephropathy, C3 glomerulopathy and other glomerulonephritis), the lungs (e.g., idiopathic pulmonary fibrosis, cystic fibrosis, bronchiectasis), liver (e.g., cirrhosis, nonalcoholic fatty liver disease), heart (e.g., myocardial infarction, atrial fibrosis, valve fibrosis, endocardial myocardial fibrosis), brain (e.g., stroke), skin (e.g., excessive wound healing, scleroderma, systemic sclerosis, keloids), blood vessels (e.g., atherosclerotic vascular disease), intestines (e.g., crohn's disease), eyes (e.g., subcapsular cataract, posterior capsular opacification), soft tissue structures of musculoskeletal (e.g., adhesive cystitis, dupuytren's contracture, myelofibrosis), reproductive organs (e.g., endometriosis, pecies disease), and some infectious diseases (e.g., alphavirus, hepatitis C, hepatitis b, etc.).
When fibrosis occurs in many tissues and diseases, there are common molecular and cellular mechanisms for its pathology. Extracellular matrix deposition by fibroblasts is accompanied by immune cell infiltration, primarily monocytes (see Wynn t., nat Rev Immunol 4 (8): 583-594, 2004, incorporated herein by reference). Robust inflammatory responses result in the expression of growth factors (TGF- β, VEGF, hepatocyte growth factor, connective tissue growth factor), cytokines and hormones (endothelin, IL-4, IL-6, IL-13, chemokines), degrading enzymes (elastase, matrix metalloproteinases, cathepsins) and extracellular matrix proteins (collagen, fibronectin, integrins).
In addition, the complement system is activated in many fibrotic diseases. Complement components, including membrane attack complexes, have been identified in many fibrotic tissue samples. For example, components of the lectin pathway have been described in renal disease (Satomura et al, nephron.92 (3): 702-4 (2002); sato et al, lupus 20 (13): 1378-86 (2011); liu et al, clin Exp Immunol,174 (1): 152-60 (2013)); liver disease (Rensen et al, hepatology 50 (6): 1809-17 (2009)); and pulmonary disease (Olesen et al, clin Immunol 121 (3): 324-31 (2006)).
Excessive (overshowing) complement activation has been identified as a key contributor to immune complex mediated and antibody independent glomerulonephritis. However, strong evidence suggests that uncontrolled complement activation is inherently involved in the pathophysiological progression of TI fibrosis in situ in non-glomerular disease (Quigg R.J, J Immunol 171:3319-3324, 2003, naik A. Et al, semin Nephrol 33:575-585, 2013, mather D.R. et al, clin JAm Soc Nephrol 10:P1636-1650, 2015). Strong pro-inflammatory signals triggered by local complement activation can be initiated by filtration of complement components entering the proximal tubule and subsequently the interstitial space, or by abnormal synthesis of complement components by tubules or other resident and infiltrating cells, or by altered expression of complement regulatory proteins on kidney cells, or by lack or loss or acquisition of functional mutations in complement regulatory components (Mather D.R. et al Clin J Am Soc Nephrol: P1636-1650, 2015, shearin N.S. et al, FASEB J22: 1065-1072, 2008). For example, in mice, a deficiency in complement regulator protein CR 1-related gene/protein y (Crry) results in Tubulointerstitial (TI) complement activation, where subsequent inflammation and fibrosis typical of lesions is seen in human TI disease (Naik A. Et al, semin Nephrol 33:575-585, 2013, bao L. Et al, J Am Soc Nephrol 18:811-822, 2007). Exposure of tubule epithelial cells to anaphylatoxin C3a results in an epithelial to mesenchymal transition (Tsang z et al, J Am Soc Nephrol 20:593-603, 2009). Blocking C3a signaling by the C3a receptor alone has recently been shown to reduce renal TI fibrosis in proteinuria and non-proteinuria animals (Tsang Z. Et al, J Am Soc Nephrol 20:593-603, 2009, bao L. Et al, kidney int.80:524-534, 2011).
As described herein, the present inventors have identified a central role for the lectin pathway in the initiation of renal tubular pathology and in disease progression, suggesting that lectin pathway activation is indicated in a variety of renal diseases including IgA nephropathy, C3 glomerulopathy and other glomerulonephritis (Endo M. Et al Nephrol Dialysis Transplant:1984-1990, 1998; hisano S. Et al, am J Kidney Dis 45:295-302, 2005; roos A. Et al, J Am Soc neprol 17:1724-1734, 2006; liu L.L. et al, clin exp. Immunol 174:152-160, 2013; lhotta K. Et al, nephrol Dialysis Transplant:881-886, 1999; pickering et al, kidney International:1079-1089, 2013), diabetic nephropathy (Hovind P. Et al, diabetes 54:1523-1527, 2005), ischemia reperfusion injury (Asgari E. Et al, FASEB J28:3996-4003, 2014) and graft rejection (Berger S.P. et al, am J Transplay 5:1361-1366, 2005).
As further described herein, the inventors have demonstrated that MASP-2 inhibition reduces inflammation and fibrosis in a mouse model of tubular interstitial disease. Thus, inhibitors of MASP-2 are expected to be useful in the treatment of renal fibrosis, including tubular interstitial inflammation and fibrosis, proteinuria, igA nephropathy, C3 glomerulopathy and other glomerulonephritis and renal ischemia reperfusion injury.
Renal diseases and disorders
According to National Kidney Foundation,2.6 million U.S. adults suffer from Chronic Kidney Disease (CKD). Most patients have progressive diseases that lead to renal failure, requiring erythropoiesis stimulating drugs, dialysis or kidney transplant therapy for survival. There are several drugs that can treat hypertension, the main symptom of CKD, but there are currently no drugs that address their root causes.
Studies have shown that progressive kidney injury is caused by capillary hypertension in the substructure of the kidney (called nephron) (Whitworth j.a., annals Acad of Med, vol 34 (1): 2005). Because the nephron (the filter unit of the kidney) is damaged or destroyed in the process, inflammation and tissue scarring occur, replacing the nephron with nonfunctional scar tissue. As a result, the ability of the kidneys to filter blood decreases over time. This is called renal fibrosis, which is a common pathway for progressive renal disease. Regardless of the nature of the initial lesion, renal fibrosis is considered a common final pathway for progression of renal disease to end stage renal failure. Improvement in renal fibrosis may be determined by one or more of the following: interstitial volume, collagen IV deposition and/or connective tissue growth mRNA levels were assessed. The compounds and methods described herein are useful for treating renal fibrosis.
Renal fibrosis and inflammation are the major features of end stage renal disease of almost any etiology (see Boor et al, boor p. Et al J ofAm Soc of Nephrology: 1508-1515, 2007 and Chevalier et al Kidney International: 1145-1152, 2009). Renal failure can be caused by a group of heterologous disorders. Progressive renal dysfunction leads to proteinuria and renal insufficiency. As patient health deteriorates, dialysis may only be necessary to prevent kidney injury and prevent multiple system failure. Renal failure and renal insufficiency can progress over time to End Stage Renal Disease (ESRD), which is a permanent loss of complete or nearly complete renal function. Depending on the form of the kidney disease, kidney function may be lost in about days or weeks, or may deteriorate slowly and gradually over decades. Once the patient has progressed to ESRD, dialysis (semi-dialysis or peritoneal dialysis) is required to prevent death. The patient must either maintain some form of dialysis protocol or must obtain a kidney transplant.
Components of the lectin pathway have been found in fibrotic lesions of kidney disease (Satomura et al, nephron.92 (3): 702-4 (2002); sato et al, lupus 20 (13): 1378-86 (2011); liu et al, clin Exp Immunol,174 (1): 152-60 (2013)). In IgA nephropathy, patients with glomerular MBL deposition have more severe proteinuria, reduced renal function, lower levels of serum albumin, more severe histology and greater hypertension than patients without MBL deposition (Liu et al Clin Exp immunol.2013Oct;174 (1): 152-60). Patients with Lupus nephritis (Sato et al, lupus,20 (13): 1378-86, 2011) and chronic renal failure (Satomura et al, nephron 92 (3): 702-4, 2002) also have increased levels of MBL and lectin pathway activity.
It has also been demonstrated that in a non-proteinuria model of primary tubular interstitial injury (i.e., unilateral Ureteral Obstruction (UUO)), C5 defects lead to significant improvements in the major component of renal fibrosis (Boor p. Et al J ofAm Soc ofNephrology 18:1508-1515, 2007). It has also been reported that C3 gene expression is increased in wild-type mice after UUFO, and collagen deposition is significantly reduced in C3-/-mice after UFO compared to wild-type mice, indicating a role of complement activation in renal fibrosis (Fearn et al, mol Immunol 48:1666-1733, 2011: abstract). However, prior to the findings described herein by the inventors, the complement components involved in renal fibrosis have not been sufficiently determined. As described in examples 14-17 herein, the inventors have unexpectedly determined that MASP-2 deficiency or MASP-2 is blocked by inhibitory antibodies that selectively block the lectin pathway while leaving the classical pathway intact, specifically protecting mice from renal fibrosis in various animal models of renal disease.
Thus, in certain embodiments, the present disclosure provides methods of inhibiting renal fibrosis in a subject having a renal disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to the subject in need thereof an inhibitor of MASP-2, e.g., an anti-MASP-2 antibody. The method comprises administering to a subject having a renal disease or disorder caused or exacerbated by fibrosis and/or inflammation a composition comprising an amount of a MASP-2 inhibitor effective to inhibit renal fibrosis.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, during surgery or local injection, by administering the composition locally, either directly or distally, for example, through a catheter. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide energy agents, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has resolved or is controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., an anti-MASP-2 antibody) is administered in combination with one or more agents or therapeutic modalities appropriate for the underlying renal disease or condition. In certain embodiments, MASP-2 inhibitors (e.g., anti-MASP-2 antibodies) are administered in combination with dialysis or plasmapheresis protocols. In certain embodiments, MASP-2 inhibitors (e.g., anti-MASP-2 antibodies) are used to reduce the frequency of need for dialysis or plasmapheresis. In certain other embodiments, MASP-2 inhibitors (e.g., anti-MASP-2 antibodies) are used in combination with kidney transplantation. In certain other embodiments, MASP-2 inhibitors (e.g., anti-MASP-2 antibodies) are used to control renal insufficiency and prevent further decline in renal function in patients awaiting kidney transplantation.
For example, in certain embodiments, anti-MASP-2 antibodies are used to inhibit renal fibrosis, thereby treating or ameliorating glomerular disorders such as focal segmental glomerulosclerosis and nephrotic syndrome (including treating or ameliorating symptoms of the disorder). Exemplary symptoms that may be treated include, but are not limited to, hypertension, proteinuria, hyperlipidemia, hematuria, and hypercholesterolemia. In some embodiments, the MASP-2 inhibitor inhibits tubular interstitial fibrosis. In certain embodiments, the treatment comprises improving kidney function, reducing proteinuria, improving hypertension, and/or reducing kidney fibrosis. In certain embodiments, the treatment comprises (i) delaying or preventing progression to renal insufficiency, renal failure, or ESRD; (ii) delay, reduce or prevent the need for dialysis; or (iii) delay or prevent the need for kidney transplantation.
Certain specific kidney diseases and conditions caused or exacerbated by fibrosis and/or inflammation are described below.
In certain embodiments, the renal disease caused or exacerbated by fibrosis and/or inflammation is a glomerular disease such as Focal Segmental Glomerulosclerosis (FSGS). Glomerular disease damages the glomeruli, causing leakage of proteins and sometimes red blood cells into the urine. Sometimes glomerular diseases also interfere with waste clearance through the kidneys, so they begin to accumulate in the blood. Symptoms of glomerular disease include proteinuria, hematuria, reduced glomerular filtration rate, hypoproteinemia, and edema. Many different diseases can lead to glomerular diseases. It may be the direct result of an infection or a drug that is toxic to the kidneys, or it may be caused by a disease affecting the whole body, such as hypertension, diabetes, or lupus. FSGS is a specific glomerular disease, but even this specific condition characterized by scarring in the kidneys can have a number of causes. FSGS patients typically progress to advanced renal disease within 5-20 years, but patients with invasive forms of disease progress to ESRD within 2-3 years.
In certain embodiments, the renal disease caused or exacerbated by fibrosis and/or inflammation is Diabetic Nephropathy (DN), a field of medical need that is significantly unmet. Diabetic nephropathy is a kidney disease or injury that results as a complication of diabetes. The condition is exacerbated by hypertension, hyperglycemia levels, and high cholesterol and lipid levels. The exact cause of diabetic nephropathy is unknown. However, without being bound by theory, it is believed that uncontrolled hyperglycemia leads to the occurrence of kidney damage, such as fibrosis and scarring of tissue. In humans, DN appears as a clinical syndrome consisting of albuminuria, progressively decreasing Glomerular Filtration Rate (GFR) and increased risk of cardiovascular disease. Diabetic albuminuria is associated with the development of characteristic histopathological features, including Glomerular Basement Membrane (GBM) thickening and mesangial distention. With the progression of albuminuria and the ensuing occurrence of renal insufficiency, glomerulosclerosis, arteriovenous transparency and tubular interstitial fibrosis occur.
Accordingly, in one embodiment, the present disclosure provides a method for treating diabetic nephropathy comprising administering to a subject in need thereof an effective amount of a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody). In certain embodiments, the treatment comprises alleviating one or more symptoms of diabetic nephropathy. In certain embodiments, the treatment comprises alleviating, delaying or eliminating the need for dialysis. In certain embodiments, the treatment comprises reducing, delaying or eliminating the need for kidney transplantation. In certain embodiments, the treatment comprises delaying, preventing, or reversing the progression of diabetic nephropathy to renal failure or end stage renal disease.
In certain embodiments, the renal disease caused or exacerbated by fibrosis and/or inflammation is lupus nephritis. As described in more detail below, lupus nephritis, which is a serious complication of Systemic Lupus Erythematosus (SLE), is another example of renal fibrosis that can be treated with MASP-2 inhibitors (e.g., anti-MASP-2 antibodies).
Thus, in one embodiment, the present disclosure provides a method for inhibiting renal fibrosis in a subject having a renal disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering an effective amount of a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody). In some embodiments, the kidney disease or disorder exacerbated by fibrosis and/or inflammation is selected from chronic kidney disease, chronic renal failure, glomerular disease (e.g., focal segmental glomerulosclerosis), immune complex disorders (e.g., igA nephropathy, membranous nephropathy), lupus nephritis, nephrotic syndrome, diabetic nephropathy, tubular interstitial injury, and C3 glomerulonephritis or other types of glomerulonephritis.
Method for preventing or treating kidney injury caused by drug-induced toxicity
Another cause of kidney injury includes drug-induced toxicity. For example, nephrotoxins can cause direct toxicity to tubular epithelial cells. As described herein, the inventors have demonstrated that MASP-2 deficient mice are protected from doxorubicin-induced nephropathy.
Nephrotoxins include, but are not limited to, therapeutic drugs (e.g., cisplatin, gentamicin, ceftiodine, cyclosporine, amphotericin, doxorubicin), radiocontrast dyes, pesticides (e.g., paraquat), and environmental pollutants (e.g., trichloroethylene and dichloroacetylene). Other examples include aminonucleoside Puromycin (PAN); aminoglycosides such as gentamicin; cephalosporins such as ceftiofur; a troponin inhibitor such as tacrolimus or sirolimus. Drug-induced nephrotoxicity may also be caused by non-steroidal anti-inflammatory drugs, antiretroviral agents, anti-cytokines, immunosuppressants, oncology drugs or ACE inhibitors. Drug-induced nephrotoxicity may be further caused by fenoprofen abuse, ciprofloxacin, clopidogrel, cocaine, cox-2 inhibitors, diuretics, foscarnet, gold, ifosfamide, immunoglobulins, chinese herbal medicine, interferons, lithium, mannitol, mesalazine, mitomycin, nitrosourea, penicillamine, penicillin, pentamidine, quinine, rifampin, streptozocin, sulfanilamide, ticlopidine, ampelopsis, valproic acid, doxorubicin, glycerol, cidofovir, tobramycin, neomycin sulfate, sulfocolicin, vancomycin, amikacin, cefotaxime, cisplatin, aciclovir, lithium, interleukin-2, cyclosporine, or indinavir.
Thus, in one embodiment, a subject at risk of developing or suffering from kidney injury may receive one or more therapeutic agents having a nephrotoxic effect. These subjects may be administered MASP-2 inhibitors of the invention prior to or concurrently with the therapeutic agent. Likewise, MASP-2 inhibitors may be administered after the therapeutic agent to treat or reduce the likelihood of developing nephrotoxicity.
Diseases and conditions associated with proteinuriaIt has been determined that impaired glomerular filtration of proteins leads to proteinuria and accelerated progressive loss of nephrons, which occur in all chronic kidney diseases (Remuzzi and Bertani, new Eng.J Med vol 339 (20): 1448-1456, 1998). For example, in the study described in Eddy et al, am J Pathol 135:719-33, 1989, interstitial damage and scarring invariably occurs after glomerular filtration of albumin. As further described in Eddy et al, 1989, complement C3 deposition on the luminal surface of the proximal tubule was observed in rats with kidney disease caused by protein-overload, suggesting that components of the complement system filtered by the glomeruli may cause interstitial damage. It has been demonstrated that complement depletion or lack of C6 improves tubular interstitial lesions in proteinuria animal models Such as mesangial proliferative glomerulonephritis, doxorubicin nephropathy, 5/6 nephrectomy and aminonucleoside puromycin nephropathy (Boor et al J of Am Soc of Nephrology: JASN 18:1508-1515, 2007). Human studies have shown that proteinuria is an independent predictor of chronic renal disease progression and that proteinuria reduction is kidney protective (Ruggenti P. Et al, J Am SocNephrol 23:1917-1928, 2012).
Thus, in one embodiment, the present disclosure provides a method of preventing or reducing proteinuria and/or preventing or reducing kidney damage in a subject suffering from a disease or condition associated with proteinuria, comprising administering an amount of a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) effective to reduce or prevent proteinuria in the subject. In some embodiments, the disease or condition associated with proteinuria is selected from nephrotic syndrome, preeclampsia, eclampsia, toxic damage to the kidney, amyloidosis, collagen vascular disease (e.g., systemic lupus erythematosus), dehydration, glomerular disease (e.g., membranous glomerulonephritis, focal segmental glomerulonephritis, morbid disease, steatonephrosis), intensive exercise, stress, benign orthotopic (postural) proteinuria, focal segmental glomerulosclerosis, igA nephropathy (i.e., begella disease), igM nephropathy, membranous proliferative glomerulonephritis, membranous nephropathy, morbid disease, sarcoidosis, albert syndrome, diabetes (diabetic nephropathy), drug-induced toxicity (e.g., NSAIDS, nicotine, penicillamine, lithium carbonate, gold and other heavy metals, ACE inhibitors, antibiotics or opiates (e.g., heroin)); fabry's disease, infection (e.g., HIV, syphilis, hepatitis a, b or c, post streptococcal infection, schistosomiasis urinary); amino acid urine syndrome, van sconey syndrome, hypertensive nephrosclerosis, interstitial nephritis, sickle cell disease, hemoglobinuria, multiple myeloma, myoglobin urine, organ rejection (e.g., kidney transplant rejection), ebola hemorrhagic fever, patella nail syndrome, familial mediterranean fever, HELLP syndrome, systemic lupus erythematosus, wegener's granulomatosis, rheumatoid arthritis, glycogen storage disease type 1, goodpasture's syndrome, allergic purpura, urinary tract infections that have spread to the kidney, sjogren's syndrome, and post-infection glomerulonephritis.
Liver disease
Liver fibrosis, also known as liver fibrosis, is caused by the accumulation of scar tissue in the liver and is a feature of most types of liver diseases. Replacement of healthy liver tissue by scar tissue compromises the ability of the liver to function properly. If the condition causing scarring is untreated, liver fibrosis may develop into cirrhosis and complete liver failure, a life threatening condition. The main causes of liver fibrosis are alcohol abuse, chronic hepatitis c virus infection, nonalcoholic steatohepatitis and hepatotoxicity (e.g., drug-induced liver damage caused by acetaminophen or other drugs).
Components of the lectin pathway have been found in fibrotic lesions of liver disease (Rensen et al, hepatology 50 (6): 1809-17 (2009)). For example, in nonalcoholic steatohepatitis (also known as fatty liver disease), there is a general activation of complement system proteins, and its expression is related to disease severity (Rensen et al, hepatology 50 (6): 1809-17 (2009), where MBL accumulation is found in addition to C3 and C9 deposition, confirming activation of the lectin pathway.
Thus, in certain embodiments, the present disclosure provides methods of inhibiting liver fibrosis in a subject having a liver disease or disorder caused or exacerbated by fibrosis and/or inflammation comprising administering to a subject in need thereof a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody. The method comprises administering to a subject having a liver disease or condition caused or exacerbated by fibrosis and/or inflammation a composition comprising an amount of a MASP-2 inhibitor effective to inhibit liver fibrosis.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying liver disease or condition.
In some embodiments, the liver disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from: cirrhosis, non-alcoholic fatty liver disease (steatohepatitis), liver fibrosis secondary to alcohol abuse, liver fibrosis secondary to acute or chronic hepatitis, biliary diseases, and toxic liver injury (e.g., liver toxicity due to drug-induced liver injury caused by acetaminophen or other drugs).
Pulmonary disease
Pulmonary fibrosis is the formation or development of excessive fibrous connective tissue in the lung, where normal lung tissue is replaced by fibrotic tissue. This scarring results in stiffness of the lungs and impaired lung structure and function. In humans, pulmonary fibrosis is thought to be caused by repeated tissue damage within and between tiny air sacs (lung cells) of the lung. Under experimental setup, various animal models have repeated aspects of human disease. For example, an external agent such as bleomycin, fluorescein isothiocyanate, silica or asbestos may be infused into the trachea of an animal (Gharaee-kerani et al, animal Models of Pulmonary fibris. Methods mol. Med.,2005, 117:251-259).
Thus, in certain embodiments, the present disclosure provides methods of inhibiting pulmonary fibrosis in a subject having a pulmonary disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit pulmonary fibrosis, reduce pulmonary fibrosis, and/or improve pulmonary function. Symptomatic improvement of lung function includes improvement of lung function and/or lung capacity, reduction of fatigue and improvement of oxygen saturation.
In some embodiments, the present disclosure provides methods of treating, inhibiting, preventing, or ameliorating pulmonary fibrosis in a subject having cystic fibrosis comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying pulmonary disease or condition.
Certain specific lung diseases and conditions caused or exacerbated by fibrosis and/or inflammation are described below.
In certain embodiments, the lung disease caused or exacerbated by fibrosis and/or inflammation is Chronic Obstructive Pulmonary Disease (COPD). COPD is a disease in which the airway wall is fibrotic with the accumulation of myofibroblasts and collagen, the leading cause of disability, and it is the fourth leading cause of death in the united states. COPD blocks air flow and makes patient breathing increasingly difficult. COPD is caused by damage to the airways, which ultimately interferes with oxygen and carbon dioxide exchange in the lungs. COPD includes chronic obstructive bronchitis and emphysema, and generally both. COPD patients whose lungs have been damaged and whose lung function has been compromised are at increased risk of complications associated with bacterial and viral infections.
Accordingly, in one embodiment, the present disclosure provides a method of treating Chronic Obstructive Pulmonary Disease (COPD), comprising administering to a subject in need thereof an amount of a MASP-2 inhibitor (e.g., an anti-MASP-2 antibody) effective to inhibit and/or reduce pulmonary fibrosis. In certain embodiments, the treatment comprises reducing one or more symptoms of COPD. Symptoms of COPD and/or pulmonary fibrosis include, but are not limited to, coughing with mucus, shortness of breath (dyspnea) where mild activity may become worsened, fatigue, frequent respiratory tract infections, wheezing, chest tightness, irregular heartbeat (arrhythmia), need for ventilator and oxygen therapy, right side heart failure or pulmonary heart disease (heart swelling and heart failure due to chronic lung disease), pneumonia, pneumothorax, severe weight loss and malnutrition. Symptoms also include reduced lung function, as assessed using one or more standard lung function tests.
In certain embodiments, the pulmonary disease caused or exacerbated by fibrosis and/or inflammation is pulmonary fibrosis associated with scleroderma. As described in more detail below, pulmonary fibrosis associated with scleroderma is another example of pulmonary fibrosis that can be treated with MASP-2 inhibitors (e.g., MASP-2 inhibitory antibodies).
In some embodiments, the pulmonary disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from: chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis associated with scleroderma, bronchiectasis, and pulmonary arterial hypertension.
Heart and vascular diseases
Many different heart and vascular diseases are caused by a common fibrosis process. Excessive deposition of fibrotic tissue in the heart leads to cardiac pathology, where excessive production of extracellular matrix proteins alters the structure, architecture, shape, and impact contractile function of the heart (Khan and Sheppard, immunology 118:10-24, 2006).
Studies have shown that fibrosis can significantly promote cardiac dysfunction in ischemic, dilated and hypertrophic cardiomyopathy. For example, patients with chronic atrial fibrillation have been shown to have higher levels of myocardial interstitial fibrosis than controls (Khan and Sheppard, immunology 118:10-24, 2006). As another example, it has been determined that in the United states, most cases of Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) exhibit fat infiltration and scarring (fibrous fatty ARVC) (Burke et al, circulation 97:1571-1580, 1998). In studies examining the histopathological characteristics of ventricular muscle in human subjects with ARVC, it has been determined that extensive fibrosis exists in biopsy samples from pediatric patients with ARVC (Nishikawa T. Et al, cardiovascular Pathology vol (4): 185-189, 1999).
Thus, in certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject suffering from a heart or vascular disease or condition caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit cardiac and/or vascular fibrosis and/or improve cardiac and/or vascular function.
In some embodiments, the present disclosure provides methods of treating, inhibiting, preventing, or ameliorating fibrosis in a subject having valve fibrosis comprising administering to the subject in need thereof an inhibitor of MASP-2, e.g., an inhibitory antibody of MASP-2.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying cardiac disease or vascular disease or condition.
In some embodiments, the heart or vascular disease or condition caused or exacerbated by fibrosis and/or inflammation is selected from: cardiac fibrosis, myocardial infarction, atrial fibrosis, cardiac endocardial fibrosis-induced Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), vascular disease, atherosclerotic vascular disease, vascular stenosis, restenosis, vasculitis, phlebitis, deep venous thrombosis, and abdominal aortic aneurysm.
Chronic infectious diseases
Chronic infectious diseases, such as hepatitis c and hepatitis b, cause tissue inflammation and fibrosis, and high lectin pathway activity may be detrimental. Inhibitors of MASP-2 may be beneficial in such diseases. For example, MBL and MASP-1 levels were found to be important predictors of the severity of liver fibrosis in Hepatitis C Virus (HCV) infection (Brown et al, clin Exp immunol.147 (1): 90-8, 2007; saadanay et al, arab J gastroenterol.12 (2): 68-73, 2011; saeed et al, clin Exp immunol.174 (2): 265-73, 2013). MASP-1 has previously been shown to be a potent activator of the MASP-2 and lectin pathways (Megyeri et al, J Biol chem.29:288 (13): 8922-34, 2013). Alphaviruses such as chikungunya and ross river viruses induce strong host inflammatory responses leading to arthritis and myositis, and this pathology is mediated by the MBL and lectin pathways (Gunn et al, PLoS pathg.8 (3): e1002586, 2012).
Thus, in certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having or having previously had a chronic infectious disease that causes inflammation and/or fibrosis, comprising administering to a subject in need thereof a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying chronic infectious disease.
In some embodiments, the chronic infectious disease that causes inflammation and/or fibrosis is selected from: alpha virus, hepatitis a, hepatitis b, hepatitis c, tuberculosis, HIV and influenza.
Autoimmune diseases
Scleroderma is a chronic autoimmune disease characterized by fibrosis, vascular changes, and autoantibodies. There are two main forms: limited system scleroderma and diffuse system scleroderma. The skin symptoms of restrictive systemic scleroderma affect the hands, arms and face. Patients with this form of scleroderma typically have one or more of the following complications: calcaneosis, raynaud's phenomenon, esophageal dysfunction, scleroderma and telangiectasia. Diffuse systemic scleroderma develops rapidly and affects a large area of skin and one or more internal organs, typically the kidneys, esophagus, heart and/or lungs.
Scleroderma affects small blood vessels called arterioles in all organs. First, the endothelial cells and smooth muscle cells of the arterioles gradually decrease through apoptosis. These cells are replaced by collagen and other fibrous materials. Inflammatory cells, particularly cd4+ helper T cells, infiltrate the arterioles and cause further damage.
The skin manifestations of scleroderma can be painful, can impair the use of affected areas (e.g., use of hands, fingers, toes, feet, etc.) and can impair appearance. Skin ulcers may occur, and such ulcers may be susceptible to infection or even necrosis. The skin of ulcers may be difficult to heal or slow to heal. The difficulty in curing skin ulcers may be particularly exacerbated in patients with impaired circulation (e.g., those with reynolds phenomenon). In certain embodiments, the methods of the present disclosure are used to treat scleroderma, e.g., a cutaneous symptom of scleroderma. In certain embodiments, treating scleroderma comprises treating a skin ulcer, such as a finger ulcer. Administration of MASP-2 inhibitors such as anti-MASP-2 antibodies may be used to reduce fibrosis and/or inflammatory symptoms of scleroderma in the affected tissues and/or organs.
In addition to skin symptoms/manifestations, scleroderma can also affect the heart, kidneys, lungs, joints and digestive tract. In certain embodiments, treating scleroderma comprises treating symptoms of the disease in any one or more of these tissues, for example, by reducing fibrotic and/or inflammatory symptoms. Pulmonary problems are one of the most serious complications of scleroderma and are responsible for the majority of mortality associated with the disease. Two major pulmonary conditions associated with scleroderma are pulmonary fibrosis and pulmonary arterial hypertension. Patients with compromised lungs may have either or both conditions. Pulmonary fibrosis associated with scleroderma is one example of pulmonary fibrosis that can be treated with MASP-2 inhibitors. Scleroderma involving the lung leads to scarring (pulmonary fibrosis). Such pulmonary fibrosis occurs in about 70% of scleroderma patients, although its progression is generally slow, and symptoms vary widely in severity among patients. For patients who do have symptoms associated with pulmonary fibrosis, symptoms include dry cough, shortness of breath, and reduced exercise capacity. About 16% of patients with a certain level of pulmonary fibrosis develop severe pulmonary fibrosis. Patients with severe pulmonary fibrosis experience a significant decrease in pulmonary function and alveolitis.
In certain embodiments, the methods of the present disclosure are used to treat scleroderma, e.g., pulmonary fibrosis associated with scleroderma. Administration of MASP-2 inhibitors, such as MASP-2 inhibitory antibodies, may be used to reduce the fibrotic symptoms of scleroderma in the lung. For example, the methods may be used to improve lung function and/or reduce the risk of mortality due to scleroderma.
The involvement of the kidneys is also common in scleroderma patients. Renal fibrosis associated with scleroderma is an example of renal fibrosis that can be treated by administration of MASP-2 inhibitors, such as anti-MASP-2 antibodies. In certain embodiments, the methods of the present disclosure are used to treat scleroderma, such as renal fibrosis associated with scleroderma. In one embodiment, administration of MASP-2 inhibitory antibodies may be used to reduce the fibrotic symptoms of scleroderma in the kidney. For example, the methods can be used to improve kidney function, reduce proteins in urine, reduce hypertension and/or reduce the risk of renal crisis that can lead to fatal renal failure.
Systemic Lupus Erythematosus (SLE) is a chronic inflammatory autoimmune disorder characterized by spontaneous B and T cell autoreactivity and multiple organ immune injury, and can affect skin, joints, kidneys, and other organs. Almost all people with SLE have joint pain, and the majority of arthritis occurs. Frequently affected joints are fingers, hands, wrists and knees. Common symptoms of SLE include: arthritis; fatigue; general malaise, anxiety or cachexia (cachexia); joint pain and swelling; muscle pain; nausea and vomiting; and rash. In addition, symptoms may also include: abdominal pain; hematuria; finger discoloration under pressure or cold; numbness and tingling; and skin erythema. In some patients, SLE has an associated lung or kidney. Without being bound by theory, inflammation and/or fibrosis of the lung and kidney damage these organs, and cause symptoms associated with lung and/or kidney damage. In some cases, patients with SLE develop a particular kidney condition known as lupus nephritis. In certain embodiments, the disclosure provides methods of treating SLE comprising administering an effective amount of a MASP-2 inhibitor, such as an anti-MASP-2 antibody. Administration of MASP-2 inhibitory antibodies may be used to reduce one or more symptoms of SLE. In certain embodiments, anti-MASP-2 antibodies are administered for the treatment of SLE in patients with lupus nephritis. In such cases, treating SLE includes treating lupus nephritis, e.g., by reducing symptoms of lupus nephritis. In certain embodiments, treating comprises treating skin symptoms of SLE. In certain embodiments, the treatment comprises reducing one or more symptoms of lupus nephritis. In certain embodiments, the treatment comprises reducing, delaying or eliminating the need for dialysis. In certain embodiments, the treatment comprises reducing, delaying or eliminating the need for kidney transplantation. In certain embodiments, the treatment comprises delaying or preventing the progression of lupus nephritis to renal failure or end stage renal disease.
Lupus nephritis is inflammation of the kidney and is a serious complication of Systemic Lupus Erythematosus (SLE). In the kidneys, lupus nephritis can result in a loss of function. Lupus nephritis patients can eventually develop renal failure and require dialysis or kidney transplantation. Related complications that can also be treated using the methods of the present disclosure include interstitial nephritis and nephrotic syndrome. Symptoms of lupus nephritis include: hematuria, urinary foamy appearance, hypertension, urinary proteins, fluid retention and oedema. Other symptoms include signs and symptoms of renal fibrosis and/or renal failure. Lupus nephritis can lead to renal failure, and even end stage renal disease if untreated.
Thus, in certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having an autoimmune disease that causes or aggravates fibrosis and/or inflammation, comprising administering a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody, to a subject in need thereof. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying autoimmune disease.
In some embodiments, the autoimmune disease that causes or aggravates fibrosis and/or inflammation is selected from: scleroderma and Systemic Lupus Erythematosus (SLE).
Central nervous system diseases and conditions
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a disease or condition of the central nervous system caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., an anti-MASP-2 antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example, by administering the composition locally, either directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying central nervous system disease or disorder.
In some embodiments, the disease or disorder of the central nervous system caused or exacerbated by fibrosis and/or inflammation is selected from the group consisting of: stroke, traumatic brain injury, and spinal cord injury.
Skin diseases and conditions
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a skin disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered topically to the fibrotic area, for example by topically applying the composition to the skin, or directly or distally (e.g., via a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying skin disease or disorder.
In some embodiments, the skin disease or condition caused or exacerbated by fibrosis and/or inflammation is selected from: skin fibrosis, wound healing, scleroderma, systemic sclerosis, keloids, connective tissue disease, scarring and hypertrophic scars.
Bone and soft tissue disorders and conditions of musculoskeletal bones
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a bone or soft tissue disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by topically applying the composition to bone or soft tissue structures, or directly or distally (e.g., via a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying bone or soft tissue disease or disorder.
In some embodiments, the bone or soft tissue disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from the group consisting of: osteoporosis and/or osteopenia associated with, for example, cystic fibrosis, myelodysplastic conditions with increased bone fibrosis, adhesive capsulitis, dupuytren's contracture, and myelofibrosis.
Joint diseases and conditions
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a joint disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by topically applying the composition to the joint, or directly or distally (e.g., via a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying joint disease or disorder.
In some embodiments, the joint disease or disorder caused or exacerbated by fibrosis and/or inflammation is joint fibrosis.
Digestive diseases and conditions
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a digestive disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by local administration directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying digestive disease or disorder.
In some embodiments, the digestive disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from: crohn's disease, ulcerative colitis and pancreatic fibrosis.
Eye diseases and conditions
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having an ocular disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by local administration directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intraarterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration to the eye (e.g., as eye drops), or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying ocular disease or disorder.
In some embodiments, the ocular disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from: subcapsular cataract, posterior opacification, macular degeneration, and retinal and vitreoretinopathy.
Diseases and conditions of the reproductive organs
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a reproductive disease or disorder caused or exacerbated by fibrosis and/or inflammation, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by local administration directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying reproductive disease or disorder.
In some embodiments, the reproductive disease or disorder caused or exacerbated by fibrosis and/or inflammation is selected from: endometriosis and pecrenet disease.
Scar formation associated with trauma
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject suffering from a disease or condition resulting from scar formation associated with a wound, comprising administering to a subject in need thereof a MASP-2 inhibitor, such as a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by local administration directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intra-arterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration, or, for non-peptide drugs, by oral administration. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying disease or disorder.
In some embodiments, the scar formation associated with the wound is selected from: surgical complications (e.g., surgical adhesions, where scar tissue may form between internal organ functions, cause contractures, pain, and may cause infertility), chemotherapy drug-induced fibrosis, radiation-induced fibrosis, and scar formation associated with burns.
Other diseases and conditions caused or exacerbated by fibrosis and/or inflammation
In certain embodiments, the present disclosure provides methods of preventing, treating, reversing, inhibiting and/or reducing fibrosis and/or inflammation in a subject having a disease or condition caused or exacerbated by fibrosis and/or inflammation selected from organ transplantation, breast fibrosis, muscle fibrosis, retroperitoneal fibrosis, thyroid fibrosis, lymph node fibrosis, bladder fibrosis and pleural fibrosis, comprising administering to a subject in need thereof a MASP-2 inhibitor, e.g., a MASP-2 inhibitory antibody. The method comprises administering a composition comprising an amount of a MASP-2 inhibitor effective to inhibit fibrosis and/or inflammation.
The MASP-2 inhibitory composition may be administered locally to the fibrotic area, for example by local administration directly or distally (e.g., through a catheter) during surgery or local injection. Alternatively, the MASP-2 inhibitor may be administered systemically to the subject, e.g., by intraarterial, intravenous, intramuscular, inhalation, nasal, subcutaneous, or other parenteral administration, by topical administration to the eye (e.g., as eye drops), or, for non-peptide drugs, may be administered orally. Administration may be repeated, as determined by the physician, until the condition has been eliminated or controlled.
In certain embodiments, a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered in combination with one or more agents or therapeutic forms suitable for the underlying disease or disorder.
In certain embodiments of any of the various methods and pharmaceutical compositions described herein, the MASP-2 inhibitory antibody selectively blocks the lectin pathway while leaving the classical pathway intact.
MASP-2 inhibitors
In various aspects, the invention provides methods of inhibiting the adverse effects of fibrosis and/or inflammation comprising administering to a subject in need thereof a MASP-2 inhibitor. The MASP-2 inhibitor is administered in an amount effective to inhibit MASP-2 dependent complement activation in a living subject. In the practice of this aspect of the invention, representative MASP-2 inhibitors include: molecules that inhibit MASP-2 biological activity (such as small molecule inhibitors, anti-MASP-2 antibodies (e.g., MASP-2 inhibitory antibodies), or blocking peptides that interact with MASP-2 or interfere with protein-protein interactions), and molecules that reduce MASP-2 expression (such as MASP-2 antisense nucleic acid molecules, MASP-2 specific RNAi molecules, and MASP-2 ribozymes), thereby preventing MASP-2 from activating the lectin complement pathway. MASP-2 inhibitors may be used alone as the primary therapy, or in combination with other therapeutic agents as an adjunct therapy to enhance the therapeutic benefits of other drug therapies.
MASP-2 dependent inhibition of complement activation is characterized by at least one of the following changes in the complement system components due to administration of a MASP-2 inhibitor according to the methods of the invention: inhibition of formation or production of MASP-2 dependent complement activation system products C4b, C3a, C5a and/or C5b-9 (MAC) (e.g., as measured in example 2), reduction of C4 cleavage and C4b deposition (e.g., as measured in example 2), or reduction of C3 cleavage and C3b deposition (e.g., as measured in example 2).
According to the invention, inhibitors of MASP-2 are used which are effective in inhibiting fibrosis and/or inflammation and which exhibit a detectable anti-fibrotic activity and/or induce a reduction in fibrosis. Within the context of the present invention, anti-fibrotic activity may comprise at least one or more of the following: reduced inflammation, e.g., by activating and recruiting macrophages and endothelial cells, as compared to the fibrotic activity in the absence of the MASP-2 inhibitor; recruiting and activating lymphocytes and/or eosinophils by secreting a number of cytokines/chemokines; release of cytotoxic mediators and fibrogenic cytokines evaluation; (2) reduced cell proliferation, ECM synthesis, or angiogenesis; and/or (3) reduced collagen deposition.
The evaluation of anti-fibrotic agents, such as MASP-2 inhibitors, may be detected using any technique known to the skilled artisan. For example, the evaluation of the anti-fibrotic agent may be evaluated in a UUO model (as described in examples 12 and 14 herein). If a detectable anti-fibrotic activity and/or decrease in fibrosis is assessed using a MASP-2 inhibitor, such a MASP-2 inhibitor is considered to be useful as a medicament for preventing, treating, reversing and/or inhibiting fibrosis.
The assessment of fibrosis may be performed periodically, e.g., weekly or monthly. The increase/decrease in fibrosis and/or the presence of anti-fibrotic activity may thus be assessed periodically, e.g. weekly or monthly. Such assessment is preferably performed at several time points for a given subject, or at one or several time points for a given subject and healthy control. The evaluation may be performed at regular time intervals, e.g. weekly or monthly. The evaluation may thus be performed regularly, for example weekly or monthly. When an assessment results in the discovery of reduced fibrosis or the presence of anti-fibrotic activity, MASP-2 inhibitors, such as MASP-2 inhibitory antibodies, are believed to exhibit detectable anti-fibrotic activity and/or induce a reduction or decrease in fibrosis.
MASP-2 inhibitors useful in the practice of this aspect of the invention include, for example, MASP-2 antibodies and fragments thereof, MASP-2 inhibitory peptides, small molecules, MASP-2 soluble receptors, and expression inhibitors. MASP-2 inhibitors may inhibit the MASP-2 dependent complement activation system by blocking the biological function of MASP-2. For example, inhibitors are effective in blocking interactions between MASP-2 proteins, interfering with MASP-2 dimerization or assembly, blocking Ca 2+ Binding, interfering with the MASP-2 serine protease active site, or may reduce MASP-2 protein expression.
In some embodiments, the MASP-2 inhibitor selectively inhibits MASP-2 complement activation, preserving the functional integrity of the C1 q-dependent complement activation system.
In one embodiment, the MASP-2 inhibitor used in the methods of the invention is a specific MASP-2 inhibitor that specifically binds to a polypeptide comprising SEQ ID NO. 6 with an affinity that is at least 10-fold greater than the affinity for binding to other antigens in the complement system. In another embodiment, the binding affinity of the MASP-2 inhibitor to bind specifically to a polypeptide comprising SEQ ID NO. 6 is at least 100-fold greater than to bind to other antigens in the complement system. In one embodiment, the MASP-2 inhibitor specifically binds to at least one of the CCP1-CCP2 domain (aa 300-431 of SEQ ID NO: 6) or the serine protease domain of MASP-2 (aa 445-682 of SEQ ID NO: 6) and inhibits MASP-2-dependent complement activation. In one embodiment, the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds MASP-2. The binding affinity of MASP-2 inhibitors may be determined using a suitable binding assay.
MASP-2 polypeptides have a molecular structure similar to that of the proteases MASP-1, MASP-3 and C1r and C1s of the C1 complement system. A representative example of a cDNA molecule shown in SEQ ID NO. 4 encoding MASP-2 (consisting of the amino acid sequence shown in SEQ ID NO. 5) provides a human MASP-2 polypeptide with a leader sequence (amino acids 1-15) that is cleaved after secretion to yield the mature form of human MASP-2 (SEQ ID NO. 6). As shown in FIG. 2, the human MASP 2 gene comprises twelve exons. Human MASP-2cDNA is encoded by exons B, C, D, F, G, H, I, J, K and L. Alternative splicing produces a 20kDa protein called MBL-associated protein 19 ("MAp 19", also called "sMAP") (SEQ ID NO: 2), encoded by exons B, C, D and E shown in FIG. 2 (SEQ ID NO: 1). The cDNA molecule shown in SEQ ID NO. 50 encodes murine MASP-2 (consisting of the amino acid sequence shown in SEQ ID NO. 51) providing the murine MASP-2 polypeptide with a leader sequence that is cleaved after secretion to yield the mature form of murine MASP-2 (SEQ ID NO. 52). The cDNA molecule shown in SEQ ID NO. 53 encodes rat MASP-2 (consisting of the amino acid sequence shown in SEQ ID NO. 54) providing the rat MASP-2 polypeptide with a leader sequence that is cleaved after secretion to yield the mature form of rat MASP-2 (SEQ ID NO. 55).
It will be appreciated by those skilled in the art that the sequences disclosed in SEQ ID NO. 4, SEQ ID NO. 50 and SEQ ID NO. 53 represent individual alleles of human, mouse and rat MASP-2, respectively, and that allelic variation and alternative splicing are contemplated. Allelic variants of the nucleotide sequences shown in SEQ ID No. 4, SEQ ID No. 50 and SEQ ID No. 53, including variants comprising silent mutations and variants in which the mutation results in an amino acid sequence change, are within the scope of the invention. Allelic variants of MASP-2 sequences may be cloned from different individuals by probing cDNA or genomic libraries according to standard methods.
The domains of the human MASP-2 protein (SEQ ID NO: 6) are shown in FIGS. 1 and 2A, and include the N-terminal C1r/C1 s/sea urchin Vegf/bone morphogenic protein (CUBI) domain (amino acids 1-121 of SEQ ID NO: 6), the epidermal growth factor-like domain (amino acids 122-166), the second CUBI domain (amino acids 167-293), and the tandem complement regulatory protein domain and serine protease domain. Alternative splicing of the MASP 2 gene results in MAP19 as shown in FIG. 1. MAp19 is a non-enzymatic protein containing the N-terminal CUBI-EGF region of MASP-2 with 4 additional residues (EQSL) from exon E shown in FIG. 1.
Some proteins have been shown to bind to MASP-2 or interact with MASP-2 through interactions between proteins. For example, MASP-2 is known to bind to and form Ca with the lectin proteins MBL, H-fiber gellin and L-fiber gellin 2+ A dependent complex. Each MASP-2/lectin complex has been shown to activate complement by MASP-2 dependent cleavage of proteins C4 and C2 (Ikeda, K., et al, J. Biol. Chem.262:7451-7454,1987; matsushita, M., et al, J. Exp. Med.176:1497-2284,2000; matsushita, M., et al, J. Immunol.168:3502-3506, 2002). Studies have shown that the CUBI-EGF domain of MASP-2 is essential for binding of MASP-2 to MBL (Thielens, N.M., et al, J.Immunol.166:5068,2001). Studies have also shown that the CUBIEGFCUBII domain mediates dimerization of MASP-2, which is required for the formation of active MBL complexes (Wallis, R., et al, J.biol. Chem.275:30962-30969,2000). Thus, MASP-2 inhibitors that bind to or interfere with a target region of MASP-2 known to be important for MASP-2 dependent complement activation may be identifiedAnd (5) setting.
anti-MASP-2 antibodies
In some embodiments of this aspect of the invention, the MASP-2 inhibitor comprises an anti-MASP-2 antibody that inhibits the MASP-2 dependent complement activation system. anti-MASP-2 antibodies useful in this aspect of the invention include polyclonal, monoclonal, or recombinant antibodies derived from any antibody-producing mammal, and may be multispecific, chimeric, humanized, anti-idiotype antibodies, and antibody fragments. Antibody fragments include Fab, fab', F (ab) 2 、F(ab') 2 Fv fragments, scFv fragments and single chain antibodies, see further description herein.
Using the assays described herein, MASP-2 antibodies can be screened for their ability to inhibit the MASP-2-dependent complement activation system and anti-fibrotic activity and/or for their ability to inhibit kidney damage associated with proteinuria or doxorubicin-induced kidney disease. Several MASP-2 antibodies have been described in the literature and some have been recently produced, some of which are listed in Table 1 below. For example, anti-MASP-2 Fab2 antibodies which block MASP-2-dependent complement activation have been identified as described in examples 10 and 11 herein. As described in example 12, and in WO2012/151481, which is incorporated herein by reference, fully human MASP-2scFv antibodies (e.g., OMS 646) that block MASP-2-dependent complement activation have been identified. As described in example 13, and in WO2014/144542, which is incorporated herein by reference, MASP-2 antibodies and fragments thereof carrying an SGMI-2 peptide with MASP-2 inhibitory activity are produced by fusion of the SGMI-2 peptide amino acid sequence (SEQ ID NO:72, 73 or 74) to the amino or carboxy terminus of the heavy and/or light chain of a human MASP-2 antibody (e.g., OMS 646-SGMI-2).
Thus, in one embodiment, the MASP-2 inhibitor for use in the methods of the invention comprises a human antibody, such as OMS646. Thus, in one embodiment, the MASP-2 inhibitors useful in the compositions and methods of the invention comprise a human antibody that binds to a polypeptide consisting of human MASP-2 (SEQ ID NO: 6), wherein the antibody comprises: (I) (a) a heavy chain variable region comprising i) a heavy chain CDR-H1 comprising the amino acid sequence of 31-35 of SEQ ID NO 67; and ii) a heavy chain CDR-H2 comprising the amino acid sequence of 50-65 of SEQ ID NO. 67; and iii) a heavy chain CDR-H3 comprising the amino acid sequence of 95-107 of SEQ ID NO. 67 and b) a light chain variable region comprising i) a light chain CDR-L1 comprising the amino acid sequence of 24-34 of SEQ ID NO. 70; and ii) light chain CDR-L2 comprising the amino acid sequence of 50-56 of SEQ ID NO. 70; and iii) light chain CDR-L3 comprising the amino acid sequence of 89-97 of SEQ ID NO. 70, or (II) variants thereof, comprising a heavy chain variable region having at least 90% identity to SEQ ID NO. 67 (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID NO. 67) and a light chain variable region having at least 90% identity to SEQ ID NO. 70 (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity).
In some embodiments, the methods comprise administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO:67 and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 70.
In some embodiments, the methods comprise administering to a subject a composition comprising a MASP-2 inhibitory antibody or antigen binding fragment thereof that specifically recognizes at least a portion of an epitope on human MASP-2 that is recognized by reference antibody OMS646, reference antibody OMS646 comprising the heavy chain variable region set forth in SEQ ID NO:67 and the light chain variable region set forth in SEQ ID NO: 70. In one embodiment, the MASP-2 inhibitor for use in the methods of the invention comprises human antibody OMS646.
Table 1: exemplary MASP-2-specific antibodies
anti-MASP-2 antibodies with reduced effector function
In some embodiments of this aspect of the invention, the anti-MASP-2 antibodies have reduced effectsSub-functions to alleviate inflammation that may result from activation of the classical complement pathway. The ability of IgG molecules to elicit the classical complement pathway has been shown to exist in the Fc portion of the molecule (Duncan, A.R., et al Nature 332:738-7401988). IgG molecules in which the Fc portion of the molecule is removed by enzymatic cleavage lack such effector function (see Harlow, antibodies: A Laboratory Manual, cold Spring Harbor Laboratory, new York, 1988). Thus, it is possible to use a human IgG or a human IgG by having a genetically engineered Fc sequence that minimizes effector function 2 Or IgG 4 Isotype, thereby removing the Fc portion of the molecule, resulting in antibodies with reduced effector function.
Standard molecular biology procedures can be performed on the Fc portion of an IgG heavy chain to generate antibodies with reduced effector function, as described herein, also in Jolliffe et al, int' l Rev. Immunol.10:241-250,1993, and Rodrigues et al, J.Immunol.151:6954-6961,1998. Antibodies with reduced effector function also include human IgG2 and IgG4 isotypes with reduced ability to activate complement and/or interact with Fc receptors (Ravetch, J.V., et al, annu. Rev. Immunol.9:457-492,1991; isaacs, J.D., et al, J.Immunol.148:3062-3071,1992;van de Winkel,J.G, et al, immunol.today 14:215-221,1993). Humanized or fully human antibodies specific for human MASP-2 composed of IgG2 or IgG4 isotypes can be produced by one of several methods known to those of ordinary skill in the art, as described by Vaughan, T.J., et al Nature Biotechnical 16:535-539, 1998.
Production of anti-MASP-2 antibodies
anti-MASP-2 antibodies may be produced using a MASP-2 polypeptide (e.g., full length MASP-2) or using a peptide bearing an antigenic MASP-2 epitope (e.g., a MASP-2 polypeptide portion). Immunogenic peptides can be as few as five amino acid residues. For example, MASP-2 polypeptides comprising the entire amino acid sequence of SEQ ID NO. 6 may be used to induce anti-MASP-2 polypeptides for use in the methods of the invention. Specific MASP-2 domains known to be involved in protein-protein interactions, such as the CUBI and CUBIEGF domains and regions comprising serine protease active sites, can be expressed as recombinant polypeptides as described in example 3 and used as antigens. In addition, peptides comprising at least a portion of the MASP-2 polypeptide (SEQ ID NO: 6) of at least 6 amino acids may also be used to induce MASP-2 antibodies. Additional examples of MASP-2 derived antigens for inducing MASP-2 antibodies are provided in Table 2 below. MASP-2 peptides and polypeptides for producing antibodies may be isolated as natural polypeptides, or recombinant or synthetic peptides, as well as catalytically inactive recombinant polypeptides such as MASP-2A, as further described herein. In some embodiments of this aspect of the invention, transgenic mouse strains are used to obtain anti-MASP-2 antibodies, as described herein.
Antigens useful in the production of anti-MASP-2 antibodies also include fusion polypeptides, such as fusion of MASP-2 or a portion thereof with an immunoglobulin polypeptide or with a maltose binding protein. The polypeptide immunogen may be a full-length molecule or a portion thereof. If the polypeptide moiety is hapten-like, it is preferred that such moiety be conjugated or otherwise linked to a macromolecular carrier such as Keyhole Limpet Hemocyanin (KLH), bovine Serum Albumin (BSA) or tetanus toxoid for immunization.
Table 2: MASP-2 derived antigens
Polyclonal antibodies
Polyclonal antibodies against MASP-2 may be prepared by immunizing an animal with a MASP-2 polypeptide or immunogenic portions thereof using methods well known to those of ordinary skill in the art. See, e.g., green et al, "Production of Polyclonal Antisera," on Immunochemical Protocols (Manson Main, inc.), page 105. Immunogenicity of MASP-2 polypeptides can be increased by the use of adjuvants including inorganic gels (such as aluminum hydroxide) or Freund's adjuvant (complete or incomplete), surfactants (such as lysolecithin), pluronic polyols (pluronic polyols), polyanions, oil emulsions, keyhole limpetHemocyanin and dinitrophenol. Polyclonal antibodies are typically produced by animals such as horses, cattle, dogs, chickens, rats, mice, rabbits, guinea pigs, goats, or sheep. Alternatively, anti-MASP-2 antibodies useful in the invention may be obtained From a primate approximating a human. General techniques for producing diagnostic and therapeutic antibodies in baboons can be found, for example, in International patent publication No. WO 91/11465 to Goldenberg et al and Losman, M.J., et al, int.J.cancer 46:310,1990. Serum containing immunologically active antibodies is then produced from the blood of these immunized animals using standard methods well known in the art. />
Monoclonal antibodies
In some embodiments, the MASP-2 inhibitor is an anti-MASP-2 monoclonal antibody. anti-MASP-2 monoclonal antibodies are highly specific against a single MASP-2 epitope. The modifier "monoclonal" as used herein refers to the nature of the antibody obtained from a population of substantially homogeneous antibodies and is not to be understood as requiring production of the antibody by any particular method. Monoclonal antibodies can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture, such as the hybridoma method described in Kohler, g., et al, nature 256:495,1975, or can be prepared by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567 to cabill). Monoclonal antibodies can also be isolated from phage antibody libraries using techniques described by Clackson, T., et al, nature 352:624-628,1991, and Marks, J.D., et al, J.mol. Biol.222:581-597, 1991. Such antibodies may be of any immunoglobulin class, including IgG, igM, igE, igA, igD and any subclass thereof.
For example, monoclonal antibodies can be obtained by injecting a composition comprising a MASP-2 polypeptide or a portion thereof into a suitable mammal (e.g., a BALB/c mouse). After a predetermined period of time, spleen cells are removed from the mice and suspended in cell culture medium. Spleen cells are then fused with an immortalized cell line to form hybridomas. The hybridomas formed are cultured in cell culture media and screened for their ability to produce anti-MASP-2 monoclonal antibodies. Examples of anti-MASP-2 monoclonal antibody production are further described herein (see also Current Protocols in Immunology, vol.1., john Wiley & Sons, pages 2.5.1-2.6.7,1991.).
Human monoclonal antibodies can be obtained by using transgenic mice that have been engineered to produce specific human antibodies in response to antigen challenge. In this technique, human immunoglobulin heavy and light chain locus elements are introduced into a mouse strain derived from an embryonic stem cell line containing endogenous immunoglobulin heavy and light chain loci that are targeted for disruption. The transgenic mice can be synthesized as human antibodies specific for human antigens, such as the MASP-2 antigen described herein, which can be used to generate hybridomas secreting human MASP-2 antibodies by fusing B cells from these animals with a suitable myeloma cell line using conventional Kohler-Milstein techniques, as further described herein. Transgenic mice with human immunoglobulin genomes are commercially available (e.g., from Abgenix, inc., fremont, CA, and Medarex, inc., annandale, n.j.). Methods for obtaining human antibodies from transgenic mice are described, for example, in Green, l.l., et al, nature genet.7:13,1994; lonberg, n., et al, nature 368:856,1994; and Taylor, L.D., et al, int.Immun.6:579,1994.
Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of established techniques. Such separation techniques include agarose gel affinity chromatography, size exclusion chromatography, and ion exchange chromatography using protein A (see, e.g., coligan, pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; baines et al, "Purification of Immunoglobulin G (IgG)," in Methods in MolecularBiology, the Humana Press, inc., volume 10, pages 79-104, 1992).
Once polyclonal, monoclonal or phage-derived antibodies are produced, their specificity for MASP-2 binding is first determined. Antibodies that specifically bind MASP-2 may be detected using a variety of assays known to those of skill in the art. Exemplary assays include Western blotting or immunoprecipitation assays (described, for example, in Ausubel et al), immunoelectrophoresis, ELISA, dot blotting, inhibition or competition assays, and sandwich assays (described in Harlow and Land, antibodies: A Laboratory Manual, cold Spring Harbor Laboratory Press, 1988) by standard methods. Once antibodies that specifically bind MASP-2 are identified, the ability of an anti-MASP-2 antibody to function as a MASP-2 inhibitor is determined by one of several assays: such as, for example, lectin-specific C4 cleavage assay (described in example 2), C3b deposition assay (described in example 2) or C4b deposition assay (described in example 2).
The affinity of anti-MASP-2 monoclonal antibodies can be readily determined by one of ordinary skill in the art (see, e.g., scatchard, A., NY Acad. Sci.51:660-672, 1949). In one embodiment, the anti-MASP-2 monoclonal antibodies used in the methods of the invention are capable of binding MASP-2 with an affinity of <100nM, preferably <10nM, most preferably <2nM.
Chimeric/humanized antibodies
Monoclonal antibodies useful in the methods of the invention include chimeric antibodies, as well as fragments of such antibodies, in which the heavy and/or light chain portions are identical or homologous to corresponding sequences from antibodies derived from a particular species or belonging to a particular antibody class or subclass, and the remainder of the chain is identical or homologous to corresponding sequences from antibodies derived from another species or belonging to another antibody class or subclass (U.S. Pat. No. 4,816,567 to Casully; and Morrison, S.L., et al, proc.Nat' l Acad.Sci.USA 81:6851-6855,1984).
One form of chimeric antibody useful in the invention is a humanized monoclonal anti-MASP-2 antibody. Humanized versions of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequences derived from non-human immunoglobulins. Humanized monoclonal antibodies are produced by transferring non-human (e.g., mouse) Complementarity Determining Regions (CDRs) from the variable heavy and variable light chains of a mouse immunoglobulin to a human variable region. Then, it is typical practice to substitute the rest of the human antibody into the framework regions of the non-human counterpart. In addition, humanized antibodies may comprise residues that are not present in the recipient antibody or the donor antibody. These modifications are used to further improve antibody performance. Generally, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the Fv framework regions are of a human immunoglobulin sequence. The humanized antibody optionally may comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For more details see Jones, P.T., et al, nature 321:522-525,1986; reichmann, L., et al, nature 332:323-329,1988; and Presta, curr.Op.struct.biol.2:593-596,1992.
Humanized antibodies useful in the present invention include human monoclonal antibodies comprising at least the MASP-2 binding CDRH3 region. Furthermore, the Fc portion may be replaced in order to produce IgA or IgM and human IgG antibodies. These humanized antibodies will have particular clinical utility because they specifically recognize human MASP-2, but will not elicit an immune response in humans against the antibody itself. They are therefore more suitable for in vivo administration in humans, especially when repeated or prolonged administration is necessary.
Examples of humanized anti-MASP-2 antibodies obtained from murine anti-MASP-2 monoclonal antibodies are provided in example 6 herein. The production of humanized monoclonal antibodies is also described, for example, in Jones, P.T., et al, nature 321:522,1986; carter, p., et al, proc.nat' l.acad.sci.usa 89:4285,1992; sandhu, J.S., crit.Rev.Biotech.12:437,1992; singer, i.i., et al, j.immun.150:2844,1993; sudhir (master), antibody Engineering Protocols, humana Press, inc.,1995; kelley, "Engineering Therapeutic Antibidies," in Protein Engineering: principles andPractice, cleland et al (Main plaited), john Wiley & Sons, inc., pages 399-434, 1996; and U.S. Pat. No. 5,693,762 to Queen, (1997). In addition, there are commercial entities such as Protein Design Labs (Mountain View, CA) that synthesize humanized antibodies from specific murine antibody regions.
Recombinant antibodies
Recombinant methods can also be used to produce anti-MASP-2 antibodies. For example, human immunoglobulin expression libraries (available from, e.g., stratagene, corp., la Jolla, calif.) can be used to generate human antibody fragments (V H 、V L Fv, fd, fab or F (ab') 2 ) To prepare human antibodies. These fragments are then used to construct fully human antibodies using techniques similar to those used to generate chimeric antibodies.
Anti-idiotype antibody
Once anti-MASP-2 antibodies having the desired inhibitory activity are identified, these antibodies can be used to generate anti-idiotype antibodies that resemble partial MASP-2 using techniques well known in the art. See, for example, greenspan, N.S., et al, FASEBJ.7:437,1993. For example, antibodies that bind MASP-2 and competitively inhibit the interaction of MASP-2 proteins required for complement activation can be used to generate anti-idiotype antibodies that resemble the MBL binding site on MASP-2 proteins, thereby binding and neutralizing binding ligands of MASP-2, such as, for example, MBL.
Immunoglobulin fragments
MASP-2 inhibitors useful in the methods of the invention include not only intact immunoglobulin molecules, but also well-known fragments, including Fab, fab', F (ab) 2 、F(ab') 2 And Fv fragments, scFv fragments, diabodies, linear antibodies, single chain antibody molecules, and multispecific antibodies formed from antibody fragments.
It is well known in the art that only a small part of the antibody molecules, paratopes, are involved in the binding of antibodies to their epitopes (see e.g. Clark, w.r., the Experimental Foundations ofModern Immunology, wiley&Sons, inc., NY, 1986). The pFc' and Fc regions of antibodies are effectors of the classical complement pathway, but are not involved in antigen binding. Antibodies in which the pFC ' region has been cleaved by an enzyme, or antibodies produced without the pFC ' region, are referred to as F (ab ') 2 Fragments which retain the antigen binding site of the intact antibody. Isolated F (ab') 2 Fragments are referred to as bivalent monoclonal fragments due to the presence of two antigen binding sites. Similarly, antibodies in which the Fc region has been cleaved by an enzyme, or antibodies produced without an Fc region, are referred to as Fab fragments, which retain one antigen binding site of the intact antibody molecule.
Antibody fragments may be obtained by proteolysis, such as digestion of the intact antibody by pepsin or papain by conventional methods. For example, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin, thereby providing a polypeptide called F (ab') 2 Is a 5S fragment of (C). This fragment can be cleaved again using thiol reducing reagents to give the 3.5S Fab' monovalent fragment. The cleavage reaction may optionally be carried out using a capping group for the sulfhydryl group generated by disulfide cleavage. As an alternative, enzymatic cleavage using pepsin directly produces two monovalent Fab fragments A fragment and an Fc fragment. Such methods are described, for example, in U.S. Pat. nos. 4,331,647 to Goldenberg; nisonoff, A., et al, arch. Biochem. Biophys.89:230,1960; porter, R.R., biochem.J.73:119,1959; edelman et al, supra, methods in Enzymology 1:422,Academic Press,1967; and pages 2.8.1-2.8.10 and pages 2.10..about. 2.10.4 of Coligan.
In some embodiments, it is preferred to use antibody fragments that lack an Fc region to avoid activation of the classical complement pathway after Fc binds to fcγ receptors. There are several methods to generate moabs that avoid interactions with fcγ receptors. For example, the Fc region of a monoclonal antibody can be removed chemically by partial digestion with proteolytic enzymes (e.g., ficin digestion), thereby producing, for example, an antigen-binding antibody fragment, such as Fab or F (ab) 2 Fragment (Mariani, M., et al, mol. Immunol.28:69-71,1991). Alternatively, human gamma 4IgG isotypes that do not bind fcgamma receptor may be used during construction of the humanized antibodies described herein. Antibodies lacking Fc domains, single chain antibodies, and antigen binding domains can also be engineered using the recombinant techniques described herein.
Single chain antibody fragments
Alternatively, single peptide chain binding molecules specific for MASP-2 may be prepared in which the heavy and light chain Fv regions are linked. Fv fragments may be joined by a peptide linker to form a single chain antigen binding protein (scFv). By constructing a vector comprising the code V H And V L The structural genes of the DNA sequences of the domains are used to prepare these single-chain antigen-binding proteins, with the domains being linked by oligonucleotides. The structural gene is inserted into an expression vector, which is then introduced into a host cell (such as E.coli). Recombinant host cells synthesize a single polypeptide chain bridging two V domains by a linker peptide. Methods for the preparation of scFv are described, for example, in Whitlow et al, "Methods: A Companion to Methods in Enzymology"2:97,1991; bird, et al, science 242:423,1988; U.S. Pat. nos. 4,946,778 to Ladner; pack, P., et al, bio/Technology 11:1271,1993.
For example, MASP-2 specific scFv can be obtained by exposing lymphocytes to MASP-2 polypeptides in vitro and selecting an antibody display library in a phage or similar vector (e.g., by using immobilized or labeled MASP-2 proteins or peptides). Genes encoding polypeptides having possible MASP-2 polypeptide binding domains can be obtained by screening random peptide libraries displayed on phage or bacteria, such as E.coli. These random peptide display libraries can be used to screen peptides that interact with MASP-2. Techniques for constructing and screening such random peptide display libraries are well known in the art (U.S. Pat. No. 5,223,409 to Lardner; U.S. Pat. No. 4,946,778 to Lardner; U.S. Pat. No. 5,403,484 to Lardner; U.S. Pat. No. 5,571,698 to Kay et al, phage Display of Peptides and Proteins Academic Press, inc., 1996), and random peptide display libraries and kits for screening such libraries are commercially available, for example, from CLONTECH Laboratories, inc. (Palo Alto, calif.), invitrogen Inc. (San Diego, calif.), new England Biolabs, inc. (Ifswick, mass.).
Another form of anti-MASP-2 antibody fragment useful in this aspect of the invention is a peptide encoding a single Complementarity Determining Region (CDR) that binds to an epitope of the MASP-2 antigen and inhibits MASP-2 dependent complement activation. CDR peptides ("minimal recognition units") can be obtained by constructing genes encoding the CDRs of the antibody of interest. These genes can be prepared, for example, by synthesizing variable regions from RNA of antibody-producing cells using the polymerase chain reaction (see, e.g., larrick et al, methods: A Companion to Methods in Enzymology 2:106,1991; courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in Monoclonal Antibodies: production, engineering and Clinical Application, ritter et al (Main plaited), pages 166, cambridge University Press,1995; and Ward et al, "Genetic Manipulation and Expression of Antibodies," in Monoclonal Antibodies: principles and Applications, birch et al (Main plaited), page 137, wiley-Lists, inc., 1995)).
The MASP-2 antibodies described herein are administered to a subject in need thereof to inhibit MASP-2 dependent complement activation. In some embodiments, the MASP-2 inhibitor is a high affinity human or humanized monoclonal anti-MASP-2 antibody with reduced effector function.
Peptide inhibitors
In some embodiments of this aspect of the invention, the MASP-2 inhibitor comprises an isolated MASP-2 peptide inhibitor, which comprises an isolated natural peptide inhibitor and a synthetic peptide inhibitor that inhibit the MASP-2 dependent complement activation system. The term "isolated MASP-2 peptide inhibitor" as used herein refers to a peptide that inhibits MASP-2-dependent complement activation by binding to MASP-2, competing with MASP-2 for binding to other recognition molecules in the lectin pathway (e.g., MBL, H-fiber gelator, M-fiber gelator, or L-fiber gelator), and/or interacting directly with MASP-2 to inhibit MASP-2-dependent complement activation, the peptide being substantially purified of other substances that are found in nature therewith to the extent practical and suitable for its intended use.
Peptide inhibitors have been used to successfully interfere with the interactions and catalytic sites between proteins in vivo. For example, recently, peptide inhibitors of adhesion molecules structurally related to LFA-1 have been approved for clinical use in coagulopathies (ohm an, E.M., et al, european Heart J.16:50-55,1995). Studies have revealed that short linear peptides (< 30 amino acids) prevent or interfere with integrin-dependent adhesion (Murayama, O., et al J.biochem.120:445-51, 1996). Longer peptides ranging in length from 25-200 amino acid residues were also used, successfully blocking integrin-dependent adhesion (Zhang, l., et al, j. Biol. Chem.271 (47): 29953-57, 1996). In general, longer peptide inhibitors have higher affinity and/or slower dissociation rates than short peptides and are therefore more potent inhibitors. Studies have also shown that cyclic peptide inhibitors are potent in vivo integrin inhibitors for the treatment of human inflammatory diseases (Jackson, D.Y., et al, J.Med. Chem.40:3359-68, 1997). One method of producing cyclic peptides involves the synthesis of peptides in which the terminal amino acids of the peptide are cysteines, thereby enabling the peptide to exist in cyclic form through disulfide bonds between the terminal amino acids, which has been demonstrated to improve affinity and in vivo half-life when used in the treatment of hematopoietic tumors (e.g., U.S. patent No. 6,649,592 to Larson).
Synthetic MASP-2 peptide inhibitors
MASP-2 inhibitory peptides for use in the methods of this aspect of the invention are exemplified by mimicking the amino acid sequence of a target region important for MASP-2 function. The inhibitory peptides used in practicing the methods of the invention range in size from about 5 amino acids to about 300 amino acids. Table 3 provides a list of exemplary inhibitory peptides useful in practicing this aspect of the invention. The ability of a candidate MASP-2 inhibitory peptide to function as a MASP-2 inhibitor may be assayed by one of several assays, including, for example, lectin-specific C4 cleavage assays (see example 2) and C3b deposition assays (see example 2).
In some embodiments, the MASP-2 inhibitory peptide is derived from a MASP-2 polypeptide and is selected from the full length mature MASP-2 protein (SEQ ID NO: 6), or from a specific domain of a MASP-2 protein, such as, for example, the CUBI domain (SEQ ID NO: 8), the CUBIEGF domain (SEQ ID NO: 9), the EGF domain (SEQ ID NO: 11), and the serine protease domain (SEQ ID NO: 12). As previously described, studies have shown that the CUBEGFCUBII region is required for dimerization and binding to MBL (Thielens et al, supra). In particular, studies to identify human subjects carrying Asp105 to Gly105 homozygous mutations have demonstrated that the peptide sequence TFRSDYN (SEQ ID NO: 16) in the CUBI domain of MASP-2 is involved in binding MBL, which mutations result in the loss of MASP-2 from the MBL complex (Stengaard-Pedersen, K., et al, new England J.Med.349:554-560, 2003).
In some embodiments, the MASP-2 inhibitory peptide is derived from a lectin protein that binds MASP-2 and participates in the lectin complement pathway. Several different lectins involved in this pathway have been identified, including mannan-binding lectin (MBL), L-fiber-gelling protein, M-fiber-gelling protein and H-fiber-gelling protein (Ikeda, K., et al, J.biol. Chem.262:7451-7454,1987; matsushita, M., et al, J.exp. Med.176:1497-2284,2000; matsushita, M., et al, J.Immunol.168:3502-3506, 2002). These lectins are present in serum as oligomers of homotrimeric subunits, each with N-terminal collagen-like fibers with carbohydrate recognition domains. These different lectins have been shown to bind MASP-2 and the lectin/MASP-2 complex activates complement by cleaving proteins C4 and C2. H-fiber gelator has an amino terminal region of 24 amino acids, a collagen-like domain with 11 Gly-Xaa-Yaa repeats, a neck domain of 12 amino acids and an fibrinogen-like domain of 207 amino acids (Matsushita, M., et al J.Immunol.168:3502-3506, 2002). H-fiber gellin binds GlcNAc and aggregates human erythrocytes coated with LPS from Salmonella typhimurium (S.tyrphimum), salmonella minnesota (S.minnesota) and E.coli. H-fiber gelator has been shown to bind MASP-2 and MAP19 and activate the lectin pathway, supra. L-fiber gelator/P35 also binds GlcNAc, has been shown to bind MASP-2 and MAp19 in human serum, and this complex has been shown to activate the lectin pathway (Matsushita, M., et al, J.Immunol.164:2281,2000). Thus, MASP-2 inhibitory peptides for use in the present invention may comprise a region of at least 5 amino acids selected from the group consisting of MBL protein (SEQ ID NO: 21), H-fiber gelling protein (Genbank accession No. NM-173452), M-fiber gelling protein (Genbank accession No. O00602) and L-fiber gelling protein (Genbank accession No. NM-015838).
More specifically, scientists have identified MASP-2 binding sites on MBL as being in 12 Gly-X-Y triplets "GKD GRD GTK GEK GEP GQG LRG LQG POG KLG POGNOG PSG SOG PKG QKG DOG KS" (SEQ ID NO: 26) located between the hinge and neck of the C-terminal portion of the MBP collagen-like domain (Wallis, R., et al, J.biol. Chem.279:14065,2004). The MASP-2 binding site region is also highly conserved in human H-fiber gel protein and human L-fiber gel protein. Studies have indicated that there is a consensus binding site in all three lectin proteins comprising the amino acid sequence "OGK-X-GP" (SEQ ID NO: 22), in which the letter "O" represents hydroxyproline and the letter "X" represents a hydrophobic residue (Wallis et al, 2004, supra). Thus, in some embodiments, MASP-2 inhibitory peptides useful in this aspect of the invention are at least 6 amino acids in length and comprise SEQ ID NO. 22. Peptides comprising the amino acid sequence "GLR GLQ GPO GKL GPO G" (SEQ ID NO: 24) and derived from MBL have been shown to bind MASP-2 in vitro (Wallis et al, 2004, supra). To enhance binding to MASP-2, peptides can be synthesized that abut two GPO triplets at each end ("GPO GPO GLR GLQ GPO GKL GPO GGP OGP O" SEQ ID NO: 25) to enhance the formation of the triple helix found in the native MBL protein (see Wallis, R., et al, J. Biol. Chem.279:14065,2004 for further description).
MASP-2 inhibitory peptides may also be derived from human H-fiber gelator protein, which includes the sequence "GAO GSO GEK GAO GPQ GPO GPO GKM GPK GEO GDO" (SEQ ID NO: 27) from the consensus MASP-2 binding region of H-fiber gelator protein. Also included are peptides derived from human L-fiber gel protein, which include the sequence "GCO GLO GAO GDK GEAGTN GKR GER GPO GPO GKA GPO GPN GAO GEO" (SEQ ID NO: 28) from the consensus MASP-2 binding region of L-fiber gel protein.
MASP-2 inhibitory peptides may also be derived from C4 cleavage sites, such as the C4 cleavage site "LQRALEILPNRVTIKANRPFLVFI" (SEQ ID NO: 29) (Glover, G.I., et al mol. Immunol.25:1261 (1988)) linked to the C-terminal portion of antithrombin III.
Table 3: exemplary MASP-2 inhibitory peptides
/>
Note that: the letter "O" represents hydroxyproline. The letter "X" is a hydrophobic residue.
Peptides derived from the C4 cleavage site, as well as other peptides that inhibit the MASP-2 serine protease site, may be chemically modified to render them irreversible protease inhibitors. For example, suitable modifications may include, but are not necessarily limited to, C-terminal, asp or Glu, or halomethyl ketone added to a functional side chain (Br, cl, I, F); haloacetyl (or other alpha-haloacetyl) groups on amino or other functional side chains; epoxy or imine-containing groups at the amino or carboxyl terminus or on functional side chains; or imidoesters at the amino or carboxyl terminus or on other functional side chains. These modifications provide the advantage of permanently inhibiting the enzyme by covalent binding of the peptide. This may result in lower effective doses and/or less frequent dosing of the peptide inhibitor.
In addition to the inhibitory peptides described above, MASP-2 inhibitory peptides for use in the methods of the invention include peptides comprising the MASP-2 binding CDRH3 region of an anti-MASP-2 MoAb obtained as described herein. The sequence of the CDR regions for the synthetic peptides can be determined by methods known in the art. The heavy chain variable region is a peptide typically ranging from 100 to 150 amino acids in length. The light chain variable region is a peptide typically ranging from 80 to 130 amino acids in length. CDR sequences within the heavy and light chain variable regions comprise approximately only 3-25 amino acid sequences, which can be readily sequenced by one of ordinary skill in the art.
It will be appreciated by those skilled in the art that substantially homologous variations of the MASP-2 inhibitory peptides described above may also have MASP-2 inhibitory activity. Exemplary variations include, but are not necessarily limited to, peptides having amino acids inserted, deleted, substituted and/or added at the carboxy-terminal or amino-terminal portion of the subject peptide, and mixtures thereof. Thus, we consider these cognate peptides having MASP-2 inhibitory activity to be useful in the methods of the invention. The peptides may also include replication motifs and other modifications with conservative substitutions. Other parts of this document describe conservative variants, including the exchange of one amino acid for another amino acid having the same charge, size, or hydrophobicity, etc.
MASP-2 inhibitory peptides may be modified to increase solubility and/or maximize positive or negative charge in order to more closely resemble segments of an intact protein. The derivatives may have or may not have the exact primary amino acid structure of the peptides disclosed herein, provided that the derivatives still retain the desired MASP-2 inhibitory properties functionally. The modification may include amino acid substitution, i.e. substitution with one or another of the 20 commonly known amino acids, substitution with a derivatized or substituted amino acid with a desired characteristic (such as resistance to enzymatic degradation) or substitution with a D amino acid, or substitution with another molecule or compound (such as a carbohydrate) that mimics the natural conformation and function of one or more amino acids or peptides; amino acid deletion; amino acid insertions, i.e. insertions of one or another of the 20 amino acids generally known, insertions of derivatized or substituted amino acids or insertions of D amino acids to which a desired characteristic (such as resistance to enzymatic degradation) is attached, or substitutions by further molecules or compounds (such as carbohydrates) which mimic the natural conformation and function of one or more amino acids or peptides; or by another molecule or compound (such as a carbohydrate or nucleic acid monomer) that mimics the native conformation, charge distribution and function of the parent peptide. Peptides may also be modified by acetylation or amidation.
The derivatized inhibitory peptides may be synthesized according to known techniques of peptide biosynthesis, carbohydrate biosynthesis, and the like. Initially, the skilled person can determine the conformation of the target peptide according to a suitable computer program. Once the conformation of the peptides disclosed herein is known, the skilled artisan can then determine in a rational design manner what class of substitutions can be made at one or more sites so that the resulting derivative retains the basic conformation and charge distribution of the parent peptide, but may possess characteristics that are not present in the parent peptide or that are superior to those present in the parent peptide. Once candidate derived molecules are identified, the derivatives can be assayed using the assays described herein to determine whether they function as MASP-2 inhibitors.
Screening of MASP-2 inhibitory peptides
Molecular modeling and rational molecular design can also be used to generate and screen peptides that mimic the molecular structure of the key binding region of MASP-2 and inhibit complement activity of MASP-2. The molecular structure used for modeling includes the CDR regions of anti-MASP-2 monoclonal antibodies, as well as regions of interest known to be important for MASP-2 function, including the regions required for dimerization as described previously, the regions involved in binding MBL, and serine protease active sites. Methods for identifying peptides that bind to a particular target are well known in the art. For example, molecular imprinting can be used to construct macromolecular structures from scratch, such as peptides that bind to specific molecules. See, e.g., shea, k.j., "Molecular Imprinting of Synthetic Network Polymers: the De Novo synthesis ofMacromolecular Binding and Catalytic Sties," TRIP2 (5) 1994.
For example, one method of preparing MASP-2 binding peptide mimetics is as follows. Functional monomers (templates) of known MASP-2 binding peptides or binding regions of anti-MASP-2 antibodies with MASP-2 inhibitory effect are polymerized. The template is then removed and the second type of monomer is then polymerized in the voids left by the template to give new molecules having one or more desired properties similar to those of the template. In addition to preparing peptides in this manner, other MASP-2 binding molecules may be prepared that act as MASP-2 inhibitors, such as polysaccharides, nucleosides, drugs, nucleoproteins, lipoproteins, carbohydrates, glycoproteins, steroids, lipids, and other bioactive materials. The method is suitable for designing a wide variety of biological mimics that are more stable than their natural counterparts, as they are typically formed by free radical polymerization of functional monomers, resulting in compounds with a biologically non-degradable backbone.
Peptide synthesis
MASP-2 inhibitory peptides may be prepared using techniques well known in the art, such as the solid phase synthesis techniques originally proposed by Merrifield (Merrifield, J. Amer. Chem. Soc.85:2149-2154, 1963). For example, automated synthesis may be accomplished using a Applied Biosystems 431A peptide synthesizer (Foster City, calif.) according to the instructions provided by the manufacturer. Other techniques are described, for example, in Bodanszky, M., et al, peptide synthesis, second edition, john Wiley & Sons,1976, and other references known to those skilled in the art.
Peptides may also be prepared using standard genetic engineering techniques known to those skilled in the art. For example, a peptide can be prepared by inserting a nucleic acid encoding the peptide into an expression vector by an enzymatic method, expressing the DNA, and translating the DNA into the peptide in the presence of the desired amino acid. The peptide is then purified using chromatographic or electrophoretic techniques, or by inserting the sequence encoding the peptide into a carrier protein in synchronization with the nucleic acid sequence encoding the carrier protein, which carrier protein may be fused to the peptide and subsequently excised from the peptide. Protein-peptide fusions can be isolated using chromatographic techniques, electrophoretic techniques, or immunological techniques (such as binding to a resin via antibodies to carrier proteins). The peptides may be cleaved using chemical or enzymatic methods (e.g., by hydrolytic enzymes).
MASP-2 inhibitory peptides for use in the methods of the invention may also be produced by recombinant host cells according to conventional techniques. In order to express MASP-2 inhibitory peptide coding sequences, the nucleic acid molecule encoding the peptide must be operably linked to regulatory sequences that control the transcriptional expression of the expression vector and then introduced into a host cell. In addition to transcriptional regulatory sequences (such as promoters and enhancers), expression vectors can include translational regulatory sequences and marker genes suitable for selection of cells carrying the expression vector.
Nucleic acid molecules encoding MASP-2 inhibitory peptides may be synthesized using a "genetic apparatus" and using phosphoramidite methods, among other methods. If chemically synthesized double-stranded DNA is required in applications such as gene or gene fragment synthesis, each complementary strand is prepared separately. The production of short genes (60-80 base pairs) is technically simple and easy to implement by synthesizing complementary strands and then annealing them. For the production of longer genes, synthetic genes (double-stranded) are assembled in modular form from single-stranded fragments of 20-100 nucleotides in length. For reviews of polynucleotide synthesis see, e.g., glick and masternak, "Molecular Biotechnology, principles andApplications ofRecombinantDNA", ASM Press,1994; itakura, K. Et al, annu. Rev. Biochem.53:323,1984; and Climie, S.et al, proc.Nat' l Acad.Sci.USA 87:633,1990.
Small molecule inhibitors
In some embodiments, the MASP-2 inhibitor is a small molecule inhibitor, including natural and synthetic substances having low molecular weight, such as, for example, peptides, peptidomimetics, and non-peptide inhibitors (including oligonucleotides and organic compounds). Small molecule inhibitors of MASP-2 may be produced based on the molecular structure of the variable region of an anti-MASP-2 antibody.
Small molecule inhibitors may also be designed and generated using in silico drug design based on the crystal structure of MASP-2 (Kuntz i.d., et al Science 257:1078, 1992). The crystal structure of rat MASP-2 has been demonstrated (Feinberg, H., et al, EMBOJ.22:2348-2359, 2003). Using the method described by Kuntz et al, MASP-2 crystal structure coordinates are entered into a computer program (such as DOCK) that will output a series of small molecule structures that are expected to bind MASP-2. The use of such computer programs is well known to those skilled in the art. For example, using the crystal structure of HIV-1 protease inhibitors, unique non-peptide ligands as HIV-1 protease inhibitors were identified by evaluating the match of compounds to enzyme binding sites as found in Cambridge crystallography database (Cambridge Crystallographic database) using the procedure DOCK (Kuntz, I.D., et al, J.mol. Biol.161:269-288,1982; desJarlais, R.L., et al, PNAS87:6644-6648, 1990).
A series of small molecule structures identified in silico as potential MASP-2 inhibitors were screened using a MASP-2 binding assay such as described in example 10. Small molecules determined to bind MASP-2 are then analyzed in a functional assay such as that described in example 2 to determine whether they inhibit MASP-2 dependent complement activation.
MASP-2 soluble receptors
Other suitable inhibitors of MASP-2 are believed to include MASP-2 soluble receptors and may be produced using techniques known to those of ordinary skill in the art.
Inhibitors of MASP-2 expression
In another embodiment of this aspect of the invention, the MASP-2 inhibitor is an inhibitor of MASP-2 expression that is capable of inhibiting MASP-2 dependent complement activation. In the practice of this aspect of the invention, representative inhibitors of MASP-2 expression include MASP-2 antisense nucleic acid molecules (such as antisense mRNA, antisense DNA or antisense oligonucleotides), MASP-2 ribozymes, and MASP-2RNAi molecules.
Antisense RNA and antisense DNA molecules act to directly block MASP-2mRNA translation by hybridizing to MASP-2mRNA and preventing translation of MASP-2 protein. The antisense nucleic acid molecule can be constructed in many different ways so long as it is capable of interfering with the expression of MASP-2. For example, antisense nucleic acid molecules can be constructed by reversing the coding region (or portion thereof) of MASP-2cDNA (SEQ ID NO: 4) relative to its normal transcription direction for transcription of its complement.
The antisense nucleic acid molecule is generally substantially identical to at least a portion of one or more target genes. However, the nucleic acids need not be identical to inhibit expression. Higher homology can generally be used to compensate for the use of shorter antisense nucleic acid molecules. The minimum percent identity is typically above about 65%, but higher percent identity may exert a more effective repression of expression of the endogenous sequence. A greater percentage identity of substantially greater than about 80% is generally preferred, although about 95% to exactly the same is generally most preferred.
The antisense nucleic acid molecule need not have the same intron or exon pattern as the target gene, and the non-coding segment of the target gene may be equivalent to the coding segment in obtaining antisense inhibition of target gene expression. DNA sequences of at least about 8 nucleotides may be used as antisense nucleic acid molecules, although longer sequences are preferred. In the present invention, a representative example of a useful MASP-2 inhibitor is an antisense MASP-2 nucleic acid molecule which has at least 90% identity to the complement of MASP-2cDNA consisting of the nucleic acid sequence set forth in SEQ ID NO. 4. The nucleic acid sequence shown in SEQ ID NO. 4 encodes a MASP-2 protein consisting of the amino acid sequence shown in SEQ ID NO. 5.
Targeting of antisense oligonucleotides to MASP-2mRNA is another mechanism that may be used to reduce the level of MASP-2 protein synthesis. For example, synthesis of polygalacturonase and muscarinic acetylcholine type 2 receptors is inhibited by antisense oligonucleotides directed against their corresponding mRNA sequences (Cheng, U.S. patent No. 5,739,119 and Shewmaker, U.S. patent No. 5,759,829). In addition, the use of nucleoprotein cyclin, multidrug resistance gene (MDG 1), ICAM-1, E-selectin, STK-1, striatal GABA A Examples of antisense inhibition are demonstrated by receptor and human EGF (see, e.g., U.S. Pat. No. 5,801,154 to Baracchini; U.S. Pat. No. 5,789,573 to Baker; U.S. Pat. No. 5,718,709 to Considine; and U.S. Pat. No. 5,610,288 to Reubenstein).
The literature describes a method that enables one of ordinary skill to determine which oligonucleotides are useful in the systems of the invention, including the use of RNase H cleavage as an indicator of sequence accessibility in transcripts to detect appropriate sites of target mRNA. Scherr, M., et al, nucleic Acids Res.26:5079-5085,1998; lloyd, et al, nucleic Acids Res.29:3665-3673,2001. An antisense oligonucleotide mixture complementary to certain regions of MASP-2 transcript is added to a MASP-2 expressing cell extract (such as hepatocytes) and hybridized to create sites that are vulnerable to RNase H. The method may be combined with computer-assisted sequence selection that predicts optimal sequence selection for antisense composition based on the relative ability of the sequences to form dimers, hairpin structures, or other secondary structures that reduce or inhibit specific binding to host cell target mRNA. These secondary structural analyses and target site selection considerations can be performed using OLIGO primer analysis software (Rychlik, i., 1997) and BLASTN 2.0.5 algorithm software (Altschul, s.f., et al, nucleic acids res.25:3389-3402, 1997). The antisense compound to the target sequence preferably comprises about 8 to about 50 nucleotides in length. Antisense oligonucleotides comprising about 9 to about 35 nucleotides are particularly preferred. The inventors believe that all oligonucleotide compositions ranging from 9 to 35 nucleotides (i.e., nucleotides around 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 bases in length) are highly preferred for practicing the antisense oligonucleotide-based methods of the invention. A highly preferred MASP-2mRNA target region is a region located at or adjacent to the AUG translation initiation codon, and a sequence which is substantially complementary to the 5' region of the mRNA, for example a sequence between the-10 and +10 region of the nucleotide sequence of the MASP 2 gene (SEQ ID NO: 4). Exemplary inhibitors of MASP-2 expression are shown in Table 4.
Table 4: exemplary inhibitors of MASP-2 expression
As described above, the term "oligonucleotide" as used herein refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or a mimetic thereof. The term also includes those oligonucleobases (oligonucleobases) consisting of naturally occurring nucleotides, sugars and internucleoside (backbone) covalent bonds, and oligonucleotides having non-naturally occurring modifications. These modifications enable the introduction of certain desirable properties not provided by naturally occurring oligonucleotides, such as reduced toxicity, improved stability against nuclease degradation, and enhanced cellular uptake. In exemplary embodiments, antisense compounds of the invention differ from the native DNA in that modification of the phosphodiester backbone extends the lifetime of the antisense oligonucleotide, wherein the phosphate substituent is replaced with a phosphorothioate. Likewise, one or both ends of the oligonucleotide may be substituted with one or more acridine derivatives interposed between adjacent base pairs in the nucleic acid strand.
Another alternative to the antisense approach is to use "RNA interference" (RNAi). Double-stranded RNA (dsRNA) can cause gene silencing in mammals. The natural function and co-suppression of RNAi appears to protect the genome from viral attack by mobile genetic elements such as retrotransposons, which when activated produce abnormal RNA or dsRNA in the host cell (see, e.g., jensen, J., et al, nat. Genet.21:209-12, 1999). Double-stranded RNA molecules can be prepared by synthesizing two RNA strands capable of forming a double-stranded RNA molecule, each strand being about 19-25 (e.g., 19-23) nucleotides in length. For example, a dsRNA molecule for use in the methods of the invention may include RNAs corresponding to the sequences listed in table 4 and their complements. Preferably at least one RNA strand has a 3' overhang of 1-5 nucleotides. The synthesized RNA strands are combined under conditions that form a double stranded molecule. The RNA sequence may comprise a portion of at least 8 nucleotides of SEQ ID NO. 4, with a total length of 25 nucleotides or less. The design of siRNA sequences for a particular target is well within the skill of one of ordinary skill in the art. Commercial services (Qiagen, valencia, calif.) are available that design siRNA sequences and ensure expression with at least 70% knockdown.
The dsRNA can be administered as a pharmaceutical composition and performed in a known manner, wherein the nucleic acid is introduced into the desired target cell. Commonly used gene transfer methods include calcium phosphate, DEAE-dextran, electroporation, microinjection and viral methods. These methods are taught in Ausubel et al Current Protocols in Molecular Biology, john Wiley & Sons, inc.
Ribozymes may also be used to reduce the amount and/or biological activity of MASP-2, such as ribozymes that target MASP-2 mRNA. Ribozymes are catalytic RNA molecules capable of cleaving nucleic acid molecules having sequences that are wholly or partially homologous to the ribozyme sequence. Ribozyme transgenes can be designed that encode RNA ribozymes that specifically pair with the target RNA and cleave the phosphodiester backbone at specific locations, thereby functionally inactivating the target RNA. In performing such cleavage, the ribozyme itself is not altered, and thus is capable of recycling and cleaving other molecules. The inclusion of a ribozyme sequence in the antisense RNA confers activity on the antisense RNA to cleave the RNA, thereby increasing the activity of the antisense construct.
Ribozymes useful in the practice of the present invention generally comprise a hybridization region of at least about 9 nucleotides complementary to at least a portion of the nucleotide sequence of a target MASP-2mRNA and a catalytic region suitable for cleavage of the target MASP-2mRNA (see generally EPA No. 032951; WO88/04300; haseloff, J. Et al, nature 334:585-591,1988; fedor, M.J. Et al, proc.Natl.Acad.Sci. USA 87:1668-1672,1990; cech, T.R. Et al, ann.Rev.biochem.55:599-629, 1986).
Ribozymes can be incorporated into the form of RNA oligonucleotides of the ribozyme sequence to target cells directly or as expression vectors encoding the desired ribozyme RNA. Ribozymes can be used and applied in much the same manner as described for antisense polynucleotides.
Antisense RNA and DNA, ribozymes, and RNAi molecules useful in the methods of the present invention can be prepared by any method known in the art for the synthesis of DNA and RNA molecules. These methods include chemical synthesis techniques for oligodeoxyribonucleotides and oligoribonucleotides well known in the art, such as solid phase phosphoramidite chemical synthesis. Alternatively, the RNA molecules can be produced by in vitro and in vivo transcription of DNA sequences encoding antisense RNA molecules. Such DNA sequences may be incorporated into a wide variety of vectors into which a suitable RNA polymerase promoter (such as the T7 or SP6 polymerase promoter) is inserted. Alternatively, antisense cDNA constructs which synthesize antisense RNA constitutively or inducibly depending on the promoter used can be stably introduced into the cell line.
Various well-known modifications of DNA molecules can be introduced to increase stability and half-life. Useful modifications include, but are not limited to, adding ribonucleotide or deoxyribonucleotide flanking sequences to the 5' and/or 3' end of the molecule, or using phosphorothioates or 2' O-methyl instead of phosphodiester linkages within the oligodeoxyribonucleotide backbone.
V. pharmaceutical compositions and methods of delivery
Administration of drugs
In another aspect, the invention provides a composition for inhibiting side effects of MASP 2-dependent complement activation in a subject having a disease or condition described herein, comprising administering to the subject a composition comprising a therapeutically effective amount of a MASP-2 inhibitor and a pharmaceutically acceptable carrier. A therapeutically effective dose of a MASP-2 inhibitor may be administered to a subject in need thereof in an amount that treats or ameliorates a disease associated with MASP-2 dependent complement activation. A therapeutically effective dose refers to an amount of a MASP-2 inhibitor sufficient to result in an improvement in symptoms associated with a disease or condition.
Toxicity and therapeutic efficacy of MASP-2 inhibitors may be determined by standard pharmaceutical methods using experimental animal models, such as the murine MASP-2-/-mouse model described in example 1 that expresses a human MASP-2 transgene. Using these animal models, NOAEL (no observed side effect levels) and MED (minimum effective dose) can be determined using standard methods. The dose ratio between NOAEL/MED effects is the therapeutic ratio, expressed as NOAEL/MED ratio. Most preferred are MASP-2 inhibitors with high therapeutic rates or indices. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of MASP-2 inhibitor is preferably within a range of circulating concentrations, including MED that is practically non-toxic or non-toxic. The dosage may vary within this range depending upon the dosage form employed and the route of administration used.
In some embodiments, the therapeutic efficacy of a MASP-2 inhibitor for treating, inhibiting, reducing, or preventing fibrosis in a mammalian subject having or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation is determined by one or more of: reduction of one or more markers of inflammation and scarring (e.g., tgfβ -1, CTFF, IL-6, apoptosis, fibronectin, laminin, collagen, EMT, infiltrating macrophages) in kidney tissue; soluble markers of inflammatory and fibrotic kidney disease are released into urine and plasma reduction (e.g., by measuring renal excretion function).
For any compound formulation, an animal model can be used to evaluate a therapeutically effective dose. For example, doses up to a range of circulating plasma concentrations including MED can be formulated in animal models. Quantitative levels of MASP-2 inhibitors in plasma may also be determined, for example, by high performance liquid chromatography.
In addition to toxicity studies, effective dosages may also be estimated based on the amount of MASP-2 protein present in the living subject and the binding affinity of the MASP-2 inhibitor. MASP-2 levels present in serum from normal subjects have been shown to be in the low level range of 500ng/ml and MASP-2 quantitative assays can be used to determine MASP-2 levels in specific subjects (described in Moller-Kristensen M., et al J.Immunol. Methods 282:159-167,2003).
The dosage of a composition comprising a MASP-2 inhibitor administered will generally vary depending on the age, weight, height, sex, general medical condition and medical history of the subject. For example, MASP-2 inhibitors, such as anti-MASP-2 antibodies, may be administered in a dosage range of about 0.010-10.0mg/kg, preferably 0.010-1.0mg/kg, more preferably 0.010-0.1mg/kg of the subject's body weight. In certain embodiments, the compositions comprise a combination of an anti-MASP-2 antibody and a MASP-2 inhibitory peptide.
The therapeutic efficacy of the MASP-2 inhibitor compositions and methods of the invention, as well as the appropriate dosage, for a particular subject may be determined according to complement assays well known to those of skill in the art. Complement produces a variety of specific products. During the last decade, sensitive and specific assays have been developed and most of these activation products are commercially available, including small activation fragments C3a, C4a and C5a and large activation fragments iC3b, C4d, bb and sC5b-9. Most of these assays utilize monoclonal antibodies that react with neoantigens (new antigens/neoantigens) that are exposed on fragments rather than on the native protein they form, which makes these assays very simple and specific. Most rely on ELISA techniques, although radioimmunoassays are sometimes used for C3a and C5a. Radioimmunoassay determines unprocessed fragments and their "desArg" fragments, which are the predominant forms present in the circulation. Unprocessed fragment and C5a desArg Is rapidly cleared by binding to cell surface receptors, therebyIs present in very low concentrations of C3a desArg Then no cells are bound and only accumulate in the plasma. The assay of C3a provides a sensitive, pathway independent marker of complement activation. Alternative pathway activation can be assessed by assaying the Bb fragment. Detection of the membrane attack pathway activated liquid phase product sC5b-9 provides evidence that complement is fully activated. Because both the lectin and classical pathways produce the same activation products C4a and C4d, determining the two fragments does not provide any information as to which of the two pathways produced the activation product.
MASP-2 dependent inhibition of complement activation is characterized by at least one of the following changes in the complement system components due to administration of a MASP-2 inhibitor according to the methods of the invention: inhibition of formation or production of MASP-2 dependent complement activation system products C4b, C3a, C5a and/or C5b-9 (MAC) (e.g., as measured in example 2), reduction of C4 cleavage and C4b deposition (e.g., as measured in example 10), or reduction of C3 cleavage and C3b deposition (e.g., as measured in example 10).
Other medicaments
In certain embodiments, the methods of preventing, treating, reversing, and/or inhibiting fibrosis and/or inflammation comprise administering a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) as part of a therapeutic regimen, and one or more other drugs, biologicals, or therapeutic interventions suitable for inhibiting fibrosis and/or inflammation. In certain embodiments, other drugs, biologicals, or therapeutic interventions are appropriate for the particular symptoms associated with the disease or condition caused or exacerbated by fibrosis and/or inflammation. For example, MASP-2 inhibitory antibodies may be administered as part of a therapeutic regimen in conjunction with one or more immunosuppressants such as methotrexate, cyclophosphamide, azathioprine, and mycophenolate. For another example, MASP-2 inhibitory antibodies may be administered as part of a therapeutic regimen in conjunction with one or more agents designed to increase blood flow (e.g., nifedipine, amlodipine, diltiazem, felodipine, or nicardipine). For another example, MASP-2 inhibitory antibodies may be administered as part of a therapeutic regimen in conjunction with one or more agents that are expected to reduce fibrosis, such as d-penicillamine, colchicine, PUVA, relaxin, cyclosporine, a TGF-beta blocker, and/or a p38 MAPK blocker. For another example, MASP-2 inhibitory antibodies may be administered as part of a therapeutic regimen in conjunction with a steroid or bronchodilator.
Compositions and methods comprising MASP-2 inhibitors (e.g., MASP-2 inhibitory antibodies) may optionally comprise one or more additional therapeutic agents that may enhance the activity of the MASP-2 inhibitor or that provide related therapeutic functions in an additive or synergistic manner. For example, in the case of treating a subject suffering from a disease or condition caused or exacerbated by fibrosis and/or inflammation, one or more MASP-2 inhibitors may be administered (including co-administration) in combination with one or more additional anti-fibrotic agents and/or one or more anti-inflammatory and/or immunosuppressive agents.
MASP-2 inhibitors (e.g., MASP-2 inhibitory antibodies) may be used in combination with other therapeutic agents such as general immunosuppressive drugs, e.g., corticosteroids, immunosuppressive or cytotoxic agents, and/or anti-fibrotic agents.
Drug carrier and delivery vehicle
In general, MASP-2 inhibitor compositions of the invention, when used in combination with any other selected therapeutic agent, are suitable for inclusion in a pharmaceutically acceptable carrier. The carrier is non-toxic, biocompatible and is selected so as not to adversely affect the biological activity of the MASP-2 inhibitor (and any other therapeutic agent used in combination therewith). Exemplary pharmaceutically acceptable carriers for peptides are described in U.S. patent No. 5,211,657 to Yamada. The anti-MASP-2 antibodies and inhibitory peptides for use in the present invention may be formulated in solid, semi-solid, gel, liquid or gaseous form preparations such as tablets, capsules, powders, granules, ointments, solutions, suppositories (suppositories), inhalants and injections for oral, parenteral or surgical administration. The present invention also includes topical administration of the compositions by coating medical devices and the like.
Suitable carriers for parenteral delivery by injection, infusion or irrigation and topical delivery include distilled water, phosphate buffered saline, standard ringer's solution or lactate ringer's solution, dextrose solution, hank's solution or propylene glycol. In addition, sterile fixed oils may be employed as a solvent or suspending medium. For this purpose, any biocompatible oil may be employed, including synthetic mono-or diglycerides. In addition, fatty acids (such as oleic acid) find use in the preparation of injectables. The carrier and drug may be formulated as a liquid formulation, suspension, gel, paste or ointment, either polymerizable or non-polymerizable.
The carrier may also include a delivery vehicle to sustain (i.e., prolong, delay or modulate) drug delivery, or enhance delivery, absorption, stability or pharmacokinetic profile of the therapeutic drug. Such delivery vehicles may include, but are not limited to, the following examples: microparticles, microspheres, nanospheres or nanoparticles consisting of proteins, liposomes, carbohydrates, synthetic organic compounds, inorganic compounds, polymeric or copolymeric hydrogels and polymeric micelles. Suitable hydrogel and micelle delivery systems include PEO as disclosed in WO 2004/009664A2, PHB, PEO copolymers and copolymer/cyclodextrin complexes, and PEO/cyclodextrin complexes as disclosed in U.S. patent application publication No. 2002/0019369A 1. These hydrogels may be injected locally at the desired site of action, or subcutaneously or intramuscularly to form a slow release depot.
For intra-articular delivery, the MASP-2 inhibitor may be loaded into the injectable liquid or gel carrier described above, the injectable slow release delivery vehicle described above, or hyaluronic acid or a hyaluronic acid derivative.
For oral administration of non-peptide drugs, the MASP-2 inhibitor may be loaded in an inert filler or diluent, such as sucrose, corn starch or cellulose.
For topical administration, the MASP-2 inhibitor may be loaded into ointments, lotions, creams, gels, drops, suppositories, sprays, liquid preparations or powders or into gel or microcapsule delivery systems via transdermal patches.
Various nasal and pulmonary delivery systems are under development, including aerosols, metered dose inhalers, dry powder inhalers and nebulized inhalers, each suitably adapted to deliver the medicament of the invention in aerosol, inhalant or nebulized delivery vehicles.
For Intrathecal (IT) or Intraventricular (ICV) delivery, suitable sterile delivery systems (e.g., liquid formulations; gels, suspensions, etc.) may be used to administer the agents of the present invention.
The compositions of the present invention may also include biocompatible excipients such as dispersing or wetting agents, suspending agents, diluents, buffers, permeation enhancers, emulsifiers, binders, thickeners, flavoring agents (for oral administration).
Pharmaceutical carriers for antibodies and peptides
More particularly, as regards anti-MASP-2 antibodies and inhibitory peptides, exemplary dosage forms may be administered parenterally in the form of solutions or suspensions of the compounds in injectable doses, the compounds being contained in physiologically acceptable diluents and pharmaceutical carriers, which may be sterile liquids, such as water, oil, saline, glycerol or ethanol. In addition, auxiliary substances such as wetting or emulsifying agents, surfactants, pH buffering substances and the like may be present in the compositions comprising anti-MASP-2 antibodies and inhibitory peptides. Additional components of the pharmaceutical composition include oils (such as oils of animal, vegetable or synthetic origin), for example soybean oil and mineral oil. In general, glycols such as propylene glycol or polyethylene glycol are the preferred liquid carriers for injection solutions.
The anti-MASP-2 antibodies and inhibitory peptides can also be administered in the form of a long-acting injectable or implantable formulation, which may be formulated in a manner that allows for sustained or pulsatile release of the active agent.
Pharmaceutically acceptable carrier for expression inhibitor
More specifically, as for the expression inhibitor used in the method of the present invention, there is provided a composition comprising the above-mentioned expression inhibitor and a pharmaceutically acceptable carrier or diluent. The composition may also comprise a colloidal dispersion.
Pharmaceutical compositions including expression inhibitors may include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be prepared from a variety of components including, but not limited to, preformed liquids, self-emulsifying solids, and self-emulsifying semisolids. The preparation of these compositions generally involves mixing the expression inhibitor with one or more of the following ingredients: buffers, antioxidants, low molecular weight polypeptides, proteins, amino acids, carbohydrates (including glucose, sucrose, or dextrins), chelating agents (such as EDTA), glutathione, and other stabilizers and excipients. Neutral buffered saline or saline mixed with non-specific serum albumin are examples of suitable diluents.
In some embodiments, the compositions may be prepared and formulated as emulsions, which are generally heterogeneous systems in which one liquid is dispersed in another liquid in the form of droplets (see Idson, load Pharmaceutical Dosage Forms, first volume, rieger and Banker (major code), mark Dekker, inc., n.y., 1988). Examples of naturally occurring emulsifiers for emulsion formulations include gum arabic, beeswax, lanolin, lecithin, and phospholipids.
In one embodiment, the composition comprising the nucleic acid may be formulated as a microemulsion. Microemulsion as used herein refers to a system of water, oil and amphiphiles that is a single optically isotropic and thermodynamically stable liquid solution (see Rosoff in Pharmaceutical Dosage Forms, first volume). The methods of the invention can also use liposomes to transfer and deliver antisense oligonucleotides to desired sites.
Pharmaceutical compositions and formulations of expression inhibitors for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquid formulations and powders. Conventional pharmaceutical carriers, aqueous, powder or oily matrices, thickeners and the like may be used.
Administration mode
Pharmaceutical compositions comprising MASP-2 inhibitors may be administered in a variety of ways, depending on whether topical or systemic administration is most appropriate for the disease to be treated. In addition, the compositions of the present invention may be delivered by coating or incorporating the compositions onto or into implantable medical devices.
Systemic delivery
The terms "systemic delivery" and "systemic administration" as used herein are meant to include, but are not limited to, oral and parenteral routes, including Intramuscular (IM), subcutaneous, intravenous (IV), intraarterial, inhalation, sublingual, buccal, topical, transdermal, nasal, rectal, vaginal and other routes of administration that effectively disperse the delivered drug to one or more sites of intended therapeutic action. Preferred routes for systemic delivery of the present invention include intravenous, intramuscular, subcutaneous, and inhalation. It will be appreciated that for a drug selected for use in a particular composition of the invention, the exact systemic route of administration will be determined in part by consideration of the sensitivity of the drug to the metabolic conversion pathway associated with that particular route of administration. For example, peptide-energy drugs may be most suitable for administration by a route other than oral.
MASP-2 inhibitory antibodies and polypeptides may be delivered to a subject in need thereof by any suitable method. Methods of delivering MASP-2 antibodies and polypeptides include administration via oral, pulmonary, parenteral (e.g., intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), inhalation (such as via a fine powder formulation), transdermal, nasal, vaginal, rectal or sublingual routes of administration, which may be formulated into dosage forms suitable for each route of administration.
For example, MASP-2 inhibitory antibodies and peptides may be introduced into a living body by applying them to body membranes capable of absorbing polypeptides, such as nasal, gastrointestinal and rectal membranes. Polypeptides are typically applied to an absorbable membrane along with a permeation enhancer (see, e.g., lee, V.H.L., crit.Rev.Ther.Drug Carrier sys.5:69,1988; lee, v.h.l., j. Controlled Release13:213,1990; lee, v.h.l., ed., peptide and Protein Drug Delivery, marcel Dekker, new York (1991); deBoer, a.g., et al, j. Controlled Release 13:241, 1990). For example, STDHF is a synthetic derivative of fusidic acid, a steroid surfactant structurally similar to bile salts, and has been used as a permeation enhancer for nasal delivery. (Lee, W.A., biopharm.22, 1990, 11/12 month).
MASP-2 inhibitory antibodies and polypeptides may be introduced in combination with other molecules, such as lipids, to protect the polypeptides from enzymatic degradation. For example, covalent attachment of polymers, particularly polyethylene glycol (PEG), has been used to protect certain proteins from enzymatic hydrolysis in vivo, thereby extending half-life (Fuertges, F., et al, J.controlled Release 11:139, 1990). A number of polymer systems for protein delivery have been reported (Bae, Y.H. et al, J.controlled Release 9:271,1989; hori, R.et al, pharm.Res.6:813,1989; yamakawa, I.et al, J.pharm.Sci.79:505,1990; yoshihiro, I.et al, J.controlled Release 10:195,1989; asano, M.et al, J.controlled Release 9:111,1989; rosenblatt, J.et al, J.controlled Release 9:195,1989; mao, K., J.controlled Release 12:235,1990; takakura, Y.et al, J.Pharm.Sci.78:117,1989, Y.Pharmi.78:38, J.38).
Recently, liposomes with improved serum stability and circulation half-life have been developed (see, e.g., U.S. patent No. 5,741,516 to Webb). Furthermore, various methods of liposomes and liposome-like preparations as possible drug carriers have been reviewed (see, e.g., U.S. Pat. No. 5,567,434 to Szoka; U.S. Pat. No. 5,552,157 to Yagi; U.S. Pat. No. 5,565,213 to Nakamori; U.S. Pat. No. 5,738,868 to Shinkanenko, and U.S. Pat. No. 5,795,587 to Gao).
For transdermal applications, MASP-2 inhibitory antibodies and polypeptides may be combined with other suitable ingredients, such as carriers and/or adjuvants. There are no limitations on the nature of these other ingredients, except that they must be pharmaceutically acceptable for their intended administration and do not reduce the activity of the active ingredient in the composition. Examples of suitable vehicles include ointments, creams, gels or suspensions with or without purified collagen. MASP-2 inhibitory antibodies and polypeptides may also be impregnated into transdermal patches, plasters and bandages, preferably in liquid or semi-liquid form.
The compositions of the present invention may be administered systemically on a periodic basis at intervals determined to maintain the desired level of therapeutic effect. For example, the compositions may be administered every 2-4 weeks or at less frequent intervals (such as via subcutaneous injection). The dosage regimen will be determined by the physician considering various factors that may affect the effect of the drug combination. These factors may include the extent of progression of the disease to be treated, the age, sex and weight of the patient, and other clinical factors. The dosage of each individual pharmaceutical ingredient will vary with the presence and nature of the MASP-2 inhibitor and any drug delivery vehicle (e.g., slow release delivery vehicle) included in the composition. Furthermore, the dosage may be adjusted after taking into account the frequency of administration and changes in the pharmacokinetic profile of the drug being delivered.
Local delivery
The term "topical" as used herein includes the application of a drug at or about a predetermined localized site of action, and may include, for example, topical delivery to the skin or other affected tissue; ocular delivery; intrathecal (IT), intraventricular (ICV), intra-articular, intra-luminal, intracranial, or intra-alveolar administration, placement, or irrigation. It may be preferable to be able to administer low doses of local administration to avoid systemic side effects, as well as to more precisely control the timing of delivery and concentration of active agent at the local delivery site. Local administration achieves a known concentration at the target site regardless of changes in metabolism, blood flow, etc. between patients. Dose control is also improved by direct delivery.
Local delivery of MASP-2 inhibitors may be achieved in the context of surgical methods for treating diseases or conditions caused or exacerbated by fibrosis and/or inflammation, for example during procedures such as surgery.
Treatment regimen
In prophylactic applications, a pharmaceutical composition comprising a MASP-2 inhibitor (e.g., a MASP-2 inhibitory antibody) is administered to a subject that is susceptible to or otherwise at risk of developing a disease or disorder caused or exacerbated by fibrosis and/or inflammation in an amount sufficient to inhibit fibrosis and/or inflammation, thereby eliminating or reducing the risk of developing symptoms of the condition. In some embodiments, the pharmaceutical composition is administered to a subject suspected of or having a disease or disorder caused or exacerbated by fibrosis and/or inflammation in a therapeutically effective amount sufficient to reduce or at least partially reduce the symptoms of the condition. In a prophylactic and therapeutic regimen, a composition comprising a MASP-2 inhibitor may be administered in several doses until sufficient therapeutic results are achieved in the subject. Administration of the MASP-2 inhibitory compositions of the invention may be performed by single administration of the compositions, or by a limited number of sequential administrations, to treat acute conditions associated with fibrosis and/or inflammation. Alternatively, the composition may be administered at regular intervals over a long period of time to treat chronic conditions associated with fibrosis and/or inflammation.
In a prophylactic and therapeutic regimen, a composition comprising a MASP-2 inhibitor may be administered in several doses until a sufficient therapeutic result is obtained in the subject. In one embodiment of the invention, the MASP-2 inhibitor comprises a MASP-2 antibody, which may suitably be administered to an adult patient (e.g., 70kg average adult body weight) at the following doses: 0.1mg to 10,000mg, more suitably 1.0mg to 5,000mg, more suitably 10.0mg to 2,000mg, more suitably 10.0mg to 1,000mg and still more suitably 50.0mg to 500mg. For pediatric patients, the dose may be adjusted in proportion to the weight of the patient. The MASP-2 inhibitor compositions of the present invention may be administered by a single administration or a limited continuous administration of the compositions to treat subjects suffering from or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation. Alternatively, the composition may be administered at regular intervals (such as daily, twice weekly, every other week, monthly or bi-monthly) over an extended period of time for treating a subject suffering from or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation.
In a prophylactic and therapeutic regimen, a composition comprising a MASP-2 inhibitor may be administered in several doses until sufficient therapeutic results are achieved in the subject.
In some embodiments, by determining that a subject has one or more symptoms of impaired renal function, the subject is identified as being at risk of developing a disease or disorder caused or exacerbated by fibrosis or inflammation, as assessed, for example, by measuring serum creatinine levels, serum creatinine clearance, blood urea nitrogen levels, protein in urine, and/or by measuring one or more biomarkers associated with renal disease or injury.
Methods for assessing kidney function are well known in the art and include, but are not limited to, measuring systemic and glomerular capillary blood pressure, proteinuria (e.g., albuminuria), microscopic and macroscopic blood urine, serum creatinine levels (e.g., one formula for estimating kidney function in humans equates a creatinine level of 2.0mg/dl to 50% of normal kidney function, and 4.0mg/dl to 25%), drop in glomerular filtration rate (e.g., creatinine clearance), and extent of tube injury. For example, the assessment of renal function can include assessing at least one renal function using biological and/or physiological parameters such as serum creatinine levels, creatinine clearance, 24 hour urinary protein secretion, glomerular filtration rate, urinary albumin creatinine ratio, albumin secretion rate, and renal biopsy (e.g., by measuring collagen and/or fibronectin deposition, determining the extent of renal fibrosis).
VI. Examples
The following examples merely illustrate the best mode presently contemplated for carrying out the invention, but are not to be construed as limiting the invention. All documents cited herein are incorporated herein by reference.
Example 1
This example describes the production of a mouse strain lacking MASP-2 (MASP-2-/-) but with sufficient MAp19 (MAp19+/+).
Materials and methods: the targeting vector pKO-NTKV 1901 was designed to disrupt the three exons encoding the C-terminus of murine MASP-2, including the exon encoding the serine protease domain, see FIG. 3. Murine ES cell line E14.1a (SV 129 Ola) was transfected with PKO-NTKV 1901. Neomycin-resistant and thymidine kinase-sensitive clones were selected. 600 ES clones were selected, in which 4 different clones were identified, which were confirmed by Southern blotting to contain the targeting event (targeting event) and recombination event of the intended choice, see FIG. 3. Chimeras were generated from these 4 positive clones by embryo transfer. The chimeras were then backcrossed against a genetic background C57/BL6 to generate transgenic male mice. Crossing transgenic male and female mice produced F1, with 50% of the offspring exhibiting disrupted heterozygosity of the MASP-2 gene. Heterozygous mice were crossed to produce homozygous MASP-2 deficient offspring, producing heterozygous and wild-type mice at a ratio of 1:2:1, respectively.
Results and phenotypes: the resulting homozygous MASP-2-/-deficient mice were found to be viable and fertile, and the lack of MASP-2 was confirmed by Southern blotting, confirming the correct targeting event, northern blotting, and Western blotting, confirming the lack of MASP-2mRNA (data not shown). The presence of MAp19 mRNA and the absence of MASP-2mRNA was further confirmed on a LightCycler machine using time-resolved RT-PCR. MASP-2-/-mice did continue to express MAP19, MASP-1 and MASP-3mRNA and protein as expected (data not shown). The presence and abundance of mRNA for properdin, factor B, factor D, C, C2 and C3 in MASP-2-/-mice was assessed by the LightCycler assay and found to be identical to that in wild-type littermate mice controls (data not shown). Plasma from homozygous MASP-2-/-mice is completely devoid of lectin pathway-mediated complement activation, as further described in example 2.
MASP-2-/-lines were generated in the context of pure C57BL 6: MASP-2-/-mice were used as experimental animal models after 9 backcrossing with the pure C57BL6 strain.
Transgenic mouse lines that were murine MASP-2-/-, MAp19+/+ and expressed human MASP-2 transgenes (murine MASP-2 knockdown and human MASP-2 knockdown) were also generated as follows:
Materials and methods: a minigene (SEQ ID NO: 49) comprising the promoter region of the human MASP2 gene and encoding human MASP-2, designated "mini hMASP-2", is constructed, see FIG. 4, which comprises the first 3 exons (exon 1 to exon 3) followed by a cDNA sequence representing the coding sequence of the next 8 exons, thus encoding the full length MASP-2 protein driven by its endogenous promoter. The mini hMASP-2 construct is injected into the fertilized egg of MASP-2-/-so that the human MASP-2 is expressed by the transgene in place of the defective murine MASP2 gene.
Example 2
This example demonstrates that MASP-2 is necessary for complement activation via the lectin pathway.
Methods and materials:
lectin pathway specific C4 cleavage assay: the C4 cleavage assay is described in Petersen, et al, J.Immunol. Methods 257:107 (2001), which determines lectin pathway activation by lipoteichoic acid (LTA) from Staphylococcus aureus, which binds L-fiber gelling proteins. The assay described in Petersen et al, (2001) was improved by adding serum from MASP-2-/-mice after coating the plates with LPS and mannan or zymosan to determine lectin pathway activation by MBL, as described below. The assay was also improved to eliminate the possibility of C4 cleavage by the classical pathway. This is achieved by using a sample dilution buffer containing 1M NaCl, which allows lectin pathway recognition components to bind their ligands with high affinity, but prevents activation of endogenous C4, thereby excluding participation of the classical pathway by dissociating the C1 complex. Briefly, in an improved assay, a serum sample (diluted in high salt (1M NaCl) buffer) is added to a ligand coated plate followed by a constant amount of pure C4 in buffer (salt with physiological concentration). Binding recognition complex containing MASP-2 cleaves C4 to cause C4b deposition.
The measuring method comprises the following steps:
1) Diluted in coating buffer (15 mM Na 2 CO 3 ,35mM NaHCO 3 1. Mu.g/ml mannan (M7504 Sigma) or any other ligand (such as the following ligand, for example) at pH 9.6) to coat the Nunc Maxisorb microtitre plate @Nunc, catalog number 442404,Fisher Scientific).
The following reagents were used in the present assay:
a. mannans (1. Mu.g/Kong Ganlou glycans (M7504 Sigma) in 100. Mu.l coating buffer);
b. zymosan (1. Mu.g/Kong Jiaomu glycan (Sigma), in 100. Mu.l coating buffer);
LTA (1. Mu.g/well in 100. Mu.l coating buffer, or 2. Mu.g/well in 20. Mu.l methanol);
d.1 mu g H-fiber gel protein specific Mab 4H5 in coating buffer;
e. PSA (2 μg/well in 100 μl coating buffer) from aerococcus aequorum (Aerococcus viridans);
f.100. Mu.l/well formalin-fixed Staphylococcus aureus DSM20233 (OD 550 =0.5) in coating buffer.
2) Plates were incubated overnight at 4 ℃.
3) After overnight incubation, the plate was incubated withBlocking buffer (0.1% (w/v) HSA in 10mM Tris-HCl, 140mM NaCl, 1.5mM NaN) for 0.1% HSA-TBS 3 After 1-3 hours incubation with solution (pH 7.4)), TBS/Tween/Ca was used 2+ (containing 0.05% Tween 20 and 5mM CaCl) 2 、1mM MgCl 2 The TBS (pH 7.4)) was washed 3 times to saturate the remaining protein binding sites.
4) Serum samples to be tested were diluted in MBL binding buffer (1M NaCl), the diluted samples were added to the plate and incubated overnight at 4 ℃. Wells with buffer only served as negative controls.
5) After incubation overnight at 4 ℃, the plates were incubated with TBS/Tween/Ca 2+ Washing 3 times. Human C4 (100. Mu.l/well, 1. Mu.g/ml was then diluted in BBS (4 mM barbital, 145mM NaCl,2mM CaCl) 2 ,1mM MgCl 2 pH 7.4) was added to the plate and incubated at 37 ℃ for 90 minutes. TBS/Tween/Ca for the plate 2+ And washed 3 more times.
6) Chicken anti-human C4C conjugated with alkaline phosphatase (diluted 1:1000 in TBS/tween/Ca 2+ In) to detect C4b deposition, which was added to the plate and incubated for 90 minutes at room temperature. The plate was then treated with TBS/Tween/Ca 2+ Washing 3 times.
7) Alkaline phosphatase was detected by adding 100. Mu.l of p-nitrophenyl phosphate substrate solution, incubating for 20 min at room temperature, and reading OD on a microtiter plate reader 405
Results: FIGS. 5A-B show the amount of C4B deposition on mannan (FIG. 5A) and zymosan (FIG. 5B) in serum dilutions from MASP-2+/+ (cross-hatching), MASP-2+/- (filled circles) and MASP-2+/- (filled triangles). Fig. 5C shows the relative activity of C4 convertase on zymosan (white bar) or mannan (shaded bar) coated plates from MASP-2-/+ mice (n=5) and MASP-2-/-mice (n=4) relative to wild-type mice (n=5), based on the amount of C4b deposition determined for wild-type serum homogenization. Error bars represent standard deviation. As shown in FIGS. 5A-C, plasma from MASP-2-/-mice was completely devoid of lectin pathway-mediated complement activation on mannan and zymosan coated plates. These results clearly demonstrate that MASP-2 is an effector component of the lectin pathway.
Lectin pathway dependent C4 activation in recombinant MASP-2-/-mouse serum to confirm that MASP-2 deficiency is a direct cause of loss of MASP-2-/-mouse lectin pathway dependent C4 activation, the effect of adding recombinant MASP-2 protein to serum samples was tested in the C4 cleavage assay described above. Functionally active murine MASP-2 and catalytically inactive murine MASP-2A recombinant proteins in which the serine residue at the active site of the serine protease domain was replaced by an alanine residue were prepared and purified as described in example 3 below. Pooled sera from 4 MASP-2-/-mice were pre-incubated with either recombinant murine MASP-2 or inactive recombinant murine MASP-2A at increasing protein concentrations and the C4 convertase activity was determined as described above.
Results: as shown in FIG. 6, addition of functionally active murine recombinant MASP-2 protein (represented by open triangles) to serum obtained from MASP-2-/-mice restored lectin pathway-dependent C4 activation in a protein concentration-dependent manner, whereas catalytically inactive murine MASP-2A protein (represented by asterisks) did not restore C4 activation. The results shown in fig. 6 were normalized to the C4 activation results observed with pooled wild-type mouse serum (indicated by dotted lines).
Example 3
This example describes recombinant expression and protein production of recombinant full-length human, rat and mouse MASP-2, MASP-2-derived polypeptides, and catalytically inactive mutant forms of MASP-2.
Expression of full-length human, mouse and rat MASP-2:
the full-length cDNA sequence of human MASP-2 (SEQ ID NO: 4) was likewise subcloned into the mammalian expression vector pCI-Neo (Promega), driving eukaryotic expression under the control of the CMV enhancer/promoter region (described in Kaufman R.J. et al, nucleic Acids Research19:4485-90,1991;Kaufman,Methods in Enzymology,185:537-66 (1991)). Full-length mouse cDNA (SEQ ID NO: 50) and rat MASP-2cDNA (SEQ ID NO: 53) were subcloned into pED expression vectors, respectively. The MASP-2 expression vector was then transfected into the adherent Chinese hamster ovary cell line DXB1 using standard calcium phosphate transfection methods (described in Maniatis et al, 1989). Cells transfected with these constructs grew very slowly, indicating cytotoxicity of the encoded protease.
In another method, a minigene construct (SEQ ID NO: 49) containing human MASP-2cDNA driven by its endogenous promoter is transiently transfected into Chinese hamster ovary Cells (CHO). Human MASP-2 protein is secreted into the medium and isolated as follows.
Full length catalytically inactive expression of MASP-2:
basic principle: MASP-2 is activated by autocatalytic cleavage after recognition of the subfraction MBL or fibrous gelator (L-fibrous gelator, H-fibrous gelator or M-fibrous gelator) binding to their respective carbohydrate patterns. Autocatalytic cleavage leading to activation of MASP-2 often occurs during isolation of MASP-2 from serum or during purification after recombinant expression. To obtain a more stable protein preparation for use as an antigen, MASP-2, called MASP-2A, is produced in a catalytically inactive form by replacing serine residues present in the catalytic triad of the protease domain with alanine residues, in rats (SEQ ID NO:55Ser617 to Ala 617); in mice (SEQ ID NO:52Ser617 to Ala 617); or in humans (SEQ ID NO:6Ser618 to Ala 618).
To produce catalytically inactive human and murine MASP-2A proteins, site-directed mutagenesis was performed using the oligonucleotides shown in Table 5. To change serine codons to alanine codons, the oligonucleotides in table 5 were designed to anneal human and murine cDNA regions encoding enzymatically active serine, the oligonucleotides containing mismatches. For example, the human MASP-2cDNA (SEQ ID NO: 4) is bound using PCR oligonucleotides SEQ ID NO:56-59 to amplify the region from the start codon to enzymatically active serine and the region from serine to the stop codon, thereby producing the mutant MASP-2A containing the Ser618 to Ala618 mutation in a completely open readable form. The PCR products were purified after agarose gel electrophoresis and band preparation, using standard tailing methods to create a monoadenosine overlap. The adenosyltailed MASP-2A was then cloned into pGEM-T easy vector and transformed into E.coli.
Catalytically inactive rat MASP-2A protein was produced by kinase (king) the two oligonucleotides SEQ ID NO. 64 and SEQ ID NO. 65 and annealing by combining the two oligonucleotides in equimolar amounts, after heating at 100℃for 2 minutes, with slow cooling to room temperature. The resulting annealed fragment had Pst1 and Xba1 compatible ends, and the fragment was inserted to replace the Pst1-Xba1 fragment of wild-type rat MASP-2cDNA (SEQ ID NO: 53) to yield rat MASP-2A.
5'GAGGTGACGCAGGAGGGGCATTAGTGTTT 3'(SEQ ID NO:64)
5'CTAGAAACACTAATGCCCCTCCTGCGTCACCTCTGCA 3'(SEQ ID NO:65)
Human, mouse and rat MASP-2A were further subcloned into the mammalian expression vectors pED or pCI-Neo, respectively, and transfected into the Chinese hamster ovary cell line DXB1, as follows.
In another approach, the method described by Chen et al was used to construct a catalytically inactive form of MASP-2 (Chen et al J. Biol. Chem.,276 (28): 25894-25902,2001). Briefly, a plasmid containing full-length human MASP-2cDNA (described in Thiel et al, nature 386:506, 1997) was digested with Xho1 and EcoR1, and MASP-2cDNA (see SEQ ID NO:4 herein) was cloned into the corresponding restriction site of the pFastBac1 baculovirus transfer vector (Life Technologies, NY). MASP-2 serine protease active site Ser618 is then changed to Ala618 by substituting the double-stranded oligonucleotides encoding the peptide region amino acids 610-625 (SEQ ID NO: 13) with the native region amino acids 610-625, thereby producing a MASP-2 full-length polypeptide with an inactive protease domain.
Construction of expression plasmids containing polypeptide regions derived from human Masp-2
MASP-2 signal peptide (residues 1-15 of SEQ ID NO: 5) was used to generate the following constructs to secrete the different MASP-2 domains. Constructs expressing the human MASP-2CUBI domain (SEQ ID NO: 8) were prepared by PCR amplification of the region encoding MASP-2 (SEQ ID NO: 6) residues 1-121 (corresponding to the N-terminal CUBI domain). Constructs expressing the human MASP-2CUBIEGF domain (SEQ ID NO: 9) were prepared by PCR amplification of the region encoding MASP-2 (SEQ ID NO: 6) residues 1-166, corresponding to the N-terminal CUBIEGF domain. Preparation of expressed human MASP-2CUBIEGFCU by PCR amplification of the region encoding MASP-2 (SEQ ID NO: 6) residues 1-293 (corresponding to the N-terminal CUBIEGFCUBII domain)Constructs of BII domain (SEQ ID NO: 10). By Vent R The polymerase, pBS-MASP-2, serves as a template to amplify the domains by PCR according to established PCR methods. Sense primer (5' -CG)GGATCCATGAGGCTGCTGACCCTC-3'SEQ ID NO: 34) introduces a BamHI restriction site (underlined) at the 5' end of the PCR product. The antisense primers for each MASP-2 domain shown in Table 5 below were designed to introduce a stop codon (bold) before the EcoRI site (underlined) at the end of each PCR product. Once amplified, the DNA fragment was digested with BamHI and EcoRI and cloned into the corresponding sites of the pFastBac1 vector. The resulting constructs were characterized by restriction mapping and confirmed by dsDNA sequencing.
Table 5: MASP-2PCR primer
Recombinant eukaryotic expression of MASP-2 and production of enzymatically inactive mouse, rat and human MASP-2A proteins the above MASP-2 and MASP-2A expression constructs were transfected into DXB1 cells using standard calcium phosphate transfection methods (Maniatis et al, 1989). MASP-2A was produced in serum-free medium to ensure that the preparation was not contaminated with other serum proteins. Media was harvested from confluent cells every other day (four total). The recombinant MASP-2A level in each of the three species was on average about 1.5 mg/liter of medium.
MASP-2A protein purification: MASP-2A (Ser-Ala mutant described above) was purified by affinity chromatography on MBP-A agarose columns. This strategy enables rapid purification without the use of external labels. MASP-2A (100-200 ml of medium was buffered with equal volumes of loading buffer (50 mM Tris-Cl, pH 7.5, containing 150mM NaCl and 25mM CaCl) 2 ) Diluted) was applied to MBP-agarose affinity column (4 ml) pre-equilibrated with 10ml loading buffer. After washing with another 10ml of loading buffer, the proteins were eluted in 1ml fractions containing 50mM Tris-Cl (pH 7.5) with 1.25M NaCl and 10mM EDTA. SDS-polyacrylamide gel electrophoresis was used to identify MASP-2A containing fractions. MASP-2A was further purified by ion exchange chromatography on a MonoQ column (HR 5/5), if necessary. Protein content 5 Dialysis was performed against 50mM Tris-Cl (pH 7.5) with 0mM NaCl and applied to a column equilibrated with the same buffer. After washing, bound MASP-2A was eluted with an additional 10ml of 0.05-1M NaCl gradient.
Results: MASP-2A protein was obtained in a yield of 0.25-0.5mg from 200ml of medium. The molecular weight of 77.5kDa, as measured by MALDI-MS, is greater than the calculated for the unmodified polypeptide (73.5 kDa). Attachment of glycans at each N-glycosylation site is responsible for the measured molecular weight. MASP-2A migrates as a single band on SDS-polyacrylamide gel, demonstrating no proteolytic processing during biosynthesis. The weight average molecular weight as measured by equilibrium ultracentrifugation was consistent with the calculated value of glycosylated polypeptide homodimer.
Production of recombinant human MASP-2 polypeptides
Another method for producing recombinant MASP-2 and MASP-2A-derived polypeptides is described in Thielens, N.M., et al, J.Immunol.166:5068-5077,2001. Briefly, spodoptera frugiperda (Spodoptera frugiperda) insect cells (Ready-Plaque Sf9 cells from Novagen, madison, wis.) were grown and maintained in Sf900II serum-free medium (Life Technologies) supplemented with 50IU/ml penicillin and 50mg/ml streptomycin (Life Technologies). Trichoplusia ni (High Five) insect cells (supplied by Jadwiga Chroboczek, institut de Biologie Structurale, grenobe, france) were maintained in TC100 medium (Life Technologies) containing 10% FCS (Dominique Dutscher, brumath, france) supplemented with 50IU/ml penicillin and 50mg/ml streptomycin. The Bac-to-Bac system (Life Technologies) was used to produce recombinant baculoviruses. The bacmid (bacmid) DNA was purified using a Qiagen medium prep purification system (Qiagen) and used to transfect Sf9 insect cells in Sf900II SFM medium (Life Technologies) of cellfectin according to the protocol described by the manufacturer. Recombinant viral particles were collected after 4 days and titrated by viral plaque assay and amplified according to the methods described by King and Possee (King and Possee, supra, the Baculovirus Expression System: A Laboratory Guide, chapman and Hall Ltd., london, pages 111-114, 1992).
In Sf900 II SFM medium, recombinant diseases containing MASP-2 polypeptideThe toxin infects High Five cells at 28℃C (1.75 x 10 7 Individual cells/175 cm 2 Tissue culture flasks) for 96 hours, the multiplicity of infection was 2. The supernatant was collected by centrifugation and diisopropylfluorophosphoric acid was added to a final concentration of 1mM.
MASP-2 polypeptide is secreted into the culture medium. The culture supernatant was subjected to 50mM NaCl, 1mM CaCl 2 50mM triethanolamine hydrochloride (pH 8.1) was dialyzed and applied to a Q-Sepharose fast flow column (Amersham Pharmacia Biotech) (2.8X 12 cm) equilibrated with the same buffer at a rate of 1.5 ml/min. Elution was performed with a linear gradient of 1.2 liters (to 350mM NaCl) in the same buffer. Fractions containing recombinant MASP-2 polypeptide were identified by Western blot analysis by addition of (NH 4 ) 2 SO 4 Precipitation was performed to 60% (w/v), and left standing overnight at 4 ℃. The pellet was resuspended in 145mM NaCl, 1mM CaCl 2 50mM triethanolamine hydrochloride (pH 7.4) was applied to a TSK G3000 SWG column (7.5 x 600 mM) (Tosohaas, montgomeryville, pa.) equilibrated with the same buffer. The purified polypeptide was then concentrated to 0.3mg/ml by ultrafiltration in a Microsep microconcentrator (molecular weight cut-off=10,000) (Filtron, karlstein, germany).
Example 4
This example describes the production of polyclonal antibodies against MASP-2 polypeptides.
Materials and methods:
MASP-2 antigen: rabbits were immunized with the following isolated MASP-2 polypeptides to produce polyclonal anti-human MASP-2 antisera: human MASP-2 (SEQ ID NO: 6) isolated from serum; recombinant human MASP-2 (SEQ ID NO: 6), MASP-2A containing an inactive protease domain (SEQ ID NO: 13), see example 3; expressed recombinant CUBI (SEQ ID NO: 8), CUBEGFI (SEQ ID NO: 9) and CUBEGFCUBII (SEQ ID NO: 10) as described in example 3 above.
Polyclonal antibodies: six week old rabbits, which had been immunized with BCG (bacillus calmette Guerin (bacillus Calmette-Guerin vaccinee)), were immunized by injection with 100. Mu.g of MASP-2 polypeptide, which was dissolved in a sterile saline solution at 100. Mu.g/ml. Injections were performed every 4 weeks and antibody titers were monitored by ELISA assays as described in example 5. Culture supernatants were collected for antibody purification by protein a affinity chromatography.
Example 5
This example describes the production of murine monoclonal antibodies against rat or human MASP-2 polypeptides.
Materials and methods:
100 μg of human or rat rMASP-2 or rMASP-2A polypeptide (prepared as described in example 3) dissolved in 200 μl of Phosphate Buffered Saline (PBS) (pH 7.4) complete Freund's adjuvant (Difco Laboratories, detroit, mich.) was subcutaneously injected into 8-12 week old male A/J mice (Harlan, houston, tex.). At two week intervals, 50 μg of human or rat rMASP-2 or rMASP-2A polypeptide dissolved in incomplete Freund's adjuvant was injected subcutaneously into mice twice. On the fourth week, mice were injected with 50. Mu.g of human or rat rMASP-2 or rMASP-2A polypeptide in PBS and fused 4 days later.
For each fusion, a single cell suspension was prepared from the spleen of the immunized mice for fusion with Sp2/0 myeloma cells. Will be 5x10 8 Sp2/0 and 5x10 8 Individual spleen cells were fused in medium containing 50% polyethylene glycol (molecular weight 1450) (Kodak, rochester, n.y.) and 5% dimethyl sulfoxide (Sigma Chemical co., st.louis, mo.). The cells were then conditioned to 1.5x10 per 200 μl of Iscove medium (Gibco, grand Island, N.Y.) suspension 5 Spleen cells were concentrated and the medium was supplemented with 10% fetal bovine serum, 100 units/ml penicillin, 100. Mu.g/ml streptomycin, 0.1mM hypoxanthine, 0.4. Mu.M aminopterin and 16. Mu.M thymidine. 200 microliters of cell suspension was added to each well of approximately twenty 96-well microplates. After about 10 days, the culture supernatant was removed for screening for reactivity with purified factor MASP-2 in an ELISA assay.
ELISA assay: coating at room temperature overnight by adding 50. Mu.l of purified 50ng/ml hMASP-2 or rat rMASP-2 (or rMASP-2A)2 (Dynatech Laboratories, chantilly, va.) wells of a microplate. The low concentration of MASP-2 used for coating enables the selection of high affinity antibodies. After gently beating the plate to remove the coating solution, 200 μl of PBS solution of BLOTTO (skimmed milk powder) was added to each well for 1 hour to block non-specific sites. After one hour, each well was then washed with buffer PBST (PBS containing 0.05% tween 20). 50 microliters of culture supernatant was collected from each fusion well and mixed with 50 microliters of BLOTTO and then added to each well of the microplate. After 1 hour incubation, each well was washed with PBST. Bound murine antibodies were then detected by reaction with horseradish peroxidase (HRP) -conjugated goat anti-mouse IgG (specific for Fc) (Jackson ImmunoResearch Laboratories, west Grove, pa.) and diluted 1:2,000 in bloto. A peroxidase substrate solution containing 0.1%3, 5-tetramethylbenzidine (Sigma, st. Louis, mo.) and 0.0003% hydrogen peroxide (Sigma) was added to each well and developed for 30 minutes. Mu.l of 2M H are added to each well 2 SO 4 The reaction was terminated. Use->ELISA reader (+)>Instruments, winooski, vt.) read the Optical Density (OD) of the reaction mixture at 450 nm.
MASP-2 binding assay:
culture supernatants that test positive in the MASP-2ELISA assay described above may be assayed in a binding assay to determine the binding affinity of MASP-2 inhibitors for MASP-2. Similar assays can also be used to determine whether an inhibitor binds to other antigens in the complement system.
Each well of a polystyrene microtiter plate (96 Kong Peiyang base binding plate, corning Costar, cambridge, mass.) was coated with MASP-2 Phosphate Buffered Saline (PBS) (pH 7.4) (20 ng/100. Mu.l/well, advanced Research Technology, san Diego, calif.) overnight at 4 ℃. After aspiration of the MASP-2 solution, each well was blocked with PBS containing 1% bovine serum albumin (BSA; sigma Chemical) for 2 hours at room temperature. Wells without MASP-2 coating were used as background controls. Hybridoma supernatants at different concentrations in blocking solution or aliquots of purified anti-MASP-2 MoAb were added to each well. After incubation for 2 hours at room temperature, each well was rinsed thoroughly with PBS. anti-MASP-2 MoAb binding to MASP-2 was detected by adding peroxidase conjugated goat anti-mouse IgG (Sigma Chemical) to the blocking solution and incubating at room temperature for 1 hour. After the plate was rinsed thoroughly again with PBS, 100. Mu.l of 3,3', 5' -Tetramethylbenzidine (TMB) substrate (Kirkegaard and Perry Laboratories, gaithersburg, MD) was added. 100 μl of 1M phosphoric acid was added to quench the TMB reaction and the plate was read in a microplate reader (SPECTRA MAX 250,Molecular Devices,Sunnyvale,CA) at 450 nm.
Culture supernatants from positive wells were then assayed for their ability to inhibit complement activation in a functional assay (such as the C4 lysis assay described in example 2). Cells in the positive wells were then cloned by limiting dilution. The reactivity of the MoAb with hMASP-2 was then determined in the ELISA assay described above. Selected hybridomas were grown in roller bottles, and supernatants from depleted cultures were collected and antibodies were purified by protein a affinity chromatography.
Example 6
This example describes the production and production of humanized murine anti-MASP-2 antibodies and antibody fragments.
Murine anti-MASP-2 monoclonal antibodies were generated in male A/J mice as described in example 5. The murine constant region is then substituted with its human counterpart to generate a chimera of IgG and antibody Fab fragments, which chimera is used to inhibit the side effects of MASP-2 dependent complement activation in human subjects of the invention, and the murine antibody is humanized to reduce its immunogenicity as follows.
1. Cloning of anti-MASP-2 variable region genes from murine hybridoma cells
Total RNA was isolated from anti-MASP-2 MoAb secreting hybridoma cells (obtained as described in example 7) using RNAzol according to the manufacturer's protocol (Biotech, houston, tex.). The first cDNA strand was synthesized from total RNA using oligo dT as a primer. 3' primers derived from immunoglobulin constant C region and derived from leader peptide or murine V H Or V K PCR was performed with the degenerate primer set of the first framework region of the gene as the 5' primer pair. Anchor PCR was performed as described by Chen and Platsucas (Chen, P.F., scand.J.Immunol.35:539-549, 1992). For clone V K The gene was used to prepare double-stranded cDNA using the Not1-MAK1 primer (5 '-TGCGGCCGCTGTAGGTGCTGTCTTT-3' SEQ ID NO: 38). Annealing adaptors AD1 (5 '-GGAATTCACTCGTTATTCTCGGA-3' SEQ ID NO: 39) and AD2 (5 '-TCCGAGAATAACGAGTG-3' SEQ ID NO: 40) were ligated to the 5 'and 3' ends of the double stranded cDNA. The 3' terminal adaptors were removed by Notl digestion. The digested product was then used as a PCR template with AD1 oligonucleotide as the 5 'primer and MAK2 (5' -CATTGAAAGCTTTGGGGTAGAAGTTGTTC-3'SEQ ID NO: 41) as the 3' primer. A DNA fragment of about 500bp was cloned into pUC 19. Several clones were selected for sequence analysis to confirm that the cloned sequences included the expected murine immunoglobulin constant regions. Not1-MAK1 and MAK2 oligonucleotides were obtained from V K The region is 182bp and 84bp downstream of the first base pair of the Cκ gene. Selection includes complete V K And cloning of the leader peptide.
For clone V H The gene, a Not1 MAG1 primer (5 '-CGCGGCCGCAGCTGCTCAGAGTGTAGA-3' SEQ ID NO: 42) was used to prepare double stranded cDNA. Annealing adaptors AD1 and AD2 are ligated to the 5 'and 3' ends of the double stranded cDNA. The 3' terminal adaptors were removed by Notl digestion. The digestion products were used as PCR templates with AD1 oligonucleotide and MAG2 (5 '-CGGTAAGCTTCACTGGCTCAGGGAAATA-3' SEQ ID NO: 43) as primers. A DNA fragment of 500-600bp in length was cloned into pUC 19. Not1-MAG1 and MAG2 oligonucleotides were obtained from the murine C.gamma.7.1 region, 180bp and 93bp downstream of the first base pair of the murine C.gamma.7.1 gene, respectively. Selection includes complete V H And cloning of the leader peptide.
2. Construction of chimeric MASP-2IgG and Fab expression vectors
V of the clone H And V K The gene was used as a template for a PCR reaction to add the Kozak consensus sequence to the 5 'end and the splice donor to the 3' end of the nucleotide sequence. After analysis of the sequence to determine that there is no PCR error, V H And V K The genes were inserted into expression vector cassettes containing human C.gamma.1 and C.kappa.respectively to give pSV2 neoVH-huC.gamma.1 and pSV2 neoV-huC.gamma.respectively. Plasmid DNA of heavy and light chain vectors purified using CsCl gradients was transfected into COS cells by electroporation. After 48 hours, the culture supernatant was assayed by ELISA, confirming the presence of approximately 200ng/ml chimeric IgG. Cells were harvested and total RNA was prepared. Using Oligo dT was used as a primer to synthesize the first cDNA strand from total RNA. The cDNA was used as a PCR template to generate Fd and κ DNA fragments. For the Fd gene, PCR was performed using 5'-AAGAAGCTTGCCGCCACCATGGATTGGCTGTGGAACT-3' (SEQ ID NO: 44) as a 5 'primer and a CH1 derived 3' primer (5 '-CGGGATCCTCAAACTTTCTTGTCCACCTTGG-3' SEQ ID NO: 45). Confirmation that the DNA sequence contains the complete human IgG 1V H And a CH1 domain. After digestion with the appropriate enzymes, the Fd DNA fragment was inserted into the HindIII and BamHI restriction sites of the expression vector cassette pSV2dhfr-TUS to give pSV2dhfrFd. The pSV2 plasmid is commercially available and consists of DNA segments of different origins: pBR322 DNA (thin line) contains a pBR322 DNA replication origin (pBR ori) and a lactamase ampicillin resistance gene (Amp); SV40 DNA, represented by thicker hatching and labeled, contains an SV40 DNA replication origin (SV 40 ori), an early promoter (5 'ends of dhfr and neo genes), and polyadenylation signals (3' ends of dhfr and neo genes). The SV 40-derived polyadenylation signal (pA) is also located at the 3' end of the Fd gene.
For the kappa gene, 5'-AAGAAAGCTTGCCGCCACCATGTTCTCACTAGCTCT-3' (SEQ ID NO: 46) was used as the 5' primer and C K The derived 3' primer (5 ' -CGGGATCCTTCTCCCTCTAACACTCT-3' SEQ ID NO: 47) was subjected to PCR. Verification that the DNA sequence contains complete V K And person C K An area. After digestion with the appropriate restriction enzymes, the kappa DNA fragment was inserted into the HindIII and BamHI restriction sites of the expression vector cassette pSV2neo-TUS to give pSV2neoK. The expression of Fd and kappa genes is driven by HCMV-derived enhancer and promoter elements. Because the Fd gene does not include cysteine amino acid residues that are involved in interchain disulfide bonds, the recombinant chimeric Fab contains non-covalently linked heavy and light chains. The chimeric Fab was designated cFab.
To obtain a recombinant Fab with disulfide bonds between the heavy and light chains, the Fd gene described above can be extended to include the coding sequence of an additional 9 amino acids (EPKSCDKTH SEQ ID NO: 48) from the hinge region of human IgG 1. The 30 amino acid BstEII-BamHI DNA segment encoding the 3' end of the Fd gene can be replaced with a DNA segment encoding an extended Fd, resulting in pSV2dhfrFd/9aa.
3. Expression and purification of chimeric anti-MASP-2 IgG
To generate chimeric anti-MASP-2 IgG secreting cell lines, NSO cells were transfected by electroporation with purified plasmid DNA for pSV2neoVH-huC. Gamma.1 and pSV2neoV-huC kappa. Transfected cells were selected in the presence of 0.7mg/ml G418. Cells were cultured in 250ml roller bottles with serum-containing medium.
100ml of culture supernatant of the swirling culture was applied to a 10ml PROSEP-A column (Bioprocessing, inc., princeton, N.J.). The column was washed with 10 bed volumes of PBS. The bound antibody was eluted with 50mM citrate buffer (pH 3.0). An equal volume of 1M Hepes (pH 8.0) was added to the fraction containing purified antibody and the pH was adjusted to 7.0. Residual salts were removed by ultrafiltration through a Millipore membrane (molecular weight cut-off: 3,000) and buffer exchange with PBS. The protein concentration of the purified antibodies was measured by BCA method (Pierce).
4. Expression and purification of chimeric anti-MASP-2 Fab
To generate a chimeric anti-MASP-2 Fab secreting cell line, CHO cells were transfected by electroporation with purified plasmid DNA for pSV2dhfrFd (or pSV2dhfrFd/9 aa) and pSV2neoκ. Transfected cells were selected in the presence of G418 and methotrexate. Selected cell lines were amplified in increasing concentrations of methotrexate. The cells were subjected to single cell subcloning by limiting dilution. The high-yielding single-cell subclone cell line was then cultured in 100ml roller bottles with serum-free medium.
Chimeric anti-MASP-2 Fab was purified by affinity chromatography using a mouse anti-idiotype MoAb against MASP-2 MoAb. Can be used with key hole Hemocyanin (KLH) -conjugated murine anti-MASP-2 MoAb mice were immunized and specific MoAb binding capable of competing with human MASP-2 was screened to prepare anti-idiotype MASP-2MoAb. For purification, 100ml of supernatant from cFab or cFab/9aa producing spinner cultured CHO cells was applied to an affinity column coupled to anti-idiotype MASP-2MoAb. The column was then washed thoroughly with PBS and the bound Fab eluted with 50mM diethylamine (pH 11.5). Residual salts were removed by buffer exchange as described above. Measured by BCA method (Pierce)The protein concentration of the purified Fab was determined.
The inhibition assays described in example 2 or example 7 can be used to determine the ability of chimeric MASP-2IgG, cFab and cFAb/9aa to inhibit the MASP-2 dependent complement pathway.
Example 7
This example describes an in vitro C4 cleavage assay for use as a functional screen to identify MASP-2 inhibitors capable of blocking MASP-2 dependent complement activation via L-fiber gelator/P35, H-fiber gelator, M-fiber gelator or mannan.
C4 cleavage assay: petersen, S.V. et al describe C4 cleavage assays (Petersen, S.V., et al, J.Immunol. Methods 257:107, 2001) and determine lectin pathway activation by lipoteichoic acid (LTA) of Staphylococcus aureus binding L-fiber gelling proteins.
Reagent: formalin-fixed staphylococcus aureus (DSM 20233) was prepared as follows: bacteria were incubated overnight at 37℃in trypticase soy blood medium (tryptic soy blood medium), washed three times with PBS, then fixed in PBS/0.5% formalin for 1 hour at room temperature, washed three times with PBS, and resuspended in coating buffer (15 mM Na 2 CO 3 、35mM NaHCO 3 pH 9.6).
Assay: nuncEach well of a microtiter plate (Nalgene Nunc International, rochester, NY) was coated with the following ingredients: 100 μl formalin-fixed Staphylococcus aureus DSM20233 (OD 550 =0.5) coating buffer with 1 μ g L-fiber gel protein. After overnight incubation, each well was blocked with a TBS solution (10 mM Tris-HCl,140mM NaCl,pH 7.4) containing 0.1% Human Serum Albumin (HSA) followed by a solution containing 0.05% Tween 20 and 5mM CaCl 2 Is washed with TBS solution (washing buffer). Human serum samples were diluted in 20mM Tris-HCl, 1M NaCl, 10mM CaCl 2 In 0.05% Triton X-100, 0.1% HSA (pH 7.4), this prevents activation of endogenous C4 and dissociates the C1 complex (consisting of C1q, C1r and C1 s). Will beDifferent concentrations of MASP-2 inhibitors (including anti-MASP-2 MoAb and inhibitory peptides) were added to serum samples. Diluted samples were added to the plates and incubated overnight at 4 ℃. After 24 hours, each plate was washed thoroughly with wash buffer, then 0.1. Mu.g of solution in 100. Mu.l of 4mM barbital, 145mM NaCl, 2mM CaCl was added to each well 2 、1mM MgCl 2 Purified human C4 (pH 7.4) (obtained as described in Dodds, A.W., methods enzyme.223: 46,1993). After 1.5 hours at 37 ℃, the plates were again washed, C4b deposition was detected using alkaline phosphatase conjugated chicken anti-human C4C (obtained from Immunsystem, uppsala, sweden), and assayed using the colorimetric substrate p-nitrophenyl phosphate.
Measurement of C4 for mannan: the above assay was modified to determine lectin pathway activation via MBL, and assay plates were coated with LSP and mannan prior to addition of serum mixed with various MASP-2 inhibitors.
C4 determination for H-fiber gel protein (Hakata Ag): the above assay was modified to determine lectin pathway activation via H-fiber gel protein and assay plates were coated with LSP and H-fiber gel protein prior to addition of serum mixed with various MASP-2 inhibitors.
Example 8
The following assay demonstrates the presence of classical pathway activation in wild-type and MASP-2-/-mice.
The method comprises the following steps: microtiter plate [ ]Nunc, catalog No. 442404,Fisher Scientific) was coated with 0.1% human serum albumin in 10mM Tris, 140mM NaCl (pH 7.4) solution at room temperature for 1 hour, then diluted 1:1000 in TBS/Tween/Ca 2+ The antisera (Scottish Antibody Production Unit, carluke, scotland) of the sheep antisera in (a) were incubated overnight at 4 ℃ to generate immune complexes in situ. Serum samples were obtained from wild-type and MASP-2-/-mice and added to the coated plates. Control samples were prepared in which C1q was depleted from wild-type and MASP-2-/-serum samples. Protein A-conjugated coated rabbit anti-human C1q IgG (Dako, glotrup, denmark) was used according to the supplier's instructions>(Dynal Biotech, oslo, norway) to prepare C1q depleted mouse serum. Each plate was incubated at 37 ℃ for 90 minutes. Diluted 1:1000 in TBS/Tween/Ca ++ In (a) was used to detect bound C3b by polyclonal anti-human C3C antibody (Dako a 062). The secondary antibody is goat anti-rabbit IgG.
Results: FIG. 7 shows the relative deposition levels of C3b on plates coated with IgG and wild-type serum, MASP-2-/-serum, C1q depleted wild-type, and C1q depleted MASP-2-/-serum. These results demonstrate that the classical pathway is intact in MASP-2-/-mouse strains.
Example 9
The following assay is used to detect whether a MASP-2 inhibitor blocks the classical pathway by assaying the effect of the MASP-2 inhibitor in the event that the classical pathway is initiated by an immune complex.
The method comprises the following steps: to examine the effect of MASP-2 inhibitors on complement activation in which the classical pathway is initiated by immune complexes, 50 μl samples containing 90% NHS were incubated in triplicate at 37℃in the presence of 10 μg/ml Immune Complex (IC) or PBS, and also in triplicate in parallel samples (+/-IC) containing 200nM anti-properdin monoclonal antibody at 37 ℃. After two hours incubation at 37 ℃, 13mM EDTA was added to all samples to terminate further complement activation and the samples were immediately cooled to 5 ℃. Samples were stored at-70℃prior to analysis of the complement activation products (C3 a and sC5 b-9) using ELISA kits (Quidel, accession numbers A015 and A009) according to the manufacturer's instructions.
Example 10
This example describes the identification of high affinity anti-MASP-2 Fab2 antibody fragments that block MASP-2 activity.
Background and rationale: MASP-2 is a complex protein with many independent functional domains, including: MBL and fibrous gelling protein binding site, serine protease catalytic site, proteolytic substrate C2 binding site, proteolytic substrate C4 binding site, MASP-2 zymogen self-activated MASP-2 cleavage site and two Ca ++ Binding site . Fab2 antibody fragments that bind to MASP-2 with high affinity are identified and the identified Fab2 fragments are assayed in a functional assay to determine whether they are capable of blocking MASP-2 functional activity.
In order to block MASP-2 functional activity, an antibody or Fab2 antibody fragment must bind to and block the structural epitope on MASP-2 that is required for MASP-2 functional activity. Thus, many or all of the high affinity binding anti-MASP-2 Fab2 cannot inhibit the functional activity of MASP-2 unless they are able to bind to structural epitopes of MASP-2 that are directly involved in the functional activity of MASP-2.
Functional assays to determine inhibition of lectin pathway C3 convertase formation were used to evaluate the "blocking activity" of anti-MASP-2 Fab 2. The primary physiological role of MASP-2 in the lectin pathway is known to be the production of the next functional component of the lectin-mediated complement pathway, lectin pathway C3 convertase. Lectin pathway C3 convertases are key enzyme complexes (C4 bC2 a) that proteolytically cleave C3 into C3a and C3 b. MASP-2 is not a structural component of the lectin pathway C3 convertase (C4 bC2 a); however, MASP-2 functional activity is required to produce the two protein components (C4 b, C2 a) that make up the lectin pathway C3 convertase. Furthermore, in order for MASP-2 to produce lectin pathway C3 convertases, it appears that all of the above-described MASP-2 independent functional activities are required. For these reasons, the preferred assay for evaluating the "blocking activity" of anti-MASP-2 Fab2 is believed to be a functional assay for determining inhibition of lectin pathway C3 convertase formation.
Production of high affinity Fab 2: phage display libraries of human variable light and heavy chain antibody sequences and automated antibody screening techniques for identifying Fab2 reactive with selected ligands of interest were used to generate high affinity Fab2 against the rat MASP-2 protein (SEQ ID NO: 55). Antibody screening was performed using known amounts of rat MASP-2 (-1 mg, purity > 85%) protein. Three rounds of amplification were used to select the antibody with the highest affinity. Approximately 250 different targets (hits) expressing antibody fragments were selected for ELISA screening. The high affinity targets are then sequenced to determine the uniqueness of the different antibodies.
50 unique anti-MASP-2 antibodies were purified and 250. Mu.g of each purified Fab2 antibody was used to characterize MASP-2 binding affinity and complement pathway function assays, as detailed below.
Assays for evaluating anti-MASP-2 Fab2 inhibition (blocking) activity
1. An assay for inhibiting lectin pathway C3 convertase formation:
background: lectin pathway C3 convertase is an enzyme complex (C4 bC2 a) that proteolytically cleaves C3 into two potent pro-inflammatory fragments, anaphylatoxin C3a and opsonin C3b. The formation of C3 convertase appears to be a key step in the lectin pathway in mediating inflammation. MASP-2 is not a structural component of the lectin pathway C3 convertase (C4 bC2 a); thus, an anti-MASP-2 antibody (or Fab 2) does not directly inhibit the activity of the pre-existing C3 convertase. However, MASP-2 serine protease activity is necessary in order to produce the two protein components (C4 b, C2 a) that make up the lectin pathway C3 convertase. Thus, anti-MASP-2 Fab2 which inhibits the functional activity of MASP-2 (i.e., blocking anti-MASP-2 Fab 2) will inhibit the de novo formation of lectin pathway C3 convertase. C3 contains a unique highly reactive thioester group as a moiety thereof. In this assay, when the C3 convertase cleaves C3, the thioester group on C3b can form a covalent bond with a hydroxyl or amino group on a macromolecule immobilized at the bottom of the plastic well via an ester or amide bond, thereby facilitating detection of C3b in an ELISA assay.
Yeast mannans are known activators of the lectin pathway. In the following method for determining C3 convertase formation, plastic wells coated with mannan were incubated with diluted rat serum at 37 ℃ for 30 minutes to activate the lectin pathway. The wells were then washed and the C3b immobilized in the wells was determined using standard ELISA methods. The amount of C3b produced in this assay directly reflects the de novo formation of lectin pathway C3 convertases. The ability of selected concentrations of anti-MASP-2 Fab2 to inhibit the formation of C3 convertase and subsequent production of C3b is determined in this assay.
The method comprises the following steps:
96-well Costar media binding plates were incubated with mannan diluted in 50mM carbonate buffer (pH 9.5) at 1. Mu.g/50. Mu.l/well overnight at 5 ℃. After overnight incubation, each well was washed three times with 200 μl PBS. Each well was then filled with 100. Mu.l/wellIs blocked with 1% bovine serum albumin in PBS and incubated for 1 hour at room temperature with gentle agitation. Each well was then washed three times with 200. Mu.l PBS. Diluting anti-MASP-2 Fab2 samples at 5℃in Ca-containing ++ And Mg (magnesium) ++ GVB buffer (4.0 mM barbital, 141mM NaCl,1.0mM MgCl) 2 ,2.0mM CaCl 2 0.1% gelatin, pH 7.4) to a selected concentration. 0.5% rat serum was added to the above samples at 5℃and 100. Mu.l was transferred to each well. Plates were covered and incubated in a 37 ℃ water bath for 30 minutes for complement activation. The plates were transferred from the 37 ℃ water bath to a vessel containing an ice-water mixture to terminate the reaction. Each well was washed 5 times with 200. Mu.l of PBS-Tween 20 (0.05% Tween 20 in PBS) and 2 times with 200. Mu.l of PBS. Mu.l/well of primary antibody (rabbit anti-human C3C, DAKO A0062) diluted 1:10,000 was added, the antibody was dissolved in PBS containing 2.0mg/ml bovine serum albumin, and incubated for 1 hour at room temperature with gentle agitation. Each well was washed 5 times with 200. Mu.l PBS. Mu.l/well of secondary antibody (peroxidase conjugated goat anti-rabbit IgG, american Qualex A102 PU) diluted at 1:10,000 was added, the antibody was dissolved in PBS containing 2.0mg/ml bovine serum albumin, and incubated with gentle agitation in a shaker at room temperature for 1 hour. Each well was washed 5 times with 200. Mu.l PBS. 100 μl/well of peroxidase substrate TMB (Kirkegaard &Perry Laboratories) were incubated for 10 minutes at room temperature. By adding 100. Mu.l/well 1.0. 1.0M H 3 PO 4 Termination of the peroxidase reaction, measurement of OD 450
2. Assay background for determining MASP-2 dependent C4 cleavage inhibition: the serine protease activity of MASP-2 is highly specific, and only two protein substrates for MASP-2 were identified: c2 and C4. Cleavage of C4 produces C4a and C4b. anti-MASP-2 Fab2 can bind to a structural epitope of MASP-2 that is directly involved in C4 cleavage (e.g., the MASP-2 binding site of C4; the MASP-2 serine protease catalytic site), thereby inhibiting the C4 cleavage functional activity of MASP-2.
Yeast mannans are known activators of the lectin pathway. In the following method for determining the C4 lytic activity of MASP-2, plastic wells coated with mannan were incubated with diluted rat serum at 37℃for 30 minutes to activate the lectin pathway. Since the primary antibody used in this ELISA assay recognizes only human C4, diluted rat serum was also supplemented with human C4 (1.0. Mu.g/ml). The wells were then washed and human C4b immobilized in the wells was determined using standard ELISA methods. In this assay, the amount of C4b produced is a measure of the C4 cleavage activity of MASP-2. In this assay, the ability of selected concentrations of anti-MASP-2 Fab2 to inhibit C4 cleavage is measured.
The method comprises the following steps: 96-well Costar media binding plates were incubated with mannan diluted in 50mM carbonate buffer (pH 9.5) at 1. Mu.g/50. Mu.l/well overnight at 5 ℃. Each well was washed 3 times with 200. Mu.l PBS. The wells were then blocked with 100 μl/well of 1% bovine serum albumin in PBS and incubated for 1 hour with gentle agitation at room temperature. Each well was washed 3 times with 200. Mu.l PBS. Diluting anti-MASP-2 Fab2 samples at 5℃in Ca-containing ++ And Mg (magnesium) ++ GVB buffer (4.0 mM barbital, 141mM NaCl,1.0mM MgCl) 2 ,2.0mM CaCl 2 0.1% gelatin, pH 7.4) to a selected concentration. 1.0 μg/ml human C4 (Quidel) is also included in these samples. 0.5% rat serum was added to the above samples at 5℃and 100. Mu.l was transferred to each well. Plates were covered and incubated in a 37 ℃ water bath for 30 minutes for complement activation. The plates were transferred from the 37 ℃ water bath to a vessel containing an ice-water mixture to terminate the reaction. Each well was washed 5 times with 200. Mu.l of PBS-Tween 20 (0.05% Tween 20 in PBS), and then each well was washed 2 times with 200. Mu.l of PBS. 100 μl/well of biotin-conjugated chicken anti-human C4C (Immunsystem AB, uppsala, sweden) in PBS (containing 2.0mg/ml Bovine Serum Albumin (BSA)) was added and incubated with gentle agitation at room temperature for 1 hour. Each well was washed 5 times with 200. Mu.l PBS. 100 μl/well of 0.1 μg/ml PBS solution of peroxidase conjugated streptavidin (Pierce Chemical # 21126) (containing 2.0mg/ml BSA) was added and incubated for 1 hour at room temperature with gentle agitation in a shaker. Each well was washed 5 times with 200. Mu.l PBS. 100 μl/well of peroxidase substrate TMB (Kirkegaard &Perry Laboratories) were incubated for 16 minutes at room temperature. By adding 100. Mu.l/well 1.0. 1.0M H 3 PO 4 Termination of the peroxidase reaction, measurement of OD 450
3. Background of binding assays against rat MASP-2Fab2 against "native" rat MASP-2: MASP-2 is typically present in plasma as a dimeric complex of MASP-2 that also includes specific lectin molecules (mannose binding protein (MBL) and fiber gelling proteins). Thus, if there is an interest in studying the binding of anti-MASP-2 Fab2 to physiologically relevant forms of MASP-2, it is important to develop binding assays in which the interaction between Fab2 and plasma "native" MASP-2 is used instead of purified recombinant MASP-2. In this binding assay, the "native" MASP-2-MBL complex from 10% rat serum is first immobilized in mannan-coated wells. The binding affinity of various anti-MASP-2 Fab2 against immobilized "native" MASP-2 was then investigated using standard ELISA methods.
The method comprises the following steps: 96-well Costar high binding plates were incubated with mannan diluted in 50mM carbonate buffer (pH 9.5) at 1. Mu.g/50. Mu.l/well overnight at 5 ℃. Each well was washed 3 times with 200. Mu.l PBS. Each well was blocked with 100 μl/well of 0.5% nonfat dry milk in PBST (PBS with 0.05% tween 20) and incubated with gentle agitation for 1 hour at room temperature. 200 μl TBS/Tween/Ca for each well ++ Washing buffer (Tris buffer, 0.05% Tween 20, containing 5.0mM CaCl) 2 pH 7.4) were washed 3 times. Preparation of high salt binding buffer of 10% rat serum on ice (20mM Tris,1.0MNaCl,10mM CaCl 2 0.05% Triton-X100,0.1% (w/v) bovine serum albumin, pH 7.4. Mu.l of each well was added and incubated overnight at 5 ℃. 200 μl TBS/Tween/Ca for each well ++ The wash buffer was washed 3 times. Each well was then washed 2 times with 200. Mu.l PBS. 100 μl/well was added to dilute the solution containing Ca ++ And Mg (magnesium) ++ GVB buffer (4.0 mM barbital, 141mM NaCl,1.0mM MgCl) 2 ,2.0mM CaCl 2 0.1% gelatin, pH 7.4) at a selected concentration of anti-MASP-2 Fab2, incubated for 1 hour at room temperature with gentle agitation. Each well was washed 5 times with 200. Mu.l PBS. Add 100. Mu.l/well HRP conjugated goat anti-Fab 2 (Biogenesis catalog number 0500-0099) in 2.0mg/ml bovine serum albumin/PBS diluted 1:5000 and incubate with gentle agitation at room temperature for 1 hour. Each well was washed 5 times with 200. Mu.l PBS. 100 μl/well of peroxidase substrate TMB (Kirkegaard&Perry Laboratories) are incubated at room temperatureIncubate for 70 minutes. By adding 100. Mu.l/well of 1.0. 1.0M H 3 PO 4 Termination of the peroxidase reaction, measurement of OD 450
Results:
approximately 250 different Fab2 were selected for ELISA screening which reacted with anti-rat MASP-2 protein with high affinity. These high affinity Fab2 were sequenced to determine the uniqueness of the different antibodies, and 50 unique anti-MASP-2 antibodies were purified for further analysis. 250 μg of each purified Fab2 antibody was used to characterize MASP-2 binding affinity and complement pathway function assays. The results of this analysis are shown in table 6 below.
Table 6: anti-MASP-2 FAB2 blocking lectin pathway complement activation
As shown in Table 6 above, 17 Fab2 out of 50 tested anti-MASP-2 Fab2 were identified as MASP-2 blocking Fab2, which effectively inhibited C3 convertase formation, IC 50 10nM Fab2 (34% positive selection). Of the 17 Fab2 identified, 8 had ICs 50 The range is less than nanomole. Furthermore, all 17 MASP-2 blocking Fab2 shown in Table 6 inhibited C3 convertase formation substantially completely in the lectin pathway C3 convertase assay. FIG. 8A illustrates the results of the C3 convertase formation assay, fab2 antibody #11, representative of the other Fab2 antibodies assayed, the results of which are shown in Table 6. Because "blocking" Fab2 is only likely to inhibit MASP-2 function very weakly, even when individual MASP-2 molecules are bound by the Fab2, this is possible in theory and care is taken.
Although mannans are known activators of the lectin pathway, it is theoretically possible that anti-mannans antibodies present in rat serum might also activate the classical pathway and produce C3b by classical pathway C3 convertases. However, each of the 17 blocking anti-MASP-2 Fab2 listed in this example effectively inhibited C3b production (> 95%), thus demonstrating the specificity of this assay for lectin pathway C3 convertases.
To calculate apparent K of each antibody d Binding assays were performed on all 17 of the blocking Fab 2. The results of the binding assays for the 6 blocking Fab2 anti-rat MASP-2Fab2 against native rat MASP-2 are also shown in Table 6. FIG. 8B graphically illustrates the results of a binding assay with Fab2 antibody # 11. Similar binding assays were also performed for other Fab2, the results of which are shown in table 6. In general, the apparent K obtained for each of the 6 Fab2 binding to "native" MASP-2 d IC with Fab2 in C3 convertase function assay 50 The correspondence of (2) is quite suitable. There is evidence that MASP-2 undergoes a conformational change from an "inactive" to an "active" form after activation of its protease activity (Feinberg et al, EMBO J22:2348-59 (2003); gal et al, J.biol. Chem.280:33435-44 (2005)). MASP-2 exists predominantly in the "inactive" zymogen conformation in normal rat plasma for the C3 convertase formation assay. In contrast, in the binding assay, MASP-2 is present as a component of a complex that binds to immobilized mannans with MBL; thus, MASP-2 may be in an "active" conformation (Petersen et al, J.Immunol Methods 257:107-16,2001). Thus, for each of the 17 blocking Fab2 assayed in these two functional assays, it may not be necessary to expect an IC 50 And K is equal to d The exact correspondence between these is due to the fact that Fab2 may bind to MASP-2 in different conformational forms in each assay. Nevertheless, each of the other 16 Fab 2's tested in both assays, except Fab2#88, IC 50 And apparent K d There appears to be a fairly close correspondence between them (see table 6).
Several blocking Fab2 were evaluated for inhibition of MASP-2 mediated C4 cleavage. FIG. 8C graphically illustrates the results of a C4 cleavage assay, indicating inhibition of IC with Fab2#41 50 =0.81 nM (see table 6). As shown in FIG. 9, all tested Fab2 were found to inhibit C4 cleavage, IC 50 Similar to IC obtained in the C3 convertase assay 50 (see Table 6).
Although mannans are known activators of the lectin pathway, it is theoretically possible that the presence of anti-mannan antibodies in rat serum might also activate the classical pathway, thereby generating C4b by C1 s-mediated C4 cleavage. However, several anti-MASP-2 Fab2 have been identified which are effective in inhibiting C4b production (> 95%), thus demonstrating the specificity of this assay for MASP-2 mediated C4 cleavage. C4, like C3, contains a unique highly reactive thioester group as a moiety. In this assay, after cleavage of C4 by MASP-2, the thioester group on C4b may form a covalent bond with a hydroxyl or amino group on a macromolecule immobilized at the bottom of the plastic well via an ester or amide bond, thus facilitating detection of C4b in an ELISA assay.
These studies clearly demonstrate that the production of high affinity Fab2 for the rat MASP-2 protein functionally blocks C4 and C3 convertase activity, thereby preventing lectin pathway activation.
Example 11
This example describes epitope mapping on several blocking anti-rat MASP-2Fab2 antibodies produced according to the method described in example 10.
The method comprises the following steps:
as shown in fig. 10, using the pED4 vector, the following proteins, each having an N-terminal 6 His-tag, were expressed in CHO cells:
rat MASP-2A, a full length MASP-2 protein, is inactivated by the change of serine to alanine at the center of activity (S613A);
rat MASP-2K, altered to reduce self-activation of full-length MASP-2 protein (R424K);
CUBI-II, an N-terminal fragment of rat MASP-2 comprising only CUBI, EGF-like and CUBII domains; and CUBI/EGF-like, an N-terminal fragment of rat MASP-2 containing only CUBI and EGF-like domains.
These proteins were purified from the culture supernatant by nickel affinity chromatography as described previously (Chen et al J.biol. Chem.276:25894-02 (2001)).
The C-terminal polypeptide (CCPII-SP) comprising the CCPII and rat MASP-2 serine protease domains was expressed in E.coli as a thioredoxin fusion protein using pTrxFus (Invitrogen). Proteins were purified from cell lysates using a Thiobond affinity resin. Thioredoxin fusion partners were expressed from pTrxFus empty vector as negative control.
All recombinant proteins were dialyzed into TBS buffer and their concentration was determined by measuring OD (280 nm).
Dot blot analysis:
serial dilutions of the above and 5 recombinant MASP-2 polypeptides shown in figure 10 (as well as thioredoxin polypeptides as negative controls for CCPII-serine protease polypeptides) were spotted onto nitrocellulose membranes. The amount of protein spotted in the 5-fold step was in the range of 100ng-6.4pg. In a later experiment, the amount of protein spotted again was reduced in the range of 5-fold steps from 50ng to 16pg. The membrane was blocked with 5% nonfat dry milk in TBS (blocking buffer) and then with 1.0. Mu.g/ml blocking buffer against MASP-2Fab2 (containing 5.0mM Ca) 2+ ) Incubation was performed. Bound Fab2 was detected with HRP conjugated anti-human Fab (AbD/Serotec; 1/10,000 dilution) and ECL detection kit (Amersham). One membrane was incubated with polyclonal rabbit anti-human MASP-2Ab (see Stover et al, JImmunol 163:6848-59 (1999)) as a positive control. In this case, bound Ab was detected with HRP conjugated goat anti-rabbit IgG (Dako; 1/2,000 dilution).
MASP-2 binding assays
ELISA plates were coated overnight at 4℃with 1.0. Mu.g/well of carbonate buffer (pH 9.0) of recombinant MASP-2A or CUBI-II polypeptide. The wells were blocked with a 1% BSA TBS solution, followed by the addition of serial dilutions of anti-MASP-2 Fab2 TBS solution (containing 5.0mM Ca) 2+ ). The plates were incubated for 1 hour at room temperature. With TBS/Tween/Ca 2+ After 3 washes, 1/10,000 dilution in TBS/Ca was added 2+ Is incubated again at room temperature for 1 hour. Bound antibodies were detected with TMB peroxidase substrate kit (Biorad).
Results:
the results of the dot blot analysis demonstrating the reactivity of Fab2 with various MASP-2 polypeptides are provided in Table 7 below. The values provided in table 7 indicate the amount of protein spotting required to provide about half of the maximum signal intensity. As shown, all polypeptides (except for thioredoxin fusion partner alone) were recognized by positive control Ab (polyclonal anti-human MASP-2 serum, produced in rabbits).
Table 7: reactivity with each recombinant rat MASP-2 polypeptide in dot blot
Fab2 antibody # MASP-2A CUBI-II CUBI/EGF-like CCPII-SP Thioredoxin
40 0.16ng NR NR 0.8ng NR
41 0.16ng NR NR 0.8ng NR
11 0.16ng NR NR 0.8ng NR
49 0.16ng NR NR >20ng NR
52 0.16ng NR NR 0.8ng NR
57 0.032ng NR NR NR NR
58 0.4ng NR NR 2.0ng NR
60 0.4ng 0.4ng NR NR NR
63 0.4ng NR NR 2.0ng NR
66 0.4ng NR NR 2.0ng NR
67 0.4ng NR NR 2.0ng NR
71 0.4ng NR NR 2.0ng NR
81 0.4ng NR NR 2.0ng NR
86 0.4ng NR NR 10ng NR
87 0.4ng NR NR 2.0ng NR
Positive control <0.032ng 0.16ng 0.16ng <0.032ng NR
Nr=no reaction. The positive control antibody was polyclonal anti-human MASP-2 serum produced in rabbits.
All Fab2 reacted with MASP-2A and MASP-2K (data not shown). Most Fab2 recognizes the CCPII-SP polypeptide but does not recognize the N-terminal fragment. Fab2#60 and fab2#57 are two exceptions. Fab2#60 recognized MASP-2A and CUBI-II fragments, but did not recognize the CUBI/EGF-like polypeptide or the CCPII-SP polypeptide, suggesting that it could bind to an epitope of CUBII or span both CUBII and EGF-like domains. Fab2#57 recognizes MASP-2A but does not recognize any of the MASP-2 fragments tested, indicating that such Fab2 recognizes the epitope of CCP 1. Fabs 2#40 and #49 bind only intact MASP-2A. In the ELISA binding assay shown in FIG. 11, fab2#60 also bound the CUBI-II polypeptide, albeit with only slightly lower apparent affinity.
These observations indicate the identification of unique blocking Fab2 for various regions of the MASP-2 protein.
Example 12
This example describes the use of phage display to identify fully human scFv antibodies that bind MASP-2 and inhibit lectin-mediated complement activation, while preserving the integrity of the classical (C1 q-dependent) pathway components of the immune system.
SUMMARY
Fully human high affinity MASP-2 antibodies were identified by screening phage display libraries. The variable light and heavy chain fragments of antibodies are isolated in scFv format and full length IgG format. Human MASP-2 antibodies may be used to inhibit cellular damage associated with lectin pathway-mediated complement pathway activation, while leaving the classical (C1 q-dependent) pathway components of the immune system intact. In some embodiments, the MASP-2 inhibitory antibody has the following characteristics: (a) High affinity for human MASP-2 (e.g., KD of 10nM or less) and (b) inhibiting MASP-2-dependent complement activity in 90% human serum with an IC50 of 30nM or less.
Method
Full length catalytically inactive expression of MASP-2:
the full length cDNA sequence (SEQ ID NO: 4) of human MASP-2 encoding the human MASP-2 polypeptide and leader sequence (SEQ ID NO: 5) was subcloned into the mammalian expression vector pCI-Neo (Promega) which drives eukaryotic expression under the control of the CMV enhancer/promoter region (described in Kaufman R.J. et al, nucleic Acids Research19:4485-90, 1991;Kaufman,Methods in Enzymology,185:537-66 (1991)).
To produce catalytically inactive human MASP-2A protein, site-directed mutagenesis was performed as described in US2007/0172483, which is incorporated herein by reference. The PCR products were purified after agarose gel electrophoresis and band preparation, and were subjected to standard tailing procedures to create a monoadenosine overlap. The adenosine-tailed MASP-2A was then cloned into pGEM-T easy vector and transformed into E.coli. Human MASP-2A was further subcloned into either of the mammalian expression vectors pED or pCI-Neo.
The MASP-2A expression construct described above was transfected into DXB1 cells using standard calcium phosphate transfection procedures (Maniatis et al, 1989). MASP-2A was produced in serum-free medium to ensure that the preparation was not contaminated with other serum proteins. Media was harvested from confluent cells every other day (four total). The level of recombinant MASP-2A was on average about 1.5 mg/liter of medium. MASP-2A (Ser-Ala mutant described above) was purified by affinity chromatography on MBP-A-agarose columns.
MASP-2AELISA against ScFv candidate clones identified by panning/scFv transformation and Filter screening
Antigen panning was performed on phage display libraries of human immunoglobulin light and heavy chain variable region sequences followed by automated antibody screening and selection to identify high affinity scFv antibodies to human MASP-2 proteins. Three rounds of panning of scFv phage libraries were performed against HIS-tagged or biotin-tagged MASP-2A. The third round of panning was first eluted with MBL and then TEA (alkaline). To monitor the specific enrichment of phages displaying scFv fragments against the target MASP-2A, a polyclonal phage ELISA was performed against immobilized MASP-2A. scFv genes from round 3 panning were cloned into the pheg expression vector and subjected to a small scale filter screen to find specific clones for MASP-2A.
Bacterial colonies containing plasmids encoding scFv fragments from the third round of panning were picked, transferred onto nitrocellulose membranes, and grown overnight on non-induction medium to yield the master plates. A total of 18,000 colonies were picked and analyzed from the third round of panning, half from competitive elution and half from subsequent TEA elution. Panning the scFv phagemid library against MASP-2A followed by scFv transformation and filter screening resulted in 137 positive clones. 108/137 clones were positive in ELISA assays for MASP-2 binding (data not shown), where 45 clones were further analyzed for their ability to block MASP-2 activity in normal human serum.
Assay for measuring inhibition of lectin pathway C3 convertase formation
Functional assays measuring inhibition of lectin pathway C3 convertase formation were used to evaluate the "blocking activity" of MASP-2scFv candidate clones. MASP-2 serine protease activity is required to produce two protein components (C4 b, C2 a) that comprise lectin pathway C3 convertases. Thus, a MASP-2scFv that inhibits the functional activity of MASP-2 (i.e., blocks a MASP-2 scFv) will inhibit the reformation of lectin pathway C3 convertases. C3 contains unusual and highly reactive thioester groups as part of its structure. In this assay, after cleavage of C3 by the C3 convertase, the thioester group on C3b can form a covalent bond with a hydroxyl or amino group on a macromolecule immobilized at the bottom of the plastic well via an ester or amide bond, thereby facilitating detection of C3b in an ELISA assay.
Yeast mannans are known activators of the lectin pathway. In the following method of measuring the formation of C3 convertase, plastic wells coated with mannan are incubated with diluted human serum to activate the lectin pathway. The wells were then washed and the C3b immobilized on the wells was determined using standard ELISA methods. The amount of C3b produced in this assay is a direct reflection of the reformation of lectin pathway C3 convertases. Selected concentrations of MASP-2scFv clones were tested in this assay for their ability to inhibit C3 convertase formation and thus C3b production.
Method:
45 candidate clones identified as above were expressed, purified and diluted to the same stock concentration, which was re-diluted in GVB buffer (4.0 mM barbital, 141mM NaCl, 1.0mM MgCl2, 2.0mM CaCl2, 0.1% gelatin, pH 7.4) containing ca++ and mg++ to ensure that all clones had the same amount of buffer. scFv clones were each tested in triplicate at a concentration of 2 μg/mL. The positive control was OMS100 Fab2 and tested at 0.4. Mu.g/mL. C3C formation was monitored in the presence and absence of scFv/IgG clones.
Mannan was diluted to a concentration of 20 μg/mL (1 μg/well) in 50mM carbonate buffer (15mM Na2CO3+35mM NaHCO3+1.5mM NaN3) pH 9.5 and coated overnight at 4 ℃ on ELISA plates. The next day, the mannan-coated plates were washed 3 times with 200 μl PBS. Mu.l of 1% HSA blocking solution was then added to the wells and incubated for 1 hour at room temperature. Plates were washed 3 times with 200 μl PBS and stored with 200 μl PBS on ice until samples were added.
Normal human serum was diluted to 0.5% in camgvb buffer and scFv clones or OMS100 Fab2 positive controls were added to the buffer in triplicate at 0.01 μg/mL, 1 μg/mL (OMS 100 control only) and 10 μg/mL and preincubated on ice for 45 min before addition to the blocked ELISA plates. The reaction was started by incubation at 37 ℃ for 1 hour and stopped by transferring the plates into an ice bath. C3b deposition was detected with rabbit α -mouse C3C antibody followed by goat α -rabbit HRP. The negative control was buffer without antibody (no antibody = maximum C3b deposition) and the positive control was buffer with EDTA (no C3b deposition). The background was determined by performing the same assay except that the wells contained no mannans. The background signal for the plate without mannan was subtracted from the signal of the well containing mannan. Cut-off criteria were set for the irrelevant scFv clone (VZV) and only half the activity of the buffer.
Results: a total of 13 clones were found to block MASP-2 activity according to cut-off criteria. Production of>All 13 clones with 50% pathway inhibition were selected and sequenced, yielding 10 unique clones. All 10 clones were found to have the same light chain subclass λ3, but three different heavy chain subclasses: VH2, VH3 and VH6. In the functional assay, using 0.5% human serum, 5/10 candidate scFv clones gave IC50 nM values of less than 25nM target standard.
To identify antibodies with improved potency, three parent scFv clones identified as described above were light chain shuffled. The process involves generating a combinatorial library consisting of VH pairs of each master clone with a library of native human lambda light chains (VL) derived from 6 healthy donors. The library is then screened for scFv clones with improved binding affinity and/or functionality.
Table 8: functional potency comparison of IC50 (nM) of major subclones and their corresponding parent clones (all in scFv form)
Heavy chain variable region (VH) sequences for the parent and child clones shown in table 8 above are provided below.
Kabat CDRs (31-35 (H1), 50-65 (H2), and 95-107 (H3)) are indicated in bold; and Chothia CDRs (26-32 (H1), 52-56 (H2) and 95-101 (H3)) are underlined.
17D20_35VH-21N11VL heavy chain variable region (VH) (SEQ ID NO:67, encoded by SEQ ID NO: 66)
d17N9 heavy chain variable region (VH) (SEQ ID NO: 68)
The light chain variable region (VL) sequences for the parent and child clones shown in table 8 above are provided below.
Kabat CDRs (24-34 (L1); 50-56 (L2); and 89-97 (L3) are indicated in bold, and Chothia CDRs (24-34 (L1); 50-56 (L2) and 89-97 (L3) are indicated by underlining.
17D20m_d3521N11 light chain variable region (VL) (SEQ ID NO:70, encoded by SEQ ID NO: 69)
17N16m_d17N9 light chain variable region (VL) (SEQ ID NO: 71)
Both MASP-2 antibody OMS100 and MoAb_d3521N11VL (comprising the heavy chain variable region shown in SEQ ID NO:67 and the light chain variable region shown in SEQ ID NO:70, also referred to as "OMS646" and "mAb 6") have demonstrated the ability to bind human MASP-2 with high affinity and to block functional complement activity, as analyzed by dot blot for epitope binding. The results indicate that OMS646 and OMS100 antibodies are highly specific for MASP-2 and do not bind MASP-1/3. None of the antibodies bound MAp19 nor the MASP-2 fragment that did not contain the CCP1 domain of MASP-2, leading to the conclusion that the binding site contains CCP1.
When compared to C1s, C1r or MASP-1, the MASP-2 antibody OMS646 was assayed to tightly bind recombinant MASP-2 (Kd 60-250 pM) with > 5000-fold selectivity (see Table 9 below):
table 9: affinity and specificity of OMS646 MASP-2 antibody-MASP-2 interaction assessed by solid phase ELISA studies
Antigens K D (pM)
MASP-1 >500,000
MASP-2 62±23*
MASP-3 >500,000
Purified human C1r >500,000
Purified human C1s ~500,000
* Mean ± SD; n=12
OMS646 specifically blocks lectin-dependent activation of terminal complement components
The method comprises the following steps:
the effect of OMS646 on Membrane Attack Complex (MAC) deposition was analyzed using pathway-specific conditions for lectin pathway, classical pathway and alternative pathway. For this purpose, the Wieslab Comp300 complement screening kit (Wieslab, lund, sweden) was used according to the manufacturer's instructions.
Results:
FIG. 12A illustrates the level of MAC deposition under lectin pathway-specific assay conditions in the presence or absence of anti-MASP-2 antibody (OMS 646). FIG. 12B illustrates the level of MAC deposition under classical pathway-specific assay conditions in the presence or absence of anti-MASP-2 antibody (OMS 646). FIG. 12C illustrates the level of MAC deposition under alternative pathway-specific assay conditions with or without anti-MASP-2 antibody (OMS 646).
As shown in FIG. 12A, OMS646 has an IC of about 1nM 50 Values block lectin pathway-mediated activation of MAC deposition. However, OMS646 had no effect on MAC deposition resulting from classical pathway-mediated activation (fig. 12B) or from alternative pathway-mediated activation (fig. 12C).
Pharmacokinetics and pharmacodynamics of OMS646 following Intravenous (IV) or Subcutaneous (SC) administration to miceThe Pharmacokinetics (PK) and Pharmacodynamics (PD) of OMS646 were evaluated in a 28 day single dose PK/PD study in mice. The study tested dosage levels of OMS646 given Subcutaneously (SC) at 5mg/kg and 15mg/kg and OMS646 given Intravenously (IV) at 5 mg/kg.
Regarding PK profile of OMS646, fig. 13 graphically illustrates OMS646 concentration (n=3 animals/group average) as a function of time after administration of a given dose of OMS 646. As shown in FIG. 13, OMS646 reached a maximum plasma concentration of 5-6 μg/mL approximately 1-2 days after administration of 5mg/kg SC. The bioavailability of OMS646 was approximately 60% at 5mg/kg SC. As further shown in FIG. 13, OMS646 reached a maximum plasma concentration of 10-12 μg/mL approximately 1-2 days after administration of 15mg/kg SC. For all groups, OMS646 cleared slowly from the systemic circulation, with a terminal half-life of about 8-10 days. OMS646 is characterized as typical for human antibodies in mice.
The PD activity of OMS646 is shown in fig. 14A and 14B. FIGS. 14A and 14B show PD responses (decrease in systemic lectin pathway activity) for each mouse in the 5mg/kg IV (FIG. 14A) and 5mg/kg SC (FIG. 14B) groups. The dashed line represents the baseline measured (maximum inhibition; in vitro addition of excess OMS646 naive mouse serum prior to the test). As shown in FIG. 14A, the systemic lectin pathway activity immediately decreased to almost undetectable levels following IV administration of 5mg/kg OMS646, and the lectin pathway activity showed only modest recovery over the 28 day observation period. As shown in FIG. 14B, a time-dependent inhibition of lectin pathway activity was observed in mice given 5mg/kg OMS646 SC. Lectin pathway activity decreased to almost undetectable levels and remained low for at least 7 days within 24 hours of drug administration. Lectin pathway activity gradually increased over time, but did not return to pre-dosing levels during the 28 day observation period. The lectin pathway activity versus time profile observed after administration of 15mg/kg SC was similar to that of 5mg/kg SC dose (data not shown), indicating saturation of the PD endpoint. The data further indicate that weekly doses of 5mg/kg OMS646 given IV or SC are sufficient to achieve sustained inhibition of systemic lectin pathway activity in mice.
Example 13
This example describes the production of recombinant antibodies that inhibit MASP-2 comprising heavy and/or light chain variable regions (comprising one or more CDRs that specifically bind MASP-2) and at least one SGMI core peptide sequence (also referred to as MASP-2 antibodies with SGMI peptides or antigen binding fragments thereof).
Background/rationale: …
The production of specific inhibitors of MASP-2, known as SGMI-2, is described in Heja et al, J Biol Chem 287:20290 (2012) and Heja et al, PNAS 109:10498 (2012), each of which is incorporated herein by reference. SGMI-2 is a 36 amino acid peptide selected from a phage library of variants of grasshopper (Schistocerca gregaria) protease inhibitor 2, in which 6 of the 8 positions of the protease binding loop are completely randomized. Subsequent in vitro evolution gave a monospecific inhibitor with single digit nM K I Values (Heja et al, J.biol. Chem.287:20290, 2012). Structural studies have shown that optimized protease binding loop formation defines the primary binding site for specificity of both inhibitors. The amino acid sequences of the extended secondary and internal binding regions are common to both inhibitors and contribute to the contact interface (Heja et al 2012.j.biol. Chem. 287:20290). Mechanistically, SGMI-2 blocks complement The lectin pathway is activated without affecting the classical pathway (Heja et al 2012.Proc. Natl. Acad. Sci.109:10498).
The amino acid sequence of the SGMI-2 inhibitor is shown below:
SGMI-2-full length:
LEVTCEPGTTFKDKCNTCRCGSDGKSAVCTKLWCNQ(SEQ ID NO:72)
SGMI-2-medium:
TCEPGTTFKDKCNTCRCGSDGKSAVCTKLWCNQ(SEQ ID NO:73)
SGMI-2-short:
………………………………TCRCGSDGKSAVCTKLWCNQ(SEQ ID NO:74)
as described in this example, and in WO2014/144542, MASP-2 antibodies and fragments thereof carrying an SGMI-2 peptide are produced by fusing an amino acid sequence (e.g., SEQ ID NO:72, 73 or 74) of the SGMI-2 peptide to the amino or carboxy terminus of the heavy and/or light chain of a human MASP-2 antibody. MASP-2 antibodies and fragments bearing the SGMI-2 peptide have improved inhibitory activity compared to naked MASP-2 scaffold antibodies that do not comprise the SGMI-2 peptide sequence when measured in a C3b or C4b deposition assay using human serum, as described in WO2014/144542, and also have improved inhibitory activity compared to naked MASP-2 scaffold antibodies when measured in a mouse in vivo model. Methods for producing MASP-2 antibodies with SGMI-2 peptides are described below.
Method
Expression constructs encoding four exemplary MASP-2 antibodies with SGMI-2 peptides were generated in which the SGMI-2 peptides were fused to the N-or C-terminus of the heavy or light chain of a representative MASP-2 inhibitory antibody OMS646 (generated as described in example 12).
Table 10: MASP-2 antibody/SGMI-2 fusion
Abbreviations in table 10:
"H-N" = amino terminus of heavy chain
"H-C" = carboxyl terminus of heavy chain
"L-N" = amino terminus of light chain
"L-C" = carboxyl terminus of light chain
"M2" =masp-2 ab scaffold (representative OMS 646)
For the N-terminal fusions shown in Table 10, a peptide linker ('GTGGGSGSSS' SEQ ID NO: 79) was added between the SGMI-2 peptide and the variable region.
For the C-terminal fusions shown in Table 10, a peptide linker ('AAGGSG' SEQ ID NO: 80) was added between the constant region and the SGMI-2 peptide, and a second peptide "GSGA" (SEQ ID NO: 81) was added at the C-terminus of the fusion polypeptide to protect the C-terminal SGMI-2 peptide from degradation.
For the following representative MASP-2 antibody/SGMI-2 fusion, the amino acid sequences are provided below:
H-M2ab6-SGMI-2-N (SEQ ID NO:75, encoded by SEQ ID NO: 82):
[491aa protein, aa 1-36=sgmi-2 (underlined), aa 37-46=linker (italic), aa 47-164=heavy chain variable region of MASP-2ab#6 (underlined), aa 165-491=igg 4 constant region with hinge mutation ]
H-M2ab6-SGMI-2-C (SEQ ID NO:76, encoded by SEQ ID NO: 83):
[491aa protein, aa 1-118=masp-2ab#6 heavy chain variable region (underlined), aa 119-445=igg 4 constant region with hinge mutation, aa 446-451=1st linker (italic), aa 452-487=sgmi-2, aa488-491=2nd linker (italic) ]
L-M2ab6-SGMI-2-N (SEQ ID NO:77, encoded by SEQ ID NO: 84):
[258aa protein, aa 1-36=sgmi-2 (underlined), aa 37-46=linker (italic), aa 47-152=light chain variable region of MASP-2ab#6 (underlined), aa 153-258=human igλ constant region ]
L-M2ab6-SGMI-2-C(SEQ ID NO:78、encodedbySEQ ID NO:85):
[258aa protein, aa 1-106=MASP-2 ab#6 light chain variable region (underlined), aa 107-212=human Ig lambda constant region, aa 213-218=1st linker, aa 219-254=SGMI-2, aa255-258=2nd linker ]
Functional assay:
four MASP-2-SGMI-2 fusion antibody constructs were transiently expressed in an Expi293F cell (Invitrogen), purified by protein A affinity chromatography, and tested for inhibition of C3b deposition in 10% normal human serum in the mannan coated bead assay described below.
MASP-2-SGMI-2 fusion was tested in a mannan coated bead assay for C3b depositionLectin pathway inhibition of MASP-2-SGMI-2 fusion antibodies was assessed on mannan-coated beads in a C3b deposition assay. The assay provides a specific ratio by determining the extent of activity by flow cytometryThe assay is of greater resolution. Lectin pathway bead assay was performed as follows: mannan was adsorbed to 7 μm diameter polystyrene beads (Bangs Laboratories; fishers, IN, USA) overnight at 4 ℃ IN carbonate-bicarbonate buffer (pH 9.6). Beads were washed in PBS and exposed to 10% human serum, or 10% serum pre-incubated with antibodies or inhibitors. The serum-bead mixture was incubated for 1 hour at room temperature while stirring. After serum incubation, the beads were washed and C3b deposition on the beads was measured by detection with anti-C3C rabbit polyclonal antibody (Dako North America; carpinteria, calif., USA) and PE-Cy5 conjugated goat anti-rabbit secondary antibody (Southern Biotech; birmingham, AL, USA). After the staining procedure, analysis was performed using a FACSCalibur flow cytometer And (3) beads. The beads were gated as a uniform population using forward and side scatter, and C3b deposition was evident as FL 3-positive particles (FL-3 or "FL-3 channel" means the 3 rd or red channel on the cytometer). For each experimental condition, the geometric Mean Fluorescence Intensity (MFI) of the population was plotted against the antibody/inhibitor concentration to evaluate lectin pathway inhibition.
IC 50 Values were calculated using GraphPad PRISM software. In particular, IC 50 Values were obtained by applying a variable slope (four parameters), a non-linear fit to the log (antibody) and mean fluorescence intensity curve obtained from the cell count assay.
The results are shown in Table 11.
Table 11: c3b deposition in 10% human serum (mannan coated bead assay)
Constructs IC 50 (nM)
Naked N2ab (mAb # 6) ≥3.63nM
H-M2-SGMI-2-N 2.11nM
L-M2-SGMI-2-C 1.99nM
H-M2-SGMI-2-N 2.24nM
L-M2-SGMI-2-N 3.71nM
Results:
control, MASP-2 "naked" scaffold antibody (mAb # 6) without SGMI, which is inhibitory in this assay, has an IC50 value of > 3.63nM, consistent with the inhibitory results observed in example 12. Clearly, as shown in table 11, all SGMI-2-MASP-2 antibody fusions tested improved the efficacy of MASP-2 scaffold antibodies in this assay, indicating that increased cost may also be beneficial for inhibition of C3b deposition.
MASP-2-SGMI-assay with 10% human serum in the mannan-coated bead assay for C4b deposition 2 fusionsThe C4b deposition assay was performed with 10% human serum using the same assay conditions as described above for the C3b deposition assay, with the following modifications. C4b detection and flow cytometry analysis were performed by staining the deposition reaction with anti-C4 b mouse monoclonal antibody (1:500, quidel) and staining with goat anti-mouse F (ab') 2 secondary antibody conjugated to PE Cy5 (1:200,Southern Biotech) prior to flow cytometry analysis.
Results:
MASP-2-N-terminal antibody fusions with SGMI-2 (H-M2-SGMI-2-N: IC50=0.34 nM), L-M2-SGMI-2-N: IC50=0.41 nM) all have increased potency compared to MASP-2 scaffold antibodies (HL-M2: IC50=0.78 nM).
Similarly, MASP-2 scaffold antibodies (HL-M2: IC 50 =1.2 nM) with a single SGMI-2 (H-M2-SGMI-2-C: IC) 50 =0.45 nM and L-M2-SGMI-2c ic 50 =0.47 nM) all had increased potency.
MASP-2-SGMI-2 was tested in a mannan-coated bead assay for C3b deposition with 10% mouse serum Fusion ofMannan-coated bead assays for C3b deposition were performed with 10% mouse serum as described above. Similar to the results observed in human serum, MASP-2 fusions with SGMI-2 were determined to have increased potency in mouse serum compared to MASP-2 scaffold antibodies.
Summary of results: the results of this example demonstrate that all SGMI-2-MASP-2 antibody fusions tested improved the efficacy of MASP-2 scaffold antibodies.
Example 14
This example provides results generated using a Unilateral Ureteral Obstruction (UUO) model of renal fibrosis in MASP-2-/-deficient and MASP-2+/+ full mice to evaluate the role of the lectin pathway in renal fibrosis.
Background/rationale:
renal fibrosis and inflammation are the major features of advanced renal disease. Tubular interstitial fibrosis is a progressive process involving sustained cell injury, abnormal healing, activation of resident and infiltrating kidney cells, cytokine release, inflammation and activation of kidney cell phenotypes to produce extracellular matrix. Tubular Interstitial (TI) fibrosis is a common endpoint of a variety of renal pathologies and represents a key target for potential therapies aimed at preventing progressive impairment of renal function in Chronic Kidney Disease (CKD). Renal TI injury is closely associated with reduced renal function in glomerular disease (Risdon R.A. et al, lancet 1:363-366, 1968;Schainuck L.I. Et al, hum Pathol 1:631-640, 1970; nath K.A., am JKidDis 20:1-17, 1992), and is a characteristic of CKD in which myofibroblasts accumulate and potential space between the tubules and capillaries surrounding the tubes is occupied by a matrix containing collagen and other proteoglycans. The source of TI myofibroblasts remains widely controversial, but fibrosis is usually followed by inflammation, initially characterized by TI accumulation of T lymphocytes followed by macrophages (Liu Y. Et al, nat Rev Nephrol 7:684-696, 2011;Duffield J.S, JClin Invest 124:2299-2306, 2014).
Rodent models of UUO produce progressive renal fibrosis, a hallmark of progressive renal disease of essentially any etiology (Chevalier et al Kidney International 75:1145-1152, 2009). Increased C3 gene expression has been reported in wild-type mice following UUO, and collagen deposition was significantly reduced in C3-/-knockout mice following UUO compared to wild-type mice, indicating a role of complement activation in renal fibrosis (Fearn et al Mol Immunol 48:1666-1733, 2011). It has also been reported that C5 defects result in significant improvement of the major component of renal fibrosis in models of tubular interstitial injury (Boor P. Et al J ofAm Soc ofNephrology:18:1508-1515, 2007). However, prior to the studies described herein by the inventors, the specific complement components involved in renal fibrosis have not been determined. Thus, the following study was conducted to evaluate MASP-2 (-/-) and MASP-2 (+/+) male mice in a Unilateral Ureteral Obstruction (UUO) model.
The method comprises the following steps:
MASP-2-/-mice were generated as described in example 1 and backcrossed to C57BL/6 for 10 passages. Male wild-type (WT) C57BL/6 mice and homozygous MASP-2 deficient (MASP-2-/-) mice on the C57BL/6 background were kept under standard conditions for 12/12 day/night cycles, fed standard feed pellets and were free to gain food and water. 10 week old mice, 6 per group, were anesthetized with 2.5% isoflurane in 1.5L/min oxygen. Two groups of 10 week old male C56/BL6 mice (wild type and MASP-2-/-) were surgically ligated right ureters. The right kidney was exposed through a 1cm lateral incision. The right ureter was completely blocked at two points using 6/0polyglactin sutures. Buprenorphine analgesia is provided every 12 hours before and after surgery, with up to 5 doses depending on the pain score. The topical bupivacaine anesthetic is administered once during surgery.
Mice were sacrificed 7 days post-surgery and kidney tissue was collected, fixed and embedded in paraffin blocks. Blood was collected from mice under anesthesia by heart puncture, and mice were eliminated by apheresis after nephrectomy. The blood was allowed to set on ice for 2 hours and serum was separated by centrifugation and kept frozen in aliquots at-80 ℃.
Immunohistochemistry of kidney tissue
To measure the extent of renal fibrosis indicated by collagen deposition, 5 micron paraffin-embedded kidney sections were stained with sirius red, a collagen specific dye, as described by Whittaker p. Et al, basic Res Cardiol 89:397-410, 1994. Briefly, kidney sections were deparaffinized, rehydrated and collagen stained with aqueous sirius red in 500mL of saturated aqueous picric acid (0.5 gm sirius red, sigma, dorset UK) for 1 hour. Slides were washed twice in acidified water (0.5% glacial acetic acid/distilled water), each for 5 minutes, then rehydrated and fixed.
To measure the degree of inflammation indicated by macrophage infiltration, kidney sections were stained with macrophage-specific antibody F4/80 as follows. Formalin-fixed, paraffin-embedded 5 micron kidney sections were deparaffinized and rehydrated. Antigen recovery was performed in citrate buffer at 95 ℃ for 20 min, followed by antigen recovery at 3%H 2 O 2 Quenching endogenous peroxidase activity by 10 minutes of incubation. Tissue sections were incubated in blocking buffer (10% heat-inactivated normal goat serum and 1% bovine serum albumin/Phosphate Buffered Saline (PBS)) for 1 hour at room temperature, followed by avidin/biotin blocking. Tissue sections were washed three times in PBS after each step for 5 minutes. F4/80 macrophage primary antibody (Santa Cruz, dallas, TX, USA) diluted 1:100 in blocking buffer was applied for 1 hour. A 1:200 dilution of biotinylated goat anti-rat secondary antibody was then applied for 30 minutes followed by horseradish peroxidase (HRP) conjugated enzyme for 30 minutes. Staining was performed using Diaminobenzidine (DAB) substrate (Vector Labs, peterborough UK) for 10 minutes and the slides were washed in water, rehydrated and fixed without counterstaining to facilitate computer-based analysis.
Image analysis
The percentage of renal cortex staining was determined as described by Furness P.N. et al, J Clin Pathol 50:118-122, 1997. Briefly, 24 color images were captured from a continuous, non-overlapping field of view of the renal cortex immediately below the renal capsule around the entire periphery of the renal slice. After each image capture, the NIH image is used to extract the red channel as an 8-bit monochromatic image. The non-uniformity of the background illumination is subtracted using a pre-recorded image of a bright microscope field without a slice in place. The image is subjected to a fixed threshold to identify areas of the image corresponding to positive staining. The percentage of black pixels is then calculated and after all images around the kidney have been measured in this way, the average percentage is recorded, providing a value corresponding to the percentage of the stained area in the kidney section.
Gene expression analysis
Expression of several genes for kidney inflammation and fibrosis in mouse kidney was measured by quantitative PCT (qPCR) as follows. Total RNA use from renal cortex(ThermoFisher Scientific, paisley, UK) was isolated according to the manufacturer's instructions. The extracted RNA was treated with a Turbo DNA-free kit (ThermoFisher Scientific) to eliminate DNA contamination, and then AMV Reverse Transcription System (Promega, madison, wis., USA) was used to synthesize first strand cDNA. cDNA integrity was confirmed by a single qPCR reaction using TaqMan GAPDH Assay (Applied Biosystems, paisley UK) followed by a qPCR reaction using Custom TaqMan Array 96-well plates (Life Technologies, paisley, UK).
In this analysis 12 genes were studied:
collagen IV type alpha 1 (col 4 alpha 1; measuring ID: mm01210125 _m1)
Transforming growth factor beta-1 (TGF beta-1; assay ID: mm01178820 _m1);
cadherin 1 (Cdh 1; assay ID: mm01247357 _m1);
fibronectin 1 (Fn 1; assay ID: mm01256744 _m1);
interleukin 6 (IL 6; assay ID Mm00446191 _m1);
interleukin 10 (IL 10; assay ID Mm00439614 _m1);
interleukin 12a (IL 12a; assay ID Mm00434165 _m1);
vimentin (Vim; assay ID Mm01333430 _m1);
Actin α1 (Actn 1; assay ID Mm01304398 _m1);
tumor necrosis factor-alpha (TNF-alpha; assay ID Mm00443260 _g1)
Complement component 3 (C3; assay ID Mm00437838 _m1);
interferon gamma (Ifn-gamma; determination of ID Mm 01168134)
The following housekeeping control genes were used:
glyceraldehyde-3-phosphate dehydrogenase (GAPDH; assay ID Mm99999915 _g1);
glucuronidase beta (Gusbeta; assay ID Mm00446953 _m1);
eukaryotic 18S rRNA (18S; assay ID Hs99999901 _s1);
inosine guanine phosphoribosyl transferase (HPRT; determination of ID Mm00446968 _m1)
mu.L of the reaction was amplified for 40 cycles using TaqMan Fast Universal Master Mix (Applied Biosystems). Real-time PCR amplification data was analyzed using Applied Biosystems 7000sds v1.4 software.
Results:
following Unilateral Ureteral Obstruction (UUO), the obstructed kidney undergoes inflammatory cell, particularly macrophage, influx followed by rapid fibrosis as evidenced by collagen accumulation and tubular dilation and thinning of the proximal tubular epithelium (see Chevalier r.l. Et al, kidneyInt 75:1145-1152, 2009).
FIG. 15 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, wherein the tissue sections were obtained from wild-type and MASP-2-/-mice 7 days after ureteric obstruction (UUO) or from sham-operated control mice. As shown in fig. 15, kidney sections of wild-type mice 7 days after ureteral obstruction showed significantly greater collagen deposition than MASP-2-/-mice (p-value = 0.0096). The mean ± standard error of the mean for UUO operated mice in wild type and MASP-2-/-groups were 24.79 ± 1.908 (n=6) and 16.58 ± 1.3 (n=6), respectively. As further shown in fig. 15, tissue sections from sham-control wild-type and sham-control MASP-2-/-mice showed very low levels of collagen staining, as expected.
FIG. 16 illustrates the results of computer-based image analysis of kidney tissue sections stained with F4/80 macrophage specific antibody, wherein the tissue sections were obtained from wild-type and MASP-2-/-mice 7 days after ureteric obstruction or from sham-operated control mice. As shown in FIG. 16, tissues of UUO kidney obtained from MASP-2-/-mice showed significantly less macrophage infiltration after ureteric obstruction for 7 days compared to wild-type mice (% macrophage staining area was WT: 2.23.+ -. 0.4 vs. MASP-2-/-: 0.53.+ -. 0.06, p=0.0035). As further shown in FIG. 16, tissue sections from sham wild type and sham MASP-2-/-mice indicated no detectable macrophage staining.
Gene expression analysis of the various genes associated with kidney inflammation and fibrosis was performed in kidney tissue sections obtained from wild-type and MASP-2-/-mice 7 days after ureteral obstruction and sham-operated wild-type and MASP-2-/-mice. The data shown in fig. 17-20 are the standard error of Log10 and bar representation mean values for the relative numbers of wild-type sham surgical samples. With respect to the results of gene expression analysis of fibrosis-related genes, fig. 17 illustrates the relative mRNA expression levels of collagen IV type α1 (collagen-4) as measured by qPCR in kidney tissue sections obtained from wild-type and MASP-2-/-mice and sham-operated control mice 7 days after ureteric obstruction. FIG. 18 illustrates the relative mRNA expression levels of transforming growth factor beta-1 (TGF beta-1) as measured by qPCR in kidney tissue sections obtained from wild-type and MASP-2-/-mice 7 days after ureteric obstruction and sham-operated control mice. As shown in fig. 17 and 18, the obstructed kidney from wild-type mice demonstrated a significant increase in expression of fibrosis-related genes collagen type IV (fig. 17) and tgfβ -1 (fig. 18) compared to sham operated kidney of wild-type mice, confirming that these fibrosis-related genes were induced following UUO injury in wild-type mice, as expected. In contrast, as further shown in fig. 17 and 18, the obstructed kidney from UUO-damaged MASP-2-/-showed significantly reduced expression of collagen type IV (fig. 17, p=0.0388) and tgfβ -1 (fig. 18, p=0.0174) compared to UUO-damaged wild-type mice.
With respect to the results of gene expression analysis of inflammation-related genes, fig. 19 illustrates the relative mRNA expression levels of interleukin-6 (IL-6), as measured by qPCR in kidney tissue sections obtained from wild-type and MASP-2-/-mice after 7 days of ureteric obstruction, as well as sham-operated control mice. FIG. 20 illustrates relative mRNA expression levels of interferon-gamma as measured by qPCR in kidney tissue sections obtained from wild-type and MASP-2-/-mice and sham-operated control mice 7 days after ureteric obstruction. As shown in fig. 19 and 20, the obstructed kidney from wild-type mice demonstrated a significant increase in the expression of the inflammation-associated genes interleukin-6 (fig. 19) and interferon-gamma (fig. 20) compared to the sham operated kidney of wild-type mice, confirming that these inflammation-associated genes were induced following UUO injury in wild-type mice. In contrast, as further shown in fig. 19 and 20, the obstructed kidney from UUO-damaged MASP-2-/-showed significantly reduced expression of interleukin-6 (fig. 19, p=0.0109) and interferon- γ (fig. 20, p=0.0182) compared to UUO-damaged wild-type mice.
Note that gene expression was all found to be significantly up-regulated for Vin, actn-1, TNFα, C3 and IL-10 in UFO kidneys obtained from both wild-type and MASP-2-/-mice, with no significant difference in the expression levels of these specific genes between wild-type and MASP-2-/-mice (data not shown). Gene expression levels of Cdh-1 and IL-12a were unchanged in the obstructed kidneys from animals of any group (data not shown).
Discussion:
rodent UUO models are recognized to induce early, active and significant injury in the obstructed Kidney, with reduced renal blood flow, interstitial inflammation and rapid fibrosis within 1-2 weeks after the obstruction, and have been widely used to understand the common mechanisms and mediators of Kidney inflammation and fibrosis (see, e.g., chevalier r.l., kidney Int 75:1145-1152, 2009; yang H. Et al, drug Discov TodayDisModels 7:13-19, 2010).
The results described in this example demonstrate that there is a significant reduction in collagen deposition and macrophage infiltration in UUO-operated kidneys of MASP-2 (-/-) mice relative to wild-type (+/-) control mice. This unexpected result, which showed a significant reduction in kidney injury in MASP-2-/-animals at 2 levels of histological and gene expression, demonstrated that the complement-activated lectin pathway significantly promoted the occurrence of inflammation and fibrosis in the obstructed kidney. While not wishing to be bound by a particular theory, it is believed that the lectin pathway decisively promotes the pathophysiology of fibrotic diseases by triggering and maintaining pro-inflammatory stimuli that maintain a vicious circle as cellular injury drives inflammation, which in turn causes further cellular injury, scarring and tissue loss. Based on these results, inhibition or blocking of MASP-2 with an inhibitor would be expected to have preventive and/or therapeutic effects in inhibiting or preventing renal fibrosis, as well as in inhibiting or preventing general fibrosis (i.e., independent of tissue or organ).
Example 15
This example describes the analysis of the efficacy of monoclonal MASP-2 inhibitory antibodies in a Unilateral Ureteral Obstruction (UUO) model, a murine model of renal fibrosis.
Background/basic principle
Improvement of tubular interstitial fibrosis (a common endpoint of multiple renal pathologies) represents a key target for therapeutic strategies aimed at preventing progressive renal diseases. In view of the lack of new and existing treatments targeting the inflammatory pro-fibrotic pathway of renal disease, there is an urgent need to develop new therapies. Many patients with proteinuria kidney disease show tubular interstitial inflammation and progressive fibrosis, which is closely accompanied by reduced renal function. Proteinuria itself induces the occurrence of tubular interstitial inflammation and proteinuria nephropathy (Brunskill N.J. et al, JAm Soc Nephrol 15:504-505, 2004). Regardless of the primary renal disease, tubular interstitial inflammation and fibrosis is always seen in patients with progressive renal injury and is closely related to reduced excretory function (Risdon r.a. et al, lancet 1:363-366, 1968;Schainuck L.I. Et al, hum Pathol 1:631-640, 1970). Therapies with the potential to block critical common cellular pathways leading to fibrosis hold the promise of widespread use of renal disorders.
As described in example 14, MASP-2-/-mice were determined to exhibit significantly less renal fibrosis and inflammation compared to wild-type control animals in a UUO model of non-proteinuria renal fibrosis, as evidenced by inflammatory cell infiltration (75% reduction) and histological markers of fibrosis, e.g., collagen (1/3 reduction), establishing a key role for the lectin pathway in renal fibrosis.
Monoclonal MASP-2 antibodies (OMS 646-SGMI-2 fusions comprising SGMI-2 peptide fused to the C-terminus of the heavy chain of OMS 646) specifically blocking human lectin pathway function have also been shown to block lectin pathway in mice as described in example 13. In this example, OMS646-SGMI-2 was analyzed in a UFO mouse model of kidney fibrosis in wild type mice to determine whether a specific inhibitor of MASP-2 was able to inhibit kidney fibrosis.
The method comprises the following steps:
the present study evaluated the effect of MASP-2 inhibitory antibodies (10 mg/kg OMS 646-SGMI-2) in male WT C57BL/6 mice compared to human IgG4 isotype control antibody (10 mg/kg ET 904) and vehicle control. Antibody (10 mg/kg) was administered to 9 mice per group by intraperitoneal (ip) injection on day 7, 4 and 1 before UUO surgery and on day 2 after surgery. Blood samples were obtained to evaluate lectin pathway functional activity prior to antibody administration and at the end of the experiment.
UUO surgery, tissue collection and staining with sirius red and macrophage specific antibody F4/80 were performed using the method described in example 14.
The hydroxyproline content of the kidney of the mice was measured using a specific colorimetric assay kit (Sigma) according to the manufacturer's instructions.
To evaluate the pharmacodynamic effects of MASP-2 inhibitory mAbs in mice, systemic lectin pathway activity was evaluated by quantifying lectin-induced C3 activation in minimal diluted serum samples collected at the indicated time after i.p. administration of MASP-2mAb or control mAb to mice. Briefly, 7. Mu.M diameter polystyrene microspheres (Bangs Laboratories, fisher IN, USA) were coated with mannan by overnight incubation with 30. Mu.g/mL mannan (Sigma) IN sodium bicarbonate buffer (pH 9.6), then washed, blocked with 1% fetal bovine serum/PBS and incubated at 1X10 8 The final concentration of individual beads/mL was resuspended in PBS. Complement deposition reactions were started by adding 2.5 μl of mannan-coated beads (-250,000 beads) to 50 μl of the minimum diluted mouse serum sample (90% final serum concentration) followed by incubation at 4 ℃ for 40 minutes. After the sedimentation reaction was stopped by adding 250. Mu.L of ice-cold flow cytometry buffer (FB: PBS containing 0.1% fetal bovine serum), the beads were collected by centrifugation and washed twice more with 300. Mu.L ice-cold FB.
To quantify lectin-induced C3 activation, beads were incubated with 50 μl of rabbit anti-human C3C antibody (Dako, carpenter ia, CA, USA) diluted in FB for 1 hour at 4 ℃. After washing twice with FB to remove unbound material, the beads were incubated with 50 μl of goat anti-rabbit antibody conjugated to PE-Cy5 (Southern Biotech, birmingham, AL, USA) diluted in FB for 30 min at 4 ℃. After washing twice with FB to remove unbound material, the beads were resuspended in FB and analyzed by FACS Calibur cytometry. Beads were gated as a uniform population using forward and side scatter, and C3b deposition in each sample was quantified as Mean Fluorescence Intensity (MFI).
Results:
evaluation of collagen deposition:
fig. 21 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, wherein tissue sections were obtained from wild-type mice treated with MASP-2 inhibitory antibodies or isotype control antibodies 7 days after ureteral obstruction. As shown in fig. 21, tissue sections of kidney harvested 7 days after obstruction (UUO) from wild-type mice treated with MASP-2 inhibitory antibodies showed a significant reduction in collagen deposition compared to the amount of collagen deposition obtained from the tissue sections of obstructed kidney from wild-type mice treated with IgG4 isotype control (p=0.0477).
Evaluation of hydroxyproline content:
hydroxyproline was measured in kidney tissue as an indication of collagen content. Hydroxyproline is a parameter that is highly indicative of the pathophysiological progression of the disease induced in this model.
Figure 22 illustrates hydroxyproline content of kidneys harvested 7 days after obstruction (UUO) from wild-type mice treated with MASP-2 inhibitory antibodies or isotype control antibodies. As shown in figure 22, obstructed kidney tissue from mice treated with MASP-2 inhibitory antibodies demonstrated significantly less hydroxyproline (an indication of collagen content) (p= 0.0439) than kidney from mice treated with IgG4 isotype control mAb.
Evaluation of inflammation:
obstructed kidneys from wild-type, isotype control antibody-treated animals and wild-type animals treated with MASP-2 inhibitory antibodies confirm active infiltration of macrophages. Careful quantification showed no significant difference in percent macrophage staining area between the two groups (data not shown). However, despite the equal amount of infiltrating macrophages, the obstructed kidney from MASP-2 inhibitory antibody-injected animals showed significantly less fibrosis than the obstructed kidney from isotype control-injected animals, as judged by sirius red staining, consistent with the result that the obstructed kidney tissue from mice treated with MASP-2 inhibitory antibodies had significantly less hydroxyproline than the kidney treated with IgG4 isotype control mAb.
Discussion of the invention
The results described in this example demonstrate that the use of MASP-2 inhibitory antibodies provides protection against renal fibrosis in a UUO model, consistent with the results described in example 14, example 14 demonstrates that MASP-2-/-mice have significantly reduced renal fibrosis and inflammation in the UUO model compared to wild-type mice. The results of this example show reduced fibrosis in mice treated with MASP-2 inhibitory antibodies. The finding of reduced fibrosis in the UUO kidney of animals that reduce or block MASP-2-dependent lectin pathway activity is a very significant new finding. Taken together, the results provided in example 14 and this example demonstrate the beneficial effects of MASP-2 inhibition on tubular interstitial inflammation, tubular cell injury, pro-fibrotic cytokine release and scarring. The reduction of renal fibrosis remains a key target for renal therapy. The UUO model is an accurate model for accelerating renal fibrosis and interventions that reduce fibrosis in this model, such as the use of MASP-2 inhibitory antibodies, may be used to inhibit or prevent renal fibrosis. Results from the UUO model may be transferred to renal diseases characterized by glomerular and/or proteinuria tubular injury.
Example 16
This example provides results generated using the proteinuria model of renal fibrosis, inflammation and tubular interstitial injury in MASP-2-/-and wild-type mice to evaluate the role of the lectin pathway in proteinuria.
Background/basic principle
Proteinuria is a risk factor for the occurrence of renal fibrosis and loss of renal excretion function, regardless of the primary renal disease (Tryggvason K. Et al, JInter Med 254:216-224, 2003, williams M., am J. Nephrol 25:77-94, 2005). The concept of proteinuria nephropathy describes the toxic effects of excess protein entering the proximal tubules as a result of impaired glomerular permselectivity (Brunskill n.j., JAm Soc Nephrol 15:504-505, 2004, baines r.j., nature Rev Nephrol 7:177-180, 2011). This phenomenon, common to many glomerular diseases, leads to a pro-inflammatory scarring environment in the kidney and is characterized by deregulation of the signaling pathway due to stimulation of proteinuria tubule fluid, alterations in proximal tubule cell growth, apoptosis, gene transcription and inflammatory cytokine production. Proteinuria nephropathy is known as a key contributor to progressive kidney injury common to various primary renal pathologies.
Chronic kidney disease affects more than 15% of the adult population in the united states and is responsible for approximately 750,000 deaths annually worldwide (Lozano r et al, lancet vol 380,Issue 9859:2095-2128, 2012). Proteinuria is an indication of chronic kidney disease and a factor that contributes to disease progression. Many patients with proteinuria kidney disease show tubular interstitial inflammation and progressive fibrosis, which is closely related to reduced renal function. Proteinuria itself induces the occurrence of tubular interstitial inflammation and proteinuria nephropathy (Brunskill N.J. et al, J Am Soc Nephrol 15:504-505, 2004). In proteinuria kidney disease, excess albumin and other macromolecules filter through the glomeruli and are reabsorbed by proximal tubular epithelial cells. This results in inflammatory malignancy cycles mediated by complement activation, resulting in cytokine and leukocyte infiltration, which causes interstitial damage and fibrosis of the renal tubules, thereby accentuating proteinuria and resulting in loss of renal function and eventual progression to end stage renal failure (see, e.g., clark et al Canadian Medical Association Journal 178:173-175, 2008). Therapies that regulate this deleterious circulation of inflammation and proteinuria are expected to improve the outcome of chronic kidney disease.
Based on the beneficial results of MASP-2 inhibition in the UUO model of tubular interstitial injury, the following experiments were conducted to determine whether MASP-2 inhibition would reduce kidney injury in the protein overload model. This study utilized protein overload to induce proteinuria kidney disease as described in Ishola et al European RenalAssociation 21:591-597, 2006.
Method:
MASP-2-/-mice were generated and backcrossed to BALB/c for 10 passages as described in example 1. Current studies compare the results of wild-type and MASP-2-/-BALB/c mice in a protein overload proteinuria model as follows.
To see the best response, one week prior to the experiment, mice were unilaterally nephrectomized prior to protein overload challenge. The proteinuria inducing agent used was low endotoxin bovine serum albumin (BSA, sigma), WT (n=7) and MASP-2-/-mice (n=7) were given i.p. in normal saline at the following doses: one dose of 2mg BSA/gm, 4mg BSA/gm, 6mg BSA/gm, 8mg BSA/gm, 10mg BSA/gm and 12mg BSA/gm body weight and 9 doses of 15mg BSA/gm body weight were each administered for 15 days for a total of i.p. Control WT (n=4) and MASP-2-/- (n=4) mice received i.p-administered saline alone. Animals were housed individually in metabolic cages for 24 hours after the last dose was given to collect urine. Blood was collected by cardiac puncture under anesthesia, allowed to coagulate on ice for 2 hours and serum was separated by centrifugation. Serum and urine samples were collected at the end of the day 15 experiment, stored and frozen for analysis.
Mice were sacrificed 24 hours after the last BSA administration on day 15 and various tissues were collected for analysis. Kidneys were harvested and treated for H&E and immunostaining. Immunohistochemical staining was performed as follows. Formalin-fixed, paraffin-embedded 5-micron kidney tissue sections from each mouse were deparaffinized and rehydrated. Antigen retrieval was performed in citrate buffer at 95℃for 20 min, followed by 3%H 2 O 2 The tissue was incubated for 10 minutes. The tissue was then incubated in blocking buffer (10% serum from the species from which the secondary antibody was derived and 1% BSA/PBS) with 10% avidin solution for 1 hour at room temperature. After each step the sections were washed three times in PBS for 5 minutes each. The primary antibody was then applied in blocking buffer containing 10% biotin solution for 1 hour at a concentration of 1:100 for antibodies F4/80 (Santa Cruz cat#sc-25830), TGF beta (Santa Cruz cat#sc-7892), IL-6 (Santa Cruz cat#sc-1265) and 1:50 for TNFα antibodies (Santa Cruz cat#sc-1348). Biotinylated secondary antibody was then applied for 30 minutes at 1:200 for F4/80, TGF beta and IL-6 sections and 1:100 for TNF alpha sections, followed by an additional HRP conjugated enzyme30 minutes. The slides were developed using Diaminobenzidine (DAB) substrate kit (Vector labs) for 10 minutes and washed in water, dehydrated and fixed without counterstaining to facilitate computer-based image analysis. Stained tissue sections from renal cortex were analyzed by digital image capture followed by quantification using automated image analysis software.
Apoptosis was assessed in tissue sections by staining with terminal deoxynucleotidyl transferase dUTP notch end marker (TUNEL) as follows. Apoptotic cell use in kidney sectionsPeroxidase kit (Millipore) was stained as follows. Paraffin-embedded, formalin-fixed kidney sections from each mouse were deparaffinized, rehydrated, and then protein infiltrated with proteinase K (20 μg/mL), which was applied to each sample for 15 minutes at room temperature. Samples were washed in PBS between steps. Endogenous peroxidase activity was demonstrated by the sequence of amino acid sequences at 3%H 2 O 2 The tissue was quenched by incubation for 10 min. The tissues were then incubated in equilibration buffer followed by incubation with TdT enzyme for 1 hour at 37 ℃. After washing in stop/wash buffer for 10 minutes, the anti-digoxigenin conjugate was applied for 30 minutes at room temperature, followed by washing. Color development was performed in DAB substrate kit for 4 minutes followed by washing in water. Tissues were counterstained in hematoxylin and fixed in DBX. The frequency of TUNEL stained (brown) apoptotic cells was counted manually using a Leica DBXM optical microscope under 20 high power fields from continuous selection of cortex.
Results:
the proteinuria assessment was to confirm the presence of proteinuria in mice, total protein in serum was analyzed on day 15 and total secreted protein in urine was measured in urine samples collected over 24 hours on day 15 of the study.
Figure 23 illustrates total amounts of serum proteins (mg/ml) measured on day 15 in wild-type control mice (n=2) receiving saline alone, wild-type mice (n=6) receiving BSA, and MASP-2-/-mice (n=6) receiving BSA. As shown in figure 23, in the wild type and MASP-2-/-groups, administration of BSA increased serum total protein levels to concentrations exceeding twice that of the saline-only control group, with no significant differences between the treatment groups.
Figure 24 illustrates total amount of secreted protein (mg) in urine collected over 24 hours on study day 15 from wild-type control mice (n=2), wild-type mice (n=6) and MASP-2-/-mice (n=6) receiving BSA, receiving saline alone. As shown in fig. 24, on day 15 of the study, there was an approximately 6-fold increase in total secreted protein in urine in the BSA-treated group compared to the sham-treated control group that received saline alone. The results shown in fig. 23 and 24 confirm that the proteinuria model was effective, as expected.
Histological change assessment of kidney
Fig. 25 shows representative H & E stained kidney tissue sections harvested on day 15 of protein overload study from the following mice groups: (panel a) wild-type control mice; (Panel B) MASP-2-/-control mice; (Panel C) wild-type mice treated with BSA; and (Panel D) MASP-2-/-mice treated with BSA. As shown in FIG. 25, under the same level of protein overload attack, there is a higher degree of tissue preservation in the MASP-2-/-overload group (panel D) than in the wild type overload group (panel C). For example, a tremendous expansion of Bowman's capsule in wild-type mice treated with BSA (overload) was observed compared to that of wild-type control group (panel A) (panel C). In contrast, bowman's capsule (panel D) of MASP-2-/-mice treated with the same level of BSA (overloaded) remained similar to the morphology of MASP-2-/-control mice (panel B) and wild-type control mice (panel A). As further shown in fig. 25, large protein cast structures have accumulated in the proximal and distal tubules of wild type kidney slices (panel C), which are larger and richer compared to MASP-2-/-mice (panel D).
Also note that analysis of kidney sections from this study by electron emission microscopy showed that mice treated with BSA had an overall injury to the cilia boundaries of distal and proximal tubule cells, with cell content and nuclei bursting into the lumen of the tubule. In contrast, tissue was preserved in MASP-2-/-mice treated with BSA.
Assessment of macrophage infiltration in the kidney
To measure the degree of inflammation indicated by macrophage infiltration, harvested tissue sections of the kidney were also stained with macrophage specific antibody F4/80 using the method described in Boor et al JofAm Soc ofNephrology 18:1508-1515, 2007.
Fig. 26 illustrates the results of computer-based image analysis of kidney tissue sections stained with macrophage specific antibody F4/80, showing the mean area of macrophage staining (%), where tissue sections were obtained from wild-type control mice (n=2), BSA-treated wild-type mice (n=6), and BSA-treated MASP-2-/-mice (n=5) on day 15 of the protein overload study. As shown in fig. 26, kidney tissue sections stained with F4/80 anti-macrophage antibody showed that while both groups treated with BSA showed a significant increase in kidney macrophage infiltration (measured as% F4/80 antibody stained area) compared to wild-type sham control group, a significant decrease in macrophage infiltration in tissue sections from BSA-treated MASP-2-/-mice was observed compared to macrophage infiltration in tissue sections from BSA-treated wild-type mice (p-value = 0.0345).
Fig. 27A illustrates the presence of macrophage-proteinuria correlation in each wild-type mouse treated with BSA (n=6) by plotting total secreted protein measured in urine from 24 hours samples against macrophage infiltration (average stained area%). As shown in fig. 27A, most samples from wild-type kidneys showed a positive correlation between the level of proteinuria present and the degree of macrophage infiltration.
Figure 27B illustrates analysis of the presence of macrophage-proteinuria correlation in each MASP-2-/-mouse treated with BSA (n=5) by plotting total secreted protein in urine versus macrophage infiltration (average stained area%) in 24 hour samples. As shown in FIG. 27B, no positive correlation between proteinuria levels and macrophage infiltration levels was observed in wild-type mice in MASP-2-/-mice (shown in FIG. 27A). While not wishing to be bound by any particular theory, these results may suggest that an inflammatory clearance mechanism exists in MASP-2-/-mice at high levels of proteinuria.
Assessment of cytokine infiltration Interleukin 6 (IL-6), transforming growth factor beta (TGF beta) and tumor necrosis factor alpha (TNF alpha) are pro-inflammatory cytokines that are known to be up-regulated in the proximal tubules of wild-type mice in the proteinuria model (Abbate M. Et al, journal of the American Society of Nephrology: JASN,17:2974-2984, 2006; david S. Et al, nephrology, didalysis, trans-placement, official Publication of the European Dialysis and Transplant Association-European RenalAssociation, 51-56, 1997). Tissue sections of the kidney were stained with cytokine-specific antibodies as described above.
Fig. 28 illustrates the results of computer-based image analysis of tissue sections stained with anti-tgfβ antibodies (measured as% tgfβ antibody staining area) in wild-type mice treated with BSA (n=4) and MASP-2-/-mice treated with BSA (n=5). As shown in fig. 28, a significant increase in tgfβ staining was observed in the wild-type BSA treated (overloaded) group compared to the MASP-2-/-BSA treated (overloaded) group (p=0.026).
Fig. 29 illustrates the results of computer-based image analysis of tissue sections stained with anti-tnfα antibodies (measured as% tnfα antibody staining area) in wild type mice treated with BSA (n=4) and MASP-2-/-mice treated with BSA (n=5). As shown in fig. 29, a significant increase in tnfα staining was observed in the wild-type BSA treated (overloaded) group compared to the MASP-2-/-BSA treated (overloaded) group (p=0.0303).
FIG. 30 graphically depicts the results of computer-based image analysis of tissue sections stained with anti-IL-6 antibody (measured as% IL-6 antibody staining area) in wild type control mice, MASP-2-/-control mice, wild type mice treated with BSA (n=7), and MASP-2-/-mice treated with BSA (n=7). As shown in fig. 30, a significant increase in IL-6 staining height was observed in the wild-type BSA treated group compared to the MASP-2-/-BSA treated group (p=0.0016).
Apoptosis assessment
Apoptosis was assessed in tissue sections by staining with terminal deoxynucleotidyl transferase dUTP notch end marker (TUNEL), and the frequency of TUNEL stained apoptotic cells was counted in 20 High Power Fields (HPF) from continuous selection of cortex.
Fig. 31 illustrates the frequency of TUNEL apoptotic cells counted in 20 High Power Fields (HPF) from sequential selection of tissue sections of kidney cortex of wild type control mice (n=1), MASP-2-/-control mice (n=1), wild type mice treated with BSA (n=6) and MASP-2-/-mice treated with BSA (n=7). As shown in fig. 31, a significantly higher rate of apoptosis in the cortex was observed in kidneys obtained from wild-type mice treated with BSA (p=0.0001) compared to kidneys obtained from MASP-2-/-mice treated with BSA.
Results and conclusions overall:
the results of this example demonstrate that MASP-2-/-mice have reduced kidney injury in the protein overload model. Thus, inhibitors of MASP-2, such as MASP-2 inhibitory antibodies, are expected to inhibit or prevent the deleterious circulation of inflammation and proteinuria, and to ameliorate the consequences of chronic kidney disease.
Example 17
This example describes an analysis of the efficacy of monoclonal MASP-2 inhibitory antibodies in a mouse protein overload proteinuria model of wild-type mice to reduce and/or prevent kidney inflammation and tubular interstitial injury.
Background/rationale:
as described in example 16, in the protein overload model of proteinuria, MASP-2-/-mice were determined to show significantly better results (e.g., less tubular interstitial damage and less renal inflammation) than wild-type mice, suggesting a pathogenic role for the lectin pathway in proteinuria renal disease.
Monoclonal MASP-2 inhibitory antibodies (OMS 646-SGMI-2) were generated as described in example 13, which specifically block the function of the human lectin pathway and also indicated blocking the lectin pathway in mice. In this example, the efficacy of MASP-2 inhibitory antibody OMS646-SGMI-2 in reducing and/or preventing kidney inflammation and tubular interstitial injury in wild-type mice was analyzed in a mouse protein overload proteinuria model.
The method comprises the following steps:
the effect of MASP-2 inhibitory antibodies (10 mg/kg OMS 646-SGMI-2) compared to human IgG4 isotype control antibody ET904 (10 mg/kg) and saline control was evaluated in this study.
Similar to the study described in example 16, the present study utilized protein overload to induce proteinuria kidney disease (Ishola et al European Renal Association 21:591-597, 2006). Proteinuria was induced in unilaterally nephrectomized Balb/c mice by daily i.p. injection of increasing doses (2 g/kg to 15 g/kg) of low endotoxin Bovine Serum Albumin (BSA) for a total of 15 days, as described in example 16.
Antibody treatment was given by i.p. injection starting two weeks 7 days prior to proteinuria induction and continuing the entire study. The dosage regimen was selected based on previous PK/PD and pharmacological studies demonstrating sustained lectin pathway inhibition (data not shown). Mice were sacrificed on day 15, kidneys were harvested and treated for H & E and immunostaining. Stained tissue sections from renal cortex were analyzed by digital image capture followed by quantification using automated image analysis software.
Immunohistochemical staining and apoptosis assessment were performed as described in example 16.
Results:
proteinuria assessment
To confirm the presence of proteinuria in mice, total secreted proteins in urine were measured in urine samples collected over 24 hours on day 15 (end of the experiment). It was determined that urine samples showed an average almost 6-fold increase in total protein levels in the BSA treated group compared to the untreated control group (data not shown), confirming the presence of proteinuria in BSA treated mice. No significant differences in protein levels were observed between BSA treated groups.
Histological change assessment
Fig. 32 shows representative H & E stained tissue sections of mice from the following groups on day 15 post treatment with BSA: (panel a) wild-type control mice treated with saline; (panel B) isotype antibody-treated control mice; and (panel C) wild-type mice treated with MASP-2 inhibitory antibodies.
As shown in figure 32, there was a higher degree of tissue preservation in the MASP-2 inhibitory antibody-treated group (panel C) compared to the wild-type group treated with either saline (panel a) or isotype control (panel B) at the same level of protein overload challenge.
Apoptosis assessment
Apoptosis was assessed in tissue sections by staining with terminal deoxynucleotidyl transferase dUTP notch end marker (TUNEL), and the frequency of TUNEL stained apoptotic cells was counted in 20 High Power Fields (HPF) from continuous selection of cortex. Fig. 33 illustrates the frequency of TUNEL apoptotic cells counted in 20 High Power Fields (HPF) from sequential selection of tissue sections of renal cortex from wild type mice treated with saline control and BSA (n=8), wild type mice treated with isotype control antibody and BSA (n=8), and wild type mice treated with MASP-2 inhibitory antibody and BSA (n=7). As shown in fig. 33, a highly significant reduction in apoptosis rate in the cortex was observed in kidneys obtained from the MASP-2 inhibitory antibody treated group compared to the saline and isotype control treated group (p=0.0002 for saline control and MASP-2 inhibitory antibodies; p=0.0052 for isotype control and MASP-2 inhibitory antibodies).
Assessment of cytokine infiltration Interleukin 6 (IL-6), transforming growth factor beta (TGF beta) and tumor necrosis factor alpha (TNF alpha), which are pro-inflammatory cytokines known to be upregulated in proximal tubules of wild-type mice in the proteinuria model, were evaluated in kidney tissue sections obtained in this study.
Fig. 34 illustrates the results of computer-based image analysis of tissue sections stained with anti-tgfβ antibodies (measured as% tgfβ antibody staining area) in wild-type mice treated with BSA and saline (n=8), wild-type mice treated with BSA and isotype control antibodies (n=7), and wild-type mice treated with BSA and MASP-2 inhibitory antibodies (n=8). As shown in fig. 34, quantification of tgfβ staining area indicated a significant decrease in tgfβ levels in MASP-2 inhibitory antibody treated mice (p-value = 0.0324 and 0.0349, respectively) compared to saline and isotype control antibody treated control group.
Fig. 35 illustrates the results of computer-based image analysis of tissue sections stained with anti-tnfα antibodies (measured as% tnfα antibody staining area) in wild-type mice treated with BSA and saline (n=8), wild-type mice treated with BSA and isotype control antibodies (n=7), and wild-type mice treated with BSA and MASP-2 inhibitory antibodies (n=8). As shown in fig. 35, analysis of the stained sections showed a significant reduction in tnfα levels in the MASP-2 inhibitory antibody treated group compared to the saline control group (p=0.011) and the isotype control group (p=0.0285).
Fig. 36 illustrates the results of computer-based image analysis of tissue sections stained with anti-IL-6 antibody (measured as% IL-6 antibody staining area) in wild-type mice treated with BSA and saline (n=8), BSA and isotype control antibodies (n=7), and BSA and MASP-2 inhibitory antibodies (n=8). As shown in fig. 36, analysis of the stained sections showed a significant decrease in IL-6 levels in the MASP-2 inhibitory antibody treated group compared to the saline control group (p=0.0269) and the isotype control group (p=0.0445).
Results and conclusions overall:
the results of this example demonstrate that the use of MASP-2 inhibitory antibodies provides protection against kidney injury in a protein overload model, consistent with the results described in example 16, example 16 demonstrates that MASP-2-/-mice have reduced kidney injury in a proteinuria model.
Example 18
This example provides results from evaluation of the role of the lectin pathway in doxorubicin-induced nephropathy using doxorubicin-induced nephropathogenic models of renal fibrosis, inflammation and tubular interstitial injury in MASP-2-/-and wild-type mice.
Background/rationale:
Doxorubicin is an anthracycline antitumor antibiotic used to treat a variety of cancers, including hematological malignancies, soft tissue sarcomas, and many types of cancers. Doxorubicin-induced kidney disease is well established in rodent models of chronic kidney disease, which can better understand the progression of chronic proteinuria (Lee and Harris, nephrology,16:30-38, 2011). The type of structural and functional impairment of doxorubicin-induced kidney disease is very similar to that of human chronic proteinuria kidney disease (Pippin et al American Journal of Renal Physiology 296: F213-29, 2009).
Doxorubicin-induced kidney disease is characterized by podocyte injury followed by glomerulosclerosis, tubular interstitial inflammation and fibrosis. Doxorubicin-induced nephropathy has been shown in many studies to be mediated by mechanisms of both immune and non-immune origin (Lee and Harris, nephrology,16:30-38, 2011). Doxorubicin-induced nephropathy has several advantages as a model of kidney disease. First, it is a highly reproducible and predictable model of kidney injury. This is because it is characterized by induction of kidney injury within days of drug administration, which allows for ease of experimental design as the time of injury is consistent. It is also a model in which the extent of tissue damage is severe, but with acceptable mortality (< 5%) and morbidity (weight loss). Thus, due to the severity and time of renal injury to doxorubicin-induced nephropathy, it is a model suitable for testing interventions that protect against renal injury.
As described in examples 16 and 17, MASP-2-/-mice and mice treated with MASP-2 inhibitory antibodies were determined to show significantly better results (e.g., less tubulointerstitial damage and less nephritis) than wild-type mice in a protein overload model of proteinuria, suggesting a pathogenic role for the lectin pathway in proteinuria kidney disease.
MASP-2-/-mice were analyzed in the doxorubicin-induced nephropathy model (AN) in comparison to wild-type mice in this example to determine if MASP-2 deficiency reduced and/or prevented doxorubicin-induced nephritis and tubular interstitial injury.
The method comprises the following steps:
1. dose and time point optimization
Initial experiments were performed to determine the dose of doxorubicin and the time point at which BALB/c mice developed nephritis at levels appropriate for testing therapeutic intervention.
Three groups of wild-type BALB/c mice (n=8) were injected with IV-administered single dose of doxorubicin (10.5 mg/kg). Mice were eliminated at three time points: one week, two weeks and four weeks after doxorubicin administration. Control mice were injected with saline alone.
Results: all mice in the three groups showed signs of glomerulosclerosis and proteinuria, as indicated by H&Determined by E-stainingAnd a gradual increase in tissue inflammation, as measured by macrophage infiltration in the kidney (data not shown). The extent of tissue damage was mild in the one week group, moderate in the two week group, and severe in the four week group (data not shown). Two week time points were selected for the rest of the study.
2. Analysis of Adriamycin-induced nephropathy in wild-type and MASP-2-/-mice
To elucidate the role of the lectin pathway of complement in doxorubicin-induced nephropathy, a panel of MASP-2-/-mice (BALB/c) was compared to wild-type mice (BALB/c) at the same dose of doxorubicin. MASP-2-/-mice were backcrossed to BALB/c mice for 10 passages.
Wild type (n=8) and MASP-2-/- (n=8) were IV injected with doxorubicin (10.5 mg/kg), and three mice of each strain were given saline alone as a control. All mice were eliminated two weeks after treatment and tissues were collected. The extent of histopathological lesions was assessed by H & E staining.
Results:
fig. 37 shows representative H & E stained tissue sections from the following groups of mice on day 14 after treatment with doxorubicin or saline only (control): (panels A-1, A-2, A-3) wild-type control mice treated with saline alone; (panels B-1, B-2, B-3) wild-type mice treated with doxorubicin; and (panels C-1, C-2, C-3) MASP-2-/-mice treated with doxorubicin. Each figure (e.g., figures A-1, A-2, A-3) represents a different mouse.
As shown in FIG. 37, there was a higher degree of tissue preservation in the MASP-2-/-group treated with doxorubicin than in the wild-type group treated with the same dose of doxorubicin.
FIG. 38 illustrates the results of computer-based image analysis of kidney tissue sections stained with macrophage specific antibody F4/80, showing the average area of macrophage staining (%) from the following groups of mice on day 14 after treatment with doxorubicin or saline only (wild type control): wild-type control mice treated with saline alone; wild-type mice treated with doxorubicin; MASP-2-/-mice treated with saline only, and MASP-2-/-mice treated with doxorubicin. As shown in fig. 38, MASP-2-/-mice treated with doxorubicin had reduced macrophage infiltration compared to wild-type mice treated with doxorubicin (p=0.007).
Fig. 39 illustrates the results of computer-based image analysis of kidney tissue sections stained with sirius red, showing collagen deposition staining areas (%) from mice of the following groups on day 14 after treatment with doxorubicin or saline only (wild type control): wild-type control mice treated with saline alone; wild-type mice treated with doxorubicin; MASP-2-/-mice treated with saline only, and MASP-2-/-mice treated with doxorubicin. As shown in fig. 39, MASP-2-/-mice treated with doxorubicin had reduced collagen deposition compared to wild-type mice treated with doxorubicin (p=0.005).
Sum-of-conclusions:
improvement of tubular interstitial inflammation is a key goal for the treatment of renal disease. The results presented herein indicate that the complement-activated lectin pathway significantly promotes the occurrence of tubular interstitial inflammation. As further demonstrated herein, inhibitors of MASP-2, such as MASP-2 inhibitory antibodies, are useful as novel therapies for treating proteinuria nephropathy, doxorubicin nephropathy, and ameliorating tubular interstitial inflammation.
Example 19
This example describes the initial results of an ongoing phase 2 clinical trial evaluating the safety and clinical efficacy of fully human monoclonal MASP-2 inhibitory antibodies in adults with steroid-dependent immunoglobulin a nephropathy (IgAN) and in adults with steroid-dependent Membranous Nephropathy (MN).
Background:
chronic kidney disease affects more than 2 million people in the united states (dragz p. Et al, ann international Med 162 (11); ITC1-16, 2015). Glomerulonephropathy (GN), including IgAN and MN, is a kidney disease in which glomerulus injury and frequently results in end stage renal disease and dialysis. There are several types of primary GNs, most commonly igans. Many of these patients have persistent kidney inflammation and progressive damage. Typically, these patients are treated with corticosteroids or immunosuppressants, which have many serious long-term adverse consequences. Many patients continue to deteriorate, even at these treatments. No treatment was approved for treatment of IgAN or MN.
IgA nephropathy
Immunoglobulin a kidney disease (IgAN) is an autoimmune kidney disease, resulting in intrarenal inflammation and kidney injury. IgAN is the most common form of primary glomerulonephritis worldwide (Magistroni et al, kidney int.88 (5): 974-89, 2015). It is estimated that IgAN will occur in 1/1400 of the United states according to an annual incidence of about 2.5/100,000. In 20 years after diagnosis, up to 40% of IgAN patients will develop advanced renal disease (ESRD) (Coppo R., D' Amico G., J Nephrol 18 (5): 503-12,2005; xie et al, PLoS One,7 (6): e38904 (2012)). Patients often present with microscopic hematuria and mild to moderate proteinuria and varying levels of renal insufficiency (Wyatt r.j. Et al, N Engl J Med 368 (25): 2402-14, 2013). Clinical signs such as impaired renal function, sustained hypertension and heavy proteinuria (over 1 g/day) are associated with poor prognosis (Goto M et al Nephrol Dial Transplant (10): 3068-74, 2009; berthoux F. Et al, J Am Soc Nephrol22 (4): 752-61, 2011). Proteinuria was the strongest prognostic factor in a number of large-scale observational studies and prospective trials, independent of other risk factors (Coppo R. Et al, JNEPHROL 18 (5): 503-12,2005; reich H.N. et al, JAm Soc Nephrol 18 (12): 3177-83, 2007). If untreated, it is estimated that 15-20% of patients reach ESRD (D' Amico G., am JKidneyDis 36 (2): 227-37, 2000) within 10 years of disease occurrence.
Diagnostic markers for IgAN are significant IgA deposition in glomerular vasculature membranes, alone or with IgG, igM, or both. In IgAN, kidney biopsies reveal glomerular deposition of mannan-binding lectin (MBL), a key recognition molecule for MASP-2 activation, which is an effector enzyme of the lectin pathway of the complement system. Glomerular MBL deposition, often co-localized with IgA and indicated complement activation, and high levels of urinary MBL associated with adverse prognosis of IgAN, where these patients demonstrated more severe histological changes and mesangial proliferation than patients without MBL deposition or high levels of urinary MBL (Matsuda M. Et al, nephron 80 (4): 408-13, 1998; liu LL et al, clin Exp Immunol 169 (2): 148-155, 2012; roos A. Et al, J Am Soc Nephrol 17 (6): 1724-34, 2006; liu LL et al, clin Exp Immunol174 (1): 152-60, 2013). The remission rate was also significantly lower for patients with MBL deposition (Liu LL et al, clin Exp Immunol174 (1): 152-60, 2013).
Current methods of treating IgAN attempt to slow, stop or delay the deterioration of kidney function. General results of renal disease improvement for glomerulonephritis (Kidney Disease Improving Global Outcomes, kdaigo) clinical practice guidelines recommend IgAN treatment programs that primarily emphasize controlling blood pressure by renin-angiotensin system (RAS) blockade [ kdaigo Work Group 2012]. For patients with 1g or more of persistent daily proteinuria, the recommended treatment includes corticosteroids and/or other immunosuppressants, such as cyclophosphamide, azathioprine or mycophenolate mofetil, despite the maximally tolerated dose of antihypertensive drug and well-controlled blood pressure. The guidelines for the overall outcome of renal disease improvement for glomerulonephritis (KDIGO) (int. Soc of neprol 2 (2): 139-274, 2012) suggest that corticosteroids should be administered to patients with proteinuria of greater than or equal to 1 g/day, typically for a treatment duration of 6 months. For patients with crescent IgAN (defined as >50% of the glomeruli having crescent) and rapid deterioration of renal clearance, another immunosuppressant (e.g. cyclophosphamide) may be added to the corticosteroid. However, even with invasive immunosuppressive therapy, which is associated with severe long-term sequelae, some patients still have progressive impairment of renal function. There is no FDA approved IgAN therapy and even the use of Angiotensin Converting Enzyme (ACE) inhibitors or Angiotensin Receptor Blockers (ARBs) to control blood pressure, proteinuria continues to increase in some patients. None of these treatments suggests stopping or even slowing the disease progression in patients at risk of rapid progression of IgAN. Alternative treatments that can reduce or eliminate the need for long-term corticosteroids and/or immunosuppressive therapies would clearly address the unmet medical need.
Membranous nephropathy
The annual incidence of Membranous Nephropathy (MN) is about 10-12/1,000,000. Patients with MN may have varying clinical course, but about 25% will develop advanced renal disease.
Membranous nephropathy is one of the most common causes of immune-mediated glomerular disease and nephrotic syndrome in adults. The disease is characterized by the formation of an immune deposit, mainly IgG4, on the outside of glomerular basement membrane, which contains podocyte antigens and antibodies specific for these antigens, resulting in complement activation. The initial manifestations of MN are related to nephrotic syndrome, proteinuria, hypoalbuminemia, hyperlipidemia and edema.
Although MN can spontaneously alleviate without treatment, up to 1/3 of patients demonstrated progressive loss of renal function and progress to ESRD with a median of 5 years after diagnosis. In general, corticosteroids are used for the treatment of MN and there is a need to develop alternative therapies. In addition, patients at moderate risk of progression are determined to be treated with prednisone-binding cyclophosphamide or a troponin (calpain) inhibitor based on the severity of proteinuria, and both treatments are often associated with severe systemic adverse effects.
The method comprises the following steps:
two phase 1 clinical trials in healthy volunteers have shown that intravenous and subcutaneous administration of the MASP-2 inhibitory antibody OMS646 resulted in sustained lectin pathway inhibition.
This example describes the provisional results from an ongoing phase 2 non-control multicenter study of MASP-2 inhibitory antibody OMS646 in subjects with IgAN and MN. Inclusion criteria required that all patients in the study, regardless of the renal disease subtype, had maintained a stable dose of corticosteroid for at least 12 weeks prior to study recruitment (i.e., the patients were steroid dependent). The study was a single trial study with 12 weeks of treatment and 6 weeks of follow-up period.
Approximately 4 subjects were enrolled per disease plan. Studies were designed to evaluate whether OMS646 could improve kidney function (e.g., improve proteinuria) and reduce corticosteroid requirements in subjects with IgAN and MN. To date, 2 patients with IgA nephropathy and 2 patients with membranous nephropathy have completed treatment in research.
At study entry, each subject must have high levels of protein in the urine, although being treated with a stable corticosteroid dose. These criteria select patients who were unlikely to spontaneously improve during the study.
Subjects were 18 or more in age at screening and included in the study only when they diagnosed one of the following: the kidney biopsy is diagnosed with IgAN or the kidney biopsy is diagnosed with primary MN. The enrolled patient must also meet all of the following inclusion criteria:
(1) Three samples collected consecutively and daily from each prior to 2 visits during the screening, with an average urinary albumin/creatinine ratio >0.6;
(2) Prednisone or equivalent doses of ≡10mg have been administered for at least 12 weeks prior to screening visit 1;
(3) If immunosuppressive therapy (e.g., cyclophosphamide, mycophenolate mofetil) is administered, a stable dose has been administered for at least 2 months prior to screening visit 1, no dose change is expected for the duration of the study;
(4) Has the concentration of not less than 30mL/min/1.73m 2 Is calculated by MDRD equation 1;
(5) A physician-directed stable optimized treatment with an Angiotensin Converting Enzyme Inhibitor (ACEI) and/or an Angiotensin Receptor Blocker (ARB), and a systolic blood pressure of <150mmHg and a diastolic blood pressure of <90mmHg at rest;
(6) Belimumab, eculizumab or rituzimab was not used within 6 months of screening visit 1; and
(7) There is no history of kidney transplantation.
1 MDRD formula: eGFR (mL/min/1.73 m) 2 )=175x(SCr) -1.154 x(Age) -0.203 x (0.742 if female) x (1.212 if african americans). Note that: SCr = serum creatinine measurement should be mg/dL.
The monoclonal antibody OMS646 used in this study was a fully human IgG4 monoclonal antibody that bound and inhibited human MASP-2. MASP-2 is an effector enzyme of the lectin pathway. As demonstrated in example 12, OMS646 tightly bound recombinant MASP-2 (apparent equilibrium dissociation constant in the 100pM range) and showed greater than 5,000-fold selectivity over homologous proteins C1s, C1r and MASP-1. OMS646 was shown to be nanomolar in potency in the functional assay (resulting in 50% inhibition [IC 50 ]About 3 nM) inhibits the human lectin pathway but has no significant effect on the classical pathway. Administration of OMS646 by Intravenous (IV) or Subcutaneous (SC) injection into mice, non-human primates and humans resulted in high plasma concentrations, which are associated with inhibition of lectin pathway activation in ex vivo assays.
In this study, OMS646 drug was provided at a concentration of 100mg/mL, which was further diluted for IV administration. An appropriate calculated volume of OMS646100mg/mL injection solution was withdrawn from the vial using a syringe for dose preparation. Infusion bags were administered within 4 hours of preparation.
The study consisted of screening (28 days), treatment (12 weeks) and follow-up (6 weeks) periods, as shown in the study design flow chart below.
The flow chart of the study design is shown in figure 44.
During the screening period and prior to the first OMS646 dose, the consented subjects provided three urine samples (collected once daily) in each of two consecutive 3 days to establish a baseline value for urine albumin to creatinine ratio. After the screening period, eligible subjects received OMS6464mg/kg IV weekly for 12 weeks (treatment period). There was a 6 week follow-up period following the last dose of OMS 646.
During the first 4 weeks of treatment with OMS646, subjects maintained their stable pre-study doses of corticosteroid. At the end of the initial 4 weeks of the 12-week treatment period, the subject underwent a corticosteroid taper (i.e., a corticosteroid dose reduction) over 4 weeks, if tolerated, followed by a 4-week hold of the resulting corticosteroid dose. The goal is to decrease to less than or equal to 6mg of prednisone (or equivalent dose) per day. At this stage, decrementing was stopped in subjects with worsening renal function as determined by the investigator. Subjects were treated with OMS646 during the corticosteroid decrease and throughout the 12 week treatment period. The patient then passed another 6 weeks after their last treatment. The decrementing of corticosteroids and OMS646 treatment allows for evaluation of whether OMS646 allows for a reduction in the corticosteroid dosage required to maintain stable renal function.
The key efficacy measures in this study were the change from baseline to 12 weeks, albumin to creatinine ratio (uACR) and 24-hour protein levels in urine. Measurement of urine proteins or albumin is routinely used to assess kidney involvement, and sustained high levels of urine proteins are associated with renal disease progression. uACR is used clinically to evaluate proteinuria.
Efficacy analysis
The analytical value of uACR is defined as the average of all values obtained for the time points. The planned value of uACR is 3 at each predetermined point in time. The baseline value of uACR is defined as the average of the analysis values of the two screening visits.
Results:
FIG. 40 illustrates uACR in two IgAN patients during the course of a 12 week study with 4mg/kg MASP-2 inhibitory antibody (OMS 646) weekly treatment. As shown in fig. 40, at time point "a" (p=0.003) by non-transformation analysis; time point "b" (p=0.007) and time point "c" (p=0.033), the change from baseline was statistically significant. Table 12 provides 24-hour urine protein data for two IgAN patients treated with OMS 646.
Table 12: 24-hour urine protein (mg/day) in OMS646 treated IgAN patients
As shown in fig. 40 and table 12, patients with IgAN demonstrated clinically and statistically significant renal function improvement over the course of the study. There was a statistically significant decrease in uACR (see fig. 40) and 24 hour urinary protein concentration (see table 12). As shown in the uACR data of fig. 40, the average baseline uACR was 1264mg/g and reached 525mg/g at the end of the treatment (p=0.011), decreasing to 128mg/g at the end of the follow-up period. As further shown in fig. 40, the therapeutic effect was maintained throughout the follow-up period. Measurement of 24 hours urine protein secretion followed the uACR, decreasing from 3156mg/24 hours on average to 1119mg/24 hours (p=0.017). The therapeutic effect is highly consistent between the two patients. Both patients experienced a decrease of about 2000 mg/day and achieved partial remission (defined as greater than 50% decrease in urine protein secretion over 24 hours and/or less than 1000 mg/day protein secretion obtained; complete remission defined as less than 300 mg/day protein secretion). The magnitude of 24-hour proteinuria reduction in both IgA nephropathy patients was associated with a significant improvement in kidney survival. Two IgA nephropathy patients were also able to significantly decrement their steroids, each reducing the daily dose to 5mg (60 mg to 0mg;30mg to 5 mg).
Two MN patients also demonstrated a decrease in uACR during treatment with OMS 646. One MN patient's uACR was reduced from 1003mg/g to 69mg/g and kept at this low level throughout the follow-up period. The uACR of the other MN patient decreased from 1323mg/g to 673mg/g with varying post-treatment course. The first MN patient showed a significant 24-hour decrease in urine protein levels (10,771 mg/24 hours of baseline to 325mg/24 hours of day 85) with partial and almost complete remission, while the other patient remained essentially unchanged (4272 mg/24 hours of baseline to 4502mg/24 of day 85). The steroid was decreased from 30mg to 15mg and from 10mg to 5mg in both MN patients.
In summary, consistent improvement in renal function was observed in IgAN and MN subjects treated with MASP-2 inhibitory antibody OMS 646. The effect of OMS646 treatment in patients with IgAN was robust and consistent, indicating a strong efficacy signal. These effects are supported by results in MN patients. The time course and amplitude of the uACR changes during treatment were consistent between all four patients with IgAN and MN. No significant security problems were observed. The patients in this study represent a treatment-difficult group and the therapeutic effects of these patients are believed to be predictive of efficacy of MASP-2 inhibitory antibodies, such as OMS646, in IgAN and MN patients, such as patients with steroid-dependent IgAN and MN (i.e., patients undergoing treatment with stable doses of corticosteroid prior to treatment with MASP-2 inhibitory antibodies), including those at risk for rapid progression to advanced renal disease.
In accordance with the foregoing, in one embodiment, the present invention provides a method of treating a human subject having IgAN or MN comprising administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation. In one embodiment, the method comprises administering to a human subject having IgAN or MN an amount of MASP-2 inhibitory antibody sufficient to improve kidney function (e.g., improve proteinuria). In one embodiment, the subject has steroid dependent IgAN. In one embodiment, the subject has a steroid dependent MN. In one embodiment, the MASP-2 inhibitory antibody is administered to a subject having a steroid-dependent IgAN or a steroid-dependent MN in an amount sufficient to improve kidney function and/or reduce corticosteroid dosage in said subject.
In one embodiment, the method further comprises identifying a human subject having steroid dependent IgAN prior to the step of administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation.
In one embodiment, the method further comprises identifying a human subject having a steroid dependent MN prior to the step of administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation.
According to any of the embodiments disclosed herein, the MASP-2 inhibitory antibody exhibits at least one or more of the following characteristics: the antibody binds to human MASP-2 with a KD of 10nM or less, the antibody binds to an epitope in the CCP1 domain of MASP-2, the antibody inhibits C3b deposition with an IC50 of 10nM or less in 1% human serum in an in vitro assay, the antibody inhibits C3b deposition with an IC50 of 30nM or less in 90% human serum, wherein the antibody is an antibody fragment selected from the group consisting of Fv, fab, fab ', F (ab) 2, and F (ab') 2, wherein the antibody is a single chain molecule, wherein the antibody is an IgG2 molecule, wherein the antibody is an IgG1 molecule, wherein the antibody is an IgG4 molecule, wherein the IgG4 molecule comprises an S228P mutation. In one embodiment, the antibody binds MASP-2 and selectively inhibits the lectin pathway and does not significantly inhibit the classical pathway (i.e., inhibits the lectin pathway while leaving the classical complement pathway intact).
In one embodiment, the MASP-2 inhibitory antibody is administered in an amount effective to improve at least one or more clinical parameters associated with renal function, such as proteinuria improvement (e.g., a decrease in uACR and/or a decrease in 24-hour urine protein concentration, such as a greater than 20% decrease in 24-hour urine protein secretion, or such as a greater than 30% decrease in 24-hour urine protein secretion, such as a greater than 40% decrease in 24-hour urine protein secretion, such as a greater than 50% decrease in 24-hour urine protein secretion).
In some embodiments, the methods comprise administering (e.g., intravenously) a MASP-2 inhibitory antibody to a subject having an IgAN (e.g., a steroid-dependent IgAN) via a catheter for a first period of time (e.g., at least one day to one week or two weeks or three weeks or four weeks or more), followed by subcutaneously administering the MASP-2 inhibitory antibody to the subject for a second period of time (e.g., a chronic stage of at least two weeks or more).
In some embodiments, the method comprises administering (e.g., intravenously) a MASP-2 inhibitor to a subject having MN (e.g., steroid-dependent MN) through a catheter for a first period of time (e.g., at least one day to one week or two weeks or three weeks or four weeks or more), followed by subcutaneously administering a MASP-2 inhibitory antibody to the subject for a second period of time (e.g., a chronic stage of at least two weeks or more).
In some embodiments, the method comprises administering intravenously, intramuscularly, or subcutaneously a MASP-2 inhibitory antibody to a subject having IgAN (e.g., steroid-dependent IgAN) or MN (e.g., steroid-dependent MN). The treatment may be chronic, administered daily to monthly, but is preferably administered at least once every two weeks or at least once a week, for example twice a week or three times a week.
In one embodiment, a method comprises treating a subject having IgAN (e.g., steroid dependent IgAN) or MN (e.g., steroid dependent MN) comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence set forth in SEQ ID NO:67 and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence set forth in SEQ ID NO: 70. In some embodiments, the composition comprises a MASP-2 inhibitory antibody comprising (a) a heavy chain variable region comprising: i) A heavy chain CDR-H1 comprising the amino acid sequence of 31-35 of SEQ ID NO. 67; and ii) a heavy chain CDR-H2 comprising the amino acid sequence of 50-65 of SEQ ID NO. 67; and iii) a heavy chain CDR-H3 comprising the amino acid sequence of 95-107 of SEQ ID NO:67, and b) a light chain variable region comprising: i) A light chain CDR-L1 comprising the amino acid sequence of 24-34 of SEQ ID NO. 70; and ii) light chain CDR-L2 comprising the amino acid sequence of 50-56 of SEQ ID NO. 70; and iii) light chain CDR-L3 comprising the amino acid sequence of 89-97 of SEQ ID NO. 70, or (II) variants thereof, comprising a heavy chain variable region having at least 90% identity (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity) to SEQ ID NO. 67 and a light chain variable region having at least 90% identity (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity) to SEQ ID NO. 70.
In some embodiments, the method comprises administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO. 67 and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO. 70.
In some embodiments, the method comprises administering to the subject a composition comprising a MASP-2 inhibitory antibody or antigen binding fragment thereof that specifically recognizes at least a portion of an epitope on human MASP-2 that is recognized by reference antibody OMS646 that comprises the heavy chain variable region set forth in SEQ ID NO 67 and the light chain variable region set forth in SEQ ID NO 70.
In some embodiments, the method comprises administering to a subject having or at risk of developing IgAN (e.g., steroid dependent IgAN) or MN (e.g., steroid dependent MN) a composition comprising a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence shown in SEQ ID NO:67 and a light chain variable region comprising the amino acid sequence shown in SEQ ID NO:70 at a dose of 1mg/kg to 10mg/kg (i.e., 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, or 10 mg/kg) for a period of at least 3 weeks, or at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks, or at least 12 weeks.
Example 20
This example describes the initial results of an ongoing phase 2 clinical trial to evaluate the safety and clinical efficacy of fully human monoclonal MASP-2 inhibitory antibodies in adults with steroid dependent Lupus Nephritis (LN).
Background:
chronic kidney disease affects more than 2 million people in the united states (dragz p. Et al, ann international Med 162 (11); ITC1-16, 2015). Glomerulonephropathy (GN), including IgAN, MN, and LN, is a kidney disease in which glomerular injury and frequently results in end stage renal disease and dialysis. Many of these patients have persistent kidney inflammation and progressive damage. Typically, these patients are treated with corticosteroids or immunosuppressants, which have many serious long-term adverse consequences. Many patients continue to deteriorate, even at these treatments.
Lupus nephritis
The major complication of Systemic Lupus Erythematosus (SLE) is nephritis, also known as lupus nephritis, which is classified as a secondary form of glomerulonephritis. Up to 60% of adults with SLE have some form of kidney accumulation late in the course of disease (Koda-Kimble et al, koda-Kimble and Young's Applied Therapeutics: the clinical use of drugs, 10) th Ed,Lippincott Williams&Wilkins: pages 792-9,2012), the prevalence in the United states is 20-70 per 10 thousands of people. Lupus nephritis is commonly present in patients with active SLE with other symptoms including fatigue, fever, rash, arthritis, serositis, or central nervous system disorders (Pisetsky D.S et al, med Clin North Am (1): 113-28, 1997). Some patients have asymptomatic lupus nephritis; however, during periodic follow-up, laboratory abnormalities such as elevated serum creatinine levels, low albumin levels, or urinary proteins or sediment suggest active lupus nephritis. Autoimmunity plays a major role in the pathogenesis of lupus nephritis. These autoantibodies form pathogenic immune complexes within the blood vessels, which deposit in the glomeruli. Self-resistance The body may also bind to antigens already located in the glomerular basement membrane, forming immune complexes in situ. Immune complexes promote inflammatory responses by activating complement and attracting inflammatory cells (D 'Agati V.D et al, lupus nephritis: pathology and pathogenesis: wallace D.J. Hahn, dubois' Lupus Erythematosus, 7) th Ed Philadelpha:Lippincott Williams&Wiklins: p1094-111, 2007). Thus, immune complex mediated complement activation plays a key role in the pathogenesis of lupus nephritis. C4d deposits are present in kidney tissue and are generally associated with immune complex deposits Clq and C3, leading to the classical pathway. In some cases, the presence of C4d deposits in the absence of Clq suggests a possible lectin pathway involvement (Kim M.K., et al, intJClin Exp Pathol 6 (10): 2157-67, 2013).
Further support of the important contribution of the lectin pathway, deposits of MBL appear in skin lesions in SLE patients (Wallim L.R et al, hum Immunol 75 (7): 629-32, 2014). In addition, strong deposition of MBL and fiber gel protein has been observed in most kidney biopsies from patients with lupus nephritis (Nisihara R.M et al, hum Immunol 74 (8): 907-10, 2013). Renal MBL deposition is most pronounced in patients with high proteinuria. In addition, plasma MBL levels in SLE patients were significantly higher than healthy controls and MBL levels correlated with disease activity, suggesting that MBL levels may represent biomarkers of SLE disease activity (Panda A.K et al Arthritis Res Ther (5): R218,2012). Corticosteroids are the primary routine treatment option for patients with mild lupus nephritis. For more severe cases, high doses of prednisone, methylprednisolone, mycophenolate mofetil, cyclophosphamide, azathioprine and cyclosporine have been used in clinical practice. Treatment options for SLE and lupus nephritis have high associated morbidity and mortality. Side effects, particularly the use of long-term corticosteroids, limit patient compliance and subsequently affect therapeutic efficacy. There is a need to develop better tolerogenic treatment regimens.
The method comprises the following steps:
as described in example 19 above, two phase 1 clinical trials in healthy volunteers have shown that intravenous and subcutaneous administration of MASP-2 inhibitory antibody OMS646 resulted in sustained lectin pathway inhibition.
This example describes the provisional results from an ongoing phase 2 non-control multicenter study of MASP-2 inhibitory antibody OMS646 in subjects with Lupus Nephritis (LN). Inclusion criteria required that all patients in the study, regardless of the renal disease subtype, had maintained a stable dose of corticosteroid for at least 12 weeks prior to study recruitment (i.e., the patients were steroid dependent). The study was a single trial study with 12 weeks of treatment and 6 weeks of follow-up period.
Studies were designed to evaluate whether OMS646 could improve kidney function (e.g., improve proteinuria) and reduce corticosteroid requirements in subjects with LN. To date, 5 patients with Lupus Nephritis (LN) have completed treatment in a study.
At study entry, each subject must have high levels of protein in the urine, although being treated with a stable corticosteroid dose. These criteria select patients who were unlikely to spontaneously improve during the study.
Subjects were not less than 18 years of age at the time of screening and included in the study only at the time of diagnosis of lupus nephritis at their kidney biopsy diagnosis. The enrolled patient must also meet all of the following inclusion criteria:
(1) Three samples collected consecutively and daily from each prior to 2 visits during the screening, with an average urinary albumin/creatinine ratio >0.6;
(2) Prednisone or equivalent doses of ≡10mg have been administered for at least 12 weeks prior to screening visit 1;
(3) If immunosuppressive therapy (e.g., cyclophosphamide, mycophenolate mofetil) is administered, a stable dose has been administered for at least 2 months prior to screening visit 1, no dose change is expected for the duration of the study;
(4) Has the concentration of not less than 30mL/min/1.73m 2 Is calculated by MDRD equation 1;
(5) A physician-directed stable optimized treatment with an Angiotensin Converting Enzyme Inhibitor (ACEI) and/or an Angiotensin Receptor Blocker (ARB), and a systolic blood pressure of <150mmHg and a diastolic blood pressure of <90mmHg at rest;
(6) Belimumab, eculizumab or rituzimab was not used within 6 months of screening visit 1; and
(7) There is no history of kidney transplantation.
1 MDRD formula: eGFR (mL/min/1.73 m) 2 )=175x(SCr) -1.154 x(Age) -0.203 x (0.742 if female) x (1.212 if african americans). Note that: SCr = serum creatinine measurement should be mg/dL.
The monoclonal antibody OMS646 used in this study was a fully human IgG4 monoclonal antibody that bound and inhibited human MASP-2. MASP-2 is an effector enzyme of the lectin pathway. As demonstrated in example 12, OMS646 tightly bound recombinant MASP-2 (apparent equilibrium dissociation constant in the 100pM range) and showed greater than 5,000-fold selectivity over homologous proteins C1s, C1r and MASP-1. OMS646 was shown to be nanomolar in potency in the functional assay (resulting in 50% inhibition [ IC 50 ]About 3 nM) inhibits the human lectin pathway but has no significant effect on the classical pathway. Administration of OMS646 by Intravenous (IV) or Subcutaneous (SC) injection into mice, non-human primates and humans resulted in high plasma concentrations, which are associated with inhibition of lectin pathway activation in ex vivo assays.
In this study, OMS646 drug was provided at a concentration of 100mg/mL, which was further diluted for IV administration. An appropriate calculated volume of OMS646100mg/mL injection solution was withdrawn from the vial using a syringe for dose preparation. Infusion bags were administered within 4 hours of preparation.
The study consisted of screening (28 days), treatment (12 weeks) and follow-up (6 weeks) periods, as shown in the study design flow chart below.
The flow chart of the study design is shown in fig. 45.
During the screening period and prior to the first OMS646 dose, the consented subjects provided three urine samples (collected once a day) in each of two consecutive 3 days to establish a baseline value for 24 hours urine protein and urine albumin to creatinine ratio. After the screening period, eligible subjects received OMS6464 mg/kg IV weekly for 12 weeks (treatment period). There was a 6 week follow-up period following the last dose of OMS 646.
During the first 4 weeks of treatment with OMS646, subjects maintained their stable pre-study doses of corticosteroid. At the end of the initial 4 weeks of the 12-week treatment period, the subject underwent a corticosteroid taper (i.e., a corticosteroid dose reduction) over 4 weeks, if tolerated, followed by a 4-week hold of the resulting corticosteroid dose. The goal is to decrease to less than or equal to 6mg of prednisone (or equivalent dose) per day. At this stage, decrementing was stopped in subjects with worsening renal function as determined by the investigator. Subjects were treated with OMS646 during the corticosteroid decrease and throughout the 12 week treatment period. Patients were then followed for another 6 weeks after their last treatment. The decrementing of corticosteroids and OMS646 treatment allows for evaluation of whether OMS646 allows for a reduction in the corticosteroid dosage required to maintain stable renal function.
Efficacy analysis
The key efficacy measure in this study is the change in protein levels from baseline to 12 weeks, 24 hours. Measurement of urine proteins or albumin is routinely used to assess kidney involvement, and sustained high levels of urine proteins are associated with renal disease progression. Partial remission is defined as a greater than 50% reduction in urine protein secretion at 24 hours.
Results:
table 13 provides 24 hour urine protein (mg/day) data for 5 LN patients treated with OMS 646.
Table 13: 24 hour urine protein (mg/day) in OMS646 treated LN patients
Note that: patient #1 experienced systemic disease exacerbation during the study.
As shown in table 13, patients with LN demonstrated clinically and statistically significant renal function improvement over the course of the study. As shown in table 13, 4 of the 5 LN patients showed a significant (69% average) decrease in proteinuria secretion at 24 hours during the treatment period. Patient 5 (patient # 1) experienced systemic disease exacerbation and showed significant increases. Most lupus responders are able to significantly decrement their steroid dose.
In summary, significant improvement in renal function was observed in 4 of the 5 LN patients treated with the MASP-2 inhibitory antibody OMS 646. The effect of OMS646 treatment was strong and consistent in patients with LN, indicating a strong efficacy signal. No significant security problems were observed. The patients in this study represent a treatment-refractory group and the therapeutic effects of these patients are believed to be predictive of efficacy in MASP-2 inhibitory antibodies, such as OMS646, in LN patients, such as patients with steroid-dependent LN (i.e., patients undergoing treatment with a stable corticosteroid dose prior to treatment with MASP-2 inhibitory antibodies), including those at risk for rapid progression to advanced renal disease.
In accordance with the foregoing, in one embodiment, the invention provides a method of treating a human subject having LN, comprising administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation. In one embodiment, the method comprises administering to a human subject having LN an amount of MASP-2 inhibitory antibody sufficient to improve kidney function (e.g., improve proteinuria). In one embodiment, the subject has steroid dependent LN. In one embodiment, the MASP-2 inhibitory antibody is administered to a subject suffering from steroid dependent LN in an amount sufficient to improve kidney function and/or reduce corticosteroid dosage in said subject.
In one embodiment, the method further comprises identifying a human subject having steroid dependent LN prior to the step of administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody effective to inhibit MASP-2-dependent complement activation.
According to any of the embodiments disclosed herein, the MASP-2 inhibitory antibody exhibits at least one or more of the following characteristics: the antibody binds to human MASP-2 with a KD of 10nM or less, the antibody binds to an epitope in the CCP1 domain of MASP-2, the antibody inhibits C3b deposition with an IC50 of 10nM or less in 1% human serum in an in vitro assay, the antibody inhibits C3b deposition with an IC50 of 30nM or less in 90% human serum, wherein the antibody is an antibody fragment selected from the group consisting of Fv, fab, fab ', F (ab) 2, and F (ab') 2, wherein the antibody is a single chain molecule, wherein the antibody is an IgG2 molecule, wherein the antibody is an IgG1 molecule, wherein the antibody is an IgG4 molecule, wherein the IgG4 molecule comprises an S228P mutation. In one embodiment, the antibody binds MASP-2 and selectively inhibits the lectin pathway and does not significantly inhibit the classical pathway (i.e., inhibits the lectin pathway while leaving the classical complement pathway intact).
In one embodiment, the MASP-2 inhibitory antibodies are administered to a subject having LN in an amount effective to improve at least one or more clinical parameters associated with renal function, such as improving proteinuria (e.g., a decrease in uACR and/or a decrease in 24-hour urine protein concentration, such as greater than a 20% decrease in 24-hour urine protein secretion, or such as a greater than a 30% decrease in 24-hour urine protein secretion, such as a greater than a 40% decrease in 24-hour urine protein secretion, such as a greater than a 50% decrease in 24-hour urine protein secretion). In some embodiments, the MASP-2 inhibitory antibody is administered to a subject having LN in an amount effective to cause partial remission of proteinuria (i.e., greater than 50% reduction in 24 hours proteinuria secretion compared to baseline).
In some embodiments, the methods comprise administering (e.g., intravenously) a MASP-2 inhibitory antibody to a subject having LN (e.g., steroid-dependent LN) through a catheter for a first period of time (e.g., at least one day to one or two weeks or three weeks or four weeks or more), followed by subcutaneously administering the MASP-2 inhibitory antibody to the subject for a second period of time (e.g., a chronic stage of at least two weeks or more).
In some embodiments, the method comprises administering intravenously, intramuscularly, or subcutaneously a MASP-2 inhibitory antibody to a subject having LN (e.g., steroid-dependent LN). The treatment may be chronic, administered daily to monthly, but is preferably administered at least once every two weeks or at least once a week, for example twice a week or three times a week.
In one embodiment, a method comprises treating a subject having LN (e.g., steroid dependent LN) comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence shown in SEQ ID NO:67 and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO: 70. In some embodiments, the composition comprises a MASP-2 inhibitory antibody comprising (a) a heavy chain variable region comprising: i) A heavy chain CDR-H1 comprising the amino acid sequence of 31-35 of SEQ ID NO. 67; and ii) a heavy chain CDR-H2 comprising the amino acid sequence of 50-65 of SEQ ID NO. 67; and iii) a heavy chain CDR-H3 comprising the amino acid sequence of 95-107 of SEQ ID NO:67, and b) a light chain variable region comprising: i) A light chain CDR-L1 comprising the amino acid sequence of 24-34 of SEQ ID NO. 70; and ii) light chain CDR-L2 comprising the amino acid sequence of 50-56 of SEQ ID NO. 70; and iii) light chain CDR-L3 comprising the amino acid sequence of 89-97 of SEQ ID NO. 70, or (II) variants thereof, comprising a heavy chain variable region having at least 90% identity (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity) to SEQ ID NO. 67 and a light chain variable region having at least 90% identity (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity) to SEQ ID NO. 70.
In some embodiments, the methods comprise administering to a subject having LN (e.g., steroid dependent LN) a composition comprising an amount of a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO:67 and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 70.
In some embodiments, the methods comprise administering to a subject having LN (e.g., steroid dependent LN) a composition comprising a MASP-2 inhibitory antibody or antigen binding fragment thereof that specifically recognizes at least a portion of an epitope on human MASP-2 that is recognized by reference antibody OMS646 that comprises the heavy chain variable region shown in SEQ ID NO. 67 and the light chain variable region shown in SEQ ID NO. 70.
In some embodiments, the method comprises administering to a subject having or at risk of developing LN (e.g., steroid dependent LN) a composition comprising a MASP-2 inhibitory antibody, or antigen binding fragment thereof, comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO:67 and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO:70, at a dose of 1mg/kg to 10mg/kg (i.e., 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, or 10 mg/kg), at least once a week (e.g., at least twice a week or at least three times a week), for a period of at least 3 weeks, or at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks, or at least 12 weeks.
Example 21
This example describes additional results obtained in an ongoing phase 2 clinical trial to assess the safety and clinical efficacy of the fully human monoclonal MASP-2 inhibitory antibody OMS646 for reducing proteinuria in adult patients with glomerulonephropathy, including IgAN as described in example 19.
The method comprises the following steps:
phase 2 trials included IgAN patients receiving corticosteroids at study entry, all patients received OMS646 in an open label format, and positive results were obtained from both IgAN patients, as described in example 19. For a total of 4 IgAN patients who have completed the trial, administration to the other two IgAN patients using the method described in example 19 has now been completed.
For inclusion in this trial, igAN patients must demonstrate: (1) IgAN, (2) uACR for biopsy diagnosis>0.6g/g,(3)eGFR≥30mL/min/1.73m 2 (4) treatment of controlled blood pressure with stabilized ACEI/ARB, and (5) stable steroid doses of prednisone of ≡10mg for at least 12 weeks.
All 4 IgAN adult patients treated with OMS646 in this trial had pre-existing kidney lesions with an estimated glomerular filtration rate (egffr) of 30 to 46mL/mg/1.73m on entry 2 And 24-hour protein measurement is 2.44 to 4.87 g/24 hours. All patients had Stable renin-angiotensin system (RAS) blockade and at least 3 months of corticosteroid treatment are received at study entry.
All patients received OMS646 IV once a week for 12 weeks. The patient underwent an infusion period of 4 weeks, followed by OMS646 treatment, which included a stable steroid dose for 4 weeks, a steroid decrease if tolerated for 4 weeks, and a steroid dose decrease for 4 weeks. Following OMS646 treatment, the patients were followed up for an additional 6 weeks in the trial. After the trial was completed, the investigator followed the patient.
In the experiments, efficacy measurements were (1) measured 6 times before treatment (baseline) and 3 times at each efficacy assessment during treatment and follow-up for urinary albumin/creatinine ratio (uACR); (2) Urine protein was measured 24 hours once prior to OMS646 treatment and once 2-4 weeks after OMS646 treatment was completed. According to the regimen, the corticosteroid is decremented between week 4 and week 8, if clinically appropriate.
Results:
the 4 IgAN patients completed a 6 week follow-up period after treatment. Table 14 provides demographic and baseline characteristics for these patients.
Table 14: demographic and baseline characteristics
The evfr-estimated glomerular filtration rate; SSA-standard surface area (1.73 m 2 ) The method comprises the steps of carrying out a first treatment on the surface of the uACR-urinary albumin/creatinine ratio.
All patients showed a significant decrease in proteinuria during OMS646 treatment. As shown in fig. 41 and 42, both statistically and clinically significant improvements were observed in both the uACR and 24 hour protein measurements.
FIG. 41 illustrates uACR (mg/g) over time for 4 IgAN patients treated with OMS646 from baseline to 120 days. As shown in fig. 41, the average uACR decreased by 1.13g/g±0.27 (77% decrease, p=0.026) from baseline to the end of the study. As further shown in fig. 41, the uACR was reduced by 94%, 86%, 47% and 89% for each patient (patient 1-4, respectively) relative to baseline at the last follow-up following OMS646 treatment.
Fig. 42 illustrates the change in urine protein from baseline on day 1 prior to treatment to 24 hours post treatment for 4 IgAN patients treated with OMS 646. As shown in FIG. 42, 24-hour urine protein was reduced by 54%, 81%, 63% and 95% (patient 1-4, respectively) from baseline.
Fig. 43 illustrates the mean change in urine protein from baseline to 24 hours post-treatment for 4 IgAN patients treated with OMS 646. As shown in FIG. 43, the average 24-hour urine protein was reduced by 2.87.+ -. 1.08 g/24-hour (73% reduction; p=0.013).
All patients were able to discontinue corticosteroid during or shortly after the study period, demonstrating that the effect of OMS646 on proteinuria is unlikely to be associated with corticosteroids. The estimated glomerular filtration rate (gfr) (calculated by dietary changes in renal disease formulation) was stable throughout the treatment and follow-up period.
All patients were well tolerated OMS646.
In summary, in this open label phase 2 clinical study, a significant and sustained decrease in uACR was observed in all IgAN patients treated with OMS646 for 12 weeks. The 24-hour proteinuria was significantly reduced in all patients. The observed degree of proteinuria reduction correlated with significant improvement in renal prognosis and clinical outcome (Inker L.A et al, am JKidney Dis 68 (3): 392-401 (2016)). Substantial reduction of proteinuria following OMS646 (a monoclonal antibody directed against MASP-2 that abrogates the effects of the lectin pathway of complement) treatment, allowed the discovery of steroid withdrawal, supporting the use of OMS646 as a therapeutic agent for improving IgA glomerular disease outcome. The role of OMS646 in IgAN patients was strong and consistent, demonstrating efficacy in this population.
Example 22
Maintenance of remission after completion of OMS646 treatment in IgA nephropathy (IgAN) patients
Background/principle
In phase 2 studies of IgAN patients, 4 IgAN patients were treated with OMS646, OMS646 being a fully human monoclonal antibody that inhibited MASP-2 activity, as described in examples 19 and 21. All patients received OMS646 IV weekly for 12 weeks as described in examples 19 and 21. As described in example 21, after OMS646 treatment, patients were followed up for another 6 weeks in the trial, and all IgAN patients treated with OMS646 achieved partial remission (defined as more than 50% reduction in urine protein secretion at 24 hours and/or less than 1000 mg/day protein secretion obtained). These four patients were followed after the trial and the duration of remission after OMS646 treatment was assessed as described in this example.
The method comprises the following steps:
after completion of the phase 2 clinical trial described in example 21, 4 IgAN patients treated with OMS646 were followed by the investigator. In the experiments, the endpoints were uACR and 24 hours proteinuria. As described in example 21, all 4 IgAN patients achieved partial remission at the end of the trial. In the post-trial follow-up, urine protein to creatinine ratio (uPCR) was measured. Each uPCR value was converted to uACR (urinary albumin/creatinine ratio) by multiplication by 0.64 (see Zhao et al, clin J Am Soc Nephrol 11:947-55,2016).
Results
All patients achieved partial remission after OMS646 treatment. The average age of 3 females and 1 male was 42 years, 3 caucasians and 1 asian. Average eGFR of 41mL/min/1.73m 2 And the average incoming steroid dose was 55mg. The follow-up range was 2 to 10 months after the last OMS646 dose. As described in example 21, the average uACR was reduced by 77% (p=0.026) during the test. 3 patients remained partially relieved during the available follow-up (54%, 93% and 78% decrease in uacr at 12, 12 and 5 months, respectively). 1 patient had 88% baseline uACR at 7 months. During follow-up, 3 patients also showed an eGFR improvement of 7, 13 and 7ml/min/1.73m 2 . The 4 th patient had stable eGFR. All patients were refractory to steroids. OMS646 is well tolerated.
In summary, proteinuria in IgAN patients was significantly reduced during 12 weeks of treatment with OMS646 and 6 weeks after treatment included in the trial, as described in example 21. The decrease in proteinuria was maintained for up to 10 months after treatment was completed. These data support the use of OMS646 as a therapeutic agent for improving IgA glomerular disease outcome.
In the update from the investigator (status of 4 patients described in this example in a follow-up of approximately one year after a single 12 week course of treatment with OMS 646), 3 of 4 patients were reported to have a retained proteinuria reduction. Of these 3 patients, uACR remained reduced by 14%, 23% and 24% of baseline values for patients prior to OMS646 treatment. Furthermore, after the trial, an improvement in estimated glomerular filtration rate (evfr), a measure of renal function, was observed in 3 out of 4 patients. Patients with the most severe decrease in renal function showed eGFR from 30mL/min/1.73m 2 Improving the concentration to 47mL/min/1.73m 2 The improvement is 57 percent.
In summary, the sustained reduction in proteinuria continues to be impressive throughout the one year follow-up after a single OMS646 session is completed. The improvement observed in the egffr is unexpected, especially in one year follow-up, as it is expected that a significant longer time is required to develop. As described above, two of the 4 patients exhibited a slight increase in egfpr, with one of the patients exhibiting a 50% improved exciting response. The improvement observed in the evfr suggests that OMS646 may provide further benefit to the patient by potentially eliminating or significantly extending the time to dialysis need and reducing the risk of complications associated with chronic renal disease progression.
In accordance with the foregoing, in one embodiment, the present invention provides a method of reducing proteinuria in a human subject suffering from IgAN, comprising administering to the subject a MASP-2 inhibitory antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence of SEQ ID NO 67 according to the following dosage regimen; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70:
c. intravenous administration of about 4mg/kg (i.e., 3.6mg/kg to 4.4 mg/kg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks; or (b)
d. Intravenous administration of about 180mg to about 725mg (i.e., 162mg to 797 mg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks,
wherein the method reduces proteinuria in the human subject.
In one embodiment, the dosage of MASP-2 inhibitory antibody is about 4mg/kg (i.e., 3.6mg/kg to 4.4 mg/kg), such as about 3.6mg/kg, about 3.7mg/kg, about 3.8mg/kg, about 3.9m/kg, about 4.0mg/kg, about 4.1mg/kg, about 4.2mg/kg, about 4.3mg/kg, or about 4.4mg/kg.
In one embodiment, the dose of MASP-2 inhibitory antibody is a fixed dose of about 180mg to about 725mg (i.e., 160mg to 800mg, or about 300mg to 500mg, e.g., about 300mg to about 300 mg), for example, about 160mg, about 165mg, about 170mg, about 175mg, about 180mg, about 185mg, about 190mg, about 195mg, about 200mg, about 205mg, about 210mg, about 215mg, about 220mg, about 225mg, about 230mg, about 240mg, about 245mg, about 250mg, about 255mg, about 260mg, about 265mg, about 270mg, about 275mg, about 280mg, about 285mg, about 290mg, about 295mg, about 300mg, about 305mg, about 310mg, about 315mg, about 320mg, about 325mg, about 330mg, about 335mg, about 340mg, about 345mg, about 350mg, about 355mg, about 360mg, about 365mg, about 370mg, about 375mg, about 380mg, about 385mg, about 390mg, about 395mg, about 400mg, about 405mg, about 410mg, about 415mg, about 420mg, about 425mg, about 430mg, about 440mg, about 445mg, about 460mg, about 470mg, about 465mg, about 475 mg. About 485mg, about 490mg, about 495mg, about 500mg, about 505mg, about 510mg, about 515mg, about 520mg, about 525mg, about 530mg, about 535mg, about 540mg, about 545mg, about 550mg, about 555mg, about 560mg, about 565mg, about 570mg, about 575mg, about 580mg, about 585mg, about 590mg, about 595mg, about 600mg, about 605mg, about 610mg, about 615mg, about 620mg, about 625mg, about 630mg, about 635mg, about 640mg, about 645mg, about 650mg, about 655mg, about 660mg, about 665mg, about 670mg, about 675mg, about 680mg, about 685mg, about 690mg, about 695mg, about 700mg, about 705mg, about 710mg, about 715mg, about 720mg, about 725mg, about 730mg, about 735mg, about 740mg, about 745mg, about 750mg, about 755mg, about 760mg, about 765mg, about 775mg, about 780mg, about 790mg, or about 770 mg.
In one embodiment, the treatment period is 12 weeks.
In one embodiment, the treatment period is followed by a rest period of at least 2 months (i.e., no MASP-2 inhibitor is administered), or a rest period of at least 3 months, or a rest period of at least 4 months, or a rest period of at least 5 months, or a rest period of at least 6 months or longer, such as a rest period of at least 7 months, or a rest period of at least 8 months, or a rest period of at least 9 months, or a rest period of at least 10 months, or a rest period of at least 11 months, or a rest period of at least 12 months or longer.
In some embodiments, the method further comprises periodically monitoring the urine protein level in the subject during the treatment period and/or the rest period, and optionally resuming treatment with a MASP-2 inhibitory antibody when proteinuria is found to recur.
In some embodiments, the method is effective to reduce proteinuria in a subject with IgAN from baseline (pre-treatment) by at least 30%, such as at least 40%, or at least 50%, or greater than 50%, as measured at the end of the treatment period and/or at the end of the rest period.
In some embodiments, the method is effective to increase estimated glomerular filtration rate (eGFR) in a subject having IgAN.
In some embodiments, the subject with IgAN has greater than 1 gram of protein per 24 hours of proteinuria of urine protein secretion prior to treatment, and the method is effective to reduce proteinuria in the subject by at least 30%, e.g., at least 40%, or at least 50%, or greater than 50% from baseline (prior to treatment), as determined at the end of the treatment period and/or at the end of the rest period, and/or to reduce proteinuria to less than 1 gram of protein per 24 hours of urine protein secretion, as determined at the end of the treatment period and/or at the end of the rest period.
In some embodiments, the subject with IgAN has more than 1 gram of protein per 24 hours of proteinuria secreted by the urine protein despite the use of the maximum tolerizing dose of antihypertensive drug and having a well-controlled blood pressure prior to treatment, and the method is effective to reduce proteinuria in the subject from baseline (pre-treatment) by at least 30%, such as at least 40%, or at least 50%, or more than 50%, and/or to less than 1 gram of protein per 24 hours of urine protein secretion as measured at the end of the treatment period and/or at the end of the rest period.
In some embodiments, the subject with IgAN is not treated with a steroid for at least one year. In some embodiments, the subject with IgAN is treated with a steroid for at least a portion of the 12 weeks of treatment with OMS 646. In some embodiments, the subject with IgAN is treated for at least a portion of the 12 weeks of treatment with OMS646 for a steroid treatment, and the method is effective to reduce proteinuria and reduce or eliminate the need for steroid treatment by the end of the treatment period and/or the end of the rest period.
Other embodiments
All publications, patent applications, and patents mentioned in this specification are herein incorporated by reference.
Various modifications and variations of the methods and compositions of the present invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific desired embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments.
In view of the above, the present invention is characterized by the following embodiments.
A method for treating, inhibiting, reducing or preventing fibrosis in a mammalian subject having or at risk of developing a disease or condition caused or exacerbated by fibrosis and/or inflammation, comprising administering to the subject an amount of a MASP-2 inhibitor effective to inhibit fibrosis.
A method according to paragraph 1A, wherein the MASP-2 inhibitor is a MASP-2 antibody or fragment thereof.
A method according to paragraph 2A, wherein the MASP-2 inhibitor is a MASP-2 monoclonal antibody or fragment thereof that specifically binds to a portion of SEQ ID NO. 6.
A method according to paragraph 2A, wherein the MASP-2 antibody or fragment thereof specifically binds to a polypeptide comprising SEQ ID NO. 6 with an affinity that is at least 10-fold greater than its binding to a different antigen in the complement system.
The method according to paragraph 2A, wherein the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies and human antibodies.
A method according to paragraph 1A, wherein the MASP-2 inhibitor selectively inhibits lectin pathway complement activation without significantly inhibiting C1 q-dependent complement activation.
A method according to paragraph 1A, wherein the MASP-2 inhibitor is administered subcutaneously, intraperitoneally, intramuscularly, intra-arterially, intravenously or as an inhalant.
The method according to any one of paragraphs 1A-7A, wherein the disease or condition caused or exacerbated by fibrosis and/or inflammation is associated with ischemic reperfusion injury.
The method according to any one of paragraphs 1A-7A, wherein the disease or condition caused or exacerbated by fibrosis and/or inflammation is not associated with ischemic reperfusion injury.
The method according to any of paragraphs 1A-7A, wherein the subject exhibits proteinuria prior to administration of the MASP-2 inhibitor, and administration of the MASP-2 inhibitor reduces proteinuria in the subject.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or condition caused or exacerbated by renal fibrosis and/or inflammation.
A method according to paragraph 11A, wherein the MASP-2 inhibitor is administered in an amount effective to inhibit tubular interstitial fibrosis.
The method according to paragraph 11A, wherein the MASP-2 inhibitor is administered in an amount effective to reduce, delay or eliminate the dialysis need in the subject.
The method according to paragraph 11A, wherein the disease or disorder is selected from the group consisting of chronic kidney disease, chronic renal failure, glomerular disease (e.g., focal segmental glomerulosclerosis), immune complex disorders (e.g., igA nephropathy, membranous nephropathy), lupus nephritis, nephrotic syndrome, diabetic nephropathy, tubular interstitial injury and glomerulonephritis (e.g., C3 glomerulopathy).
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by pulmonary fibrosis and/or inflammation.
A method according to paragraph 15A, wherein the disease or disorder is selected from the group consisting of chronic obstructive pulmonary disease, cystic fibrosis, scleroderma-associated pulmonary fibrosis, bronchiectasis, and pulmonary arterial hypertension.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by liver fibrosis and/or inflammation.
A method according to paragraph 17A, wherein the disease or disorder is selected from the group consisting of liver cirrhosis, non-alcoholic fatty liver disease (steatohepatitis), liver fibrosis secondary to alcohol abuse, liver fibrosis secondary to acute or chronic hepatitis, biliary disease, and toxic liver injury (e.g., liver toxicity due to drug-induced liver injury caused by acetaminophen or other drugs, such as nephrotoxins).
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by cardiac fibrosis and/or inflammation.
A method according to paragraph 19A, wherein the disease or condition is selected from the group consisting of cardiac fibrosis, myocardial infarction, valve fibrosis, atrial fibrosis, endocardial myocardial fibrosis, arrhythmogenic Right Ventricular Cardiomyopathy (ARVC).
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by vascular fibrosis.
A method according to paragraph 21A, wherein the disease or disorder is selected from the group consisting of vascular disease, atherosclerotic vascular disease, vascular stenosis, restenosis, vasculitis, phlebitis, deep vein thrombosis, and abdominal aortic aneurysm.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by fibrosis of the skin.
A method according to paragraph 23A, wherein the disease or disorder is selected from the group consisting of excessive wound healing, scleroderma, systemic sclerosis, keloids, connective tissue disease, scarring and hypertrophic scarring.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by fibrosis of the joint.
A method according to paragraph 25A, wherein the disease or disorder is joint fibrosis.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or condition caused or exacerbated by fibrosis of the central nervous system.
A method according to paragraph 27A, wherein the disease or disorder is selected from stroke, traumatic brain injury, and spinal cord injury.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or condition caused or exacerbated by fibrosis of the digestive system.
A method according to paragraph 29A, wherein the disease or disorder is selected from Crohn's disease, pancreatic fibrosis and ulcerative colitis.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by ocular fibrosis.
A method according to paragraph 31A, wherein the disease or condition is selected from the group consisting of subcapsular cataract, posterior capsular opacification, macular degeneration and retinopathy and vitreoretinopathy.
The method of any of paragraphs 1A-7A, wherein the subject has a disease or condition caused or exacerbated by fibrosis of the bone or soft tissue structure of the musculoskeletal bone.
A method according to paragraph 33A, wherein the disease or disorder is selected from the group consisting of osteoporosis and/or osteopenia associated with cystic fibrosis, myelodysplastic conditions with increased bone fibrosis, adhesive capsulitis, dupuytren's contracture, and myelofibrosis.
The method according to any one of paragraphs 1A-7A, wherein the subject has a disease or disorder caused or exacerbated by fibrosis of the reproductive organ.
A method according to paragraph 35A, wherein the disease or disorder is selected from endometriosis and Paeninie's disease.
The method according to any one of paragraphs 1A-7A, wherein the subject has a chronic infectious disease that causes fibrosis and/or inflammation.
A method according to paragraph 37A, wherein the infectious disease is selected from the group consisting of alphavirus, hepatitis A, hepatitis B, hepatitis C, tuberculosis, HIV and influenza.
39A. The method according to any of paragraphs 1A-7A, wherein the subject has an autoimmune disease that causes fibrosis and/or inflammation.
A method according to paragraph 39A, wherein the autoimmune disease is selected from scleroderma and Systemic Lupus Erythematosus (SLE).
The method according to any one of paragraphs 1A-7A, wherein the subject has scarring associated with the wound.
A method according to paragraph 41A, wherein the scar formation associated with the wound is selected from the group consisting of surgical complications (e.g., surgical adhesions, wherein scar tissue may form between internal organ organs, resulting in contracture, pain, and may result in infertility), chemotherapeutic drug-induced fibrosis, radiation-induced fibrosis, and scar formation associated with burns.
The method according to any one of paragraphs 1A-7A, wherein the disease or condition caused or exacerbated by fibrosis and/or inflammation is selected from the group consisting of organ transplantation, breast fibrosis, muscle fibrosis, retroperitoneal fibrosis, thyroid fibrosis, lymph node fibrosis, bladder fibrosis and pleural fibrosis.
A method of preventing or reducing kidney damage in a subject having a disease or condition associated with proteinuria, comprising administering an amount of a MASP-2 inhibitor effective to reduce or prevent proteinuria in the subject.
A method according to paragraph 1B, wherein the MASP-2 inhibitor is a MASP-2 inhibitory antibody or fragment thereof.
A method according to paragraph 1B or 2B, wherein the MASP-2 inhibitor is administered in an amount and for a time effective to achieve at least a 20% reduction in 24-hour urine protein secretion compared to baseline 24-hour urine protein secretion prior to treatment.
The method according to any of paragraphs 1B-3B, wherein the disease or condition associated with proteinuria is selected from nephrotic syndrome, preeclampsia, eclampsia, toxic damage to the kidney, amyloidosis, collagen vascular disease (e.g., systemic lupus erythematosus), lupus nephritis, dehydration, glomerular disease (e.g., membranous glomerulonephritis, focal segmental glomerulonephritis, C3 glomerulopathy, morbid state, steatopathy), intensive exercise, stress, benign orthotopic (posture) proteinuria, focal segmental glomerulosclerosis, igA nephropathy (i.e., begelosis), igM nephropathy, membranous proliferative glomerulonephritis, membranous nephropathy, morbid state, sarcoidosis, alport syndrome, diabetes (diabetic nephropathy), drug-induced toxicity (e.g., NSAIDS, nicotine, penicillamine, lithium carbonate, gold and other heavy metals, inhibitors, antibiotics (e.g., doxorubicin) or formulations (e.g., heroin) or other nephrotoxins); fabry's disease, infection (e.g., HIV, syphilis, hepatitis a, b or c, post streptococcal infection, schistosomiasis urinary); amino acid urine syndrome, van sconey syndrome, hypertensive nephrosclerosis, interstitial nephritis, sickle cell disease, hemoglobinuria, multiple myeloma, myoglobin urine, organ rejection (e.g., kidney transplant rejection), ebola hemorrhagic fever, patella nail syndrome, familial mediterranean fever, HELLP syndrome, systemic lupus erythematosus, wegener's granulomatosis, rheumatoid arthritis, glycogen storage disease type 1, goodpasture's syndrome, allergic purpura, urinary tract infections that have spread to the kidney, sjogren's syndrome, and post-infection glomerulonephritis.
The method according to any one of paragraphs 1B-3B, wherein the disease or condition associated with proteinuria is IgA nephropathy (i.e., begonia disease).
The method according to any one of paragraphs 1B-3B, wherein the disease or condition associated with proteinuria is membranous nephropathy.
The method according to any one of paragraphs 1B-3B, wherein the disease or condition associated with proteinuria is lupus nephritis.
A method of inhibiting the progression of chronic kidney disease comprising administering an amount of a MASP-2 inhibitor effective to reduce or prevent tubular interstitial fibrosis in a subject in need thereof.
A method according to paragraph 1C, wherein the MASP-2 inhibitor is a MASP-2 inhibitory antibody or fragment thereof.
A method according to paragraph 1C, wherein the subject in need thereof exhibits proteinuria prior to administration of the MASP-2 inhibitor, and administration of the MASP-2 inhibitor reduces proteinuria in the subject such that the subject has at least a 20% reduction in 24 hours urine protein secretion compared to baseline 24 hours urine protein secretion in the subject prior to treatment.
4C. the method according to paragraph 1C, wherein the MASP-2 inhibitor is administered in an amount effective to reduce, delay or eliminate the dialysis need in the subject.
A method of protecting a kidney from kidney injury in a subject who has undergone, is undergoing or will undergo treatment with one or more nephrotoxic agents, comprising administering an amount of a MASP-2 inhibitor effective to prevent or ameliorate the incidence of drug-induced kidney disease.
A method according to paragraph 1D, wherein the MASP-2 inhibitor is a MASP-2 inhibitory antibody or fragment thereof.
A method according to paragraph 1D, wherein the MASP-2 inhibitor is administered prior to the nephrotoxic agent.
4D. the method according to paragraph 1D, wherein the MASP-2 inhibitor is co-administered simultaneously with the nephrotoxic agent.
A method according to paragraph 1D, wherein a MASP-2 inhibitor is administered after the nephrotoxic agent to treat nephrotoxicity.
A method of treating a human subject having immunoglobulin a kidney disease (IgAN) comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody or antigen-binding fragment thereof effective to inhibit MASP-2-dependent complement activation.
The method according to paragraph 1E, wherein the subject has steroid dependent IgAN.
A method according to paragraph 1E or 2E, wherein the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2.
The method according to any one of paragraphs 1E-3E, wherein the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies and human antibodies.
The method according to any one of paragraphs 1E-4E, wherein the MASP-2 inhibitory antibody does not significantly inhibit the classical pathway.
The method according to any one of paragraphs 1E-3E, wherein the MASP-2 inhibitory antibody is administered in an IC of 30nM or less 50 Inhibition of C3b deposition in 90% human serum.
A method according to paragraph 2E, wherein the method further comprises identifying a human subject having steroid dependent IgAN prior to the step of administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody or antigen binding fragment thereof effective to improve kidney function.
The method according to any one of paragraphs 1E-7E, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function.
A method according to paragraph 8E, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount and for a time effective to achieve at least a 20% reduction in 24-hour urine protein secretion compared to baseline 24-hour urine protein secretion in the subject prior to treatment.
A method according to paragraph 1E, wherein the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in said subject.
The method according to any one of paragraphs 1E-10E, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence of SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
A method of treating a human subject having Membranous Nephropathy (MN), comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody, or antigen-binding fragment thereof, effective to inhibit MASP-2-dependent complement activation.
A method according to paragraph 1F, wherein the subject has a steroid dependent MN.
A method according to paragraph 1F or 2F, wherein the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2.
The method according to any one of paragraphs 1F-3F, wherein the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies, and human antibodies.
The method according to any one of paragraphs 1F-4F, wherein the MASP-2 inhibitory antibody does not significantly inhibit the classical pathway.
The method according to any one of paragraphs 1F-5F, wherein the MASP-2 inhibitory antibody is administered in an IC of 30nM or less 50 Inhibition of C3b deposition in 90% human serum.
A method according to paragraph 1F, wherein the method further comprises identifying a human subject having a steroid dependent MN prior to the step of administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody or antigen binding fragment thereof effective to improve kidney function.
The method according to any one of paragraphs 1F-7F, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function.
A method according to paragraph 8F, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount and for a time effective to achieve at least a 20% reduction in 24-hour urine protein secretion compared to baseline 24-hour urine protein secretion in the subject prior to treatment.
A method according to paragraph 1F or 2F, wherein the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject.
The method according to any one of paragraphs 1F-10F, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence of SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
A method of treating a human subject having Lupus Nephritis (LN) comprising administering to the subject a composition comprising an amount of a MASP-2 inhibitory antibody, or antigen-binding fragment thereof, effective to inhibit MASP-2-dependent complement activation.
A method according to paragraph 1G, wherein the subject has a steroid dependent MN.
A method according to paragraph 1G or 2G, wherein the MASP-2 inhibitory antibody is a monoclonal antibody or fragment thereof that specifically binds human MASP-2.
The method according to any one of paragraphs 1G-3G, wherein the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies, and human antibodies.
The method according to any one of paragraphs 1G-4G, wherein the MASP-2 inhibitory antibody does not significantly inhibit the classical pathway.
The method according to any one of paragraphs 1G-5G, wherein the MASP-2 inhibitory antibody is administered in an IC of 30nM or less 50 Inhibition of C3b deposition in 90% human serum.
A method according to paragraph 1G, wherein the method further comprises identifying a human subject having steroid dependent LN prior to the step of administering to the subject a composition comprising an amount of MASP-2 inhibitory antibody or antigen binding fragment thereof effective to improve kidney function.
The method according to any one of paragraphs 1G-7G, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount effective to improve kidney function.
A method according to paragraph 8G, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof is administered in an amount and for a time effective to achieve at least a 20% reduction in 24-hour urine protein secretion compared to baseline 24-hour urine protein secretion in the subject prior to treatment.
A method according to paragraph 1G or 2G, wherein the composition is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject.
The method according to any one of paragraphs 1G-10G, wherein the MASP-2 inhibitory antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence of SEQ ID NO. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
While exemplary embodiments have been illustrated and described, it will be appreciated that various changes can be made therein without departing from the spirit and scope of the invention.

Claims (15)

1. Use of a MASP-2 inhibitory monoclonal antibody or antigen-binding fragment thereof that specifically binds human MASP-2 and inhibits MASP-2 dependent complement activation in the manufacture of a medicament for treating a human subject suffering from Lupus Nephritis (LN).
2. The use according to claim 1, wherein the subject has steroid dependent LN.
3. The use according to claim 1, wherein the antibody or fragment thereof is selected from the group consisting of recombinant antibodies, antibodies with reduced effector function, chimeric antibodies, humanized antibodies and human antibodies.
4. The use according to claim 1, wherein the MASP-2 inhibitory antibody does not significantly inhibit the classical pathway.
5. The use according to claim 1, wherein the MASP-2 inhibitory antibody is in an IC of 30nM or less 50 Inhibition of C3b deposition in 90% human serum.
6. Use according to claim 1, wherein prior to the step of administering the medicament, a human subject having steroid dependent LN is identified.
7. The use according to claim 1, wherein the MASP-2 inhibitory antibody or antigen-binding fragment thereof is administered in an effective amount and for a sufficient time to achieve at least a 20% reduction in 24-hour urinary protein secretion compared to baseline 24-hour urinary protein secretion in the pre-treatment subject.
8. The use according to claim 1, wherein the medicament is administered in an amount sufficient to improve kidney function and reduce corticosteroid dosage in the subject.
9. The use according to claim 1, wherein the MASP-2 inhibitory antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence depicted in SEQ ID No. 67; and a light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence shown in SEQ ID NO. 70.
Use of a MASP-2 inhibitory monoclonal antibody or antigen-binding fragment thereof in the manufacture of a medicament for reducing proteinuria in a human subject suffering from IgAN, wherein said MASP-2 inhibitory monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising CDR-H1, CDR-H2 and CDR-H3 of the amino acid sequence depicted in SEQ ID No. 67; and
A light chain variable region comprising CDR-L1, CDR-L2 and CDR-L3 of the amino acid sequence depicted in SEQ ID No. 70: and the medicament is administered according to the following dosage regimen:
a. intravenous administration of about 4mg/kg (i.e., 3.6mg/kg to 4.4 mg/kg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks; or (b)
b. Intravenous administration of about 180mg to about 725mg (i.e., 162mg to 797 mg) of the antibody to a subject having IgAN once a week for a treatment period of at least 12 weeks,
wherein the method reduces proteinuria in the human subject.
11. The use according to claim 10, wherein the treatment period is followed by a rest period of at least 2 months to at least 6 months (i.e. no MASP-2 inhibitor is administered).
12. Use according to claim 10, wherein proteinuria in the subject at the end of the treatment period and/or at the end of the rest period is reduced by at least 20% from baseline (pre-treatment) to at least 50% from baseline (i.e. uACR reduction and/or 24 hour reduction in urine protein concentration); and/or wherein the estimated glomerular filtration rate (eGFR) in the subject is increased, e.g., wherein the subject with IgAN has more than 1 gram of protein per 24 hours of proteinuria secreted by urine protein prior to treatment, and the method is effective to reduce proteinuria in the subject by at least 30%.
13. Use according to claim 10, wherein the urine protein level in the subject is monitored periodically during the treatment period and/or the rest period.
14. The use according to claim 10, wherein the subject stops or significantly reduces the corticosteroid dose at the end of the treatment period and/or at the end of the rest period, as compared to the corticosteroid dose taken prior to the start of treatment with a MASP-2 inhibitory antibody.
15. The use according to claim 10, wherein the MASP-2 inhibitory antibody or fragment thereof comprises a heavy chain variable region comprising the amino acid sequence shown in SEQ ID No. 67 and comprises a light chain variable region shown in SEQ ID No. 70.
CN202310450575.2A 2016-10-13 2017-10-12 Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease Pending CN116726163A (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US201662407979P 2016-10-13 2016-10-13
US62/407979 2016-10-13
US15/399524 2017-01-05
US15/399,524 US10736960B2 (en) 2016-01-05 2017-01-05 Methods for inhibiting fibrosis in a subject in need thereof
US15/470,647 US20170253667A1 (en) 2016-01-05 2017-03-27 Methods for inhibiting fibrosis in a subject in need thereof
US15/470647 2017-03-27
US201762527926P 2017-06-30 2017-06-30
US62/527926 2017-06-30
PCT/US2017/056386 WO2018071701A1 (en) 2016-10-13 2017-10-12 Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy
CN201780060710.5A CN110177557A (en) 2016-10-13 2017-10-12 Method for reducing the albuminuria in the people experimenter for suffering from IgANP

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201780060710.5A Division CN110177557A (en) 2016-10-13 2017-10-12 Method for reducing the albuminuria in the people experimenter for suffering from IgANP

Publications (1)

Publication Number Publication Date
CN116726163A true CN116726163A (en) 2023-09-12

Family

ID=61906454

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201780060710.5A Pending CN110177557A (en) 2016-10-13 2017-10-12 Method for reducing the albuminuria in the people experimenter for suffering from IgANP
CN202310450575.2A Pending CN116726163A (en) 2016-10-13 2017-10-12 Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201780060710.5A Pending CN110177557A (en) 2016-10-13 2017-10-12 Method for reducing the albuminuria in the people experimenter for suffering from IgANP

Country Status (18)

Country Link
EP (1) EP3525798A4 (en)
JP (1) JP6893554B2 (en)
KR (1) KR102348939B1 (en)
CN (2) CN110177557A (en)
AU (1) AU2017342428B2 (en)
BR (1) BR112019007426A2 (en)
CA (2) CA3039927C (en)
CL (2) CL2019000909A1 (en)
GE (1) GEP20247587B (en)
IL (1) IL265981A (en)
JO (1) JOP20190068A1 (en)
MA (1) MA46541A (en)
MX (1) MX2019004074A (en)
PH (1) PH12019500711A1 (en)
SG (1) SG11201902941UA (en)
UA (1) UA126908C2 (en)
WO (1) WO2018071701A1 (en)
ZA (1) ZA201902933B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111419860A (en) * 2020-03-19 2020-07-17 长春市儿童医院 Glomerular lobular nephropathy modeling method
CN115215937B (en) * 2021-04-15 2023-05-26 上海麦济生物技术有限公司 Anti-human MASP-2 antibody, preparation method and application thereof
CN117016486A (en) * 2023-07-20 2023-11-10 澎立生物医药技术(上海)股份有限公司 Animal model construction method for IgA nephropathy combined membranous nephropathy

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE024996T2 (en) * 2003-05-12 2016-01-28 Helion Biotech Aps Antibodies to masp-2
US20140056873A1 (en) * 2004-06-10 2014-02-27 University Of Leicester Methods for Treating Conditions Associated with MASP-2 Dependent Complement Activation
US8840893B2 (en) * 2004-06-10 2014-09-23 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
US7919094B2 (en) * 2004-06-10 2011-04-05 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
US7626730B2 (en) 2006-03-31 2009-12-01 Eastman Kodak Company Method of making a multilevel halftone screen
BR112012008970A2 (en) * 2009-10-16 2019-12-10 Omeros Corp use of a composition and composition
US9102721B2 (en) * 2011-01-21 2015-08-11 Fibrogen, Inc. Therapeutic method
MX355648B (en) 2011-04-08 2018-04-26 Univ Leicester Methods for treating conditions associated with masp-2 dependent complement activation.
US9644035B2 (en) * 2011-04-08 2017-05-09 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
HUE049154T2 (en) * 2011-05-04 2020-09-28 Omeros Corp Compositions for inhibiting masp-2 dependent complement acitivation
WO2014144542A2 (en) * 2013-03-15 2014-09-18 Omeros Corporation Methods of generating bioactive peptide-bearing antibodies and compositions comprising the same
DK3057993T3 (en) * 2013-10-17 2020-11-16 Omeros Corp Method for Treating Conditions Associated with MASP-2 Dependent Complement
SG11201805695SA (en) * 2016-01-05 2018-07-30 Univ Leicester Methods for inhibiting fibrosis in a subject in need thereof
JOP20170170B1 (en) * 2016-08-31 2022-09-15 Omeros Corp Highly concentrated low viscosity masp-2 inhibitory antibody formulations, kits, and methods

Also Published As

Publication number Publication date
ZA201902933B (en) 2023-05-31
CN110177557A (en) 2019-08-27
EP3525798A4 (en) 2020-07-08
JP6893554B2 (en) 2021-06-23
PH12019500711A1 (en) 2019-11-18
KR20190063475A (en) 2019-06-07
BR112019007426A2 (en) 2019-07-02
JP2019534271A (en) 2019-11-28
AU2017342428B2 (en) 2021-02-04
KR102348939B1 (en) 2022-01-12
MX2019004074A (en) 2019-06-10
UA126908C2 (en) 2023-02-22
WO2018071701A1 (en) 2018-04-19
GEP20247587B (en) 2024-01-25
EP3525798A1 (en) 2019-08-21
CA3039927A1 (en) 2018-04-19
CA3039927C (en) 2023-10-10
AU2017342428A1 (en) 2019-05-23
IL265981A (en) 2019-06-30
SG11201902941UA (en) 2019-05-30
JOP20190068A1 (en) 2019-04-01
MA46541A (en) 2019-08-21
NZ753260A (en) 2021-11-26
CA3210384A1 (en) 2018-04-19
CL2019000909A1 (en) 2019-06-14
CL2019003485A1 (en) 2020-04-13

Similar Documents

Publication Publication Date Title
JP7430208B2 (en) Methods for inhibiting fibrosis in a subject in need thereof
CN117582494A (en) Methods of inhibiting angiogenesis in a subject in need thereof
US20210355236A1 (en) Methods for inhibiting fibrosis in a subject in need thereof
US20210079116A1 (en) Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy
CN115335120A (en) Method for inhibiting MASP-2 for the treatment and/or prevention of coronavirus induced acute respiratory distress syndrome
CN116726163A (en) Methods for reducing proteinuria in human subjects with immunoglobulin A kidney disease
EA043102B1 (en) METHOD FOR REDUCING PROTEINURIA IN A HUMAN SUFFERING FROM IMMUNOGLOBULIN A NEPHROPATHY
OA18820A (en) Methods for inhibiting fibrosis in a subject in need thereof.
OA19229A (en) Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy.

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40099516

Country of ref document: HK