WO2025186364A1 - Anticorps ciblant angpt2 destinés à être utilisés dans le traitement de maladies fibrotiques, en particulier d'une ou de plusieurs maladies rénales, d'un ou plusieurs troubles ou d'une lésion associés à une fibrose rénale - Google Patents
Anticorps ciblant angpt2 destinés à être utilisés dans le traitement de maladies fibrotiques, en particulier d'une ou de plusieurs maladies rénales, d'un ou plusieurs troubles ou d'une lésion associés à une fibrose rénaleInfo
- Publication number
- WO2025186364A1 WO2025186364A1 PCT/EP2025/056090 EP2025056090W WO2025186364A1 WO 2025186364 A1 WO2025186364 A1 WO 2025186364A1 EP 2025056090 W EP2025056090 W EP 2025056090W WO 2025186364 A1 WO2025186364 A1 WO 2025186364A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- antibody
- antigen
- amino acid
- acid sequence
- 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
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/22—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
Definitions
- ANGPT2-targeting antibodies for use in the treatment of fibrotic diseases, particularly kidney disease(s), disorder(s) or injury associated with kidney fibrosis.
- the present invention relates to ANGPT2-targeting antibodies for use in the treatment or prevention of a fibrotic disease, particularly kidney disease(s), disorder(s) or injuries in a subject, wherein the kidney disease(s), disorder(s) or injury is associated with kidney fibrosis.
- CKD Chronic kidney disease
- Angiopoietins are a family of vascular growth factors, comprised of three angiopoietins (ANGPT1 , ANGPT2, ANGPT3/ANGPT4 (mouse/human, respectively), that share multiple cellular functions related to cell survival, proliferation, migration, and inflammation.
- ANGPTs play physiological and pathological roles through the TIE tyrosine kinase receptors.
- the ANGPT-TIE2 signalling pathway participates in the developmental and tumour-induced angiogenesis and is also involved in many disease settings, such as vascular diseases, systemic inflammation, and cancers.
- angiopoietin signalling is involved in the regulation of renal vascular development and in response to injury.
- the endothelial growth factor TIE2 (also known as TEK), is a tyrosine kinase receptor mainly expressed on endothelial cells. TIE2 signalling promotes vascular quiescence and reduces vascular permeability, inflammation, and endothelial apoptosis. TIE2 activity is regulated by angiopoietin (ANGPT) ligands ANGPT1 and ANGPT2, as well as the vascular endothelial tyrosine phosphatase (VEPTP). ANGPT2-binding antibodies have been reported (e.g., US 7,658,924, US 8,987,420 and US 11,498,962) largely in the context of cancer therapeutics.
- ANGPT2 angiopoietin
- vascular endothelial fibrosis include the peptide-Fc fusion protein; trebananib (also known as AMG386), and the ANGPT2- targeting monoclonal antibodies; MEDI3617, LY3127804 and nesvacumab (also known as REGN910), neutralize the interaction of ANGPT2 and TIE2 (Saharinen et al., 2017).
- trebananib also known as AMG386
- ANGPT2- targeting monoclonal antibodies include peptide-Fc fusion protein; trebananib (also known as AMG386), and the ANGPT2- targeting monoclonal antibodies; MEDI3617, LY3127804 and nesvacumab (also known as REGN910), neutralize the interaction of ANGPT2 and TIE2 (Saharinen et al., 2017).
- MEDI3617 also known as AMG386
- LY3127804 also known as REGN910
- renin-angiotensin system RAS
- GLP-1 receptor agonists GLP-1 receptor agonists
- Atrasentan endothelin-1 blocker
- Tolvaptan vasopressin receptor 2 antagonist
- Finerenone non-steroidal antimineralocorticoid
- TIE2-activating therapeutics have shown great potential for the treatment of fibrotic diseases, for example kidney disease.
- certain disadvantages are associated with the use of said TIE2-activating therapeutics which have limited their clinical use, such as, for instance, limitations related to production, storage, half-life, specificity, and efficacy of these compounds. Accordingly, there is an unmet need for TIE2-activating therapeutics for the treatment of fibrotic diseases, for example kidney disease, having differential characteristics.
- ANGPT2-binding antibodies such as ABTAA
- ABTAA have shown great therapeutic potential in cancer, sepsis and neovascular age-related macular degeneration, however, it is not known whether ABTAA has efficacy in treating the complex pathophysiology associated with fibrotic diseases, for example, kidney disease wherein said kidney disease is associated with kidney fibrosis.
- the present inventors have found that the ANGPT2-binding and TIE2-activating antibody, ABTAA, has therapeutic efficacy in the treatment of fibrotic diseases, and particularly kidney diseases, wherein said kidney disease is associated with kidney fibrosis, as outlined herein.
- the present examples demonstrate the unexpected therapeutic effect of ABTAA- induced ANGPT2 and TIE2-activation in an in vivo model of kidney disease, wherein ABTAA prevents the development of tubulointerstitial fibrosis compared to IgG-injected and WT mice.
- TIE2 iECK0 mice had aggravated disease and significantly more tubulointerstitial fibrosis compared to WT and ABTAA-treated mice.
- the present examples demonstrate the unexpected therapeutic effect of ABTAA- induced ANGPT2 and TIE2-activation in an in vivo model of early diabetic complications, wherein ABTAA normalised plasma blood urea nitrogen levels, indicating protection against nephropathy.
- ABTAA also significantly preserved capillary density in the renal cortex and maintained normal fenestrations in peritubular capillaries, which are crucial for kidney function.
- ABTAA treatment prevented the increase of plasma creatinine kinase, suggesting protection against cardiac muscle injury.
- ABTAA significantly reduced vessel regression (i.g. the number of empty collagen sleeves) in the retina, indicating preserved capillary structure.
- the present examples demonstrate the unexpected therapeutic effect of ABTAA- induced ANGPT2 and TIE2-activation in an in vivo model of coagulopathy, wherein ABTAA completely prevented the TNFa-induced decrease in tail bleeding time, indicating a protective effect on coagulopathy.
- ABTAA exhibits an unexpected dual action of normalising pathological, unstable blood vessels and preventing the progression of fibrosis in kidney disease.
- this avoids adverse effects associated with over-activation of TIE2, such as vascular enlargement and venous malformation (Vikkula et al., 1996).
- This anti-fibrotic effect of ABTAA has not previously been reported.
- ABTAA as a therapeutic for fibrotic diseases, for example, kidney disease is advantageous due to its long half-life in circulation: 198 hours, compared to other TIE2 activators, such as COMP-ANGPT1 which has a half-life of 30 minutes (Han et al., 2016).
- the present invention is directed to antibodies or antigen-binding fragments thereof, that bind to ANGPT2 and bind to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a fibrotic disease.
- the present invention is directed to antibodies or antigen-binding fragments thereof, that bind to ANGPT2 and bind to TIE2 receptor via ANGPT2, for use in the treatment or prevention of fibrosis.
- the present invention is directed to antibodies or antigen-binding fragments thereof, that bind to ANGPT2 and bind to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- the antibodies or antigen-binding fragments thereof are characterised by binding to a site from position 336 to position 434 of Angiopoietin-2 (ANGPT2) (SEQ ID NO: 30).
- ANGPT2 Angiopoietin-2
- the antibodies or antigen-binding fragments thereof comprise: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1 , SEQ ID NO: 24 or SEQ ID NO: 35, the LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 25 or SEQ ID NO: 36, and the LCDR3 comprising an amino acid sequence selected from the
- the antibodies or antigen-binding fragments thereof comprise a variable heavy chain of SEQ ID NO: 8 or SEQ ID NO: 10 or SEQ ID NO: 31 or SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 7 or SEQ ID NO: 11 or SEQ ID NO: 32 or SEQ ID NO: 34 or SEQ ID NO: 38.
- the antibodies or antigen-binding fragments thereof comprise a variable heavy chain of SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 34.
- the antibodies or antigen-binding fragments thereof comprise complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence of SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence of SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence of SEQ ID NO: 29, and complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence of SEQ ID NO: 35, the LCDR2 comprising an amino acid sequence SEQ ID NO: 36, and the LCDR3 comprising an amino acid sequence of SEQ ID NO: 26.
- CDRs complementarity determining regions
- the antibodies or antigen-binding fragments thereof described herein are encoded by the nucleotide sequences as described in US Patent No. 9,994,632 B2.
- the present invention is directed to methods of: a) decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in fibrotic diseases, b) normalising pathological blood vessels and treating and reducing and preventing the progression of fibrotic diseases, c) treating and/or reducing and/or preventing the progression of fibrotic diseases, d) preventing or treating a reduction in perfusion in a subject, wherein the disease, disorder or injury is associated with fibrosis, e) preventing or reducing capillary rarefaction associated with the disease, disorder or injury in a subject, wherein the disease, disorder or injury is associated with fibrosis, and/or comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the
- the present invention is directed to methods of: a) decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in a diseased kidney, wherein the diseased kidney is fibrotic, b) normalising pathological blood vessels and treating and reducing and preventing the progression of fibrosis in a diseased kidney, c) treating and/or reducing and/or preventing the progression of fibrosis in a diseased kidney, d) preventing or treating a reduction in kidney perfusion in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis, e) preventing or reducing capillary rarefaction associated with the kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis, and/or f) preventing or treating a reduction of fenestrations of peritubular capillaries associated with a kidney
- FIG. 1 Schematic diagram of ABTAA administration, experimental setup, and (B) inducible conditional mouse lines.
- FIG. 3 TIE2-activation reduced after UUO.
- FIG. 4 TIE2-activation by ABTAA reduces UUO-induced renal perfusion. Renal perfusion measured after intravenous injection of the circulating vascular marker MicroMarker and contrast imaging ultrasound in 2-day UUO and CL kidneys, expressed as UUO/CL from ABTAA treated (ABT), Veptp iECKO (Ve KO ), 77E2 iECKO (T2 KO ), and Pdgfb' KO (Pb KO ) mice and their controls. Data is presented as mean (SD), n as dots in graphs. # P ⁇ 0.05, # # # # # P ⁇ 0.001 compared to WT/IgG UUO within the same group using Student’s t test (unpaired, 2-tailed). **P ⁇ 0.01 , ****P ⁇ 0.0001 compared to WT/IgG CL using one-way ANOVA followed by Bonferroni post hoc test.
- FIG. 6 TIE2-activation by ABTAA reduces UUO-induced endothelial injury.
- FIG. 7 TIE2-activation by ABTAA reduces UUO-induced tubulointerstitial fibrosis.
- D Renal Col1a1 expression in ABTAA treated mice.
- Tubular segments with pathological vacuoles indicated with white arrow heads in 3-day UUO kidney sections stained with Toluidine blue.
- B Quantification of tubular segments with vacuoles for ABTAA treated (ABT), Veptp' ECKO (Ve KO ), and 77E2 iECKO (T2 KO ) mice. Data is presented as mean (SD), n as dots in graphs. # P ⁇ 0.05compared to WT/IgG UUO within the same group using Student’s t test (unpaired, 2-tailed).
- A-C Expression of Pdgfb mRNA (A, C) and PDGFB protein (B) in 3-day UUO kidneys from ABTAA treated (ABT), Veptp' ECKO (VeKO), and Pdgfb KO (Pb KO ) mice.
- FIG. 10 Late onset treatment with ABTAA reduces UUO-induced injury.
- A Schematic diagram of administration of ABTAA for evaluation in 10-day UUO kidneys.
- E Protein concentration for PDGFB in 10-day UUO kidneys from ABTAA treated (ABT) mice. Data is presented as mean (SD), n as dots in graphs.
- ANGPT2 staining in glomeruli of diabetic BTBRob/ob mice compared to non-diabetic mice (BTBRLean).
- B Preliminary results from the first cohort of BTBRob/ob studies treated with 25 mg/kg ABTAA (ABT) or IgG compared to non-diabetic controls (ND). Mice started treatment at 4-weeks of age and were studied until 11-weeks of age.
- ABT ABTAA
- ND non-diabetic controls
- Mice started treatment at 4-weeks of age and were studied until 11-weeks of age.
- C Kidney function (nephropathy) was evaluated by plasma blood urea nitrogen (BUN).
- D Capillary density of peritubular capillaries in renal cortex was quantified with endomucin (EMCN) staining.
- Plasma creatinine kinase was used as a marker of cardiac injury.
- FIG. 12 ABTAA therapy is protective in coagulopathy.
- TNFa triggers release of ANGPT2 in mouse plasma.
- B ABX treatment in TNFa induced coagulopathy with the tail bleeding model. Data presented as mean (SD). **P ⁇ 0.01, ****P ⁇ 0.0001 compared to controls Student’s t test (unpaired, 2-tailed).
- antibody and “antibodies” (immunoglobulins) may be an oligoclonal antibody, a polyclonal antibody, a monoclonal antibody (including full-length monoclonal antibodies), a camelised antibody, a chimeric antibody, a CDR-grafted antibody, a multi-specific antibody, a bi-specific antibody, a catalytic antibody, a chimeric antibody, a humanized antibody, a fully human antibody, an anti-idiotypic antibody and antibodies that can be labelled in soluble or bound form as well as fragments, variants or derivatives thereof, either alone or in combination with other amino acid sequences provided by known techniques.
- An antibody may be from any species.
- An antibody comprises a polypeptide or group of polypeptides that are comprised of at least one binding domain that is formed from the folding of polypeptide chains having three-dimensional binding spaces with internal surface shapes and charge distributions complementary to the features of an antigenic determinant of an antigen.
- An antibody typically has a tetrameric form, comprising two identical pairs of polypeptide chains, each pair having one "light” and one "heavy” chain. The variable regions of each light/heavy chain pair form an antibody binding site.
- Native antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains.
- Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes.
- Each heavy and light chain also has regularly spaced intrachain disulfide bridges.
- Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains.
- Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
- Light chains are classified as either lambda chains or kappa chains based on the amino acid sequence of the light chain constant region.
- variable domain of a kappa light chain may also be denoted herein as VK.
- variable region may also be used to describe the variable domain of a heavy chain or light chain. Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains.
- the variable regions of each light/heavy chain pair form an antibody binding site.
- antibody fragment refers to any derivative of an antibody that is less than full-length. In one aspect, the antibody fragment retains at least a significant portion of the full-length antibody's specific binding ability, specifically, as a binding partner. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, scFv, Fv, dsFv diabody, and Fd fragments.
- the antibody fragment may be produced by any means. For example, the antibody fragment may be enzymatically or chemically produced by fragmentation of an intact antibody, or it may be recombinantly produced from a gene encoding the partial antibody sequence. Alternatively, the antibody fragment may be wholly or partially synthetically produced.
- the antibody fragment may comprise a single chain antibody fragment.
- the fragment may comprise multiple chains that are linked together, for example, by disulfide linkages.
- the fragment may also comprise a multimolecular complex.
- a functional antibody fragment may typically comprise at least about 50 amino acids and more typically will comprise at least about 200 amino acids.
- the term “monoclonal antibody” refers to an antibody that is derived from a single cell clone, including any eukaryotic or prokaryotic cell clone, or a phage clone, and not the method by which it is produced. Thus, the term “monoclonal antibody” is not limited to antibodies produced through hybridoma technology.
- ANGPT2-binding antibody refers to antibody that binds to ANGPT2 resulting in alteration of the biological activity of ANGPT2 and is used interchangeably with “anti-ANGPT2 antibody”, “ANGPT2-binding antibody”, “antibody specifically binding to ANGPT2”.
- the “antibody” used herein is an immunoglobulin molecule which is immunologically reactive to a specific antigen and means a protein molecule acting as a receptor that specifically recognizes an antigen, and may include all of a polyclonal antibody, a monoclonal antibody (single clone antibody), a whole antibody, and an antibody fragment. Further, the antibody may include a chimeric antibody (e.g., humanized murine antibody) and a bivalent or bispecific molecule (e.g., bispecific antibody), a diabody, a triabody, and a tetrabody. In some preferred embodiments, the antibody described herein is a whole antibody.
- ABSTAA refers to a group of angiopoietin-2-binding, TIE2- activating antibodies which bind to ANGPT-2 and bind to TIE2 receptor via ANGPT2, thereby activating the TIE2 receptor.
- ABTAA can be used interchangeably with “ABT”, “ABTAA”, and “ABX”.
- the group includes the anti-ANGPT2 antibody of US 11 ,498,962 and the anti-ANGPT2 antibody of US 2015/337033 A1.
- pathological blood vessel refers to a pathological condition or pathophysiological process wherein blood vessels exhibit abnormal or pathological characteristics that deviate from the normal vascular architecture and function. These aberrant vessels may be associated with various medical conditions, including but not limited to, cancer, inflammatory disorders, vascular malformations, and certain ocular diseases. Characteristics of pathological blood vessels may include excessive angiogenesis, irregular vessel branching, increased permeability, loss of fenestrations, loss of capillary density, and compromised structural integrity.
- endothelial dysfunction refers to a pathological condition or pathophysiological process characterized by impaired or abnormal functioning of the endothelium, the inner lining of blood vessels. Endothelial dysfunction involves a disturbance in the regulation of various physiological processes, including the control of vascular tone, blood clotting, and the inflammatory response. It is associated with an imbalance in the production and availability of endothelium-derived factors, including, but not limited to ANGPT2, thrombomodulin, nitric oxide and endothelin, which play crucial roles in vascular homeostasis.
- fibrotic disease refers to any disease or condition caused by or associated with fibrosis.
- Fibrosis is a pathological condition or pathophysiological process characterized by the excessive accumulation of extracellular matrix (ECM) components, leading to the replacement of normal tissue architecture with fibrous tissue. This process results in the disruption of normal organ function and can affect various organs, including the kidneys and heart. Fibrotic disease involves an imbalance in the regulation of ECM production and degradation, often associated with chronic inflammation, tissue injury, and abnormal wound healing responses. Fibrosis may be associated with several chronic conditions, including type 2 diabetes. Fibrosis may also be induced by, propagated by and/or associated with coagulopathy. As used herein, the term “fibrotic tissue” refers to any tissue that is associated with fibrosis. Fibrotic tissue can be identified as it is typically hard, fibrous, thick, and inflexible, when compared to healthy tissue. Fibrotic tissue may also be referred to herein as “diseased tissue”.
- Fibrosis is defined as the replacement of functional tissue parenchyma with extracellular matrix (ECM) and can be used interchangeably with “sclerosis” and/or “scarring”.
- ECM extracellular matrix
- fibrosis of a transplanted kidney predominantly affects the tubulointerstitium which represents 90 percent of kidney volume.
- IFTA tubulointerstitial fibrosis/interstitial fibrosis/tubular atrophy
- IFTA can be the result of various renal insults, such as ischemia-reperfusion injury, rejection, infection, diabetes, and pharmacological treatment (e.g., anticalcineurinic agents).
- kidney fibrosis refers to a pathological condition or pathophysiological process characterized by the excessive accumulation of extracellular matrix (ECM) proteins, including, but not limited to, collagen, smooth muscle actin, and vimentin, within the renal tissue. This fibrotic process involves the activation of myofibroblasts, leading to the formation of scar tissue and disruption of the normal architecture of the kidney. Kidney fibrosis is often associated with various chronic kidney diseases and represents a progressive and irreversible condition that can result in impaired renal function.
- ECM extracellular matrix
- kidney disease(s), disorder(s) or injuries, wherein the kidney disease, disorder or injury is associated with kidney fibrosis refers to any condition having kidney fibrosis as a symptom or cause of the condition, or a condition that can be worsened by the development of kidney fibrosis, or a condition the progression of which is linked to the progression of kidney fibrosis.
- a condition associated with kidney fibrosis can therefore benefit therapeutically by preventing/inhibiting and/or delaying the progression of kidney fibrosis.
- Conditions associated with kidney fibrosis include, but are not limited to, diabetic nephropathy, chronic kidney disease, end-stage renal disease, systemic lupus erythematosis, vasculitis, IgA nephropathy, other autoimmune diseases, paraprotein diseases, diabetes. Since chronic kidney disease associated with kidney fibrosis is a very important risk factor for cardiovascular disease, it would be apparent to a skilled artisan that a therapeutic that prevented or reduced kidney fibrosis would have a beneficial effect on cardiac and vascular disease throughout the body.
- tubulointerstitial fibrosis refers to a pathological condition or pathophysiological process characterized by the excessive accumulation of fibrous tissue, including, but not limited to, collagen, smooth muscle actin, and vimentin, within the tubulointerstitial compartment of the kidney. This fibrotic process involves the activation of myofibroblasts and the deposition of extracellular matrix, leading to structural changes, tissue scarring, and impaired kidney function. Tubulointerstitial fibrosis is often associated with various kidney disorders, including chronic kidney diseases and inflammatory conditions.
- glomerulosclerosis refers to a pathological condition or pathophysiological process characterized by scaring and excessive accumulation of extracellular matrix, including, but not limited to, collagen and smooth muscle actin within the glomerular compartment of the kidney.
- the scaring process involves the activation of mesangial cells and the deposition of extracellular matrix, leading to structural changes, tissue scaring, and impaired kidney function.
- Glomerulosclerosis is often associated with various kidney disorders, including chronic kidney diseases.
- capillary rarefaction refers to a pathological condition or pathophysiological process characterized by a diminution in the microvascular density, specifically within the capillary network of a given tissue or organ. This phenomenon involves the quantifiable reduction in the number of capillaries, leading to compromised microcirculatory perfusion, tissue hypoxia, and perturbed physiological function.
- Capillary rarefaction may manifest in diverse biological contexts and is associated with pathological states wherein the microvascular architecture undergoes structural alterations, thereby adversely impacting tissue perfusion and homeostasis.
- tubular atrophy refers to a pathological condition or pathophysiological process characterized by the degeneration and necrosis of renal tubular epithelial cells within the kidney nephron. This process entails the loss of cellular integrity and functional capacity of the renal tubules, commonly observed in the context of chronic kidney diseases and various renal pathologies. Tubular atrophy contributes significantly to the disruption of normal renal histology, compromises tubular integrity, and is indicative of progressive renal damage.
- tubular injury refers to a pathological condition or pathophysiological process characterized by deleterious alterations in the structural and functional integrity of renal tubules within the kidney nephron. Such injury encompasses morphological changes, compromised cellular integrity, and functional impairment of the tubular epithelial cells. T ubular injury is a hallmark of various renal pathologies, including acute kidney injury (AKI) and chronic kidney diseases (CKD).
- AKI acute kidney injury
- CKD chronic kidney diseases
- oxidative stress refers to an imbalance between the production and manifestation of excessive levels of molecular oxygen or reactive oxygen species (ROS) and a biological system's ability to readily detoxify the reactive intermediates or to repair the resulting damage via endogenous antioxidant systems. Disturbances in the normal redox state of tissues can cause toxic effects through the production of peroxides and free radicals that damage all components of the cell, including proteins, lipids, and DNA via mechanisms including lipid peroxidation, protein oxidation and aggregation and DNA damage. Some reactive oxidative species can even act as messengers through a phenomenon called redox signalling.
- ROS reactive oxygen species
- hypoxia refers to a pathological condition or pathophysiological process in which the kidney or a region of the kidney is deprived of adequate oxygen supply at the tissue level. Such injury may be the result of decreased perfusion of renal blood vessels, loss of capillary density, or altered capillary function. Hypoxia is a hallmark of various renal pathologies, including chronic kidney diseases (CKD).
- CKD chronic kidney diseases
- treatment is an approach for obtaining beneficial or desired results including clinical results.
- beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease or disorder, stabilized (/.e., not worsening) state of disease or disorder, prevention of the disease or disorder, delay or slowing of disease or disorder progression, amelioration or palliation of the disease state, and remission (whether partial or total) whether detectable or undetectable.
- chronic kidney disease refers to the presence of kidney damage including, but not limited to, tubulointerstitial fibrosis, glomerulosclerosis and/or an estimated glomerular filtration rate (eGFR) less than 60 ml/min per 1.73 square meters, persisting for 90 days or more. It is a state of progressive loss of kidney function ultimately resulting in the need for renal replacement therapy (dialysis or transplantation).
- CKD has its general meaning in the art and is used to classify numerous conditions that affect the kidney, destruction of the renal parenchyma and the loss of functional nephrons or glomeruli. Examples of aetiology of CKD include, but are not limited to, cardiovascular diseases, hypertension, diabetes, glomerulonephritis, polycystic kidney diseases, and kidney graft rejection.
- coagulopathy refers to a pathological condition or pathophysiological process in which the blood coagulation system is impaired, which may cause formation of blood clots/thrombosis.
- the present invention is directed to antibodies or antigen-binding fragments that bind to ANGPT2 and bind to TIE2 receptor via ANGPT2, for use in the treatment or prevention of fibrotic diseases.
- the present invention is directed to antibodies or antigen-binding fragments that bind to ANGPT2 and bind to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- the antibodies or antigen-binding fragments thereof are characterised by binding to a site from position 336 to position 434 of Angiopoietin-2 (ANGPT2) (SEQ ID NO: 30).
- the binding site is about 2 to about 20 contiguous amino acids in length, such about 2 to about 18, 5 to about 20, or about 10 to about 20, such as about 15 to 20.
- the antibodies or antigen-binding fragments thereof are characterised by binding to the epitope of SEQ ID NO: 9 or SEQ ID NO: 30 of human Angiopoietin-2 (ANGPT2).
- ANGPT2 human Angiopoietin-2
- the antibodies or antigen-binding fragments thereof comprise: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ I D NO: 1 , SEQ I D NO: 24 or SEQ I D NO: 35, the LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 25 or SEQ ID NO: 36, and the LCDR3 comprising an amino acid sequence
- the antibodies or antigen-binding fragments thereof comprise: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3, or the LCDR1 amino acid sequence of SEQ ID NO
- the antibodies or antigen-binding fragments thereof comprise: a variable heavy chain of SEQ ID NO: 8 or SEQ ID NO: 10 or SEQ ID NO: 31 or SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 7 or SEQ ID NO: 11 or SEQ ID NO: 32 or SEQ ID NO: 34 or SEQ ID NO: 38.
- the antibodies or antigen-binding fragments thereof comprise: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- the antibodies or antigen-binding fragments thereof comprise: a variable heavy chain of SEQ ID NO: 8 or SEQ ID NO: 10, and a variable light chain of SEQ ID NO: 7 or SEQ ID NO: 11.
- the antibodies or antigen-binding fragments thereof are characterised by binding to the epitope of SEQ ID NO: 9 of human Angiopoietin-2 (ANGPT2).
- the antibodies or antigen-binding fragments thereof comprise: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of SEQ ID NO: 25 and the LCDR3 amino acid sequence of SEQ ID NO: 26, or the LCDR1 amino acid sequence of SEQ ID NO: 35, the LCDR2 amino acid sequence of SEQ ID NO: 36 and the LCDR3 amino acid sequence of SEQ ID NO: 26.
- CDRs complementarity determining regions
- the antibodies or antigen-binding fragments thereof comprise: a variable heavy chain of SEQ ID NO: 31 or SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 32 or SEQ ID NO: 34 or SEQ ID NO: 38.
- the antibodies or antigen-binding fragments thereof are characterised by binding to the epitope of SEQ ID NO: 30 of human Angiopoietin-2 (ANGPT2).
- ANGPT2 human Angiopoietin-2
- the amino acid sequence of the antibodies or antigen-binding fragments of the present disclosure may be substituted by conservative substitution.
- conservative substitution refers to modification of polypeptide including substitution of at least one amino acid with an amino acid having similar biochemical properties to corresponding polypeptide without causing loss of biological or biochemical function.
- Consservative amino acid substitution refers to a substitution in which an amino acid residue is replaced with an amino acid residue having similar side chains. Classes of the amino acid residues having similar side chains are defined in the art.
- amino acids having basic side chains e.g., lysine, arginine, histidine
- amino acids having acidic side chains e.g., aspartic acid, glutamic acid
- amino acids having uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
- amino acids having non-polar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
- amino acids having beta-branched side chains e.g., threonine, valine, isoleucine
- amino acids having aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
- the antibody of the present invention is able to still retain an activity while having the conservative amino acid substitution.
- the antibody or antigen-binding fragment thereof is characterized by containing the complementary determining regions (CDRs) of an antibody produced from a cell line deposited with accession number KCLRF-BP-00417 at the Korean Cell Line Bank (KCLB) at Cancer Research Institute, Seoul National University, College of Medicine, 28 Yongon-dong, Chongno-Gu, Seoul, 110-744, Korea on Jan. 30, 2018, or a hybridoma deposited with accession number KCLRF- BP-00295 at the Korean Cell Line Bank located at Yongon-dong, Chongno-gu, Seoul, South Korea, as of Apr. 23, 2013.
- CDRs complementary determining regions
- sequences of the antibodies or antigen-binding fragments of the invention may vary from the sequences provided in the present application.
- amino sequences may vary from those set out above in that (a) the variable regions may be segregated away from the constant domains of the light chains, (b) the amino acids may vary from those set out above while not drastically affecting the chemical properties of the residues thereby (so-called conservative substitutions), (c) the amino acids may vary from those set out above by a given percentage, e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology.
- the nucleic acids encoding the antibodies may (a) be segregated away from the constant domains of the light chains, (b) vary from those set out above while not changing the residues coded thereby, or (c) may vary from those set out above by a given percentage, e.g., 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology.
- the hydropathic index of amino acids may be considered.
- the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art. It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
- substitution of like amino acids can be made effectively on the basis of hydrophilicity.
- hydrophilicity the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein.
- an amino acid can be substituted for another having a similar hydrophilicity and produce a biologically or immunologically modified protein.
- substitution of amino acids whose hydrophilicity values are within +1-2 is preferred, those that are within +/-1 are particularly preferred, and those within +/-0.5 are even more particularly preferred.
- amino acid substitutions generally are based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
- Exemplary substitutions that take into consideration the various foregoing characteristics are well known to those of skill in the art and include arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
- the antibody or antigen-binding fragment thereof may bind to human and/or mouse ANGPT2. In one embodiment, the antibody or antigen-binding fragment thereof binds to human ANGPT2. In one embodiment, the antibody or antigen-binding fragment thereof binds to mouse ANGPT2.
- the antibody or antigen-binding fragment thereof is polyclonal or monoclonal. In one embodiment, the antigen-binding fragment thereof is a scFv or a Fab.
- the antibody or antigen-binding fragment thereof may be humanized or human.
- the complete antibody has a structure having two full-length light chains and two full- length heavy chains, wherein each light chain may be linked to the corresponding heavy chain by a disulfide bond.
- the whole antibody may include IgA, IgD, IgE, IgM, an/or IgG, and the IgG is a subtype, and may include lgG1, lgG2, lgG3, and/or lgG4. In one embodiment, the antibody is of human subclass lgG1.
- the heavy chain constant region has gamma (y), mu (p), alpha (a), delta (5) and epsilon (E) types and is subclassified into gamma 1 (y1), gamma 2 (y2), gamma 3 (y3), gamma 4 (y4), alpha 1 (a1) and alpha 2 (a2).
- the constant region of the light chain has kappa (K) and lambda (A) types.
- the basic four-chain antibody unit is a heterotetramer glycoprotein consisting of two identical light chains (L) and two identical heavy chains (H).
- the light chain has a variable region (VL) at the N-terminus and a constant region at another terminus.
- the heavy chain has a variable region (VH) at the N-terminus and three constant regions (CH) for a and y chains, and four CH regions for p and E isoforms.
- VH variable region
- CH constant regions
- p and E CH regions
- the term “variable” means that a particular portion of the variable region is significantly different in sequence between antibodies.
- the V region mediates antigen binding and defines the specificity of a particular antibody for a particular antigen thereof.
- Variability is concentrated in three segments called “hypervariable region (HVR)”, that is, CDR, in both light- and heavy-chain variable regions.
- HVR hypervariable region
- the more highly conserved portion of the variable region is called a “framework region (FR)”.
- the heavy and light-chain variable regions have FR1, CDR1 , FR2, CDR2, FR3, CDR3 and FR4 structures from the N-terminus to the C-terminus.
- heavy chain encompasses both a full-length heavy chain, which comprises a variable domain (VH) comprising an amino acid sequence having a sufficient variable region sequence for imparting specificity to an antigen and three constant domains (CH1, CH2 and CH3), and a fragment thereof.
- VH variable domain
- CH1, CH2 and CH3 constant domains
- light chain encompasses both a full-length light chain, which comprises a variable domain (VL) comprising an amino acid sequence having a sufficient variable region sequence for imparting specificity to an antigen and a constant domain (CL), and a fragment thereof.
- VL variable domain
- CL constant domain
- ANGPT2 is also known to act as an agonist to induce activation of the TIE2 receptor in several processes, including lymphatic tube formation and maintenance, and thus it is believed that ANGPT2 performs various functions depending on the context. Pathological expression of ANGPT2 dysregulates these processes.
- the antibody or antigen-binding fragment : a. Decreases or inhibits human angiopoietin-2 (ANGPT2), and/or b. Induces TIE2 activation, and c. Induces TIE2 phosphorylation, in a fibrotic tissue.
- ANGPT2 human angiopoietin-2
- the induction of TIE2-activation is ANGPT2-dependent.
- the antibody or antigen-binding fragment thereof normalises pathological blood vessels and/or prevents the progression of fibrosis.
- a further advantage of the present invention is that the antibody described herein that binds to ANGPT2 and activates TIE2 is dependent on the presence of ANGPT2. Therefore, TIE2 is only activated in locations with elevated ANGPT2. So, the antibody described herein will only function to activate TIE2 in diseased and/or injured tissues, thereby leading to a reduction in off-target effects.
- ANGPT2-TIE2 signalling plays a role in various pathophysiological processes associated with fibrosis.
- ANGPT2-TIE2 signalling plays a role in various renal pathophysiological processes.
- the antibody or antigen-binding fragment thereof prevents or reduces a reduction in perfusion, for example kidney perfusion associated with fibrotic disease, for example kidney disease, disorder or injury.
- the antibody or antigen-binding fragment thereof prevents or reduces capillary rarefaction associated with fibrotic disease, for example kidney disease, disorder or injury.
- the prevention or reduction of capillary rarefaction is evaluated via quantification of endothelial markers, such as endomucin or podocalyxin.
- the antibody or antigen-binding fragment thereof prevents or reduces a reduction of fenestrations of peritubular capillaries associated with the kidney disease, disorder or injury.
- the antibody or antigen-binding fragment thereof prevents or reduces fibrosis, for example fibrosis associated with the kidney disease, disorder or injury.
- the fibrosis is tubulointerstitial fibrosis and/or glomerulosclerosis.
- the prevention or reduction of fibrosis is evaluated via quantification of fibrotic markers, such as a-Smooth muscle actin (aSMA), vimentin, Type I collagen (Coll), or platelet-derived growth factor receptor-p (PDGFR-P).
- aSMA smooth muscle actin
- Coll Type I collagen
- PDGFR-P platelet-derived growth factor receptor-p
- the antibody or antigen-binding fragment thereof for use as described herein, wherein the kidney disease, disorder or injury is induced by, propagated by and/or associated with increased extracellular matrix such as a-Smooth muscle actin (aSMA), vimentin, Type I collagen (Coll), or platelet-derived growth factor receptor-p (PDGFR-P).
- aSMA smooth muscle actin
- Coll Type I collagen
- PDGFR-P platelet-derived growth factor receptor-p
- the term "pharmaceutical composition” refers to the combination of an active agent ⁇ e.g., a composition of the present invention) with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
- the term "pharmaceutical composition” can be a formulation containing a composition of the present invention and/or an agent for use in the treatment or prevention of a fibrotic disease, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis, in a form suitable for administration to a subject.
- the present invention relates to a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof as an active ingredient.
- the pharmaceutical composition is characterised by containing a pharmaceutically effective amount of the antibody or an antigen-binding fragment thereof according to the invention and a pharmaceutically acceptable carrier and/or excipient.
- the present invention relates to a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof according to any of the preceding items, for use in the treatment or prevention of a fibrotic disease.
- the disease is kidney disease, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- the pharmaceutical composition may further include a small molecule inhibitor used in chemotherapy or a vascular endothelial growth factor (VEGF) antagonist.
- VEGF vascular endothelial growth factor
- the VEGF antagonist may be an anti-VEGF antibody, a VEGF inhibiting fusion protein, or a small molecule kinase inhibitor. It also may prove effective, in particular, to combine antibodies or antigen-binding fragments thereof of the present disclosure with other therapies that target different aspects of ANGPT2 function.
- the antibodies or antigen-binding fragments thereof of the present disclosure may be linked to at least one agent to form an antibody conjugate in order to increase the efficacy of antibody molecules as diagnostic or therapeutic agents.
- the present invention includes antibodies and antigen-binding fragments thereof, compositions and methods for use in treatment or prevention of a fibrotic disease in a subject.
- the present invention includes antibodies and antigen-binding fragment thereof, compositions and methods for use in treatment or prevention of a kidney disease, disorder or injury in a subject.
- Administration of the antibodies and antigenbinding fragment thereof and/or compositions according to the present invention will typically be via any common route. This includes, but is not limited to parenteral, orthotopic, intradermal, subcutaneous, intramuscular, intraperitoneal, intranasal, or intravenous injection. Additional formulations which are suitable for other modes of administration include oral formulations.
- Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained-release formulations, or powders and contain about 10% to about 95% of active ingredient, preferably about 25% to about 70%.
- compositions of the invention are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective.
- the quantity to be administered depends on the subject to be treated. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner.
- the manner of application may be varied widely. Any of the conventional methods for administration of an antibody are applicable. These are believed to include oral application on a solid physiologically acceptable base or in a physiologically acceptable dispersion, parenterally, by injection, and the like.
- the dosage of the pharmaceutical composition will depend on the route of administration and will vary according to the size and health of the subject.
- the course of the administrations may be followed by assays for organ function, for example kidney function and/or the assessment of a fibrotic disease, disorder or injury or status thereof, for example kidney disease, disorder or injury or status thereof, including but not limited to measurements of relevant biomarkers, tissue capillary density, and structural integrity of affected tissues, as well as evaluations of associated complications such as cardiomyopathy, coagulopathy, and retinopathy.
- phrases “pharmaceutically acceptable” or “pharmacologically acceptable” refer to molecular entities and compositions that do not produce an adverse, allergic, or other untoward reaction when administered to an animal, or human.
- pharmaceutically acceptable excipient includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well-known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredients, its use in therapeutic compositions is contemplated.
- compositions may be formulated into a neutral or salt form.
- Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine, and the like.
- the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
- polyol for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like
- suitable mixtures thereof and vegetable oils.
- the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
- unit dose refers to physically discrete units suitable for use in a subject, each unit containing a predetermined quantity of the composition calculated to produce the desired responses discussed above in association with its administration, i.e. , the appropriate route and regimen.
- the quantity to be administered depends on the result and/or protection desired. Precise amounts of the composition also depend on the judgment of the practitioner and are peculiar to each individual. Factors affecting dose include the physical and clinical state of the subject, route of administration, the intended goal of treatment (alleviation of symptoms versus cure), and potency, stability, and toxicity of the particular composition.
- solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically or prophylactically effective. The formulations are easily administered in a variety of dosage forms.
- the antibody or antigen-binding fragment thereof is administered before the onset of the fibrotic disease, disorder or injury. In one embodiment, the antibody or antigen-binding fragment thereof is administered at the onset of fibrotic disease, disorder or injury. In one embodiment, the antibody or antigen-binding fragment thereof is administered after the onset of the fibrotic disease, disorder or injury.
- the antibody or antigen-binding fragment thereof is administered before the onset of the kidney disease, disorder or injury. In one embodiment, the antibody or antigen-binding fragment thereof is administered at the onset of the kidney disease, disorder or injury. In one embodiment, the antibody or antigen-binding fragment thereof is administered after the onset of the kidney disease, disorder or injury. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with endothelial dysfunction. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with kidney fibrosis.
- the fibrotic disease is selected from the following: kidney fibrosis and myocardial fibrosis.
- the kidney fibrosis is tubulointerstitial fibrosis. In some embodiments, the kidney fibrosis is glomerulosclerosis. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with renal capillary rarefaction. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with tubular atrophy. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with tubular injury. In one embodiment, the kidney disease, disorder or injury is induced by, propagated by and/or associated with oxidative stress.
- the kidney disease, disorder or injury is acute kidney injury (AKI). In some embodiments, the kidney disease, disorder or injury is acute kidney disease (AKD). In some embodiments, the kidney disease, disorder or injury is chronic kidney disease (CKD). In some embodiments, the CKD is induced by, propagated by and/or associated with coagulopathy.
- Kidney disease can be divided into acute kidney injury (AKI) and chronic kidney disease (CKD).
- AKI is a sudden and typically reversible decline in kidney function that occurs over hours to days. It is often triggered by conditions such as dehydration, infections (sepsis), drug toxicity, or obstruction in the urinary tract.
- AKI is marked by a rapid increase in blood urea nitrogen (BUN) and creatinine levels and may present symptoms like reduced urine output, swelling, and fatigue. Sepsis is the leading cause of AKI. Early intervention can often restore normal kidney function.
- BUN blood urea nitrogen
- Sepsis is the leading cause of AKI.
- Early intervention can often restore normal kidney function.
- CKD Chronic Kidney Disease
- GFR glomerular filtration rate
- CKD is associated with tubulointerstitial fibrosis, which is not seen in AKI.
- the kidney disease is CKD associated with tubulointerstitial fibrosis.
- the kidney disease is CKD associated with glomerulosclerosis.
- AKD is sudden and potentially reversible
- CKD is gradual and permanent. Both conditions require prompt medical attention but differ in their management, outcomes, and implications for long-term health.
- ABTAA has shown protective effects in two preclinical models of chronic kidney disease, one model with fibrosis as a clinical endpoint and one model of diabetes type 2 with complications in the kidney and heart. That ABTAA has an effect on fibrosis was new and unexpected.
- Chronic kidney disease encompasses several clinical kidney diseases all associated with a fibrosis/connective tissue component (fibrosis) in either the tubulointerstitium or glomeruli from the European Renal Association (Venkat-Raman et al. Nephrol Dial Transpl, 2012).
- the kidney disease, disorder or injury is selected from the group comprising: chronic renal impairment, chronic renal failure syndrome, glomerulonephritis, type I diabetes mellitus, type II diabetes mellitus, ischaemic nephropathy, focal segmental glomerulosclerosis, nephrotic syndrome, cardiorenal syndrome, renal tubulo-interstitial disorders associated with metabolic disease, minimal change disease, hypertensive renal disease, malignant hypertensive renal disease, IgA nephropathy, IgM nephropathy, mesangiocapillary glomerulonephritis, crescentic glomerulonephritis, diffuse endocapillary proliferative glomerulonephritis, mesangial proliferative glomerulonephritis, focal and segmental proliferative glomerulonephritis, systemic vasculitis, goodpasture's syndrome, dent's disease, systemic lupus erythematosus glomer
- the kidney fibrosis is induced by, propagated by and/or associated with chronic renal impairment, chronic renal failure syndrome, glomerulonephritis, type I diabetes mellitus, type II diabetes mellitus, ischaemic nephropathy, focal segmental glomerulosclerosis, nephrotic syndrome, cardiorenal syndrome, renal tubulo-interstitial disorders associated with metabolic disease, minimal change disease, hypertensive renal disease, malignant hypertensive renal disease, IgA nephropathy, IgM nephropathy, mesangiocapillary glomerulonephritis, crescentic glomerulonephritis, diffuse endocapillary proliferative glomerulonephritis, mesangial proliferative glomerulonephritis, focal and segmental proliferative glomerulonephritis, systemic vasculitis, goodpasture's syndrome, dent's disease, systemic lupus erythemato
- fibrotic disease for example kidney disease, disorder or injury is induced by, propagated by and/or associated with type-2 diabetes.
- the subject suffers from type-2 diabetes.
- the fibrotic disease is a caused by type-2 diabetes.
- ABTAA treatment does not affect the obesity and diabetic status of the subject, suggesting its potential for combination therapy with other drugs targeting obesity and blood glucose levels.
- the invention provides an antibody or antigen-binding fragment thereof, that binds to ANGPT2 and binds to TIE2 receptor via ANGPT2, for use in the treatment or prevention of diabetic complications.
- the fibrotic disease is myocardial fibrosis.
- the myocardial fibrosis is induced by, propagated by and/or associated with myocardial infarction, hypertensive heart disease, or cardiomyopathy.
- the fibrotic disease is cardiomyopathy.
- the cardiomyopathy is hypertrophic cardiomyopathy.
- the cardiomyopathy is idiopathic dilated cardiomyopathy.
- the invention provides an antibody or antigen-binding fragment thereof, that binds to ANGPT2 and binds to TIE2 receptor via ANGPT2, for use in the treatment or prevention of coagulopathy.
- the coagulopathy is caused by or associated with a fibrotic disease.
- the methods disclosed in the section herein, entitled ‘indications/treatment regime’ may further comprise administering to the subject in need thereof the pharmaceutical compositions disclosed in the section ‘pharmaceutical compositions’ herein.
- treatment or prevention not only comprises preventing and/or treating the disease, but also generally comprises preventing the onset of the disease, slowing or reversing the progress of disease, preventing or slowing the onset of one or more symptoms associated with the disease, reducing and/or alleviating one or more symptoms associated with the disease, reducing the severity and/or the duration of the disease and/or of any symptoms associated therewith and/or preventing a further increase in the severity of the disease and/or of any symptoms associated therewith, preventing, reducing or reversing any physiological damage caused by the disease, and generally any pharmacological action that is beneficial to the patient being treated.
- the subject to be treated may be any warm-blooded animal but is, in particular, a mammal, and more in particular a human being.
- the subject to be treated will, in particular, be a person suffering from, or at risk of, the diseases and disorders mentioned herein.
- the present invention relates to methods of treating fibrotic diseases, disorder or injury, comprising administering a therapeutically effective amount of the antibody or antigen-binding fragments.
- the present invention relates to methods of treating a kidney disease, disorder or injury, wherein the kidney disease, disorder or injury is associated with kidney fibrosis, comprising administering a therapeutically effective amount of the antibody or antigen-binding fragments.
- the present invention relates to the manufacture of a medicament for treatment of fibrotic diseases.
- the present invention relates to the manufacture of a medicament for treatment of a kidney disease, disorder or injury, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- the “therapeutically effective amount” means an amount sufficient to treat diseases at a reasonable benefit/risk ratio applicable for medical treatment, and an amount of a composition comprising the anti-ANGPT2 antibody.
- the exact amount may vary depending on a number of factors that include components and physical characteristics of a therapeutic composition, intended patient population, individual patient considerations, etc., but are not limited thereto, and may be easily determined by those skilled in the art. When completely considering these factors, it is important to administer the minimum amount sufficient to obtain the maximum effect without the side effect, and this dosage may be easily determined by an expert in the field.
- the dosage of the pharmaceutical composition of the present invention is not specifically limited but is changed according to various factors including a health state and weight, severity of the disease of a patient, and a drug type, an administration route, and administration time.
- the composition may be administered in routes that are typically allowed in mammals including rat, mouse, cattle, human, etc., for example, orally, rectally, intravenously, subcutaneously, intrarenally, intrauterinely or intracerebrovascularly in a single dose amount per day or every other day or every week or every other week.
- the present invention is directed to methods of: a) decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in a fibrotic tissue, for example, diseased kidney, wherein the diseased kidney is fibrotic, b) normalising pathological blood vessels and treating and reducing and preventing the progression of fibrosis in a tissue, for example a diseased kidney, c) treating and/or reducing and/or preventing the progression of fibrosis in a tissue, for example a diseased kidney, d) preventing or treating a reduction in perfusion in a subject, for example wherein the perfusion is kidney perfusion wherein the disease is kidney disease, disorder or injury is associated with kidney fibrosis, e) preventing or reducing capillary rarefaction associated with fibrotic diseases, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney
- the present invention is directed to methods of: a) decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in fibrotic tissue, for example a diseased kidney, wherein the diseased kidney is fibrotic, b) normalising pathological blood vessels and treating and reducing and preventing the progression of fibrosis in a tissue, for example diseased kidney, c) treating and/or reducing and/or preventing the progression of fibrosis in a tissue, for example a diseased kidney, d) preventing or treating a reduction in perfusion in a subject, for example wherein the perfusion is kidney perfusion wherein the disease is kidney disease, disorder or injury is associated with kidney fibrosis, e) preventing or reducing capillary rarefaction associated with the fibrotic disease, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis,
- the induction of TIE2-activation is ANGPT2-dependent.
- the prevention or reduction of capillary rarefaction is evaluated via quantification of endothelial markers, such as endomucin or podocalyxin.
- the fibrosis is tubulointerstitial fibrosis.
- the fibrosis is glomerulosclerosis.
- the prevention or reduction of fibrosis is evaluated via quantification of fibrotic markers, such as a-Smooth muscle actin (aSMA), vimentin, Type I collagen (Coll), or platelet-derived growth factor receptor-p (PDGFR-P).
- the present invention relates to a nucleic acid(s) encoding the antibodies or antigen-binding fragments thereof.
- the nucleic acid(s) may be present in a cell, a cell lysate, or may also be present in a partially purified form or a substantially pure form.
- the nucleic acid(s) is “isolated” or “is substantially pure” when it is purified from other cell components or other contaminants, for example, other cell nucleic acid or protein by standard techniques including alkaline/SDS treatment, CsCI banding, column chromatography, agarose gel electrophoresis, and other techniques well-known in the art.
- the nucleic acid(s) of the present invention may be, for example, DNA or RNA, and may include an intron sequence, or may not include the intron sequence.
- the present invention relates to a recombinant expression vector including the nucleic acid.
- DNA encoding the light chain and the heavy chain having a partial length or a full length may be obtained by standard molecular biology techniques (for example, PCR amplification or cDNA cloning using a hybridoma that expresses a target antibody), and the DNA may be “operably bound” to transcription and translation control sequences to be inserted into the expression vector.
- operably bound used herein may indicate that an antibody gene is ligated into the vector so that the transcription and translation control sequences in the vector have an intended function to control transcription and translation of the antibody gene.
- the expression vector and an expression control sequence are selected so as to have compatibility with a host cell for expression to be used.
- the light chain gene of the antibody and the heavy chain gene of the antibody are inserted into a separate vector, or both genes are inserted into the same expression vector.
- the antibody is inserted into the expression vector by a standard method (for example, ligation of an antibody gene fragment and a complementary restriction enzyme site on a vector or when the restriction enzyme site is not present at all, blunt end ligation).
- the recombinant expression vector may encode a signal peptide that facilitates secretion of the antibody chain from the host cell.
- the antibody chain gene may be cloned into the vector so that the signal peptide is bound to an amino terminal of the antibody chain genes according to a frame.
- the signal peptide may be an immunoglobulin signal peptide or a heterologous signal peptide (/.e. signal peptide derived from proteins except for immunoglobulin).
- the recombinant expression vector has a regulatory sequence that controls the expression of the antibody chain genes in the host cell.
- the “regulatory sequence” may include a promoter, an enhancer and other expression control element (for example, polyadenylation signal) controlling the transcription or translation of the antibody chain gene.
- a promoter for example, an enhancer and other expression control element controlling the transcription or translation of the antibody chain gene.
- expression control element for example, polyadenylation signal
- design of the expression vector may vary by changing the regulatory sequences according to factors such as selection of the host cell to be transformed, an expression level of the protein, etc.
- the present invention relates to a cell transformed with the recombinant expression vector.
- the cell used to produce the antibodies or antigenbinding fragments thereof of the present disclosure may be a prokaryote, yeast or higher eukaryotic cell, but is not limited thereto.
- strains of the genus Bacillus such as Escherichia coli, Bacillus subtilis and Bacillus tuligensis, Streptomyces, Pseudomonas, and prokaryotic host cells such as Proteus mirabilis and Staphylococcus can be used.
- animal cells can be used. Examples include, but are not limited to, COS-7, BHK, CHO, CHOK1 , DXB-11, DG-44, CHO/-DHFR, CV1, COS-7, HEK293, BHK, TM4, VERO, HELA, MDCK, BRL 3A, W138, Hep G2, SK-Hep, MMT, TRI, MRC 5, FS4, 3T3, RIN, A549, PC12, K562, PER.C6, SP2/0, NS-0, U20S, or HT1080.
- COS-7 COS-7, BHK, CHO, CHOK1 , DXB-11, DG-44, CHO/-DHFR
- the nucleic acid or the vector is transfected into the host cell.
- various kinds of generally used techniques such as electrophoresis, calcium phosphate precipitation, DEAE-dextran transfection, lipofection, etc., may be used to introduce an exogenous nucleic acid (DNA or RNA) into a prokaryotic host cell or a eukaryotic host cell.
- the antibody according to the present invention may be expressed in a eukaryotic cell, preferably, in a mammalian host cell, in consideration of applicability into a mammalian cell.
- the mammalian host cells suitable for expression of the antibody may include a Chinese hamster ovary (CHO) cell (for example, including a dhfr- CHO cell used together with a DHFR selection marker), an NSO myeloma cell, a COS cell, or a SP2 cell, etc., as examples.
- CHO Chinese hamster ovary
- the present invention relates to a method for producing the anti-ANGPT2 antibodies or antigen-binding fragments thereof, including culturing the host cells and expressing the antibody or antigen-binding fragment thereof.
- the antibody may be produced by culturing the host cell for a sufficient period of time so that the antibody is expressed in the host cell, or more preferably, for a sufficient period of time so that the antibody is secreted into a culture medium in which the host cell is cultured.
- the expressed antibody may be separated from the host cell and purified for uniformity.
- the separation or the purification of the antibody may be performed by a separation method, a purification method generally used for protein, for example, chromatography.
- the chromatography may include, for example, affinity chromatography, ion exchange chromatography or hydrophobic chromatography including protein A column and protein G column.
- the antibody may be separated and purified by additionally combining with filtration, ultrafiltration, salting out, dialysis, etc.
- mice Inducible mouse lines utilised in the present examples are summarised in Figure 1B. Experimental mice groups utilised in the present examples are summarised in Table 1.
- Floxed Veptp and TIE2 mice were crossed to tamoxifen inducible Cd/75-Cre ERT2 mice (Pitulescu et al., 2010) to generate endothelial cell specific knockout of the gene (77E2 iECKO and Veptp' ECKO , respectively).
- TIE2 mice were also crossed to a reporter mouse, Ai14-TdTomato (Gt(ROSA)26Sor tm14(CAG - tdTomat °’ Hze , JAX stock 007914) (Madisen et al., 2010), to get a lineage tag of endothelial cells.
- Littermate controls were mice with wt/wt alleles for TIE2, and Veptp (WT).
- Floxed Pdgfb mice (Pdgfb tm2Cbet ) (Enge et al., 2002) were crossed to tamoxifen inducible Acfb-Cre ERT2 mice (Tg(CAG- cre/Esr1*)5Amc, JAX stock 004682) (Hayashi et al., 2002) to generate whole body knockout of Pdgfb (Pdgfb' KO ).
- Littermate controls were Cre negative Pdgfb wt/lox or lox/lox mice.
- Knockout was induced with 3 doses of tamoxifen (2 mg) in peanut oil by oral gavage at 4 weeks of age (77E2 iECKO and Veptp' ECKO ), or 1 week prior to experiments (Pdgfb' KO ). Mice were on a mixed background. Mice were genotyped with primers described in Table 2. Mice were injected with ABTAA (25 mg/kg body weight in PBS) intraperitoneally at indicated time points in Figure 1A and 11 A. Control mice were injected with the same dose of human IgG Fc (AG714, Millipore) in PBS.
- mice for treatment with ABTAA and for early gene regulation analysis were C57BL6/J mice from inhouse breeding or WT mice from the above-mentioned breeding’s.
- Adult mice (8-16 weeks) were used for all experiments.
- CKD was induced by UUO as previously described (Loganathan et al., 2018; Chevalier et al., 2009). Comparisons were made between the unobstructed, contralateral (CL) kidney and the obstructed, UUO kidney. Both female and male mice were used in all experiments as the UUO model has not shown any gender differences (Yang et al., 2010).
- Table 2 Genotyping primers
- Renal perfusion measured with contrast imaging ultrasound Renal perfusion was measured in isoflurane anesthetized mice on a heated platform utilizing a Vevo 2100 ultrasound system with a MS250 transducer and contrast imaging functionality software (Visual Sonics, Fujifilm). Mice were kept at a body temperature of 36-37°C, continuously measured with an anal probe. Mice were imaged from the back and the ultrasound transducer was fixed in place with a mechanical positioning system. Regular B-mode images were used to optimally positioning the mice to enable imaging of both kidneys at the same time.
- mice were tail vein injected with 100 pl microbubble-based contrast agent (Vevo MicroMarker, VS-11913, Fujifilm Visual Sonics) from vials resuspended with 2 ml saline.
- Non-linear contrast images were acquired with General imaging in the Vascular package at 21 GHz. Images were analysed for contrast intensity with Vevo Lab 3.2.6 (Fujifilm Visual Sonics) after manual marking region of interest (ROI), cortex, of both CL and UUO kidneys. Perfusion was normalized to CL kidney for each mouse. Five sham operated WT mice were used in ultrasound graphs as comparison presented as the left kidney normalized to the right kidney.
- Immunohistochemistry was performed in well plates on floating 100 pm thick vibratome sections after protein block (X0909, DAKO) with 0.25% Triton X100 for 2 hours. Sections were incubated with primary antibodies overnight at 4°C on a shaker. Antibodies are described in Table 3. Appropriate secondary antibodies were added after washing with PBS containing 0.05% Tween-20 and incubated for 2 hours at room temperature. Nuclei were stained with Hoechst 33342 (Thermo Fisher Scientific). Table 3: Antibodies
- Kidney cortex was imaged (5 images/mouse) at original magnification x400 using a Leica SP8 confocal microscope.
- aSMA and vimentin positive staining was quantified in each image using Otzu thresholding in Imaged (NIH). Large arteries and glomeruli were excluded. Fibrotic area was expressed a percentage of the whole image area. Quantification of vascular area was done in the same way as above for endomucin and podocalyxin and correlated to the total number of nuclei per image. The number of nuclei were counted by utilizing Analyze Particles in Imaged (NIH) with selection of positive areas of 50-800 pixels.
- RNAeasy Micro or Minikit Qiagen was used to extract mRNA according to the manufacturer’s protocol, followed by cDNA synthesis of 1 pg mRNA using iScript reverse transcription supermix (170-8841 , BioRad).
- Real time PCR was performed using cDNA with Taqman Gene Expression master mix (4369016, Thermo Fisher Scientific) together with probes on a CFX-96 real time PCR system (BioRad). Probes are listed in Table 4. Expression results were normalized to endogenous control Hprt or Gapdh and relative quantification was done using the Livak method (2’ AACT ).
- kidneys were cut in 1 mm cubes, and immersion fixed in 2.5% glutaraldehyde (Ted Pella) and 1% paraformaldehyde (Merck) in 0.1 M phosphate buffer pH 7.4 and stored at 4°C until further processed. Samples were rinsed in 0.1 M phosphate buffer for 10 min followed by 1 h incubation in 1 % osmium tetroxide (TAAB Laboratories Equipment) in 0.1 phosphate buffer. After rinsing in phosphate buffer, samples were dehydrated in alcohol followed by 5 min incubation in propylene oxide (TAAB Laboratories Equipment).
- Sections were contrasted with 5% uranyl acetate and Reynold’s lead citrate and visualized in a Tecnai G2 Spirit BioTwin electron microscope (Thermo Fisher Scientific/FEI) at 80 kV with an Orius SC200 CCD camera and Gatan Digital Micrograph software (both from Gatan Inc.). Micrographs were taken of peritubular capillaries and the number of capillary fenestrations was semi- quantitatively graded and expressed as a score between 0-4, with 0: 0-5%, 1 :6-25%, 2:26-50%, 3:51-75%, and 4:76-100% of the capillary length having fenestrations (i.e, a score of 4 in healthy kidneys).
- a tubular segment was defined as affected when vacuoles were present in the whole epithelial cell, from the luminal to the basolateral side (Fig. 8A).
- Example 1 - TIE2-activation byABTAA improves renal perfusion and protects from capillary rarefaction following UUO.
- the ABTAA antibody is as defined by SEQ ID NOs 33 and 34 described herein.
- Renal perfusion was evaluated as above-mentioned, a contrast imaging ultrasound after intravenous injection of a circulating vascular marker (MicroMarker). Capillary density in renal cortex was evaluated with staining for the endothelial markers endomucin and podocalyxin (excluding glomeruli).
- ABTAA significantly rescues renal perfusion and capillary rarefaction in an in vivo model of kidney disease.
- Fenestrations of peritubular capillaries are vital structures for reabsorption of water and solutes from the tubular system. Fenestrations are reduced in CKD and can be used to estimate capillary dysfunction (Babickova et al., 2000). Fenestrations of peritubular capillaries were semi-quantitatively analysed with transmission electron microscopy in T/E2 iECKO , ⁇ /eptp iECKO , and ABTAA treated groups in 3-day UUO and CL kidneys.
- ABTAA significantly rescues renal endothelial injury in an in vivo model of kidney disease.
- Example 3 TIE2-activation by ABTAA reduces UUO-induced tubulointerstitial fibrosis.
- perivascular mesenchymal cells to myofibroblasts is characterized by expression of alpha-smooth muscle actin (aSMA) and production of extracellular matrix components including type I collagens, fibronectin, and vimentin.
- aSMA alpha-smooth muscle actin
- Tubulointerstitial fibrosis was quantified in renal cortex by staining for aSMA and vimentin (glomeruli excluded).
- UUO-induced tubulointerstitial fibrosis were significantly reduced in ABTAA-treated and Veptp' ECKO mice compared to IgG injected and WT mice, respectively (Fig. 7A-C).
- the reduction in fibrosis was also reflected in decreased UUO-induced Col1a1 and Pdgfrb expression in ABTAA treated and Veptp' ECKO mice, respectively (Fig. 7D, E).
- Pdgfb' KO mice had significantly reduced tubulointerstitial fibrosis, supporting the notion of PDGFB as a major mesenchymal activator.
- 77E2 iECKO mice had aggravated disease and significantly more tubulointerstitial fibrosis in 3-day UUO kidneys as well as significantly increased expression for Col1a1, Tagln, and Fn1 (Fig. 7F).
- ABTAA significantly reduces renal fibrosis in an in vivo model of kidney disease.
- Example 4 - TIE2-activation by ABTAA reduces UUO-induced tubular injury and PDGFB expression.
- ABTAA significantly reduces tubular injury in an in vivo model of kidney disease.
- Example 5 Post injury treatment with ABTAA reduces UUO-induced injury.
- Late-onset ABTAA treatment was initiated 3 days after UUO and 10-day UUO kidneys were analysed (Fig. 10A). Fibrosis was evaluated via aSMA and vimentin staining, and tubular injury was evaluated via PDGFB expression.
- ABTAA significantly reduces tubulointerstitial fibrosis and tubular injury in an in vivo model of kidney disease.
- Example 6 - ABTAA therapy is protective in early diabetic complications.
- BTBRob/ob also referred to ob/ob
- ob/ob mouse model
- This model has significant loss of capillary density in the kidney already at 6-weeks of age (Zhou et al., 2024).
- the BTBRob/ob model also develops cardiomyopathy (Ye et al., 2017) and retinopathy (Lee et al., 2018) making it an ideal model to investigate therapeutic interventions in diabetic complications.
- the inventors confirmed an upregulation of ANGPT2 in renal tissue of ob/ob mice as this is a requirement for ABTAA mode of action (Fig. 11 A). Non-diabetic heterozygous mice served as controls.
- mice were treated with once weekly subcutaneous injections of either 25 mg/kg ABTAA or human IgG until they reached 11 weeks of age.
- Various measurements were taken, including plasma blood urea nitrogen levels, capillary density in the renal cortex (using endomucin staining of renal cortex), fenestrations in peritubular capillaries (visualized via electron microscopy), plasma creatinine kinase levels.
- Retinopathy was assessed by staining for capillaries with Pecaml and for the basement membrane with Collagen IV, followed by quantifying the number of empty collagen sleeves left behind when capillaries disappear.
- ABTAA treatment normalized blood urea nitrogen levels in ob/ob mice, indicating protection against nephropathy (Fig. 11C).
- Capillary density in the renal cortex was preserved in ABTAA-treated mice compared to controls (Fig. 11D). Additionally, ABTAA maintained normal fenestrations in peritubular capillaries, suggesting improved kidney function (Fig. 11 E).
- ABTAA prevented the increase in plasma creatinine kinase levels, indicating protection against cardiac muscle injury (Fig. 11 F).
- Fig. 11 G For retinopathy, ABTAA reduced the number of empty collagen sleeves in retinal capillaries, demonstrating its protective effect (Fig. 11 G).
- ABTAA therapy effectively protects against early diabetic complications in the BTBRob/ob mouse model, preserving kidney function, capillary integrity, and preventing cardiac muscle injury.
- Example 7 - ABTAA therapy is protective in coagulopathy.
- ANGPT2 can inhibit thrombomodulin (TM) mediated activation of protein C (Hultstrom et al., 2022), suggesting additional roles for ANGPT2 in endothelial dysfunction and coagulopathy, which is also seen in diabetic patients.
- TM thrombomodulin
- the inventors developed a coagulopathy model. Mice were injected with the inflammatory cytokine TNFa to trigger endogenous ANGPT2 release (Fig. 12A). The coagulation capacity was then evaluated by measuring tail bleeding time.
- BTBR ob/ob mouse model of type 2 diabetes exhibits early loss of retinal function and retinal inflammation followed by late vascular changes. Diabetologia. 2018;61(11):2422-2432
- An antibody or antigen-binding fragment thereof that binds to ANGPT2 and binds to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a fibrotic disease.
- An antibody or antigen-binding fragment thereof that binds to ANGPT2 and binds to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- An antibody or antigen-binding fragment thereof that binds to ANGPT2 and binds to TIE2 receptor via ANGPT2, for use in the treatment or prevention of a coagulopathy.
- binding site is about 2 to about 20 contiguous amino acids in length, such about 2 to about 18, about 5 to about 20, or about 10 to about 20, such as about 15 to about 20.
- said antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ I D NO: 1 , SEQ I D NO: 24 or SEQ I D NO: 35, the LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 25
- antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of S
- the antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein the antibody or antigen-binding fragment thereof comprises a variable heavy chain of SEQ ID NO: 8 or SEQ ID NO: 10 or SEQ ID NO: 31 or SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 7 or SEQ ID NO: 11 or SEQ ID NO: 32 or SEQ ID NO: 34 or SEQ ID NO: 38.
- An antibody or antigen-binding fragment thereof for use in the treatment or prevention of a kidney disease, disorder or injury in a subject, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- An antibody or antigen-binding fragment thereof for use in the treatment or prevention of a fibrotic disease, disorder or injury in a subject, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3. 13.
- CDRs complementarity determining regions
- the antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein the antibody or antigen-binding fragment thereof comprises a variable heavy chain of SEQ ID NO: 8 or SEQ ID NO: 10, and a variable light chain of SEQ ID NO: 7 or SEQ ID NO: 11.
- antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of SEQ ID NO: 25 and the LCDR3 amino acid sequence of SEQ ID NO: 26, or the LCDR1 amino acid sequence of SEQ ID NO: 35, the LCDR2 amino acid sequence of SEQ ID NO: 36 and the LCDR3 amino
- the antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein the antibody or antigen-binding fragment thereof comprises a variable heavy chain of SEQ ID NO: 31 or SEQ ID NO: 33, and a variable light chain of SEQ ID NO: 32 or SEQ ID NO: 34 or SEQ ID NO: 38.
- the antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein the antibody or antigen-binding fragment thereof: a) Decreases or inhibits human angiopoietin-2 (ANGPT2), and/or b) Induces TIE2 activation, and c) Induces TIE2 phosphorylation.
- ANGPT2 human angiopoietin-2
- the antibody or antigen-binding fragment thereof for use according to any of the preceding items, wherein the antibody or antigen-binding fragment thereof: a) Decreases or inhibits human angiopoietin-2 (ANGPT2), and/or b) Induces TIE2 activation, and c) Induces TIE2 phosphorylation, in a fibrotic and/or diseased tissue, for example diseased kidney.
- ANGPT2 human angiopoietin-2
- fibrosis is evaluated via quantification of fibrotic markers, such as a-Smooth muscle actin (aSMA), vimentin, Type I collagen (Coll), or platelet-derived growth factor receptor-p (PDGFR-P), optionally wherein the fibrosis is kidney fibrosis.
- aSMA smooth muscle actin
- Coll Type I collagen
- PDGFR-P platelet-derived growth factor receptor-p
- a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof according to any of the preceding items, for use in the treatment or prevention of kidney disease, wherein the kidney disease, disorder or injury is associated with kidney fibrosis.
- a pharmaceutical composition comprising the antibody or antigen-binding fragment thereof according to any of the preceding items, for use in the treatment or prevention of fibrotic disease.
- fibrotic disease for example kidney disease, disorder or injury is induced by, propagated by and/or associated with oxidative stress.
- fibrotic disease for example kidney disease, disorder or injury is induced by, propagated by and/or associated with hypoxia.
- kidney disease, disorder or injury is chronic kidney disease (CKD).
- kidney disease, disorder or injury is selected from the group comprising: chronic renal impairment, chronic renal failure syndrome, glomerulonephritis, type I diabetes mellitus, type II diabetes mellitus, ischaemic nephropathy, focal segmental glomerulosclerosis, nephrotic syndrome, cardiorenal syndrome, renal tubulo-interstitial disorders associated with metabolic disease, minimal change disease, hypertensive renal disease, malignant hypertensive renal disease, IgA nephropathy, IgM nephropathy, mesangiocapillary glomerulonephritis, crescentic glomerulonephritis, diffuse endocapillary proliferative glomerulonephritis, mesangial proliferative glomerulonephritis, focal and segmental proliferative glomerulonephritis, systemic vasculitis, goodpasture's syndrome,
- kidney fibrosis is induced by, propagated by and/or associated with chronic renal impairment, chronic renal failure syndrome, glomerulonephritis, type I diabetes mellitus, type II diabetes mellitus, ischaemic nephropathy, focal segmental glomerulosclerosis, nephrotic syndrome, cardiorenal syndrome, renal tubulo-interstitial disorders associated with metabolic disease, minimal change disease, hypertensive renal disease, malignant hypertensive renal disease, IgA nephropathy, IgM nephropathy, mesangiocapillary glomerulonephritis, crescentic glomerulonephritis, diffuse endocapillary proliferative glomerulonephritis, mesangial proliferative glomerulonephritis, focal and segmental proliferative glomerulonephritis, systemic vasculitis, goodpasture's
- a method of treating a kidney disease, disorder or injury, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering a therapeutically effective amount of the antibody or antigen-binding fragment thereof according to any one of the preceding items or the pharmaceutical composition according to any one of the preceding items.
- a method of treating a fibrotic disease comprising administering a therapeutically effective amount of the antibody or antigen-binding fragment thereof according to any one of the preceding items or the pharmaceutical composition according to any one of the preceding items.
- CDRs complementarity determining regions
- a method of decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in fibrotic tissue, and/or diseased tissue, for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 12, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3, wherein the diseased kidney is fibrotic.
- CDRs complementarity
- a method of decreasing or inhibiting human angiopoietin-2 (ANGPT2) signalling, and/or inducing TIE2 activation, and/or inducing TIE2 phosphorylation in fibrotic tissue, and/or diseased tissue, for example diseased kidney, comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ
- a method of normalising pathological blood vessels and treating and reducing and preventing the progression of fibrosis in fibrotic and/or diseased tissue for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- a method of normalising pathological blood vessels and treating and reducing and preventing the progression of fibrosis in fibrotic and/or diseased tissue for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of SEQ ID NO: 25 and the LCDR3 amino acid sequence of SEQ
- a method of treating and/or reducing and/or preventing the progression of fibrosis in fibrotic and/or diseased tissue for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ I D NO: 1 , SEQ I D NO: 24 or
- a method of treating and/or reducing and/or preventing the progression of fibrosis in fibrotic and/or diseased tissue for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- a method of treating and/or reducing and/or preventing the progression of fibrosis in fibrotic and/or diseased tissue for example diseased kidney comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of SEQ ID NO: 25 and the LCDR3 amino acid sequence of SEQ ID NO:
- aSMA smooth muscle actin
- Coll Type I collagen
- PDGFR-P platelet-derived growth factor receptor-p
- a method of preventing or treating a reduction in perfusion in a subject for example kidney perfusion, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ I D NO: 1 , SEQ I D
- a method of preventing or treating a reduction in perfusion in a subject for example kidney perfusion, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a. complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b. CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- a method of preventing or treating a reduction in perfusion in a subject for example kidney perfusion, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of SEQ ID NO: 25 and the LCDR3 amino acid
- a method of preventing or reducing capillary rarefaction associated with diseased and/or fibrotic tissue, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 29, and b) complementarity determining regions (CDRs) of a light chain variable region comprising the LCDR1 comprising an amino acid sequence selected from
- a method of preventing or reducing capillary rarefaction associated with diseased and/or fibrotic tissue, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- a method of preventing or reducing capillary rarefaction associated with diseased and/or fibrotic tissue, for example kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence of
- a method of preventing or treating a reduction of fenestrations of peritubular capillaries associated with a kidney disease, disorder or injury in a subject, wherein a kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 27, the HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 28, or SEQ ID NO: 37, and the HCDR3 comprising an amino acid sequence selected from the
- a method of preventing or treating a reduction of fenestrations of peritubular capillaries associated with a kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of SEQ ID NO: 4, the HCDR2 amino acid sequence of SEQ ID NO: 5 and the HCDR3 amino acid sequence of SEQ ID NO: 6; and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 1 , the LCDR2 amino acid sequence of SEQ ID NO: 2 and the LCDR3 amino acid sequence of SEQ ID NO: 3.
- CDRs complementarity determining regions
- a method of preventing or treating a reduction of fenestrations of peritubular capillaries associated with a kidney disease, disorder or injury in a subject, wherein the kidney disease, disorder or injury is associated with kidney fibrosis comprising administering an antibody or antigen-binding fragment thereof to a subject in need thereof, wherein said antibody or antigen-binding fragment thereof comprises: a) complementarity determining regions (CDRs) of a heavy chain variable region comprising the HCDR1 amino acid sequence of HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 28 and the HCDR3 amino acid sequence of SEQ ID NO: 29, or the HCDR1 amino acid sequence of SEQ ID NO: 27, the HCDR2 amino acid sequence of SEQ ID NO: 37 and the HCDR3 amino acid sequence of SEQ ID NO: 29, and b) CDRs of a light chain variable region comprising the LCDR1 amino acid sequence of SEQ ID NO: 24, the LCDR2 amino acid sequence
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Urology & Nephrology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
Abstract
L'invention concerne des anticorps ciblant ANGPT2 destinés à être utilisés dans le traitement ou la prévention d'une maladie fibrotique, en particulier d'une ou plusieurs maladies rénales, d'un ou de plusieurs troubles ou lésions chez un sujet, la/le ou les maladies rénales, troubles ou lésion étant associés à la fibrose rénale.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP24161836 | 2024-03-06 | ||
| EP24161836.2 | 2024-03-06 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2025186364A1 true WO2025186364A1 (fr) | 2025-09-12 |
| WO2025186364A8 WO2025186364A8 (fr) | 2025-10-02 |
Family
ID=90362990
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2025/056090 Pending WO2025186364A1 (fr) | 2024-03-06 | 2025-03-06 | Anticorps ciblant angpt2 destinés à être utilisés dans le traitement de maladies fibrotiques, en particulier d'une ou de plusieurs maladies rénales, d'un ou plusieurs troubles ou d'une lésion associés à une fibrose rénale |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025186364A1 (fr) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7658924B2 (en) | 2001-10-11 | 2010-02-09 | Amgen Inc. | Angiopoietin-2 specific binding agents |
| US8987420B2 (en) | 2009-07-29 | 2015-03-24 | Regeneron Pharmaceuticals, Inc. | High affinity human antibodies to human angiopoietin-2 |
| US20150337033A1 (en) | 2014-05-26 | 2015-11-26 | Samsung Electronics Co., Ltd. | Humanized or affinity-matured anti ang-2 antibody and uses thereof |
| US11498962B2 (en) | 2018-02-20 | 2022-11-15 | Institute For Basic Science | Anti-angiopoietin-2 antibodies that induce Tie2 activation |
-
2025
- 2025-03-06 WO PCT/EP2025/056090 patent/WO2025186364A1/fr active Pending
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7658924B2 (en) | 2001-10-11 | 2010-02-09 | Amgen Inc. | Angiopoietin-2 specific binding agents |
| US8987420B2 (en) | 2009-07-29 | 2015-03-24 | Regeneron Pharmaceuticals, Inc. | High affinity human antibodies to human angiopoietin-2 |
| US20150337033A1 (en) | 2014-05-26 | 2015-11-26 | Samsung Electronics Co., Ltd. | Humanized or affinity-matured anti ang-2 antibody and uses thereof |
| US9994632B2 (en) | 2014-05-26 | 2018-06-12 | Samsung Electronics Co., Ltd. | Humanized or affinity-matured anti Ang-2 antibody and uses thereof |
| US11498962B2 (en) | 2018-02-20 | 2022-11-15 | Institute For Basic Science | Anti-angiopoietin-2 antibodies that induce Tie2 activation |
Non-Patent Citations (24)
| Title |
|---|
| BABICKOVA JKLINKHAMMER BMBUHL EMDJUDJAJ SHOSS MHEYMANN F ET AL.: "Regardless of etiology, progressive renal disease causes ultrastructural and functional alterations of peritubular capillaries", KIDNEY INT, vol. 91, no. 1, 2017, pages 70 - 85 |
| BOLIGNANO DLACQUANITI ACOPPOLINO GDONATO VCAMPO SFAZIO MR ET AL.: "Neutrophil gelatinase-associated lipocalin (NGAL) and progression of chronic kidney disease", CLINICAL JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY : CJASN, vol. 4, no. 2, 2009, pages 337 - 44, XP055006845, DOI: 10.2215/CJN.03530708 |
| CHEVALIER RLFORBES MSTHORNHILL BA: "Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy", KIDNEY INT, vol. 75, no. 11, 2009, pages 1145 - 52 |
| ENGE MBJARNEGARD MGERHARDT HGUSTAFSSON EKALEN MASKER N ET AL.: "Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy", EMBO J., vol. 21, no. 16, 2002, pages 4307 - 16, XP002962654, DOI: 10.1093/emboj/cdf418 |
| FANG-FANG HE ET AL: "Angiopoietin-Tie signaling in kidney diseases: an updated review", FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 593, no. 19, 11 August 2019 (2019-08-11), pages 2706 - 2715, XP071256418, ISSN: 0014-5793, DOI: 10.1002/1873-3468.13568 * |
| HAMILTON TGKLINGHOFFER RACORRIN PDSORIANO P: "Evolutionary divergence of platelet-derived growth factor alpha receptor signaling mechanisms", MOLECULAR AND CELLULAR BIOLOGY, vol. 23, no. 11, 2003, pages 4013 - 25 |
| HAN SANGYEUL ET AL: "Amelioration of sepsis by TIE2 activation-induced vascular protection", SCIENCE TRANSLATIONAL MEDICINE, vol. 8, no. 335, 20 April 2016 (2016-04-20), XP055875476, ISSN: 1946-6234, DOI: 10.1126/scitranslmed.aad9260 * |
| HAN SLEE SJKIM KELEE HSOH NPARK IKO EOH SJLEE YSKIM D: "Amelioration of sepsis by TIE2 activation-induced vascular protection", SCI TRANSL MED, vol. 8, no. 335, 20 April 2016 (2016-04-20), pages 335 - 55, XP055875476, DOI: 10.1126/scitranslmed.aad9260 |
| HAYASHI SMCMAHON AP: "Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse", DEV BIOL, vol. 244, no. 2, 2002, pages 305 - 18, XP055112559, DOI: 10.1006/dbio.2002.0597 |
| HULTSTROM M, FROMELL K, LARSSON A, PERSSON B, NILSSON B, QUAGGIN SE, BETSHOLTZ C,FRITHIOF R, LIPCSEY M, JEANSSON M.: "Angiopoietin-2 Inhibition of Thrombomodulin-Mediated Anticoagulation-A Novel Mechanism That May Contribute to Hypercoagulation in Critically III COVID-19 Patients", BIOMEDICINES, vol. 10, no. 6, 2022 |
| JIANG LU ET AL: "Reduction of renal interstitial fibrosis by targeting Tie2 in vascular endothelial cells", vol. 95, no. 4, 27 November 2023 (2023-11-27), US, pages 959 - 965, XP093189156, ISSN: 0031-3998, Retrieved from the Internet <URL:https://www.nature.com/articles/s41390-023-02893-8> DOI: 10.1038/s41390-023-02893-8 * |
| KIM W ET AL: "COMP-angiopoietin ameliorates renal fibrosis in a unilateral ureteral obstruction model", JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY, WILLIAMS AND WILKINS, BALTIMORE, MD, US, vol. 17, no. 9, 1 January 2006 (2006-01-01), pages 2474 - 2483, XP008104736, ISSN: 1046-6673, DOI: 10.1681/ASN.2006020109 * |
| KRAMANN RTANAKA MHUMPHREYS BD: "Fluorescence microangiography for quantitative assessment of peritubular capillary changes after AKI in mice", J AM SOC NEPHROL, vol. 25, no. 9, 2014, pages 1924 - 31 |
| LEE VK, HOSKING BM, HOLENIEWSKA J, KUBALA EC, LUNDH VON LEITHNER P, GARDNER PJ, FOXTON RH, SHIMA DT.: "BTBR ob/ob mouse model of type 2 diabetes exhibits early loss of retinal function and retinal inflammation followed by late vascular changes.", DIABETOLOGIA., vol. 61, no. 11, 2018, pages 2422 - 2432 |
| LOGANATHAN KSALEM SAID EWINTERROWD EOREBRAND MHE LVANLANDEWIJCK M ET AL.: "Angiopoietin-1 deficiency increases renal capillary rarefaction and tubulointerstitial fibrosis in mice", PLOS ONE, vol. 13, no. 1, 2018, pages 0189433 |
| MADISEN LZWINGMAN TASUNKIN SMOH SWZARIWALA HAGU H ET AL.: "A robust and high-throughput Cre reporting and characterization system for the whole mouse brain", NAT NEUROSCI, vol. 13, no. 1, 2010, pages 133 - 40, XP055199562, DOI: 10.1038/nn.2467 |
| MIIKKA VIKKULALAURENCE M BOONKERMIT L.CARRAWAY LIIJENNIFER T CALVERTA.JOHN DIAMONTIBOYAN GOUMNEROVKRYSTYNA A PASYKDOUGLAS A MARCHU: "Vascular Dysmorphogenesis Caused by an Activating Mutation in the Receptor Tyrosine Kinase TIE2", CELL, vol. 87, 1996, pages 1181 - 1190, XP029186414, DOI: 10.1016/S0092-8674(00)81814-0 |
| PITULESCU MESCHMIDT IBENEDITO RADAMS RH: "Inducible gene targeting in the neonatal vasculature and analysis of retinal angiogenesis in mice", NAT PROTOC, vol. 5, no. 9, 2010, pages 1518 - 34 |
| SAHARINEN, P.EKLUND, L.ALITALO, K.: "Therapeutic targeting of the angiopoietin-TIE pathway", NAT REV DRUG DISCOV, vol. 16, 2017, pages 635 - 661, Retrieved from the Internet <URL:https://doi.orq/10.1038/nrd.2016.278> |
| SYRJ�L� S O ET AL: "Angiopoietin-2 Inhibition Prevents Transplant Ischemia-Reperfusion Injury and Chronic Rejection in Rat Cardiac Allografts", AMERICAN JOURNAL OF TRANSPLANTATION, BLACKWELL MUNKSGAARD, DK, vol. 14, no. 5, 7 April 2014 (2014-04-07), pages 1096 - 1108, XP072347925, ISSN: 1600-6135, DOI: 10.1111/AJT.12672 * |
| VENKAT-RAMAN ET AL., NEPHROL DIAL TRANSPL, 2012 |
| YANG HCZUO YFOGO AB: "Models of chronic kidney disease", DRUG DISCOVERY TODAY DISEASE MODELS, vol. 7, no. 1-2, 2010, pages 13 - 9, XP055960654, DOI: 10.1016/j.ddmod.2010.08.002 |
| YE YBAJAJ MYANG HCPEREZ-POLO JRBIRNBAUM Y: "SGLT-2 Inhibition with Dapagliflozin Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Cardiomyopathy in Mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor", CARDIOVASC DRUGS THER, vol. 31, no. 2, 2017, pages 119 - 132, XP036231803, DOI: 10.1007/s10557-017-6725-2 |
| ZHOU AAJEANSSON MAHE L ET AL.: "Renal endothelial single-cell transcriptomics reveals spatiotemporal regulation and divergent roles of differential gene transcription and alternative splicing in murine diabetic nephropathy", INT J MOL SCI, 2024 |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2025186364A8 (fr) | 2025-10-02 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US8841424B2 (en) | Humanized AXL antibodies | |
| JP7767281B2 (ja) | 線維性疾患の予防及び処置のための抗クローディン-1モノクローナル抗体。 | |
| KR102513989B1 (ko) | 간 질환을 치료하거나 예방하는 방법 | |
| KR20160021125A (ko) | 항-cxcl1, cxcl7 및 cxcl8 항체 및 이들의 용도 | |
| CN113735974B (zh) | 针对Claudin18.2的抗体及其用途 | |
| CN111741975A (zh) | 抗血管生成素-2抗体及其用途 | |
| JP2024164139A (ja) | Egfl6特異的モノクローナル抗体及びそれらの使用方法 | |
| JP2023511686A (ja) | 腎疾患の治療に使用するための抗αvβ8インテグリン抗体 | |
| US12018074B2 (en) | Anti CCL24 (eotaxin2) antibodies for use in the treatment of hepatic diseases | |
| JP7511241B2 (ja) | リシル-tRNA合成酵素N末端に特異的に結合する抗体を有効成分として含む免疫細胞の遊走関連疾患の予防又は治療用薬学的組成物 | |
| CA3227057A1 (fr) | Utilisation d'un antagoniste d'alpha-enolase dans le traitement de fibroses | |
| CN104470950B (zh) | 抗cxadr抗体 | |
| KR102654035B1 (ko) | 도펠 단백질 억제제 | |
| WO2025186364A1 (fr) | Anticorps ciblant angpt2 destinés à être utilisés dans le traitement de maladies fibrotiques, en particulier d'une ou de plusieurs maladies rénales, d'un ou plusieurs troubles ou d'une lésion associés à une fibrose rénale | |
| US10654925B2 (en) | Anti-marinobufagenin antibodies and uses thereof | |
| CN114901313B (zh) | 急性期的视神经脊髓炎的预防或治疗剂 | |
| JP7698842B2 (ja) | 糖尿病性自律神経障害の予防又は治療剤 | |
| WO2025217258A1 (fr) | Anticorps nptx1 | |
| WO2025070749A1 (fr) | Agent préventif ou thérapeutique pour les symptômes associés accompagnant une maladie induite par un trouble de la moelle épinière ou du cerveau | |
| JP2025522462A (ja) | 腎疾患の予防、治療または改善のための融合タンパク質 | |
| JP2023501316A (ja) | 腎機能障害を治療するための抗il-33治療薬 | |
| WO1999058150A1 (fr) | Agents preventifs/remedes contre la cirrhose du foie | |
| HK40013366A (en) | Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25711041 Country of ref document: EP Kind code of ref document: A1 |