US20170137506A1 - Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator - Google Patents

Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator Download PDF

Info

Publication number
US20170137506A1
US20170137506A1 US15/320,101 US201515320101A US2017137506A1 US 20170137506 A1 US20170137506 A1 US 20170137506A1 US 201515320101 A US201515320101 A US 201515320101A US 2017137506 A1 US2017137506 A1 US 2017137506A1
Authority
US
United States
Prior art keywords
gdf15
subject
seq
renal
antibody
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.)
Abandoned
Application number
US15/320,101
Other languages
English (en)
Inventor
Jeno Gyuris
Lorena Lerner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aveo Pharmaceuticals Inc
Original Assignee
Aveo Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aveo Pharmaceuticals Inc filed Critical Aveo Pharmaceuticals Inc
Priority to US15/320,101 priority Critical patent/US20170137506A1/en
Publication of US20170137506A1 publication Critical patent/US20170137506A1/en
Assigned to AVEO PHARMACEUTICALS, INC. reassignment AVEO PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GYURIS, JENO, LERNER, LORENA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to methods of using, and compositions containing, a GDF15 modulator for treating a subject having a renal disorder or renal dysfunction, particularly chronic kidney disease, acute, chronic or end stage renal failure, glomerulonephritis, anemia, diabetic nephropathy and insulin resistance.
  • a renal disorder or renal dysfunction particularly chronic kidney disease, acute, chronic or end stage renal failure, glomerulonephritis, anemia, diabetic nephropathy and insulin resistance.
  • Kidney disease is a common and expensive condition that is a major source of morbidity and mortality in humans. From a clinical perspective, kidney diseases can be classified as acute kidney injury (AM) and chronic kidney disease (CKD).
  • AKI also termed acute kidney failure or acute renal failure, is a rapid loss of renal function, and may end up with full, partial or no recovery of normal renal function.
  • AKI is an abrupt (within 48 hours) reduction in kidney function, which may include an absolute increase in serum creatinine of at least 0.3 mg/dl, a percentage increase in serum creatinine of at least 50%, or a reduction in urine output of less than 0.5 ml/kg per hour for more than six hours. See Metha et al., 2007, C RITICAL C ARE, 11:R31.
  • AKI may be brought about post-renally, through obstruction of the urinary collection system by either intrinsic or extrinsic masses.
  • AKI may originate within the renal system itself, by disorders of or injury affecting the structures of the nephron, such as the glomeruli, tubules, vessels, or interstitium.
  • CKD is a progressive loss of function over a prolonged period of time. An AKI patient who does not recover renal function may progress to CKD. Moreover, CKD-affected patients are prone to suffer AKI-like events.
  • GDF15 Growth Differentiation Factor-15
  • TGF- ⁇ transforming growth factor-beta
  • BMPs bone morphogenetic proteins
  • GDNF glial cell-line derived neurotrophic factor
  • myostatin also known as GDF-8
  • GDF15 was initially isolated from tissues such as prostate and placenta, and has been known by the additional names macrophage inhibitory cytokine 1 (or MIC1), NSAID-activated gene 1 protein (or NAG1), NSAID-regulated gene 1 protein (or NRG-1), placental TGF-beta (or PTGFB), placental bone morphogenetic protein (or PLAB), and prostate differentiation factor (or PDF).
  • macrophage inhibitory cytokine 1 or MIC1
  • NAG1 NSAID-activated gene 1 protein
  • NRG-1 NSAID-regulated gene 1 protein
  • PTGFB placental TGF-beta
  • PLAB placental bone morphogenetic protein
  • PDF prostate differentiation factor
  • the present invention provides methods and compositions useful for detecting, preventing, and treating conditions and disorders that involve disease, dysfunction, hypertrophy or hypotrophy of kidneys or renal tissue.
  • Such conditions include, for example, chronic kidney or end stage renal failure, uremic syndrome, anemia and/or reduced erythropoietin production from the kidneys, diabetes, insulin resistance and reduced kidney function or kidney size.
  • compositions which reduce or inhibit the activity of GDF15 for example, by reducing the ability of GDF15 to bind to an endogenous binding partner (also referred to as cognate receptor or binding partner), for example, by competitively binding to GDF15 or to an endogenous binding partner, or by otherwise neutralizing the activity of GDF15.
  • an endogenous binding partner also referred to as cognate receptor or binding partner
  • such a composition may comprise an antibody that binds to GDF15 or an endogenous binding partner, as well as a peptide or fusion molecule that comprises such an antibody.
  • the composition may comprise a peptide or small molecule that binds, for example, competitively binds, to GDF15 or to an endogenous binding partner, such that the activity of GDF15 is reduced or inhibited, for example, by reducing or inhibiting the ability of GDF15 to bind to its endogenous binding partner or otherwise neutralizing the activity of GDF 15.
  • the invention provides a method of increasing renal function in a subject in need thereof, the method comprising administering an effective amount of a composition comprising a GDF15 modulator thereby to increase renal function in the subject.
  • Renal function can include any of the biochemical and physiological parameters discussed below.
  • the invention provides a method of treating a subject having a renal disorder or renal dysfunction, the method comprising administering an effective amount of a composition comprising a GDF15 modulator thereby to ameliorate a symptom of the renal disorder or renal dysfunction.
  • the symptoms can include any of the biochemical and physiological parameters discussed below.
  • the invention provides a method of reducing or reversing renal hypotrophy in a subject in need thereof wherein the subject has one or more symptoms of chronic kidney disease, the method comprising administering an effective amount of a composition comprising a GDF15 modulator thereby to reduce or reverse renal hypotrophy in the subject.
  • the symptoms can include any of the biochemical and physiological parameters discussed below.
  • the invention provides a method of treating or preventing chronic kidney disease (CKD) in a subject in need thereof, the method comprising administering to the subject an effective amount of a composition comprising a GDF15 modulator thereby to treat or prevent CKD in the subject.
  • CKD chronic kidney disease
  • the subject exhibits a glomerular filtration rate (GFR) below 90 ml creatinine/minute/1.73 m 2 body-surface area.
  • GFR glomerular filtration rate
  • albuminuria e.g., urinary excretion of albumin in excess of 30 mg per day, 30 mg per liter of urine, or 30 ⁇ g/mg of creatinine in urine.
  • the subject exhibits hyperuricemia, exhibits a serum uric acid level of at least 6.3 mg/dL, exhibits iron deficiency, or exhibits transferrin saturation of below 25% and a low ferritin level.
  • the subject has been diagnosed as having chronic kidney disease.
  • the GDF15 modulator of the invention can decrease or inhibit GDF15 activity in the subject. In certain embodiments, the GDF15 modulator inhibits the activity, expression or binding of GDF15 to its cognate receptor.
  • the GDF15 modulator can be an anti-GDF15 antibody, which can be humanized or human.
  • the invention comprises a method of treating a subject exhibiting one or more of the following characteristics, which can be indicative of renal dysfunction or disease, such as chronic kidney disease.
  • renal-related characteristics include:
  • useful biomarkers indicative of renal dysfunction include one or more of the following: cystatin C in the urine or plasma; urinary C-reactive protein (uCRP); urinary retinol-binding protein (uRBP); neutrophil gelatinase-associated lipocalin (NGAL); hepcidin; creatinine; hemojuvelin; uric acid and/or urea; beta trace protein; kidney injury molecule-1 (KIM-1); urinary N-acetyl-beta-(D)-glucosaminidase (NAG); urinary interleukin-18 (uIL-18); liver fatty acid binding protein-1 (L-FABP-1); blood urea nitrogen (BUN); micro-RNA 21 (miRNA-21); and electrolytes.
  • cystatin C in the urine or plasma include urinary C-reactive protein (uCRP); urinary retinol-binding protein (uRBP); neutrophil gelatinase-associated lipocalin (NGAL); hepcidin; creatin
  • Subjects with abnormal levels of these biomarkers may be candidates for treatment with GDF15 modulators.
  • the clinician will use one or more of the above characteristics in combination with other observations, such as family history of kidney disease, or whether the subject has had, or has been diagnosed as requiring renal intervention, such as kidney transplant or dialysis.
  • the above renal-related characteristics and biomarkers can also be used to monitor the subject's progress in response to treatment with a GDF15 modulator in accordance with the present invention, and to modify the dosing regimen if deemed clinically appropriate.
  • the subject having a renal disorder such as chronic kidney disease (CKD)
  • CKD chronic kidney disease
  • the present invention provides methods and compositions for avoiding or reducing the occurrence and/or severity of at least one of the above renal-related characteristics, and may also avoid or reduce the need for further renal treatments, by administering to the subject a GDF15 inhibitor.
  • the present invention comprises methods of improving at least one of the following characteristics in a subject, wherein the subject has been diagnosed as, or considered to be at risk of developing chronic kidney disease:
  • the GDF15 modulator of the invention can decrease or inhibit GDF15 activity in the subject. In certain embodiments, the GDF15 modulator inhibits the activity, expression or binding of GDF15 to its cognate receptor.
  • the GDF15 modulator can be an anti-GDF15 antibody, which can be humanized or human.
  • the above renal-related characteristics can be monitored to confirm the subject's progress in response to treatment with GDF15 binding inhibitors in accordance with the present invention, and to modify the dosage regime if deemed clinically appropriate.
  • the subject having a renal disorder such as chronic kidney disease (CKD)
  • CKD chronic kidney disease
  • the present invention provides methods and compositions for avoiding or reducing the occurrence and/or severity of at least one of the above characteristics, and may also avoid or reduce the need for one of the renal interventions described above.
  • FIG. 1 is a graph showing the effect on mice of intra-peritoneal administration of 40 ⁇ g recombinant murine Fc-GDF15 protein or phosphate buffered saline (PBS), where body weight decreased significantly in animals treated with mFC-GDF15 ( ⁇ ) but not with PBS ( ⁇ ).
  • PBS phosphate buffered saline
  • FIGS. 2A-2B are bar charts showing the effect of a single dose of 40 ⁇ g murine Fc-GDF15 recombinant protein injected intraperitoneally into mice, where both gonadal ( FIG. 2A ) and gastroc muscle ( FIG. 2B ) mass decreased significantly.
  • left hand bar is PBS
  • right-hand bar is mFc-GDF15.
  • FIG. 3 is a bar chart showing the effect on mice following injection of recombinant murine Fc-GDF15.
  • ALT alanine aminotransferase
  • ALK alkaline phosphatase
  • FIG. 4A is a graph showing that body weight loss is reversed in immune-incompetent mice (ICR-SCID) bearing an HT-1080 fibrosarcoma tumor xenograft model following administration of 2 mg/kg of an anti-GDF15 antibody (01G06). Arrows indicate intra-peritoneal injection of antibody.
  • FIG. 4B is a bar chart showing that administration of the anti-GDF15 antibody increased organ mass (liver, heart, spleen, kidney) and increased tissue mass (gonadal and gastrocnemius) compared to negative control (murine IgG ( ⁇ ) and baseline (day 1).
  • FIG. 5A is a graph showing the effect of systemic administration of anti-GDF15 antibody (Hu01G06-127) on ICR Scid mice bearing HT-1080 human tumor xenografts. Such mice exhibit significant body weight loss. Administration of anti-GDF15 antibody ( ⁇ ) but not human IgG ( ⁇ ) reversed the loss of body weight.
  • FIG. 5B is a bar chart showing that administration of an anti-GDF antibody, but not human IgG, restored gonadal mass.
  • FIG. 6 is a bar chart showing the effect on serum levels of urea nitrogen following administration of antibody to GDF15.
  • Mice bearing HT-1080 human tumor xenografts exhibited increased levels of serum urea nitrogen, a marker of kidney impairment. This increase is reversed by the administration of an anti-GDF15 antibody. Bars from left to right are SHAM, Baseline (93%), hIgG 10 mpk, and Hu01G06-127 10 mpk.
  • the present invention provides methods and compositions for treating a subject having any of a number of kidney-related or renal disease states, for example, a subject having chronic kidney disease, end stage renal failure, diabetes, insulin resistance, kidney hypertrophy, kidney hypotrophy, polycystic kidney disease, proteinuria, hyperglycemia, hyperuricemia, uremic syndrome, gout, kidney stones, hypertension or hypertensive nephropathy, dyslipidemia, anemia and/or reduced erythropoietin production; iron deficiency or hyperfiltration, where such disease state is associated with a renal disorder.
  • kidney-related or renal disease states for example, a subject having chronic kidney disease, end stage renal failure, diabetes, insulin resistance, kidney hypertrophy, kidney hypotrophy, polycystic kidney disease, proteinuria, hyperglycemia, hyperuricemia, uremic syndrome, gout, kidney stones, hypertension or hypertensive nephropathy, dyslipidemia, anemia and/or reduced erythropoie
  • the methods and compositions may be useful in treating a subject with chronic kidney disease, which may be caused by the above disease states, or who exhibits at least one characteristic that is symptomatic of chronic kidney disease, renal failure, uremic syndrome, renal dystrophies, other renal conditions or disorders, acute kidney injury, acute kidney disease or adverse clinical outcomes resulting in acute renal failure, for example, obstructive nephropathy, including one or more of the following:
  • the subject exhibits chronic kidney disease (CKD).
  • CKD chronic kidney disease
  • a subject having CKD or another kidney-related disease or disorder who exhibits symptoms of CKD or who is diagnosed as having CKD or at risk of having CKD may not be optimally treated by existing treatments.
  • treatment in accordance with the methods and compositions of the present invention may be especially beneficial in improving one or more of the characteristics above.
  • the subject exhibits one or more of the following characteristics such that the subject is considered to have CKD or considered to be suffering from CKD, such that the subject may benefit from treatment according to the present invention.
  • the term “considered to have CKD” or “considered to be suffering from CKD” means that following the disclosure of this application, one skilled in the art would expect that a subject would benefit from the administration of GDF15 inhibitors in accordance with the present invention.
  • a subject is also “considered to have CKD” or “considered to be suffering from CKD” if a qualified clinical professional, after examination of information related to the subject, has made the professional judgment or diagnosis that the subject presently suffers from CKD.
  • CKD considered to be at risk of developing CKD
  • a subject is also term “considered to be at risk of developing CKD” if a qualified clinical professional, after examination of information related to the subject, has made the professional judgment or diagnosis that the subject presently a risk of developing CKD, sufficient to justify prophylactic or therapeutic intervention.
  • treat means the treatment of a disease in a mammal, e.g., in a human. This includes: (a) inhibiting the disease, i.e., arresting its development; and (b) relieving the disease, i.e., causing regression of the disease state.
  • GFR glomerular filtration rate
  • GFR is measured in terms of clearance or filtration of ml of creatinine per minute per 1.73m 2 body-surface area.
  • cystatin C another marker for CKD prognosis, is a potential alternative to serum creatinine for estimating GFR. See, Inker et al., 2012, N. E. J. M EDICINE, 367:20-29.
  • a subject exhibiting GFR less than 90 is considered to have CKD.
  • a subject exhibiting GFR greater than or equal to 90 may be considered to be at CKD stage 1, or at an increased risk of CKD, after taking into consideration other indicators of CKD.
  • indicators of CKD risk factors include higher than normal levels of creatinine or urea in the blood, blood or protein in the urine, and a family history of polycystic kidney disease.
  • a subject exhibiting kidney damage with a GFR between 60 and 89 ml/minute/1.73 m 2 body-surface area is considered to be at CKD stage 2.
  • a subject exhibiting a GFR between 30 and 59 ml/minute/1.73 m 2 body-surface area is considered to be at CKD stage 3, at which many clinicians consider the possibilities of renal intervention.
  • the interventions are strict dietary modification plans, in which daily intake of phosphorus, calcium, carbohydrates and sodium may be recommended. Supplementation of the diet with protein, water-soluble vitamin B complex, and vitamin C also may be recommended.
  • a subject exhibiting GFR between 15 and 29 ml/minute/1.73 m 2 body-surface area is considered to be at CKD stage 4, at which many clinicians consider dialysis and/or preparation for kidney replacement.
  • a subject exhibiting GFR of less than 15 ml/minute/1.73 m 2 body-surface area is considered to be at CKD stage 5, or kidney failure, an urgent stage at which intervention such as kidney replacement may be urgently needed, particularly if uremia is present.
  • a subject presenting with CKD stages 1-5 may benefit from therapy according to the present invention, in order to fully or partially restore the loss of kidney function, or to slow or prevent further deterioration of kidney function.
  • a subject is considered to be suffering from chronic kidney disease if their urinary NGAL levels are in excess 50 ⁇ g/L; and more serious CKD if the urinary or plasmatic NGAL levels are in excess of 100 ⁇ g/L.
  • a subject can be considered to be suffering from chronic kidney disease if they exhibit microalbuminuria: urinary excretion of albumin in excess of 30 mg per day, 30 mg per liter of urine, or 30 ⁇ g/mg of creatinine in urine.
  • a subject is considered to be suffering from serious CKD if they exhibit macroalbuminuria: their urinary excretion of albumin is 300 mg or more per day, 300 mg or more per liter of urine, or 300 ⁇ g or more/mg of creatinine in urine. See Levey and Coresh, 2012, T HE L ANCET, 379:165-180.
  • a subject is considered to be suffering from chronic kidney disease if they exhibit uric acid in the blood in excess of 530 micromol/L (6 mg/dL) for women and 619 micromol/L (7 mg/dL) for men or if they suffer from kidney stones. See also, peak VO 2 , Jalal et al., 2012, A M. J. OF K IDNEY D ISEASE, 62:134-146.
  • a subject is considered to be suffering from chronic kidney disease if they exhibit iron deficiency, particularly a low transferrin saturation ( ⁇ 25%) coupled with low ferritin ( ⁇ 200 ng/mL in dialysis patients; ⁇ 100 ng/mL in non-dialysis patients). See Larson and Coyne, 2013, K IDNEY R ESEARCH AND C LINICAL P RACTICE, 32:11-15.
  • Kidney hypertrophy may be compensatory, for loss of kidney tissue function. For example, hypertrophy is frequently observed in patients with a solitary functional kidney.
  • Cell hypertrophy can be assessed by total protein/cell and electronic volume. See Huang et al., 2014, M OLECULAR AND C ELLULAR E NDOCRINOLOGY, 390:45-53.
  • a subject is considered to be suffering from chronic kidney disease if they experience renal hypotrophy, hypoplasia, or significant decrease in kidney mass. Renal hypotrophy can be assessed, for example, by magnetic resonance imaging. See Chang et al., 2007, J. U ROLOGY, 178:2550-2554.
  • the subject exhibits one or more additional symptoms of chronic kidney disease.
  • This may include one or more of the following indicators of chronic kidney disease, renal failure or kidney dysfunction: increased serum creatinine levels, decreased serum bilirubin levels, increased urine albumin concentration, increased urinary creatinine levels, increased urinary albumin-to-creatine ratio (albumin-to-creatinine ratio of 25 mg/g or higher in women and 17 mg/g or higher in men, with a value of 30 mg/g indicative of serious CKD), increased urinary protein-to-creatinine ratio (protein-to-creatinine ratio of 200 mg/g is considered to be too high and indicative of CKD), hypertension (defined as systolic blood pressure of 140 mm Hg or above; diastolic blood pressure of 90 mm Hg or above; or undergoing current antihypertensive drug treatment), diabetes mellitus (defined as fasting glucose level of 126 mg/dL or higher; or the use of insulin or oral hypogly
  • useful biomarkers indicative of renal dysfunction include one or more of the following: cystatin C in the urine or plasma; urinary C-reactive protein (uCRP); urinary retinol-binding protein (uRBP); neutrophil gelatinase-associated lipocalin (NGAL); hepcidin; creatinine; hemojuvelin; uric acid and/or urea; beta trace protein; kidney injury molecule-1 (KIM-1); urinary N-acetyl-beta-(D)-glucosaminidase (NAG); urinary interleukin-18 (uIL-18); liver fatty acid binding protein-1 (L-FABP-1); blood urea nitrogen (BUN); micro-RNA 21 (miRNA-21); and electrolytes.
  • cystatin C in the urine or plasma include urinary C-reactive protein (uCRP); urinary retinol-binding protein (uRBP); neutrophil gelatinase-associated lipocalin (NGAL); hepcidin; creatin
  • solutes including phosphates, dimethyarginines (asymmetric, or ADMA, and symmetric, or SDMA), uric acid, parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), beta-2 microalbumin (B2M), interleukin-6 (IL-6), protein-bound indoxyl sulfate (IS) and p-cresol and its conjugates, p-cresylsulfate (PCS) and p-cresylglucuronide (PCG), may be indicative of decreased renal function or a renal disorder, e.g., “uremic syndrome.” See, Liabeuf et al., 2014, S EMIN N EPHROL, 34(2):164-179, http://dx.doi.org/10.1016/j.copephro.2014.02.008.
  • Subjects with abnormal levels of these biomarkers may be candidates for treatment with GDF15 modulators.
  • the clinician will use one or more of the above characteristics in combination with other observations, such as family history of kidney disease, or whether the subject has had, or has been diagnosed as requiring renal intervention, such as kidney transplant or dialysis.
  • the above renal-related characteristics and biomarkers can also be used to monitor the subject's progress in response to treatment with a GDF15 modulator in accordance with the present invention, and to modify the dosing regimen if deemed clinically appropriate.
  • the subject having a renal disorder such as chronic kidney disease (CKD)
  • CKD chronic kidney disease
  • the present invention provides methods and compositions for avoiding or reducing the occurrence and/or severity of at least one of the above renal-related characteristics, and may also avoid or reduce the need for further renal treatments, by administering to the subject a GDF15 inhibitor.
  • the subject may also exhibit elevated levels of GDF15 activity relative to a baseline activity level present in subjects without the renal disorder or dysfunction.
  • Elevated levels of GDF15 activity can determined by measuring the level of GDF15 in a sample from a subject.
  • the amount regarded as an “elevated level” of GDF15 may vary according to the particular tissue or body fluid of interest, as well as the particular assay that is utilized.
  • an “elevated level” of GDF15 may be determined relative to a control distribution of subjects, for example, subjects without a renal disease or dysfunction, for example, CKD, and may be determined at a pre-specified cutoff of, for example, the 75 th percentile (i.e., upper quartile or 25%); 90 th percentile (i.e., upper 10%); or 95 th percentile (i.e., upper 5%).
  • An “elevated level” of GDF15 may also be determined at a pre-specified GDF15 level above the mean, for example one standard deviation above the mean, or two standard deviations above the mean average GDF15 level of a group of control subjects without renal disease or dysfunction, for example, CKD. See, for example, Brown et al., 2002, T HE L ANCET 359:2159-2163; Kempf et al., 2011, N ATURE M EDICINE, 17:581-588.
  • the preferred body sample is a body fluid, for example, a sample of blood plasma, however a sample of amniotic fluid, placental extract, whole blood, serum, buffy coat, urine, cerebrospinal fluid, seminal fluid, synovial fluid, or a tissue biopsy may also be suitable.
  • the amount of GDF15 present in a body sample may be readily determined by, for example, immunoassays (e.g., with a body fluid) or immunohistochemistry (e.g., with sectionalized samples of a tissue biopsy) using an anti-GDF15 antibody. See Tsai et al., 2013, PLOS O NE, 8:e55174.
  • the present invention includes methods of increasing renal function in a subject in need thereof, the method comprising administering an effective amount of a composition comprising a GDF15 inhibitor to increase renal function in a subject who.
  • the subject suffering from renal dysfunction or CKD may exhibit a comorbidity of cachexia, chronic or congestive heart failure, anemia, diabetes or hypertension.
  • GDF15 modulator is understood to mean an agent that reduces or inhibits GDF15 activity, which can result from reduced expression, amount, or biological activity or function, of GDF15.
  • GDF15 modulators or modulating agents useful in the practice of the invention may comprise an anti-GDF15 antibody, an anti-GDF15 receptor antibody, soluble GDF15 mimetics or analogs that prevent GDF15 from binding to its cognate binding partner, a soluble GDF15 receptor mimetic or analog that prevents GDF15 from binding to its cognate binding partner.
  • Additional exemplary GDF15 modulating agents include small molecule inhibitors of GDF15 or a GDF15 receptor, interfering nucleic acids (for example, interfering RNA or antisense nucleic acids (for example, antisense DNA or RNA) that interfere with expression of endogenous GDF15 or a cognate receptor.
  • interfering nucleic acids for example, interfering RNA or antisense nucleic acids (for example, antisense DNA or RNA) that interfere with expression of endogenous GDF15 or a cognate receptor.
  • the GDF15 modulating agent can comprise an anti-GDF15 antibody, which is humanized or human.
  • antibody is understood to mean an intact antibody (e.g., an intact monoclonal antibody) or antigen-binding fragment of an antibody, including an intact antibody or antigen-binding fragment of an antibody (e.g., a phage display antibody including a fully human antibody, a semisynthetic antibody or a fully synthetic antibody) that has been optimized, engineered or chemically conjugated.
  • antibodies that have been optimized are affinity-matured antibodies.
  • antibodies that have been engineered are Fc optimized antibodies, and multispecific antibodies (e.g., bispecific antibodies).
  • antigen-binding fragments examples include Fab, Fab′, F(ab′) 2 , Fv, single chain antibodies (e.g., scFv), minibodies and diabodies.
  • An antibody conjugated to a toxin moiety is an example of a chemically conjugated antibody.
  • the antibody comprises: (a) an immunoglobulin heavy chain variable region comprising the structure CDR H1 -CDR H2 -CDR H3 and (b) an immunoglobulin light chain variable region, wherein the heavy chain variable region and the light chain variable region together define a single binding site for binding GDF15 or a GDF15 receptor.
  • the CDR H1 , CDR H2 , and CDR H3 sequences are interposed between immunoglobulin framework (FR) sequences.
  • the antibody comprises (a) an immunoglobulin light chain variable region comprising the structure CDR L1 -CDR L2 -CDR L3 , and (b) an immunoglobulin heavy chain variable region, wherein the IgG light chain variable region and the IgG heavy chain variable region together define a single binding site for binding GDF15 or a GDF15 receptor.
  • the CDR L1 , CDR L2 , and CDR L3 sequences are interposed between immunoglobulin FR sequences.
  • the antibody comprises: (a) an immunoglobulin heavy chain variable region comprising the structure CDR H1 -CDR H2 -CDR H3 and (b) an immunoglobulin light chain variable region comprising the structure CDR L1 -CDR L2 -CDR L3 , wherein the heavy chain variable region and the light chain variable region together define a single binding site for binding GDF15 or a GDF15 receptor.
  • an immunoglobulin heavy chain variable region comprising the structure CDR H1 -CDR H2 -CDR H3
  • an immunoglobulin light chain variable region comprising the structure CDR L1 -CDR L2 -CDR L3
  • the heavy chain variable region and the light chain variable region together define a single binding site for binding GDF15 or a GDF15 receptor.
  • Exemplary anti-GDF15 antibodies are described, for example, in U.S. Patent Publication No. US 2014-0193427-A1, the disclosure of which is incorporated by reference herein for all purposes.
  • Exemplary anti-GDF15 antibodies useful in the methods and compositions of the invention may, for example, include a heavy chain variable region comprising any one of the nine sets of CDR H1 , CDR H2 , and CDR H3 region sequences set forth in Table 1 below.
  • Exemplary anti-GDF15 antibodies useful in the methods and compositions of the invention may, for example, include a light chain variable region comprising any one of the four sets of CDR L1 , CDR L2 , and CDR L3 region sequences set forth in Table 2 below.
  • anti-GDF15 antibodies useful in the practice of the invention are described in U.S. Patent Publication No. US 2014-0193427-A1, including 01G06, 03G05, 04F08, 06C11, 08G01, 14F11, 17B11, as well as human or humanized forms thereof.
  • the antibodies disclosed herein e.g., 01G06, 03G05, 04F08, 06C11, 08G01, 14F11, or 17B11, or humanized forms thereof
  • the antibodies reverse a symptom or characteristic of CKD or another kidney-related disease or disorder by at least 2%, 5%, 10%, 15%, 20%, 25%, 30% or 35%.
  • an anti-GDF15 antibody useful in the practice of the invention is referred to as 01G06 in U.S. Patent Publication No. US 2014-0193427-A1.
  • Humanized forms of the 01G06 antibody are listed below together with the amino acid sequences of their respective heavy and light chain variable regions.
  • Exemplary humanized anti-GDF15 antibodies include: Hu01G06-1; Hu01G06-46; Hu01G06-52; Hu01G06-100; Hu01G06-101; Hu01G06-102; Hu01G06-103; Hu01G06-104; Hu01G06-105; Hu01G06-106; Hu01G06-107; Hu01G06-108; Hu01G06-109; Hu01G06-110; Hu01G06-111; Hu01G06-112; Hu01G06-113; Hu01G06-114; Hu01G06-122; Hu01G06-127; Hu01G06-135; Hu01G06-138; Hu01G06-146; Hu06C11-1; Hu06C11-27; Hu06C11-30; Hu14F11-1; Hu14F11-23; Hu14F11-24; Hu14F11-39; and Hu14F11-47.
  • the amino acid sequences for the heavy chain and light chain for each of the aforementioned antibodies is set forth below in Table 3.
  • SEQ ID NO: 25 1 diqmtqspas lsasvgetvt itcrtsenlh nylawyqqkq gkspqllvyd aktladgvps 61 rfsgsgsgtq yslkinslqp edfgsyycqh fwsspytfgg gtkleikrad aaptvsifpp 121 sseqltsgga svvcflnnfy pkdinvkwki dgserqngvl nswtdqdskd stysmsstlt 181 ltkdeyerhn sytceathkt stspivksfn rnec SEQ ID NO: 26 1 diqmtqspas lsasvgetvt itcrtsenlh nylawyqqkq gkspq
  • the antibody may be a neutralizing antibody, which reduces GDF15 activity.
  • the antibody may reduce GDF15 activity in an in vivo assay (see, e.g., Johnen et al., 2007, N ATURE M EDICINE 13:1333-1340) by at least 10%, preferably 20%, 30% or 40%, and more preferably at least about 50%, 60%, 80% or 90% of GDF15 compared to GDF15 activity measured in the same assay under the same conditions in the absence of the antibody.
  • the antibody may selectively and/or significantly reduce or inhibit the binding of GDF15 to its endogenous receptor.
  • the term “significantly reduces or inhibits binding” of GDF15 to its receptor is understood to mean that the antibody inhibits GDF15 binding with a potency or percent inhibition that measures at least 10%, preferably 20%, 30% or 40%, and more preferably at least about 50%, 60%, 80% or 90% of GDF15 [serum level/activity] in the absence of said antibody. Binding can be measured using a direct or sandwich enzyme-linked immunosorbent assay (ELISA), as described, e.g., in Tsai et al., 2013, PLOS O NE, 8:e55174.
  • ELISA enzyme-linked immunosorbent assay
  • the term “selectively” in the context of an antibody that binds to GDF15 or GDF15 receptor is understood to mean that the antibody binds GDF15 or a GDF15 receptor with a binding affinity that is at least two, three, four, five or ten times greater than that of a functionally unrelated protein or another member of the TGF- ⁇ superfamily or a receptor of a member of the TGF- ⁇ superfamily.
  • the antibodies are to be administered to a human, the antibodies preferably are “humanized” to reduce or eliminate antigenicity in humans.
  • each humanized antibody has the same or substantially the same affinity for the antigen as the non-humanized mouse antibody from which it was derived.
  • chimeric proteins are created in which mouse immunoglobulin constant regions are replaced with human immunoglobulin constant regions. See, e.g., Morrison et al.,1984, P ROC. N AT. A CAD. S CI. 81:6851-6855, Neuberger et al., 1984, N ATURE 312:604-608; U.S. Pat. No. 6,893,625 (Robinson); U.S. Pat. No. 5,500,362 (Robinson); and U.S. Pat. No. 4,816,567 (Cabilly).
  • CDR grafting the CDRs of the light and heavy chain variable regions are grafted into frameworks from another species.
  • murine CDRs can be grafted into human FRs.
  • the CDRs of the light and heavy chain variable regions of an anti-GDF15 antibody are grafted into human FRs or consensus human FRs.
  • consensus human FRs FRs from several human heavy chain or light chain amino acid sequences are aligned to identify a consensus amino acid sequence.
  • CDR grafting is described in U.S. Pat. No. 7,022,500 (Queen); U.S. Pat. No. 6,982,321 (Winter); U.S. Pat. No. 6,180,370 (Queen); U.S.
  • human CDR sequences are chosen from human germline genes, based on the structural similarity of the human CDRs to those of the mouse antibody to be humanized. See, e.g., U.S. Pat. No. 6,881,557 (Foote); and Tan et al., 2002, J. I MMUNOL. 169:1119-1125.
  • ACTIVMABTM technology Vaccinex, Inc., Rochester, N.Y.
  • ACTIVMABTM technology Vaccinex, Inc., Rochester, N.Y.
  • High levels of combinatorial diversity of IgG heavy and light chains are said to be produced. See, e.g., U.S. Pat. No. 6,706,477 (Zauderer); U.S. Pat. No. 6,800,442 (Zauderer); and U.S. Pat. No. 6,872,518 (Zauderer).
  • HUMAN ENGINEERINGTM technology Another approach for modifying a mouse antibody into a form suitable for medical use in humans is HUMAN ENGINEERINGTM technology, which is practiced commercially by XOMA (US) LLC. See, e.g., PCT Publication No. WO 93/11794 and U.S. Pat. No. 5,766,886 (Studnicka); U.S. Pat. No. 5,770,196 (Studnicka); U.S. Pat. No. 5,821,123 (Studnicka); and U.S. Pat. No. 5,869,619 (Studnicka).
  • Any suitable approach including any of the above approaches, can be used to reduce or eliminate human immunogenicity of an antibody.
  • Fully human mAbs lacking any non-human sequences can be prepared from human immunoglobulin transgenic mice by techniques referenced in, e.g., Lonberg et al., N ATURE 368:856-859, 1994; Fishwild et al., N ATURE B IOTECHNOLOGY 14:845-851, 1996; and Mendez et al., N ATURE G ENETICS 15:146-156, 1997.
  • Fully human mAbs can also be prepared and optimized from phage display libraries by techniques referenced in, e.g., Knappik et al., J. M OL. B IOL. 296:57-86, 2000; and Krebs et al., J. I MMUNOL. M ETH. 254:67-84 2001).
  • variants and derivatives of GDF15 that act as decoys can be useful in the practice of the invention. For example, through deletion analysis, it may be possible to identify smaller biologically active fragments of GDF15 that compete with endogenous GDF15 for its cognate receptor. Similarly, it is possible to create soluble biologically active fragments of the GDF15 receptor that compete with endogenous GDF15 receptor for available GDF.
  • biologically active fragments include, but are not limited to, fragments of a naturally-occurring GDF15 (or homolog) or a GDF15 receptor (or homolog) that compete with endogenous GDF15 or an endogenous GDF15 receptor, respectively, for binding to a cognate binding partner (e.g., GDF15 receptor or GDF15, respectively).
  • antisense nucleic acids DNA and RNA
  • small interfering nucleic acids e.g., siRNAs
  • siRNA inhibitors of GDF15 include siRNAs from Santa Cruz Biotech (Catalog No. sc-39799, targeting mouse GDF15; and Catalog No. sc-39798, targeting human GDF15), siRNAs from Life Technologies (Cat. Nos. AM16708, 4392420, and 1299001, targeting human GDF15; and Cat. Nos. 1320001 and 4390771, targeting mouse GDF15; and Cat. Nos. 1330001 and 4390771, targeting rat GDF15), siRNAs from Fisher Scientific (Catalog No.
  • compositions containing GDF15 modulators such as those disclosed herein, can be formulated into dosage forms or dosage units using standard formulation techniques. However, the pharmaceutical composition should be formulated to be compatible with its intended route of administration.
  • compositions described herein can be administered to a subject via any route, including, but not limited to, intravenous (e.g., by infusion pumps), intraperitoneal, intraocular, intra-arterial, intrapulmonary, oral, inhalation, intravesicular, intramuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, transdermal, transpleural, intraarterial, topical, inhalational (e.g., as mists of sprays), mucosal (such as via nasal mucosa), subcutaneous, transdermal, gastrointestinal, intraarticular, intracistemal, intraventricular, rectal (i.e., via suppository), vaginal (i.e., via pessary), intracranial, intraurethral, intrahepatic, and intratumoral.
  • intravenous e.g., by infusion pumps
  • intraperitoneal intraocular, intra-arterial, intrapulmonary, oral, inhalation, intravesicular, intramuscular
  • compositions are administered systemically (for example by intravenous injection). In some embodiments, the compositions are administered locally (for example by intraarterial or intraocular injection).
  • a preferred route of administration for GDF15 modulators, such as an antibody, is via intravenous infusion.
  • Formulation components suitable for parenteral administration include a sterile diluent such as bacteriostatic water for injection, physiological saline, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA; buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a sterile diluent such as bacteriostatic water for injection, physiological saline, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl paraben
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as EDTA
  • buffers such as acetates, citrates
  • a GDF15 modulator e.g., an antibody
  • a GDF15 modulator is lyophilized, and then reconstituted in buffered saline, at the time of administration.
  • a GDF15 modulator e.g., an antibody
  • a pharmaceutically acceptable carrier means buffers, carriers, and excipients suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the carrier(s) should be “acceptable” in the sense of being compatible with the other ingredients of the formulations and not deleterious to the recipient.
  • Pharmaceutically acceptable carriers include buffers, solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is known in the art.
  • compositions preferably are sterile. Sterilization can be accomplished, for example, by filtration through sterile filtration membranes. Where the composition is lyophilized, filter sterilization can be conducted prior to or following lyophilization and reconstitution.
  • a therapeutically effective amount of active component is in the range of 0.1 mg/kg to 100 mg/kg, e.g., 1 mg/kg to 100 mg/kg, 1 mg/kg to 10 mg/kg.
  • the amount administered will depend on variables such as the type and extent of disease or indication to be treated, the overall health of the patient, the in vivo potency of a GDF15 modulator (e.g., an antibody), the pharmaceutical formulation, and the route of administration.
  • the initial dosage can be increased beyond the upper level in order to rapidly achieve the desired blood-level or tissue-level. Alternatively, the initial dosage can be smaller than the optimum, and the daily dosage may be progressively increased during the course of treatment.
  • Human dosage can be optimized, e.g., in a conventional Phase I dose escalation study designed to run from 0.5 mg/kg to 20 mg/kg.
  • Dosing frequency can vary, depending on factors such as route of administration, dosage amount, serum half-life of the GDF15 modulator (e.g., an antibody), and the disease being treated. Exemplary dosing frequencies are once per day, once per week and once every two weeks.
  • the optimal effective amount of the compositions can be determined empirically and will depend on the type and severity of the disease, route of administration, disease progression and health, mass and body area of the subject. Such determinations are within the skill of one in the art.
  • Examples of dosages of GDF15 modulator molecules which can be used for methods described herein include, but are not limited to, an effective amount within the dosage range of any of about 0.01 ⁇ g/kg to about 300 mg/kg, or within about 0.1 ⁇ g/kg to about 40 mg/kg, or with about 1 ⁇ g/kg to about 20 mg/kg, or within about 1 ⁇ g/kg to about 10 mg/kg.
  • the composition when administered subcutaneously, may be administered at low microgram ranges, including for example about 0.1 ⁇ g/kg or less, about 0.05 ⁇ g/kg or less, or 0.01 ⁇ g/kg or less.
  • the amount of GDF15 modulators administered to a subject is about 10 ⁇ g to about 500 mg per dose, including for example any of about 10 ⁇ g to about 50 ⁇ g, about 50 ⁇ g to about 100 ⁇ g, about 100 ⁇ g to about 200 ⁇ g, about 200 ⁇ g to about 300 ⁇ g, about 300 ⁇ g to about 500 ⁇ g, about 500 ⁇ g to about 1 mg, about 1 mg to about 10 mg, about 10 mg to about 50 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 300 mg, about 300 mg to about 400 mg, or about 400 mg to about 500 mg per dose.
  • a GDF15 modulator is administered at a dose from about 0.025 mg to about 4 mg, from about 0.035 mg to about 2 mg, from about 0.05 mg to about 2 mg, from about 0.1 mg to about 2 mg, from about 0.2 mg to about 1 mg, or from about 0.2 mg to about 0.8 mg of the GDF15 modulator can be administered.
  • 0.5 mg of GDF15 modulator is administered locally.
  • from about 0.05 mg to about 2 mg, from about 0.2 mg to about 2 mg, from about 0.05 mg to about 1.5 mg, from about 0.15 mg to about 1.5 mg, from about 0.4 mg to about 1 mg, or from about 0.5 mg to about 0.8 mg of GDF15 modulator is administered locally.
  • the GDF15 modulator compositions may be administered in a single daily dose, or the total daily dose may be administered in divided dosages of two, three, or four times daily.
  • the compositions can also be administered less frequently than daily, for example, six times a week, five times a week, four times a week, three times a week, twice a week, once a week, once every two weeks, once every three weeks, once a month, once every two months, once every three months, or once every six months.
  • the compositions may also be administered in a sustained release formulation, such as in an implant which gradually releases the composition for use over a period of time, and which allows for the composition to be administered less frequently, such as once a month, once every 2-6 months, once every year, or even a single administration.
  • the sustained release devices (such as pellets, nanoparticles, microparticles, nanospheres, microspheres, and the like) may be administered by injection or surgical implanted in various locations in the body.
  • the dosing of the GDF15 modulator is titrated such that the dose is sufficient to reduce or prevent adverse effects, but yet fully or partially inhibit the activity of the GDF15.
  • the activity of GDF15 can be modulated in a target cell using antisense nucleic acids or small interfering nucleic acids. Modulation can be achieved using expression constructs known in the art, e.g., naked DNA constructs, DNA vector based constructs, and/or viral vector and/or viral based constructs to express nucleic acids encoding an anti-GDF15 siRNA or antisense molecule.
  • DNA constructs and the therapeutic use of such constructs are well known to those of skill in the art (see, e.g., Chiarella et al., 2008, R ECENT P ATENTS A NTI -I NFECT. D RUG D ISC., 3:93-101; Gray et al., 2008, E XPERT O PIN. B IOL. T HER., 8:911-922; Melman et al., 2008, H UM. G ENE T HER., 17:1165-1176).
  • Naked DNA constructs typically include one or more therapeutic nucleic acids (e.g., GDF15 modulators) and a promoter sequence.
  • a naked DNA construct can be a DNA vector, commonly referred to as pDNA.
  • naked DNA typically do not integrate into chromosomal DNA.
  • naked DNA constructs do not require, or are not used in conjunction with, the presence of lipids, polymers, or viral proteins.
  • Such constructs may also include one or more of the non-therapeutic components described herein.
  • DNA vectors are known in the art and typically are circular double stranded DNA molecules. DNA vectors usually range in size from three to five kilo-base pairs (e.g., including inserted therapeutic nucleic acids). Like naked DNA, DNA vectors can be used to deliver and express one or more therapeutic proteins in target cells. DNA vectors do not integrate into chromosomal DNA.
  • DNA vectors include at least one promoter sequence that allows for replication in a target cell. Uptake of a DNA vector may be facilitated by combining the DNA vector with, for example, a cationic lipid, and forming a DNA complex.
  • viral vectors are double stranded circular DNA molecules that are derived from a virus. Viral vectors typically are larger in size than naked DNA and DNA vector constructs and have a greater capacity for the introduction of foreign (i.e., not virally encoded) genes. Like naked DNA and DNA vectors, viral vectors can be used to deliver and express one or more therapeutic nucleic acids in target cells. Unlike naked DNA and DNA vectors, certain viral vectors stably incorporate themselves into chromosomal DNA.
  • viral vectors include at least one promoter sequence that allows for replication of one or more vector encoded nucleic acids, e.g., a therapeutic nucleic acid, in a host cell.
  • Viral vectors may optionally include one or more non-therapeutic components described herein.
  • uptake of a viral vector into a target cell does not require additional components, e.g., cationic lipids. Rather, viral vectors transfect or infect cells directly upon contact with a target cell.
  • the approaches described herein include the use of retroviral vectors, adenovirus-derived vectors, and/or adeno-associated viral vectors as recombinant gene delivery systems for the transfer of exogenous genes in vivo, particularly into humans. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY, Ausubel, F. M. et al. (eds.) Greene Publishing Associates, 1989, Sections 9.10-9.14, and other standard laboratory manuals.
  • Viruses that are used as transduction agents of DNA vectors and viral vectors such as adenoviruses, retroviruses, and lentiviruses may be used in practicing the present invention.
  • Illustrative retroviruses include, but are not limited to: Moloney murine leukemia virus (M-MuLV), Moloney murine sarcoma virus (MoMSV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), gibbon ape leukemia virus (GaLV), feline leukemia virus (FLV), spumavirus, Friend murine leukemia virus, Murine Stem Cell Virus (MSCV) and Rous Sarcoma Virus (RSV)) and lentivirus.
  • M-MuLV Moloney murine leukemia virus
  • MoMSV Moloney murine sarcoma virus
  • Harvey murine sarcoma virus HaMuSV
  • murine mammary tumor virus
  • lentivirus refers to a group (or genus) of complex retroviruses.
  • Illustrative lentiviruses include, but are not limited to: HIV (human immunodeficiency virus; including HIV type 1, and HIV type 2); visna-maedi virus (VMV) virus; the caprine arthritis-encephalitis virus (CAEV); equine infectious anemia virus (EIAV); feline immunodeficiency virus (FIV); bovine immune deficiency virus (BIV); and simian immunodeficiency virus (SIV).
  • HIV human immunodeficiency virus
  • VMV visna-maedi virus
  • CAEV caprine arthritis-encephalitis virus
  • EIAV equine infectious anemia virus
  • FV feline immunodeficiency virus
  • BIV bovine immune deficiency virus
  • SIV simian immunodeficiency virus
  • an adenovirus can be used in accordance with the methods described herein.
  • the genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle.
  • Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus are known to those skilled in the art.
  • Recombinant adenoviruses can be advantageous in certain circumstances in that they are not capable of infecting nondividing cells and can be used to infect a wide variety of cell types, including epithelial cells
  • the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity.
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situ where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors.
  • Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle. It is also one of the few viruses that may integrate its DNA into non-dividing cells, and exhibits a high frequency of stable integration.
  • one or more viral vectors that expresses a therapeutic transgene or transgenes encoding a GDF15 modulator is administered by direct injection to a cell, tissue, or organ of a subject, in vivo.
  • cells are transduced in vitro or ex vivo with such a vector encapsulated in a virus, and optionally expanded ex vivo. The transduced cells are then administered to the subject.
  • Cells suitable for transduction include, but are not limited to stem cells, progenitor cells, and differentiated cells.
  • the transduced cells are embryonic stem cells, bone marrow stem cells, umbilical cord stem cells, placental stem cells, mesenchymal stem cells, neural stem cells, liver stem cells, pancreatic stem cells, cardiac stem cells, kidney stem cells, or hematopoietic stem cells.
  • host cells transduced with viral vector of the invention that expresses one or more polypeptides are administered to a subject to treat and/or prevent an auditory disease, disorder, or condition.
  • Other methods relating to the use of viral vectors can be found in, e.g., Kay, 1997, C HEST, 111(6 Supp.):138S-142S; Ferry et al., 1998, H UM. G ENE T HER., 9:1975-81; Shiratory et al., 1999, L IVER, 19:265-74; Oka et al., 2000, C URR. O PIN.
  • Certain embodiments of the invention provide conditional expression of a polynucleotide of interest.
  • expression is controlled by subjecting a cell, tissue, organism, etc., to a treatment or condition that causes the polynucleotide to be expressed or that causes an increase or decrease in expression of the polynucleotide encoded by the polynucleotide of interest.
  • inducible promoters/systems include, but are not limited to, steroid-inducible promoters such as promoters for genes encoding glucocorticoid or estrogen receptors (inducible by treatment with the corresponding hormone), metallothionine promoter (inducible by treatment with various heavy metals), MX-1 promoter (inducible by interferon), the “GeneSwitch” mifepristone-regulatable system (Sirin et al., 2003, G ENE, 323:67), the cumate inducible gene switch (WO 2002/088346), tetracycline-dependent regulatory systems, etc.
  • steroid-inducible promoters such as promoters for genes encoding glucocorticoid or estrogen receptors (inducible by treatment with the corresponding hormone), metallothionine promoter (inducible by treatment with various heavy metals), MX-1 promoter (inducible by interferon), the “GeneSwitch”
  • Conditional expression can also be achieved by using a site specific DNA recombinase.
  • the vector comprises at least one (typically two) site(s) for recombination mediated by a site specific recombinase.
  • recombinase or “site specific recombinase” include excisive or integrative proteins, enzymes, co-factors or associated proteins that are involved in recombination reactions involving one or more recombination sites (e.g., two, three, four, five, seven, ten, twelve, fifteen, twenty, thirty, fifty, etc.), which may be wild-type proteins (see Landy, 1993, C URRENT O PINION IN B IOTECHNOLOGY, 3:699-707), or mutants, derivatives (e.g., fusion proteins containing the recombination protein sequences or fragments thereof), fragments, and variants thereof
  • Illustrative examples of recombinases suitable for use in particular embodiments of the present invention include, but are not limited to: Cre, Int, IHF, Xis, Flp, Fis, Hin, Gin, OC31 , Cin, Tn3 resolvase, TndX, XerC,
  • the vectors may comprise one or more recombination sites for any of a wide variety of site specific recombinases. It is to be understood that the target site for a site specific recombinase is in addition to any site(s) required for integration of a vector (e.g., a retroviral vector or lentiviral vector).
  • a vector e.g., a retroviral vector or lentiviral vector.
  • vectors comprise a selection gene, also termed a selectable marker.
  • selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, hygromycin, methotrexate, Zeocin, Blastocidin, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli. Any number of selection systems may be used to recover transformed cell lines.
  • herpes simplex virus thymidine kinase (Wigler et al., 1977, C ELL, 11:223-232) and adenine phosphoribosyltransferase (Lowy et al., 1990, C ELL, 22:817-823) genes which can be employed in tk- or aprt-cells, respectively.
  • DNA delivery may occur parenterally, intravenously, intramuscularly, or even intraperitoneally as described, for example, in U.S. Pat. Nos. 5,543,158; 5,641,515; and 5,399,363 (each specifically incorporated herein by reference in its entirety).
  • Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • DNA delivery may occur by use of liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, optionally mixing with cell penetrating polypeptides, and the like, for the introduction of the compositions of the present invention into suitable host cells.
  • the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, a nanoparticle or the like.
  • the formulation and use of such delivery vehicles can be carried out using known and conventional techniques.
  • Exemplary formulations for ex vivo DNA delivery may also include the use of various transfection agents known in the art, such as calcium phosphate, electroporation, heat shock and various liposome formulations (i.e., lipid-mediated transfection).
  • various transfection agents known in the art, such as calcium phosphate, electroporation, heat shock and various liposome formulations (i.e., lipid-mediated transfection).
  • Particular embodiments of the invention may comprise other formulations, such as those that are well known in the pharmaceutical art, and are described, for example, in R EMINGTON: T HE S CIENCE AND P RACTICE OF P HARMACY, 20th Edition. Baltimore, Md.: Lippincott Williams & Wilkins, 2000.
  • GDF15 activity is inhibited by contacting a body fluid with a composition comprising a GDF15 modulator ex vivo under conditions that permit the GDF15 modulators to reduce or inhibit GDF15 activity.
  • Suitable body fluids include those that can be returned to the individual, such as blood, plasma, or lymph.
  • Affinity adsorption apheresis is described generally in Nilsson et al., 1988, B LOOD, 58(1):38-44; Christie et al., 1993, T RANSFUSION, 33:234-242; Richter et al., 1997, ASAIO J., 43(1):53-59; Suzuki et al., 1994, A UTOIMMUNITY, 19: 105-112; U.S. Pat. No. 5,733,254; Richter et al., 1993, M ETABOL. C LIN. E XP., 42:888-894; and Wallukat et al., 1996, I NT'L J. C ARD., 54:1910195.
  • the invention includes methods of treating one or more diseases described herein in a subject comprising treating the subject's blood extracoporeally (i.e., outside the body or ex vivo) with a composition comprising a GDF15 modulator under conditions that permit the modulator to reduce or inhibit GDF15 activity in the blood of the subject.
  • ICR SCID spontaneous mutant T&B cell deficient mice were injected intraperitoneally with a PBS solution (control) or 40 ⁇ g of a murine, recombinant Fc-GDF15 protein.
  • Body weight, and molecular markers for muscle degradation (Atrogin, MurF1), adipogenesis (Glut4, Leptin, C/EPB ⁇ ) and lipid accumulation (Stearoyl-CoA desaturase, Fatty acid synthase) were measured after injection.
  • FIG. 1 shows that mice injected with Fc-GDF15 exhibited significant body weight loss. Similarly, mice injected with Fc-GDF15 exhibited significant loss of gonadal mass ( FIG. 2A ) and gastroc mass ( FIG. 2B ). The treated mice also exhibited upregulation of markers for muscle degradation (Atrogin, MurF1) and downregulation of markers of adipogenesis (Glut4, leptin) and lipid accumulation (stearoyl-CoA desaturase; fatty acid synthase).
  • Atrogin, MurF1 markers for muscle degradation
  • Glut4, leptin markers of adipogenesis
  • lipid accumulation stearoyl-CoA desaturase; fatty acid synthase
  • mice treated with Fc-GDF15 also exhibited lower levels of liver enzymes (alanine aminotransferase (ALT), alkaline phosphatase (ALK); and an increase in urea levels in blood (urea nitrogen, a marker of kidney impairment) ( FIG. 3 ), consistent with a role of GDF15 in kidney function.
  • liver enzymes alanine aminotransferase (ALT), alkaline phosphatase (ALK)
  • ALK alkaline phosphatase
  • urea levels in blood urea nitrogen, a marker of kidney impairment
  • This Example demonstrates the treatment of renal hypotrophy (as indicated by kidney weight loss) by an anti-GDF15 antibody (01G06) in an HT-1080 fibrosarcoma xenograft model.
  • HT-1080 cells were grown in culture at 37° C. in an atmosphere containing 5% CO 2 , using Eagle's Minimum Essential Medium (ATCC, Catalog No. 30-2003) containing 10% FBS. Cells were inoculated subcutaneously into the flank of 8-week old female ICR SCID mice with 5 ⁇ 10 6 cells per mouse in 50% matrigel. Body weight was measured daily. When body weight reached 80%, the mice were randomized into two groups of five mice each.
  • mice received one of the following treatments via intraperitoneal injection: murine IgG control, murine 01G06 dosed at 2 mg/kg on day 1 and day 7.
  • a group of five mice were sacrificed at the time of dosing (baseline or 80% body weight loss, without treatment) and at the end of study (seven days post dose, either mIgG or 01G06).
  • Liver, heart, spleen, kidney, gonadal fat and the gastrocnemius muscles were removed surgically and weighed.
  • Treatment with anti-GDF15 antibody 01G06 resulted in body weight increase to initial weight or 100% (p ⁇ 0.001) ( FIG. 4A ), and in kidney weight increase ( FIG. 4B ).
  • mice treated with anti-GDF15 antibody 01G06 displayed significant liver and gonadal muscle gain compared to the baseline group ( FIG. 4B ).
  • FIGS. 4A-B indicate that an anti-GDF15 antibody can reverse kidney weight loss in an HT-1080 fibrosarcoma xenograft model.
  • the results indicate that an anti-GDF15 antibody can reverse the loss of organ mass, such as kidney mass, loss of muscle mass, loss of fat and involuntary weight loss in an HT-1080 xenograft tumor model.
  • ICR SCID spontaneous mutant T&B cell deficient mice bearing HT-1080 human tumor xenografts, as in Example 2, exhibit cachexia.
  • Administration of the anti-GDF15 antibody also reversed elevated levels of urea observed in mice bearing HT-1080 human tumor xenografts ( FIG. 6 ).
  • Sub-total nephrectomy mimics the progressive renal failure after loss of renal mass in humans.
  • K IDNEY I NTERNATIONAL, 64:350-355 one kidney is removed and approximately 2/3 of the remaining kidney is ablated, resulting in reduced kidney functional relevant to progressive kidney disease.
  • Animals are dosed with either anti-GDF15 antibody or control at the time of treatment. Animals are assessed for body and kidney size and weight, glomerular filtration rate, serum creatinine and other markers for kidney disease.
  • UUO Unilateral ureteral obstruction
  • K IDNEY I NTERNATIONAL 75:1145-1152
  • Ureteral obstruction can be accomplished by ligation, or by placement of a reversible obstruction.
  • Animals are dosed with either anti-GDF15 antibody or control at the time of treatment. Animals are assessed for body and kidney size and weight, glomerular filtration rate, systolic blood pressure, atrophy of the tubules and tubular apoptosis. Glomerular filtrate rate can be measured following relief of UUO using standard clearance techniques.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Urology & Nephrology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US15/320,101 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator Abandoned US20170137506A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/320,101 US20170137506A1 (en) 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462015242P 2014-06-20 2014-06-20
PCT/US2015/036794 WO2015196145A1 (en) 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator
US15/320,101 US20170137506A1 (en) 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/036794 A-371-Of-International WO2015196145A1 (en) 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/179,166 Continuation US11897948B2 (en) 2014-06-20 2018-11-02 Treatment of chronic kidney disease and other renal dysfunction using a GDF15 modulator

Publications (1)

Publication Number Publication Date
US20170137506A1 true US20170137506A1 (en) 2017-05-18

Family

ID=53724443

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/320,101 Abandoned US20170137506A1 (en) 2014-06-20 2015-06-19 Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator
US16/179,166 Active US11897948B2 (en) 2014-06-20 2018-11-02 Treatment of chronic kidney disease and other renal dysfunction using a GDF15 modulator

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/179,166 Active US11897948B2 (en) 2014-06-20 2018-11-02 Treatment of chronic kidney disease and other renal dysfunction using a GDF15 modulator

Country Status (9)

Country Link
US (2) US20170137506A1 (pt)
EP (1) EP3157953B1 (pt)
JP (1) JP6768527B2 (pt)
AU (2) AU2015276800B2 (pt)
BR (1) BR112016029820A2 (pt)
CA (1) CA2952032C (pt)
EA (1) EA201790051A1 (pt)
MX (1) MX2016016826A (pt)
WO (1) WO2015196145A1 (pt)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10588980B2 (en) 2014-06-23 2020-03-17 Novartis Ag Fatty acids and their use in conjugation to biomolecules
US10603359B2 (en) * 2014-10-30 2020-03-31 Acceleron Pharma Inc. Methods and compositions using GDF15 polypeptides for increasing red blood cells
US11137408B2 (en) * 2016-01-29 2021-10-05 Washington University GDF15 in glaucoma and methods of use thereof
US11725047B2 (en) 2012-12-21 2023-08-15 Aveo Pharmaceuticals, Inc. Anti-GDF-15 antibodies

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108367053A (zh) * 2015-12-22 2018-08-03 诺华股份有限公司 使用生长分化因子15(gdf-15)治疗或改善代谢性疾病的方法
US11312766B2 (en) 2016-04-27 2022-04-26 Novartis Ag Antibodies against growth differentiation factor 15 and uses thereof

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US6893625B1 (en) 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US5733254A (en) 1987-10-15 1998-03-31 Cypress Bioscience, Inc. Method for treating patients suffering from immune thrombocytopenic purpura
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US6066718A (en) 1992-09-25 2000-05-23 Novartis Corporation Reshaped monoclonal antibodies against an immunoglobulin isotype
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
IE80468B1 (en) 1995-04-04 1998-07-29 Elan Corp Plc Controlled release biodegradable nanoparticles containing insulin
US6872518B2 (en) 1997-09-22 2005-03-29 University Of Rochester Methods for selecting polynucleotides encoding T cell epitopes
AU2002257420A1 (en) 2001-05-01 2002-11-11 National Research Council Of Canada A system for inducible expression in eukaryotic cells
EP1539233B1 (en) 2001-07-12 2011-04-27 FOOTE, Jefferson Super humanized antibodies
US7919084B2 (en) 2002-06-17 2011-04-05 St. Vincent's Hospital Sydney Limited Methods of diagnosis, prognosis and treatment of cardiovascular disease
EP2441466B1 (en) * 2004-04-13 2014-07-23 St Vincent's Hospital Sydney Limited MIC-1 inhibiting agent
CN101854947A (zh) 2007-08-16 2010-10-06 圣文森特医院悉尼有限公司 用于调节巨噬细胞抑制因子(mic-1)活性的药剂和方法
WO2009038533A1 (en) 2007-09-17 2009-03-26 Bioneris Ab Method and means for the treatment of cachexia
AU2008310310A1 (en) * 2007-10-09 2009-04-16 St Vincent's Hospital Sydney Limited A method of treating cachexia with the removal or inactivation of macrophage inhibitory cytokine-1
WO2010048670A1 (en) * 2008-10-31 2010-05-06 St Vincent's Hospital Sydney Limited Method of prognosis in chronic kidney disease
EP3590537A1 (en) * 2012-09-26 2020-01-08 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
AR094271A1 (es) * 2012-12-21 2015-07-22 Aveo Pharmaceuticals Inc Anticuerpos anti-gdf15

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11725047B2 (en) 2012-12-21 2023-08-15 Aveo Pharmaceuticals, Inc. Anti-GDF-15 antibodies
US10588980B2 (en) 2014-06-23 2020-03-17 Novartis Ag Fatty acids and their use in conjugation to biomolecules
US10786576B2 (en) 2014-06-23 2020-09-29 Novartis Ag Fatty acids and their use in conjugation to biomolecules
US11752211B2 (en) 2014-06-23 2023-09-12 Novartis Ag Fatty acids and their use in conjugation to biomolecules
US10603359B2 (en) * 2014-10-30 2020-03-31 Acceleron Pharma Inc. Methods and compositions using GDF15 polypeptides for increasing red blood cells
US11642394B2 (en) 2014-10-30 2023-05-09 Acceleron Pharma Inc. Methods and compositions using GDF15 polypeptides for increasing red blood cells
US11137408B2 (en) * 2016-01-29 2021-10-05 Washington University GDF15 in glaucoma and methods of use thereof
US11933791B2 (en) 2016-01-29 2024-03-19 Washington University GDF15 in glaucoma and methods of use thereof

Also Published As

Publication number Publication date
MX2016016826A (es) 2017-08-28
CA2952032A1 (en) 2015-12-23
US20190292252A1 (en) 2019-09-26
EP3157953B1 (en) 2021-12-22
AU2015276800B2 (en) 2021-03-04
US11897948B2 (en) 2024-02-13
AU2021203523A1 (en) 2021-07-01
AU2015276800A1 (en) 2017-01-05
EP3157953A1 (en) 2017-04-26
JP6768527B2 (ja) 2020-10-14
BR112016029820A2 (pt) 2017-10-24
EA201790051A1 (ru) 2017-04-28
CA2952032C (en) 2023-03-14
WO2015196145A1 (en) 2015-12-23
JP2017519764A (ja) 2017-07-20

Similar Documents

Publication Publication Date Title
US11897948B2 (en) Treatment of chronic kidney disease and other renal dysfunction using a GDF15 modulator
US20220403015A1 (en) Treatment of congestive heart failure and other cardiac dysfunction using a gdf15 modulator
US20230295285A1 (en) Methods of reversing cachexia and prolonging survival comprising administering a gdf15 modulator and an anti-cancer agent
EP3135298B1 (en) Therapeutic agents for diseases involving choroidal neovascularization
JP5068253B2 (ja) 心臓血管疾患の治療
JP6942356B2 (ja) 制御性t細胞の活性化剤及びその使用
CN112592402B (zh) 抗dkk2抗体、包含该抗dkk2抗体的组合物及其用途
KR102239752B1 (ko) 종양 특이적 약물복합체와 항pd-l1 항체를 유효성분으로 하는 암 예방 또는 치료를 위한 병용 투여용 약학 조성물
EA042620B1 (ru) Лечение хронического заболевания почек и другого нарушения функции почек при помощи модулятора gdf15
EP3353196B1 (en) Polypeptides capable of inhibiting the binding between leptin and neuropilin-1
CA3020339C (en) Compositions and methods for treating pancreatitis and pain with death receptor agonists
WO2022092326A1 (ja) 眼組織線維化の阻害に使用するためのgdf15調節物質

Legal Events

Date Code Title Description
AS Assignment

Owner name: AVEO PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GYURIS, JENO;LERNER, LORENA;SIGNING DATES FROM 20150323 TO 20150401;REEL/FRAME:042459/0323

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION