EP1812044A2 - Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique - Google Patents

Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique

Info

Publication number
EP1812044A2
EP1812044A2 EP05796707A EP05796707A EP1812044A2 EP 1812044 A2 EP1812044 A2 EP 1812044A2 EP 05796707 A EP05796707 A EP 05796707A EP 05796707 A EP05796707 A EP 05796707A EP 1812044 A2 EP1812044 A2 EP 1812044A2
Authority
EP
European Patent Office
Prior art keywords
use according
ghrelin
secretagogue
salt
phe leu
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.)
Withdrawn
Application number
EP05796707A
Other languages
German (de)
English (en)
Inventor
Birgitte Holst Lange
Hans T. Schambye
Tina Geritz Nielsen
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.)
Gastrotech Pharma AS
Original Assignee
Gastrotech Pharma AS
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 Gastrotech Pharma AS filed Critical Gastrotech Pharma AS
Publication of EP1812044A2 publication Critical patent/EP1812044A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/25Growth hormone-releasing factor [GH-RF], i.e. somatoliberin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2264Obesity-gene products, e.g. leptin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH], i.e. somatotropin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/30Insulin-like growth factors, i.e. somatomedins, e.g. IGF-1, IGF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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

Definitions

  • the present invention relates to agents, compositions and methods for the stimulation of appetite, food intake and/or weight gain in individuals suffering from, or at risk of suffering from, renal failure and/or liver failure.
  • Ghrelin is a bioactive peptide which originally was described to be involved in the control of GH secretion but later found to be a major regulator of appetite, food intake and energy homeostasis ( Kojima M et al., Trends Endocrinol Metab 12:118- 122; Nakazato M et al., 2001 , Nature 409:194-198). Similar to many other bioactive peptides, ghrelin probably acts both as a hormone, a paracrine substance and as a neurotransmitter. The story of ghrelin, its receptor and synthetic compounds acting through this receptor unraveled in a unique "reverse" order.
  • ghrelin is secreted in the pre-meal situation which results in a sharp, short-lived surge in plasma levels of ghrelin before the meal and starting 1-2 hours before and lasting a short while after initiation of the meal. Since ghrelin is the only peripherally produced orexigenic (appetite promoting) substance it is believed that the increase in plasma levels of ghrelin is crucial for the initiation of the meal.
  • ghrelin released from the endocrine cells in the mucosa of the Gl tract may act both locally as a paracrine substance and centrally as a hormone.
  • ghrelin has been administered by continuous infusions for 270 minutes, which has shown that an increase in food intake can be obtained through intravenous administration of ghrelin (Wren et al JCEM 2001 ; 86(12)5992-5995).
  • Simren M Stotzer PO
  • Abrahamsson H Bjomsson ES. Delay in gastric emptying in patients with chronic renal failure. Scand. J. Gastroenterol. 2004;39(6):516-20.).
  • One subgroup of patients with hypoalbumineamia who are unresponsive to dialysis and adequate prescription of dietary intake may have delayed gastric emptying ( Silang R, Regalado M, Cheng TH, Wesson DE. Prokinetic agents increase plasma albumin in these patients with delayed gastric emptying. AmJ. Kidney Dis. 2001 ;37(2):287- 93.).
  • the delay in gastric emptying is not correlated to any specific biochemical parameter but is closely associated to the feeling of malaise and tiredness. Liver Failure
  • Liver failure is a state in which the liver is unable to adequately perform its functions.
  • Chronic liver failure may be caused by e.g. alcoholic liver disease, viral hepatitis, hemochromatosis, drug-induced hepatitis, Wilson's disease, sclerosing cholangitis, primary biliary cirrhosis, Budd-Chiari syndrome, venoocclusive disease, autoimmune hepatitis, steatohepatitis, partial hepatectomy, or Reye's syndrome.
  • Alcoholic liver failure, hepatitis C and drug induced liver failure constitute the three major groups of patients with liver failure. They all present a large number of gastro ⁇ intestinal complaints including abdominal pain, early satiety, nausea, vomiting, abdominal distension and anorexia. In some cases these symptoms may be explained by the presence of organic disorders such as peptic ulcer disease or gastroesophagal reflux disease. However in many patients with cirrhosis no obvious cause is apparent for the gastrointestinal symptoms ( Verne GN 1 Soldevia-Pico C, Robinson ME, Spicer KM, Reuben A. Autonomic dysfunction and gastroparesis in cirrhosis. J.Clin. Gastroenterol. 2004;38(1):72-6.).
  • Biliary atresia is the most common cause of liver failure in children. Other causes include hepatitis, Alagille's syndrome, familial intrahepatic cholestasis and metabolic diseases, such as ⁇ -1 -antitrypsin deficiency, tyrosinaemia, type 1 , Wilson's disease, cystic fibrosis, or glycogen storage disease type IV.
  • liver failure may present with jaundice, malaise, fatigue, weakness, nausea, anorexia, pruritus, increased bleeding tendency, esophageal varices, ascites, hepatic encephalopathy, and generally failing health. In children liver failure is often complicated by failure to thrive and growth failure.
  • Liver failure is diagnosed by assessment of liver function tests, such as alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase, albumin, and bilirubin. Liver biopsy is the definitive test for determining the cause of liver failure.
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • alkaline phosphatase albumin
  • bilirubin bilirubin
  • the etiology of anorexia in chronic liver failure is not fully clarified, but may involve elevated cytokine levels, gastroparesis, malaise, and satiety caused by ascitis or hepatomegalia.
  • Prolonged gastrointestinal transit and delayed gastric emptying may be responsible for part of the feeling of satiety.
  • paracentesis to empty ascetic fluid does not alter the delayed gastric emptying but it decreases the feeling of satiety ( Scolapio JS, Ukleja A, McGreevy K, Burnett OL, O'Brien PC.
  • Nutritional problems in end-stage liver disease contribution of impaired gastric emptying and ascites. J.Clin. Gastroenterol. 2002;34(1 ):89-93.).
  • the presence of autonomic dysfunction is positively correlated to the delay in gastric emptying but it is probably not the only cause of the gastroparesis.
  • the initiation of delayed gastric emptying is not correlated to any specific biochemical parameter but is closely associated to the feeling of malaise and tiredness.
  • Treatment of liver failure is aimed at alleviating complications and preventing progressive liver damage.
  • Commonly used therapies include colestyramine, ondansetron, rifampicin, variceal sclerotherapy, lactulose, metronidazole, ascitis paracentesis, sodium and fluid restriction, spironolactone.
  • transplantation remains the only therapeutic option.
  • the invention relates to the use of a growth hormone-(GH-) secretagogue compound for the preparation of a medicament for the stimulation of appetite in an individual suffering from renal failure and/or liver failure. Stimulation of appetite does not necessarily lead to an increase in food intake or stimulate weight gain and accordingly, the present invention further relates to another aspect: the stimulation of food intake and/or weight gain by administering a GH-secretagogue, such as a ghrelin-like compound.
  • a GH-secretagogue such as a ghrelin-like compound.
  • the invention relates to a method for stimulating appetite, food intake and/or weight gain in an individual suffering from liver failure and/or renal failure, said method comprising administration of a GH-secretagogue to said patient.
  • the invention relates to the use of a GH-secretagogue compound for the preparation of a medicament for a) stimulation of appetite, and/or b) stimulation of food intake, and/or c) stimulation of weight gain, and/or d) increasing body fat mass, and/or e) increasing lean body mass, and/or f) increasing the muscle strength and/or g) increasing the muscle perseverance, and/or h) improving Quality of Life (QoL), and/or i) decreasing nausea
  • Preferred combinations are: a); b); c); d); e); f); g); h); and i); in isolation; as well as a)+b); a) + c); a) + d); a) + e); b) + c); b) + d); b) + e); d) +e); c)+ d)+ e) ; a) + c) + d); a) + c) + e); ; a) + d) + e); a) + c) + d); b) + c) + d); b) + c) + d); b) + c) + e); b) + d) + e); and b) + c) + d) + e); a) + b) + i); f) + g) + h); e) + f) + g) + h); c) +
  • the invention relates to the use of a GH-secretagogue compound for the preparation of a medicament for the stimulation of gastric emptying in an individual suffering from liver failure and/or renal failure, in particular gastric emptying in relation to meals.
  • said GH-secretagogue is a ghrelin-like compound which comprises e.g. a structure defined herein below.
  • Affinity the strength of binding between receptors and their ligands, for example between the GHRIa receptor and a ghrelin-like compound,
  • the affinity may be e.g. described using a dissociation constant, Kd.
  • Amino Acid Residue An amino acid formed upon chemical digestion (hydrolysis) of a polypeptide at its peptide linkages.
  • the amino acid residues described herein are preferably in the "L" isomeric form.
  • the amino acid encompasses every amino acid such as L-amino acid, D-amino acid, alpha -amino acid, beta -amino acid, gamma -amino acid, natural amino acid and synthetic amino acid or the like as long as the desired functional property is retained by the polypeptide.
  • NH 2 refers to the free amino group present at the amino terminus of a polypeptide.
  • COOH refers to the free carboxy group present at the carboxy terminus of a polypeptide.
  • amino acid residue sequences represented herein by formulae have a left-to-right orientation in the conventional direction of amino terminus to carboxy terminus.
  • amino acid residue is broadly defined to include the amino acids listed in the Table of Correspondence and modified and non-naturally occurring amino acids.
  • a dash at the beginning or end of an amino acid residue sequence indicates a peptide bond to a further sequence of one or more amino acid residues or a covalent bond to an amino-terminal group such as NH 2 or acetyl or to a carboxy- terminal group such as COOH.
  • Appetite in an individual is assessed by measuring the amount of food ingested and by assessing the individual's desire to eat.
  • Appetite for example hunger
  • Appetite is typically assessed with a short questionnaire given to individuals on a random basis several times a week.
  • subjects rate their hunger, preoccupation with food, and desire to eat greater quantities and different types of food by answering the questions using analogue scales ranging from 1, not at all, to 5, extremely.
  • BMI measures your height/weight ratio. It is determined by calculating weight in kilograms divided by the square of height in meters. The BMI "normal" range is 19- 25.
  • Body fat mass can be measured e.g. by the fat fold technique: In this technique, a pincer-type caliper is used to measure subcutaneous fat by determining skin fold thickness at representative sites on the body. These skin fold measurements are then used to compute body fat by either adding the scores from the various measurements and using this value as an indication of the relative degree of fatness among individuals or by using the measurements in mathematical equations that have been developed to predict percent body fat.
  • Body composition can also be assessed by Dual Energy X-ray Absorptiometry (DEXA) scanning, a non-invasive test which accurately quantifies the lean body mass, total fat mass and regional body fat (e.g. abdominal fat).
  • DEXA Dual Energy X-ray Absorptiometry
  • a concentration equivalent is an Equivalents dosage being defined as the dosage of a ghrelin-like compound having in vitro and/or in vivo the same response as evaluated from a dosage-response curve of wild-type ghrelin.
  • Kd Dissociation constant
  • Fusion Polypeptide A polypeptide comprised of at least two polypeptides and a linking sequence to operatively link the two polypeptides into one continuous polypeptide.
  • the two polypeptides linked in a fusion polypeptide are typically derived from two independent sources, and therefore a fusion polypeptide comprises two linked polypeptides not normally found linked in nature.
  • Ghrelin a polypeptide as described in Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K 1999 Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402:656-660. Human 28 aa ghrelin has the amino acid of SEQ ID NO: 1.
  • Ghrelin-like compound refers to any compound which mimics the function of wild-type ghrelin, such as any of the compounds described herein, such as further described in the section below entitled “Functionality", for example wild-type human ghrelin.
  • the ghrelin-like compound may be defined by the formula I:
  • Z 1 - (X 1 ) m - (X 2 ) - (X 3 ) n - Z 2 , wherein Z 1 is an optionally present protecting group
  • each X 1 is independently selected from an amino acid, wherein said amino acid is selected from naturally occurring and synthetic amino acids,
  • X 2 is any amino acid selected from naturally occurring and synthetic occurring amino acids, said amino acid being modified with a bulky hydrophobic group, preferably an acyl group, or a fatty acid,
  • each X 3 is independently selected from an amino acid, wherein said amino acid is selected from naturally occurring and synthetic amino acids,
  • X 1 and X 3 optionally may be modified by a bulky hydrophobic group, preferably an acyl group, or a fatty acid,
  • Z 2 is an optionally present protecting group
  • n is an integer in the range of from 1-10
  • n is 0 or an integer in the range of from 1-35.
  • a ghrelin-like compound has a functionality leading to the desired therapeutic effects described herein.
  • GHS growth hormone secretagogue
  • GHS-R 1a the receptor for GHS.
  • GHS-R 1a is also denoted GHS 1a, or the ghrelin receptor 1a.
  • the subject is a mammal, including humans and non-human mammals such as dogs, cats, pigs, cows, sheep, goats, horses, rats, and mice. In the most preferred embodiment, the subject is a human.
  • Isolated is used to describe the various ghrelin-like compounds, polypeptides and nucleotides disclosed herein, that have been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In preferred embodiments, the polypeptide will be purified.
  • Lean body mass and "increased or maintained lean body mass” Lean body mass is defined herein as body weight minus body fat; primarily muscle, bone and other non-fat tissue. A number of methods may be used for calculating an individual's percentage lean body mass:
  • Body mass index calculated as body weight (kg) divided by the square of height (m) is used to describe the severity of obesity.
  • BMI body mass index
  • WHR Waist-to-hip ratio
  • CT Computed Tomography
  • CT scan for the assessment of body composition can be performed as either multi-slice covering the whole body and wherefrom calculations of whole body and regional AT can be derived.
  • the CT method has been extensively validated. Animal and human cadaver studies have been used to verify the accuracy of the AT-estimate and its anatomical distribution showing a high correlation and therefore by some suggested as a "gold standard" against which to compare other non-invasive methods (Kvist, H., et al., 1988, INTJ. OBES. 12:249- 266).
  • the most accurate in vivo methods of measuring body composition are multi-slice MRI and CT (Rossner,S., 1990, INT.J.OBES. 14:893-902).
  • DXA Dual energy X-ray Absorptiometry
  • DEXA Dual energy X-ray Absorptiometry
  • DXA is a direct, operator independent, non-invasive method to estimate body composition. Measurements are based on the differential attenuation of two X-rays as they pass through the body. It distinguishes bone mineral from soft tissue and subsequently divides the latter into FM and FFM (Pietrobelli.A-, et al., 1996, Am J Physiol 271 :E941-E951). The analyzed data yield information about composition of the whole body but also permits regional body composition determination. DXA has been widely validated and appear to be a precise (CV ⁇ 1%(bone), CV ⁇ 3%(LBM and FM) and simple way of measuring total and regional FM and LBM. Exposure to radiation is minimal (2-5 ⁇ Sv) in most DXA machines.
  • the increase or maintenance of lean body mass is measured using DXA. In another preferred embodiment, said increase or maintenance of lean body mass is measured using MRI.
  • Increasing lean body mass can mean e.g. either increasing total lean body mass in an individual and/or increasing an individual's overall percentage lean body mass (e.g. as compared to total body mass), such as increasing an individual's percentage lean body mass by more than 0.5 %, such as more than 0.75%, such as more than 1%, for example more than 1.25%, such as more than 1.5%, for example more than 1.75%, such as more than 2%, for example more than 2.25%, such as more than 2.5%, for example more than 2.75%, such as more than 3%, for example more than 3.25%, such as more than 3.5%, for example more than 3.75%, such as more than 4%, for example more than 4.25%, such as more than 4.5%, for example more than 4.75%, such as more than 5%, for example more than 5.25%.
  • 0.5 % such as more than 0.75%, such as more than 1%, for example more than 1.25%, such as more than 1.5%, for example more than
  • the increase lean body mass is caused by an increase in muscle mass, as measured using for instance MRI or CT.
  • said increase of lean body mass may be with respect to a control group of individuals not treated with the GH secretagogue.
  • maintaining lean body mass and grammatical variants thereof is meant that said GH secretagogue acts to counteract loss of an individual's lean body mass, by preventing or reducing a decrease in total amount of lean body mass (as measured using for instance DEXA scans).
  • Modified amino acid an amino acid wherein an arbitrary group thereof is chemically modified.
  • a modified amino acid chemically modified at the alpha - carbon atom in an alpha -amino acid is preferable.
  • Monoclonal Antibody The phrase monoclonal antibody in its various grammatical forms refers to a population of antibody molecules that contains only one species of antibody combining site capable of immunoreacting with a particular antigen.
  • Muscle strength is the maximal force that a muscle can exert. It is usually measured by determination of maximum voluntary isometric contraction, e.g. the handgrip strength test. Muscle strength can also be assessed by measurement of isokinetic strength of isolated muscles/muscle groups, such as the knee extension/flexor muscles.
  • Muscle endurance is the ability of a muscle to exert repeated submaximal contractions over time. Muscular endurance can be assessed by a multitude of tests, such as a test consisting of dynamic contractions of the quadriceps muscle at a given percentage of maximal voluntary contraction at a regular pace until exhaustion.
  • One method of measuring muscle endurance is to measure functional capacity. Functional capacity can be assessed by e.g. determination of the 6-minute walking distance or the minimum time the patient spends on walking 1 kilometer.
  • Non-acylated ghrelin-like compound a ghrelin like-compound as defined herein, which does not contain an acyl group attached to any of its constitutent amino acids.
  • Individual suffering from renal failure An individual having reduced kidney function, e.g. compared to a healthy individual.
  • Renal failure is for example the inability of the kidneys to excrete waste products leading to accumulation of these waste products above normal levels in the blood.
  • Patients with mild renal failure are often asymptomatic, whereas patients with moderate to severe chronic renal failure typically present with one or more of: fatigue, anaemia, itching, loss of appetite, nausea, vomiting, weight loss, dyspnoea, fluid retention, congestive heart failure, bleeding, hypertension, hyperkalemia, acidosis, muscular cramps, nocturia, neuropathies, bleeding, stomatitis, pericarditis, osteodystrophy.
  • the clinical presentation of renal failure not only depends on the degree of impairment but also on the underlying disease and superimposed complications.
  • Renal failure can e.g. be diagnosed by determination of the plasma concentrations of creatinine and urea and by measurement of the glomerular filtration rate (GFR). Assessment of the degree of renal impairment is based on GFR; a GFR of 60-89 ml/min indicates mild renal failure, a GFR of 30-59 ml/min moderate renal failure, a GFR of 15-29 ml/min severe renal failure and a GFR ⁇ 15 ml/min end-stage renal disease.
  • GFR glomerular filtration rate
  • Chronic renal failure may be caused by primary glomerular diseases, such as IgA nephropathy, focal glomerulosclerosis, membranous nephropathy, membranoproliferative glomerulonephritis, idiopathic crescentic glomerulonephritis.
  • primary glomerular diseases such as IgA nephropathy, focal glomerulosclerosis, membranous nephropathy, membranoproliferative glomerulonephritis, idiopathic crescentic glomerulonephritis.
  • Chronic renal failure may also be caused by glomerulopathies associated with systemic disease such as diabetes mellitus, post infectious glomerulonephritis, systemic lupus erythromatosis, Wegener's granulomatosis, haemolytic ureic syndrome, amyloidosis, chronic interstitial nephropathies, hereditary nephropathies, such as polycystic kidney disease, Alport's syndrome, medullary cystic disease, nail- patella syndrome, hypertension, renal macrovascular disease (vascolupathy of the renal arteries), obstructive uropathy.
  • systemic disease such as diabetes mellitus, post infectious glomerulonephritis, systemic lupus erythromatosis, Wegener's granulomatosis, haemolytic ureic syndrome, amyloidosis, chronic interstitial nephropathies, hereditary nephropathies, such as polycystic kidney disease,
  • congenital renal diseases such as renal hypoplasia, renal dysplasia, and obstructive uropathy
  • hereditary diseases include juvenile nephritis, cystic kidney, and Alport syndrome. The most frequent metabolic causes are cystinosis and oxalosis, and the principal acquired etiology is chronic glomerulonephritis.
  • the therapeutic options in chronic renal failure include fluid and potassium restriction, diuretics, high-energy low-protein diet, erythropoietin- ⁇ , iron, resin, sodium bicarbonate or calcium carbonate (for correction of acidosis), 1- ⁇ -cholecalciferole and 1 , 25-(OH)-D3, other prokinetics.
  • Optimal control of underlying and/or concurrent diseases is essential to reduce the risk of progressive renal impairment and include antihypertensive agents (e.g. ACE inhibitors) and antidiabetic agents. Children with growth failure may benefit from growth hormone and/or IGF-1 therapy.
  • Anorexia in renal failure is assumed primarily to be caused by retension in the body fluid by one or more toxic substances as a consequence of reduced renal clearance
  • liver failure An individual having reduced liver function.
  • the severity of liver failure can, for example, be assessed by the Child-Turcotte-Pugh classification or the MELD scoring system.
  • Child-Pugh classification (sometimes termed the Child-Turcotte-Pugh score) is based on a combination of clinical and biochemical parameters.
  • the patient group treated have a Child-Pugh classification of 1 point or 2 points or 3 points.
  • the MELD scoring system was initially evaluated as a predictor of 3-month survival following placement of a transjugular intrahepatic portosystemic shunt (TIPS), but the MELD model was later confirmed to be an accurate predictor of survival in a heterogenous population of patients with end-stage liver disease.
  • This scoring system assesses severity of liver failure based on three objective variables: total bilirubin, international normalized ratio (INR) of prothrombin time, and serum creatinine.
  • the MELD score can be calculated as follows: 0.957 x loge (creatinine) + 0.378 x loge (bilirubin mg/dL) + 1.12 x loge (INR) + 0.643 x 10.
  • Polyclonal antibodies are a mixture of antibody molecules recognising a specific given antigen hence polyclonal antibodies may recognise different epitopes within said antigen.
  • Polypeptide refers to a molecule comprising amino acid residues which do not contain linkages other than amide linkages between adjacent amino acid residues.
  • a receptor is a molecule, such as a protein, glycoprotein and the like, that can specifically (non-randomly) bind to another molecule.
  • GH-Secretagogue a growth hormone secretagogue, i.e. a substance stimulating growth hormone release, such as ghrelin or a ghrelin-like compound.
  • a GH secretagogue according to the invention may for example be selected from the group of: L-692-429, L-692-585 (Benzoelactam compounds)
  • G-7203, G-7039, G-7502 (Isonipecotic acid peptidomimetic)
  • the secretagogue is a ghrelin-like compound, including 28 aa human ghrelin.
  • the secretagogue may in one embodiment be non-acylated, for instance a non-acylated form of ghrelin or a non-acylated ghrelin-like compound.
  • another bulky group such as e.g. a cholesterol or tryptophan moiety replaces the usual acyl group on said non-acylated ghrelin-like compound.
  • Surfactant molecule Molecule comprising a hydrophobic part and a hydrophilic part, i.e. molecule capable of being present in the interphase between a lipophilic phase and a hydrophilic phase.
  • the present invention is in one embodiment directed to the treatment of individuals suffering from renal failure.
  • the renal failure may be caused by a variety of factors, such as infections or immunologic causes, as well as idiopathic.
  • the invention is useful at any stage of renal failure, in particular useful in patients with end-stage renal failure and/or euremic individuals.
  • One group of patients that can be treated with the present invention are those with hypoalbumineamia who are unresponsive to dialysis and adequate prescription of dietary intake.
  • One preferred patient group is patients suffering from chronic renal failure.
  • the present invention is directed to the treatment of individuals suffering from liver failure.
  • the liver failure may be caused by a variety of factors, such as infections, alcoholic or chemical causes, as well as idiopathic. Accordingly, the liver failure may be due to alcoholic liver failure, hepatitis, such as hepatitis C and drug induced liver failure.
  • said liver failure is chronic liver failure.
  • the liver failure can be caused by e.g. alcoholic liver disease, viral hepatitis, hemochromatosis, drug- induced hepatitis, Wilson's disease, sclerosing cholangitis, primary biliary cirrhosis, Budd-Chiari syndrome, venoocclusive disease, autoimmune hepatitis, steatohepatitis, partial hepatectomy, or Reye's syndrome.
  • the individual is suffering, or at risk of suffering from, alcoholic liver failure, hepatitis C or drug-induced liver failure constitute the three major groups of patients with liver failure.
  • the individual is suffering from biliary atresia, such as childhoodbiliary atresia.
  • the individual is suffering from one or more of the following: hepatitis, Alagille's syndrome, familial intrahepatic cholestasis and metabolic diseases, such as D -1 -antitrypsin deficiency, tyrosinaemia, type 1 , Wilson's disease, cystic fibrosis, or glycogen storage disease type IV.
  • one group of patients particularly in need of treatment are those having a lean body mass of less than 80% of normal, such as less than 60% of normal and/or a body mass index below 17 kg/m2.
  • the medicament is preferably given until the lean body mass is more than 60 % of normal, preferably more than 80% of normal, more preferably more than more 90% of normal.
  • Facilitating a weight gain or facilitating maintenance of weight, in particular in individuals suffering from a pathological weight loss, is not only a matter of stimulating appetite and/or food intake but rather correcting the imbalance between energy intake and energy consumption, i.e. total body metabolism.
  • the present invention relates to the stimulation of appetite by administering a GH-secretagogue, such as a ghrelin-like compound.
  • a GH-secretagogue such as a ghrelin-like compound.
  • the stimulation of appetite may be measured using for instance a visual analog scale for measuring appetite, feeling of hunger or satiety level as described in e.g. Example 8 of published patent application WO 2005/014032 (Gastrotech Pharma: "Use of secretagogues like ghrelin in cancer cachexia and for stimulating appetite").
  • the stimulation is at least 5% compared to prior to the treatment (i.e. total appetite at least 105% of appetite prior to treatment), such as 10% higher, more preferably 20% higher or even more preferably 30%, 40% or 50% higher
  • the present invention further relates to another aspect: the stimulation of food intake by administering a GH-secretagogue, such as a ghrelin-like compound.
  • a GH-secretagogue such as a ghrelin-like compound.
  • the food intake can be measured using a multitude of techniques including self-reporting using e.g. diaries or questionnaires, measurements of calorie-intake from a buffet meal, using weighing of food prior to ingestion, or weighing and analysis of paired quantities of food.
  • the food intake may be measured on a meal basis, a daily basis, a weekly basis or a monthly basis.
  • the treatment results in at least a 1% increase in food intake, such as an increase of at least 2%, more preferably least 3%, or least 5% or 7%, and even more preferred least 10% above average food intake prior to initiation of treatment.
  • the treatment leads to increase in calorie intake irrespective of changes in food intake, since amount of food ingested may not be directly related to the ingested calorie intake, as the various food items such as fat, carbohydrates and proteins, contain different amounts of calories per amount food.
  • the treatment results in an least 1% increase in calorie intake, such as an increase of least 2%, more preferably least 3%, or least 5% or 7%, and even more preferred 10% in calorie intake.
  • the present invention relates to stimulation of weight gain, reducing a weight loss or maintaining a stable body-weight by administering a ghrelin-like compound to an individual suffering from renal failure and/or liver failure.
  • a ghrelin-like compound is useful for stimulating food intake and weight gain
  • the secretagogue such as a ghrelin-like compound
  • the secretagogue is useful for stimulating weight gain, for reducing a weigth loss or for maintaining stable body weight.
  • the GH-secretagogue such as a ghrelin-like compound
  • a meal such as within 180 minutes prior to a meal, such as within 150 minutes prior to a meal, for example within 120 minutes prior to a meal, such as within 100 minutes prior to a meal, for example within 80 minutes prior to a meal, such as within 60 minutes prior to a meal, for example within 45 minutes prior to a meal, such as within 30 minutes prior to a meal, for example within 15 minutes prior to a meal.
  • the present invention is useful for treatment of under weight subjects, or for preventing loss of weight to a stage of under weight.
  • Under weight subjects include those having a body weight about 3% or less, 5% or less, 10% or less, 20% or less, or 30% or less, than the lower end of "normal” weight range or Body Mass Index ("BMI").
  • BMI Body Mass Index
  • "Normal” weight ranges are well known in the art and take into account factors such as a patient age, height, and body type.
  • the invention is suitable for treating patients who have experienced an involuntary weight-loss prior to commencement of treatment, such as a weight-loss of 1% or less per month, 2% or less per month, or 5% or less per 6 months.
  • an increase in the body fat mass of an individual can be readily assessed by the skilled person using a number of state of the art techniques.
  • the invention relates to an increase in body fat mass without the individual gaining weight overall.
  • a preferred embodiment of the invention leads to an increase in body fat of at least 1%, such as at least 2% compared to prior to the initiation of treatment, more preferably at least 4%, such as at least 5%, and at least 8% and at least 10 %, even more preferably at least 20% or at least 40% above pre-treatment values.
  • the invention leads to an increase in lean body mass of at least 1%, such as at least 2% compared to prior to the initiation of treatment, more preferably at least 3%, such as at least 4%, such as at least 5%, such as at least 6%, such as at least 7%, such as at least 8% or at least 10 %, or least 15% above pre-treatment values.
  • administration of a GH secretagogue may be used as part of the "supportive care regimen" for treatment of a patient in need thereof, e.g. to encourage intake of food and/or counteract any metabolic changes caused by the patient's state and/or therapy, and/or increase the patients functional status and/or increase the patient's life quality.
  • Such patients in need thereof include, but are not restricted to, transplant patients (both before and after transplant, e.g. liver or kidney transplant) and patients having undergone, or who are going to undergo, a major operative procedure.
  • said GH secretagogue is administered as supportive care to optimise nutritional status in a patient preparing to undergo, undergoing or having undergone an anorectic therapy, such as cytoxic therapy, such as cytotoxic chemotherapy, more preferably acting to increase or maintain lean body mass.
  • anorectic therapy such as cytoxic therapy, such as cytotoxic chemotherapy
  • the administration of a GH secretagogue as part of a supportive care regimen may be started up to a month before, such as two weeks before, for example a week before, the primary treatment (such as anorectic therapy or major operation) is due to start, with the GH secretagogue being administered e.g. 1-2 times daily.
  • GH secretagogue administration may preferably act to counteract loss of lean body mass, such as acting to increase the individual's lean body mass, and/or increasing the individual's percentage lean body mass.
  • the invention leads to an increase in muscle strength and/or muscle perseverance, e.g. measured as the maximal distance the patient is capable to walk in 5 minutes, the minimum time the patient spends on walking 1 kilometer, the length of time the patient can walk at maximum pace, grip strength, knee extension power.
  • the increase is preferably at least 5% compared to prior to initation of treatment, such as at least 8% compared to prior to initiation of treatment, such as at least 12% compared to prior to initation of treatment, such as at least 20%, or at least 50% compared to prior to initiation of treatment.
  • grip strength the increase is preferably at least 10%, such as at least 20%, such as at least 30%, such as at least 50%, such as at least 75%, such as at least 100% compared to prior to initiation of the treatment.
  • the treatment method and/or pharmaceutical compositions and/or compounds of the present invention are capable of affording the individual thus treated an improved quality of life (QOL), for example as is caused by improved appetite and/or body weight and/or nutritional status.
  • QOL quality of life
  • the invention relates to improvements of Quality of Life using a secretagogue, such as ghrelin or a ghrelin-like compound as described herein.
  • said improvement in an individual's life quality is assessed using a "Quality of life" questionnaire, as is known to one skilled in the art.
  • SF-36 Medical Outcomes Study Short-Form Health Survey
  • the SF-36 contains 36 questions that assess eight aspects of the patients' QOL; physical functioning (PF), role-physical functioning (RP), bodily pain (BP), general health (GH) 1 vitality (VT), social functioning (SF), role emotional functioning (RE), and mental health (MH).
  • PF physical functioning
  • RP role-physical functioning
  • BP bodily pain
  • GH general health
  • VT general health
  • VT general health
  • SF role emotional functioning
  • MH mental health
  • responses to questions within scales are summed and linearly transformed to scale scores that range from 0, representing poor health status, to 100, representing optimal health status.
  • the Swedish version has been validated and normative data have been presented for the general Swedish population (Sullivan MKJ, Ware J. Ha ⁇ soenkat: svensk manual och tolkningsguide (SF-36 Health Survey. Swedish manual and interpretation guide).
  • G ⁇ teborg Sahlgrenska University Hospital; 1994.
  • EORTC QLQ-C30 (+3) questionnaire.
  • the EORTC QLQ-C30 (version 1.0) is a 30 item core questionnaire intended for assessment of QOL among patients, the instrument is developed by the EORTC Quality of Life Study group. The first version has been validated in cancer patients and reference data from general populations have been published. The questionnaire comprises five functional scales; physical functioning (five questions), role functioning (two questions), emotional functioning (four questions), cognitive functioning (two questions) and social functioning (two questions). There are three symptom scales; fatigue (three questions), nausea and vomiting (two questions) and pain (two questions), and there are six single items on dyspnoea, insomnia, loss of appetite, constipation, diarrhea and financial difficulties.
  • Example 8 of published patent application WO 2005/014032 (Gastrotech Pharma: "Use of secretagogues like ghrelin in cancer cachexia and for stimulating appetite"): "Examples of questionnaires assessing patient quality of life: A) EORTC QLQ-C30 B) Taste questionnaire
  • treatment of patients with the described conditions results in a significant improvement in the patient's quality of life.
  • the treatment results in a significant increase in quality of life as measured using any method for testing the quality of life including, but not limited to, the above mentioned questionnaires, e.g. an increase in the quality of life score(s), or a composite quality of life score, as appropriate for the individual measuring tool, or a decrease in score(s) related to the symptoms and/or problems, respectively.
  • This increase or decrease, respectively is preferably 1 % above the score obtained prior to initiation of the treatment, more preferably 2% above, even more preferred 5%, such as 10%, even more preferred 20%, 50% or 75% above the pre-treatment score.
  • the treatment results in measurable increases in quality of life score such that the score after treatment is equal to the average score found in a comparable healthy subject pool, or close to such a "normal" score, i.e. more than 50% of the score, even more preferably 60% of the score, or more preferably 75% of the score.
  • the treatment results in a decrease in the score(s) related to the symptoms and/or problems of at least 1 %, more preferably 3%, even more preferably 5% or more preferred 10%, 20%, 30% or 50% of the score(s) prior to initiation of treatment.
  • These increases or reductions, respectively may refer to one, several, or all of the aspects of the individual quality of life measuring tool, or a composite score when appropriate.
  • GH-secretagogue such as ghrelin or a ghrelin-like compound
  • ghrelin-like compound Any GH-secretagogue, such as ghrelin or a ghrelin-like compound, may be used in the present invention.
  • One preferred type of ghrelin-like compound according to the invention described herein is a compound comprising a structure defined by formula I:
  • Z 1 is an optionally present protecting group each X 1 is independently selected from an amino acid, wherein said amino acid is selected from naturally occurring and synthetic amino acids,
  • X 2 is any amino acid selected from naturally occurring and synthetic occurring amino acids, said amino acid being modified with a bulky hydrophobic group, preferably an acyl group, or a fatty acid,
  • each X 3 is independently selected from an amino acid, wherein said amino acid is selected from naturally occurring and synthetic amino acids,
  • X 1 and X 3 optionally may be modified by a bulky hydrophobic group, preferably an acyl group, or a fatty acid,
  • Z 2 is an optionally present protecting group
  • n is an integer in the range of from 1-10
  • n is 0 or an integer in the range of from 1-35.
  • the term "GH-secretagogue” includes the naturally occurring 28 aa human ghrelin, the amino acid of which is shown in SEQ ID NO: 1 , as well as the naturally occurring 27 aa human ghrelin, the amino acid of which is shown in SEQ ID NO: 2.
  • the present invention relates to the use of ghrelin or a peptide homologous thereto. Ghrelin is described by Kojima in Nature (1999), vol. 402,656- 660.
  • the present invention includes diastereomers as well as their racemic and resolved enantiomerically pure forms.
  • Secretagogues can contain D-amino acids, L-amino acids, alpha-amino acid, beta-amino acid, gamma-amino acid, natural amino acid and synthetic amino acid or the like or a combination thereof.
  • amino acids present in a ghrelin-like compound are the L-enantiomer.
  • the ghrelin-like compound preferably comprises an amino acid modified with a bulky hydrophobic group.
  • the number of amino acids N-terminally to the modified amino acid is preferably within the range of from 1-9.
  • m is preferably an integer in the range of from 1-9, such as of from 1-8, such as of from 1-7, such as of from 1-6, such as of from 1-5, such as of from 1-4, such as of from 1-3, such as of from 1-2, such as 2.
  • the number of amino acids N-terminally to the modified amino acid is low, such as of from 1-3, such as of from 1-2. Most preferably 2 amino acids are positioned N-terminal to the modified amino acid.
  • (X 1 ) m has a GIy residue in the N-terminal part of the sequence. Accordingly, in preferred embodiment (X 1 ) m is selected from the sequences:
  • GIy Gly-Ser, Gly-Cys, Gly-Lys, Gly-Asp, GIy-GIu, Gly-Arg, Gly-His, Gly-Asn, GIy- GIn, Gly-Thr, and Gly-Tyr.
  • (X 1 ) m is Gly-Ser or Gly-Cys, most preferably Gly-Ser.
  • the ghrelin-like compound is selected from a compound of
  • the ghrelin-like compound has formula III.
  • X 2 may be any amino acid modified with a bulky hydrophobic group.
  • X 2 is selected from the group of modified Ser, Cys, Asp, Lys, Trp, Phe, lie, and Leu. More preferably X 2 is selected from the group of modified Ser, modified Cys and modified Lys, and most preferably X 2 is modified Ser.
  • (X 1 ) m - (X 2 ) is preferably Gly-Xaa-Ser*, or Gly-Xaa-Cys * , wherein Xaa is any amino acid, more preferably (X 1 ) m - (X 2 ) is Gly-Ser-Ser * , or Gly-Ser-Cys * , wherein * indicates that the amino acid residue is modified with a bulky hydrophobic group.
  • (X 3 ) n preferably comprises a sequences which is a fragment of ghrelin, such as human ghrelin. Accordingly, (X 3 ) n preferably comprises a sequence selected from one or more of the sequences shown below:
  • n is preferably an integer in the range of from 1-25, such as of from 1-24, such as from 1-15, such as of from 1-10, or such as of from 10-25, such as of from 10-24, such as of from 15-25, such as of from 15-24.
  • n may equally be e.g. any of: 5-9, 5-24, 5-20, 9-25, 9-18, 9-12, 3-25, 3-22, 3-15, 3-10 etc.
  • a ghrelin-like compound includes the naturally occurring 28 aa human ghrelin, the amino acid of which is shown in SEQ ID NO: 1, as well as the naturally occurring 27 aa human ghrelin, the amino acid of which is shown in SEQ ID NO: 2.
  • (X 3 ) n may be selected from any fragment of ghrelin, such as human ghrelin, and accordingly, (X 3 ) n may be selected from one or more of the sequences shown below or a homologue thereof: Phe Leu Ser Pro GIu His GIn Arg VaI GIn GIn Arg Lys GIu Ser Lys Lys Pro Pro Ala Lys Leu GIn Pro Arg
  • (X 3 ) n comprises or consists of a sequence selected from the sequences
  • GH secretagogues for use in the present invention are disclosed in PCT patent application no. PCT/DK2004/000529, Danish patent application no. PA 200401875, and PCT applications with publication numbers WO0192292 (Merck and Co. Inc), WO0134593 (Novo Nordisk AS) and WO0107475 ("Novel peptides", Kangawa et al.); said documents all being incorporated herein by reference.
  • Methods for production of GH secretagogues are well known to thoese skilled in the art, for example in Example 2 of PCT patent application PCT/DK2004/000519 (Gastrotech Pharma), incorporated herein by reference.
  • the GH secretagogues used herein are active at the receptor for GHS as described above, i.e. the receptor GHS-R 1a.
  • the compounds can bind to the receptor, and preferably, stimulate receptor activity.
  • the receptor activity can be measured using different techniques such as detecting a change in the intracellular conformation of the receptor, in the G-protein coupled activities, and/or in the intracellular messengers.
  • One simple measure of the ability of a ghrelin like compound to activate the ghrelin receptor is to measure its EC50, i.e. the dose at which the compound is able to activate the signalling of the receptor to half of the maximal effect of the compound.
  • the receptor can either be expressed endogenously on primary cells cultures, for example pituitary cells, or heterologously expressed on cells transfected with the ghrelin receptor.
  • Whole cell assays or assays using membranes prepared form either of these cell types can be used depending on the type of assay.
  • any suitable assay which monitor activity in the Gq/G11 signalling pathway can be used, for example:
  • an assay measuring the activation of Gq / G11 performed for example by measurement of GTPgS binding combined with, e.g., anti-G-alpha-q or -11 antibody precipitation in order to increase the signal to noise ratio.
  • This assay may also detect coupling to other G-proteins than Gq/11.
  • PLC phopholipase C
  • a ghrelin-like compound used in the invention preferably has at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, functional activity relative to 28 aa human ghrelin as determined using the assay described herein above, and/or an EC50 greater than about 1 ,000 nM, greater than about 100 nM, or greater than about 50 nM, or greater than about 10 nM or greater than 1 nM. Greater refers to potency and thus indicates a lesser amount is needed to achieve binding inhibition.
  • the compound has a potency (EC50) on the GHS-R 1A of less than 500 nM. In another embodiment the compound has a potency (EC50) on the GHS-R 1A of less than 100 nM, such as less than 80 nM, for example less than 60 nM, such as less than 40 nM, for example less than 20 nM, such as less than 10 nM, for example less than 5 nM, such as less than 1 nM, for example less than 0.5 nM, such as less than 0.1 nM, for example less than 0.05 nM, such as less than 0.01 nM.
  • the dissociation constant (Kd) of the compound is less than 500 nM. In a still further embodiment the dissociation constant (Kd) of the ligand is less than 100 nM, such as less than 80 nM, for example less than 60 nM, such as less than 40 nM, for example less than 20 nM, such as less than 10 nM, for example less than 5 nM, such as less than 1 nM, for example less than 0.5 nM, such as less than 0.1 nM, for example less than 0.05 nM, such as less than 0.01 nM.
  • Binding assays can be performed using recombinantly-produced receptor polypeptides present in different environments.
  • environments include, for example, cell extracts and purified cell extracts containing the receptor polypeptide expressed from recombinant nucleic acid or naturally occurring nucleic acid; and also include, for example, the use of a purified GHS receptor polypeptide produced by recombinant means or from naturally occurring nucleic acid which is introduced into a different environment.
  • the receptor can be expressed in a cell line such as HEK 293, COS 7, and CHO not normally expressing the receptor by an expression vector, wherein the same cell line without the expression vector can act as a control.
  • a GH secretagogue may be demonstrated in mammals using e.g. the test for the absolute bioavailability of iv administered Ghrelin and sc administered Ghrelin described in Example 3 of PCT patent application PCT/DK2004/000519 (Gastrotech Pharma), or the method in Example 6 "Efficacy of subcutaneous administration of Ghrelin" from PCT patent application PCT/DK2004/000519 (Gastrotech Pharma), or the trial described in Example 11 "Administering ghrelin to cancer cachexia patients" from PCT patent application PCT/DK2004/000519 (Gastrotech Pharma). All these Examples are hereby incorporated herein by reference
  • identity shall be construed to mean the percentage of amino acid residues in the candidate sequence that are identical with the residue of a corresponding sequence to which it is compared, after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known in the art. Sequence identity may be measured using sequence analysis software (e.g., Sequence Analysis Software Package, Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Ave., Madison, Wis. 53705). This software matches similar sequences by assigning degrees of homology to various substitutions, deletions, and other modifications.
  • sequence analysis software e.g., Sequence Analysis Software Package, Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Ave., Madison, Wis. 53705
  • a ghrelin homologue of one or more of the sequences specified herein may vary in one or more amino acids as compared to the sequences defined, but is capable of performing the same function, i.e. a homologue may be envisaged as a functional equivalent of a predetermined sequence.
  • a ghrelin homologue is preferably a ghrelin-like compound as defined above.
  • homologue of any of the predetermined sequences herein may be defined as:
  • homologues comprising an amino acid sequence capable of being recognised by an antibody, said antibody also recognising the 28 aa human ghrelin, preferably the acylated 28 aa human ghrelin, and/or
  • homologues comprising an amino acid sequence capable of binding selectively to GHS-R 1a, and/or
  • homologues having a substantially similar or higher binding affinity to GHS-R 1a than the 28 aa human ghrelin, preferably the acylated 28 aa human ghrelin.
  • the 28 aa human ghrelin has the sequence shown in SEQ ID NO: 1 , and when acylated is acylated in position 3.
  • the antibodies used herein may be antibodies binding the N-terminal part of ghrelin or the C-terminal part of ghrelin, preferably the N-terminal part of ghrelin.
  • the antibodies may be antibodies as described in Ariyasu et al. "Delayed short-term secretory regulation of ghrelin in obese animals: Evidenced by a specific RIA for the active form of ghrelin, Endocrinology 143(9):3341-3350, 2002.
  • Examples of homologues comprises one or more conservative amino acid substitutions including one or more conservative amino acid substitutions within the same group of predetermined amino acids, or a plurality of conservative amino acid substitutions, wherein each conservative substitution is generated by substitution within a different group of predetermined amino acids.
  • Homologues may thus comprise conservative substitutions independently of one another, wherein at least one glycine (GIy) of said homologue is substituted with an amino acid selected from the group of amino acids consisting of Ala, VaI, Leu, and lie, and independently thereof, homologues, wherein at least one of said alanines (Ala) of said homologue thereof is substituted with an amino acid selected from the group of amino acids consisting of GIy, VaI, Leu, and lie, and independently thereof, homologues, wherein at least one valine (VaI) of said homologue thereof is substituted with an amino acid selected from the group of amino acids consisting of GIy, Ala, Leu, and lie, and independently thereof, homologues thereof, wherein at least one of said leucines (Leu) of said homologue thereof is substituted with an amino acid selected from the group of amino acids consisting of GIy, Ala, VaI, and He, and independently thereof, homologues thereof, wherein at least one isoleucine (lie)
  • Conservative substitutions may be introduced in any position of a preferred predetermined sequence. It may however also be desirable to introduce non- conservative substitutions, particularly, but not limited to, a non-conservative substitution in any one or more positions.
  • a non-conservative substitution leading to the formation of a functionally equivalent homologue of the sequences herein would for example i) differ substantially in polarity, for example a residue with a non-polar side chain (Ala, Leu, Pro, Trp, VaI, lie, Leu, Phe or Met) substituted for a residue with a polar side chain such as GIy, Ser, Thr, Cys, Tyr, Asn, or GIn or a charged amino acid such as Asp, GIu, Arg, or Lys, or substituting a charged or a polar residue for a non-polar one; and/or ii) differ substantially in its effect on polypeptide backbone orientation such as substitution of or for Pro or GIy by another residue; and/or iii) differ substantially in electric charge, for example substitution of a negatively charged residue such as GIu or Asp for a positively charged residue such as Lys, His or Arg (and vice versa); and/or iv) differ substantially in steric bulk,
  • Substitution of amino acids may in one embodiment be made based upon their hydrophobicity and hydrophilicity values and the relative similarity of the amino acid side-chain substituents, including charge, size, and the like.
  • Exemplary amino acid substitutions which take various of the foregoing characteristics into consideration 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 binding domain comprises a homologue having an amino acid sequence at least 60 % homologous to SEQ ID NO 1.
  • the homology is at least 65 %, such as at least 70 % homologous, such as at least 75 % homologous, such as at least 80 % homologous, such as at least 85 % homologous, such as at least 90 % homologous, such as at least 95 % homologous, such as at least 98 % homologous to SEQ ID NO:1.
  • the percentages mentioned above relates to the identity of the sequence of a homologue as compared to SEQ ID NO:1.
  • Homologues to SEQ ID NO:1 may be 27 aa human ghrelin SEQ ID NO: 2, or rat ghrelin SEQ ID NO:3.
  • Other homologues are the variants described in EP 1197496 (Kangawa) and WO 01/92292 (Merck) and WO 01/56592 (Novo Nordisk) incorporated herein by reference.
  • the bulky hydrophobic group of the secretagogue according to the invention is any bulky hydrophobic group capable of providing the des-acylated 28 aa human ghrelin, or an analogue thereof, with binding affinity to GHS-R 1a.
  • Any suitable amino acid may be modified with any suitable bulky hydrophobic group; in a preferred embodiment, a Ser residue (preferably amino acid number 3 in the amino acid chain) is modified with the bulky hydrophobic group.
  • the amino acid being modified contains e.g. - OH, -SH, -NH or -NH 2 as a substituent group in a side chain thereof
  • a group formed by acylating such a substituent group is preferred.
  • the mode of linkage may thus be selected from the group consisting of ester, ether, thioester, thioether, amide and carbamide.
  • the modified amino acid is serine, threonine, tyrosine or oxyproline
  • the amino acid has a hydroxy! group in the side chain.
  • the modified amino acid is cysteine
  • the amino acid has a mercapto group in the side chain.
  • the modified amino acid is lysine, arginine, histidine, tryptophan, proline oroxyproline, it has an amino group or imino group in the side chain.
  • the hydroxyl group, mercapto group, amino group and imino group described above may thus have been chemically modified. That is, the hydroxyl group or mercapto group may be etherized, esterified, thioetherified or thioesterified.
  • the imino group may have been iminoetherified, iminothioetherified or alkylated.
  • the amino group may have been amidated, thioamidated or carbamidated.
  • the mercapto group may have been disulfidated, the imino group may have been amidated or thioamidated, and the amino group may have been alkylated or thiocarbamidated.
  • the modified amino acid is Ser coupled through an ester linkage to the hydrophobic group.
  • the hydrophobic group may be any group with a saturated or unsaturated alkyl or acyl group containing one or more carbon atoms.
  • the bulky hydrophobic group is an acyl group, including groups formed by removing a hydroxyl group from an organic carboxylic acid, organic sulfonic acid or organic phosphoric acid.
  • the organic carboxylic acid includes e.g. fatty acids, and the number of carbon atoms thereof is preferably 1 to 35. In the organic sulfonic acid or organic phosphoric acid, the number of carbon atoms thereof is preferably 1 to 35.
  • the acyl group is preferably selected from a C1-C35 acyl group, such as a C1 - C20 acyl group, such as a C1 - C15 acyl group, such as a C6 - C15 acyl group, such as a C6 - C12 acyl group, such as a C8 - C12 acyl group.
  • the acyl group is selected from the group of C7 acyl group, C8 acyl group, C9 acyl group, C10 acyl group, C11 acyl group, and C12 acyl group.
  • Such acyl group may be formed from octanoic acid (preferably caprylic acid), decanoic acid (preferably capric acid), or dodecanoic acid (preferably lauric acid), as well as monoene or polyene fatty acids thereof.
  • the acyl group is selected from the group of C8 acyl group, and C10 acyl group.
  • Such acyl groups may be formed from octanoic acid (preferably caprylic acid), or decanoic acid (preferably capric acid).
  • the acyl group is selected from the group of C7 acyl group, C9 acyl group, and C11 acyl group, such as from the group of C9 acyl group and C11 acyl group.
  • modified amino acid may be any amino acid wherein a group is modified as described in EP 1 197 496 (Kangawa), which is hereby incorporated by reference.
  • the ghrelin-like compound according to the invention may comprise a protecting group at the N-terminus or the C-terminus or at both.
  • a protecting group covalently joined to the N-terminal amino group reduces the reactivity of the amino terminus under in vivo conditions.
  • Amino protecting groups include - C1-10 alkyl, -C1-10 substituted alkyl, -C2-10 alkenyl, -C2-10 substituted alkenyl, aryl, -C1-6 alkyl aryl, -C(O)- (CH2) 1-6-COOH, -C(O)-CI -6 alkyl, -C(O)-aryl,
  • the amino terminus protecting group is acetyl, propyl, succinyl, benzyl, benzyloxycarbonyl or tbutyloxycarbonyl.
  • a protecting group covalently joined to the C-terminal carboxy group reduces the reactivity of the carboxy terminus under in vivo conditions.
  • the carboxy terminus protecting group is preferably attached to the a-carbonyl group of the last amino acid.
  • Carboxy terminus protecting groups include amide, methylamide, and ethylamide.
  • the secretagogue such as a ghrelin-like compound, to be used in the present invention may be provided in the form of a secretagogue conjugate, i.e. a molecule comprising the secretagogue conjugated to another entity.
  • the other entity may be any substance that is capable of conferring improved properties to the secretagogue, e.g. in terms of improved stability, half-life, etc.
  • the secretagogue may be conjugated to a peptide, such as a peptide having effect on nociceptin receptor ORL1.
  • the conjugate is a conjugate of ghrelin or a derivative or homologue thereof and a peptide having effect on ORL1 , e.g. the peptide AC-RYY(RK)(WI)RK)-NH 2 , where the brackets show allowable variation of amino acid residues. Examples of other suitable peptides are found in US patent applications 2003040472 and US2002004483, and US patent 5869046.
  • a secretagogue such as ghrelin or a ghrelin-like compound
  • the polymer molecule may be any suitable polymer molecule, such as a natural or synthetic polymer, typically with a molecular weight in the range of about 1-100 kDa, such as about 3-20, kDa, e.g. 5-10 kDa.
  • the polymer is attached to a reactive group present on the secretagogue, e.g. an amine group or a thiol group.
  • suitable polymer molecules include polymer molecules selected from the group consisting of polyalkylene oxide (PAO), including polyalkylene glycol (PAG), such as linear or branched polyethylene glycol (PEG) and polypropylene glycol (PPG), poly-vinyl alcohol (PVA), poly-carboxylate, poly-(vinylpyrolidone), polyethylene-co-maleic acid anhydride, polystyrene-co-maleic acid anhydride, dextran, including carboxymethyl-dextran.
  • PAO polyalkylene oxide
  • PAG polyalkylene glycol
  • PEG polyalkylene glycol
  • PPG polypropylene glycol
  • PVA poly-vinyl alcohol
  • PVA poly-carboxylate
  • poly-(vinylpyrolidone) polyethylene-co-maleic acid anhydride
  • polystyrene-co-maleic acid anhydride dextran, including carboxymethyl-dextran
  • the polymer molecule is a PEG
  • polymer molecules can be activated by conventional methods known in the art, e.g., as disclosed in WO 90/13540.
  • activated PEG polymers include the following linear PEGs: NHS-PEG (e.g., SPA- PEG, SSPA-PEG, SBA-PEG, SS-PEG, SSA-PEG, SC-PEG, SG-PEG, and SCM- PEG), and NOR-PEG), BTC-PEG, EPOX-PEG, NCO-PEG, NPC-PEG, CDI-PEG, ALD-PEG, TRES-PEG, VS-PEG, IODO-PEG, and MAL-PEG, and branched PEGs such as PEG2-NHS and those disclosed in U.S. Pat. No. 5,932,462 and U.S. 5,643,575, both of which are incorporated herein by reference.
  • linear PEG e.g., SPA- PEG, SSPA-PEG, SBA-PEG, SS-PEG, SSA-PEG, SC-PEG, SG-PEG, and SCM- PEG
  • the PEGylation i.e. conjugation of the secretagogue polypeptide and the activated polymer molecule
  • PEGylation is conducted in accordance with established procedures, e.g., as described in the following references (which also describe suitable methods for activation of polymer molecules): R. F. Taylor, (1991 ), “Protein immobilisation. Fundamental and applications", Marcel Dekker, N.Y.; S. S. Wong, (1992), “Chemistry of Protein Conjugation and Crosslinking", CRC Press, Boca Raton; G. T. Hermanson et al., (1993), “Immobilized Affinity Ligand Techniques", Academic Press, N. Y.).
  • linker it is also contemplated according to the invention to couple the polymer molecules to the secretagogue through a linker.
  • Suitable linkers are well known to the skilled person.
  • a preferred example is cyanuric chloride (Abuchowski et al., (1977), J. Biol. Chem., 252, 3578-3581 ; U.S. Pat. No. 4,179,337; Shafer et al., (1986), J. Polym. Sci. Polym. Chem. Ed., 24, 375-378.
  • the secretagogue is conjugated to an oligosaccharide molecule, such as dextran, glycan, transferrin, etc.
  • an oligosaccharide molecule such as dextran, glycan, transferrin, etc.
  • conjugation may be achieved in accordance with established technologies, e.g. those available from Neose Technologies, Inc. Horsham, PA.
  • the secretagogue is conjugated to an Fc region of an IgG molecule, typically in the form of a fusion protein.
  • a salvage receptor binding epitope of the Fc region of an IgG i.e. the Fc portion of an immunoglobulin of the isotype IgG
  • This can take place by any means, such as by mutation of the appropriate region in the secretagogue to mimic the Fc region or by incorporating the epitope into a peptide tag that is then fused to the secretagogue at either end or in the middle or by DNA or peptide synthesis.
  • the salvage receptor binding epitope is any suitable such epitope as known to the person skilled in the art, and its nature will depend, e.g., on the type of secretagogue being modified.
  • the epitope is introduced into the secretagogue such that the biological activity of the secretagogue is maintained, i.e., the epitope does not adversely affect the conformation of the secretagogue or affect its binding to ligands that confers its biological activity.
  • the secretagogue may be modified to include suitable reactive groups, whereby the thus modified secretagogue is capable of forming a conjugate in vivo (after having been administered to an individual) through covalent bonding with available reactive functionalities on blood components.
  • the invention also relates to such modified secretagogues, and methods for their use.
  • the invention relates to conjugates formed in vitro between a modified secretagogue as described above and a blood component.
  • the conjugates formed in accordance with this embodiment are contemplated to have an increased in vivo half life as compared to the corresponding non-modified secretagogue.
  • the secretagogue is modified with a chemically reactive group (reactive entity).
  • the reactive entity may, e.g., be selected from the wide variety of active carboxyl groups, particularly esters, where the hydroxyl moiety is physiologically acceptable.
  • Such groups may be selected from the group consisting of N-hydroxysuccinimide (NHS), N-hydroxy-sulfosuccinimide (sulfo-NHS), maleimide-benzoyl-succinimide (MBS), gamma-maleimido-butyryloxy succinimide ester (GMBS) and maleimidopropionic acid (MPA).
  • the principal targets for this group of entities are primary amines on the blood component.
  • Another group of active entities is constituted by a maleimido-containing group such as MPA and gamma-maleimide-butrylamide (GMBA) Such groups react with thiol groups present on the blood component.
  • the blood component with which the modified secretagogue is designed to react may be any blood component having an available target group, e.g. an amine or a thiol group, and which is suitable as a carrier for binding the modified secretagogue in vivo and thereby extend the circulating half-life thereof.
  • blood components are serum albumin and IgG.
  • the covalent bonding of a modified secretagogue to a blood component may be achieved in vivo by administration of the modified secretagogue directly to the patient.
  • the administration may be done in any suitable form, such as in the form of a bolus or introduced slowly over time by infusion using metered flow or the like.
  • the secretagogue/blood component conjugate may also be prepared ex vivo by combining blood with the modified secretagogue, allowing covalent bonding of the modified secretagogue to reactive functionalities on blood components and then returning or administering the conjugated blood to the individual.
  • the above may also be accomplished by first purifying an individual blood component or limited number of components, such as red blood cells, immunoglobulins, serum albumin, or the like, and combining the component or components ex vivo with the chemically reactive secretagogues.
  • an individual blood component or limited number of components such as red blood cells, immunoglobulins, serum albumin, or the like.
  • GH-secretagogues such as ghrelin-like compounds
  • a polypeptide region of a ghrelin-like compound can be chemically or biochemically synthesized and modified. Techniques for chemical synthesis of polypeptides are well known in the art. (See e. g., Vincent in Peptide and Protein Drug Delivery, New York, N. Y., Dekker, 1990.).
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a GH secretagogue (or pharmaceutically acceptable salt thereof) suitable for use in the present invention.
  • the pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier, vehicle and/or excipient.
  • the carrier, vehicle and/or excipient should be compatible with the GH secretagogue or salt thereof.
  • the pharmaceutical composition is not immunogenic when administered to a human in accordance with the present invention.
  • compositions, carriers, diluents and reagents are used interchangeably and represent that the materials are capable of administration to or upon a human without the production of undesirable physiological effects such as nausea, dizziness, gastric upset and the like.
  • compositions that contains active ingredients dissolved or dispersed therein are well understood in the art.
  • compositions are prepared as sterile injectables either as liquid solutions or suspensions, aqueous or non-aqueous, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared.
  • the preparation can also be emulsified.
  • Suitable pharmaceutical carriers include sterile aqueous solution and various organic solvents and inert solid diluents or fillers.
  • solid carriers are lactose, terra alba, sucrose, cyclodextrin, talc, gelatine, agar, pectin, acacia, magnesium stearate, stearic acid or lower alkyl ethers of cellulose.
  • liquid carriers are syrup, peanut oil, olive oil, phospholipids, fatty acids, fatty acid amines, polyoxyethylene or water.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient.
  • the formulation has a pH within the range of 3.5-8, such as in the range 4.5-7.5, such as in the range 5.5-7, such as in the range 6-7.5, preferably around 7.3.
  • the pH range may be adjusted according to the individual treated and the administration procedure. For example, some GH secretagogues may be easily stabilised at a lower pH, so in another preferred embodiment of the the invention the formulation has a pH within the range 3.5-7, such as 4-6, such as 5-6, such as 5.3-5.7, such as 5.5.
  • Liquid compositions can also contain liquid phases in addition to and to the exclusion of water.
  • additional liquid phases are glycerin, vegetable oils such as cottonseed oil, organic esters such as ethyl oleate, and water-oil emulsions.
  • the pharmaceutical composition can include a pharmaceutically acceptable salt of the GH secretagogue therein.
  • the salt will be one which is acceptable in its therapeutic use. By that it is meant that the salt will retain the biological activity of the GH secretagogue and the salt will not have untoward or deleterious effects in its application and use in treating diseases.
  • GH secretagogue is a base it is treated with an excess of an organic or inorganic acid in a suitable solvent. If the GH secretagogue is an acid, it is treated with an inorganic or organic base in a suitable solvent.
  • the pharmaceutically acceptable salt may be an acid addition salts including salts of inorganic acids as well as organic acids. Acid addition salts are formed with free amino groups of the GH secretagogue.
  • suitable inorganic acids include hydrochloric, hydrobromic, hydriodic, metaphosphoric, phosphoric, sulpfuric and nitric acids and the like.
  • suitable organic acids include formic, acetic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, citric, fumaric, glycolic, lactic, maleic, malic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methanesulfonic, ethanesulfonic, tartaric, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, aspartic, stearic, palmitic, ethylenediaminetetraacetic (EDTA), p-aminobenzoic, glutamic, benzenesulfonic and ptoluenesulfonic acids and the like.
  • EDTA ethylenediaminetetraacetic
  • the metal salt may be an alkali metal or earth alkali metal salt.
  • metal salts include lithium, sodium, potassium and magnesium salts and the like.
  • ammonium and alkylated ammonium salts include ammonium, methylammonium, dimethylammonium, trimethylammonium, ethylammonium, hydroxyethylammonium, diethylammonium, butylammonium and tetramethylammonium salts and the like.
  • Salts formed with the free carboxyl groups can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like.
  • inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides
  • organic bases such as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like.
  • hydrate hydrated form thereof.
  • the pharmaceutical composition comprises at least one acylated GH secretagogue, or a pharmaceutically acceptable salt thereof, in combination with a desacylated Ghrelin-like compound, or a pharmaceutically acceptable salt thereof, such as any of the desacylated ghrelin-like compounds described in WO03051389 (Theratechnologies: "Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof), incorporated herein by reference.
  • the invention relates to the use of a pharmaceutical composition
  • a pharmaceutical composition comprising a mixture of at least two different GH-secretagogues (such as e.g. two different ghrelin-like compounds), such as a mixture of a ghrelin-like compound being acylated with a C8 acyl and a ghrelin-like compound being acylated with a C10 acyl.
  • GH-secretagogues such as e.g. two different ghrelin-like compounds
  • the pharmaceutical composition used comprises one or more acylated GH secretagogue(s) (preferably ghrelin-like compound(s)), optionally compounds having different acyl chain lengths preferably selected from the group of C7 acyl group, C9 acyl group, and C11 acyl group, such as from the group of C9 acyl group and C11 acyl group, further optionally in combination with a desacylated secretagogue, such as a desacylated Ghrelin-like compound.
  • acylated GH secretagogue(s) preferably ghrelin-like compound(s)
  • optionally compounds having different acyl chain lengths preferably selected from the group of C7 acyl group, C9 acyl group, and C11 acyl group, such as from the group of C9 acyl group and C11 acyl group, further optionally in combination with a desacylated secretagogue, such as a desacylated Ghrelin-like compound.
  • the GH-secretagogue (such as a ghrelin-like compound) is administered with a substance capable of increasing the half-life of the GH-secretagogue, for example by incorporating the secretagogue into liposomes, micelles, iscoms, and/or microspheres or other transport molecules.
  • a substance capable of increasing the half-life of the GH-secretagogue for example by incorporating the secretagogue into liposomes, micelles, iscoms, and/or microspheres or other transport molecules.
  • the invention relates to the use of a pharmaceutical composition
  • a pharmaceutical composition comprising any secretagogue, such as any ghrelin-like compound as defined above or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable carriers, vehicles and/or excipients said composition further comprising transport molecules.
  • the transport molecules are primarily added in order to increase the half-life of the acylated compound, preventing premature des-acylation, since the des-acylated ghrelin is not active at the GHS-R 1a.
  • Transport molecules act by having incorporated into or anchored to it the compound according to the invention.
  • transport molecules Any suitable transport molecules known to the skilled person may be used. Examples of transport molecules are those described in the conjugate section. Other preferred examples are liposomes, micelles, and/or microspheres.
  • liposomes are typically composed of phospholipids (neutral or negatively charged) and/or cholesterol.
  • the liposomes are vesicular structures based on lipid bilayers surrounding aqueous compartments. They can vary in their physiochemical properties such as size, lipid composition, surface charge and number and fluidity of the phospholipids bilayers.
  • lipid for liposome formation 1 ,2-Dilauroyl-sn-Glycero-3-Phosphocholine (DLPC), 1 ,2- Dimyristoyl-sn-Glycero-S-Phosphocholine (DMPC) 1 1 ,2-Dipalmitoyl-sn-Glycero-3- Phosphocholine (DPPC), i ⁇ -Distearoyl-sn-Glycero-S-Phosphocholine (DSPC), 1 ,2- Dioleoyl-sn-Glycero-3-Phosphocholine (DOPC), 1 ,2-Dimyristoyl-sn-Glycero-3- Phosphoethanolamine (DMPE), 1 ⁇ -Dipalmitoyl-sn-Glycero-S-Phosphoethanolamine (DPPE), 1 ⁇ -Dioleoyl-sn-Glycero-S-Phosphoethanolamine (DOPE), 1 ,
  • DPPC in combination with other lipid or modifiers of liposomes are preferred e.g. in combination with cholesterol and/or phosphatidylcholine.
  • Long-circulating liposomes are characterized by their ability to extravasate at body sites where the permeability of the vascular wall is increased.
  • the most popular way to produce long circulating liposomes is to attach hydrophilic polymer polyethylene glycol (PEG) covalently to the outer surface of the liposome.
  • PEG polyethylene glycol
  • Some of the preferred lipids are: 1 ⁇ -Dipalmitoyl-sn-Glycero-S-Phosphoethanolamine-N- [Methoxy(Polyethylene glycol)-2000] (Ammonium Salt), 1 ,2-Dipalmitoyl-sn-Glycero- 3-Phosphoethanolamine-N-[Methoxy(Polyethylene glycol)-5000] (Ammonium Salt), 1 ,2-Dioleoyl-3-Trimethylammonium-Propane (Chloride Salt) (DOTAP).
  • the liposome suspension may include lipid-protective agents which protect lipids against free-radical and lipid-peroxidative damages on storage.
  • Lipophilic free-radical quenchers such as alpha-tocopherol and water- soluble iron-specific chelators, such as ferrioxianine, are preferred.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), U.S. Pat. Nos. 4, 235,871 , 4,501 ,728 and 4,837,028, all of which are incorporated herein by reference.
  • One method produces multilamellar vesicles of heterogeneous sizes.
  • the vesicle-forming lipids are dissolved in a suitable organic solvent or solvent system and dried under vacuum or an inert gas to form a thin lipid film.
  • the film may be redissolved in a suitable solvent, such as tertiary butanol, and then lyophilized to form a more homogeneous lipid mixture which is in a more easily hydrated powder like form.
  • a suitable solvent such as tertiary butanol
  • This film is covered with an aqueous solution of the targeted drug and the targeting component and allowed to hydrate, typically over a 15-60 minute period with agitation.
  • the size distribution of the resulting multilamellar vesicles can be shifted toward smaller sizes by hydrating the lipids under more vigorous agitation conditions or by adding solubilizing detergents such as deoxycholate.
  • the liposome suspension may include lipid-protective agents which protect lipids against free-radical and lipid-peroxidative damages on storage. Lipophilic free-radical quenchers, such as alpha-tocopherol and water- soluble iron-specific chelators, such as ferrioxianine, are preferred.
  • Micelles are formed by surfactants (molecules that contain a hydrophobic portion and one or more ionic or otherwise strongly hydrophilic groups) in aqueous solution. As the concentration of a solid surfactant increases, its monolayers adsorbed at the air/water or glass/water interfaces become so tightly packed that further occupancy requires excessive compression of the surfactant molecules already in the two monolayers. Further increments in the amount of dissolved surfactant beyond that concentration cause amounts equivalent to the new molecules to aggregate into micelles. This process begins at a characteristic concentration called "critical micelle concentration".
  • Suitable surfactants include sodium laureate, sodium oleate, sodium lauryl sulfate, octaoxyethylene glycol monododecyl ether, octoxynol 9 and PLURONIC F-127 (Wyandotte Chemicals Corp.).
  • Preferred surfactants are nonionic polyoxyethylene and polyoxypropylene detergents compatible with IV injection such as, TWEEN-80., PLURONIC F-68., n-octyl-.beta.-D-glucopyranoside, and the like.
  • phospholipids such as those described for use in the production of liposomes, may also be used for micelle formation.
  • One suitable formulation for preparing pharmaceutical compositions for use in the present invention is described in Example 9 of PCT patent application PCT/DK2004/000519 (Gastrotech Pharma).
  • An example of how one skilled in the art may investigate the pharmacokinetics of different formulations is given in Example 10 of PCT patent application PCT/DK2004/000519 (Gastrotech Pharma). Both Examples are incorporated herein by reference.
  • the compounds of the present invention are formulated as described in the literature for an administration route selected from: buccal delivery, sublingual delivery, transdermal delivery, inhalation and needle-free injection, such as using the methods developed by Powderjet.
  • the compounds of the present invention can be formulated as using methods known to those skilled in the art, for example an aerosol, dry powder or solubilized such as in micro droplets, preferably in a device intended for such delivery (such as commercially available from Aradigm, Alkerme or Nektar).
  • compositions for parenteral administration are provided.
  • the GH secretagogue or a salt thereof may be formulated for parenteral administration (e.g., by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • a pharmaceutical composition for parenteral administration may include sterile aqueous and non-aqueous injectable solutions, dispersions, suspensions or emulsions in oily or aqueous vehicles, for example solutions in aqueous polyethylene glycol, as well as sterile powders to be reconstituted in sterile injectable solutions or dispersions prior to use.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution for constitution before use with a suitable vehicle, e.g., sterile, pyrogen-free water.
  • a suitable vehicle e.g., sterile, pyrogen-free water.
  • Aqueous solutions should be suitably buffered if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • the aqueous solutions are particularly suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • the sterile aqueous media employed are all readily available by standard techniques known to those skilled in the art.
  • Solutions of GH secretagogues or pharmaceutically acceptable salts thereof can be prepared in water or saline, and optionally mixed with a nontoxic surfactant.
  • compositions for intravenous or intra-arterial administration may include sterile aqueous solutions that may also contain buffers, liposomes, diluents and other suitable additives.
  • oily or nonaqueous carriers, diluents, solvents or vehicles for parental use examples include propylene glycol, polyethylene glycol, animal, synthetic or vegetable oils, and injectable organic esters, and may contain formulatory agents such as preserving, wetting, emulsifying or suspending, stabilizing and/or dispersing agents.
  • oils useful in such compositions include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral.
  • Suitable fatty acids for use in parenteral compositions include oleic acid, stearic acid, and isostearic acid.
  • Suitable organic esters include fatty acid esters such as ethyl oleate and isopropyl myristate.
  • Suitable soaps for use in parenteral compositions include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides; (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl- imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • compositions typically will contain from about 0.5 to about 25% by weight of the active ingredient in solution. Preservatives and buffers may be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such compositions will typically range from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-lipophile balance
  • parenteral compositions can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions comprising the active ingredient that are adapted for administration by encapsulation in liposomes. In all cases, the ultimate dosage form must be sterile, fluid and stable under the conditions of manufacture and storage.
  • Sterile injectable solutions are prepared by incorporating the compound(s) or pharmaceutically acceptable salt(s) thereof in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • compositions for oral delivery are provided.
  • GH secretagogue types capable of remaining biologically active in an individual after oral administration (such as short peptides) can be formulated in a wide range of oral administration dosage forms.
  • the pharmaceutical compositions and dosage forms may comprise the compounds of the invention or its pharmaceutically acceptable salt or a crystal form thereof as the active component.
  • the pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, and dispersible granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, wetting agents, tablet disintegrating agents, or an encapsulating material.
  • the composition will be about 0.5% to 75% by weight of a compound or compounds of the invention, with the remainder consisting of suitable pharmaceutical excipients.
  • excipients include pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, glucose, gelatin, sucrose, magnesium carbonate, and the like.
  • the carrier is a finely divided solid which is a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets preferably contain 1-70% of the active compound.
  • Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • preparation is intended to include the composition of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is in association with it.
  • carrier which is in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be as solid forms suitable for oral administration.
  • Drops can be used according to the present invention and may comprise sterile or non-sterile aqueous or oil solutions or suspensions, and may be prepared by dissolving the active ingredient in a suitable aqueous solution, optionally including a bactericidal and/or fungicidal agent and/or any other suitable preservative, and optionally including a surface active agent.
  • a suitable aqueous solution optionally including a bactericidal and/or fungicidal agent and/or any other suitable preservative, and optionally including a surface active agent.
  • the resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98-100 0 C for half an hour.
  • the solution may be sterilized by filtration and transferred to the container aseptically.
  • bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0.002%), benzalkonium chloride (0.01%) and chlorhexidine acetate (0.01%).
  • Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol.
  • Other forms suitable for oral administration include toothpaste, gel dentrifrice orchewing gum.
  • Emulsions may be prepared in solutions in aqueous propylene glycol solutions or may contain emulsifying agents such as lecithin, sorbitan monooleate, or acacia.
  • Aqueous solutions can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents.
  • Aqueous suspensions can be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well known suspending agents.
  • Solid form preparations include solutions, suspensions, emulsions, syrups and elixirs and may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
  • compositions for topical administration are provided.
  • the compounds of the invention can be delivered topically. Regions for topical administration include the skin surface. Compositions for topical administration via the skin and mucous membranes should not give rise to signs of irritation, such as swelling or redness.
  • Transdermal administration typically involves the delivery of a pharmaceutical agent for percutaneous passage of the drug into the systemic circulation of the patient.
  • the skin sites include anatomic regions for transdermally administering the drug and include the forearm, abdomen, chest, back, buttock, mastoidal area, and the like.
  • the GH secretagogues may be formulated for topical administration to the epidermis as ointments, creamse, gels or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also containing one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • compositions suitable for topical administration in the mouth include lozenges comprising active agents in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • compositions for aerosol, nasal or inhalation delivery are provided.
  • the GH secretagogues may be formulated for administration to the respiratory tract and including intranasal administration, and for nasal administration.
  • the solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the compositions may be provided in a single or multidose form. In the latter case of a dropper or pipette this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray this may be achieved for example by means of a metering atomizing spray pump.
  • a suitable formulation for nasal administration is described in EP 1 466 610.
  • the compounds can be formulated as using methods known to those skilled in the art, for example an aerosol, dry powder or solubolized such as in microdroblets, preferably in a device intended for such delivery (such as commercially available from Aradigm, Alkerme or Nektar).
  • compositions administered by aerosols may be prepared, for example, as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, employing fluorocarbons, and/or employing other solubilizing or dispersing agents in accordance with methods known in the art.
  • the GH secretagogues to be used herein may also be formulated for administration as suppositories.
  • a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted and the active component is dispersed homogeneously, for example, by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and to solidify.
  • the active compound may be formulated into a suppository comprising, for example, about 0.5% to about 50% of a compound of the invention, disposed in a polyethylene glycol (PEG) carrier (e.g., PEG 1000 [96%] and PEG 4000 [4%].
  • PEG polyethylene glycol
  • compositions for other types of delivery are Compositions for other types of delivery
  • Subcutaneous administration Any parenteral administration form that will ensure that the ghrelin receptors which normally are the target for peripherally produced ghrelin in the premeal situation will be exposed to sufficient levels of the bioactive form of a GH secretagogueor a salt thereof to ensure robust and appropriate appetite stimulation, without causing desensitization of the system, may be part of the present invention. However, taken into consideration that the individuals to be treated possibly will have to receive treatment for a longer period, such as weeks or months, it is preferred that the administration form is well suited herefor.
  • the GH secretagogue or a salt thereof is administered subcutaneously in an amount sufficient to allow sufficient levels of the bioactive form, e.g. an acylated or anchorgroup containing form, to reach the receptors in time, such as prior to the forthcoming meal.
  • Dose and frequency of administration by the subcutaneous route are as described above.
  • receptor internalization i.e. physical removal of the receptor from the cell surface where it could bind the agonist
  • receptor down regulation i.e. decreased production / expression of the receptor
  • the present invention relates in one aspect to administration of a secretagogue, such as a ghrelin-like compound, in boluses, preferably a bolus prior to each main meal.
  • a secretagogue such as a ghrelin-like compound
  • the medicament can be administered as a bolus injection or by fast running infusion, i.e. an infusion preferably lasting less than 120 minutes, such as less than 90 minutes, for example less than 60 minutes, such as less than 45 minutes, such as less than 30 minutes, for example less than
  • 25 minutes such as less than 20 minutes, such as less than 15 minutes, for example less than 12 minutes, such as less than 10 minutes, such as less than 8 minutes, for example less than 6 minutes, such as less than 5 minutes, such as less than 4 minutes, for example less than 3 minutes, such as less than 2 minutes, such as less than 1 minute.
  • the bolus injection or the fast running infusion can be administered prior to a meal or during a meal as described in more detail herein below.
  • the medicament is administered as a bolus.
  • the bolus is preferably administered subcutaneously.
  • the present invention preferably deals with methods for administering a secretagogue, such as ghrelin, in a way which mimics the physiologically pre-meal situation as closely as possible yet providing patients in need of increased food intake, for example post operative patients with a sufficient extra stimulatory input to their appetite regulating ghrelin receptors, which normally are reached by ghrelin in the pre-meal situation.
  • a secretagogue such as ghrelin
  • Suitable dosing regimens for the various compounds and methods of the present invention are preferably determined taking into account factors well known in the art including type of subject being dosed; age, weight, sex and medical condition of the subject; the route of administration; the renal and hepatic function of the subject; the desired effect; and the particular compound employed.
  • Optimal precision in achieving concentrations of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug.
  • the secretagogue such as ghrelin or a ghrelin-like compound, is administered subcutaneously.
  • the secretagogue such as ghrelin or a ghrelin-like compound
  • the administration form may be any suitable parenteral form.
  • the secretagogue such as ghrelin or a ghrelin-like compound, is administered subcutaneously in a premeal bolus.
  • the secretagogue such as ghrelin or a ghrelin-like compound, can also be administered during a meal as a bolus.
  • the mode of administration during a meal includes subcutaneous administration, such as a subcutaneously administered bolus.
  • compositions for parenteral administration include sterile aqueous and non-aqueous injectable solutions, dispersions, suspensions or emulsions, as well as sterile powders to be reconstituted in sterile injectable solutions or dispersions prior to use.
  • Other suitable administration forms include suppositories, sprays, ointments, cremes, gels, inhalants, dermal patches, implants, pills, tablets, lozenges and capsules.
  • Ghrelin is primarily cleared by the kidneys and ghrelin administration yields markedly higher plasma levels in patients with renal failure than in healthy subjects (Wynne et al. J Am Soc Nephrol 2005; Aug (54)(8):2390-5. Accordingly a dosing regimen will be developed based on ghrelin pharmacokinetics and pharmacodynamics in patients with renal impairment.
  • a typical dosage of a compound employed according to the invention is in a concentration equivalent to from 10 ng to 10 mg ghrelin per kg bodyweight.
  • concentrations and amounts herein are given in equivalents of amount ghrelin, wherein the ghrelin is the 28 aa human ghrelin. Equivalents may be tested as described in the section entitled "Functionality", above.
  • the medicament is administered in a concentration equivalent to from 0.1 ⁇ g to 1 mg ghrelin per kg bodyweight, such as from 0.5 ⁇ g to 0.5 mg ghrelin per kg bodyweight, such as from 1.0 ⁇ g to 0.1 mg ghrelin per kg bodyweight, such as from 1.0 ⁇ g to 50 ⁇ g ghrelin per kg bodyweight, such as from 1.0 ⁇ g to 10 ⁇ g ghrelin per kg bodyweight.
  • the secretagogue such as ghrelin or a ghrelin-like compound
  • the medicament is administered as a bolus prior to a meal, said bolus comprising an amount of the secretagogue or a salt thereof equivalent to from 0.3 ⁇ g to 150 mg ghrelin. More preferably, the medicament is administered as a bolus prior to a meal, said bolus comprising an amount of the secretagogue or a salt thereof equivalent to from 2.0 ⁇ g to 200 mg ghrelin, such as from 5.0 ⁇ g to 100 mg ghrelin, such as from
  • 10 ⁇ g to 50 mg ghrelin such as from 10 ⁇ g to 5 mg ghrelin, such as from 10 ⁇ g to
  • the ghrelin-like compound is administered as a bolus in an amout equivalent to 10 ⁇ g ghrelin per kg bodyweight.
  • a dosing regimen for treatment of live failure patients is preferably based on ghrelin pharmacokinetics and pharmacodynamics in patients with liver failure, as known by one skilled in the art.
  • ghrelin is administered as a subcutaneous injection at a dose of 0.1-1000 ⁇ g/kg, such as at a dose of 1- 1000 ⁇ g/kg, such as at a dose of 1-100 ⁇ g/kg, such as at a dose of 1-50 ⁇ g/kg, such as at a dose of about 10 ⁇ g/kg.
  • compositions of the present invention contemplates pharmaceutical compositions useful for practicing the therapeutic methods described herein.
  • Pharmaceutical compositions of the present invention contain a physiologically tolerable carrier together with at least one species of a secretagogue, such as ghrelin or a ghrelin-like compound as described herein, dissolved or dispersed therein as an active ingredient.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one secretagogue, such as ghrelin or a ghrelin-like compound as defined above in formula I.
  • the pharmaceutical composition comprises at least two different ghrelin-like compounds as defined above in formula I in order to increase the effect of the treatment.
  • the difference may for example be compounds having different acylations, for example wild-type ghrelin and L-692-429 (Merck).
  • the formulation comprises the secretagogue or a salt thereof as a lyophilisate and the formulation further comprises a solvent, said lyophilisate and said solvent being in separate compartments until administration.
  • the formulation is a solution of the secretagogue or a salt thereof.
  • the present compounds may be administered in combination with further pharmacologically active substances or therapeutic method or other pharmacologically active material.
  • in combination with another substance(s) and/or therapeutic method(s) is meant herein that said another substance(s) and/or therapeutic method(s) is administered to the individual thus treated before, during (including concurrently with) and/or after treatment of an individual with a secretagogue.
  • the combination may be in the form of kit-in-part systems, wherein the combined active substances may be used for simultaneous, sequential or separate administration.
  • any of the herein-mentioned medicaments are administered in pharmaceutically effective amounts, i.e. an administration involving a total amount of each active component of the medicament or pharmaceutical composition or method that is sufficient to show a meaningful patient benefit.
  • the GH-secretagogue is administered to an individual suffering from, or at risk of suffering from, renal failure, in combination with one or more drugs used for treating or alleviating complications of renal failure.
  • drugs used for treating or alleviating complications of renal failure such as one or more of: EPO, dietary potassium/fluid and protein restriction/resin, correction of acidose (sodium bicarbonate or Calcium carbonate), 1-alfa-cholecalciferole and 1 , 25-(OH)-D3, prokinetics, antihypertensive treatment (e.g.ACE inhibitors), antidiabetic therapy, growth hormone and/or IGF-1.
  • the secretagogue is administered to the individual suffering from liver failure in combination with one or more drugs used for treating or alleviating liver failure, such as e.g. one or more of: colestyramine, ondansetron, rifampicin, variceal sclerotherapy, lactulose, metronidazole, ascitis paracentesis, sodium and fluid restriction, spironolactone, liver surgery therapies, liver transplant and associated medications and treatment regimes.
  • drugs used for treating or alleviating liver failure such as e.g. one or more of: colestyramine, ondansetron, rifampicin, variceal sclerotherapy, lactulose, metronidazole, ascitis paracentesis, sodium and fluid restriction, spironolactone, liver surgery therapies, liver transplant and associated medications and treatment regimes.
  • the secretagogue(s) according to the invention can also be administered in combination with other appetite-regulating agents, including more than one type of growth hormone secretagogue, such as another ghrelin-like compound, such as a ghrelin-like compound comprising a structure defined by formula I, described herein.
  • Other secretagogues suitable for combination administration with another secretagogue compound are any of the secretagogue compounds described herein.
  • wild type ghrelin most preferably human wild type ghrelin
  • a different, ghrelin-like compound - this combination is envisaged to enhance and/or prolong the effect of the secretagogues on the ghrelin receptor.
  • a ghrelin-like compound that is not wild type ghrelin is administered in combination with a different ghrelin-like compound that is not wild-type ghrelin- again, this combination is envisaged to enhance and/or prolong the effect of the secretagogues on the ghrelin receptor.
  • several different secretagogues may be administered to an individual to increase efficacy on the ghrelin receptor - such as greater than 2 different secretagogue types, such as 3, such as 4, such as 5, such as 6, such as 7, such as greater than 8 different secretagogue types.
  • the secretagogue according to the invention, such as ghrelin or a ghrelin-like compound(s) can also be administered in combination with a pharmaceutically effective amount of a growth hormone, including hGH.
  • the secretagogue such as ghrelin or a ghrelin-like compound
  • the rationale behind this combination treatment is to increase the level of IGF-1 , IGFBP-3, and/or ALS found to be low in cachectic individuals.
  • the secretagogues such as ghrelin or a ghrelin-like compound
  • compounds known to stimulate appetite such as melanocortin receptor antagonists, neuropeptide Y receptor agonists including agonists selective for individual subtypes of the neuropeptide Y receptors, leptin or leptin receptor agonists, cannabinoids including marijuana and marijuana derivatives, antipsychotics, especially atypical antipsychotics such as
  • the GH secretagogue may also be administered in combination with an ingredient or therapy useful in a supportive care regimen, such as one or more of the following:
  • Androgen(s) such as SARM and androgen receptor modulators.
  • compositions used in the invention may be administered alone or in combination with pharmaceutically acceptable carriers or excipients, in either single or multiple doses.
  • the formulations may conveniently be presented in unit dosage form by methods known to those skilled in the art.
  • kits typically contains an active compound in dosage forms for administration.
  • a dosage form contains a sufficient amount of active compound such that a desirable effect can be obtained when administered to a subject, preferably prior to at least one meal a day, more preferably prior to each main meal, such as three times a day, during the course of 1 or more days.
  • a dosage unit preferably comprises an amount of the ghrelin-like compound or a salt thereof equivalent to from 0.3 ⁇ g to 150 mg ghrelin, such as of from 2.0 ⁇ g to 100 mg ghrelin, such as from 5.0 ⁇ g to 75 mg ghrelin, such as from 10 ⁇ g to 50 mg ghrelin, such as from 10 ⁇ g to 5 mg ghrelin, such as from 10 ⁇ g to 1.0 mg ghrelin.
  • said dosage unit comprises an amount of the GH secretagogue or a salt thereof equivalent to from 0.3 ⁇ g to 600 mg ghrelin, such as of from 2.0 ⁇ g to 200 mg ghrelin, such as from 5.0 ⁇ g to 100 mg ghrelin.
  • the medical packaging may be in any suitable form for parenteral, in particular subcutaneous administration.
  • the packaging is in the form of a cartridge, such as a cartridge for an injection pen, the injection pen being such as an injection pen known from insulin treatment.
  • the medical packaging comprises more than one dosage unit
  • the medical packaging is provided with a mechanism to adjust each administration to one dosage unit only.
  • a kit contains instructions indicating the use of the dosage form to achieve a desirable affect and the amount of dosage form to be taken over a specified time period.
  • the medical packaging comprises instructions for administering the pharmaceutical composition.
  • said instructions may include instructions referring to administration of said pharmaceutical composition either during a meal, or preferably at the most 45 minutes prior to a meal, such as at the most 30 minutes prior to a meal, such as at the most 25 minutes prior to a meal, such as at the most 20 minutes prior to a meal, such as at the most 15 minutes prior to a meal, such as at the most 10 minutes prior to a meal, such as at the most 5 minutes prior to a meal. Examples
  • Transfected COS-7 cells were transferred to culture plates one day after transfection at a density of 1 x 10 5 cells per well aiming at 5 - 8 % binding of the radioactive ligand.
  • Two days after transfection competition binding experiments were performed for 3 hours at 4°C using 25 pM of 125 l-ghrelin (Amersham, Little Chalfont, UK).
  • Binding assays were performed in 0.5 ml of a 5O mM Hepes buffer, pH 7.4, supplemented with 1 mM CaCI 2 , 5 mM MgCI 2 , and 0.1 % (w/v) bovine serum albumin, 40 microgram/ml bacitracin.
  • Non-specific binding was determined as the binding in the presence of 1 micromole of unlabeled ghrelin. Cells were washed twice in 0.5 ml of ice-cold buffer and 0.5-1 ml of lysis buffer (8 M Urea, 2 % NP40 in 3 M acetic acid) was added and the bound radioactivity was counted. Determinations were made in duplicate. Initial experiments showed that steady state binding was reached with the radioactive ligand under these conditions.
  • Chronic renal failure will be induced in 20 rats by one-step 5/6 nephrectomy. All surgical interventions will be performed under anesthesia, induced by intraperitoneal administration of chloral hydrate (400 mg/kg). The experimental CRF is induced by removal of the upper and the lower pole of the left kidney, followed by complete removal of the right kidney 7 d later. In the 20 control rats, sham procedures involving kidney decapsulation will be performed performed. Simultaneously with the initial partial nephrectomy/sham surgery, all rats will be bilaterally orchidectomized.
  • a tablet (diameter, 0.5 cm), containing either 15 mg of testosterone (Innovative Research of America, Toledo, OH) or placebo, is inserted subcutaneously through a tunnel extending to approximately 2 cm lateral of the nephrectomy incision site. Gradual resorption of the tablets guaranteed stable plasma testosterone concentrations in the low normal range of adult male rats for approximately 3 wk. All rats are given a standard diet.
  • Both control and CRF rats will be divided into two group with 10 rats in each group receiving either saline or ghrelin 200ug/kg s.c. for 14 days.
  • the treatment will start four days after the surgical procedure.
  • Plasma hormone level IGF-I, leptin. Insulin,
  • Trial product Single dose, subcutaneously administered ghrelin at a dose of e.g. 5- 10 ug/kg.
  • Endpoints Appetite as assessed by visual analogue scales, eating related symptoms, food intake, body weight, lean body mass and quality of life.
  • Trial product Ghrelin e.g. 5-10 ⁇ g/kg administrered as a subcutaneous injection 1-2 times daily for 3-6 months.
  • Trial product Single dose, subcutaneously administered ghrelin at a dose of e.g. 10 ug/kg.
  • Endpoints Appetite as assessed by visual analogue scales, eating related symptoms, food intake, body weight, lean body mass and quality of life.
  • Trial product Ghrelin e.g. 10 ⁇ g/kg administered as a subcutaneous injection 1-2 times daily for 3-6 months. SEQUENCE LISTING

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Genetics & Genomics (AREA)
  • Obesity (AREA)
  • Nutrition Science (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Utilisation d'un composé sécrétagogue pour la préparation d'un médicament destiné à traiter un individu souffrant d'une insuffisance rénale et/ou hépatique. Procédé de stimulation de l'appétit, de l'ingestion d'aliments et/ou du gain de poids chez un individu souffrant d'une insuffisance hépatique et/ou rénale, le procédé consistant à administrer un sécrétagogue audit patient.
EP05796707A 2004-10-27 2005-10-27 Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique Withdrawn EP1812044A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DKPA200401655 2004-10-27
DKPA200401654 2004-10-27
PCT/DK2005/000694 WO2006045319A2 (fr) 2004-10-27 2005-10-27 Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique

Publications (1)

Publication Number Publication Date
EP1812044A2 true EP1812044A2 (fr) 2007-08-01

Family

ID=36101732

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05796707A Withdrawn EP1812044A2 (fr) 2004-10-27 2005-10-27 Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique

Country Status (2)

Country Link
EP (1) EP1812044A2 (fr)
WO (1) WO2006045319A2 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7666833B2 (en) 2001-12-18 2010-02-23 Alizé Pharma SAS Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
CN101291683B (zh) 2004-11-24 2011-08-17 纽普罗研究有限公司 治疗疾病的方法和组合物
EP2015765A4 (fr) 2006-03-28 2011-11-23 Neopro Labs Llc Méthodes et compositions pour traiter des conditions
AU2008241532A1 (en) 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
WO2008144675A1 (fr) 2007-05-17 2008-11-27 Neopro Labs, Llc Formes cristallines et amorphes d'un peptide
US8318664B2 (en) 2007-05-31 2012-11-27 Alize Pharma Sas Unacylated ghrelin fragments as therapeutic agent in the treatment of obesity
US8222217B2 (en) 2007-05-31 2012-07-17 Alize Pharma Sas Unacylated ghrelin as therapeutic agent in the treatment of metabolic disorders
US8476408B2 (en) 2008-06-13 2013-07-02 Alize Pharma Sas Unacylated ghrelin and analogs as therapeutic agents for vascular remodeling in diabetic patients and treatment of cardiovascular disease
EP2401367B1 (fr) 2009-02-26 2016-11-30 Transposagen Biopharmaceuticals, Inc. Transposases piggybac hyperactives
LT2790721T (lt) 2011-12-15 2019-02-11 Millendo Therapeutics Sas Neacilinto grelino fragmentai, skirti naudoti preiderio-vilio sindromo gydymui

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020111461A1 (en) * 1999-05-21 2002-08-15 Todd C. Somers Low molecular weight peptidomimetic growth hormone secretagogues
US7385026B1 (en) * 1999-07-23 2008-06-10 Kenji Kangawa Modified ghrelin polypeptides
WO2001056592A1 (fr) * 2000-02-01 2001-08-09 Novo Nordisk A/S Utilisation de composes pour la regulation de l'absorption de nourriture
US6967237B2 (en) * 2000-05-30 2005-11-22 Merck & Co., Inc. Ghrelin analogs
WO2005014032A2 (fr) * 2003-08-06 2005-02-17 Gastrotech Pharma A/S Utilisations de secretagogues

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006045319A2 *

Also Published As

Publication number Publication date
WO2006045319A2 (fr) 2006-05-04
WO2006045319A3 (fr) 2006-09-28

Similar Documents

Publication Publication Date Title
EP1999146B1 (fr) Utilisation d'une variante de la ghréline pour traiter la cachexie, et/ou l'anorexie-cachexie, et/ou la malnutrition, et/ou la lipodystrophie, et/ou la perte de muscles, et/ou stimuler l'appétit;
US20080300180A1 (en) Growth Hormone Secretagogue Receptor 1A Ligands
WO2006045319A2 (fr) Utilisations de secretagogues d'hormones de croissance dans le traitement d'individus souffrant d'insuffisance renale et/ou hepatique
US20080171700A1 (en) Use Of Secretagogue For The Teatment Of Ghrelin Deficiency
EP1553969B1 (fr) Utilisation de la ghreline dans le traitement de la malnutrition chez des individus gastrectomises
WO2006045314A2 (fr) Utilisation d'un secretagogue de l'hormone de croissance en vue d'accroitre ou de maintenir la masse maigre et/ou pour le traitement de la maladie pulmonaire obstructive chronique
US10526383B2 (en) Ghrelin splice variant for treating neuronal damage, neurodegenerative disease, parkinsons disease, alzheimers disease, and/or depression
US7763707B2 (en) Use of ghrelin splice variant for treating cachexia and/or anorexia and/or anorexia-cachexia and/or malnutrition and/or lipodystrophy and/or muscle wasting and/or appetite-stimulation
WO2005014032A2 (fr) Utilisations de secretagogues
US20070037751A1 (en) Uses of secretagogues like ghrelin in cancer cachexia and for stimulating appetite
EP1812045A2 (fr) Utilisations de secretagogues pour le traitement de patients ayant subi une greffe d'organe
WO2005097174A2 (fr) Utilisations de combinaisons de secretagogue gh et d'une hormone de croissance

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070529

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090501