WO2005079888A2 - Delivery device for delivering pyy - Google Patents

Delivery device for delivering pyy Download PDF

Info

Publication number
WO2005079888A2
WO2005079888A2 PCT/DK2005/000111 DK2005000111W WO2005079888A2 WO 2005079888 A2 WO2005079888 A2 WO 2005079888A2 DK 2005000111 W DK2005000111 W DK 2005000111W WO 2005079888 A2 WO2005079888 A2 WO 2005079888A2
Authority
WO
WIPO (PCT)
Prior art keywords
delivery device
pyy
treatment
pharmaceutical composition
container
Prior art date
Application number
PCT/DK2005/000111
Other languages
French (fr)
Other versions
WO2005079888A3 (en
Inventor
Christian Hansen
Henrik Nilsson
Original Assignee
Aditech Pharma Ab
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 Aditech Pharma Ab filed Critical Aditech Pharma Ab
Priority to PCT/DK2005/000327 priority Critical patent/WO2005110467A1/en
Priority to JP2007516968A priority patent/JP2007538017A/en
Priority to US11/596,973 priority patent/US20080125360A1/en
Priority to PCT/DK2005/000329 priority patent/WO2005110454A2/en
Priority to PCT/DK2005/000328 priority patent/WO2005110457A2/en
Priority to EP05739487A priority patent/EP1750753A1/en
Priority to AU2005244475A priority patent/AU2005244475A1/en
Publication of WO2005079888A2 publication Critical patent/WO2005079888A2/en
Publication of WO2005079888A3 publication Critical patent/WO2005079888A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/178Syringes
    • A61M5/28Syringe ampoules or carpules, i.e. ampoules or carpules provided with a needle
    • A61M5/285Syringe ampoules or carpules, i.e. ampoules or carpules provided with a needle with sealing means to be broken or opened
    • A61M5/286Syringe ampoules or carpules, i.e. ampoules or carpules provided with a needle with sealing means to be broken or opened upon internal pressure increase, e.g. pierced or burst
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/178Syringes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/178Syringes
    • A61M5/19Syringes having more than one chamber, e.g. including a manifold coupling two parallelly aligned syringes through separate channels to a common discharge assembly
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/178Syringes
    • A61M5/28Syringe ampoules or carpules, i.e. ampoules or carpules provided with a needle
    • A61M5/284Syringe ampoules or carpules, i.e. ampoules or carpules provided with a needle comprising means for injection of two or more media, e.g. by mixing

Definitions

  • the invention relates to a delivery device, such as a single or multi dose delivery device for delivering of PYY or a functional equivalent thereof.
  • a delivery device such as a single dose or multi dose delivery device, comprising a pharmaceutical composition comprising PYY or a functional equivalent for the treatment of overweight, obesity and/or disorders of appetite regulation.
  • Said delivery device may further be used for the treatment of gastrointestinal disorders such as IBS, FD and/or abdominal pain. It is further included that the delivery device is for the treatment of treatment of eating disorders, such as bulimia nervosa, binge eating disorder and or night eating syndrome.
  • the invention further relates to a method of treatment comprising administration of PYY or a functional equivalent thereof by use of a delivery device, such as a single or multi dose delivery device.
  • Overweight and obesity is associated with severe health risk as well as loss of the sense of well- being and quality of life for the individual.
  • the cause of overweight and obesity may be disease related, caused by unsuitable eating habits or lack of exercise or a combination of these.
  • Bodymass index Overweight and obesity may be defined by the body mass index (BMI) of the individual. BMI is calculated, by dividing the bodyweight in kilograms with the square of the height in meters. bodyweight(kg)
  • Overweight and obesity may be caused by metabolic disorders as well as disorders of appetite regulation other than eating disorders as described here below.
  • Eating disorders A great number of people suffer from eating disorders, such as bulimia nervosa, binge eating disorder and/or night eating syndrome and thereby have an increased risk of acquiring additional health problems as well as a lack of quality of life. These types of syndromes have been getting more attention lately and are now being characterized as clinical diseases. A major problem is the diagnosis of these dis- eases, as the syndromes are merely characterised by a behavioural pattern of the patient. The characteristics of the three syndromes are summarised below.
  • Bulimia nervosa is a serious eating disorder marked by a destructive pattern of binge-eating and recurrent inappropriate behaviour to control one's weight. Binge eating is defined as the consumption of excessively large amounts of food within a short period of time. "Inappropriate compensatory behaviour" to control one's weight may include purging behaviours (such as self-induced vomiting, abuse of laxatives, diuretics, or enemas) or non-purging behaviours (such as fasting or excessive exer- cise).
  • BED Binge Eating Disorder
  • BED is a type of eating disorder not otherwise specified and is characterized by recurrent binge eating without the regular use of compensatory measures to counter the binge eating. People with binge eating disorder frequently eat large amounts of food and feel a loss of control over their eating behaviour. This disorder is different from binge-purge syndrome (bulimia nervosa) because people with binge eating disorder usually do not purge afterward by vomiting or using laxa- tives.
  • Night-eating syndrome has not yet been formally defined as an eating disorder.
  • a person suffering from NES shows little or no appetite for breakfast and delays first meal for several hours after waking up. Often the patient eats more food after dinner than during the meal. This may mean that more than half of the daily food intake occurs after dinner but before breakfast. This pattern should persist for at least two months before being characterised as a night eating syndrome.
  • the behaviour of an NES patient is different from the behaviour of an individual suffering from a binge eating disorder. During binge eating, relatively large amounts of food are consumed over a relatively short space of time. Night-eating syndrome involves continuous eating throughout evening hours.
  • Cravings may be characterised as an intense desire for certain foods that is almost impossible to withstand, it may even be experienced as an addiction. It remains unclear whether a legitimate physiological basis for this intense desire for certain foods truly exists. Cravings may arise in an attempt to supply the body with nutrients it lacks. Thus deprival of carbohydrates by diets or depletion of glycogen stores by heavy exercisers may result in carbohydrate cravings by which carbohydrates are replenished, although food with a high sugar and fat content is normally preferred.
  • Gastrointestinal disorders are very common in the population. Some of these are very well characterised and thus suitable treatment regimes have been developed. It is more difficult to develop treatments for functional gastrointestinal disorder with unknown aetiology such as functional dyspepsia (FD) and irritable bowel syndrome (IBS) also called irritable colon.
  • FD functional dyspepsia
  • IBS irritable bowel syndrome
  • Irritable Bowel Syndrome The predominant symptoms of IBS are abdominal pain, altered bowel habit, discomfort associated with disturbed defecation and bloating. Patients have an increased mucus and nausea and feeling of constipation and distension.
  • the criteria for irritable bowel syndrome (according to Rome II) are pain or discomfort for 12 weeks of the previous 12 months associated with two of the following; relief with defecation, looser or more frequent stools, harder or less frequent stools (reviewed by Talley, NJ and Spiller R (2002) and Talley, NJ 2003).
  • the symptoms may be chronic and impair the quality of life for the patient.
  • the patients are grouped in three groups based on there predominat bowel habit (diarrhoea and/or constipation): IBS associated with abdominal pain, fecal urgency and diarrhoea IBS associated with abdominal discomfort, bloating and constipation IBS associated with alternating diarrhoea and constipation
  • FD functional dyspepsia
  • the disorders mentioned are all affected or related to food intake and absorption of nutrients and solutes from the gut.
  • Energy uptake is regulated by hormones that regulate food intake, by modifying the sensing of appetite, and regulate absorption of nutrients and solutes from the gut. Hormones can be separated into long and short term regulating hormones. Long term regulating hormones as insulin and leptin act slowly to promote the stability of body fat stores whereas the short term regulating hormones, as ghrelin and cholecystokinin, act rapidly to influence the individual meal by sensing of hunger, satiety and "fullness". Insulin and leptin are released into the blood in proportion to the amount of body fat. When body fat stores are reduced, declining levels of these hormones are sensed by the brain and are transduced into increases in appetite and metabolic efficiency that persists until the lost weight is recovered.
  • Ghrelin and cholecystokinin are factors that trigger the onset and termination of eating and thereby ghrelin and cholecystokini function in a meal-to-meal control system that itself is sensitive to changes in insulin and leptin levels. In this way, the size and frequency of individual meals can be adjusted so as to minimize changes in body fat content.
  • the uptake of nutrients from the gut is regulated by various factors including gut hormones.
  • the gut hormones VIP, CCK, and motilin relate to the motility of the upper gastrointestinal tract whereas polypeptide YY (PYY) and Neuropeptide Y (NPY) affect the absorption in the intestine.
  • the gut hormone peptide YY (PYY), and the neuropeptide, neuropeptide Y (NPY), are structurally related to pancreatic polypeptide (PP) (figure 1).
  • PYY and NPY exert their action through NPY receptors (Y1 R, Y2R, Y4R and Y5R).
  • the PP, NPY and PYY peptides consist of 36 amino acids with an amidated C-terminal. Two forms of PYY, PYY1-36 and PYY3-36, the latter being a truncated form of the former, have been found in circulation.
  • PYY3-36 is produced by the cleavage of PYY1-36 by the enzyme dipeptidyl peptidase IV (DPP-IV).
  • DPP-IV dipeptidyl peptidase IV
  • PYY1-36 binds to and activates at least three NPY receptor subtypes (Y1 , Y2 and Y5) whereas PYY3-36 is more selective for the Y2 receptor (Y2R). Only the C-terminal part of the PYY3-36 peptide is required for the binding to the Y2 receptor.
  • PYY covers both PYY1-36 and PYY3-36 (Berglund, M.M. et al., 2003).
  • PYY was initially isolated from porcine intestine (Tatemoto, K. and Mutt, C, 1980) and named Peptide YY due to the tyrosine residues present in the N- and C- terminal of the molecule.
  • PYY is expressed in endocrine cells lining the gastrointestinal tract and particularly in the distal portion.
  • PYY is secreted in response to food ingestion.
  • the maximum level of PYY reached is proportional to the calories ingested, suggesting that PYY may function as a sensor of food ingestion.
  • PYYY is also expressed by neurones, such as in peripheral neurons, particularly enteric neurons.
  • PYY is found in a restricted set of central neurons.
  • the expression pattern of PYY in both endocrine cells and neurons suggest that PYY may be involved in regulation of multiple functions in the individual (Ekblad, E. and Sundler, F., 2002).
  • PYY may be to regulate the secretion and absorbance of fluid and electrolytes in the gastrointestinal tract and intestine and PYY have therefore been suggested as treatment of diarrhoea (US 6,588, 708) by prolonging of the residence time.
  • PYY3-36 has been suggested to be involved in the system regulating feeding behaviour. It has been found that peripheral administration of PYY3-36 inhibited food intake in rodents. Moreover, direct intra- arcuate administration of PYY3-36 inhibited food intake.
  • the hypothalamic arcuate nucleus a key brain area regulating appetite, has access to nutrients and hormones within the peripheral circulation.
  • NPY neurons within the arcuate nucleus express the Y2R.
  • the arcuate nucleus contains two distinct subgroups of neurons that control food intake. On group of neurons produces NPY, which acts in the brain to stimulate feeding (Stanley, B. G. et al, 1986), whereas an adjacent subgroup of neurones produces melanocortin peptides, which act in the same brain areas as NPY, but inhibit eating (Fan, W. et al, 1997). Typically, when one of these subsets is activated, the other one is inhibited.
  • Weight loss as modest as 5-15 % of total body weight, reduces the risk factors for many diseases. Thus, independent of the cause of overweight or obesity, treatment that decreases the calorie intake of the individual, may be beneficial. Treatment
  • injection pens used in the treatment of diabetes have improved the treatment and the quality of life for the individual suffering of diabetes.
  • injection pens are mostly used for insulin and growth hormone, but treatments with other medicaments using injection pens are emerging.
  • the injection devices exist in a variety of designs with various different functional characteristics. Some of the variants include disposable pens and reusable pens, prefilled syringes and cartridges, single or multiple doses, liquid, lyophilized or powder drugs.
  • Injection pens are produced by companies such as Becton Dickinson, Eli Lilly, Novo Nordisk, Ypsomed, Meridian Medical, Schering, Owen Mumford, Serono, Innoject and Amgen.
  • the present invention relates to a delivery device, such as a multi or single dose delivery device specially adapted for delivering PYY or a functional equivalent thereof, comprising at least one container assembly, dosing means, ejecting means and optionally a needle assembly.
  • a delivery device such as a multi or single dose delivery device specially adapted for delivering PYY or a functional equivalent thereof, comprising at least one container assembly, dosing means, ejecting means and optionally a needle assembly.
  • the delivery device is adapted for storing PYY or a functional equivalent thereof for a prolonged period of time, as well as for delivering dosages of PYY or the functional equivalent thereof parenterally to an individual in need thereof.
  • the delivery device is for use in the treatment of adipositas or obese or any other disease or disorder treatable with PYY or a functional equivalent thereof.
  • Such other disorder may be an eating disorder, as NES, BED, food cravings or Bulimia Nervosa and further gastrointestinal disorders such as functional dyspepsia and irritable bowel syndrome may according to the invention be treated using PYY or a functional equivalent thereof by use of the delivery device described herein.
  • the container assembly normally comprises at least one container comprising PYY or a functional equivalent thereof as well as means for engaging a needle assembly to the container for injecting PYY or a functional equivalent thereof.
  • the delivery device is for parenteral administration of PYY.
  • the container may be any suitable container known in the art, such as a glass con- tainer or a plastic container.
  • the container is preferably formed with a plunger at one end and a sealing at the other end of said container.
  • the inner surface of the container comprising PYY or a functional equivalent thereof is coated with a coating.
  • Said coating is preferably a coating that reduces or avoids adherence of PYY or a functional equivalent thereof to container surface.
  • the coating may serve to reduce degradation of PYY or the functional equivalent thereof, and to secure stabilization of the PYY or the functional equivalent thereof.
  • the container may be a single use container.
  • the device may be able to release the container to receive a new filled container, when the former con- tainer has been used, in the present context called a changeable container.
  • the container may be a refillable container.
  • the delivery device may comprise at least two containers.
  • each container may comprise PYY or a functional equivalent thereof.
  • the delivery device may also comprise at least two containers wherein at least one container comprises PYY or a functional equivalent thereof and at least one other container comprises a solvent to be mixed with the PYY or a functional equivalent thereof before or during administration of the PYY or the functional equivalent thereof.
  • the delivery device such as the single or multi dose delivery device may comprise two containers.
  • the container of the delivery device according to the inven- tion may comprise at least two compartments, wherein each compartment may comprise PYY or a functional equivalent thereof.
  • the container may also comprise at least two compartments wherein at least one compartment comprises PYY or a functional equivalent thereof and at least one other compartment comprises a solvent to be mixed with the PYY or a functional equivalent thereof before or during administration of the PYY or the functional equivalent thereof.
  • the contents of the two containers or two compartments can be mixed prior to use.
  • said mixing occurs via a liquid channel.
  • Said liquid channel may be formed by a needle or by opening of a breakable sealing by means of for example a plunger or a needle.
  • An embodiment of the invention relates to a single or multi dose delivery device comprising at least one container assembly wherein, said container assembly comprise a distal end comprising coupling means for realisably mounting a needle assembly and said container assembly comprise a proximal end comprising coupling means for engaging the dosing assembly.
  • the dosing assembly comprises means for adjusting ejection of one dosage per activation.
  • the dosing assembly comprises means for adjusting ejection of individual dosages in accordance with an individual dosage scheme.
  • a further embodiment of the invention include a delivery device, such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or - any combination hereof.
  • a delivery device such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or - any combination hereof.
  • a delivery device such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or and combination thereof.
  • a delivery device such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia
  • the PYY or a functional equivalent thereof comprised in the delivery device may be in the form of a pharmaceutical composition optionally as a pharmaceutically acceptable salt or pharmaceutically acceptable acid addition salt, and optionally with pharmaceutically acceptable carriers and/or diluents.
  • the pH of the pharmaceutical composition is preferably between 3 and 5.5 and preferably buffered using an acetate buffer.
  • the pharmaceutical composition comprising PYY or a functional equivalent thereof may be mixed with a solvent prior to use or during use.
  • the mixing preferably takes place within the single or multi dose delivery device.
  • the pharmaceutical composition is a dry composition, such as a freeze dried or spray dried composition, to be mixed with a solvent before or during use.
  • a method of treatment comprising the use of a delivery device, such as a single dose or multi dose delivery device according to the invention.
  • the single or multi dose delivery device according to the invention is suitable for self administration, i.e. administration wherein the individual in need of the treatment administrates the pharmaceutical composition himself or herself.
  • the dosage to be administered per administration is determined by a physician prescribing the composition. As described above, it is preferred that the delivery device allows individual adjustment of the size of dosage. Normally such as 0.25-20 nmol of PYY or a functional equivalent is administrated per day, in one or more dosages. Preferably PYY or the functional equivalent thereof is administered in at least 1 dos- age per day corresponding to the main meal of the day.
  • the medicament is administrated with in 120 minutes before a meal, such as within 60 minutes before a meal, such as within 30 minutes before a meal, such as within 15 minutes before a meal such as at the be- ginning of a meal.
  • the PYY or functional equivalent thereof can be administered less frequently than once per day.
  • PYY may be administered every second day, every third day, every fourth day, every fifth day, every sixth day or once a week depending on the needs of the individual patient.
  • the dosage for such administration may be as described herein for daily dosages, optionally times the number of days between administration. For instance, if administered every second day the dosage can be twice that determined for an every day administration.
  • the medicament is administrated dependent on the sensing of appetite in the individual.
  • the medicament is administered in combination with a second active ingredient.
  • PP-fold peptides NPY, PYY, and PP share a common hairpin-like three-dimensional structure called the PP-fold. All three peptides are 36 amino acids long with an amidated carboxy- terminus.
  • the general structure of the PP-fold peptides has been established using x-ray crystallography of avian PP and confirmed in several studies using nuclear magnetic resonance.
  • Amino acid residues 1-8 form a type II proline helix followed by a loop.
  • Residues 15-32 form an ⁇ -helix, and the four most carboxy-terminal residues are in a flexible loop conformation.
  • the amino acid sequence of mammalian NPY is highly conserved, PPY display 8 varible amino acids, whereas the amino acid sequence of PP is the least conserved PP-fold peptide.
  • the general three- dimensional structure seems to be conserved in all PP-fold peptides (Berglund MM et al, 2003).
  • Affinity The term refers to the binding strength between receptors and their ligands, for example PYY and its receptor.
  • Agonist A molecule that stimulates the action of the target.
  • Amino acid Entity comprising an amino terminal part (NH 2 ) and a carboxy terminal part (COOH) separated by a central part comprising a carbon atom, or a chain of carbon atoms, comprising at least one side chain or functional group.
  • NH 2 refers to the amino group present at the amino terminal end of an amino acid or peptide
  • COOH refers to the carboxy group present at the carboxy terminal end of an amino acid or peptide.
  • the generic term amino acid comprises both natural and non- natural amino acids. Natural amino acids of standard nomenclature as listed in J. Biol.
  • Appetite in an individual is assessed by measuring the amount of food ingested and by assessing the individual's desire to eat.
  • Appetite i.e., 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.
  • Amino acid residue the term "amino acid residue” is meant to encompass amino acids, either standard amino acids, non-standard amino acids or pseudo-amino ac- ids, which have been reacted with at least one other species, such as 2, for example 3, such as more than 3 other species.
  • amino acid residues may comprise an acyl bond in place of a free carboxyl group and/or an amine-bond and/or amide bond in place of a free amine group.
  • reacted amino acids residues may comprise an ester or thioester bond in place of an amide bond Antagonist: A molecule that inhibits that action of its target.
  • Antibody Are immunoglobulin molecules and active portions of immunoglobulin- molecules. Antibodies are for example intact immunoglobulin molecules or fragments there of retaining the immunologic activity.
  • BMI Body mass index measures an individual's height/weight ratio. It is determined by calculating weight in kilograms divided by the square of height in meters. The
  • 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.
  • Dissociation constant, Kd a measure to describe the strength of binding (or affin- ity or avidity) between receptors and their ligands, for example an antibody and its antigen. The smaller the Kd, the stronger binding.
  • Concentration equivalent is an equivalent dosage being defined as the dosage of compound having in vitro or/and in vivo the same response (as evaluated e.g. from a dosage-response curve) as a known compound.
  • 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.
  • Half-life See T1/2. Individual: A living animal or human.
  • the subject is a mammal, including humans and non-human mammals such as dogs, cats, pigs, cows, sheep, goats, horses, rats, and mice.
  • the subject is a human.
  • Isolated is used to describe any of the PYY peptides or functional analogues thereof 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, hor- mones, and other proteinaceous or non-proteinaceous solutes.
  • the polypeptide will be purified.
  • “Loss of body weight” defined herein as a reduction in BMI.
  • “Loss of body fat” defined herein as either a reduction of an individual's overall fat mass or a reduction in the percentage of an individual's body fat.
  • Medical disorder by “medical disorder” is meant any disease or syndrome having a detrimental effect on an individual's physical and/or mental health. Said medical disorder may have a genetic cause. Preferably, said medical disorder leads to one or more undesirable symptoms including increase of fat mass, increase of weight, increase of appetite, and/or causes a reduction in the individual's PYY levels.
  • Peptide Plurality of covalently linked amino acid residues defining a sequence and linked by amide bonds.
  • the term is used analogously with oligopeptide and polypeptide.
  • the amino acids may be both natural amino acids and non-natural amino acids, including any combination thereof.
  • the natural and/or non-natural amino acids may be linked by peptide bonds or by non-peptide bonds.
  • the term peptide also embraces post-translational modifications introduced by chemical or enzyme-catalyzed reactions, as are known in the art. Such post-translational modifications can be introduced prior to partitioning, if desired.
  • Amino acids as specified herein will preferentially be in the L-stereoisomeric form. Amino acid analogs can be employed instead of the 20 naturally-occurring amino acids.
  • PYY Composition This term is intended to indicate a pharmaceutical composition comprising PYY or a functional equivalent thereof.
  • Receptor A receptor is a molecule, such as a protein, glycoprotein and the like, that can specifically (non-randomly) bind to another molecule
  • rDNA Recombinant DNA
  • a recombinant DNA molecule is a hybrid DNA molecule comprising at least two nucleotide sequences not normally found together in nature.
  • T 1 2 The half-life is the time for the concentration of a compound to decrease 50 %.
  • An individual with a body mass index above 25 is considered overweight or obese.
  • the cause of overweight and obesity may have many different reasons and may be due to a combination of several factors such as disorders, lack of exercise, excess calorie intake, a diet of food with inadequate nutritional composition, and/ or social habits.
  • Metabolic disorders may affect the ability of the individual to control the bodyweight, as signalling from the energy storage compartments may be lacking or incorrect.
  • Some disorders of appetite regulation are caused by a direct malfunction of the appetite regulating system.
  • Compulsive (compensatory) eating as NES, BED food cravings and bulimia nervosa may be at least partly be characterised as psychological disorders. Theses disorders may be associated with overweight or underweight. Alternatively the patient may be of normal weight although the nutritional status is affected in an undesirable way. Gastrointestinal disorders
  • IBS or functional dyspepsia are functional gastrointestinal disorders that may also affect the nutritional status and physical well being of the patients.
  • Weight gain is caused by the intake of excess calories, thus the control of body weight need balance of calorie intake and catabolism.
  • the level of catabolism is controlled by the body activities, thus exercise increases catabolism and prevents weight gain when food consumption is constant.
  • the composition of the diet may also affect the assimilation as different categories of food are absorbed differently.
  • any suitable individual who may draw benefit from usage of the single or multi dose delivery device of the invention may be considered an individual in need.
  • said individual is suffering from overweight or obesity.
  • the individual to be treated has a BMI in the range of 25-30.
  • the individual to be treated has a BMI in the range of 30-35.
  • the individual to be treated has a BMI of 27 or above.
  • Individuals suffering from an eating disorder as described herein, or a disorder of the gastro intestinal system, such as IBS or functional dyspepsia may benefit from usage of the delivery device.
  • the delivery device of the invention may be used for inhibition of appe- tite and decreasing the rate of weight gain in animals.
  • PYY1-36 The human pancreatic polypeptide PYY1-36 is identified by SEQ ID NO:1.
  • PYY3-36 is a truncated form of PYY1-36 where the two most N-terminal residues are deleted.
  • PYY1-36 is a 36 AA polypeptide with a C- as well as an N-terminal tyrosine amino acid residue.
  • the polypeptides are produced by cleavage of a pre-polypeptide and PYY1-36 furthermore cleaved by dipeptidyl peptidase IV yielding PYY3-36, as described above.
  • peptide YY (PYY) is used as a general term covering PYY1-36 and PYY3-36.
  • Functional equivalents of PYY1-36 and PYY3-36 include PYY molecules originating from different species, such as mouse, rat, monkey, swine, bovine or other mammalian species. Furthermore, functional equivalents comprise homologues of PYY molecules.
  • a homologue shall be construed as a molecule which shares some identity to the molecule, here PYY1-36 and PYY3-36.
  • the homology may be expressed as 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.
  • 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 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 perform- ing the same function, i.e. a homologue may be envisaged as a "functional equivalent" of a predetermined sequence.
  • a functional equivalent of any of the predetermined sequences herein may be defined as: i) homologues comprising an amino acid sequence capable of being recognised by an antibody, said antibody also recognising PYY1-36 or PYY3-36, and/or
  • homologues comprising an amino acid sequence capable of binding selectively to an NPY receptor, and/or
  • the 36 aa PYY1-36 has the sequence shown in SEQ ID NO: 1.
  • PYY3-36 is 34 amino acids long and has the sequence shown in SEQ ID NO: 1 except for the deletion of the two N-terminal amino acids.
  • 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 (Gly) of said homologue is substituted with an amino acid selected from the group of amino acids consisting of Ala, Val, 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 Gly, Val, Leu, and He, and independently thereof, homologues, wherein at least one valine (Val) of said homologue thereof is substituted with an amino acid selected from the group of amino acids consisting of Gly, 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 Gly, Ala, Val, and He, and independently thereof, homologues thereof, wherein at least one isoleucine (He) of said homologues thereof is substitute
  • 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 polar- ity, for example a residue with a non-polar side chain (Ala, Leu, Pro, Trp, Val, lie, Leu, Phe or Met) substituted for a residue with a polar side chain such as Gly, Ser, Thr, Cys, Tyr, Asn, or Gin or a charged amino acid such as Asp, Glu, 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 Gly by another residue; and/or iii) differ substantially in electric charge, for example substitution of a negatively charged residue such as Glu or Asp for a positively charged residue such as Lys, His or Arg (and vice versa); and/or iv) differ substantially in steric bulk, for example substitution of
  • Substitution of amino acids may in one embodiment be made based upon their hy- drophobicity 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 functional equivalent 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.
  • the functional equivalent comprises the amino acids corresponding to the 6 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO.1 (Tyr Pro lie Lys Pro Glu).
  • the functional equivalent may comprise 8 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro), or such as 10 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu) or such as 12 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala), or such as 14 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro) ), or such as 16 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ty
  • Glu Glu Leu Asn Arg Tyr Tyr Ala or such as 24 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu), or such as 26 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His), or such as 28 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu), or such as 30 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro
  • the functional equivalent comprise the amino acids corresponding to the 6 N- terminal amino acids of PYY3-36 as defined in SEQ ID NO.1 (lie Lys Pro Glu Ala Pro), or such as 8 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu) or such as 10 N- terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala), or such as 12 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro) ), or such as 14 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu), or such as 16 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO.
  • the functional equivalent comprise the amino acids corresponding to the 6 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Val Thr Arg Gin Arg Tyr), or such as the 8 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 10 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 12 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 14 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 16 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ser Leu Arg His Tyr
  • the functional equivalent may comprises internal amino acids of PYY1-36 such as amino acid 16-21 of PYY 1-36 as defined in sequence ID NO 1 (Glu Leu Asn Arg Tyr Tyr ), or such as amino acid 15-22 of PYY 1-36 as defined in sequence ID NO 1 (Glu Glu Leu Asn Arg Tyr Tyr Ala), or such as amino acid 14-23 of PYY 1-36 as defined in sequence ID NO 1 (Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser), or such as amino acid 13-24 of PYY 1-36 as defined in sequence ID NO 1 (Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu), or such as amino acid 12-25 of PYY 1-36 as defined in sequence ID NO 1 (Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg), or such as amino acid 11-26 of PYY 1-36 as defined in sequence ID NO 1 (Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg
  • the functional equivalent may be PYY 2-36 or alternatively, the functional equivalent may be PYY4-36.
  • the functional equivalent comprise any of the above sequences with conservative amino acid substitutions, such as one substitution, or such as two substitutions, or such as two substitutions, or such as more than two substitutions, or such as more than four substitutions.
  • the functional equivalent may comprise any type of modifications. Nearly 200 structurally distinct covalent modifications have been identified thus far, ranging in size and complexity from conversion of amides to carboxylic acids, to the attachment of multiple complex oligosaccharides. Such modifications include phosphorylation, acetylation, ubiquination, lipidation (acetylation, prenylation, farnesylation, geranyla- tion, palmitoylation, myristoylation), methylation, carboxylation, sulfunation and O- or N-glycosylations.
  • vitamin C as a cofactor. This include proline and lysine hydroxylations and carboxy terminal amidation.
  • PYY or the functional equivalent comprise a C-terminal amida- tion.
  • the C-terminal tyrosine residue of PYY or a functional equivalent is amidated.
  • the functional equivalent may comprise 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 al- kenyl, aryl, -C1-6 alkyl aryl, -C(O)- (CH2) 1-6-COOH, -C(0)-C1-6 alkyl, -C(0)-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 ethyla- mide.
  • PYY or the functional equivalent of PYY may conjugated to another entity, in order for example, to prolong its half-life. Conjugation can improve the delivery of targeted doses, prevent breakdown, and increase bioavailablity in circulation.
  • the other entity may be any molecule capable of providing the desired effect.
  • the preparation of conjugates is well known in the art se for example Hermanson GT. Bioconjugate Techniques. New York: Academic Press; 1996, Aslam M, Dent AH. Bioconjugation: protein coupling techniques for the biomedical sciences. Houndsmills, England: Macmillan Publishers; 1999, and Wong SS. Chemistry of protein conjugation and cross/inking. Boca Raton, FL: CRC Press; 1991.
  • linkers that contain a disulfide bond that is sterically "hindered” are often preferred, due to their greater stability in vivo, thus preventing release of the active moiety prior to binding at the site of action. It is generally desired to have a conjugate that will remain intact under conditions found everywhere in the body except the intended site of action, at which point it is desirable that the conjugate have good "release" characteristics.
  • the molecules may be conjugated as described above, or by peptide bonds, before or after synthesis and purification.
  • PYY or a functional equivalent thereof may be fused to the Fc portion of an antibody or to albumin in order to ex- tend the half-life of the conjugated molecule.
  • the fusion may be obtained by any suitable methods, for example, but not exclusively, by recombinant DNA technology.
  • the fusion of is made by recombinant DNA technology, such as a fusion of the nucleotide sequence encoding PYY and the nucleotide sequence encoding Fc portion or albumin is made and the fusion is thereby encoded by a single nucleotide sequence.
  • the fusion polypeptide may be expressed and purified as a single polypeptide molecule, using any suitable method, as described for the purification of PYY.
  • the fusion polypeptide may include insertion of a linker, such as a peptide of at least 2 AA, such as at least 5 AA, such as at least 8 AA, such as at least 15 AA, such as at least 20 AA. Suitable linkers are known in the art. Methods for production and purification of PYYY
  • PYY or functional equivalents thereof can be produced using techniques well known in the art.
  • the PYY or functional equivalent for use herein is of GMP grade.
  • a polypeptide region of a PYY can be chemically or biochemi- cally synthesized and modified.
  • Techniques for chemical synthesis, such as solid- phase synthesisof polypeptides (see example 2) are well known in the art. (See e.g. Vincent in Peptide and Protein Drug Delivery, New York, N.
  • PYY or the functional equivalent may be produced by chemical synthesis, e.g. as described described in brief below, or by recombinant methods.
  • PYY or the functional equivalent may be produced by the following process:
  • the host cell may be cotransfected with a second vector for optimization of the production process.
  • the two vectors may contain different selectable markers.
  • the cod- ing sequences of PYY may comprise cDNA or genomic DNA or both.
  • the host cell used to express the PYY may be either a bacterial cell such as Es- cherichia coli, or a eukaryotic cell, such as S. cerevisiea or P. pastoris.
  • a mammalian cell line may be used, such as Hela, CHO or any other suitable host cell known by the person skilled in the art.
  • the general methods by which the vectors may be constructed, transfection methods required to produce the host cell and culture methods required to produce PYY or functional equivalent from such host cells are all conventional techniques.
  • the PYY or functional equivalent may be purified according to standard procedures as described below.
  • the method of purification used is dependent upon several factors, including the purity required, the method of production and the intended use. Such methods are known in the art.
  • Any suitable conventional methods of purifying polypeptides including precipitation and column chromatography are well known to one of skill in the purification arts, including cross-flow filtration, HPLC, ammonium sulphate precipitation, affinity col- umn chromatography, gel electrophoresis and the like may be used.
  • the PYY composition to be used in the delivery device of the invention comprises PYY or a functional equivalent thereof as the active ingredient.
  • the PYY composi- tion is preferably free of any contaminants present in blood such as infectious agents.
  • the PYY composition is formulated so as to be suitable for administration via a delivery device of the invention.
  • the PYY composition have a concentration of at least 10 nM such as at least 50 nM, such as at least 0.2 ⁇ M, such as at least 0.5 ⁇ M, such as at least 1 ⁇ M, such as at least 2 ⁇ M, such as at least 5 ⁇ M, such as at least 10 ⁇ M, such as at least 20 ⁇ M, such as at least 50 ⁇ M, such as at least 0.2 mM, such as at least 0.5 mM, such as at least 1 mM, such as at least 2 mM, such as at least 5 mM of PYY or a functional equivalent thereof.
  • the PYY composition may be stored as a dry composition, for example lyophilized (freeze dried) or spray dried to improve the stability of PYY.
  • Such compositions are reconstituted with liquid solutions prior to use.
  • the protein concentration of the reconstituted formulation is about 2-40 times greater than the protein concen- tration in the mixture before lyophilization, thus this allows the production of PYY composition of high concentration.
  • a diluent comprising a preservative (such as bacteriostatic water for injection, BWFI)
  • the reconstituted formulation may be used as a single-dose or multi-dose formulation.
  • a formulation is useful, for example, where the patient requires frequent subcutaneous administrations.
  • the PYY composition may be a liquid composition of high stability.
  • the PYY composition may be meant for mixing with a suitable diluent prior to use.
  • the PYY composition to be used in the delivery device of the invention may be mixed with a suitable diluent to form a pharmaceutical composition prior to use.
  • the mixing may be performed by the patient prior to insertion of the container in the delivery device, such as the single or multi dose delivery device, or by insertion of a two compartment container in a single or multi dose deliver device especially adapted to allow mixing of two components.
  • the single or multi dose delivery device may accommodate two containers and allow mixing of the content of the two prior to use.
  • compositions comprising PYY or a functional equivalent thereof to be used in the delivery device of the present invention may be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy 1995, edited by E. W. Martin, Mack Publishing Company, 19th edition, Easton, Pa.
  • the compositions may appear in conventional forms, for example solutions or suspensions.
  • compositions, carriers, diluents and reagents are used interchangeably and represent that the materials are capable of administration to or upon an individual without the production of undesirable physiological effects such as nausea, dizziness, gastric upset and the like.
  • the pharmaceutical composition preferably further comprises pharmaceutically acceptable salts, a pharmaceutically acceptable carrier or a diluent.
  • the pharmaceutical composition may further comprise transport molecules.
  • the compositions may preferably be delivered to an individual in any way so as to achieve a beneficial effect, preferably by inhibiting appetite and/or preventing malnutrition, and/or improving the individual's sense of well-being or quality of life.
  • the delivery device of the invention is useful for parenteral administration of a PYY composition as described herein, such as via a subcutaneous, intradermal or intramuscular route via a delivery device according to the invention.
  • the composition is administered via the subcutaneous route.
  • the PYY or functional equivalent thereof may be administered with at least one other compound as discussed further below.
  • the compounds may be administered simultaneously, either as separate compositions or combined in a unit dosage form, or administered sequentially.
  • the pharmaceutical composition may be produced prior to use by mixing of a PYY composition with an appropriate diluent.
  • salts of PYY or a functional equivalent where they can be prepared, are also intended to be covered by this invention. These salts will be ones which are acceptable in their application to a pharmaceutical use. By that it is meant that the salt will retain the biological activity of PYY or the functional equivalent and the salt will not have untoward or deleterious effects in its application and use in treating diseases.
  • compositions are prepared in a standard manner. If PYY or the functional equivalent is a base it is treated with an excess of an organic or inorganic acid in a suitable solvent. If PYY or the functional equivalent is an acid, it is treated with an inorganic or organic base in a suitable solvent.
  • Such salts include pharmaceutically acceptable acid addition salts, pharmaceutically acceptable metal salts, ammonium salts and alkylated ammonium salts.
  • Acid addition salts include salts of inorganic acids as well as organic acids. Examples of pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, p-toluenesulphonic acids, and aryl- sulphonic acid.
  • metal salts examples include lithium, sodium, potassium and magnesium salts and the like.
  • organic acid salts of organic acids such as for example acetic acid is preferred.
  • ammonium and alkylated ammonium salts include ammonium, methyl- ammonium, dimethylammonium, trimethylammonium, ethylammonium, hy- droxyethylammonium, diethylammonium, butylammonium and tetramethylammo- nium salts and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 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.
  • 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.
  • the active ingredient or a salt thereof can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein.
  • 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.
  • 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.
  • Suitable pharmaceutical carriers include inert solid diluents or fillers, sterile aqueous solution and various organic solvents.
  • 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.
  • the active compound of the invention may be unstable, thus the composition preferably contain stabilizers, preservatives or conservatives to increase the stability of the compounds.
  • Tween 60, Span 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate, mannitol and sodium lauryl sulphate are possible stabilizers.
  • mannitol may be used as stabilizer.
  • ad lyoprotectant may be used to stabilize the active ingredient (Townsend and DeLuca, "Use of lyoprotectants in the freeze-drying of a model protein, ribonuclease A" Journal of Parenteral Science & Technology 42 (6): 190-199 (Nov.-Dec. 1988)).
  • the lyoprotectant may preferably be a sugar such as sucrose or trehalose such as sucrose, dextran, or hydroxypropyl-/142-cyclodextrin.
  • a pH-buffering agent may be used to stabilize PYY or functional equivalent in the composition.
  • the buffering agent may be acetate, carbonate, bi- carbonate, phosphate, citrate, tris or hepes. In a preferred embodiment the buffering agent is acetate.
  • the composition preferably has a pH between 2.0 and 9.0, or such as between 2.5 and 8.0, or such as 3.0 and 7.0, or such as between 3.5 and 6.0, or such as between 3.5 and 5.0 or such as between 4.0 and 5.5, or such as between 4.0 and 5.0, or such as between 4.0 and 4.5.
  • the pH of the compositions is less than 6, preferably less than 5.5, preferably less than 5, preferably less than 4.8, preferably less than 4.6, preferably less than 4.4, preferably less than 4.2.
  • Transport molecules act by having incorporated into or anchored to it PYY or functional equivalent. Any suitable transport molecules known to the skilled person may be used. Examples of transport molecules may be liposomes, micelles, and/or mi- crospheres.
  • 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". The shape of micelles formed in dilute surfactant solutions is approximately spherical.
  • the polar head groups of the surfactant molecules are arranged in an outer spherical shell whereas their hydrocarbon chains are oriented toward the center, forming a spherical core for the micelle.
  • the hydrocarbon chains are randomly coiled and entangled and the micellar interior has a nonpolar, liquid-like character.
  • the polyoxyethylene moieties are oriented outward and permeated by water. This arrangement is energetically favourable since the hydrophilic head groups are in contact with water and the hydrocarbon moieties are removed from the aqueous medium and partly shielded from contact with water by the polar head groups.
  • the hydrocarbon tails of the surfactant molecules, located in the interior of the micelle interact with one another by weak van der Waals forces.
  • the size of a micelle or its aggregation number is governed largely by geometric factors.
  • the radius of the hydrocarbon core cannot exceed the length of the extended hydrocarbon chain of the surfactant molecule. Therefore, increasing the chain length or ascending homologous series increases the aggregation number of spherical micelles. If the surfactant concentration is increased beyond a few percent and if electrolytes are added (in the case of ionic surfactants) or the temperature is raised (in the case of nonionic surfactants), the micelles increase in size. Under these conditions, the micelles are too large to remain spherical and become ellipsoidal, cylindrical or finally lamellar in shape.
  • Suitable surfactants include sodium laureate, sodium oleate, sodium lauryl sulfate, octaoxyethylene glycol monododecyl ether, octoxynol 9 and PLURONIC F-127 (Wy- andotte 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.
  • phos- pholipids such as those described for use in the production of liposomes, may also be used for micelle formation.
  • the PYY composition may further include the transport molecules as described above.
  • the pharmaceutical compositions described herein are then readily administered in a variety of dosage forms suitable for the disclosed routes of administration via a delivery device of the invention.
  • the compositions may conveniently be presented in unit dosage form or in multiple dosage form by methods known in the art of phar- macy.
  • compositions for. parenteral administration are provided.
  • compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example solutions in aqueous polyethylene glycol.
  • oily or nonaqueous carriers, diluents, solvents or vehicles include propyl- ene glycol, polyethylene glycol, vegetable oils (e.g., olive oil), and injectable organic esters (e.g., ethyl oleate), and may contain formulatory agents such as preserving, wetting, emulsifying or suspending, stabilizing and/or dispersing agents.
  • 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 suit- able vehicle, e.g., sterile, pyrogen-free water.
  • a suit- able vehicle e.g., sterile, pyrogen-free water.
  • Aqueous solutions should be suitably buffered if necessary, and the liquid diluents first rendered isotonic with sufficient saline or glucose.
  • the aqueous solutions are particularly suitable for intramuscular, subcutaneous and intradermal administration.
  • the sterile aqueous media employed are all readily available by standard techniques known to those skilled in the art.
  • the composition comprising PYY or a functional equivalent thereof or a salt thereof is a lyophilised composition and the composition may further comprises a solvent.
  • the composition is a solution of PYY or a functional equivalent thereof or a salt thereof.
  • the solvent may be any suitable solvents, such as described herein, and preferably the solvent is saline or a physiological buffer like phosphate buffer.
  • compositions for administration with the delivery device of the invention may include sterile aqueous solutions that may also contain buffers, liposomes, diluents and other suitable additives.
  • Oils useful in parenteral compositions include animal, vegetable, or synthetic oils. Specific examples of 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. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral compositions include fatty alkali metal, ammonium, and triethanolamine salts, and 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 alkanola- mides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-.beta.-aminopropionates, and 2-alkyl-imidazoline quater- nary ammonium salts, and (e) mixtures
  • the parenteral compositions typically will contain from about 0.00002 % to about 2 % 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 compo- sitions 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 sus- pensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the pharmaceutical dosage forms suitable for injection can include sterile aqueous solutions or dispersions comprising the active ingredient that are adapted for ad- ministration 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 PYY or a functional equiva- lent thereof or pharmaceutically acceptable salt thereof in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the pharmaceutical composition may be prepared so it is suitable for one or more particular administration methods via the delivery device of the invention.
  • composition comprising PYY or a functional equivalent thereof can be administered parenterally by injection using a delivery device of the invention, such as, in- tramuscularly, subcutaneously or intradermally.
  • a suitable dose of the compositions described herein is administered in pharmaceutically effective amounts to an individual in need of such treatment.
  • pharmaceutically effective amounts is defined as a total amount of the pharmaceutical composition or the total amount of each active component of the pharmaceutical composition that is sufficient to show a meaningful patient benefit.
  • the pharmaceutical compositions containing PYY or a functional equivalent thereof are conventionally administered as a subcutaneous injection of a unit dose.
  • unit dose and "unit dosage form” as used herein refer to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of PYY or functional equivalent thereof, alone or in combination with other agents, calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier, or vehicle.
  • the specifications for the unit dosage forms depend on the particular compound or compounds employed and the effect to be achieved, as well as the pharmacodynamics associated with each compound in the individual to be treated.
  • the dose administered should be an "effective amount” or an amount necessary to achieve an "effective level” in the individual patient.
  • the pharmaceutical composition may be prepared in multiple dose form and by use of the multi dose delivery device, single doses are administrated when needed.
  • the effective level is used as the preferred endpoint for dosing, the actual dose and schedule can vary, depending on individual differences in pharmacokinetics, drug distribution, and metabolism.
  • the "effective level” can be defined, for example, as the blood or tissue level desired in the patient that corresponds to a concentration of one or more compounds to be used in accordance with the invention.
  • the effective level may be defined as the dose required to achieve a plasma concentration of at least, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140 or 150 pmol/l or alternatively a plasma concentration of 20-50, 30-100, 40-80, 50 -70 or approximately 60 pmol/l.
  • the optimal quantity and spacing of individual dosages of a compound or a pharmaceutically acceptable salt thereof will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular patient being treated, and that such optimums can be determined by conventional techniques. It will also be appreciated by one of skill in the art that the optimal course of treatment, i.e., the number of doses of a compound or a pharmaceutically acceptable salt thereof given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests.
  • treatment using the delivery system of the invention is capable of reducing symptoms of the condition being treated or a melioration of the quality of life of the patient. This may be a decrease in food intake, a decrease in pain sensation or a normalisation of bowel habits, or a normalised eating pattern. Such effects are readily recognised by the skilled artisan.
  • the dosage will vary depending on the compound employed and the mode of administration. Dosage levels may vary between about 4 ng/kg body weight to 20 ⁇ g/kg body weight daily, preferably between about 10 ng/kg body weight to 1 ⁇ g/kg body weight, more preferably between 50 to 750 ng/kg body weight.
  • the dosage may be administered when needed, such as up to ten times daily, such as one to five times daily, such as two or three times daily, or preferably such as once a day, thus the daily dosage maybe up to 2-5 times the dosages mentioned above. Alternatively, the dosage may be administered less frequently than once daily as described herein above.
  • a preferred dosage of a composition employed according to the invention is in a concentration equivalent to PYY or a functional equivalent there of from 4 ng to about 20 ⁇ g per kg bodyweight, or such as from 10 ng to 1 ⁇ g per kg bodyweight, more preferably from 50 to 750 ng per kg bodyweight.
  • the dosage of PYY is preferably 20-200 ng/kg, 20-160 ng/kg, 40-160 ng/kg, 40-120 ng/kg, 40-80 ng/kg, 60- 120 ng/kg or such as approximately 60 ng/kg or 80 ng/kg.
  • the preferred dosages may be in a concentration equivalent to PYY or a functional equivalent there of from 1 pmol/kg to 5 nmol/ kg, such as from 5 pmol/kg to 1 nmol/kg, or such as from 20 pmol/kg to 500 pmol/kg, such as from 40 to160 pmol- /kg, such as from 75 to 120 pmol/kg.
  • the dosage may be 5-50 pmol/kg, 5-40 pmol/kg, 5 to 30 pmol/kg, 10-40 pmol/kg, 10-30 pmol/kg such as 5 to 25 pmol/kg, such as 5 to 20 pmol/kg, and most preferably 10-20 pmol/kg, 15-30 pmol/kg or approximately 15 pmol/kg or 20 pmol/kg.
  • the dosages are preferably administrated once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day, or such as more than five times a day.
  • the compositions are administered in dosages of PYY or a functional equivalent from about 400 ng to about 2 mg, more preferably from about 10 ⁇ g to about 200 ⁇ g, or from about 5 ⁇ g to about 250 ⁇ g, more preferably from about 20 ⁇ g to about 200 ⁇ g, more preferably from about 20 ⁇ g to about 100 ⁇ g.
  • the dosage may be 1-20 ⁇ g, 2-16 ⁇ g, 4-16 ⁇ g, 4-12 ⁇ g, 4-8 ⁇ g, 6-12 ⁇ g, 6-10 ⁇ g or approximately 8 ⁇ g.
  • the composition is administered in dosages of PYY or a functional equivalent from 100 pmol to 500 nmole, or such from 500 pmol to 100 nmol, or such as from 1 nmol to 50 nmol, or such as from 2 to 25 nmol, or such as from 4 to 20 nmol.
  • the preferred dosage may be 0,25-5 nmol, 0,5-4 nmol, 1-4 nmol, 1-3 nmol, 1-2 nmol, 1 ,5-3 nmol or more preferably 1 ,5-2,5 nmol or most preferably approximately 2 nmol.
  • the PYY or functional equivalent is administered subcuta- neously in a dosage of 5-30 pmol/kg, such as 5-25 pmol/kg, such as 5-20 pmol/kg or such as 10-20 pmol/kg bodyweight, in order to achieve an effective level in the indi- vidual treated.
  • the presently preferred dosage is 10-20 pmol/kg bodyweight of
  • the dosages of PYY or the functional equivalent is preferably administered once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day.
  • the pharmaceutical preparations described herein may also be arranged in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as powders in compartments.
  • the powders may be mixed with a solvent prior to or during use.
  • the PYY composition is administrated in unit dosage form, from about 400 ng to about 2 mg of PYY or a functional equivalent thereof, more preferably from about 10 ⁇ g to about 200 ⁇ g, or from about 5 ⁇ g to about 250 ⁇ g, more preferably from about 20 ⁇ g to about 200 ⁇ g, more preferably from about 20 ⁇ g to about 100 ⁇ g.
  • the unit dosage form may comprise from 100 pmol to 500 nmole, or such from 500 pmol to 100 nmol, or such as from 1 nmol to 50 nmol, or such as from 2 to 25, such as from 4 nmol to 20 nmol. of PYY or a functional equivalent thereof.
  • the compositions are preferably administered once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day.
  • the dosage requirements will vary with the particular drug composition employed, the route of administration and the particular subject being treated.
  • a patient to be treated by the present method will receive a pharmaceutically effective amount of the compound in the maximum tolerated dose, generally no higher than that required before drug resistance develops
  • Suitable dosing regimens 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 normal PYY response which occurs during the cause of a meal is a short-lived surge in plasma concentrations of PYY and that due to the relative short half life of the peptide an injection, e.g. subcutaneous injection, of PYY will ensure that a similar short-lived peak on PYY concentrations can be obtained. This may depend on the thickness of the adipose tissue, as the blood flow decrease pro- portionally to the thickness of the fat layer, and therefore the clearance from the tissue is assumed to follow a similar curve.
  • administration may using a formulation with a prolonged half life may give a stable concentration of PYY, thus decreasing the need for regular meal associated injections.
  • each administration being within 120 minutes of a meal, such as within 110 minutes of a meal, such as within 100 minutes of a meal, such as within 90 minutes of a meal, such as within 85 minutes of a meal, such as within 80 minutes of a meal, such as within 75 minutes of a meal, such as within 70 minutes of a meal, such as within 65 minutes of a meal, such as within 60 minutes of a meal, such as within 55 minutes of a meal, such as within 50 minutes of a meal, such as within 45 minutes of a meal, such as within 40 minutes of a meal, such as within 35 minutes of a meal, such as within 30 minutes of a meal, such as within 25 minutes of a meal, such as within 20 minutes of a meal, such as within 15 minutes
  • the PYY composition is administrated dependent on the sensing of appetite in the individual.
  • the dosing regime may be different.
  • Administration may aim at obtaining a low basal level of PYY and additionally obtaining PYY peeks associated with the meals. This may include administration of different PYY composition with different timing.
  • PYY or a functional equivalent may be administered with at least one other com- pound.
  • the compounds may be administered simultaneously, either as separate compositions or combined in a unit dosage form, or administered sequentially.
  • Administration of the PYY composition may be used in combination with any suitable therapy.
  • treatment “in combination” is meant that another treatment regime may be carried out before, during (including concurrently with) and after treatment of an individual with a PYY composition as described herein.
  • the PYY composition of the invention is administered in combination with a second pharmaceutical composition.
  • Said second pharmaceuti- cal composition may be administered together or separately from the PYY composition.
  • said second pharmaceutical composition may comprise compound(s) selected from the group of; anti-hypertensives including (but not lim- ited to) beta-adrenoceptor antagonists, alpha-adrenoceptor antagonists, calcium antagonists, angiotensin converting enzyme (ACE) inhibitors, angiotensin II receptor antagonists and diuretics (including, but not limited to, thiazide diuretics and loop diuretics) , insulin, DPP-IV inhibitors, anti-depressant medications including (but not limited to) selective serotonin reuptake inhibitors (SSRIs), mianserine and mirtazap- ine, classical as well as atypical antipsychotic drugs (neuroleptics), corticosteroids as well as any other drug(s) that may decrease body weight or body fat mass or that may be indicated for the treatment of hypertension, overweight, obesity, syndrome X and/or diabetes mellitus.
  • anti-hypertensives including (but not lim- ited to)
  • delivery device may be used according to the invention. Dependent on the treatment to be administrated the recipient and other factors the most suitable delivery device may be selected.
  • a single dose delivery device comprises a therapeutically effective amount of PYY or a functional equivalent thereof, suitable for injection by a single injection.
  • a multi dose delivery device comprise at least two dosages of a therapeutically effective amount of PYY or a functional equivalent thereof, suitable for injection by at least two injection.
  • An aspect of the invention relates to a delivery device for delivering PYY or a functional equivalent thereof, comprising at least one container or compartment comprising a therapeutically effective amount of PYY or a functional equivalent thereof, dosing means, ejecting means and optionally a needle assembly.
  • a disposable injection device may be preferred due to convenience. There is no handling of containers and needles involved in the use and further there is no need for appropriate storage of the injection device in between injections. Disposable de- livery devices are generally smaller and therefore more discrete than conventionally reusable delivery devices.
  • a disposable delivery device may be used for delivery of PYY or a functional equivalent thereof.
  • the disposable delivery device for delivering of PYY or a functional equivalent thereof comprises according to the invention a pharmaceutical composition, a needle and ejection means and optionally dosing means.
  • Multi dose delivery device The advantage of a multi-dose delivery device is that it facilitates ease of use for the patient, reduces waste by allowing complete use of container contents, and results in a significant cost savings for the manufacturer since several doses are packaged in a single container (lower filling and shipping costs).
  • a multi dose delivery device may be used for delivery of PYY or a functional equivalent thereof.
  • a multi dose delivery device for delivering of PYY or a functional equivalent thereof comprise at least one container assembly, dosing means, ejecting means and optionally a needle assembly.
  • the delivery device comprise containers and/or compartments for storage of the therapeutically effective amount of PYY or functional equivalent thereof.
  • the delivery device may further comprise assembly means of the container or compartment.
  • the delivery device comprises at least one container assembly capable of accepting a container.
  • the container has one compartment filled with a pharmaceutical composition.
  • the container in the delivery device has two compartments. Either of said two compartments may comprise a pharmaceutical composition according to the invention and the second may comprise either a solvent or solute meant for mixing with the pharmaceutical composition or a second pharmaceutical composition.
  • the two compartments are constructed to allow mixing of the contents of the two compartments prior to use.
  • the compartments may be connected via a liquid channel that can be open manually/mechanically or electronically prior to use to allow mixing of the contents of the two compartments or alternatively the two compartments are connected by a breakable sealing.
  • sealing that can be broken mechanically or electronically prior to use to allow mixing of the contents of the two compartments prior to injection.
  • the sealing may for example be broken by means of a plunger or a needle which may be controlled manually/mechanically or by electronic means.
  • the sealing may be a movable plunger or piston.
  • the delivery device comprises two compartments.
  • the two compartments are located in continuation of each other and separated by an intervening first plunger.
  • the first compartment closest to the needle contains PYY or a functional equivalent, preferably in lyophilized form.
  • the second compartment is closed with a second plunger. A long the axis of the first compartment a channel formed as an axial indentation in the wall of the compartment is present. When the device is used the second plunger is pressed into the device and through the second compartment.
  • the pressure on the second plunger will result in pressure on the first plunger and moving of the first plunger past the opening of the channel allowing the liquid of the second compartment to access to the channel.
  • the solvent can pass the first plunger and flow from the second compartment into the first compartment, in which the PYY or functional equivalent thereof is dissolved to obtain a solution.
  • the second plunger is pressed further in the pressure on the first plunger is increased whereby the solution now present in the first compartment is injected through the needle located in the front part of the first compartment.
  • the delivery device comprises two containers.
  • Either of said two containers may comprise a pharmaceutical composition according to the invention and the second may comprise either a solvent or solute meant for mixing with the pharmaceutical composition or a second pharmaceutical composition.
  • the content of said two containers can be mixed prior to use.
  • the containers may be connected by a liquid channel; said liquid channel may be formed by a needle. Said liquid channel may be opened manually, mechanically or by electronic means prior to use. Thereby, the content of the two containers can be mixed before injection into the patient.
  • the delivery device may be reusable.
  • at least one container in the delivery device can be replaced.
  • at least one container in the delivery device may be refillable.
  • the two containers may be replaced or refilled separately.
  • An embodiment of the invention relates to a delivery device comprising at least one container assembly, wherein said container assembly comprise a distal end comprising coupling means for realisably mounting a needle assembly, and said container assembly comprise a proximal end comprising coupling means for engaging the dosing assembly.
  • the pen may further comprise an automated injection system, for example controlled by a spring.
  • the multi dose delivery device may be designed with a dosing assembly to allow the dosing of the pharmaceutical composition to be changed by the patient.
  • the dosing assembly may be adjustable for different doses.
  • the dosages of the compositions may controlled separately.
  • the dosing assembly may for example be controlled by means of a plunger or by electronic means.
  • the length of the needle may be important for correct delivery of the PYY composition.
  • the preferred delivery device may be able to accept needles of different length.
  • the needles used may be at least 0.5 cm in length, such as more than 1.0 cm, such as more than 1.5 cm, such as more than 2.0 cm or such as more than 2.5 cm in length, such as more that 3 cm in length, such as more than 4 cm in length, such as more than 5 cm in length, such as more than 6 cm in length, such as more than 7 cm in length, such as more than 8 cm in length, such as more than 9 cm in length, such as more than 10 cm in length.
  • the needle is 0.5-1.0 cm long, 1.0-1.5 cm long, or such as 1.5-2.0 cm long, or such as 2.0-2.5 cm long, or such as 2.5-3,0 cm long, or such as 3.0-4.0 cm long, or such as 4.0-6.0 cm long, or such as 6.0-8.0 cm long, or such as 8.0-10.0 cm long.
  • the delivery device maybe designed to accept needles of different diameter.
  • the diameter of the needle relate to the outer diameter of the needle.
  • the diameter of the needle is at the most 0.50 mm, such as at the most 0.40 mm, such as at the most 0.35 mm, such as at the most 0.33 mm, such as at the most 0.30 mm, such as at the most 0.20 mm, such as at the most 0.15 mm.
  • the diameter of the needle is 0.30-0.35 mm, or such as 0.30-0.33 mm, or such as 0.33mm -0.35 mm, or such as 0.35-0.40 mm.
  • the needle may be changed by the patient.
  • the needle may be hidden for the user. This may be beneficial for people suffering from fear of injection.
  • a needle based delivery device of the present invention can be developed on the basis of devices manufactured by Innoject, Ypsomed, Becton Dickenson and Owen
  • the delivery system may be needle free.
  • the pharmaceutical composition may be delivered by pressure whereby a micro-thin stream of the pharmaceutical composition penetrates the skin and is deposited into the subcutaneous (fatty) tissue.
  • needle-free systems are those manufactured by Cross Ject, BioJect, Aradigm, Antares Pharma, Alza, Injext, and Biovalve.
  • the injection device includes means for controlling the depth of injection, thereby it is possible to reach different preferred injection sites using the same type of needles.
  • the depth of injection may vary from 1.0 cm to 10 cm.
  • the injection depth may be 0.5-1.0 cm, 1.0-1.5 cm, or such as 1.5-2.0 cm, or such as 2.0-2.5 cm, or such as 2.5-3,0 cm, or such as 3.0-4.0 cm, or such as 4.0-6.0 cm, or such as 6.0-8.0 cm, or such as 8.0-10.0 cm.
  • the pen may include the possibility to control the injection pressure and/or the injection rate.
  • the pen may comprise an electronic control unit, such control unit may allow the user to store information, such as preselected dosages, timing and mode of injection.
  • the control unit may include an alarm function and thereby alert the patient about the need for injection by making a sound, light signal, or physical movement (vibration).
  • the alarm signal may be transmitted to a unit carried by the patient.
  • the control unit may indicate positions for injections, dosage, and further indicate if the pen is positioned correct for injection. Injection may be blocked by the electronic control unit to avoid incorrect usage.
  • the injection pressure may be controlled.
  • control unit is capable of registering the force (or measure the resistance) used to insert the needle and there upon control the depth/tissue of injection. Using this function the pharmaceutical composition is always injected correctly also in difficult positions and the possible injection sites is thereby increased.
  • the material used for production of the container may be any suitable material.
  • the product should be suitable for storage of the pharmaceutical composition this include being non-sticky for the components of the pharmaceutical composition and stable at various temperatures used for storage of pharmaceutical compositions.
  • at least one container may be at least partly transparent and may further comprise a scale.
  • at least one container is made of plastic or glass.
  • the container may be made of both metallic and non-metallic materials.
  • the metallic surfaces can be comprised of, for example, titanium, stainless steel and other suitable materials.
  • the non-metallic container surfaces can be comprised, for example, of a glass material of, a polymeric material, such as a polyurethane, a polyethylene, a polypropylene, a polyvinylchloride or for example a rubber such as bromobutyl rubber or chlorobutyl rubber, or other similar polymeric materials.
  • the container can be made of a polymeric material, such as those listed above, or can be formed from a polymer laminate (e.g., two or more layers of different polymeric materials).
  • the inner surface of at least one container may comprise a coating.
  • the coating may comprise polymeric materials such as hydrophilic polyurethanes, polyureas, acrylics, polycarbonates, polyvinyl pyrollidone or other hydrophilic materials, in particular materials such as polyethylene glycols, polyethylene/polypropylene glycol copolymers or other poloxamers which may be chemically (covalently) attached to the surface.
  • the coating may further comprise polymers of the various paraxylylenes.
  • the preferred coating may be a silicon-containing coating, such as a poly(dialkyl siloxane) oil or copolymer.
  • the poly(dialkyl siloxane) is selected from poly(dimethyl siloxane) (PDMS), poly(dipropyl siloxane) or poly(dihexyl siloxane).
  • the coating comprises a linear or branched hydrophilized poly(dialkyl siloxane) oil.
  • the coating comprises a crosslinked or gelled silicon oil, such as a hydrophilized poly(dialkyl siloxane) oil, or a mixture of a crosslinked and a non-crosslinked oil.
  • a cross-linked, or cured, silicon oil is typically obtained by applying a linear, or branched, silicon oil with reactive functionalities which are used to cross-link the coating in a subsequent step.
  • the coating comprises a hydrophilized poly(dialkyl siloxane) block and graft copolymer.
  • the copolymer may be any block and graft copolymer which comprises polymeric segments of poly(dialkyl siloxane), such as
  • the polymeric segments may, for example, be combined with polymeric segments of polystyrene, polyolefins, polyamides, or polyurethane to form the desired copolymer.
  • the copolymer may be prepared by any method available, for example by sequential anionic polymerization, or different grafting procedures. Hydrophilicity of the coating according may be obtained by any appropriate method.
  • the coating is subjected to an oxidative treatment, such as plasma treatment or corona treatment after having been applied to the component.
  • the coating comprises a copolymer which is made hydrophilic by end-capping the copolymer with hydrophilic group or chain segments.
  • the hydrophilic group may, for example, be a negatively charged chemical group or phosphorylcholine (PC) groups
  • the chain segment may, for example, be poly(ethylene oxide) (PEO) or poly(2-hydroxyethyl methacrylate) (pHEMA).
  • the surfaces may be further modified in order to further decrease the protein adsorption by coupling of hydrophilic polymer segments or functional groups.
  • These polymer segments or functional groups may be of the same kind as those described above, and may further be coupled to the functional groups generated during the plasma treatment.
  • the hydrophilic groups at the coating will tend to seek into the coating leaving the surface hydrophobic due to the hydrophobicity of the surrounding air. Accordingly, it is of great importance that the coating remains hydrophilic during storage until the medical article is filled with the protein solution. This may be secured by placing the coated article in a hydrophilic environment, such as by filling the medical article with the protein formulation shortly after the coating process.
  • the thickness of the coating depends on the specific coating, and is preferably from 0.005 to 10 ⁇ m, such as from 0.01 to 1 ⁇ m or such as 0.05 to 0.4 ⁇ m, or such as from 0.0 5 to 0.25 ⁇ m, more preferably approximately 0.2 ⁇ m.
  • the optimal thickness depends on the dimensions and shape of the container and it can easily be performed by one skilled in the art. If the coating is too thin the coating may be torn in use, thereby increasing the friction between the two components. When the thickness of the coating has reached a certain plateau value the friction forces are approximately constant even when the thickness is further increased.
  • the delivery device such as a single dose or multi dose delivery device comprises at least one container or compartment comprising a pharmaceutical composition.
  • the pharmaceutical composition present in the delivery device is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or any combinations hereof.
  • the pharmaceutical composition present in the delivery device is; a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or any combinations hereof.
  • the pharmaceutical composition present in the delivery device is; a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia
  • said pharmaceutical composition comprises PYY or a functional equivalent thereof.
  • a single or multi dose delivery device may be used for the administration of a PYY composition as described above.
  • the delivery device such as the single or multidose delivery device comprises in addition to the PYY composition also a second pharmaceutical composition.
  • the single or multidose delivery device may comprise separate dosing and administration means for the PYY composition and the second pharmaceutical composition.
  • compositions of the present invention are self-administered.
  • COS-7 cells can be grown Dulbecco's Modified Eagle'e Medium 1885 supplemented with 10% fetal calf serum, 2mM glutamine and
  • the expression plasmids containing the cDNAs encoding the wild type or the mutated receptors can be transiently expressed after transfetion according to the calcium phosphate precipitation method and assay can be performed 48 hour after transfection.
  • Binding assay One day after transfection the cells will be transferred and seeded in multi-well plates for assay. The number of cells to be plated per well will be chosen so as to obtain 5 to 10% binding of the radioligand added. Two days after transfection the cells will be assayed in competition binding assays using 125 l- PYY(3-36) as a tracer.
  • Radioligand will be bound in a buffer composed of 0.5 ml of 50 mM Hepes buffer, pH 7.4, supplemented with 1 mM CaCI 2 , 5 mM MgCI 2 , and 0.1% BSA, and displaced in a dose dependent manner by unlabelled ligands.
  • the assay will be performed in duplicate for 3 hours at 4 °C, and stopped by washing twice in the buffer.
  • Cell associated, receptor bound radioligand will be determined by the addition of lysis buffer (48% urea, 2% NP-40 in 3M acetic acid). The concentration of radioli- gand in the assay corresponds to a final concentration of approximately 20 pM.
  • COS-7 can be cultured as described above and contransfections can be performed.
  • the activation of Phospholipase C by chimeric G-proteins (Conklin B) formed between both G ⁇ q and G ⁇ i the Y2 receptor can be measured through the inositol phosphate (IP) turnover in the cell.
  • IP inositol phosphate
  • COS-7 cells are incubated for 24 hours with 5 :Ci of [ 3 H]- myo-inositol (Amersham, PT6-271) in 1 ml medium supplemented with 10% fetal calf serum, 2 mM glutamine and 0.01 mg/ml gentamicin per well.
  • Cells are washed twice in buffer, 20 mM HEPES, pH 7.4, supplemented with 140 mM NaCl, 5 mM
  • KCl 1 mM MgS0 4 , 1 mM CaCI 2 , 10 mM glucose, 0.05 % (w/v) bovine serum; and are incubated in 0.5 ml buffer supplemented with 10 mM LiCI at 37EC for 30 min.
  • the indicated curves are furthermore incubated with adenosine deaminase ADA (200U/mg, Boeringer Mannheim, Germany) for 30 min in a concentration of 1 U/ml .
  • adenosine deaminase ADA 200U/mg, Boeringer Mannheim, Germany
  • polypeptide of the present invention may be produced by a conventional peptide synthesis method.
  • Amino acid derivatives and synthesis reagents can be obtained from commercial sources.
  • Peptide chain extension is performed by mainly using Applied Biosystem 433A synthesizer produced by Perkin Elmer, and a protected peptide derivative- resin is constructed by the Boc or Fmoc method.
  • the protected peptide resin ob- tained by the Boc method is deprotected with anhydrous hydrogen fluoride (HF) in the presence of p-cresol thereby releasing the peptide, which is then purified.
  • the protected peptide resin obtained by the Fmoc method is deprotected with trifluoroacetic acid (TFA) or dilute with TFA containing various scavengers, and the released peptide is purified.
  • Purification is performed in reversed phase HPLC on a C4 or C18 column. The purity of the purified product is confirmed by reverse phase HPLC, and its structure is confirmed by amino acid composition analysis and mass spectrometry.

Landscapes

  • Health & Medical Sciences (AREA)
  • Vascular Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Anesthesiology (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to a delivery device comprising a PYY composition for treatment of obesity, eating disorders and gastrointestinal disorders. The delivery device is particular useful as it allows self administration of the medicament comprised therein, and thereby eases administration and further increases the effectiveness of the treatment as very flexible administration schemes may be employed.

Description

Delivery device for delivering PYY
All patent and non-patent references cited in the application, or in the present application, are also hereby incorporated by reference in their entirety.
Field of invention
The invention relates to a delivery device, such as a single or multi dose delivery device for delivering of PYY or a functional equivalent thereof. The invention relates to a delivery device, such as a single dose or multi dose delivery device, comprising a pharmaceutical composition comprising PYY or a functional equivalent for the treatment of overweight, obesity and/or disorders of appetite regulation. Said delivery device may further be used for the treatment of gastrointestinal disorders such as IBS, FD and/or abdominal pain. It is further included that the delivery device is for the treatment of treatment of eating disorders, such as bulimia nervosa, binge eating disorder and or night eating syndrome.
The invention further relates to a method of treatment comprising administration of PYY or a functional equivalent thereof by use of a delivery device, such as a single or multi dose delivery device.
Background of invention
Obesity
Overweight and obesity is associated with severe health risk as well as loss of the sense of well- being and quality of life for the individual. The cause of overweight and obesity may be disease related, caused by unsuitable eating habits or lack of exercise or a combination of these.
Bodymass index Overweight and obesity may be defined by the body mass index (BMI) of the individual. BMI is calculated, by dividing the bodyweight in kilograms with the square of the height in meters. bodyweight(kg)
Bodymass index: height2 (in) According to guidelines from the US National institute of health three BMI categories have been defined as follows. • Normal weight = 18.5-24.9
• Overweight = 25-29.9
• Obesity = BMI of 30 or greater
Overweight and obesity increase the risk of heart diseases, type 2 diabetes, several cancer types, breathing problems, and an overall risk of premature death, thus the treatment or prevention of overweight and obesity is desirable.
Overweight and obesity may be caused by metabolic disorders as well as disorders of appetite regulation other than eating disorders as described here below.
Eating disorders A great number of people suffer from eating disorders, such as bulimia nervosa, binge eating disorder and/or night eating syndrome and thereby have an increased risk of acquiring additional health problems as well as a lack of quality of life. These types of syndromes have been getting more attention lately and are now being characterized as clinical diseases. A major problem is the diagnosis of these dis- eases, as the syndromes are merely characterised by a behavioural pattern of the patient. The characteristics of the three syndromes are summarised below.
Bulimia Nervosa
Bulimia nervosa is a serious eating disorder marked by a destructive pattern of binge-eating and recurrent inappropriate behaviour to control one's weight. Binge eating is defined as the consumption of excessively large amounts of food within a short period of time. "Inappropriate compensatory behaviour" to control one's weight may include purging behaviours (such as self-induced vomiting, abuse of laxatives, diuretics, or enemas) or non-purging behaviours (such as fasting or excessive exer- cise).
Binge Eating Disorder
Binge Eating Disorder (BED) is a type of eating disorder not otherwise specified and is characterized by recurrent binge eating without the regular use of compensatory measures to counter the binge eating. People with binge eating disorder frequently eat large amounts of food and feel a loss of control over their eating behaviour. This disorder is different from binge-purge syndrome (bulimia nervosa) because people with binge eating disorder usually do not purge afterward by vomiting or using laxa- tives.
Night eating syndrome
Night-eating syndrome (NES) has not yet been formally defined as an eating disorder. A person suffering from NES shows little or no appetite for breakfast and delays first meal for several hours after waking up. Often the patient eats more food after dinner than during the meal. This may mean that more than half of the daily food intake occurs after dinner but before breakfast. This pattern should persist for at least two months before being characterised as a night eating syndrome. The behaviour of an NES patient is different from the behaviour of an individual suffering from a binge eating disorder. During binge eating, relatively large amounts of food are consumed over a relatively short space of time. Night-eating syndrome involves continuous eating throughout evening hours.
Food cravings Cravings may be characterised as an intense desire for certain foods that is almost impossible to withstand, it may even be experienced as an addiction. It remains unclear whether a legitimate physiological basis for this intense desire for certain foods truly exists. Cravings may arise in an attempt to supply the body with nutrients it lacks. Thus deprival of carbohydrates by diets or depletion of glycogen stores by heavy exercisers may result in carbohydrate cravings by which carbohydrates are replenished, although food with a high sugar and fat content is normally preferred.
Gastrointestinal disorders
Gastrointestinal disorders are very common in the population. Some of these are very well characterised and thus suitable treatment regimes have been developed. It is more difficult to develop treatments for functional gastrointestinal disorder with unknown aetiology such as functional dyspepsia (FD) and irritable bowel syndrome (IBS) also called irritable colon.
Irritable Bowel Syndrome The predominant symptoms of IBS are abdominal pain, altered bowel habit, discomfort associated with disturbed defecation and bloating. Patients have an increased mucus and nausea and feeling of constipation and distension. The criteria for irritable bowel syndrome (according to Rome II) are pain or discomfort for 12 weeks of the previous 12 months associated with two of the following; relief with defecation, looser or more frequent stools, harder or less frequent stools (reviewed by Talley, NJ and Spiller R (2002) and Talley, NJ 2003). The symptoms may be chronic and impair the quality of life for the patient.
The patients are grouped in three groups based on there predominat bowel habit (diarrhoea and/or constipation): IBS associated with abdominal pain, fecal urgency and diarrhoea IBS associated with abdominal discomfort, bloating and constipation IBS associated with alternating diarrhoea and constipation
Functional dyspepsia
The symptoms of functional dyspepsia (FD) are partially overlapping with the symptoms of IBS. Patients are grouped in dysmotility-like FD and ulcer-like FD based on the prevalent symptom centred in the upper abdomen (discomfort and pain respectivly). Many patients experience symptoms after meal ingestion, including epigastric discomfort, fullness and pain. Further symptoms include inability to finish a normal-sized meal, bloating, belching, nausea and vomiting (Feinle- Bisset, C. et al. 2003). The underlying mechanism of functional dyspepsia is unclear. The role of delay gastric emptying is debated and currently not the favoured model.
The disorders mentioned are all affected or related to food intake and absorption of nutrients and solutes from the gut.
Regulation of energy uptake
Energy uptake is regulated by hormones that regulate food intake, by modifying the sensing of appetite, and regulate absorption of nutrients and solutes from the gut. Hormones can be separated into long and short term regulating hormones. Long term regulating hormones as insulin and leptin act slowly to promote the stability of body fat stores whereas the short term regulating hormones, as ghrelin and cholecystokinin, act rapidly to influence the individual meal by sensing of hunger, satiety and "fullness". Insulin and leptin are released into the blood in proportion to the amount of body fat. When body fat stores are reduced, declining levels of these hormones are sensed by the brain and are transduced into increases in appetite and metabolic efficiency that persists until the lost weight is recovered. Ghrelin and cholecystokinin are factors that trigger the onset and termination of eating and thereby ghrelin and cholecystokini function in a meal-to-meal control system that itself is sensitive to changes in insulin and leptin levels. In this way, the size and frequency of individual meals can be adjusted so as to minimize changes in body fat content.
The uptake of nutrients from the gut is regulated by various factors including gut hormones. The gut hormones VIP, CCK, and motilin relate to the motility of the upper gastrointestinal tract whereas polypeptide YY (PYY) and Neuropeptide Y (NPY) affect the absorption in the intestine.
PYY
The gut hormone peptide YY (PYY), and the neuropeptide, neuropeptide Y (NPY), are structurally related to pancreatic polypeptide (PP) (figure 1). PYY and NPY exert their action through NPY receptors (Y1 R, Y2R, Y4R and Y5R). The PP, NPY and PYY peptides consist of 36 amino acids with an amidated C-terminal. Two forms of PYY, PYY1-36 and PYY3-36, the latter being a truncated form of the former, have been found in circulation. PYY3-36 is produced by the cleavage of PYY1-36 by the enzyme dipeptidyl peptidase IV (DPP-IV). PYY1-36 binds to and activates at least three NPY receptor subtypes (Y1 , Y2 and Y5) whereas PYY3-36 is more selective for the Y2 receptor (Y2R). Only the C-terminal part of the PYY3-36 peptide is required for the binding to the Y2 receptor. Throughout this document, the notation
PYY covers both PYY1-36 and PYY3-36 (Berglund, M.M. et al., 2003).
PYY was initially isolated from porcine intestine (Tatemoto, K. and Mutt, C, 1980) and named Peptide YY due to the tyrosine residues present in the N- and C- terminal of the molecule. PYY is expressed in endocrine cells lining the gastrointestinal tract and particularly in the distal portion. PYY is secreted in response to food ingestion. Within 15 minutes the plasma level of PYY rise and the level of PYY will reach a plateau after approximately 90 min. The maximum level of PYY reached is proportional to the calories ingested, suggesting that PYY may function as a sensor of food ingestion. In addition PYY is also expressed by neurones, such as in peripheral neurons, particularly enteric neurons. Furthermore, PYY is found in a restricted set of central neurons. The expression pattern of PYY in both endocrine cells and neurons suggest that PYY may be involved in regulation of multiple functions in the individual (Ekblad, E. and Sundler, F., 2002).
A suggested role of PYY may be to regulate the secretion and absorbance of fluid and electrolytes in the gastrointestinal tract and intestine and PYY have therefore been suggested as treatment of diarrhoea (US 6,588, 708) by prolonging of the residence time. Furthermore, PYY3-36 has been suggested to be involved in the system regulating feeding behaviour. It has been found that peripheral administration of PYY3-36 inhibited food intake in rodents. Moreover, direct intra- arcuate administration of PYY3-36 inhibited food intake. A linkage between the PYY effect on feeding behaviour and the NPY 2 receptor have been suggested by the demonstration that NPY receptor Y2 null mice are resistant to the anorectic effects of peripherally administered PYY3-36 (Batterham, R.L. and Bloom, S.R., 2003).
The hypothalamic arcuate nucleus, a key brain area regulating appetite, has access to nutrients and hormones within the peripheral circulation. NPY neurons within the arcuate nucleus express the Y2R. The arcuate nucleus contains two distinct subgroups of neurons that control food intake. On group of neurons produces NPY, which acts in the brain to stimulate feeding (Stanley, B. G. et al, 1986), whereas an adjacent subgroup of neurones produces melanocortin peptides, which act in the same brain areas as NPY, but inhibit eating (Fan, W. et al, 1997). Typically, when one of these subsets is activated, the other one is inhibited.
Weight loss, as modest as 5-15 % of total body weight, reduces the risk factors for many diseases. Thus, independent of the cause of overweight or obesity, treatment that decreases the calorie intake of the individual, may be beneficial. Treatment
Although there are great advantages of treating the disorders mentioned above, such as reducing the weight in overweight patients, treatment of eating disorders and gastrointestinal disorders only few such treatments are available. Thus there is a need for flexible and self administrated treatments.
Injection pen
The injection pens used in the treatment of diabetes have improved the treatment and the quality of life for the individual suffering of diabetes. Currently, injection pens are mostly used for insulin and growth hormone, but treatments with other medicaments using injection pens are emerging. The injection devices exist in a variety of designs with various different functional characteristics. Some of the variants include disposable pens and reusable pens, prefilled syringes and cartridges, single or multiple doses, liquid, lyophilized or powder drugs. Injection pens are produced by companies such as Becton Dickinson, Eli Lilly, Novo Nordisk, Ypsomed, Meridian Medical, Schering, Owen Mumford, Serono, Innoject and Amgen.
References
Batterham, R.L. and Bloom, S.R. (2003). The gut hormone peptide YY regulates appetite. Ann. N. Y. Acad. Sci. Jun;994:162-8.
Berglund, M.M., Hipskind, P.A. and Gehlert, D.R. (2003). Recent developments in our understanding of the physiological role of PP-fold peptide receptor subtypes. Exp Biol Med (Maywood). Mar;228(3):217-44.
Drossman, D.A.; Camilleri, M.; Mayer, E.A.; Whitehead WE (2002). AGA technical review on irritable bowel syndrome. Gastroenterology. Dec; 123(6):2108-31.
Ekblad, E. and Sundler, F. (2002). Distribution of pancreatic polypeptide and peptide YY. Peptides. Feb;23(2):251-61. Fan, W.; Boston, B.A.; Kesterson, R.A.; Hruby, V.J.; Cone, R.D (1997). Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature. Jan 9;385(6612):165-168.
Feinle-Bisset, C; Vozzo, R.; Horowitz, M., and Talley, N.J.(2003). . American
Journal of Gastroenterology, Jan.;99 (1):170-181
Stanley, B. G.; Kyrkouli, S. E.; Lampert, S. and Leibowitz, S. F. (1986). Peptides 7, 1189-1192).
Stanley, B. G.; Kyrkouli, S. E.; Lampert, S. and Leibowitz, S. F. (1986). Peptides 7, 1189-1192).
Talley, N.J. and Spiller R (2002). Irritable bowel syndrome: a little understood organic bowel disease. The Lancet, Vol 360, August 17, 555-564.
Talley, N.J. (2003). Evaluation of drug treatment in irritable bowel syndrome. J. Clin. Pharmacol, 56, 362-369.
Tatemoto, K. and Mutt, V. (1980). Isolation of two novel candidate hormones using a chemical method for finding naturally occurring polypeptides. Nature, Jun 5;285(5764):417-8.
Summary of invention
The present invention relates to a delivery device, such as a multi or single dose delivery device specially adapted for delivering PYY or a functional equivalent thereof, comprising at least one container assembly, dosing means, ejecting means and optionally a needle assembly.
The delivery device according to the invention is adapted for storing PYY or a functional equivalent thereof for a prolonged period of time, as well as for delivering dosages of PYY or the functional equivalent thereof parenterally to an individual in need thereof. In particular the delivery device is for use in the treatment of adipositas or obese or any other disease or disorder treatable with PYY or a functional equivalent thereof. Such other disorder may be an eating disorder, as NES, BED, food cravings or Bulimia Nervosa and further gastrointestinal disorders such as functional dyspepsia and irritable bowel syndrome may according to the invention be treated using PYY or a functional equivalent thereof by use of the delivery device described herein.
The container assembly normally comprises at least one container comprising PYY or a functional equivalent thereof as well as means for engaging a needle assembly to the container for injecting PYY or a functional equivalent thereof. Thus the delivery device is for parenteral administration of PYY.
The container may be any suitable container known in the art, such as a glass con- tainer or a plastic container. The container is preferably formed with a plunger at one end and a sealing at the other end of said container. In a preferred embodiment the inner surface of the container comprising PYY or a functional equivalent thereof is coated with a coating. Said coating is preferably a coating that reduces or avoids adherence of PYY or a functional equivalent thereof to container surface. Also, the coating may serve to reduce degradation of PYY or the functional equivalent thereof, and to secure stabilization of the PYY or the functional equivalent thereof.
The container may be a single use container. In this embodiment the device may be able to release the container to receive a new filled container, when the former con- tainer has been used, in the present context called a changeable container.
In another embodiment the container may be a refillable container.
The delivery device according to the invention may comprise at least two containers. In this embodiment each container may comprise PYY or a functional equivalent thereof. However, the delivery device may also comprise at least two containers wherein at least one container comprises PYY or a functional equivalent thereof and at least one other container comprises a solvent to be mixed with the PYY or a functional equivalent thereof before or during administration of the PYY or the functional equivalent thereof. Accordingly, the delivery device, such as the single or multi dose delivery device may comprise two containers.
In another embodiment, the container of the delivery device according to the inven- tion may comprise at least two compartments, wherein each compartment may comprise PYY or a functional equivalent thereof. However, the container may also comprise at least two compartments wherein at least one compartment comprises PYY or a functional equivalent thereof and at least one other compartment comprises a solvent to be mixed with the PYY or a functional equivalent thereof before or during administration of the PYY or the functional equivalent thereof.
Thus, in an embodiment of the invention the contents of the two containers or two compartments can be mixed prior to use.
In a further embodiment of the invention said mixing occurs via a liquid channel.
Said liquid channel may be formed by a needle or by opening of a breakable sealing by means of for example a plunger or a needle.
An embodiment of the invention relates to a single or multi dose delivery device comprising at least one container assembly wherein, said container assembly comprise a distal end comprising coupling means for realisably mounting a needle assembly and said container assembly comprise a proximal end comprising coupling means for engaging the dosing assembly.
The dosing assembly comprises means for adjusting ejection of one dosage per activation. In a preferred embodiment the dosing assembly comprises means for adjusting ejection of individual dosages in accordance with an individual dosage scheme.
A further embodiment of the invention include a delivery device, such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or - any combination hereof.
In a further embodiment of the invention include a delivery device, such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or and combination thereof.
In an even further embodiment of the invention include a delivery device, such as a single dose or multi dose delivery device, comprising PYY or a functional equivalent thereof, wherein the delivery device is a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia
The PYY or a functional equivalent thereof comprised in the delivery device may be in the form of a pharmaceutical composition optionally as a pharmaceutically acceptable salt or pharmaceutically acceptable acid addition salt, and optionally with pharmaceutically acceptable carriers and/or diluents.
The pH of the pharmaceutical composition is preferably between 3 and 5.5 and preferably buffered using an acetate buffer.
As described above the pharmaceutical composition comprising PYY or a functional equivalent thereof may be mixed with a solvent prior to use or during use. The mixing preferably takes place within the single or multi dose delivery device.
In one embodiment of the invention the pharmaceutical composition is a dry composition, such as a freeze dried or spray dried composition, to be mixed with a solvent before or during use. Another aspect of the invention relates to a method of treatment comprising the use of a delivery device, such as a single dose or multi dose delivery device according to the invention.
The single or multi dose delivery device according to the invention is suitable for self administration, i.e. administration wherein the individual in need of the treatment administrates the pharmaceutical composition himself or herself.
The dosage to be administered per administration is determined by a physician prescribing the composition. As described above, it is preferred that the delivery device allows individual adjustment of the size of dosage. Normally such as 0.25-20 nmol of PYY or a functional equivalent is administrated per day, in one or more dosages. Preferably PYY or the functional equivalent thereof is administered in at least 1 dos- age per day corresponding to the main meal of the day.
In one embodiment of the invention the medicament is administrated with in 120 minutes before a meal, such as within 60 minutes before a meal, such as within 30 minutes before a meal, such as within 15 minutes before a meal such as at the be- ginning of a meal.
Alternatively, the PYY or functional equivalent thereof can be administered less frequently than once per day. For instance, PYY may be administered every second day, every third day, every fourth day, every fifth day, every sixth day or once a week depending on the needs of the individual patient. The dosage for such administration may be as described herein for daily dosages, optionally times the number of days between administration. For instance, if administered every second day the dosage can be twice that determined for an every day administration.
In a further embodiment of the invention the medicament is administrated dependent on the sensing of appetite in the individual.
In an embodiment the medicament is administered in combination with a second active ingredient. Description of the figure
Structure of the family of the PP-fold peptides NPY, PYY, and PP share a common hairpin-like three-dimensional structure called the PP-fold. All three peptides are 36 amino acids long with an amidated carboxy- terminus. The general structure of the PP-fold peptides has been established using x-ray crystallography of avian PP and confirmed in several studies using nuclear magnetic resonance. Amino acid residues 1-8 form a type II proline helix followed by a loop. Residues 15-32 form an α-helix, and the four most carboxy-terminal residues are in a flexible loop conformation. The amino acid sequence of mammalian NPY is highly conserved, PPY display 8 varible amino acids, whereas the amino acid sequence of PP is the least conserved PP-fold peptide. The general three- dimensional structure seems to be conserved in all PP-fold peptides (Berglund MM et al, 2003).
Sequence listing
SEQ ID NO: 1 Human PYY
Definitions:
Affinity: The term refers to the binding strength between receptors and their ligands, for example PYY and its receptor. Agonist: A molecule that stimulates the action of the target.
Amino acid: Entity comprising an amino terminal part (NH2) and a carboxy terminal part (COOH) separated by a central part comprising a carbon atom, or a chain of carbon atoms, comprising at least one side chain or functional group. NH2 refers to the amino group present at the amino terminal end of an amino acid or peptide, and COOH refers to the carboxy group present at the carboxy terminal end of an amino acid or peptide. The generic term amino acid comprises both natural and non- natural amino acids. Natural amino acids of standard nomenclature as listed in J. Biol. Chem., 243:3552-59 (1969) and adopted in 37 C.F.R., section 1.822(b)(2) belong to the group of amino acids listed in Table 1 herein below. Non-natural amino acids are those not listed in Table 1. Examples of non-natural amino acids are those listed e.g. in 37 C.F.R. section 1.822(b)(4), all of which are incorporated herein by reference. Further examples of non-natural amino acids are listed herein below. Amino acid residues described herein can be in the "D" or or "L" isomeric form.
Symbols Amino acid 1 -Letter 3-Letter
Y Tyr tyrosine G Gly glycine F Phe phenylalanine M Met methionine A Ala alanine S Ser serine I lie isoleucine L Leu leucine T Thr threonine V Val valine P Pro proline K Lys lysine H His histidine Q Gin glutamine E Glu glutamic acid W Trp tryptophan R Arg arginine D Asp aspartic acid N Asn asparagine C Cys cysteine
Table 1. Natural amino acids and their respective codes.
Appetite: Appetite in an individual is assessed by measuring the amount of food ingested and by assessing the individual's desire to eat. Appetite (i.e., hunger) is typically assessed with a short questionnaire given to individuals on a random basis several times a week. Typically, 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. Amino acid residue: the term "amino acid residue" is meant to encompass amino acids, either standard amino acids, non-standard amino acids or pseudo-amino ac- ids, which have been reacted with at least one other species, such as 2, for example 3, such as more than 3 other species. In particular amino acid residues may comprise an acyl bond in place of a free carboxyl group and/or an amine-bond and/or amide bond in place of a free amine group. Furthermore, reacted amino acids residues may comprise an ester or thioester bond in place of an amide bond Antagonist: A molecule that inhibits that action of its target.
Antibody: Are immunoglobulin molecules and active portions of immunoglobulin- molecules. Antibodies are for example intact immunoglobulin molecules or fragments there of retaining the immunologic activity. BMI: Body mass index measures an individual's height/weight ratio. It is determined by calculating weight in kilograms divided by the square of height in meters. The
BMI "normal" range is 18,5-24,9.
Body fat mass: 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. Dissociation constant, Kd: a measure to describe the strength of binding (or affin- ity or avidity) between receptors and their ligands, for example an antibody and its antigen. The smaller the Kd, the stronger binding.
Concentration equivalent: A concentration equivalent is an equivalent dosage being defined as the dosage of compound having in vitro or/and in vivo the same response (as evaluated e.g. from a dosage-response curve) as a known compound. 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. Half-life: See T1/2. Individual: A living animal or human. In preferred embodiments, 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 any of the PYY peptides or functional analogues thereof 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, hor- mones, and other proteinaceous or non-proteinaceous solutes. In preferred embodiments, the polypeptide will be purified. "Loss of body weight": defined herein as a reduction in BMI. "Loss of body fat": defined herein as either a reduction of an individual's overall fat mass or a reduction in the percentage of an individual's body fat. Medical disorder: by "medical disorder" is meant any disease or syndrome having a detrimental effect on an individual's physical and/or mental health. Said medical disorder may have a genetic cause. Preferably, said medical disorder leads to one or more undesirable symptoms including increase of fat mass, increase of weight, increase of appetite, and/or causes a reduction in the individual's PYY levels. Peptide: Plurality of covalently linked amino acid residues defining a sequence and linked by amide bonds. The term is used analogously with oligopeptide and polypeptide. The amino acids may be both natural amino acids and non-natural amino acids, including any combination thereof. The natural and/or non-natural amino acids may be linked by peptide bonds or by non-peptide bonds. The term peptide also embraces post-translational modifications introduced by chemical or enzyme-catalyzed reactions, as are known in the art. Such post-translational modifications can be introduced prior to partitioning, if desired. Amino acids as specified herein will preferentially be in the L-stereoisomeric form. Amino acid analogs can be employed instead of the 20 naturally-occurring amino acids. Several such analogs are known, including fluorophenylalanine, norleucine, azetidine-2- carboxylic acid, S-aminoethyl cysteine, 4-methyl tryptophan and the like. Furthermore, it should be noted that 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 NH2 or acetyl or to a carboxy-terminal group such as COOH. PYY: Peptide YY, throughout this document PYY is used to describe the PYY1-36 and PYY3-36 molecules.
PYY Composition: This term is intended to indicate a pharmaceutical composition comprising PYY or a functional equivalent thereof. Receptor: A receptor is a molecule, such as a protein, glycoprotein and the like, that can specifically (non-randomly) bind to another molecule Recombinant DNA (rDNA) molecule: A DNA molecule produced by operatively linking two DNA segments. Thus, a recombinant DNA molecule is a hybrid DNA molecule comprising at least two nucleotide sequences not normally found together in nature.
T1 2: The half-life is the time for the concentration of a compound to decrease 50 %.
Detailed description of the invention
Overweight and obesity
An individual with a body mass index above 25 is considered overweight or obese. The cause of overweight and obesity may have many different reasons and may be due to a combination of several factors such as disorders, lack of exercise, excess calorie intake, a diet of food with inadequate nutritional composition, and/ or social habits.
Metabolic disorders
Metabolic disorders may affect the ability of the individual to control the bodyweight, as signalling from the energy storage compartments may be lacking or incorrect.
Disorder in appetite regulation
Some disorders of appetite regulation are caused by a direct malfunction of the appetite regulating system.
Eating disorders Compulsive (compensatory) eating as NES, BED food cravings and bulimia nervosa may be at least partly be characterised as psychological disorders. Theses disorders may be associated with overweight or underweight. Alternatively the patient may be of normal weight although the nutritional status is affected in an undesirable way. Gastrointestinal disorders
IBS or functional dyspepsia are functional gastrointestinal disorders that may also affect the nutritional status and physical well being of the patients.
Calorie intake/exercise/diet
Weight gain is caused by the intake of excess calories, thus the control of body weight need balance of calorie intake and catabolism. The level of catabolism is controlled by the body activities, thus exercise increases catabolism and prevents weight gain when food consumption is constant. The composition of the diet may also affect the assimilation as different categories of food are absorbed differently.
Individual in need
According to the invention, any suitable individual who may draw benefit from usage of the single or multi dose delivery device of the invention may be considered an individual in need.
Preferably, said individual is suffering from overweight or obesity. In one embodiment, the individual to be treated has a BMI in the range of 25-30. In another embodiment, the individual to be treated has a BMI in the range of 30-35. In a further embodiment, the individual to be treated has a BMI of 27 or above.
Individuals suffering from an eating disorder as described herein, or a disorder of the gastro intestinal system, such as IBS or functional dyspepsia may benefit from usage of the delivery device.
In addition a person in need of losing weight for athletic performance or for cosmetic reasons may also be considered as an individual in need of treatment.
Furthermore, the delivery device of the invention may be used for inhibition of appe- tite and decreasing the rate of weight gain in animals.
PYY
The human pancreatic polypeptide PYY1-36 is identified by SEQ ID NO:1. PYY3-36 is a truncated form of PYY1-36 where the two most N-terminal residues are deleted. PYY1-36 is a 36 AA polypeptide with a C- as well as an N-terminal tyrosine amino acid residue. The polypeptides are produced by cleavage of a pre-polypeptide and PYY1-36 furthermore cleaved by dipeptidyl peptidase IV yielding PYY3-36, as described above. Throughout this document, peptide YY (PYY) is used as a general term covering PYY1-36 and PYY3-36.
Functional equivalents
Functional equivalents of PYY1-36 and PYY3-36 include PYY molecules originating from different species, such as mouse, rat, monkey, swine, bovine or other mammalian species. Furthermore, functional equivalents comprise homologues of PYY molecules.
Homologues
A homologue shall be construed as a molecule which shares some identity to the molecule, here PYY1-36 and PYY3-36. The homology may be expressed as 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.
A 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 perform- ing the same function, i.e. a homologue may be envisaged as a "functional equivalent" of a predetermined sequence.
As described above a functional equivalent of any of the predetermined sequences herein may be defined as: i) homologues comprising an amino acid sequence capable of being recognised by an antibody, said antibody also recognising PYY1-36 or PYY3-36, and/or
ii) homologues comprising an amino acid sequence capable of binding selectively to an NPY receptor, and/or
iii) homologues having a substantially similar or higher binding affinity to NPY receptors than PYY1-36 or PYY3-36
In the above examples, the 36 aa PYY1-36 has the sequence shown in SEQ ID NO: 1. PYY3-36 is 34 amino acids long and has the sequence shown in SEQ ID NO: 1 except for the deletion of the two N-terminal amino acids.
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 (Gly) of said homologue is substituted with an amino acid selected from the group of amino acids consisting of Ala, Val, 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 Gly, Val, Leu, and He, and independently thereof, homologues, wherein at least one valine (Val) of said homologue thereof is substituted with an amino acid selected from the group of amino acids consisting of Gly, 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 Gly, Ala, Val, and He, and independently thereof, homologues thereof, wherein at least one isoleucine (He) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Gly, Ala, Val and Leu, and independently thereof, homologues thereof wherein at least one of said aspartic acids (Asp) of said homo- logue thereof is substituted with an amino acid selected from the group of amino acids consisting of Glu, Asn, and Gin, and independently thereof, homologues thereof, wherein at least one of said phenylalanines (Phe) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Tyr, Trp, His, Pro, and preferably selected from the group of amino acids consisting of Tyr and Trp, and independently thereof, homologues thereof, wherein at least one of said tyrosines (Tyr) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Phe, Trp, His, Pro, preferably an amino acid selected from the group of amino acids consisting of Phe and Trp, and independently thereof, homologues thereof, wherein at least one of said arginines (Arg) of said fragment is substituted with an amino acid selected from the group of amino acids consisting of Lys and His, and independently thereof, homologues thereof, wherein at least one lysine (Lys) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Arg and His, and independently thereof, homologues thereof, wherein at least one of said aspargines (Asn) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Asp, Glu, and Gin, and independently thereof, homologues thereof, wherein at least one glutamine (Gin) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Asp, Glu, and Asn, and independently thereof, homologues thereof, wherein at least one praline (Pro) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Phe, Tyr, Trp, and His, and independently thereof, homologues thereof, wherein at least one of said cysteines (Cys) of said homologues thereof is substituted with an amino acid selected from the group of amino acids consisting of Asp, Glu, Lys, Arg, His, Asn, Gin, Ser, Thr, and Tyr.
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 polar- ity, for example a residue with a non-polar side chain (Ala, Leu, Pro, Trp, Val, lie, Leu, Phe or Met) substituted for a residue with a polar side chain such as Gly, Ser, Thr, Cys, Tyr, Asn, or Gin or a charged amino acid such as Asp, Glu, 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 Gly by another residue; and/or iii) differ substantially in electric charge, for example substitution of a negatively charged residue such as Glu or Asp for a positively charged residue such as Lys, His or Arg (and vice versa); and/or iv) differ substantially in steric bulk, for example substitution of a bulky residue such as His, Trp, Phe or Tyr for one having a minor side chain, e.g. Ala, Gly or Ser (and vice versa).
Substitution of amino acids may in one embodiment be made based upon their hy- drophobicity 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.
In a preferred embodiment the functional equivalent comprises a homologue having an amino acid sequence at least 60 % homologous to SEQ ID NO 1.
More preferably 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.
In a more preferred embodiment the percentages mentioned above relates to the identity of the sequence of a homologue as compared to SEQ ID NO 1.
In a preferred embodiment the functional equivalent comprises the amino acids corresponding to the 6 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO.1 (Tyr Pro lie Lys Pro Glu). The functional equivalent may comprise 8 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro), or such as 10 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu) or such as 12 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala), or such as 14 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro) ), or such as 16 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu), or such as 18 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn), or such as 20 N-terminal amino acids of PYY1- 36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr), or such as 22 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro
Glu Glu Leu Asn Arg Tyr Tyr Ala), or such as 24 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu), or such as 26 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His), or such as 28 N- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu), or such as 30 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu), or such as 32 N-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr), or such as 34 N-terminal amino acids of PYY1 -36 as defined in SEQ ID NO 1 (Tyr Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin).
In a further preferred embodiment the functional equivalent comprise the amino acids corresponding to the 6 N- terminal amino acids of PYY3-36 as defined in SEQ ID NO.1 (lie Lys Pro Glu Ala Pro), or such as 8 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu) or such as 10 N- terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala), or such as 12 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro) ), or such as 14 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu), or such as 16 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn), or such as 18 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Ar9 Tyr), or such as 20 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr
Ala), or such as 22 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu), or such as 24 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (Tyr Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His), or such as 26 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu), or such as 28 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu), or such as 30 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (lie Lys
Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr), or such as 32 N-terminal amino acids of PYY3-36 as defined in SEQ ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin).
In a preferred embodiment the functional equivalent comprise the amino acids corresponding to the 6 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Val Thr Arg Gin Arg Tyr), or such as the 8 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 10 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 12 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 14 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as the 16 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ser Leu Arg His Tyr Leu Asn
Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 18 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 20 C- terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 22 C-terminal amino acids of PYY1 -36 as defined in SEQ ID NO 1 (Glu Glu Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 24 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ser Pro Glu Glu Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 26 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Asp Ala Ser Pro Glu Glu Leu
Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 28 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 30 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg
Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 32 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr Tyr Ala), or such as the 34 C-terminal amino acids of PYY1-36 as defined in SEQ ID NO 1 (Tyr Ala He Lys Pro Glu Ala Pro Gly Glu Asp
Ala Ser Pro Glu Glu Leu Asn Arg Tyr Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr).
In another preferred embodiment the functional equivalent may comprises internal amino acids of PYY1-36 such as amino acid 16-21 of PYY 1-36 as defined in sequence ID NO 1 (Glu Leu Asn Arg Tyr Tyr ), or such as amino acid 15-22 of PYY 1-36 as defined in sequence ID NO 1 (Glu Glu Leu Asn Arg Tyr Tyr Ala), or such as amino acid 14-23 of PYY 1-36 as defined in sequence ID NO 1 (Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser), or such as amino acid 13-24 of PYY 1-36 as defined in sequence ID NO 1 (Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu), or such as amino acid 12-25 of PYY 1-36 as defined in sequence ID NO 1 (Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg), or such as amino acid 11-26 of PYY 1-36 as defined in sequence ID NO 1 (Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His), or such as amino acid 10-27 of PYY 1-36 as defined in sequence ID NO 1 (Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr), or such as amino acid 9-28 of PYY 1-36 as defined in sequence ID NO 1 (Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu), or such as amino acid 8-29 of PYY 1-36 as defined in sequence ID NO 1 (Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn), or such as amino acid 7-30 of PYY 1-36 as defined in sequence ID NO 1 (Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu), or such as amino acid 6-31 of PYY 1-36 as defined in sequence ID NO 1 (Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val), or such as amino acid 5-32 of PYY 1-36 as defined in sequence ID NO 1 (Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser
Leu Arg His Tyr Leu Asn Leu Val Thr), or such as amino acid 4-33 of PYY 1-36 as defined in sequence ID NO 1 (Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg), or such as amino acid 3-34 of PYY 1-36 as defined in sequence ID NO 1 (He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr
Leu Asn Leu Val Thr Arg Gin), or such as amino acid 2-35 of PYY 1-36 as defined in sequence ID NO 1 (Pro lie Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg), or such as amino acid 2-36 of PYY 1-36 as defined in sequence ID NO 1 (Pro He Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu
Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr), or such as amino acid 4-36 of PYY 1-36 as defined in sequence ID NO 1 (Lys Pro Glu Ala Pro Gly Glu Asp Ala Ser Pro Glu Glu Leu Asn Arg Tyr Tyr Ala Ser Leu Arg His Tyr Leu Asn Leu Val Thr Arg Gin Arg Tyr).
The functional equivalent may be PYY 2-36 or alternatively, the functional equivalent may be PYY4-36.
In an embodiment the functional equivalent comprise any of the above sequences with conservative amino acid substitutions, such as one substitution, or such as two substitutions, or such as two substitutions, or such as more than two substitutions, or such as more than four substitutions.
Further included are functional equivalent know from the literature, such as the PYY agonists described in WO 03/057235 and references therein.
Covalent modifications
The functional equivalent may comprise any type of modifications. Nearly 200 structurally distinct covalent modifications have been identified thus far, ranging in size and complexity from conversion of amides to carboxylic acids, to the attachment of multiple complex oligosaccharides. Such modifications include phosphorylation, acetylation, ubiquination, lipidation (acetylation, prenylation, farnesylation, geranyla- tion, palmitoylation, myristoylation), methylation, carboxylation, sulfunation and O- or N-glycosylations.
A subset of modifications is dependent on vitamin C as a cofactor. This include proline and lysine hydroxylations and carboxy terminal amidation.
In an embodiment PYY or the functional equivalent comprise a C-terminal amida- tion. In a preferred embodiment the C-terminal tyrosine residue of PYY or a functional equivalent is amidated.
Protecting group
The functional equivalent may comprise 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 al- kenyl, aryl, -C1-6 alkyl aryl, -C(O)- (CH2) 1-6-COOH, -C(0)-C1-6 alkyl, -C(0)-aryl,
-C (0)-0-C1-6 alkyl, or-C (O)-O-aryl. Preferably, 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 ethyla- mide.
Conjugates
PYY or the functional equivalent of PYY may conjugated to another entity, in order for example, to prolong its half-life. Conjugation can improve the delivery of targeted doses, prevent breakdown, and increase bioavailablity in circulation.The other entity may be any molecule capable of providing the desired effect. The preparation of conjugates is well known in the art se for example Hermanson GT. Bioconjugate Techniques. New York: Academic Press; 1996, Aslam M, Dent AH. Bioconjugation: protein coupling techniques for the biomedical sciences. Houndsmills, England: Macmillan Publishers; 1999, and Wong SS. Chemistry of protein conjugation and cross/inking. Boca Raton, FL: CRC Press; 1991.
Most methods use amine-reactive reagents or thiol-reactive reagent. In the preparation of conjugates advantages may be achieved through the use of certain linkers. For example, linkers that contain a disulfide bond that is sterically "hindered" are often preferred, due to their greater stability in vivo, thus preventing release of the active moiety prior to binding at the site of action. It is generally desired to have a conjugate that will remain intact under conditions found everywhere in the body except the intended site of action, at which point it is desirable that the conjugate have good "release" characteristics.
Different conjugates have been described, for example, use of the A chain of ricin is described in US 4,340,535 incorporated herein by reference. Examples of peptide conjugates based on Ac-RYY(RK)(WI)RK)-NH2 (where the brackets show allowable variation of amino acid residues) may be found in US patent application 2003040472.
The molecules may be conjugated as described above, or by peptide bonds, before or after synthesis and purification. For instance PYY or a functional equivalent thereof may be fused to the Fc portion of an antibody or to albumin in order to ex- tend the half-life of the conjugated molecule. The fusion may be obtained by any suitable methods, for example, but not exclusively, by recombinant DNA technology. In a preferred embodiment the fusion of is made by recombinant DNA technology, such as a fusion of the nucleotide sequence encoding PYY and the nucleotide sequence encoding Fc portion or albumin is made and the fusion is thereby encoded by a single nucleotide sequence. The fusion polypeptide may be expressed and purified as a single polypeptide molecule, using any suitable method, as described for the purification of PYY.. The fusion polypeptide may include insertion of a linker, such as a peptide of at least 2 AA, such as at least 5 AA, such as at least 8 AA, such as at least 15 AA, such as at least 20 AA. Suitable linkers are known in the art. Methods for production and purification of PYY
PYY or functional equivalents thereof can be produced using techniques well known in the art. Preferably, the PYY or functional equivalent for use herein is of GMP grade. For example, a polypeptide region of a PYY can be chemically or biochemi- cally synthesized and modified. Techniques for chemical synthesis, such as solid- phase synthesisof polypeptides (see example 2) are well known in the art. (See e.g. Vincent in Peptide and Protein Drug Delivery, New York, N. Y., Dekker, 1990.) Examples of techniques for biochemical synthesis involving the introduction of a nucleic acid into a cell and expression of nucleic acids are provided in Ausubel, Cur- rent Protocols in Molecular Biology, John Wiley, 1987-1998, and Sambrook et al., in
Molecular Cloning, A Laboratory Manual, 2 d Edition, Cold Spring Harbor Laboratory Press, 1989.
PYY or the functional equivalent may be produced by chemical synthesis, e.g. as described described in brief below, or by recombinant methods.
E.g. PYY or the functional equivalent may be produced by the following process:
(a) constructing, by conventional techniques, an expression vector containing an operon with a DNA sequence encoding human PYY or a functional equivalent thereof;
(b) transfecting the expression vectors into a host cell by conventional techniques; and
(c) culturing the transfected cell by conventional techniques to produce the PYY or functional equivalent.
The host cell may be cotransfected with a second vector for optimization of the production process. The two vectors may contain different selectable markers. The cod- ing sequences of PYY may comprise cDNA or genomic DNA or both.
The host cell used to express the PYY may be either a bacterial cell such as Es- cherichia coli, or a eukaryotic cell, such as S. cerevisiea or P. pastoris. In particular a mammalian cell line may be used, such as Hela, CHO or any other suitable host cell known by the person skilled in the art. The general methods by which the vectors may be constructed, transfection methods required to produce the host cell and culture methods required to produce PYY or functional equivalent from such host cells are all conventional techniques. Like- wise, once produced, the PYY or functional equivalent may be purified according to standard procedures as described below.
The method of purification used is dependent upon several factors, including the purity required, the method of production and the intended use. Such methods are known in the art.
Any suitable conventional methods of purifying polypeptides including precipitation and column chromatography are well known to one of skill in the purification arts, including cross-flow filtration, HPLC, ammonium sulphate precipitation, affinity col- umn chromatography, gel electrophoresis and the like may be used.
PYY composition
The PYY composition to be used in the delivery device of the invention comprises PYY or a functional equivalent thereof as the active ingredient. The PYY composi- tion is preferably free of any contaminants present in blood such as infectious agents. The PYY composition is formulated so as to be suitable for administration via a delivery device of the invention.
In a preferred embodiment the PYY composition have a concentration of at least 10 nM such as at least 50 nM, such as at least 0.2 μM, such as at least 0.5 μM, such as at least 1 μM, such as at least 2 μM, such as at least 5 μM, such as at least 10 μM, such as at least 20 μM, such as at least 50 μM, such as at least 0.2 mM, such as at least 0.5 mM, such as at least 1 mM, such as at least 2 mM, such as at least 5 mM of PYY or a functional equivalent thereof.
The PYY composition may be stored as a dry composition, for example lyophilized (freeze dried) or spray dried to improve the stability of PYY. Such compositions are reconstituted with liquid solutions prior to use. Generally, the protein concentration of the reconstituted formulation is about 2-40 times greater than the protein concen- tration in the mixture before lyophilization, thus this allows the production of PYY composition of high concentration. When reconstituted with a diluent comprising a preservative (such as bacteriostatic water for injection, BWFI), the reconstituted formulation may be used as a single-dose or multi-dose formulation. Such a formulation is useful, for example, where the patient requires frequent subcutaneous administrations. In another embodiment the PYY composition may be a liquid composition of high stability. The PYY composition may be meant for mixing with a suitable diluent prior to use.
The PYY composition to be used in the delivery device of the invention may be mixed with a suitable diluent to form a pharmaceutical composition prior to use. The mixing may be performed by the patient prior to insertion of the container in the delivery device, such as the single or multi dose delivery device, or by insertion of a two compartment container in a single or multi dose deliver device especially adapted to allow mixing of two components. Alternatively the single or multi dose delivery device may accommodate two containers and allow mixing of the content of the two prior to use.
Pharmaceutical compositions comprising PYY or a functional equivalent thereof to be used in the delivery device of the present invention may be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy 1995, edited by E. W. Martin, Mack Publishing Company, 19th edition, Easton, Pa. The compositions may appear in conventional forms, for example solutions or suspensions.
As used herein, the terms "pharmaceutically acceptable", "physiologically tolerable" and grammatical variations thereof, as they refer to compositions, carriers, diluents and reagents, are used interchangeably and represent that the materials are capable of administration to or upon an individual without the production of undesirable physiological effects such as nausea, dizziness, gastric upset and the like.
The pharmaceutical composition preferably further comprises pharmaceutically acceptable salts, a pharmaceutically acceptable carrier or a diluent. The pharmaceutical composition may further comprise transport molecules. The compositions may preferably be delivered to an individual in any way so as to achieve a beneficial effect, preferably by inhibiting appetite and/or preventing malnutrition, and/or improving the individual's sense of well-being or quality of life.
The delivery device of the invention is useful for parenteral administration of a PYY composition as described herein, such as via a subcutaneous, intradermal or intramuscular route via a delivery device according to the invention. Preferably, the composition is administered via the subcutaneous route.
The PYY or functional equivalent thereof may be administered with at least one other compound as discussed further below. The compounds may be administered simultaneously, either as separate compositions or combined in a unit dosage form, or administered sequentially.
The pharmaceutical composition may be produced prior to use by mixing of a PYY composition with an appropriate diluent.
Pharmaceutically acceptable salts
Pharmaceutically acceptable salts of PYY or a functional equivalent, where they can be prepared, are also intended to be covered by this invention. These salts will be ones which are acceptable in their application to a pharmaceutical use. By that it is meant that the salt will retain the biological activity of PYY or the functional equivalent and the salt will not have untoward or deleterious effects in its application and use in treating diseases.
Pharmaceutically acceptable salts are prepared in a standard manner. If PYY or the functional equivalent is a base it is treated with an excess of an organic or inorganic acid in a suitable solvent. If PYY or the functional equivalent is an acid, it is treated with an inorganic or organic base in a suitable solvent.
Such salts include pharmaceutically acceptable acid addition salts, pharmaceutically acceptable metal salts, ammonium salts and alkylated ammonium salts. Acid addition salts include salts of inorganic acids as well as organic acids. Examples of pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, p-toluenesulphonic acids, and aryl- sulphonic acid. formic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, maleic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methanesulfonic, ethanesulfonic, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, as- partic, stearic, palmitic, ethylenediaminetetraacetic (EDTA), p-aminobenzoic, glu- tamic, benzenesulfonic and the like. Further examples of pharmaceutically acceptable inorganic or organic acid addition salts include the pharmaceutical acceptable salts listed in J. Pharm. Sci. 1977,66,2, which is incorporated herein by reference.
Examples of metal salts include lithium, sodium, potassium and magnesium salts and the like.
According to the invention organic acid salts of organic acids such as for example acetic acid is preferred.
Examples of ammonium and alkylated ammonium salts include ammonium, methyl- ammonium, dimethylammonium, trimethylammonium, ethylammonium, hy- droxyethylammonium, diethylammonium, butylammonium and tetramethylammo- nium salts and the like.
Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like.
Also included within the scope of PYY or a functional equivalent thereof or pharmaceutical acceptable acid addition salts thereof in the context of the present invention are any hydrates (hydrated forms) thereof.
The preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art. Typically such 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. Pharmaceutically acceptable carriers and diluents
The active ingredient or a salt thereof can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof. In addition, if desired, 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.
Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, organic esters such as ethyl oleate, and water-oil emulsions.
Suitable pharmaceutical carriers include inert solid diluents or fillers, sterile aqueous solution and various organic solvents. Examples of solid carriers are lactose, terra alba, sucrose, cyclodextrin, talc, gelatine, agar, pectin, acacia, magnesium stearate, stearic acid or lower alkyl ethers of cellulose. Examples of liquid carriers are syrup, peanut oil, olive oil, phospholipids, fatty acids, fatty acid amines, polyoxyethylene or water.
Stabilizers
The active compound of the invention may be unstable, thus the composition preferably contain stabilizers, preservatives or conservatives to increase the stability of the compounds.
Tween 60, Span 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate, mannitol and sodium lauryl sulphate are possible stabilizers. In a preferred embodiment mannitol may be used as stabilizer.
For the preparation of a lyophilised composition ad lyoprotectant may be used to stabilize the active ingredient (Townsend and DeLuca, "Use of lyoprotectants in the freeze-drying of a model protein, ribonuclease A" Journal of Parenteral Science & Technology 42 (6): 190-199 (Nov.-Dec. 1988)). The lyoprotectant may preferably be a sugar such as sucrose or trehalose such as sucrose, dextran, or hydroxypropyl-/142-cyclodextrin.
Furthermore, a pH-buffering agent may be used to stabilize PYY or functional equivalent in the composition. The buffering agent may be acetate, carbonate, bi- carbonate, phosphate, citrate, tris or hepes. In a preferred embodiment the buffering agent is acetate.
According to the invention the composition preferably has a pH between 2.0 and 9.0, or such as between 2.5 and 8.0, or such as 3.0 and 7.0, or such as between 3.5 and 6.0, or such as between 3.5 and 5.0 or such as between 4.0 and 5.5, or such as between 4.0 and 5.0, or such as between 4.0 and 4.5. Preferably the pH of the compositions is less than 6, preferably less than 5.5, preferably less than 5, preferably less than 4.8, preferably less than 4.6, preferably less than 4.4, preferably less than 4.2.
Transport molecules
Transport molecules act by having incorporated into or anchored to it PYY or functional equivalent. Any suitable transport molecules known to the skilled person may be used. Examples of transport molecules may be liposomes, micelles, and/or mi- crospheres.
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.
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". The shape of micelles formed in dilute surfactant solutions is approximately spherical. The polar head groups of the surfactant molecules are arranged in an outer spherical shell whereas their hydrocarbon chains are oriented toward the center, forming a spherical core for the micelle. The hydrocarbon chains are randomly coiled and entangled and the micellar interior has a nonpolar, liquid-like character. In the micelles of polyoxyethylated nonionic detergents, the polyoxyethylene moieties are oriented outward and permeated by water. This arrangement is energetically favourable since the hydrophilic head groups are in contact with water and the hydrocarbon moieties are removed from the aqueous medium and partly shielded from contact with water by the polar head groups. The hydrocarbon tails of the surfactant molecules, located in the interior of the micelle, interact with one another by weak van der Waals forces.
The size of a micelle or its aggregation number is governed largely by geometric factors. The radius of the hydrocarbon core cannot exceed the length of the extended hydrocarbon chain of the surfactant molecule. Therefore, increasing the chain length or ascending homologous series increases the aggregation number of spherical micelles. If the surfactant concentration is increased beyond a few percent and if electrolytes are added (in the case of ionic surfactants) or the temperature is raised (in the case of nonionic surfactants), the micelles increase in size. Under these conditions, the micelles are too large to remain spherical and become ellipsoidal, cylindrical or finally lamellar in shape.
Common surfactants well known to one of skill in the art can be used in the micelles. Suitable surfactants include sodium laureate, sodium oleate, sodium lauryl sulfate, octaoxyethylene glycol monododecyl ether, octoxynol 9 and PLURONIC F-127 (Wy- andotte 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. In addition, phos- pholipids, such as those described for use in the production of liposomes, may also be used for micelle formation.
The PYY composition may further include the transport molecules as described above. The pharmaceutical compositions described herein are then readily administered in a variety of dosage forms suitable for the disclosed routes of administration via a delivery device of the invention. The compositions may conveniently be presented in unit dosage form or in multiple dosage form by methods known in the art of phar- macy.
Compositions for. parenteral administration
The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example solutions in aqueous polyethylene glycol. Examples of oily or nonaqueous carriers, diluents, solvents or vehicles include propyl- ene glycol, polyethylene glycol, vegetable oils (e.g., olive oil), and injectable organic esters (e.g., ethyl oleate), and may contain formulatory agents such as preserving, wetting, emulsifying or suspending, stabilizing and/or dispersing agents. Alternatively, 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 suit- able vehicle, e.g., sterile, pyrogen-free water. Aqueous solutions should be suitably buffered if necessary, and the liquid diluents first rendered isotonic with sufficient saline or glucose. The aqueous solutions are particularly suitable for intramuscular, subcutaneous and intradermal administration. The sterile aqueous media employed are all readily available by standard techniques known to those skilled in the art.
In a preferred embodiment of the invention the composition comprising PYY or a functional equivalent thereof or a salt thereof, is a lyophilised composition and the composition may further comprises a solvent. In another embodiment the composition is a solution of PYY or a functional equivalent thereof or a salt thereof. Prefera- bly, the solvent may be any suitable solvents, such as described herein, and preferably the solvent is saline or a physiological buffer like phosphate buffer.
The pharmaceutical composition comprises PYY or a functional equivalent thereof or a pharmaceutically acceptable salt thereof, and for example protease inhibitors. Such compositions can be prepared in water or saline, and optionally mixed with a nontoxic surfactant. Compositions for administration with the delivery device of the invention may include sterile aqueous solutions that may also contain buffers, liposomes, diluents and other suitable additives. Oils useful in parenteral compositions include animal, vegetable, or synthetic oils. Specific examples of 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. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
Suitable soaps for use in parenteral compositions include fatty alkali metal, ammonium, and triethanolamine salts, and 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 alkanola- mides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-.beta.-aminopropionates, and 2-alkyl-imidazoline quater- nary ammonium salts, and (e) mixtures thereof.
The parenteral compositions typically will contain from about 0.00002 % to about 2 % 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 compo- sitions 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. The 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. Extemporaneous injection solutions and sus- pensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
The pharmaceutical dosage forms suitable for injection can include sterile aqueous solutions or dispersions comprising the active ingredient that are adapted for ad- ministration 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 PYY or a functional equiva- lent thereof or pharmaceutically acceptable salt thereof in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filter sterilization.
Administration The pharmaceutical composition may be prepared so it is suitable for one or more particular administration methods via the delivery device of the invention.
The composition comprising PYY or a functional equivalent thereof can be administered parenterally by injection using a delivery device of the invention, such as, in- tramuscularly, subcutaneously or intradermally.
In one aspect of the present invention, a suitable dose of the compositions described herein is administered in pharmaceutically effective amounts to an individual in need of such treatment. Herein, "pharmaceutically effective amounts", is defined as a total amount of the pharmaceutical composition or the total amount of each active component of the pharmaceutical composition that is sufficient to show a meaningful patient benefit. The pharmaceutical compositions containing PYY or a functional equivalent thereof are conventionally administered as a subcutaneous injection of a unit dose. The terms "unit dose" and "unit dosage form" as used herein refer to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of PYY or functional equivalent thereof, alone or in combination with other agents, calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier, or vehicle. The specifications for the unit dosage forms depend on the particular compound or compounds employed and the effect to be achieved, as well as the pharmacodynamics associated with each compound in the individual to be treated. The dose administered should be an "effective amount" or an amount necessary to achieve an "effective level" in the individual patient. Alternatively, the pharmaceutical composition may be prepared in multiple dose form and by use of the multi dose delivery device, single doses are administrated when needed. Furthermore, since the "effective level" is used as the preferred endpoint for dosing, the actual dose and schedule can vary, depending on individual differences in pharmacokinetics, drug distribution, and metabolism. The "effective level" can be defined, for example, as the blood or tissue level desired in the patient that corresponds to a concentration of one or more compounds to be used in accordance with the invention. The effective level may be defined as the dose required to achieve a plasma concentration of at least, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140 or 150 pmol/l or alternatively a plasma concentration of 20-50, 30-100, 40-80, 50 -70 or approximately 60 pmol/l.
It will also be recognized by one of skill in the art that the optimal quantity and spacing of individual dosages of a compound or a pharmaceutically acceptable salt thereof will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular patient being treated, and that such optimums can be determined by conventional techniques. It will also be appreciated by one of skill in the art that the optimal course of treatment, i.e., the number of doses of a compound or a pharmaceutically acceptable salt thereof given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests.
It is further contemplated that treatment using the delivery system of the invention is capable of reducing symptoms of the condition being treated or a melioration of the quality of life of the patient. This may be a decrease in food intake, a decrease in pain sensation or a normalisation of bowel habits, or a normalised eating pattern. Such effects are readily recognised by the skilled artisan.
For the present invention the dosage will vary depending on the compound employed and the mode of administration. Dosage levels may vary between about 4 ng/kg body weight to 20 μg/kg body weight daily, preferably between about 10 ng/kg body weight to 1 μg/kg body weight, more preferably between 50 to 750 ng/kg body weight.
The dosage may be administered when needed, such as up to ten times daily, such as one to five times daily, such as two or three times daily, or preferably such as once a day, thus the daily dosage maybe up to 2-5 times the dosages mentioned above. Alternatively, the dosage may be administered less frequently than once daily as described herein above.
A preferred dosage of a composition employed according to the invention is in a concentration equivalent to PYY or a functional equivalent there of from 4 ng to about 20 μg per kg bodyweight, or such as from 10 ng to 1 μg per kg bodyweight, more preferably from 50 to 750 ng per kg bodyweight. The dosage of PYY is preferably 20-200 ng/kg, 20-160 ng/kg, 40-160 ng/kg, 40-120 ng/kg, 40-80 ng/kg, 60- 120 ng/kg or such as approximately 60 ng/kg or 80 ng/kg.
The preferred dosages may be in a concentration equivalent to PYY or a functional equivalent there of from 1 pmol/kg to 5 nmol/ kg, such as from 5 pmol/kg to 1 nmol/kg, or such as from 20 pmol/kg to 500 pmol/kg, such as from 40 to160 pmol- /kg, such as from 75 to 120 pmol/kg. In a preferred embodiment the dosage may be 5-50 pmol/kg, 5-40 pmol/kg, 5 to 30 pmol/kg, 10-40 pmol/kg, 10-30 pmol/kg such as 5 to 25 pmol/kg, such as 5 to 20 pmol/kg, and most preferably 10-20 pmol/kg, 15-30 pmol/kg or approximately 15 pmol/kg or 20 pmol/kg. The dosages are preferably administrated once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day, or such as more than five times a day.
In one preferred embodiment of the present invention, the compositions are administered in dosages of PYY or a functional equivalent from about 400 ng to about 2 mg, more preferably from about 10 μg to about 200 μg, or from about 5 μg to about 250 μg, more preferably from about 20 μg to about 200 μg, more preferably from about 20 μg to about 100 μg. Most preferably the dosage may be 1-20 μg, 2-16 μg, 4-16 μg, 4-12 μg, 4-8 μg, 6-12 μg, 6-10 μg or approximately 8 μg. In a preferred embodiment the composition is administered in dosages of PYY or a functional equivalent from 100 pmol to 500 nmole, or such from 500 pmol to 100 nmol, or such as from 1 nmol to 50 nmol, or such as from 2 to 25 nmol, or such as from 4 to 20 nmol. Alternatively the preferred dosage may be 0,25-5 nmol, 0,5-4 nmol, 1-4 nmol, 1-3 nmol, 1-2 nmol, 1 ,5-3 nmol or more preferably 1 ,5-2,5 nmol or most preferably approximately 2 nmol. In another embodiment, the PYY or functional equivalent is administered subcuta- neously in a dosage of 5-30 pmol/kg, such as 5-25 pmol/kg, such as 5-20 pmol/kg or such as 10-20 pmol/kg bodyweight, in order to achieve an effective level in the indi- vidual treated. The presently preferred dosage is 10-20 pmol/kg bodyweight of
PYY1-36 or PYY 3-36.
The dosages of PYY or the functional equivalent is preferably administered once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day.
The pharmaceutical preparations described herein may also be arranged in unit dosage forms. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as powders in compartments. In this embodiment the powders may be mixed with a solvent prior to or during use.
In a preferred embodiment the PYY composition is administrated in unit dosage form, from about 400 ng to about 2 mg of PYY or a functional equivalent thereof, more preferably from about 10 μg to about 200 μg, or from about 5 μg to about 250 μg, more preferably from about 20 μg to about 200 μg, more preferably from about 20 μg to about 100 μg. The unit dosage form may comprise from 100 pmol to 500 nmole, or such from 500 pmol to 100 nmol, or such as from 1 nmol to 50 nmol, or such as from 2 to 25, such as from 4 nmol to 20 nmol. of PYY or a functional equivalent thereof. The compositions are preferably administered once a day, or such as two times a day, or such as three times a day, or such as four times a day, or such as five times a day.
The dosage requirements will vary with the particular drug composition employed, the route of administration and the particular subject being treated. Ideally, a patient to be treated by the present method will receive a pharmaceutically effective amount of the compound in the maximum tolerated dose, generally no higher than that required before drug resistance develops Suitable dosing regimens 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.
It should be noted that the normal PYY response which occurs during the cause of a meal is a short-lived surge in plasma concentrations of PYY and that due to the relative short half life of the peptide an injection, e.g. subcutaneous injection, of PYY will ensure that a similar short-lived peak on PYY concentrations can be obtained. This may depend on the thickness of the adipose tissue, as the blood flow decrease pro- portionally to the thickness of the fat layer, and therefore the clearance from the tissue is assumed to follow a similar curve.
Contrary, administration may using a formulation with a prolonged half life may give a stable concentration of PYY, thus decreasing the need for regular meal associated injections.
The administration route must ensure that the non-degraded, bioactive form of the peptide will be the dominating form in the circulation, which will reach the PYY receptors and stimulate these. Thus, in order to obtain the maximum effect of the pharmaceutical composition it is preferably administered from one to three times daily, each administration being within 120 minutes of a meal, such as within 110 minutes of a meal, such as within 100 minutes of a meal, such as within 90 minutes of a meal, such as within 85 minutes of a meal, such as within 80 minutes of a meal, such as within 75 minutes of a meal, such as within 70 minutes of a meal, such as within 65 minutes of a meal, such as within 60 minutes of a meal, such as within 55 minutes of a meal, such as within 50 minutes of a meal, such as within 45 minutes of a meal, such as within 40 minutes of a meal, such as within 35 minutes of a meal, such as within 30 minutes of a meal, such as within 25 minutes of a meal, such as within 20 minutes of a meal, such as within 15 minutes of a meal, such as within 10 minutes of a meal, such as within 5 minutes of a meal. More preferred the pharma- ceutical composition is administered prior to each main meal, such as administered three times daily. Most preferably the pharmaceutical composition is administered once during the day prior to a meal.
In one embodiment of the invention the PYY composition is administrated dependent on the sensing of appetite in the individual.
In special situation, for example for an individual suffering from PYY deficiency the dosing regime may be different. Administration may aim at obtaining a low basal level of PYY and additionally obtaining PYY peeks associated with the meals. This may include administration of different PYY composition with different timing.
Combination therapy
PYY or a functional equivalent may be administered with at least one other com- pound. The compounds may be administered simultaneously, either as separate compositions or combined in a unit dosage form, or administered sequentially.
Administration of the PYY composition may be used in combination with any suitable therapy. By treatment "in combination" is meant that another treatment regime may be carried out before, during (including concurrently with) and after treatment of an individual with a PYY composition as described herein.
In a preferred embodiment the PYY composition of the invention is administered in combination with a second pharmaceutical composition. Said second pharmaceuti- cal composition may be administered together or separately from the PYY composition.
In a particular embodiment said second pharmaceutical composition may comprise compound(s) selected from the group of; anti-hypertensives including (but not lim- ited to) beta-adrenoceptor antagonists, alpha-adrenoceptor antagonists, calcium antagonists, angiotensin converting enzyme (ACE) inhibitors, angiotensin II receptor antagonists and diuretics (including, but not limited to, thiazide diuretics and loop diuretics) , insulin, DPP-IV inhibitors, anti-depressant medications including (but not limited to) selective serotonin reuptake inhibitors (SSRIs), mianserine and mirtazap- ine, classical as well as atypical antipsychotic drugs (neuroleptics), corticosteroids as well as any other drug(s) that may decrease body weight or body fat mass or that may be indicated for the treatment of hypertension, overweight, obesity, syndrome X and/or diabetes mellitus.
Delivery devices
Many forms of delivery device may be used according to the invention. Dependent on the treatment to be administrated the recipient and other factors the most suitable delivery device may be selected.
A single dose delivery device comprises a therapeutically effective amount of PYY or a functional equivalent thereof, suitable for injection by a single injection. A multi dose delivery device comprise at least two dosages of a therapeutically effective amount of PYY or a functional equivalent thereof, suitable for injection by at least two injection.
An aspect of the invention relates to a delivery device for delivering PYY or a functional equivalent thereof, comprising at least one container or compartment comprising a therapeutically effective amount of PYY or a functional equivalent thereof, dosing means, ejecting means and optionally a needle assembly.
Disposable injection device
A disposable injection device may be preferred due to convenience. There is no handling of containers and needles involved in the use and further there is no need for appropriate storage of the injection device in between injections. Disposable de- livery devices are generally smaller and therefore more discrete than conventionally reusable delivery devices.
According to the invention a disposable delivery device may be used for delivery of PYY or a functional equivalent thereof. The disposable delivery device for delivering of PYY or a functional equivalent thereof comprises according to the invention a pharmaceutical composition, a needle and ejection means and optionally dosing means.
Multi dose delivery device The advantage of a multi-dose delivery device is that it facilitates ease of use for the patient, reduces waste by allowing complete use of container contents, and results in a significant cost savings for the manufacturer since several doses are packaged in a single container (lower filling and shipping costs).
According to the invention a multi dose delivery device may be used for delivery of PYY or a functional equivalent thereof. According to the invention, a multi dose delivery device for delivering of PYY or a functional equivalent thereof, comprise at least one container assembly, dosing means, ejecting means and optionally a needle assembly.
Containers and compartments
According to the invention the delivery device comprise containers and/or compartments for storage of the therapeutically effective amount of PYY or functional equivalent thereof.
The delivery device may further comprise assembly means of the container or compartment.
In an embodiment the delivery device comprises at least one container assembly capable of accepting a container. In a preferred embodiment the container has one compartment filled with a pharmaceutical composition. In a second preferred embodiment the container in the delivery device has two compartments. Either of said two compartments may comprise a pharmaceutical composition according to the invention and the second may comprise either a solvent or solute meant for mixing with the pharmaceutical composition or a second pharmaceutical composition. In a preferred embodiment the two compartments are constructed to allow mixing of the contents of the two compartments prior to use. The compartments may be connected via a liquid channel that can be open manually/mechanically or electronically prior to use to allow mixing of the contents of the two compartments or alternatively the two compartments are connected by a breakable sealing. Said sealing that can be broken mechanically or electronically prior to use to allow mixing of the contents of the two compartments prior to injection. The sealing may for example be broken by means of a plunger or a needle which may be controlled manually/mechanically or by electronic means. The sealing may be a movable plunger or piston.
In a further specific embodiment the delivery device comprises two compartments. The two compartments are located in continuation of each other and separated by an intervening first plunger. The first compartment, closest to the needle contains PYY or a functional equivalent, preferably in lyophilized form. Behind the first plunger, furthest away from the needle, is a second compartment, comprising the relevant solvent. The second compartment is closed with a second plunger. A long the axis of the first compartment a channel formed as an axial indentation in the wall of the compartment is present. When the device is used the second plunger is pressed into the device and through the second compartment. As the solvent cannot be compressed, the pressure on the second plunger will result in pressure on the first plunger and moving of the first plunger past the opening of the channel allowing the liquid of the second compartment to access to the channel. As the channel is longer than the first plunger the solvent can pass the first plunger and flow from the second compartment into the first compartment, in which the PYY or functional equivalent thereof is dissolved to obtain a solution. When the second plunger is pressed further in the pressure on the first plunger is increased whereby the solution now present in the first compartment is injected through the needle located in the front part of the first compartment.
In another embodiment the delivery device comprises two containers. Either of said two containers may comprise a pharmaceutical composition according to the invention and the second may comprise either a solvent or solute meant for mixing with the pharmaceutical composition or a second pharmaceutical composition. In a preferred embodiment the content of said two containers can be mixed prior to use. The containers may be connected by a liquid channel; said liquid channel may be formed by a needle. Said liquid channel may be opened manually, mechanically or by electronic means prior to use. Thereby, the content of the two containers can be mixed before injection into the patient.
According to the invention the delivery device may be reusable. In a further embodiment at least one container in the delivery device can be replaced. In a further embodiment at least one container in the delivery device may be refillable. In further embodiments the two containers may be replaced or refilled separately.
An embodiment of the invention relates to a delivery device comprising at least one container assembly, wherein said container assembly comprise a distal end comprising coupling means for realisably mounting a needle assembly, and said container assembly comprise a proximal end comprising coupling means for engaging the dosing assembly.
The pen may further comprise an automated injection system, for example controlled by a spring.
Dosing The multi dose delivery device, according to the invention, may be designed with a dosing assembly to allow the dosing of the pharmaceutical composition to be changed by the patient. The dosing assembly may be adjustable for different doses.
In case the injection device comprise more than one pharmaceutical composition the dosages of the compositions may controlled separately.
The dosing assembly may for example be controlled by means of a plunger or by electronic means.
Needle The length of the needle may be important for correct delivery of the PYY composition. According to the invention the preferred delivery device may be able to accept needles of different length. The needles used may be at least 0.5 cm in length, such as more than 1.0 cm, such as more than 1.5 cm, such as more than 2.0 cm or such as more than 2.5 cm in length, such as more that 3 cm in length, such as more than 4 cm in length, such as more than 5 cm in length, such as more than 6 cm in length, such as more than 7 cm in length, such as more than 8 cm in length, such as more than 9 cm in length, such as more than 10 cm in length.
In further embodiments of the invention the needle is 0.5-1.0 cm long, 1.0-1.5 cm long, or such as 1.5-2.0 cm long, or such as 2.0-2.5 cm long, or such as 2.5-3,0 cm long, or such as 3.0-4.0 cm long, or such as 4.0-6.0 cm long, or such as 6.0-8.0 cm long, or such as 8.0-10.0 cm long.
The delivery device maybe designed to accept needles of different diameter. The diameter of the needle relate to the outer diameter of the needle.
In a preferred embodiment the diameter of the needle is at the most 0.50 mm, such as at the most 0.40 mm, such as at the most 0.35 mm, such as at the most 0.33 mm, such as at the most 0.30 mm, such as at the most 0.20 mm, such as at the most 0.15 mm. In a preferred embodiment the diameter of the needle is 0.30-0.35 mm, or such as 0.30-0.33 mm, or such as 0.33mm -0.35 mm, or such as 0.35-0.40 mm.
Furthermore, in a preferred embodiment the needle may be changed by the patient.
In a preferred embodiment the needle may be hidden for the user. This may be beneficial for people suffering from fear of injection.
A needle based delivery device of the present invention can be developed on the basis of devices manufactured by Innoject, Ypsomed, Becton Dickenson and Owen
Mumford.
Needle-free injection
In further embodiments the delivery system may be needle free. The pharmaceutical composition may be delivered by pressure whereby a micro-thin stream of the pharmaceutical composition penetrates the skin and is deposited into the subcutaneous (fatty) tissue. Examples of needle-free systems are those manufactured by Cross Ject, BioJect, Aradigm, Antares Pharma, Alza, Injext, and Biovalve.
Depth of injection
In an embodiment the injection device includes means for controlling the depth of injection, thereby it is possible to reach different preferred injection sites using the same type of needles. In an embodiment the depth of injection may vary from 1.0 cm to 10 cm. The injection depth may be 0.5-1.0 cm, 1.0-1.5 cm, or such as 1.5-2.0 cm, or such as 2.0-2.5 cm, or such as 2.5-3,0 cm, or such as 3.0-4.0 cm, or such as 4.0-6.0 cm, or such as 6.0-8.0 cm, or such as 8.0-10.0 cm.
Injection
Both using needle containing and needle free injection devices the pen may include the possibility to control the injection pressure and/or the injection rate.
Control unit
In further embodiments the pen may comprise an electronic control unit, such control unit may allow the user to store information, such as preselected dosages, timing and mode of injection. The control unit may include an alarm function and thereby alert the patient about the need for injection by making a sound, light signal, or physical movement (vibration). The alarm signal may be transmitted to a unit carried by the patient. The control unit may indicate positions for injections, dosage, and further indicate if the pen is positioned correct for injection. Injection may be blocked by the electronic control unit to avoid incorrect usage.
In some cases it is difficult for the patient to correctly position the pen and to know the correct mode of injection, such as for example the depth of injection. In a further Embodiment the injection pressure may be controlled.
In an embodiment the control unit is capable of registering the force (or measure the resistance) used to insert the needle and there upon control the depth/tissue of injection. Using this function the pharmaceutical composition is always injected correctly also in difficult positions and the possible injection sites is thereby increased.
Container
The material used for production of the container may be any suitable material. The product should be suitable for storage of the pharmaceutical composition this include being non-sticky for the components of the pharmaceutical composition and stable at various temperatures used for storage of pharmaceutical compositions. According to the invention at least one container may be at least partly transparent and may further comprise a scale. In a preferred embodiment at least one container is made of plastic or glass.
The container may be made of both metallic and non-metallic materials. The metallic surfaces can be comprised of, for example, titanium, stainless steel and other suitable materials. The non-metallic container surfaces can be comprised, for example, of a glass material of, a polymeric material, such as a polyurethane, a polyethylene, a polypropylene, a polyvinylchloride or for example a rubber such as bromobutyl rubber or chlorobutyl rubber, or other similar polymeric materials.
The container can be made of a polymeric material, such as those listed above, or can be formed from a polymer laminate (e.g., two or more layers of different polymeric materials).
Coating
In order to stabilize the polypeptides of the pharmaceutical composition the inner surface of at least one container may comprise a coating.
The coating may comprise polymeric materials such as hydrophilic polyurethanes, polyureas, acrylics, polycarbonates, polyvinyl pyrollidone or other hydrophilic materials, in particular materials such as polyethylene glycols, polyethylene/polypropylene glycol copolymers or other poloxamers which may be chemically (covalently) attached to the surface. The coating may further comprise polymers of the various paraxylylenes.
In an embodiment the preferred coating may be a silicon-containing coating, such as a poly(dialkyl siloxane) oil or copolymer. In a preferred embodiment the poly(dialkyl siloxane) is selected from poly(dimethyl siloxane) (PDMS), poly(dipropyl siloxane) or poly(dihexyl siloxane). In one embodiment of the invention the coating comprises a linear or branched hydrophilized poly(dialkyl siloxane) oil.
In a preferred embodiment of the invention the coating comprises a crosslinked or gelled silicon oil, such as a hydrophilized poly(dialkyl siloxane) oil, or a mixture of a crosslinked and a non-crosslinked oil. A cross-linked, or cured, silicon oil is typically obtained by applying a linear, or branched, silicon oil with reactive functionalities which are used to cross-link the coating in a subsequent step. In another preferred embodiment of the invention the coating comprises a hydrophilized poly(dialkyl siloxane) block and graft copolymer. The copolymer may be any block and graft copolymer which comprises polymeric segments of poly(dialkyl siloxane), such as
PDMS. The polymeric segments may, for example, be combined with polymeric segments of polystyrene, polyolefins, polyamides, or polyurethane to form the desired copolymer. The copolymer may be prepared by any method available, for example by sequential anionic polymerization, or different grafting procedures. Hydrophilicity of the coating according may be obtained by any appropriate method.
In a preferred embodiment of the invention the coating is subjected to an oxidative treatment, such as plasma treatment or corona treatment after having been applied to the component.
In another preferred embodiment the coating comprises a copolymer which is made hydrophilic by end-capping the copolymer with hydrophilic group or chain segments. The hydrophilic group may, for example, be a negatively charged chemical group or phosphorylcholine (PC) groups, and the chain segment may, for example, be poly(ethylene oxide) (PEO) or poly(2-hydroxyethyl methacrylate) (pHEMA).
The surfaces may be further modified in order to further decrease the protein adsorption by coupling of hydrophilic polymer segments or functional groups. These polymer segments or functional groups may be of the same kind as those described above, and may further be coupled to the functional groups generated during the plasma treatment.
Depending on the migration ability of the coating the hydrophilic groups at the coating will tend to seek into the coating leaving the surface hydrophobic due to the hydrophobicity of the surrounding air. Accordingly, it is of great importance that the coating remains hydrophilic during storage until the medical article is filled with the protein solution. This may be secured by placing the coated article in a hydrophilic environment, such as by filling the medical article with the protein formulation shortly after the coating process. The thickness of the coating depends on the specific coating, and is preferably from 0.005 to 10 μm, such as from 0.01 to 1 μm or such as 0.05 to 0.4 μm, or such as from 0.0 5 to 0.25 μm, more preferably approximately 0.2 μm. The optimal thickness depends on the dimensions and shape of the container and it can easily be performed by one skilled in the art. If the coating is too thin the coating may be torn in use, thereby increasing the friction between the two components. When the thickness of the coating has reached a certain plateau value the friction forces are approximately constant even when the thickness is further increased.
Treatment
In an embodiment of the invention the delivery device, such as a single dose or multi dose delivery device comprises at least one container or compartment comprising a pharmaceutical composition.
In an embodiment of the invention the pharmaceutical composition present in the delivery device is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or any combinations hereof.
In a further embodiment of the invention the pharmaceutical composition present in the delivery device is; a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or any combinations hereof.
In an even further embodiment of the invention the pharmaceutical composition present in the delivery device is; a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia
In an embodiment of the invention said pharmaceutical composition comprises PYY or a functional equivalent thereof.
According to the invention a single or multi dose delivery device may be used for the administration of a PYY composition as described above.
In an embodiment the invention the delivery device, such as the single or multidose delivery device comprises in addition to the PYY composition also a second pharmaceutical composition. The single or multidose delivery device may comprise separate dosing and administration means for the PYY composition and the second pharmaceutical composition.
As described above, in one aspect of the invention, the compositions of the present invention are self-administered.
Examples
The following examples illustrate the invention without limiting it thereto. Example 1
Binding assay and functional assay
Transfections and tissue culture: COS-7 cells can be grown Dulbecco's Modified Eagle'e Medium 1885 supplemented with 10% fetal calf serum, 2mM glutamine and
0.01 mg/ml gentamicin. The expression plasmids containing the cDNAs encoding the wild type or the mutated receptors can be transiently expressed after transfetion according to the calcium phosphate precipitation method and assay can be performed 48 hour after transfection. Binding assay: One day after transfection the cells will be transferred and seeded in multi-well plates for assay. The number of cells to be plated per well will be chosen so as to obtain 5 to 10% binding of the radioligand added. Two days after transfection the cells will be assayed in competition binding assays using 125l- PYY(3-36) as a tracer. Radioligand will be bound in a buffer composed of 0.5 ml of 50 mM Hepes buffer, pH 7.4, supplemented with 1 mM CaCI2, 5 mM MgCI2, and 0.1% BSA, and displaced in a dose dependent manner by unlabelled ligands. The assay will be performed in duplicate for 3 hours at 4 °C, and stopped by washing twice in the buffer. Cell associated, receptor bound radioligand will be determined by the addition of lysis buffer (48% urea, 2% NP-40 in 3M acetic acid). The concentration of radioli- gand in the assay corresponds to a final concentration of approximately 20 pM.
Functional assay.
COS-7 can be cultured as described above and contransfections can be performed. The activation of Phospholipase C by chimeric G-proteins (Conklin B) formed between both Gαq and Gαi the Y2 receptor can be measured through the inositol phosphate (IP) turnover in the cell. The IP turnover may be recorded by use of following assay:
One day after transfection COS-7 cells are incubated for 24 hours with 5 :Ci of [3H]- myo-inositol (Amersham, PT6-271) in 1 ml medium supplemented with 10% fetal calf serum, 2 mM glutamine and 0.01 mg/ml gentamicin per well. Cells are washed twice in buffer, 20 mM HEPES, pH 7.4, supplemented with 140 mM NaCl, 5 mM
KCl, 1 mM MgS04, 1 mM CaCI2, 10 mM glucose, 0.05 % (w/v) bovine serum; and are incubated in 0.5 ml buffer supplemented with 10 mM LiCI at 37EC for 30 min. The indicated curves are furthermore incubated with adenosine deaminase ADA (200U/mg, Boeringer Mannheim, Germany) for 30 min in a concentration of 1 U/ml . After stimulation with various concentrations of peptide for 45 min at 37 C, cells will be extracted with 10 % ice-cold perchloric acid followed by incubation on ice for 30 min. The resulting supernatants are neutralized with KOH in HEPES buffer, and the generated [3H]-inositol phosphate is purified on Bio-Rad AG 1-X8 anion-exchange resin. Determinations will be made in duplicates.
Example 2
Synthetic production of PYY and functional equivalents thereof The polypeptide of the present invention may be produced by a conventional peptide synthesis method.
Amino acid derivatives and synthesis reagents, can be obtained from commercial sources. Peptide chain extension is performed by mainly using Applied Biosystem 433A synthesizer produced by Perkin Elmer, and a protected peptide derivative- resin is constructed by the Boc or Fmoc method. The protected peptide resin ob- tained by the Boc method is deprotected with anhydrous hydrogen fluoride (HF) in the presence of p-cresol thereby releasing the peptide, which is then purified. The protected peptide resin obtained by the Fmoc method is deprotected with trifluoroacetic acid (TFA) or dilute with TFA containing various scavengers, and the released peptide is purified. Purification is performed in reversed phase HPLC on a C4 or C18 column. The purity of the purified product is confirmed by reverse phase HPLC, and its structure is confirmed by amino acid composition analysis and mass spectrometry.

Claims

Claims
1. A delivery device for delivering PYY or a functional equivalent thereof, comprising at least one container comprising a therapeutically effective amount of PYY or a functional equivalent thereof, dosing means, ejecting means and optionally a needle assembly.
2. The delivery device according to claim 1 comprising at least two dosages of PYY or a functional equivalent thereof.
3. The delivery device according to claim 1 or 2, wherein the delivery device is for parenteral administration.
4. The delivery device according to claim 1 , 2 or 3, comprising one container.
5. The delivery device according to claim 4, wherein the container has one compartment.
6. The delivery device according to claim 4, wherein the container has two com- partments.
7. The delivery device according to claim 6, wherein the contents of the two compartments can be mixed prior to use. 8. The delivery device according to claim 6-7, wherein the two compartments are connected via a liquid channel.
9. The delivery device according to claim 6-8, wherein the two compartments are separated by a breakable sealing.
10. The delivery device according to claim 9, wherein the breakable sealing is a plunger.
11. The delivery device according to claim 10, wherein the sealing is broken by plunger means.
12. The delivery device according to claim 10, wherein the sealing is broken by a needle.
13. The delivery device according to claim 1 , 2 or 3, comprising two containers.
14. The delivery device according to claim 13, where in the contents of the two containers can be mixed prior to use.
15. The delivery device according to claim 14, wherein the containers are connected by a liquid channel.
16. The delivery device according to claim 15, wherein said liquid channel is formed by a needle.
17. The delivery device according to claim 1 , 2 or 3, wherein the delivery device comprises a needle assembly.
18. The delivery device according to claim 1 , 2 or 3, wherein said at least one container is refillable.
19. The delivery device according to claim 1 , 2 or 3, wherein said at least one container is having a distal end comprising coupling means for realisably mounting a needle assembly, and having a proximal end comprising coupling means for engaging the dosing assembly.
20. The delivery device according to claim 1 , 2 or 3, wherein said dosing means comprise plunger means.
21. The delivery device according to any of the previous claims wherein the dosing assembly is adjustable for different dosages.
22. The delivery device according to any of the previous claims wherein a needle is located in the needle assembly.
23. The delivery device according to claim 22 wherein the needle is at least 2 cm long.
24. The delivery device according to any of the previous claims wherein the di- ameter of the needle is at the most 0.50 mm.
25. The delivery device according to any of the previous claims, wherein said at least one container is moulded of a plastic material.
26. The delivery device according to any of the previous claims, wherein the inner surface of said at least one container comprises a coating.
27. The delivery device according to any of the previous claims, wherein said at least one container is at least partly transparent.
28. The delivery device according to any of the previous claims, wherein said at least one container further comprise a scale.
29. The delivery device according to any of the previous claims, wherein at least one container comprise a PYY composition.
30. The delivery device according to claim 29 wherein the PYY composition is a. for the treatment of overweight or b. for the treatment of obesity or c. for the treatment of disorders of appetite regulation or d. for the treatment of metabolic disorders or e. for the regulation of appetite or f. for the inhibition of appetite or or any combination hereof.
31. The delivery device according to claim 29 wherein the PYY composition is a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia
32. The delivery device according to claim 29 wherein the PYY composition is a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or any combinations hereof.
33. The delivery device according to claim 29-32, wherein the pharmaceutical composition comprises PYY1-36
34. The delivery device according to claim 29-32, wherein the pharmaceutical composition comprises PYY3-36.
35. The delivery device according to claim 29-34, wherein the pharmaceutical composition comprise at least 1 nmole of PYY or a functional equivalent thereof.
36. The delivery device according to claim 29-35, wherein the pharmaceutical composition further comprises pharmaceutical acceptable salts.
37. The delivery device according to claim 29-36, wherein the pharmaceutical composition further comprises pharmaceutical acceptable carriers and dilu- ents.
38. The delivery device according to claim 29-37, wherein the pH of the pharmaceutical composition is between 3.0 and 5.5
39. The delivery device according to claim 29-38, wherein the pharmaceutical composition is present in one compartment and a solvent is present in the second compartment.
40. The delivery device according to claim 29-39, wherein the pharmaceutical composition is present in one compartment and second pharmaceutical composition is present in the second compartment.
41. The delivery device according to claim 29-40, wherein the pharmaceutical composition is a semi-dry composition, such as a paste.
42. The delivery device according to claim 29-41 , wherein the pharmaceutical composition is a dry composition
43. The delivery device according to claim 29-42, wherein the pharmaceutical composition is a freeze dried.
44. The delivery device according to claim 29-43, wherein the pharmaceutical composition is spray dried.
45. A method of treatment of an individual in need of PYY treatment comprising administering PYY or a functional equivalent thereof by use of a delivery device according to any of the previous claims.
46. The method according to claim 45, wherein the individual in need is treated for a. overweight b. obesity c. disorders of appetite regulation d. metabolic disorders e. regulation of appetite f. inhibition of appetite or any combination hereof.
47. The method according to claim 45, wherein the individual in need is treated for a. for the treatment of IBS and/or b. for the treatment of functional dyspepsia,
48. The method according to claim 44, wherein the individual in need is treated for a. for the treatment of BED or b. for the treatment of NES or c. for the treatment of Bulima nervosa or d. for food cravings or any combinations hereof.
49. The method of treatment according to any of the claims claim 45-48 compris- ing an administration scheme.
50. The method of treatment according to any of the claims 45 - 49 where in the treatment is self administrated.
51. The method of treatment according to any of the claims 45- 50, wherein such as 0.25-20 nmol of PYY or a functional equivalent thereof is administrated per day.
52. The method of treatment according to any of the claims 45-51 , wherein the pharmaceutical composition is administrated as a single dose or in multiple dosages.
53. The method of treatment according to any of the claims 45-52, wherein the administration is performed within 60 minutes before a meal.
54. The method of treatment according to any of the claims 45-53, wherein the administration is performed within 30 minutes before a meal.
55. The method of treatment according to any of the claims 45-54, wherein the administration is performed within 15 minutes before a meal.
56. The method of treatment according to any of the claims 45-55, wherein the administration is performed at the beginning of a meal.
7. The method of treatment according to any of the claims 45-56, wherein the administration is dependent on the sensing of appetite in the individual.
PCT/DK2005/000111 2004-02-19 2005-02-18 Delivery device for delivering pyy WO2005079888A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
PCT/DK2005/000327 WO2005110467A1 (en) 2004-05-18 2005-05-18 A composition comprising pyy for the treatment of gastrointestinal disorders
JP2007516968A JP2007538017A (en) 2004-05-18 2005-05-18 PPY-containing composition for the treatment of gastrointestinal diseases
US11/596,973 US20080125360A1 (en) 2004-05-18 2005-05-18 Composition Comprising Pyy for the Treatment of Gastrointestinal Disorders
PCT/DK2005/000329 WO2005110454A2 (en) 2004-05-18 2005-05-18 Protective binding members of pyy for the treatment of metabolic disorders
PCT/DK2005/000328 WO2005110457A2 (en) 2004-05-18 2005-05-18 Treatment of eating disorders and induction of lipolysis
EP05739487A EP1750753A1 (en) 2004-05-18 2005-05-18 A composition comprising pyy for the treatment of gastrointestinal disorders
AU2005244475A AU2005244475A1 (en) 2004-05-18 2005-05-18 A composition comprising pyy for the treatment of gastrointestinal disorders

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
DKPA200400255 2004-02-19
DKPA200400255 2004-02-19
DKPA200400793 2004-05-18
DKPA200400793 2004-05-18
DKPA200401338 2004-09-03
DKPA200401338 2004-09-03

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/596,973 Continuation-In-Part US20080125360A1 (en) 2004-05-18 2005-05-18 Composition Comprising Pyy for the Treatment of Gastrointestinal Disorders

Publications (2)

Publication Number Publication Date
WO2005079888A2 true WO2005079888A2 (en) 2005-09-01
WO2005079888A3 WO2005079888A3 (en) 2005-09-22

Family

ID=34890745

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2005/000111 WO2005079888A2 (en) 2004-02-19 2005-02-18 Delivery device for delivering pyy

Country Status (1)

Country Link
WO (1) WO2005079888A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009029795A1 (en) * 2007-08-31 2009-03-05 Amgen Inc. Solid-state protein formulation

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3076456A (en) * 1960-03-07 1963-02-05 Elsie B Hunt Hypodermic syringe
US3255752A (en) * 1965-01-28 1966-06-14 Dick Peter Hypodermic syringe
DE3618318A1 (en) * 1986-05-30 1987-12-03 Rau Roland Hypodermic syringe
WO2001030424A1 (en) * 1999-10-22 2001-05-03 Antares Pharma, Inc. Medicament cartridge and injection device
EP1157703A2 (en) * 1994-09-02 2001-11-28 Delab Sustained release of peptides from pharmaceutical compositions
WO2002072171A2 (en) * 2001-03-13 2002-09-19 Mdc Investment Holdings Inc. Pre-filled safety diluent injector

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3076456A (en) * 1960-03-07 1963-02-05 Elsie B Hunt Hypodermic syringe
US3255752A (en) * 1965-01-28 1966-06-14 Dick Peter Hypodermic syringe
DE3618318A1 (en) * 1986-05-30 1987-12-03 Rau Roland Hypodermic syringe
EP1157703A2 (en) * 1994-09-02 2001-11-28 Delab Sustained release of peptides from pharmaceutical compositions
WO2001030424A1 (en) * 1999-10-22 2001-05-03 Antares Pharma, Inc. Medicament cartridge and injection device
WO2002072171A2 (en) * 2001-03-13 2002-09-19 Mdc Investment Holdings Inc. Pre-filled safety diluent injector

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE MEDLINE [Online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; 1997, ETO B ET AL: "Comparison of the antisecretory effect of endogenous forms of peptide YY on fed and fasted rat jejunum." XP002336348 Database accession no. NLM9396069 & PEPTIDES. 1997, vol. 18, no. 8, 1997, pages 1249-1255, ISSN: 0196-9781 *
DATABASE MEDLINE [Online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; 4 September 2003 (2003-09-04), BATTERHAM RACHEL L ET AL: "Inhibition of food intake in obese subjects by peptide YY3-36." XP002336358 Database accession no. NLM12954742 & THE NEW ENGLAND JOURNAL OF MEDICINE. 4 SEP 2003, vol. 349, no. 10, 4 September 2003 (2003-09-04), pages 941-948, ISSN: 1533-4406 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009029795A1 (en) * 2007-08-31 2009-03-05 Amgen Inc. Solid-state protein formulation
JP2011507558A (en) * 2007-08-31 2011-03-10 アムジエン・インコーポレーテツド Solid protein formulation
AU2008293425B2 (en) * 2007-08-31 2014-09-18 Amgen Inc. Solid-state protein formulation

Also Published As

Publication number Publication date
WO2005079888A3 (en) 2005-09-22

Similar Documents

Publication Publication Date Title
JP6608799B2 (en) Highly soluble leptin
JP6618854B2 (en) Engineered polypeptides with increased duration of action and reduced immunogenicity
EP2185178B1 (en) Use of an fgf-21 compound and a glp-1 compound for the treatment of obesity
JP5290177B2 (en) GLP-2 mimetibodies, polypeptides, compositions, methods and uses
AU2003240754B2 (en) Use of osteoprotegerin for the prevention and/or treatment of fibrosis/sclerosis
AU2008257448B2 (en) Unacylated ghrelin as therapeutic agent in the treatment of metabolic disorders
KR20170078668A (en) Compositions and peptides having dual glp-1r and glp-2r agonist activity
KR20120062777A (en) Modified vasoactive intestinal peptide
TW201922279A (en) GLP-2 analogs and PEPTIBODIES for administration before, during or after surgery
KR102569494B1 (en) Isolated polypeptides of cd44 and uses therof
WO2006063596A2 (en) A composition comprising pp for the treatment of gastrointestinal disorders
EP1750753A1 (en) A composition comprising pyy for the treatment of gastrointestinal disorders
WO2005079888A2 (en) Delivery device for delivering pyy
US20080125360A1 (en) Composition Comprising Pyy for the Treatment of Gastrointestinal Disorders
EA005973B1 (en) Use of sarp-1 for the treatment and/or prevention of scleroderma
KR20050059195A (en) THERAPIES FOR CHRONIC INFLAMMATORY DEMYELINATING POLYNEUROPATHY USING INTERFERON-β
ES2324306T3 (en) USE OF IL-17F FOR THE TREATMENT AND / OR PREVENTION OF NEUROLOGICAL DISEASES.
US7803910B2 (en) Soluble CD164 polypeptides
WO2005110454A2 (en) Protective binding members of pyy for the treatment of metabolic disorders
US20050143301A1 (en) Use of osteoprotegerin for the treatment and/or prevention of fibrotic disease
US9353171B2 (en) Use of pituitary adenylate cyclase-activating polypeptide (PACAP) and PACAP analogs for treating contrast-induced nephropathy
WO2005110457A2 (en) Treatment of eating disorders and induction of lipolysis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase
WWP Wipo information: published in national office

Ref document number: 11596973

Country of ref document: US