CA2521379A1 - Pharmaceutical formulations containing methylnaltrexone - Google Patents

Pharmaceutical formulations containing methylnaltrexone Download PDF

Info

Publication number
CA2521379A1
CA2521379A1 CA002521379A CA2521379A CA2521379A1 CA 2521379 A1 CA2521379 A1 CA 2521379A1 CA 002521379 A CA002521379 A CA 002521379A CA 2521379 A CA2521379 A CA 2521379A CA 2521379 A1 CA2521379 A1 CA 2521379A1
Authority
CA
Canada
Prior art keywords
pharmaceutical preparation
methylnaltrexone
preparation
solution
agent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA002521379A
Other languages
French (fr)
Other versions
CA2521379C (en
Inventor
Suketu P. Sanghvi
Thomas A. Boyd
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Progenics Pharmaceuticals Inc
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=33299842&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2521379(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to CA2811272A priority Critical patent/CA2811272C/en
Publication of CA2521379A1 publication Critical patent/CA2521379A1/en
Application granted granted Critical
Publication of CA2521379C publication Critical patent/CA2521379C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/047Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates having two or more hydroxy groups, e.g. sorbitol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives

Abstract

Stable pharmaceutical compositions useful for administering methylnaltrexone are described, as are methods for making the same. Kits, including these pharmaceutical compositions, also are provided.

Description

PHARMACEUTICAL FORIvIULATIONS CONTATNIN~ METHS~LNALTRE~ONE
FIELD OF THE INVENTION
This invention relates to methylnaltrexone pharmaceutical preparations, methylnaltrexone formulations, methylnaltrexone kits, and methods of making the same.
BACKGROUND OF THE INVENTION
Quaternary amine opioid antagonist derivatives have been shown to have utility in a number of contexts. They are considered peripherally acting only, and, therefore, to find particular utility in reducing the side-effects of opioids without reducing the analgesic effect of opioids. Such side effects include nausea, emesis, dysphoria, pruritis, urinary retention, bowel hypomotility, constipation, gastric hypomotility, delayed gastric emptying and immune suppression. The utility of these peripherally acting opioid antagonists is not limited to reducing side-effects stemming from opioid analgesic ~s treatment. Instead, these derivatives also have utility in circumstances where endogenous opioids alone (or in conjunction with exogenous opioid treatment) cause undesirable conditions such as ileus and other such conditions including, but not limited to, those mentioned above.
Methylnaltrexone is a quaternary amine opioid antagonist derivative, discovered 2o in the mid-70s. Methylnaltrexone and some of its uses are described in U.S.
Patents 4,176,186, 4,719,215, 4,861,781, 5,102,887, 5,972,954, and 6,274,591. Stable formulations of methylnaltrexone, however, have heretofore not existed.
Methylnaltrexone apparently was assumed to have a structure that was inherently stable.
The stability of a pharmaceutical composition in solution, however, is not necessarily 2s predictable either over time when stored at room temperature or when autoclaved.
Naloxone is an opioid antagonist that acts both centrally and peripherally. It differs structurally from methylnaltrexone and would be expected to have a different stability in solution. An allegedly stable formulation of naloxone is described in U.S.
Patent No. 5,866,154.
so Surprisingly, it has been discovered that methylnaltrexone is unusually unstable.
It further has been discovered that methylnaltrexone has certain degradation products different from those of naloxone. It also has been discovered that critical parameters and conditions are required for stable formulations of methylnaltrexone.
-2-SUMMARY OF THE INVENTION
In one aspect, the invention provides a composition or preparation that is a solution of methylnaltrexone or a salt thereof, wherein the preparation after autoclaving has a concentration of methylnaltrexone degradation products that does not exceed 2% of s the methylnaltrexone or salt thereof in the preparation. Preferably, the concentration of such degradation products does not exceed 1.5%, 1%, 0.5%, 0.25%, or even 0.125% of the methylnaltrexone or salt thereof in the preparation. The composition or preparation can contain one of, any combination of, or all of a chelating agent, a buffering agent, an anti-oxidant, a cryoprotecting agent, an isotonicity agent and an opioid. The preferred chelating agent is disodium edetate or a derivative thereof. The disodium edetate preferably is at a concentration ranging from between 0.001 and 100 mg/ml, more preferably 0.05 to 25.0 mg/ml, and even more preferably, 0.1 to 2.5 mg/ml. A
preferred buffering agent is citrate buffer. The citrate buffer typically is in a concentration ranging from 0.001 to 100.0 mM, preferably from 0.1 to 10 mM, and more preferably, 0.1 to 5.0 15 mM. A preferred cryoprotecting agent is mannitol.
The composition or preparation preferably has a pH that does not exceed 4.25.
More prefer ably, the pH ranges from 2.0 to 4~.0, 3.0 to 4~.0, and most preferably, from 3.0 to 3.5.
According to another aspect of the invention, a composition or preparation is 2o provided, which in dudes a solution of methylnaltrexone or a salt thereof wherein the preparation after storage at about room temperature for six months has a'concentration of methylnaltrexone degradation products that does not exceed 2% of the methylnaltrexone in the preparation. The concentration of the methylnaltrexone degradation products preferably does not exceed 1.5%, 1.0%, 0.5%, 0.25°/~, and even 0.125%
of the 2s methylnaltrexone in the preparation. The composition or preparation can contain one of, any combination of, or all of a chelating agent, a buffering agent, an anti-oxidant, a cryoprotecting agent, an isotonicity agent and an opioid. The preferred chelating agent and concentrations are as described above. The preferred buffering agent and concentrations are as described above. Preferably, the composition or preparation has a so pH that does not exceed 4.25. The preferred pHs and ranges are as described above.
According to another aspect of the invention, a stable composition or preparation is provided. The composition or preparation is a solution of methylnaltrexone or a salt
3 PCT/US2004/010997 thereof wherein the pH is below 4.25. Preferably, the pH is between 2.75 and
4.25, more preferably, between 3.0 and 4.0, and most preferably, between 3.0 and 3.5.
According to conventional procedures, pH can be adjusted with an acid. Examples of acids useful for this purpose include hydrochloric acid, citric acid, sulfuric acid, acetic acid, and s phosphoric acid. The stable composition or preparation can also include any one of, any combination of, or all of a chelating agent, a buffering agent, an isotonicity agent, an antioxidant, a cryogenic agent, and an opioid.
According to another aspect of the invention, a stable composition or preparation is provided. The composition or preparation is a solution of methylnaltrexone or salt thereof, wherein the solution further comprises a chelating agent in an amount sufficient to inhibit degradation of the methylnaltrexone or salt thereof, whereby the amount is such that the composition or preparation after autoclaving has a concentration of methylnaltrexone degradation products that does not exceed 0.5%, 0.25% or even 0.125% of the methylnaltrexone or salt thereof in the composition or preparation. The is composition or preparation can further include any one of, any combination of, or all of a buffering agent, an isotonicity agent, an antioxidant and an opioid. PrefeiTed chelating agents, buffering agents and pHs are as described above.
According to another aspect of the invention, a composition or preparation is provided. The composition or preparation is a solution of methylnaltrexone or salt 2o thereof in at least one methylnaltrexone degradation inhibiting agent. The agent can be any one of, any combination of, or all of a chelating agent, a buffering agent, and an antioxidant, provided that the solution has a pH ranging from 2.0 to 6Ø The degradation inhibiting agent is present in an amount sufficient to render the composition or preparation stable, wherein the composition or preparation is processed under at least one 2s sterilization technique, and wherein the composition or preparation is substantially free of methylnaltrexone degradation products. The composition or preparation can be stable to storage for at least six months, at least twelve months, or at least twenty-four months, at about room temperature. Preferably, the composition or preparation is stable after autoclaving. The composition or preparation further may include either or both of an 3o isotonicity agent and an opioid. Preferably, the pH of the solution is between 2.75 and 4.25, more preferably, between 3.0 and 4.0, and most preferably, between 3.0 and 3.5.

In any one of the foregoing aspects of the invention, the composition or preparation can be a pharmaceutical composition.
In any one of the foregoing aspects of the invention, the methylnaltrexone can be present in a therapeutically effective amount. In some embodiments, the concentration s of methylnaltrexone ranges from 0.01 to 100 mg/ml. In other embodiments, the methylnaltrexone concentration ranges between 0.1 and 100.0 mg/ml. In other embodiments, the methylnaltrexone ranges between 1.0 and 50.0 mg/ml.
In any one of the foregoing embodiments, the methylnaltrexone can be present in an amount sufficient to treat nausea, emesis, dysphoria, pruritus, urinary retention, ileus, 1o post-operative ileus, post-partum ileus, paralytic ileus, bowel hypomotility, constipation, gastric hypomotility, delayed gastric emptying, decreased biliary secretion, decreased pancreatic secretion, biliary spasm, increased sphincter tone, cutaneous flushing, impaction, sweating, inhibition of gastrointestinal motility, inhibition of gastric emptying, gastrointestinal dysfunction, incomplete evacuation, bloating, abdominal ~s distention, increased gastroesophageal reflux, hypotension, bradycardia, irritable bowel syndrome, or imanunosuppression.
In any of the foregoing embodiments, the methylnaltrexone can be present in an amount sufficient to accelerate discharge from hospital post-surgery (including abdominal surgeries such as rectal resection, colectomy, stomach, esophageal, duodenal, 2o appendectomy9 hysterectomy, or non-abdominal surgeries such as orthopedic, trauma injuries, thoracic or transplantation), for example, by accelerating bowel sounds after surgery, or speeding the time to f rst food intal~e or first bowel movement.
In other important embodiments, the amount is sufficient to induce taxation. This has particular application where the subject is a chronic opioid user.
2s In any one of the foregoing embodiments, the solution of methylnaltrexone or salt thereof may be contained in a sealed container such as a bottle, an infusion bag, a syringe, a vial, a vial with a septum, an ampoule, an ampoule with a septum, or a syringe.
The container may include indicia indicating that the solution has been autoclaved or otherwise subjected to a sterilization technique.
3o According to another aspect of the invention, any of the foregoing embodiments is lyophilized, preferably in the presence of a cryoprotecting agent. The invention therefore provides a lyophilized preparation of methylnaltrexone. Preferably, the
-5-lyophilized preparation is a stable preparation, containing less than 1%, less than 0.5%, less than 0.25% and even less than 0.125% methylnaltrexone degradation product. The preparation can contain a cryoprotecting agent, which preferably is neutral or acidic in water.
s According to another aspect of the invention, a product is provided. The product is a stable lyophilized formulation of methylnaltrexone, wherein the formulation upon reconstitution and water at a concentration of 20 mg/ml has a pH of between 2 and 6. In some embodiments, the formulation upon reconstitution has a pH of about 2, about 3, about 4, about 5, or about 6. The formulation can include a cryoprotecting agent present io in amounts sufficient to render the formulation stable. The cryoprotecting agent in important embodiments are polymerized carbohydrates. A preferred cryoprotecting agent is mannitol. Any one of the foregoing solutions described above can be lyophilized. It therefore is an aspect of the invention that such materials include one or any combination of a buffering agent, a chelating agent, an antioxidant, and an ~s isotonicity agent. Preferred materials are as described above.
According to still another aspect of the invention, a product is provided that includes methylnaltrexone and the degradation inhibiting agent selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations thereof, wherein the degradation inhibiting agent is present in an amount sufficient to 2o render stable the solution of the product containing a concentration of 20 mg/ml methylnaltrexone in water. Preferably, the product when in solution at a concentration of 20 mg/ml methylnaltrexone yields a pH of between 2 and 6.
According to another aspect of the invention, a pharmaceutical preparation is provided. The pharmaceutical preparation contains methylnaltrexone, sodium chloride, 2s citric acid, trisodium citrate, and disodium edetate. In one important embodiment, the methylnaltrexone is present between 20 and 40 mg/ml, the sodium chloride is present between 2 and 6 mg/ml, the citric acid is present between 0.05 and 0.1 mg/ml, the trisodium citrate is present between 0.025 and 0.075 mg/ml, and the disodium edetate is present between 0.5 and 1.0 mg/ml.
so The buffering agent may be any pharmaceutically acceptable buffering agent.
Common buffering agents include citric acid, sodium citrate, sodium acetate, acetic acid, sodium phosphate and phosphoric acid, sodium ascorbate, tartaric acid, malefic acid,
-6-glycine, sodium lactate, lactic acid, ascorbic acid, imidazole, sodium bicarbonate and carbonic acid, sodium succinate and succinic acid, histidine, and sodium benzoate and benzoic acid. The preferred buffering agent is a citrate buffering agent.
The chelating agent may be any pharmaceutically acceptable chelating agent.
s Common chelating agents include ethylenediaminetetraacetic acid (EDTA) and derivatives thereof, citric acid and derivatives thereof, niacinamide and derivatives thereof, and sodium desoxycholate and derivatives thereof. The preferred chelating agent is disodium edetate.
The antioxidant may be any pharmaceutically acceptable antioxidant. Common Io antioxidants include those selected from the group consisting of an ascorbic acid derivative, butylated hydroxy anisole, butylated hydroxy toluene, alkyl gallate, sodium meta-bisulfate, sodium bisulfate, sodium dithionite, sodium thioglycollic acid, sodium formaldehyde sulfoxylate, tocopherol and derivatives thereof, monothioglycerol, and sodium sulf te. The preferred antioxidant is monothioglycerol.
1s 'The ciyoprotecting agent may be any phaumaceutically acceptable cryoprotecting agent. Common cryoprotecting agents include histidine, polyethylene qlycol, polyvinyl pyrrolidine, lactose, sucrose, and mannitol. Improtant cryoprotecting agents are polyols.
The preferred cryoprotecting agent of the invention is mannitol.
The opioid can be any pharmaceutically acceptable opioid. Common opioids are 2o those selected from the group consisting of alfentanil, anileridine, aSiIlladolme, bremazocine, burprenoiphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, 25 nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
The isotonicity agent can be any pharmaceutically acceptable isotonicity agent.
Common isotonicity agents include those selected from the group consisting of sodium chloride, mannitol, lactose, dextrose, glycerol, and sorbitol. The preferred isotonicity 3o agent is mannitol.

The pharmaceutical preparation may optionally comprise a preservative.
Common preservatives include those selected from the group consisting of chlorobutanol, parabens, thimerosol, benzyl alcohol, and phenol.
According to another aspect of the invention, a method is provided for preparing s an autoclaved preparation of a solution of methylnaltrexone or salts thereof, whereby the autoclaved preparation has a concentration of methylnaltrexone degradation products that does not exceed 2% of the methylnaltrexone or salt thereof in the preparation. The method involves providing a solution, having a pH of 4.25 or less, of methylnaltrexone or a salt thereof, and being substantially free of methylnaltrexone degradation products, io and autoclaving the solution. The solution can contain, optionally, any one of, any combination of, or all of a chelating agent, an isotonicity agent, a buffering agent, an antioxidant, a cryoprotecting agent, and an opioid. Preferably, the pH of the solution ranges from 2.0 to 4Ø More preferably, from 3.0 to 4.0, and most preferably from 3.0 to 3.5. Preferred chelating agents, isotonicity agents, buffer ing agents, antioxidants, ~s cryoprotecting agents, and opioids are as described above. PrefciTed concentrations of methylnaltrexone, likewise, are as described above.
According to another aspect of the invention, a method is provided for preparing an autoclaved preparation. The preparation has a concentration of methylnaltrexone degradation products that does not exceed 2% of the methylnaltrexone or salt thereof in ~o the preparation. The method involves providing a solution containing methylnaltrexone or salt thereof and a chelating agent, the solution being substantially free of methylnaltrexone degradation products, and then autoclaving the solution. The chelating agent is present in an amount sufficient to protect the preparation against substantial unwanted degradation of methylnaltrexone or its salt, and maintain the solution to be 2s substantially free of methylnaltrexone degradation products. Preferred chelating agents and concentrations thereof are as described above. The preparation may include, optionally, any one of, any combination of, or all of a buffering agent, an isotonicity agent, an antioxidant, a cryoprotecting agent, and an opioid. Preferred buffering agents, isotonicity agents, antioxidants and opioids, as well as concentrations, are as described 3o above. Preferred pHs of the solution likewise are as described above.
Preferably, the degradation products after autoclaving do not exceed 1.5%, 1%, 0.5%, 0.25%or even 0.125%.

_g_ According to another aspect of the invention, a method is provided for inhibiting the formation of methylnaltrexone degradation products in a preparation that is a solution of methylnaltrexone or salts thereof. The method involves preparing an aqueous solution containing at least one methylnaltrexone degradation inhibiting agent selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, a cryoprotecting agent, and combinations thereof. A powdered source of methylnaltrexone or salt thereof is dissolved into the solution to form the preparation. The preparation has or is adjusted without addition of a pH-adjusting base to have a pH of between 2 and 6. More preferably, the pharmaceutical preparation is adjusted to have a pH ranging from 3 to 5, 1o more preferably, 3 to 4, and most preferably, 3.0 to 3.5. An isotonicity agent may be added to the solution. Likewise, an opioid may be added to the solution.
In any one of the foregoing aspects of the invention, the preparation can be a pharmaceutical preparation.
According to another aspect of the invention, a method is provided for preparing ~s a stable pharmaceutical preparation that is an aqueous solution of methylnaltrexone or salts thereof to inhibit formation of methylnaltrexone degradation products. A
solution is provided containing methylnaltrexone or salts thereof and at least one methylnaltrexone degradation inhibiting agent. The solution is processed under at least one sterilization technique prior to and/or after terminal filling the solution in a sealable container to form 2o the stable pharmaceutical preparations wherein the method is carried out without the addition of pH-adjusting base to the solution. The methylnaltrexone degradation inhibiting agent can be selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations thereof. An isotonicity agent can be added. A cryoprotecting agent can also be added. Likewise, an opioid can be added.
2s Preferred chelating agents, buffering agents, antioxidants, isotonicity agents, cryoprotecting agents, and opioids are as described above. Preferred concentrations are as described above. The solution may be processed to adjust the pH. This is preferably done using an acid. Most preferably, the solution is adjusted to a range between a pH of 2 and 6, more preferably, between 3 and 5, 3 and 4, and most preferably between 3.0 and so 3.5. The material can be contained in a sealed container. The container can be purged with nitrogen and/or sparged to eliminate oxygen.

In some embodiments of the invention, parenteral formulations are provided. In one embodiment, the formulation made by dissolving methylnaltrexone diluted in water, to which mannitol is added. The solution is then filter sterilized followed by lyophilization. Therefore, the product may be provided in lyophilized form, and in s combination with certain cryoprotectants such as mannitol or lactose.
Optionally, a reconstituting diluent is provided, such as a physiological saline diluent.
According to another aspect of the invention, a kit is provided. The kit is a package containing a sealed container comprising any one of the preparations described above, together with instructions for use. The kit can also include a diluent container containing a pharmaceutically acceptable diluent. The kit can further comprise instructions for mixing the preparation and the diluent. The diluent can be any pharmaceutically acceptable diluent. Well known diluents include 5°/~
dextrose solution and physiological saline solution. The container can be an infusion bag, a sealed bottle, a vial, a vial with a septum, an ampoule, an ampoule with a septum, an infusion bag or a ~s syringe. The lcit further can contain an opioid container containing an opioid. The containers can optionally include indieia indicating that the containers have been autoclaved or otherwise subjected to sterilization techniques. The kit can include instructions for administering the various solutions contained in the containers to subjects.
20 The invention also involves methods of treatment. According to another aspect of the invention, a method is provided for treating a subject in need of such treatment with an effective amount of methylnaltrexone or a salt thereof. The method involves administering to the subject an effective amount of methylnaltrexone or salt thereof in any one of the pharmaceutical preparations described above, detailed herein, and/or set zs forth in the claims. In one aspect, the method is a method for inhibiting a peripheral opioid receptor in a human subject. In another aspect, the method is for reducing a side-effect of opioid treatment. In another aspect, the method is for treating any one of a condition selected from the group consisting of nausea, emesis, dysphoria, pruritus, urinary retention, ileus, post-operative ileus, post-partumileus, parallytic ileus, bowel so hypomotility, constipation, gastric hypomotility, delayed gastric emptying, decreased biliary secretion, decreased pancreatic secretion, biliary spasm, increased sphincter tone, cutaneous flushing, impaction, sweating, inhibition of gastrointestinal motility, inhibition of gastric emptying, gastrointestinal dysfunction, incomplete evacuation, bloating, abdominal distention, increased gastroesophageal reflux, hypotension, bradycardia, irritable bowel syndrome, or immunosuppression.
In any of the foregoing embodiments, the methylnaltrexone can be present in an s amount sufficient to accelerate discharge from hospital post-surgery, accelerate bowel sounds after surgery, or induce laxation.
The subject can be any subject in need of such treatment. Important subjects include those receiving opioids including opioids for pain, cancer or surgical patients, or immunosuppressed or immunocompromised patients (including HIV infected patients), io patients with advanced medical illness, terminally ill patients, patients with neuropathies, patients with rheumatoid arthritis, patients with osteoarthritis, patients with chronic pack pain, patients with spinal cord injury, patients with chronic abdominal pain, patients with chronic pancreatic pain, patients with pelvic/perineal pain, patients with fibromyalgia, patients with chronic fatigue syndrome, patients with migraine or tension headaches, ~s patients on hemodialysis, and patients with sickle cell anemia.
In the foregoing descr iption, applicants have described the invention in connection with methylnaltrexone or salts thereof. Such salts include, but are not limited to, bromide salts, chloride salts, iodide salts, carbonate salts, and sulfate salts. It should be understood, however, that methylnaltrexone is a member of a class of compounds 20 known as quaternary derivatives of noroxymorphone9 as disclosed in LT.S.
Patent 110.
4,176,1 ~6, the entire disclosure of which is incorporated herein by reference. It is believed that the invention extends to any such quaternary derivative of noroxymorphone, and the invention is intended to embrace pharmaceutical preparations, methods and kits containing such derivatives. Another aspect of the invention then 2s embraces the foregoing summary but read in each aspect as if any such derivative is substituted wherever "methylnaltrexone" appears. Likewise, the invention also embraces each and every claim read as if the term "quaternary derivative of noroxymorphone"
were substituted whenever "methylnaltrexone" appears.

BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graph depicting methylnaltrexone degradation products eluting from a column at time zero (peak Nos. l, 2 and 4 are degradation products; peak No 4 is methylnaltrexone; peak no 5. O-methylnaltrexone bromide).
Figure 2 is a graph depicting methylnaltrexone degradation products eluting from a column at 12 months (peak Nos. 1, 2 and 4 are degradation products; peak No 4 is methylnaltrexone; peak no 5. O-methylnaltrexone bromide).
Figure 3 is a schematic representation of a kit according to the invention containing the formulations described herein.
DETAILED DESCRIPTION OF THE INVENTION
Applicants have discovered that during the autoclaving process, methylnaltrexone in aqueous solution tends to degrade to a surprising extent. The amount of degradation resulting from simple autoclaving (122 °C, 15 lbs. pressure for 20 min.) can be as high as Is 10°f~. The degradation products are depicted in Figure 1, and appear to include at least two predominant degradants having relative retention times (RRT) of 0.72 (2.~2~
minutes) and 0.~9 (3.435 minutes) and, with other minor forms as can be observed. The degradant identified by the 0.72 RRT peak appears in small amounts, 0.074, immediately upon dissolving the methylnaltrexone into solution and increases overtime with storage 20 or autoclaving 0.25°/~. The degradant identified by the 0.~~ RRT
peak appears only after storage over time or after autoclaving (<0.05°/~ and 0.724°/~, respectively). Applicants also have discovered that methylnaltrexone is unstable in aqueous solutions when stored at room temperature or even at 4 °C for significant (but commercially necessary) periods of time such as 6 months, 12 months or even two years. Degradation occurs without 2s regard to whether the aqueous solution was previously autoclaved or filter sterilized. It would be desirable to stabilize formulations of methylnaltrexone such that following the autoclaving process or following storage (or both autoclaving and storage), the amount of the total degradation products would be less than 2.0%, 1.5%, 1.0%, 0.5%, 0.25%, and even 0.125%.
3o The invention provides stable formulations of methylnaltrexone. By stable solutions of methylnaltrexone, it is meant that following autoclaving at 122 °C, 15 lbs.
pressure for 20 minutes, the methylnaltrexone degradation products resulting from such conditions are not more than 2% of the total methylnaltrexone present in a given solution. By stable solution of methylnaltrexone, it also is meant that following storage of an unautoclaved solution at room temperature for twelve months, the methylnaltrexone degradation products resulting from such conditions are not more than s 2% of the total methylnaltrexone present in a given solution. By stable solutions of methylnaltrexone, it is also meant that following storage of an unautoclaved solution at room temperature for two months, the methylnaltrexone degradation products resulting from such conditions are not more than 1.0 % of the total methylnaltrexone present in a given solution. By stable lyophilized formulations of methylnaltrexone, it is meant that io following lyophilization and storage at room temperature of methylnaltrexone for two months, and their reconstitution in water the methylnaltrexone degradation products resulting from such conditions are not more than 1.0 % of the total methylnaltrexone present in a given solution.
It was surprisingly discovered that pH alone can solve the problem of excessive Is methyhlaltrexone degradation products. In particular, it was discovered that when the pH of a methylnaltrexone solution containing 2mghmL of methylnaltrexone was at about 4.25 pH or less, there was a steep drop-off in the amount of methylnaltrexone degradation products following autoclaving. When the pH of the solution containing methylnaltrexone was adjusted to between 3.5 and 4.0, then the total percentage of 2o degradants fell below 2°/~, and in certain instances even below 1.39°/~. S~Jhen the pH was adj usted to between 3.0 and 3.5, the percentage of total degradants dropped to about 0.23% after autoclaving. It was also noted that there was a significant drop, before a plateau, when the pH of the methylnaltrexone solution was brought to below 6.0 prior to autoclaving. Adjusting pHs to between 4.25 and 6 was not sufficient to produce stable 2s formulations of methylnaltrexone (through the adjustment of pH alone). As will be seen below, however, manipulating other parameters in concert with pH resulted in stable formulations of methylnaltrexone anywhere in a range from a pH of 2.0 to 6Ø
The benefits of a low pH on the stability of methylnaltrexone formulations persisted in the presence of chelating agents, isotonicity agents, buffering agents, and antioxidants.
so Thus, the invention in one aspect provides stable formulations of methylnaltrexone in solution, wherein the pH is below 4.25, preferably between 3.0 and 4.0, and most preferably between 3.0 and 3.5.

Applicants also noted that despite setting the pH of a methylnaltrexone solution at points between 3.0 and 6.0 using a pH-adjusting acid or pH-adjusting base prior to autoclaving and despite the benefits obtained from lower pH, the pH of the autoclaved sample drifted almost immediately to about 7Ø It was therefore tested, in particular, s whether buffering agents could eliminate the pH drift that resulted from autoclaving without negatively affecting the ability to protect against heat degradation resulting from autoclaving. Applicants discovered that buffering agents indeed could be employed to stabilize the pH of methylnaltrexone solutions throughout the autoclaving process without permitting degradation products to exceed acceptable minimums. Buffers were to used in concentrations ranging from 0.25 mM to 25 mM. Acceptable levels of degradation products were obtained at all buffer concentrations tested. It was noted, however, that citrate buffer had properties more desirable than those of acetate buffer. In particular, the addition of citrate buffer did not seem to alter in any material respects the amount of degradation products resulting from autoclaving the methylnaltrexone ~s solution, resulting in less than 0.23°/~ of degradation products at pH of 3.5. The addition of acetate buffer, however, appeared to increase somewhat the amount of methylnaltrexone degradation products, although not to unacceptable levels, resulting in less than 1.39% of degradation products at pH of 3.6. Nonetheless, citrate buffer surprisingly is preferable to acetate buffer. The preferred citrate buffer range is between 2o about 2 and 5 mM.
Buffers in general are well known to those of ordinary skill in the art.
Buffer systems include citrate buffers, acetate buffers, borate buffers, and phosphate buffers.
Examples of buffers include citric acid, sodium citrate, sodium acetate, acetic acid, sodium phosphate and phosphoric acid, sodium ascorbate, tartartic acid, malefic acid, 2s glycine, sodium lactate, lactic acid, ascorbic acid, imidazole, sodium bicarbonate and carbonic acid, sodium succinate and succinic acid, histidine, and sodium benzoate and benzoic acid.
Applicants also discovered, surprisingly, that a chelating agent alone was capable of reducing the amount of degradation products to acceptable levels. In particular, pH
so was not adjusted and disodium edetate was added at concentrations of 0.01, 0.1, 0.25, 0.5, 0.75, and 1.0 mglmL. The disodium edetate stabilized methylnaltrexone against heat degradation in a concentration-dependent manner. As little as 0.01 mg/mL
had a substantial effect on the amount of degradants, yielding approximately 2.3%
total degradants. A concentration of 0.1 mg/mL resulted in under 1.5% total degradants.
There was a critical point at approximately 0.3 - 0.4 mg/mL where the total degradants became slightly under 0.5% and leveled off with increasing amounts of disodium s edetate. Thus, disodium edetate alone was sufficient to render stable an unbuffered solution of methylnaltrexone with no adjustment to pH. This was a surprising result.
Applicants believe that the result is not limited to disodium edetate.
Instead, other chelating agents well known to those of ordinary skill in the art will be useful according to the invention. Chelating agents are chemicals which form water soluble coordination compounds with metal ions in order to trap or remove the metal irons from solution, thereby avoiding the degradative effects of the metal ions.
Chelating agents include ethylenediaminetetraacetic acid (also synonymous with EDTA, edetic acid, versene acid, and sequestrene), and EDTA derivatives, such as dipotassium edetate, disodium edetate, edetate calcium disodium, sodium edetate, trisodium edetate, and ~s potassium edetate. ~ther chelating agents include citric acid and derivatives thereof.
Citric acid also is known as citric acid monohydrate. Derivatives of citric acid include anhydrous citric acid and trisodiumcitrate-dihydrate. Still other chelating agents include niacinamide and derivatives thereof and sodium desoxycholate and derivatives thereof.
A synergistic effect of pH and disodium edetate was also observed. At pH 3 -3.5, in the 2o presence of citrate buffer (25 mI~I), and 0.01 mg/mL disodium edetate, the total degradants after autoclaving amounted to less than 0.4°/~. Under the same conditions, except increasing the concentration of disodium edetate to 1 mg/mL, there was no detectable difference. That is, the degradants were on the order of approximately 0.4%
after autoclaving. The circumstance, however, differed when pH was adjusted upwardly 2s to between 6.0 and 7.0 in an unbuffered system. In particular, at a pH
adjusted upwardly to between 6.0 and 7.0, the total degradants were above 3 - 6% at a concentration of 0.01 mg/mL disodium edetate and approximately 2.8% at 1.0 mg/mL disodium edetate.
This at first glance appears anomalous with the results described above, where disodium edetate alone was sufficient to bring total degradants under 0.5% at concentrations above so approximately 0.3 disodium edetate mg/mL. It was discovered, however, that the increase in degradation was due to the addition of a pH-adjusting base to the solution containing methylnaltrexone to upwardly adjust the pH to 6.0 - 7Ø Therefore, it was discovered unexpectedly that the addition of a pH-adjusting base, such as sodium hydroxide, to a solution containing methylnaltrexone should be avoided in order to minimize the presence of degradants.
The same results were achieved through a combination of acetate buffer and s disodium edetate at 0.01 mg/mL and 1.0 mg/mL, although, once again, citrate buffer seemed to work surprisingly better than acetate buffer in protecting methylnaltrexone from heat degradation. Higher levels of disodium edetate in the presence of acetate buffer could compensate, however, for the differential effect that was observed when using citrate buffer versus acetate buffer. It is to be noted that citrate buffer also is a io chelating agent, which might contribute to its apparent superior properties. However, there was no concentration-dependent stabilization due to citrate buffer and it would appear that the chelating effect of citrate is not wholly responsible for the differential effects observed between citrate buffer and acetate buffer.
Applicants also believe that antioxidants will be useful according to the is invention. Antioxidants are substances capable of inhibiting oxidation by removing free radicals fi~om solution. Antioxidants are well known to those of ordinary skill in the art and include materials such as ascorbic acid, ascorbic acid derivatives (e.g., ascorbylpalmitate, ascorbylstearate, sodium ascorbate, calcium ascorbate, etc.), butylated hydroxy anisole, buylated hydroxy toluene, alkylgallate, sodium meta-bisulfate, sodium 2o bisulfate, sodium dithionite, sodium thioglycollic acid, sodium formaldehyde sulfoxylate, tocopherol and derivatives thereof, (d-alpha tocopherol, d-alpha tocopherol acetate, dl-alpha tocopherol acetate, d-alpha tocopherol succinate, beta tocopherol, delta tocopherol, gamma tocopherol, and d-alpha tocopherol polyoxyethylene glycol 1000 succinate) monothioglycerol, and sodium sulfite. Such materials are typically added in ranges from 2s 0.01 to 2.0%.
The pharmaceutical preparations of the invention also may include isotonicity agents. This term is used in the art interchangeably with iso-osmotic agent, and is known as a compound which is added to the pharmaceutical preparation to increase the osmotic pressure to that of 0.9% sodium chloride solution, which is iso-osmotic with human 3o extracellular fluids, such as plasma. Preferred isotonicity agents are sodium chloride, mannitol, sorbitol, lactose, dextrose and glycerol.

Optionally, the pharmaceutical preparations of the invention may further comprise a preservative. Suitable preservatives include but are not limited to:
chlorobutanol (0.3 - 0.9% W/V), parabens (0.01- 5.0%), thimerosal (0.004 -0.2%), benzyl alcohol (0.5 - 5%), phenol (0.1-1.0%), and the like.
In view of the success achieved with disodium edetate alone in an unbuffered system, it would have been expected that stable formulations could be prepared at virtually any pH simply by optimizing the various potential methylnaltrexone degradation inhibiting agents. Such agents include those as described above, that is, chelating agents, buffering agents, antioxidants, and the like. It was discovered, however, that stable formulations of methylnaltrexone in solution could not be obtained with such degradation inhibiting agents at pHs above 6. Thus, in one aspect of the invention, stable pharmaceutical preparations containing methylnaltrexone in solution are permitted, wherein the solution further includes an agent selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations is thereof, provided that the solution has a pH ranging from between 2 to 6.
The stable pharmaceutical preparations of the invention arc stable not only to heat degradation resulting from autoclaving, but also to other sterilization processes used during manufacturing. Sterilization processes or techniques as used herein include aseptic techniques such as one or more filtration (0.45 or 0.22 micron filters) steps, ao autoclaving, and a combination of f ltration and autoclaving. Theg~ also are stable to long term storage. The stable formulations of the invention are stable for at least six months at temperatures of 30 °C or less, preferably a range from 5 °C
to 30 °C, and, more preferably, they are stable at a temperature above 15 °C for at least six months. More particularly, the stable pharmaceutical preparations are stable for periods of at least six 2s months, at least twelve months, and even at least twenty-four months at about room temperature or 25 °C. Such preparations remain substantially free of methylnaltrexone degradation products, that is, such solutions contain less than 2%
methylnaltrexone degradation products compared to the total amount of methylnaltrexone in the solution.
Applicants also discovered, surprisingly, that lyophilizing conditions could so dramatically affect the amount of methylnaltrexone degradation products.
The pharmaceutical preparations of the invention therefore may advantageously include cryoprotective agents, which protect methylnaltrexone from the harmful effects of freezing. Such agents also can prevent caking and flaking, which can be problematic in reconstituting a solution and in manufacturing processing. Important cryoprotecting agents are mannitol, lactose, sucrose, polyethylene glycol and polyvinyl pyrrolidine.
Most preferred is mannitol. It is believed that cryoprotecting agents which result in a s reconstitution pH of 6.0 and higher or which are basic will contribute also to degradation of methylnaltrexone due to pH effects discussed above. Thus, preferred cryoprotecting agents are those which, together with the other components of the formulation, result in a pH in the preferred ranges described above. Preferably, the cryoprotecting agent is neutral or acidic.
1o The amount of methylnaltrexone in the solution is effective to treat completely, ameliorate, or even prevent conditions associated with activation of endogenous opioid receptors, in particular, peripheral opioid receptors such as mu opioid receptors. Such conditions include nausea, emesis, dysphoria, pruritus, urinary retention, ileus, post-operative ileus, post-partumileus, parallytic ileus, bowel hypomotility, constipation, is gastric hypomotility, delayed gastric emptying, decreased biliary secretion, decreased pancreatic secretion, biliary spasm, increased sphincter tone, cutaneous flushing, impaction, sweating, inhibition of gastr ointestinal motility, inhibition of gastric emptying, gastrointestinal dysfunction, incomplete evacuation, bloating, abdominal distention, increased gastroesophageal reflux, hypotension, bradycardia, irritable bowel 2o syndrome, or immunosuppression. ~ne important use is in the treatment of constipation, i.e., less than one bowel movement in 3 days or less than 3 bowel movements in a week.
In any of the foregoing embodiments, the methylnaltrexone can be present in an amount sufficient to accelerate discharge from hospital post-surgery, accelerate bowel sounds after surgery, or induce Taxation. Such amounts are well known to those of 2s ordinary skill in the art and are described in the literature, including the patents listed in the background of the invention. The methylnaltrexone may also be in a salt form, including the bromide, chloride, iodide, carbonate, and sulfate salts of methylnaltrexone.
Patients treatable with the formulations of the invention include those receiving opioids including opioids for pain, cancer or surgical patients, immunosuppressed or 3o immunocompromised patients (including HIV infected patients), patients with advanced medical illness, terminally ill patients, patients with neuropathies, patients with rheumatoid arthritis, patients with osteoarthritis, patients with chronic pack pain, patients with spinal cord injury, patients with chronic abdominal pain, patients with chronic pancreatic pain, patients with pelvic perineal pain, patients with fibromyalgia, patients with chronic fatigue syndrome, patients with migraine or tension headaches, patients on hemodialysis, and patients with sickle cell anemia.
The pharmaceutical preparations of the invention also can include an opioid.
The therapeutic use of opioids is well known and, again, is described in both the literature and the patents mentioned above. Opioids include alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, io hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
It should be understood that the pharmaceutical preparations of the invention will 1s typically be held in bottles, vials, ampoules, infusion bags, and the like, any one of which may be sparged to eliminate oxygen or purged with nitrogen. In some embodiments, the bottles vials and ampoules are opaque, such as when amber in color. Such sparging and purging protocols are well known to those of ordinary skill in the art and should contribute to maintaining the stability of the pharmaceutical preparations.
The 2o pharmaceutical preparations also, in certain embodiments, are expected to be contained within syringes.
According to another aspect of the invention, kits also are provided.
Referring to Figure 3, a kit 10 is depicted. The kit 10 includes a pharmaceutical preparation vial 12, a pharmaceutical preparation diluent vial 14, an opioid vial 16, and an opioid diluent vial 2s 1 ~. The kit also includes instructions 20. The vial 14 containing the diluent for the pharmaceutical preparation is optional. The vial 14 contains a diluent such as physiological saline for diluting what could be a concentrated solution of methylnaltrexone contained in vial 12. The instructions can include instructions for mixing a particular amount of the diluent with a particular amount of the concentrated so pharmaceutical preparation, whereby a final formulation for injection or infusion is prepared. The instructions may include instructions for use in a patient controlled analgesia (PCA) device. Likewise, the kit optionally contains an opioid in the opioid vial 16, which also optionally may be in a concentrated form. The optional vial 18 contains a diluent for a concentrated opioid. The instructions also may include instructions for mixing the opioid with the pharmaceutical preparation and/or diluting the opioid with the opioid diluent contained in the opioid diluent vial 18. The instructions, s therefore, would take a variety of forms depending on the presence or absence of diluent and opioid. The instructions 20 can include instructions for treating a patient with an effective amount of methylnaltrexone. It also will be understood that the containers containing the pharmaceutical preparation, whether the container is a bottle, a vial with a septum, an ampoule with a septum, an infusion bag, and the like, can contain indicia such as conventional markings which change color when the pharmaceutical preparation has been autoclaved or otherwise sterilized.
The pharmaceutical preparations of the invention, when used in alone or in cocktails, are administered in therapeutically effective amounts. A
therapeutically effective amount will be determined by the parameters discussed below; but, in any ~s event, is that amount which establishes a level of the drugs) effective for treating a subject , such as a human subject, having one of the conditions described herein. An effective amount means that amount alone or with multiple doses, necessary to delay the onset of, inhibit completely or lessen the progression of or halt altogether the onset or progression of the condition being treated. When administered to a subject, effective 2o amounts will depend, of course, on the particular condition being treated;
the severity of the condition; individual patient parameters including age, physical condition, size and weight; concurrent treatment; frequency of treatment; and the mode of administration.
These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum 2s dose be used, that is, the highest safe dose according to sound medical judgment.
The pharmaceutical preparations of the present invention may include or be diluted into a pharmaceutically-acceptable carrier. The term "pharmaceutically-acceptable carrier" as used herein means one or more compatible solid, or semi-solid or liquid fillers, diluants or encapsulating substances which are suitable for administration so to a human or other mammal such as a dog, cat, horse, cow, sheep, or goat.
The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The carriers are capable of being commingled with the preparations of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy or stability. Carriers suitable for oral, subcutaneous, intravenous, intramuscular, etc. formulations can be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
A variety of administration routes are available. The particular mode selected will depend of course, upon the particular drug selected, the severity of the disease state being treated and the dosage required for therapeutic efficacy. The methods of this invention, generally speaking, may be practiced using any mode of administration that is medically io acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include oral, rectal, sublingual, topical, nasal, transdermal or parenteral routes. The term "parenteral" includes subcutaneous, intravenous, intramuscular, or infusion.
Dosage may be adjusted appropriately to achieve desired drug levels, locally or ~s systemically. Generally, daily oral doses of active compounds will be from about 0.1 mg/kg per day to 30mg/lcg per day. It is expected that IV doses in the range of 0.01 -1.00 mg/lbg will be effective. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
Continuous zo IV dosing over, for example, 24 hours or multiple doses per day also are contemplated to achieve appropriate systemic levels of compounds. Preferred subcutaneous doses for chronic opioid users to induce taxation are 0.1-0.3 mg/kg, and preferred oral doses for the same patient population are 1.0-3.0 mg/kg. Preferred IV doses to treat post operative ileus are 0.15 mg/kg.
2s The invention also involves methods for preparing autoclaved pharmaceutical preparations that have concentrations of methylnaltrexone degradation products that do not exceed 2% of the methylnaltrexone or salt thereof in the preparation.
Aqueous solutions of methylnaltrexone are prepared. A pH-adjusting acid is added to adjust the pH to 4.25 or less, preferably to a range of between 3.0 and 3.5. The solution is then 3o autoclaved according to standard procedures. One such procedure involves autoclaving at 122 °C and 15 pounds of pressure for 20 minutes. The pharmaceutical preparation can contain any one, any combination of or all of a chelating agent, an isotonicity agent, a buffering agent, an antioxidant, a cryoprotective agent, and an opioid.
According to another aspect of the invention, a pharmaceutical preparation containing methylnaltrexone in a aqueous solution is prepared by combining a chelating agent with the methylnaltrexone solution and then autoclaving the solution. The aqueous solution s of methylnaltrexone may contain any one of, any combination of or all of a buffering agent, an antioxidant, an isotonicity agent and an opioid.
According to yet another aspect of the invention, a pharmaceutical preparation containing methylnaltrexone in a lyophilized formulation is prepared by combining a cryoprotective agent, such as mannitol, with the methylnaltrexone formulation.
The to lyophilized preparation may also contain any one of, any combination of, or all of a buffering agent, an antioxidant, an isotonicity agent and an opioid.
The invention also involves methods of inhibiting the formation of methylnaltrexone degradation products in a solution containing methylnaltrexone by combining any one of, any combination of or all of a chelating agent, a buffering agent is and an antioxidant with methylnaltrexone or salt thereof in solution. In one preferred embodiment, the aqueous solution containing the chelating agent, buffering agent and/or antio:~idant is first prepared, then a powdered source of methylnaltrexone or salt thereof is dissolved into the aqueous solution.
The invention also involves methods of inhibiting the formation of 20~ methylnaltrexone degradation products in a gel containing methylnaltrexone by combining any one of, any combination of or all of a chelating agent, a buffering agent and an antioxidant with methylnaltrexone or salt thereof in a gel matrix. In one preferred embodiment, the gel containing the chelating agent, buffering agent and/or antioxidant is first prepared, then a powdered source of methylnaltrexone or salt thereof is dissolved zs into the gel. As used herein, solution embraces gels.
The pharmaceutical preparations of the invention may be provided in particles.
Particles as used herein means nano or microparticles (or in some instances larger) which can consist in whole or in part of the peripheral opioid antagonists or the other therapeutic agents) as described herein. The particles may contain the therapeutic 3o agents) in a core surrounded by a coating, including, but not limited to, an enteric coating. The therapeutic agents) also may be dispersed throughout the particles. The therapeutic agents) also may be adsorbed into the particles. The particles may be of any order release kinetics, including zero order release, first order release, second order release, delayed release, sustained release, immediate release, and any combination thereof, etc. The particle may include, in addition to the therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited s to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof. The particles may be microcapsules which contain the antagonist in a solution or in a semi-solid state. The particles may be of virtually any shape.
Both non-biodegradable and biodegradable polymeric materials can be used in the manufacture of particles for delivering the therapeutic agent(s). Such polymers may Io be natural or synthetic polymers. The polymer is selected based on the period of time over which release is desired. Bioadhesive polymers of particular interest include bioerodible hydrogels described by H.S. Sawhney, C.P. Pathak and J.A. Hubell in Maen~naolecules, (1993) 26:581-587, the teachings of which are incorporated herein.
These include polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic is acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate).
20 The invention also provides methods for preparing stable pharmaceutical preparations containing aqueous solutions of methylnaltrexone or salts thereof to inhibit formation of methylnaltrexone degradation products. A solution is provided that contains methylnaltrexone or salts thereof and at least one methylnaltrexone inhibiting agent. The solution is processed under at least one sterilization technique prior to and/or 2s after terminal filing the solution in the sealable container to form a stable pharmaceutical preparation, wherein the method is carried out without the addition of a pH-adjusting base to the solution.

EXAMPLES
Example 1 Manufacturing Process for a Pharmaceutical Formulation of Methylnaltrexone A manufacturing process can be outlined as follows:
1. Add required amount of water for injection (~80% or final volume) to a stainless steel tank.
2. Add chelating agent to the tank and stir till dissolved.
3. Add buffering agent to the tank and stir till dissolved.
4. Add methylnaltrexone to the tank and stir till dissolved.
5. Add isotonicity agent to the tank and stir till dissolved.

6. Adjust the pH of the solution to pH 3.25.
7. Add water for injection to increase the volume to the required amount.

1s 8. Transfer material to supply pressure vessel.

9. Sterile filter into a sterile stainless steel pressure vessel.

10. Fill into bottlcs/vials, purge with nitrogen and then stopper the bottles/vials.
11. Sterilize the filled vials by autoclaving.
2o Exact amount of cxcibients to be used:
I2isodimn cdctate = 0.75 mghnl Added in step 2 Sodium Citrate = 0.199 mg/ml Added in step 3 Citric acid = 0.35 mg/ml Added in step 3 Sodium Chloride = 8.5 mg/ml Added in step 5 2s The order of addition of excipients is described above. Steps 2 to 5 can take place in any order.
When all excipients and drug have been added, step 6, pH of the solution is adjusted by addition of acid. If a buffering agent is used in the solution, pH
adjustment may not be required.
so There are no specifics on the temperature or the stirring speed during the formulation. The temperature during formulation can be as high as 80 °C.

Example 2 Preferred Manufacturing Process for a Pharmaceutical Formulation of Methylnaltrexone s A preferred manufacturing process is as follows:
100 ml of 20 mg/ml solution of methylnaltrexone solutions 1. Add 80 ml of water for injection (~80% or final volume) to a stainless steel tank.
2. Add 75 mg of disodium edetate, a chelating agent, to the tank and stir till dissolved.
Io 3. Add 19.9 mg of sodium citrate and 35 mg of citric acid (as buffering agents) to the tank and stir till dissolved.
4. Add 2000 mg of methylnaltrexone to the tank and stir till dissolved.
5. Add 850 mg of sodium chloride, an isotonicity agent, to the tank and stir till dissolved.
is 6. Adjust the pH of the solution if necessary.
7. Add water for injection to increase the volume to 100 ml.
8. Transfer the material to supply pressure vessel.
9. Sterile filter using a 0.22 micron filter into a sterile stainless steel pressure vessel.
10. Fill, purge with nitrogen and then stopper the bottles/vials.
20 11 a Sterilize the filled vials by autoclaving.
Example 3 12 Month Stability of Pharmaceutical Preparation Methylnaltrexone 2s Methylnaltrexone (bromide salt) and its degradation products in an isotonic saline solution were tested upon manufacture of the solution (no added stabilizers, sterile filtered, not autoclaved) and upon storage at room temperature for 12 months using a Hewlett-Packard HP1100 series, HPLC system equipped with quaternary gradient pump, programmable variable wavelength UV detector and a Millennium data acquisition 3o system. Two mobile phases were prepared as follows:
The reagents, standards and media included naltrexone methobromide as a reference standard, trifluoroacetic acid (ACS grade), acetonitrile (HPLC
grade), Milli-Q

water (or equivalent), and methanol (HPLC grade). The solutions were prepared as follows. Mobile phase A (85:15:0.1) (water:methanolarifluoroacetic acid): 850 mL of Milli-Q water was added to a suitable container, to which 150 mL of methanol and 1.0 mL of trifluoroacetic acid were added. The solution was mixed well and allowed to s equilibrate to room temperature. The solution was degassed by helium sparge.
Mobile phase B (methanol): Methanol was added to a suitable container and degassed by helium sparge.
Instrumental Conditions to Analytical Column: Metachem Inertsil ODS3, S~m, 150 x 4.6 mm or equivalent Mobile phase: A mixture of Mobile phase A and B is used as shown in Table I:
Table I
Time (minutes) %A %B

15.1 100 0 Column temperature:50 C

is Detection: U~ at 280 nm Injection volume:20 ~.L

Run time: 20 minutes Flow rate: 1.5 mL/minute Quantitation: Peak area responses Results:
20mg/ml saline drug product lot CTM-02085 Peak Initial 12 N - months o. RRT % DegradantsRRT % Degradants 1 degradation 0.72 0.07 0.74 0.25 product 2 degradation 0.89 <0.05 0.89 0.72 product 3 methylnaltrexone1.00 99.7 1.00 98.6 , 4 degradation 1.48 0.06 1.40 0.16 product O-Methylnaltrexone1.57* 0.17 ~ 1.54* 0.17 Bromide (process impurity) Samples from the methylnaltrexone saline formulation (not autoclaved) were analyzed for methylnaltrexone degradation products before and after storage for 12 s months at 25 °C.
The starting material was analyzed by HPLC. As shown in Fig. 1, methylnaltrexone is a peak having an I~T of 1.0 (4.364 minutes). An additional peak was identified as O-methyl naltrexone methobromide, RRT about 1.57 (6.868 minutes).
The O-methyl-naltrexone is not a degradant of methylnaltrexone but a result from the to methylnaltrexone (drug substance) manufacturing process.
The material stored for 12 months was similarly analyzed by HPLC. The chromatogram is shown in Fig. 2.
As in the starting material, the HPLC analysis of the sample stored for 12 months showed methylnaltrexone RRT of 1.00 (3.839 minutes), O-methyl-methylnaltrexone Is RRT of about 1.53 (5.866 minutes). However, HPLC analysis revealed that the methylnaltrexone saline formulation which was stored for 12 months had at least three degradation products formed during the manufacturing or during storage of the finished drug product. The degradant peak RRT's were approximately 0.74 (2.828 minutes) , 0.89 (3.435 minutes) and 1.40(5.326 minutes).
2o HPLC analysis was also conducted, prior to storage, on a methylnaltrexone solution manufactured using an isotonic saline solution (no added stabilizers), sterile filtered, and autoclaved. This saline, autoclaved solution contained the degradation products formed during manufacturing or storage, as described above (data not shown).
s Example 4 Preparation of a Subcutaneous Formulation The degradation products seen with very low citrate level were the same as those seen with normal saline solution. These low citrate formulas were autoclaved and after to three months the amount of degradation products seen were less than 0.1%
for each degradation product. The formula used for the citrate/EDTA formulation is listed below:
mg/mL

Methynaltrexone 30mg is Sodium Chloride 4mg Citric acid 0.0~75mg Trisodium Citrate 0.0496mg Disodium edetate 0.75mg Water for injectionq.s. to lgram 20 . _.

The pH of this solution is 3.5 and can withstand autoclaving process.
Example 5 2s Manufacturing Process for a Lyophilized Pharmaceutical For mutation of Methylnaltrexone The lyophilization cycle listed below is standard procude well known to one of ordinary skill in the art. This cycle was used for the preparation of lyophilized 3o preparation of methylnaltrexone analyzed in Examples 6 and 7.
1. Load chamber at room temperature (20-25C) _ 28 _ 2. Lower shelf temp to -45 degrees C at 1.0 degrees C/min 3. Hold shelf temp at -45 for 120 minutes 4. When condenser is below -50 degrees C, evacuate the chamber to 100-125 mt.
5. Ramp shelf to -20 degrees C at 0.5 degrees C/min.
6. Hold at -20 degrees C for 16 hours 7. Ramp shelf to +27 degrees C at 0.10 degrees C/min.
8. Hold for a minimum of 8 hours. Maintain chamber pressure at 100-125mt for the entire cycle.
9. Restore chamber to 11.0 PSIA + or- 1.0 with sterile filtered Nitrogen and then to seat the closures (2" Hg), then bleed to atmospheric pressure with Nitrogen to unload.
Example 6 1s Stability of Lyophilized Formulations of Methylnaltrexona The following data repouts the stability of lyophilized formulations of methylnaltrexone using different cryoprotecting agents.
~~.yoprotscting pH total degradation Agent products Mannitol 5.0 0.34 %

Polyvinyl pyrrolidone4.1 0.37%

Polyethylene 5.7 0.44%
glycol Histidine 7.4 0.55%

Example 7 Stability of Lyophilized Formulations of Methylnaltrexone The following data reports the stability of lyophilized formulations of 2s methylnaltrexone in comparison to buffered formulations.

Amount of total related substances at various stages of manufacturing 1 ~ 2 3 4 5 6 Key Monothio-CitrateCitrateAcetateLyophilizedLyophilized IngredientglycerolBuffer Buffer Buffer using using pH 3.5 pH 5 pH 3.6 Mannitol Lactose Unautoclaved0.13 0.12 0.16 0.20 0.14 0.12 Autoclaved0.91 0.23 0.61 1.39 n/a n/a Stability 1.10 0.16 0.48 1.26 0.15 0.15 (2 mths at room temp) It should be understood that various changes and modifications to the preferred embodiments described herein will be apparent to those of ordinary skill in the art. Such changes and modifications can be made without departing from the spirit and scope of this invention without diminishing its advantages. It is therefore intended that such changes and modifications, including equivalents, be covered by the appended claims.
All of the patents, patent applications and references listed herein are incorporated by reference in their entirety.
VVe claim:

Claims (315)

1. A pharmaceutical preparation comprising a solution of methylnaltrexone or a salt thereof, wherein the preparation after autoclaving has a concentration of methylnaltrexone degradation products that does not exceed 2% of the methylnaltrexone or salt thereof in the preparation.
2. The pharmaceutical preparation of claim 1, wherein the concentration of methylnaltrexone degradation products does not exceed 1.5% of the methylnaltrexone or salt thereof in the preparation.
3. The pharmaceutical preparation of claim 2, wherein the concentration of methylnaltrexone degradation products does not exceed 1.0% of the methylnaltrexone or salt thereof in the preparation.
4. The pharmaceutical preparation of claim 3, wherein the concentration of methylnaltrexone degradation products does not exceed 0.5% of the methylnaltrexone or salt thereof in the preparation.
5. The pharmaceutical preparation of claim 4, wherein the concentration of methylnaltrexone degradation products does not exceed 0.25% of the methylnaltrexone or salt thereof in the preparation.
6. The pharmaceutical preparation of claim 5, wherein the concentration of methylnaltrexone degradation products does not exceed 0.125% of the methylnaltrexone or salt thereof in the preparation.
7. The pharmaceutical preparation of claim 1, wherein the pharmaceutical preparation further comprises a chelating agent.
8. The pharmaceutical preparation of claim 7, wherein the chelating agent is ethylenediaminetetraacetic acid (EDTA) or a derivative thereof.
9. The pharmaceutical preparation of claim 8, wherein the derivative is disodium edetate.
10. The pharmaceutical preparation of claim 8, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.001 to 100.0 mg/ml.
11. The pharmaceutical preparation of claim 10, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.05 to 25.0 mg/ml.
12. The pharmaceutical preparation of claim 11, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.1 to 2.5 mg/ml.
13. The pharmaceutical preparation of claim 7, further comprising a buffering agent.
14. The pharmaceutical preparation of claim 13, wherein the buffering agent is citrate buffer.
15. The pharmaceutical preparation of claim 10, further comprising citrate in a concentration ranging from 0.0010 to 100.0 mM.
16. The pharmaceutical preparation of claim 10, further comprising citrate in a concentration ranging from 0.10 to 50 mM.
17. The pharmaceutical preparation of claim 1, further comprising a buffering agent.
18. The pharmaceutical preparation of claim 17, wherein the buffering agent is citrate buffer.
19. The pharmaceutical preparation of claim 18, wherein the citrate is present in a concentration ranging from 0.01 to 100.0 mM.
20. The pharmaceutical preparation of claim 19, wherein the citrate is present in a concentration ranging from 0.10 to 10.0 mM.
21. The pharmaceutical preparation of claim 20, wherein the citrate is present in a concentration ranging from 0.10 to 5.0 mM.
22. The pharmaceutical preparation of any one of claims 1 to 21, wherein the pH of the preparation does not exceed 4.25.
23. The pharmaceutical preparation of claim 22, wherein the pH ranges from 2.0 to 4Ø
24. The pharmaceutical preparation of claim 23, wherein the pH ranges from 3.0 to 4Ø
25. The pharmaceutical preparation of claim 24, wherein the pH ranges from 3.0 to 3.5.
26. The pharmaceutical preparation of any one claims 1 to 21 wherein, the concentration of methylnaltrexone ranges from 0.01 to 100 mg/ml.
27. The pharmaceutical preparation of claim 26 wherein, the concentration of methylnaltrexone ranges from 0.1 to 100.0 mg/ml.
2i1. The pharmaceutical preparation of claim 27 wherein, the concentration of methylnaltrexone ranges from 1.0 to 50.0 mg/ml.
29. The pharmaceutical preparation of claim 26 wherein, the pH of the pharmaceutical preparation does not exceed 4.25.
30. The pharmaceutical preparation of claim 29 wherein, the pH ranges from 2.0 to 4Ø
31. The pharmaceutical preparation of claim 29 wherein, the pH ranges from 3.0 to 4Ø
32. The pharmaceutical preparation of claim 29 wherein, the pH ranges from 3.0 to 3.5.
33. The pharmaceutical preparation of any one of claims 1 to 21, further comprising an anti-oxidant.
34. The pharmaceutical preparation of any one of claims 1 to 21, further comprising an isotonicity agent.
35. The pharmaceutical preparation of any one of claims 1 to 21, further comprising an opioid.
36. The pharmaceutical preparation of any one of claims 1 to 21, further comprising a cryoprotective agent.
37. The pharmaceutical preparation of claim 369 wherein the cryoprotective agent is ~.
polyol.
38. The pharmaceutical preparation of claim 1 to 21, wherein the solution is provided in a vial or ampoule with a septum.
39. The pharmaceutical preparation of claim 1 to 21, wherein the solution is provided in a syringe, infusion bag or sealable bottle.
40. The pharmaceutical preparation of claim 22, wherein the solution is provided in a vial or ampoule with a septum.
41. The pharmaceutical preparation of claim 22, wherein the solution is provided in a syringe, infusion bag or sealable bottle.
42. The pharmaceutical preparation of claim 26, wherein the solution is provided in a vial or ampoule with a septum.
43. The pharmaceutical preparation of claim 26, wherein the solution is provided in a syringe, infusion bag, or sealable bottle.
44. The pharmaceutical preparation of claim 1 to 21, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
45. The pharmaceutical preparation of claim 22, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
46 The pharmaceutical preparation of claim 35, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
47. The pharmaceutical preparation of claim 22, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
48. The pharmaceutical preparation of claim 25, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
49. The pharmaceutical preparation of claim 26, wherein the solution is provided in a container including indicia indicating that the pharmaceutical preparation has been autoclaved.
50. ~A method for preparing an autoclaved pharmaceutical preparation that has a concentration of methylnaltrexone degradation products that does not exceed 2%
of the methylnaltrexone or salt thereof in the preparation comprising:
providing a solution having a pH of 4.25 or less comprising methylnaltrexone or salt thereof and being substantially free of methylnaltrexone degradation products; and autoclaving the solution.
51. ~The method of claim 50, wherein the pH ranges from 2.0 to 4Ø
52. ~The method of claim 51, wherein the pH ranges from 3.0 to 4Ø
53. ~The method of claim 51, wherein the pH ranges from 3.0 to 3.5
54. ~The method of claim 50, 51, 52 or 53, wherein the solution contains a chelating agent.
55. ~The method of claim 54, wherein the solution further comprises an isotonicity agent.
56. ~The method of claim 50, S l, 52 or 53, wherein the solution contains a buffering agent.
57. ~The method of claim 56, wherein the solution contains a chelating agent.
58. ~The method of claim 50, 51, 52 or 53, wherein the solution contains an anti-oxidant.
59. ~The method of claim 58, wherein the solution contains a chelating agent.
60. ~The method of claim 58, wherein the solution contains a buffering agent.
61. ~The method of claim 54, wherein the chelating agent is EDTA or derivative thereof.
62. ~The method of claim 56, wherein the buffering agent is citrate buffer.
63. ~The method of claim 50, 51, 52 or 53, further comprising lyophilizing the solution.
64. ~The method of claim 63, further comprising adding a cryoprotecting agent to the solution.
65. ~The method of claim 63, wherein the cryoprotective agent is a polyol.
66. ~A method for preparing an autoclaved pharmaceutical preparation that has a concentration of methylnaltrexone degradation products that does not exceed 2%
of the methylnaltrexone or salt thereof in the preparation comprising:
providing a solution comprising methylnaltrexone or salt thereof and a chelating agent, the solution being substantially free of methylnaltrexone degradation products;
and autoclaving the solution.
67. ~The method of claim 66, wherein the chelating agent is EDTA or derivative thereof.
68. ~The method of claim 67, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.001 to 100.0 mg/ml.
69. ~The method of claim 68, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.05 to 25.0 mg/ml.
70. ~The method of claim 68, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.1 to 2.5 mg/ml.
71. ~The method of claim 66, 67, 68, 69 or 70, wherein the solution contains a buffering agent.
72. ~The method of claim 71, wherein the buffering agent is citrate buffer.
73. ~The method of claim 66, wherein the solution is adjusted to have a pH of 4.25 or less.
74. ~The method of claim 71, wherein the solution is adjusted to have a pH of 4.25 or less.
75. ~The method of claim 66, wherein the solution is adjusted to have a pH
ranging from 3.0 to 3.5.
76. ~The method of claim 71, wherein the solution is adjusted to have a pH
ranging from 3.0 to 3.5.
77. ~The method of claim 66, wherein the solution contains an anti-oxidant.
78. ~The method of claim 66, wherein the solution contains an isotonicity agent.
79. ~The method of claim 66, 67, 68, 69 or 70, wherein the degradation products after autoclaving do not exceed 1.0 %.
80. ~The method of claim 71, wherein the degradation products after autoclaving do not exceed 1.0%.
81. ~The method of claim 66, 67, 68, 69, or 70, wherein the degradation products after autoclaving do not exceed 0.5%.
82. ~The method of claim 71, wherein the degradation products after autoclaving do not exceed 0.5%.
83. ~The method of claim 66, further comprising lyophilizing the solution.
84. ~The method of claim 83, further comprising adding a cryoprotecting agent to the solution.
85. ~The method of claim 84, wherein the cryoprotective agent is a polyol.
86. ~A pharmaceutical preparation comprising a solution of methylnaltrexone or a salt thereof, wherein the preparation after storage at about room temperature for six months has a concentration of methylnaltrexone degradation products that does not exceed 2% of the methylnaltrexone in the preparation.
87. ~The pharmaceutical preparation of claim 86, wherein the concentration of methylnaltrexone degradation products does not exceed 1.5% of the methylnaltrexone in the preparation.
88. ~The pharmaceutical preparation of claim 87, wherein the concentration of methylnaltrexone degradation products does not exceed 1.0% of the methylnaltrexone in the preparation.
89. ~The pharmaceutical preparation of claim 88, wherein the concentration of methylnaltrexone degradation products does not exceed 0.5% of the methylnaltrexone in the preparation.
90. ~The pharmaceutical preparation of claim 89, wherein the concentration of methylnaltrexone degradation products does not exceed 0.25% of the methylnaltrexone in the preparation.
91. ~The pharmaceutical preparation of claim 90, wherein the concentration of methylnaltrexone degradation products does not exceed 0.125% of the methylnaltrexone in the preparation.
92. ~The pharmaceutical preparation of claim 88, wherein the pharmaceutical preparation further comprises a chelating agent.
93. ~The pharmaceutical preparation of claim 92, wherein the chelating agent is EDTA or derivative thereof.
94. ~The pharmaceutical preparation of claim 93, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.001 to 100.0 mg/ml.
95. ~The pharmaceutical preparation of claim 94, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.05 to 25.0 mg/ml.
96. ~The pharmaceutical preparation of claim 95, wherein the EDTA or derivative thereof is present in a concentration ranging from 0.1 to 2.5 mg/ml.
97. ~The pharmaceutical preparation of claim 92, further comprising a buffering agent.
98. ~The pharmaceutical preparation of claim 97, wherein the buffering agent is citrate buffer.
99. ~The pharmaceutical preparation of claim 94, further comprising citrate in a concentration ranging from 0.0010 to 100.0 mM.
100. The pharmaceutical preparation of claim 94, further comprising citrate in a concentration ranging from 0.10 to 50 mM.
101. The pharmaceutical preparation of claim 86, wherein the pharmaceutical preparation further comprises a buffering agent.
102. The pharmaceutical preparation of claim 86, wherein the buffering agent is citrate buffer.
103. The pharmaceutical preparation of claim 102, wherein the citrate is present in a concentration ranging from 0.01 to 100.0 mM.
104. The pharmaceutical preparation of claim 103, wherein the citrate is present in a concentration ranging from 0.10 to 10.0 mM.
105. The pharmaceutical preparation of claim 104, wherein the citrate is present in a concentration ranging from 0.10 to 5.0 mM.
106. The pharmaceutical preparation of any one of claims 86 to 105, wherein the pH
does not exceed 4.25.
107. The pharmaceutical preparation of claim 106, wherein the pH ranges from 2.0 to 4Ø
108. The pharmaceutical preparation of claim 107, wherein the pH ranges from 3.0 to 4Ø
109. The pharmaceutical preparation of claim 108, wherein the pH ranges from 3.0 to 3.5.
110. The pharmaceutical preparation of any one claims 86 to 105, wherein the concentration of methylnaltrexone ranges from 0.01 to 100 mg/ml.
111. The pharmaceutical preparation of claim 110, wherein the concentration of methylnaltrexone ranges from 0.1 to 100.0 mg/ml.

-41-~
112. The pharmaceutical preparation of claim 111, wherein the concentration of methylnaltrexone ranges from 1.0 to 50.0 mg/ml.
113. The pharmaceutical preparation of claim 111, wherein the pH does not exceed 4.25.
114. The pharmaceutical preparation of claim 113, wherein the pH ranges from 2.0 to 4Ø
115. The pharmaceutical preparation of claim 113, wherein the pH ranges from 3.0 to 4Ø
116. The pharmaceutical preparation of claim 113, wherein the pH ranges from 3.0 to 3.5.
117. The pharmaceutical preparation of any one of claims 86 to 105, further comprising an anti-oxidant.
118. The pharmaceutical preparation of any one of claims 86 to 105, further comprising an isotonicity agent.
119. The pharmaceutical preparation of any of claims 86 to 105, further comprising a cryoprotective agent.
120. The pharmaceutical preparation of claim 119, wherein the cryoprotective agent is a polyol.
121. The pharmaceutical preparation of any one of claims 86 to 105, further comprising an opioid.
122. The pharmaceutical preparation of claim 97, further comprising an isotonicity agent, wherein the pH does not exceed 4.25.
123. The pharmaceutical preparation of claim 121, wherein the pH is between 3.0 and 3.5.
124. The pharmaceutical preparation of claim 122, wherein the buffering agent is a citrate buffer and chelating agent is EDTA or a derivative thereof.
125. The pharmaceutical preparation of claim 124, wherein the citrate is present in a range between 0.001 and 100 mM and the chelating agent is present in a range between 0:001 and 100.0 mg/mL.
126. The pharmaceutical preparation of claim 122, 123, 124 or 125, further comprising an isotonicity agent.
127. The pharmaceutical preparation of claim 122, 123, 124 or 125, further comprising an antioxidant.
128. The pharmaceutical preparation of claim 127, further comprising an isotonicity agent.
129. The pharmaceutical preparation of claim 86, wherein the solution is provided in a vial or ampoule with a septum, in a syringe, an infusion bag, or a sealable bottle.
130. The pharmaceutical preparation of claim 106, wherein the solution is provided in a vial or ampoule with a septum, in a syringe, an infusion bag, or a sealable bottle.
131. The pharmaceutical preparation of claim 122, wherein the solution is provided in a vial or ampoule with a septum.
132. The pharmaceutical preparation of claim 122, wherein the solution is provided in a syringe, an infusion bag, or a sealable bottle.
133. The pharmaceutical preparation of claim 86, wherein the solution is provided in a container including indicia indicating that the solution has been autoclaved.
134. The pharmaceutical preparation of claim 106, wherein the solution is provided in a container including indicia indicating that the solution has been autoclaved.
135. The pharmaceutical preparation of claim 124, wherein the solution is provided in a container including indicia indicating that the solution has been autoclaved.
136. A stable pharmaceutical preparation comprising a solution of methylnaltrexone or salt thereof, wherein the pH is below 4.25.
137. The pharmaceutical preparation of claim 136, wherein the pH is between 2.75 and 4.25.
138. The pharmaceutical preparation of claim 136, wherein the pH is between 3.0 and 4Ø
139. The pharmaceutical preparation of claim 136, wherein the pH is between 3.0 and 3.5.
140. The pharmaceutical preparation of claim 136, 137, 138, or 139, wherein the pH is adjusted with an acid selected from the group consisting of HCl, citric acid, sulfuric acid, acetic acid, or phosphoric acid.
141. The pharmaceutical preparation of claim 136, 137, 138, or 139, wherein the preparation further comprises a buffering agent.
142. The pharmaceutical preparation of claim 141, wherein the buffering agent is selected from the group consisting of citric acid, sodium citrate, sodium acetate, acetic acid, sodium phosphate and phosphoric acid, sodium ascorbate, tartartic acid, maleic acid, glycine, sodium lactate, lactic acid, ascorbic acid, imidazole, sodium bicarbonate -44-~

and carbonic acid, sodium succinate and succinic acid, histidine, and sodium benzoate and benzoic acid.
143. The pharmaceutical preparation of claim 141, wherein the buffering agent is a citrate buffer.
144. The pharmaceutical preparation of claim 143, wherein the citrate buffer concentration ranges from 0.001 mM to 100 mM.
145. The pharmaceutical preparation of claim 143, wherein the citrate buffer concentration ranges from 0.01 mM to 50 mM.
146. The pharmaceutical preparation of claim 143, wherein the citrate buffer concentration ranges from 0.1 mM to 25 mM.
147. The pharmaceutical preparation of claim 136, further comprising a chelating agent.
148. The pharmaceutical preparation of claim 141, further comprising a chelating agent.
149. The pharmaceutical preparation of claim 148, wherein the chelating agent is selected from the group consisting of EDTA and derivatives thereof, citric acid and derivatives thereof, niacinamidc and derivatives sodium desoaycholate and derivatives thereof.
150. The pharmaceutical preparation of claim 149, wherein the chelating agent is EDTA or derivative thereof.
151. The pharmaceutical preparation of claim 150, wherein the EDTA or derivative thereof concentration ranges from 0.001 to 100 mg/ml.
152. The pharmaceutical preparation of claim 151, wherein the EDTA or derivative thereof concentration ranges from 0.05 to 25.0 mg/ml.
153. The pharmaceutical preparation of claim 151, wherein the EDTA or derivative thereof concentration ranges from 0.1 to 2.5 mg/ml.
154. The pharmaceutical preparation of claim 151, wherein the EDTA or derivative thereof concentration ranges from 0.5 to 0.75 mg/ml.
155. The pharmaceutical preparation of claim 136 or 147, wherein the preparation is substantially free of methylnaltrexone degradation products.
156. The pharmaceutical preparation of claim 141, wherein the preparation is substantially free of methylnaltrexone degradation products.
157. The pharmaceutical preparation of claim 148, wherein the preparation is substantially free of methylnaltrexone degradation products.
158. The pharmaceutical preparation of claim 148, wherein the pharmaceutical preparation has been autoclaved and the concentration of methylnaltrexone degradation products is less than 2.0% of the methylnaltrexone in the preparation.
159. The preparation of claim 158, wherein the concentration of methylnaltrexone degradation products is less than 1.0% of the methylnaltrexone in the preparation.
160. The preparation according to claim 158, wherein the concentration of methylnaltrexone degradation products is less than 0.5% of the methylnaltrexone in the preparation.
161. The preparation according to claim 158, wherein the concentration of methylnaltrexone degradation products is less than 0.25% of the methylnaltrexone in the preparation.
162. The preparation according to claim 158, wherein the concentration of methylnaltrexone degradation products is less than 0.125% of the methylnaltrexone in the preparation.
163. The pharmaceutical preparation of claim 136 or 147, wherein the methylnaltrexone or salt thereof is present in an amount effective to treat a side effect associated with opioid treatment when administered to a human subject.
164. The pharmaceutical preparation of claim 163, wherein the concentration of methylnaltrexone or salt thereof is sufficient to treat constipation.
165. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.01 to 100 mg/ml.
166. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.05 to 100 mg/ml.
167. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.1 to 100 mg/ml.
168. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof is about 50 mg/ml.
169. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof is about 10.0 mg/ml.
170. The pharmaceutical preparation of claim 136 or 147, wherein the concentration of methylnaltrexone or salt thereof is about 0.1 mg/ml.
171. The pharmaceutical composition of claim 136 or 147, further comprising an isotonicity agent.
172. The pharmaceutical composition of claim 141, further comprising an isotonicity agent.
173. The pharmaceutical composition of claim 148, further comprising an isotonicity agent.
174. The composition of claim 171, wherein the isotonicity agent is selected from the group consisting of sodium chloride, mannitol, lactose, dextrose, sorbitol, and glycerol.
175. The preparation of claim 174, wherein the isotonicity agent is sodium chloride.
176. The preparation of claim 136 or 147, further comprising an antioxidant.
177. The preparation of claim 141, further comprising an antioxidant.
178. The preparation of claim 148, further comprising an antioxidant.
179. The preparation of claim 176, wherein the antioxidant is selected from the group consisting of an ascorbic acid derivative, butylated hydroxy anisole, butylated hydroxy toluene, alkyl gallate, sodium meta-bisulfate, sodium bisulfate, sodium dithionite, sodium thioglycollic acid, sodium formaldehyde sulfoxylate, tocopherol and derivatives thereof, monothioglycerol, and sodium sulfite.
180. The preparation of claim 136 or 147, further comprising a cryoprotective agent.
181. The preparation of claim 141, further comprising a cryoprotective agent.
182. The preparation of claim 148, further comprising a cryoprotective agent.
183. The preparation of claim 180 wherein the cryoprotective agent is a polyol.
184. The preparation of claim 136 or 147, further comprising an opioid.
185. The preparation of claim 141, further comprising an opioid.
186. The preparation of claim 148, further comprising an opioid.
187. The preparation of claim 184, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
188. A stable pharmaceutical preparation comprising a solution of methylnaltrexone or salt thereof, wherein the solution further comprises a chelating agent in an amount sufficient to inhibit degradation of the methylnaltrexone or salt thereof, whereby the amount is such that the preparation after autoclaving has a concentration of mcthylnaltrexone degradation products that does not emceed 0.5% of the methylnaltrexone or salt thereof in the preparation.
189. The pharmaceutical preparation of claim 188, wherein the chelating agent is selected from the group consisting of EDTA and derivatives thereof, citric acid and derivatives thereof, niacinamide and derivatives thereof, and sodium desoxycholate and derivatives thereof.
190. The pharmaceutical preparation of claim 189, wherein the chelating agent is EDTA or derivative thereof.
191. The pharmaceutical preparation of claim 190, wherein the EDTA or derivative thereof concentration ranges from 0.4 to 100 mg/ml.
192. The pharmaceutical preparation of claim 191, wherein the EDTA or derivative concentration ranges from 0.5 to 25.0 mg/ml.
193. The pharmaceutical preparation of claim 191, wherein the EDTA or derivative concentration ranges from 0.5 to 10.0 mg/ml.
194. The pharmaceutical preparation of claim 191, wherein the EDTA or derivative concentration ranges from 0.5 to 2.5 mg/ml.
195. The pharmaceutical preparation of claim 188, 189, 190, 191 or 192, wherein the preparation further comprises a buffering agent.
196. The pharmaceutical preparation of claim 195, wherein the buffering agent is selected from the group consisting of citric acid, sodium citrate, sodium acetate, acetic acid, sodium phosphate and phosphoric acid, sodium ascorbate, tartartic acid, maleic acid, glycine, sodium lactate, lactic acid, ascorbic acid, imidazole, sodium bicarbonate and carbonic acid, sodium succinate and succinic acid, histidine, and sodium benzoate and benzoic acid.
197. The pharmaceutical preparation of claim 195, wherein the buffering agent is a citrate buffer.
198. The pharmaceutical preparation of claim 197, wherein the citrate buffer concentration ranges from 0.001 mM to 100 mM.
199. The pharmaceutical preparation of claim 197, wherein the citrate buffer concentration ranges from 0.01 mM to 50 mM.
200. The pharmaceutical preparation of claim 197, wherein the citrate buffer concentration ranges from 0.1 mM to 25 mM.
201. The pharmaceutical preparation of claim 197, wherein the citrate buffer concentration ranges from 0.25 mM to 15 mM.
202. The pharmaceutical preparation of claim 188, wherein the preparation is substantially free of methylnaltrexone degradation products.
203. The pharmaceutical preparation of claim 195, wherein the preparation is substantially free of methylnaltrexone degradation products.
204. The pharmaceutical preparation of claim 197, wherein the preparation is substantially free of methylnaltrexone degradation products.
205. The preparation according to claim 188, wherein the concentration of methylnaltrexone degradation products is less than 0.25% of the methylnaltrexone in the preparation.
206. The pharmaceutical preparation of claim 188, wherein the methylnaltrexone or salt thereof is present in an amount effective to treat a side effect associated with opioid treatment when administered to a human subject.
207. The pharmaceutical preparation of claim 206, wherein the concentration of methylnaltrexone or salt thereof is sufficient to treat constipation.
208. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.01 to 100 mg/ml.
209. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.05 to 100 mg/ml.
210. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof ranges from 0.1 to 100 mg/ml.
211. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof is in a range of 25 to 75 mg/ml.
212. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof is in a range of 1 to 20 mg/ml.
213. The pharmaceutical preparation of claim 188, wherein the concentration of methylnaltrexone or salt thereof is in a range of 0.05 to 0.5 mg/ml.
214. The pharmaceutical composition of claim 188, further comprising an isotonicity agent.
215. The pharmaceutical composition of claim 195, further comprising an isotonicity agent.
216. The composition of claim 214, wherein the isotonicity agent is selected from the group consisting of sodium chloride, mannitol, lactose, dextrose, glycerol, and sorbitol.
217. The composition of claim 215, wherein the isotonicity agent is selected from the group consisting of sodium chloride, mannitol, lactose, dextrose, glycerol, and sorbitol.
218. The preparation of claim 215, wherein the isotonicity agent is sodium chloride.
219. The preparation of claim 188, further comprising an antioxidant.
220. The preparation of claim 195, further comprising an antioxidant.
221. The preparation of claim 219, wherein the antioxidant is selected from the group consisting of an ascorbic acid derivative, butylated hydroxy anisole, butylated hydroxy toluene, alkyl gallate, sodium meta-bisulfate, sodium bisulfate, sodium dithionite, sodium thioglycollic acid, sodium formaldehyde sulfoxylate, tocopherol and derivatives thereof, monothioglycerol, and sodium sulfite.
222. The preparation of claim 220, wherein the antioxidant is selected from the group consisting of an ascorbic acid derivative, butylated hydroxy anisole, butylated hydroxy toluene, alkyl gallate, sodium meta-bisulfite, sodium dithionite, sodium thioglycollic acid, sodium formaldehyde sulfoxylate, tocopherol and derivatives thereof, monothioglycerol, sodium bisulfite, and sodium sulfite.
223. The preparation of any of claims 188, 195 or 219, further comprising a cryoprotective agent.
224. The preparation of claim 213 wherein the cryoprotective agent is a polymerized carbohydrate.
225. The preparation of claim 188, further comprising an opioid.
226. The preparation of claim 195, further comprising an opioid.
227. The preparation of claim 219, further comprising an opioid.
228. The preparation of claim 225, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
229. The preparation of claim 226, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
230. The preparation of claim 227, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
231. A pharmaceutical preparation comprising a solution of methylnaltrexone or salt thereof and at least one methylnaltrexone degradation inhibiting agent selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations thereof, wherein the solution has a pH ranging from 2 to 6, wherein the degradation inhibiting agent is present in an amount sufficient to render the preparation sable, wherein the preparation is processed under at least one sterilization technique and wherein the preparation is substantially free of methylnaltrexone degradation products.
232. The pharmaceutical preparation of claim 231, wherein the preparation is stable to storage for 6 months at about room temperature.
233. The pharmaceutical preparation of claim 232, wherein the preparation is stable to storage for 12 months at about room temperature.
234. The pharmaceutical preparation of claim 233, wherein the preparation is stable to storage for 24 months at about room temperature.
235. The pharmaceutical preparation of claim 231, wherein the preparation is stable to autoclaving.
236. The pharmaceutical preparation of claim 231, further comprising an isotonicity agent.
237. The preparation of claim 231, further comprising a cryoprotective agent.
238. The pharmaceutical preparation of claim 231, further comprising an opioid.
239. The pharmaceutical preparation of claim 237, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
240. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is provided in a vial or an ampoule with a septum.
241. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is provided in an infusion bag.
242. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is provided in a syringe.
243. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is provided in a sealable bottle.
244. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is suitable for parenteral administration.
245. The pharmaceutical preparation of claim 136, 188 or 231, wherein the preparation is suitable for oral imbibing.
246. The pharmaceutical preparation of claim 136, 188 or 231, wherein the solution is provided in a container including indicia indicating the preparation has been processed under at least one sterilization technique.
247. A method of inhibiting formation of methylnaltrexone degradation products in a pharmaceutical preparation comprising methylnaltrexone or salts thereof, the method comprising:
preparing an aqueous solution comprising at least one methylnaltrexone degradation inhibiting agent selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations thereof;
dissolving a powdered source of methylnaltrexone or salt thereof with the solution to form the pharmaceutical preparation.
248e The method of claim 247, wherein the methylnaltrexone degradation inhibiting agent is a chelating agent.
249. The method of claim 247, wherein the methylnaltrexone degradation inhibiting agent is a buffering agent.
250. The method of claim 247, wherein the methylnaltrexone degradation inhibiting agent is an antioxidant.
251. The method of claim 247, wherein the methylnaltrexone degradation inhibiting agent comprises a chelating agent and a buffering agent.
252. The method of claim 247, further comprising adjusting with an acid the pH
of the solution or the preparation to a pH ranging from 2 to 6.
253. The method of claim 247, further comprising adjusting with an acid the pH
of the solution or the preparation to a pH ranging from about 3 to 5.
254. The method of claim 247, further comprising adjusting with an acid the pH
of the solution or the preparation to a pH ranging from about 3 to 4.
255. The method of claim 247, further comprising adding an isotonicity agent to the solution.
256. A method of preparing a stable pharmaceutical preparation comprising an aqueous solution of methylnaltrexone or salts thereof to inhibit formation of methylnaltrexone degradation products, comprising:
providing a solution comprising methylnaltrexone or salts thereof and at least one methylnaltrexone degradation inhibiting agent;
processing the solution under at least one sterilization technique prior to and/or after terminal filling the solution in a sealable container to form the stable pharmaceutical preparation, wherein the method is carried out without the addition of a pH-adjusting-base to the solution.
257. The method according to claim 256, wherein the concentration of methylnaltrexone degradation products is less than 2.0% of the total methylnaltrexone in the preparation.
258. The method according to claim 256, wherein the concentration of methylnaltrexone degradation products is less than 1.0 % of the total methylnaltrexone in the preparation.
259. The method according to claim 256, wherein the concentration of methylnaltrexone degradation products is less than 0.5% of the total methylnaltrexone in the preparation.
260. The method according to claim 256, wherein the concentration of methylnaltrexone degradation products is less than 0.25% of the total methylnaltrexone in the preparation.
261. The method of claim 256, wherein the methylnaltrexone degradation inhibiting agent is selected from the group consisting of a chelating agent, a buffering agent, an antioxidant, and combinations thereof.
262. The method of claim 256, wherein the methylnaltrexone degradation inhibiting agent is a chelating agent.
263. The method of claim 256, wherein the methylnaltrexone degradation inhibiting agent is a buffering agent.
264. The method of claim 256, wherein the buffering agent is citrate buffer.
265. The method of claim 256, wherein the methylnaltrexone degradation inhibiting agent is an antioxidant.
266. The method of claim 256, wherein the methylnaltrexone degradation inhibiting agent comprises a chelating agent and a buffering agent.
267. The method of claim 256, 261, 262, 263, 264, 265 or 266, wherein the initial solution is adjusted to a pH ranging from 2 to 6 prior to the processing under the at least one sterilization technique.
268. The method of claim 267, wherein the initial solution is adjusted to a pH
ranging from 2 to 5.
269. The method of claim 268, wherein the initial solution is adjusted to a pH
ranging from 3 to 5.
270. The method of claim 269, wherein the initial solution is adjusted to a pH
ranging from 3 to 4.
271. The method of claim 256, wherein the aseptic technique is autoclaving after terminal filling the sealable container.
272. The method of claim 256, wherein the processing comprises sterile filtration prior to terminal filling followed by autoclaving after terminal filling the sealable container.
273. The method of claim 256, further comprising sealing the container, wherein the container is purged with nitrogen.
274. The method of claim 256, further comprising sealing the container, wherein the container is sparged to eliminate oxygen.
275. The method of claim 256, wherein the initial solution further comprises an isotonicity agent.
276. The method of claim 275, wherein the isotonicity agent is sodium chloride.
277. The method of claim 256, wherein the initial solution further comprising a cryoprotective agent.
278. The method of claim 277 wherein the cryoprotective agent is a polyol.
279. The method of claim 256, further comprising adding at least one opioid to the initial solution.
280. The method of claim 279, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
281. The method of claim 279, wherein the opioid is solubilized in a nonaqueous solvent prior to addition to the initial solution.
282. The method of claim 281, wherein the nonaqueous solvent is an oil, wax, or alcohol.
283. A product comprising a stable lyophilized formulation of methylnaltrexone, wherein the formulation upon reconstitution in water at a concentration of 20 mg/ml has a pH of between 2 and 6.
284. The product of claim 283, wherein the formulation upon reconstitution in water has a pH of between 3 and 5.
285. The product of claim 283, wherein the formulation comprises a cryoprotecting agent present in an amount sufficient to render the formulation stable.
286. The product of claim 284, wherein the formulation comprises a cryoprotecting agent present in an amount to render the formulation stable.
287. The product of claim 285, wherein the cryoprotecting agent is a polyol.
288. The product of claim 286, wherein the cryoprotecting agent is a polyol.
289. The product of claim 285, wherein the cryoprotecting agent is mannitol.
290. The product of claim 286, wherein the cryoprotecting agent is mannitol.
291. The product of claims 283-390, further comprising any one or more of a buffering agent, a chelating agent and an antioxidant.
292. The product of claims 283-290, further comprising citrate buffer.
293. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of claims 1-21.
294. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of claim 36.
295. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of claim 37.
296. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of claims 86-105.
297. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of 119.
298. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of 120.
299. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of 136-139.
300. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of 180.
301. A product comprising a lyophilized formulation of methylnaltrexone prepared from a solution comprising the solution of 181.
302. A product comprising methylnaltrexone and a degradation inhibiting agent selected from the group consisting of a chelating agent, a buffering agent, an anti-oxidant, and combinations thereof wherein the degradation inhibiting agent is present in an amount sufficient to render stable a solution of the product containing a concentration of 20 mg/ml methylnaltrexone.
303. The product of claim 302, wherein the product when in solution at a concentration of 20 mg/ml methylnaltrexone yields a solution with a pH of between 2 and 6.
304. The product of claim 303, wherein the product has less than 1%
methylnaltrexone degradation products when stored at room temperature in the solution for 6 months.
305. The product of claim 303, wherein the product has less than 1%
methylnaltrexone degradation products when stored at room temperature in the solution for 12 months.
306. The product of claim 303, wherein the product has less than 1%
methylnaltrexone degradation products when stored at room temperature in the solution for 24 months.
307. A pharmaceutical preparation comprising methylnaltrexone;
sodium chloride, citric acid, trisodium citrate, and disodium edetate.
308. The pharmaceutical preparation of claim 307, wherein the preparation is a solution and the methylnaltrexone is present at between 20 and 40 mg/ml, the sodium chloride is present between 2 and 6 mg/ml, the citric acid is present between 0.05 and 0.1 mg/ml, the trisodium citrate is present between 0.025 and 0.075 mg/ml and the disodium edetate is present between 0.5 and 1.0 mg/ml.
309. A kit comprising a package containing a sealed container comprising the pharmaceutical preparation of claim 136, 188, 231, or 283, and instructions for use.
310. The kit of claim 309, further comprising a diluant container containing a pharmaceutically acceptable diluant.
311. The kit of claim 310, further comprising instructions for mixing the preparation and diluant.
312. The kit of claim 310, wherein the diluant is selected from the group consisting of a 5% dextrose solution and a physiological saline solution.
313. The kit of claim 310, wherein the diluant is contained in a sealable bottle or an infusion bag.
314. The kit of claim 309, further comprising an opioid container containing an opioid.
315. The kit of claim 314, wherein the opioid is selected from the group consisting of alfentanil, anileridine, asimadoline, bremazocine, burprenorphine, butorphanol, codeine, dezocine, diacetylmorphine (heroin), dihydrocodeine, diphenoxylate, fedotozine, fentanyl, funaltrexamine, hydrocodone, hydromorphone, levallorphan, levomethadyl acetate, levorphanol, loperamide, meperidine (pethidine), methadone, morphine, morphine-6-glucoronide, nalbuphine, nalorphine, opium, oxycodone, oxymorphone, pentazocine, propiram, propoxyphene, remifentanyl, sufentanil, tilidine, trimebutine, and tramadol.
CA2521379A 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone Expired - Lifetime CA2521379C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2811272A CA2811272C (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US46161103P 2003-04-08 2003-04-08
US60/461,611 2003-04-08
PCT/US2004/010997 WO2004091623A1 (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA2811272A Division CA2811272C (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone

Publications (2)

Publication Number Publication Date
CA2521379A1 true CA2521379A1 (en) 2004-10-28
CA2521379C CA2521379C (en) 2013-07-02

Family

ID=33299842

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2521379A Expired - Lifetime CA2521379C (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone
CA2811272A Expired - Lifetime CA2811272C (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA2811272A Expired - Lifetime CA2811272C (en) 2003-04-08 2004-04-08 Pharmaceutical formulations containing methylnaltrexone

Country Status (19)

Country Link
US (8) US20040266806A1 (en)
EP (3) EP2368553B1 (en)
JP (3) JP2006522818A (en)
CN (2) CN104383542B (en)
AU (2) AU2004229463B2 (en)
BR (1) BRPI0409133B8 (en)
CA (2) CA2521379C (en)
CY (3) CY1116272T1 (en)
DK (3) DK1615646T4 (en)
ES (3) ES2528669T3 (en)
HK (2) HK1082181A1 (en)
HR (3) HRP20150039T1 (en)
IL (1) IL171228A (en)
MX (1) MXPA05010817A (en)
PL (3) PL2368553T3 (en)
PT (3) PT2368553E (en)
RU (1) RU2362560C2 (en)
SI (3) SI2368554T1 (en)
WO (1) WO2004091623A1 (en)

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6274591B1 (en) * 1997-11-03 2001-08-14 Joseph F. Foss Use of methylnaltrexone and related compounds
US20030158220A1 (en) * 1997-11-03 2003-08-21 Foss Joseph F. Use of methylnaltrexone and related compounds to treat chronic opioid use side effects
US6375957B1 (en) 1997-12-22 2002-04-23 Euro-Celtique, S.A. Opioid agonist/opioid antagonist/acetaminophen combinations
ES2415876T3 (en) 1997-12-22 2013-07-29 Euro-Celtique S.A. Oral pharmaceutical dosage form comprising a combination of an opioid agonist and an opioid antagonist
EP1387673B1 (en) 2001-05-11 2010-12-29 Endo Pharmaceuticals Inc. Abuse-resistant controlled-release opioid dosage form
EP3241548A1 (en) 2002-04-05 2017-11-08 Euro-Celtique S.A. Matrix for sustained, invariant and independent release of active compounds
US7407955B2 (en) 2002-08-21 2008-08-05 Boehringer Ingelheim Pharma Gmbh & Co., Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
RU2373936C2 (en) * 2003-04-08 2009-11-27 Проджиникс Фармасьютикалз, Инк. Application of methyl naltrexone for treating irritable colon syndrome
CA2521420A1 (en) * 2003-04-08 2004-10-28 Progenics Pharmaceuticals, Inc. Combination therapy for constipation comprising a laxative and a peripheral opioid antagonist
US20040202717A1 (en) 2003-04-08 2004-10-14 Mehta Atul M. Abuse-resistant oral dosage forms and method of use thereof
DK1615646T4 (en) 2003-04-08 2022-10-10 Progenics Pharm Inc PHARMACEUTICAL FORMULATIONS CONTAINING METHYLNALTREXONE
EP1604666A1 (en) 2004-06-08 2005-12-14 Euro-Celtique S.A. Opioids for the treatment of the Chronic Obstructive Pulmonary Disease (COPD)
EP1845989A1 (en) * 2005-01-20 2007-10-24 Progenics Pharmaceuticals, Inc. Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction
EP1702558A1 (en) 2005-02-28 2006-09-20 Euro-Celtique S.A. Method and device for the assessment of bowel function
US9662325B2 (en) 2005-03-07 2017-05-30 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
ES2714198T3 (en) 2005-03-07 2019-05-27 Univ Chicago Use of opioid antagonists to attenuate the proliferation and migration of endothelial cells
US8518962B2 (en) 2005-03-07 2013-08-27 The University Of Chicago Use of opioid antagonists
US8524731B2 (en) 2005-03-07 2013-09-03 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
AR057325A1 (en) 2005-05-25 2007-11-28 Progenics Pharm Inc SYNTHESIS OF (S) -N-METHYLNTREXONE, PHARMACEUTICAL COMPOSITIONS AND USES
AR057035A1 (en) 2005-05-25 2007-11-14 Progenics Pharm Inc SYNTHESIS OF (R) -N-METHYLNTREXONE, PHARMACEUTICAL COMPOSITIONS AND USES
LT1962805T (en) 2005-12-08 2016-10-25 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections
AU2007240993B2 (en) * 2006-04-03 2013-08-01 Ceva Animal Health Pty Ltd Stabilized pentosan polysulfate (PPS) formulations and methods of analyzing them
PE20080251A1 (en) 2006-05-04 2008-04-25 Boehringer Ingelheim Int USES OF DPP IV INHIBITORS
EP1852108A1 (en) 2006-05-04 2007-11-07 Boehringer Ingelheim Pharma GmbH & Co.KG DPP IV inhibitor formulations
KR101541791B1 (en) 2006-05-04 2015-08-04 베링거 인겔하임 인터내셔날 게엠베하 Polymorphs
TWI489984B (en) * 2006-08-04 2015-07-01 Wyeth Corp Formulations for parenteral delivery of compounds and uses thereof
TW200815451A (en) * 2006-08-04 2008-04-01 Wyeth Corp 6-carboxy-normorphinan derivatives, synthesis and uses thereof
AU2013263750C1 (en) * 2006-08-04 2017-01-12 Wyeth Llc Formulations for parenteral delivery of compounds and uses thereof
TW200817048A (en) * 2006-09-08 2008-04-16 Wyeth Corp Dry powder compound formulations and uses thereof
US20080207669A1 (en) * 2006-11-22 2008-08-28 Progenics Pharmaceuticals, Inc. (S)-N-Stereoisomers of 7,8-Saturated-4,5-Epoxy-Morphinanium Analogs
WO2008075997A1 (en) * 2006-12-21 2008-06-26 Sergey Konstantinovich Sudakov Method for treating opioid withdrawal syndrome
EP2116241B1 (en) * 2007-01-26 2016-04-27 Pola Pharma Inc. Pharmaceutical composition
TWI466671B (en) 2007-03-29 2015-01-01 Progenics Pharm Inc Peripheral opioid receptor antagonists and uses thereof
CA2865389A1 (en) 2007-03-29 2008-10-09 Progenics Pharmaceuticals, Inc. Crystal forms and uses thereof
SI2565195T1 (en) 2007-03-29 2015-09-30 Wyeth Llc Peripheral opioid receptor and antagonists and uses thereof
BRPI0809651A2 (en) * 2007-03-30 2014-10-07 Tioga Pharmaceuticals Inc KAPPA-OPIATE AGONISTS FOR THE TREATMENT OF IRRITABLE COLD SYNDROME OF PREDOMINANT OR ALTERNATED DIARRHEA
EP2151241B1 (en) 2007-04-26 2012-03-21 Toray Industries, Inc. Stable solid preparation comprising 4,5-epoxymorphinan derivative
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
WO2009009292A2 (en) 2007-07-11 2009-01-15 Mallinckrodt Inc. Crystalline forms of naltrexone methobromide
JP5905198B2 (en) * 2007-10-01 2016-04-20 ザ ユニヴァーシティー オヴ シカゴ Treatment of drug-induced nausea with opioid antagonists
EP2065038A1 (en) * 2007-11-30 2009-06-03 Pharnext New therapeutic approaches for treating Charcot-Marie-Tooth disease
AU2008349873B2 (en) 2008-02-06 2014-02-13 Progenics Pharmaceuticals, Inc. Preparation and use of (R),(R)-2,2'-bis-methylnaltrexone
AU2009225434B2 (en) 2008-03-21 2014-05-22 The University Of Chicago Treatment with opioid antagonists and mTOR inhibitors
AR071175A1 (en) 2008-04-03 2010-06-02 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITION THAT INCLUDES AN INHIBITOR OF DIPEPTIDIL-PEPTIDASA-4 (DPP4) AND A COMPARING PHARMACO
MX355683B (en) * 2008-04-18 2018-04-26 Angiochem Inc Pharmaceutical compositions of paclitaxel, paclitaxel analogs or paclitaxel conjugates and related methods of preparation and use.
JP5985824B2 (en) 2008-07-01 2016-09-06 ザ ユニヴァーシティー オヴ シカゴ Opioid receptor antagonist-containing particles and methods of use
KR20200118243A (en) 2008-08-06 2020-10-14 베링거 인겔하임 인터내셔날 게엠베하 Treatment for diabetes in patients inappropriate for metformin therapy
CA2676881C (en) 2008-09-30 2017-04-25 Wyeth Peripheral opioid receptor antagonists and uses thereof
AU2009304560A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Conjugates of GLP-1 agonists and uses thereof
US20200155558A1 (en) 2018-11-20 2020-05-21 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients with insufficient glycemic control despite therapy with an oral antidiabetic drug
AU2009307367B2 (en) 2008-10-24 2015-04-23 Toray Industries, Inc. Stable tablet containing 4,5-epoxymorphinan derivative
AU2009322043A1 (en) 2008-12-05 2011-07-07 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
MX347106B (en) 2009-03-10 2017-04-12 Euro-Celtique S A * Immediate release pharmaceutical compositions comprising oxycodone and naloxone.
ES2729261T3 (en) 2009-04-20 2019-10-31 Angiochem Inc Ovarian cancer treatment using an anticancer agent conjugated to an Angiopep-2 analog
CA2766537A1 (en) 2009-07-02 2011-01-06 Angiochem Inc. Multimeric peptide conjugates and uses thereof
ES2560210T3 (en) * 2009-07-22 2016-02-17 Grünenthal GmbH Tamper-resistant dosage form for oxidation-sensitive opiates
KR20210033559A (en) 2009-11-27 2021-03-26 베링거 인겔하임 인터내셔날 게엠베하 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
US8524276B2 (en) 2010-03-11 2013-09-03 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone
WO2011141489A1 (en) 2010-05-10 2011-11-17 Euro-Celtique S.A. Manufacturing of active-free granules and tablets comprising the same
JP5671609B2 (en) 2010-05-10 2015-02-18 ユーロ−セルティーク エス.エイ. Pharmaceutical composition comprising hydromorphone and naloxone
BR112012028656A2 (en) 2010-05-10 2016-08-09 Euro Celtique Sa combination of active loaded granules with additional assets
PT2616064T (en) 2010-10-21 2019-11-06 Rtu Pharmaceuticals Llc Ready to use ketorolac formulations
AR083878A1 (en) 2010-11-15 2013-03-27 Boehringer Ingelheim Int VASOPROTECTORA AND CARDIOPROTECTORA ANTIDIABETIC THERAPY, LINAGLIPTINA, TREATMENT METHOD
CN102525911B (en) * 2012-03-20 2013-09-18 南京臣功制药股份有限公司 Methyhaaltrexone bromide injection and preparation method thereof
RU2483731C1 (en) * 2012-04-24 2013-06-10 Федеральное государственное унитарное предприятие "Московский эндокринный завод" Solution for injections containing nalbuphine
US20130303462A1 (en) 2012-05-14 2013-11-14 Boehringer Ingelheim International Gmbh Use of a dpp-4 inhibitor in podocytes related disorders and/or nephrotic syndrome
US10485798B2 (en) 2012-08-22 2019-11-26 Aptapharma Inc. Methylnaltrexone nasal formulations, methods of making, and use thereof
RU2675859C2 (en) 2012-11-29 2018-12-25 Инсмед Инкорпорейтед Stabilised vancomycin formulations
US20140179727A1 (en) 2012-12-14 2014-06-26 Trevi Therapeutics, Inc. Methods for treating pruritus
US8637538B1 (en) 2012-12-14 2014-01-28 Trevi Therapeutics, Inc. Methods for treatment of pruritis
US8987289B2 (en) 2012-12-14 2015-03-24 Trevi Therapeutics, Inc. Methods for treating pruritus
EP2968729B1 (en) 2013-03-14 2018-08-15 Fresenius Kabi Deutschland GmbH Packaging system for oxygen-sensitive drugs
CA2902343C (en) * 2013-03-14 2022-08-02 Becton Dickinson France S.A.S. Injectable morphine formulations
EP3024461B1 (en) 2013-07-23 2020-05-13 Euro-Celtique S.A. A combination of oxycodone and naloxone for use in treating pain in patients suffering from pain and a disease resulting in intestinal dysbiosis and/or increasing the risk for intestinal bacterial translocation
KR102194174B1 (en) 2013-11-13 2020-12-23 유로-셀티큐 에스.에이. Hydromorphone and naloxone for treatment of pain and opioid bowel dysfunction syndrome
EP3142643B1 (en) 2014-05-15 2019-07-10 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
CN104116707B (en) * 2014-08-03 2016-11-23 张星一 A kind of pharmaceutical composition containing methylnaltrexone bromide
CN104224734A (en) * 2014-08-15 2014-12-24 南京优科生物医药有限公司 Dezocine freeze-dried powder injection and preparation method thereof
CN107249327A (en) 2014-10-17 2017-10-13 萨利克斯药品公司 Slow down tumour progression using methyl naltrexone
CN105769755A (en) * 2014-12-23 2016-07-20 北大方正集团有限公司 Methyhaaltrexone bromide injection and preparation method thereof
CA2989400A1 (en) 2015-06-15 2016-12-22 Angiochem Inc. Ang1005 for the treatment of leptomeningeal carcinomatosis
JP2019517542A (en) 2016-06-10 2019-06-24 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination of linagliptin and metformin
CN106265493A (en) * 2016-08-09 2017-01-04 成都佳迪璐莎生物科技有限公司 Improve the pharmaceutical composition of Codeine Hydrochloride drug injection preparation stability
CN106176588A (en) * 2016-08-09 2016-12-07 成都佳迪璐莎生物科技有限公司 Improve the preparation method of composition of Codeine Hydrochloride ejection preparation stability
CA3047325A1 (en) * 2016-12-21 2018-06-28 Tioga Pharmaceuticals Inc. Solid pharmaceutical formulations of asimadoline
WO2019055880A2 (en) * 2017-09-15 2019-03-21 Ampersand Biopharmaceuticals, Inc. Method of administration and treatment
WO2019191627A1 (en) 2018-03-30 2019-10-03 Insmed Incorporated Methods for continuous manufacture of liposomal drug products
SG11202100580UA (en) 2018-07-23 2021-02-25 Trevi Therapeutics Inc Treatment of chronic cough, breathlessness and dyspnea
WO2020033402A1 (en) * 2018-08-06 2020-02-13 Summit Biosciences Inc. Drug products for nasal administration and uses thereof
CN114207101A (en) 2019-07-31 2022-03-18 埃科莱布美国股份有限公司 Deliming composition free of personal protective equipment
IT202000002860A1 (en) * 2020-02-13 2021-08-13 Tred S R L COMPOSITIONS FOR THE DISINFECTANT TREATMENT OF THE RHINOPHARYNGAL CAVITY

Family Cites Families (280)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1420015B1 (en) * 1959-10-16 1971-08-26 Boehringer Sohn Ingelheim 2'-Hydroxy-5,9-dimethyl-6,7-benzomorphane
GB1202148A (en) 1968-03-06 1970-08-12 Sankyo Co Pharmaceutical compositions
US3854480A (en) 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US3714159A (en) * 1971-03-30 1973-01-30 Janssen Pharmaceutica Nv 2,2-diaryl-4-(4'-aryl-4'-hydroxy-piper-idino)-butyramides
US3884916A (en) * 1971-03-30 1975-05-20 Janssen Pharmaceutica Nv 2,2-Diaryl-4-(4-aryl-4-hydroxy-piperidino)-butyramides
US4025652A (en) * 1975-03-31 1977-05-24 William H. Rorer, Inc. Amidinoureas
US4326074A (en) * 1972-09-22 1982-04-20 William H. Rorer, Inc. Amidinoureas
US3937801A (en) * 1973-07-10 1976-02-10 American Home Products Corporation Reducing the incidence of gastrointestinal side effects during the treatment of inflammatory conditions with antiinflammatory drugs
US4060635A (en) 1975-03-31 1977-11-29 William H. Rorer, Inc. Amidinoureas for treating diarrhea
US4203920A (en) * 1975-03-31 1980-05-20 William H. Rorer, Inc. Amidinoureas
US4066654A (en) * 1975-04-16 1978-01-03 G. D. Searle & Co. 1-triarylalkyl-4-phenyl-4-piperidine carboxylic acids and derivatives
US4072686A (en) * 1975-04-16 1978-02-07 G. D. Searle & Co. 1-(3,3,3-Triarylalkyl)-4-phenyl-piperidinealkanols
US3996214A (en) 1976-02-23 1976-12-07 G. D. Searle & Co. 5-(1,1-Diphenyl-4-(cyclic amino) but-2-trans-en-1-yl)-2-alkyl-1,3,4-oxadiazoles and intermediates thereto
US4013668A (en) * 1976-03-10 1977-03-22 G. D. Searle & Co. 5-(1,1-diphenyl-3-(5- or 6-hydroxy-2-azabicyclo(2.2.2)oct-2-yl)propyl)-2-alkyl-1,3,4-oxadiazoles and related compounds
US4012393A (en) * 1976-03-22 1977-03-15 G. D. Searle & Co. 2-[5-(CYCLIC AMINO) ETHYL-10,11-DIHYDRO-5H-dibenzo[a,d]-cyclohepten-5- yl]-5
US4115400A (en) 1976-05-27 1978-09-19 Eli Lilly And Company 1-Azoniabicyclo[3.1.0]hexanes
GB1593191A (en) 1977-03-23 1981-07-15 Reckitt & Colmann Prod Ltd Derivatives of morphine
US4125531A (en) 1977-04-18 1978-11-14 G. D. Searle & Co. 2-Substituted-1-azabicyclo[2.2.2]octanes
US4069223A (en) * 1977-05-02 1978-01-17 G. D. Searle & Co. 4-Aminomethyl-1-(3,3,3-triarylpropyl)-4-arylpiperidine and derivatives thereof
US4116963A (en) 1977-05-23 1978-09-26 G.D. Searle & Co. 3,3,3-triarylalkyl-4-phenylalkyl-4-hydroxy piperidines and related compounds
US4194045A (en) * 1977-12-27 1980-03-18 G. D. Searle & Co. 1-(3,3-Diaryl-3-oxadiazolalkyl)-4-phenyl-4-piperidinomethanols and related compounds
IT1096665B (en) 1978-06-14 1985-08-26 Tecnofarmaci Spa PHARMACEUTICAL COMPOSITION FOR THE THERAPY OF FRIGIDITY AND IMPOTENCE STATES
US4176186A (en) 1978-07-28 1979-11-27 Boehringer Ingelheim Gmbh Quaternary derivatives of noroxymorphone which relieve intestinal immobility
JPS5535031A (en) * 1978-09-04 1980-03-11 Shin Etsu Chem Co Ltd Enteric coating composition
US4311833A (en) * 1979-03-06 1982-01-19 Daicel Chemical Industries Ltd. Process for preparing ethylcarboxymethylcellulose
US4277605A (en) 1980-03-07 1981-07-07 Bristol-Myers Company Chemical compounds
US4322426A (en) * 1980-04-28 1982-03-30 E. I. Du Pont De Nemours And Company 17-Substituted-6-desoxy-7,8-dihydro-6α-methylnoroxymorphone narcotic antagonists
US4675189A (en) 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
US4466968A (en) 1980-11-24 1984-08-21 Dermall, Ltd. Method for prophylaxis or treatment of emesis and nausea
US4427676A (en) * 1980-12-19 1984-01-24 John Wyeth & Brother Ltd. Thiomorpholine derivatives
US4377568A (en) * 1981-08-12 1983-03-22 Merck Sharp & Dohme (I.A.) Corp. Preparation of aqueous alcoholic dispersions of pH sensitive polymers and plasticizing agents and a method of enteric coating dosage forms using same
DK150008C (en) * 1981-11-20 1987-05-25 Benzon As Alfred PROCEDURE FOR THE PREPARATION OF A PHARMACEUTICAL ORAL POLYDEPOT PREPARATION
WO1983003197A1 (en) 1982-03-16 1983-09-29 Univ Rockefeller Method for controlling gastrointestinal dysmotility
US4987136A (en) * 1982-03-16 1991-01-22 The Rockefeller University Method for controlling gastrointestinal dysmotility
US4870084A (en) 1982-03-16 1989-09-26 Pfizer Inc. Bicyclic benzo fused pyran compounds used for nausea treatment and prevention
US4430327A (en) * 1982-05-18 1984-02-07 Eli Lilly And Company Method for treating pregnant females for pain and anxiety
US4457907A (en) * 1982-08-05 1984-07-03 Clear Lake Development Group Composition and method for protecting a therapeutic drug
US4533739A (en) 1982-10-12 1985-08-06 G. D. Searle & Co. 2-[(Aminophenyl and amidophenyl)amino]-1-azacycloalkanes having antidiarrheal activity
US4518433A (en) * 1982-11-08 1985-05-21 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4452775A (en) 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
US4462839A (en) * 1983-06-16 1984-07-31 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4556552A (en) * 1983-09-19 1985-12-03 Colorcon, Inc. Enteric film-coating compositions
EP0143949B1 (en) * 1983-11-01 1988-10-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Pharmaceutical composition containing urokinase
US4689332A (en) 1984-04-09 1987-08-25 Research Corporation Growth regulation and related applications of opioid antagonists
US5266574A (en) * 1984-04-09 1993-11-30 Ian S. Zagon Growth regulation and related applications of opioid antagonists
US4666716A (en) 1984-09-04 1987-05-19 Richardson-Vicks Inc. Antidiarrheal compositions and use thereof
JPS6229515A (en) * 1985-07-30 1987-02-07 Shinjiro Tsuji Method for film-coating of hard capsule
JPH0676314B2 (en) 1985-09-30 1994-09-28 花王株式会社 Suppository base and suppository
US4824853A (en) * 1985-10-11 1989-04-25 Janssen Pharmaceutica N.V. α,α-diaryl-4-aryl-4-hydroxy-1-piperidinebutanamide, N-oxides and method of treating diarrhea
US4730048A (en) * 1985-12-12 1988-03-08 Regents Of The University Of Minnesota Gut-selective opiates
US4806556A (en) * 1985-12-12 1989-02-21 Regents Of The University Of Minnesota Gut-selective opiates
US4719215A (en) 1986-03-07 1988-01-12 University Of Chicago Quaternary derivatives of noroxymorphone which relieve nausea and emesis
EP0306575B1 (en) 1987-09-10 1992-07-29 The University Of Chicago Quaternary derivatives of noroxymorphone which relieve nausea and emesis
US4861781A (en) * 1986-03-07 1989-08-29 The University Of Chicago Quaternary derivatives of noroxymorphone which relieve nausea and emesis
DE3609073C2 (en) 1986-03-18 1995-08-10 Hans J Prof Dr Rer Nat Schmitt Measuring device for non-invasive detection of peripheral drainage and flow disorders in human extremities
US4990521A (en) * 1986-07-03 1991-02-05 Janssen Pharmaceutica 4-(aroylamino)piperidine-butanimide derivatives
EP0319545B1 (en) * 1986-08-28 1992-03-18 Enzacor Properties Limited Animal growth promotant
US5597564A (en) * 1986-08-28 1997-01-28 Enzacor Properties Limited Method of administering a microgranular preparation to the intestinal region of animals
US4888346A (en) * 1986-10-07 1989-12-19 Bernard Bihari Method for the treatment of persons infected with HTLV-III (AIDS) virus
US4765978A (en) 1986-12-16 1988-08-23 Schering Corporation Novel vaginal suppository
FR2609632B1 (en) 1987-01-21 1991-03-29 Shelly Marc NOVEL THERAPEUTIC APPLICATION OF 17- (CYCLOPROPYLMETHYL) -4,5-EPOXY-3,14-DIHYDROXYMORPHINON-6-ONE AND PHARMACEUTICAL COMPOSITIONS FOR THIS USE
NL8700842A (en) 1987-04-10 1988-11-01 Duphar Int Res
US4891379A (en) * 1987-04-16 1990-01-02 Kabushiki Kaisha Kobe Seikosho Piperidine opioid antagonists
IL83086A (en) 1987-07-06 1991-03-10 Teva Pharma Stable,injectable solutions of vincristine salts
US4857833A (en) * 1987-08-27 1989-08-15 Teradyne, Inc. Diagnosis of faults on circuit board
CA1315689C (en) 1987-09-03 1993-04-06 Leon I. Goldberg Quarternary derivatives of noroxymorphone which relieve nausea and emesis
US4912114A (en) * 1988-03-18 1990-03-27 Sandoz Ltd. Morphinan derivatives
SG64368A1 (en) * 1988-06-30 1999-04-27 Astra Ab Dermorphin analogs their methods of preparation pharmaceutical compositions and methods of therapeutic treatment using the same
JPH0653683B2 (en) 1988-07-14 1994-07-20 ザ ロックフェラー ユニバーシティ Oral composition for treating chronic pain or cough
EP0352361A1 (en) 1988-07-29 1990-01-31 The Rockefeller University Method of treating patients suffering from chronic pain or chronic cough
US4999342A (en) * 1988-08-16 1991-03-12 Ortho Pharmaceutical Corporation Long lasting contraceptive suppository composition and methods of use
US4857533A (en) 1988-12-15 1989-08-15 Baker Cummins Pharmaceuticals, Inc. Method of treatment for autoimmune diseases
US4863928A (en) 1989-01-04 1989-09-05 Baker Cummins Pharmaceuticals, Inc. Method of treatment for arthritic and inflammatory diseases
US5102887A (en) 1989-02-17 1992-04-07 Arch Development Corporation Method for reducing emesis and nausea induced by the administration of an emesis causing agent
US5116868A (en) * 1989-05-03 1992-05-26 The Johns Hopkins University Effective ophthalmic irrigation solution
US5133974A (en) 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
US4965269A (en) * 1989-12-20 1990-10-23 Ab Hassle Therapeutically active chloro substituted benzimidazoles
US5236947A (en) 1990-02-28 1993-08-17 Jouveinal S.A. Propanamines, their pharmacological properties and their application as an antidiarrheal
JPH0813748B2 (en) 1990-04-23 1996-02-14 帝國製薬株式会社 Colon disintegrating polypeptide oral preparation
BR9106438A (en) 1990-05-11 1993-05-18 Pfizer SYNERGISTIC THERAPEUTIC COMPOSITIONS AND PROCESSES
US5780012A (en) 1990-06-21 1998-07-14 Huland; Edith Method for reducing lung afflictions by inhalation of cytokine solutions
JP3160862B2 (en) 1990-11-15 2001-04-25 雪印乳業株式会社 Bone-fortified foods, feeds and pharmaceuticals
JPH04230625A (en) * 1990-12-27 1992-08-19 Standard Chem & Pharmaceut Corp Ltd Method for production of finely dispersed tablet composition consisting of microcapsule containing sprayed and dried sodium dichlofenac and having enteric coating
JPH06505747A (en) 1991-02-25 1994-06-30 トラスティーズ・オブ・ボストン・ユニバーシティ Opiate receptor antagonists that modulate hyperkinetic movement disorder
JP2916290B2 (en) 1991-03-22 1999-07-05 帝國製薬株式会社 Colon-disintegrating oral preparation containing bioactive polypeptide
US5250542A (en) 1991-03-29 1993-10-05 Eli Lilly And Company Peripherally selective piperidine carboxylate opioid antagonists
US5159081A (en) * 1991-03-29 1992-10-27 Eli Lilly And Company Intermediates of peripherally selective n-carbonyl-3,4,4-trisubstituted piperidine opioid antagonists
CA2064373C (en) 1991-03-29 2005-08-23 Buddy Eugene Cantrell Piperidine derivatives
US5270328A (en) * 1991-03-29 1993-12-14 Eli Lilly And Company Peripherally selective piperidine opioid antagonists
US5220017A (en) 1991-04-10 1993-06-15 Merck & Co., Inc. Cholecystokinin antagonists
DK0590060T3 (en) * 1991-06-21 1998-05-11 Univ Cincinnati Orally administrable therapeutic proteins and method of preparation
US5407686A (en) * 1991-11-27 1995-04-18 Sidmak Laboratories, Inc. Sustained release composition for oral administration of active ingredient
US5614219A (en) * 1991-12-05 1997-03-25 Alfatec-Pharma Gmbh Oral administration form for peptide pharmaceutical substances, in particular insulin
US5256154A (en) * 1992-01-31 1993-10-26 Sterling Winthrop, Inc. Pre-filled plastic syringes and containers and method of terminal sterilization thereof
JPH05213763A (en) 1992-02-10 1993-08-24 Sanwa Kagaku Kenkyusho Co Ltd Readily absorbable activated calcium preparation
WO1993020826A1 (en) 1992-04-10 1993-10-28 Vsesojuzny Nauchno-Issledovatelsky Institut Meditsinskikh Polimerov Pharmaceutical composition
US5686072A (en) 1992-06-17 1997-11-11 Board Of Regents, The University Of Texas Epitope-specific monoclonal antibodies and immunotoxins and uses thereof
US5472943A (en) * 1992-09-21 1995-12-05 Albert Einstein College Of Medicine Of Yeshiva University, Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other opioid agonists
US6096756A (en) * 1992-09-21 2000-08-01 Albert Einstein College Of Medicine Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other bimodally-acting opioid agonists
US5580876A (en) * 1992-09-21 1996-12-03 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other bimodally-acting opioid agonists
US5512578A (en) * 1992-09-21 1996-04-30 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by exogenous and endogenous opiod agonists
USRE36547E (en) * 1992-09-21 2000-02-01 Albert Einstein College Of Medicine Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by exogenous and endogenous opioid agonists
ATE348110T1 (en) 1992-10-28 2007-01-15 Genentech Inc HVEGF RECEPTOR AS A VEGF ANTAGONIST
CA2086631C (en) * 1992-12-22 1998-10-06 J. Gabriel Michael Oral administration of immunologically active biomolecules and other therapeutic proteins
DE4303214A1 (en) 1993-02-04 1994-08-11 Wolfgang Marks Treatment of diseases of viral, viroidal or oncogenic origin by steroid saponins or their aglycones
US5585348A (en) 1993-02-10 1996-12-17 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Use of excitatory opioid receptor antagonists to prevent growth factor-induced hyperalgesia
US5656290A (en) * 1993-02-26 1997-08-12 The Procter & Gamble Company Bisacodyl dosage form with multiple enteric polymer coatings for colonic delivery
US5391372A (en) * 1993-06-28 1995-02-21 Campbell; Elizabeth Methods of treating colic and founder in horses
JP3843456B2 (en) 1993-07-23 2006-11-08 東レ株式会社 Morphinan derivatives and pharmaceutical applications
US5358970A (en) 1993-08-12 1994-10-25 Burroughs Wellcome Co. Pharmaceutical composition containing bupropion hydrochloride and a stabilizer
GB2281205A (en) 1993-08-24 1995-03-01 Euro Celtique Sa Oral opioid analgesic
SE9303744D0 (en) * 1993-11-12 1993-11-12 Astra Ab Pharmaceutical emulsion
US5434171A (en) 1993-12-08 1995-07-18 Eli Lilly And Company Preparation of 3,4,4-trisubstituted-piperidinyl-N-alkylcarboxylates and intermediates
US6190691B1 (en) * 1994-04-12 2001-02-20 Adolor Corporation Methods for treating inflammatory conditions
WO1995030774A1 (en) * 1994-05-05 1995-11-16 Beckman Instruments, Inc. Oligonucleotide repeat arrays
IT1269826B (en) 1994-05-24 1997-04-15 Paolo Minoia USE OF OPTIACEAN ANTAGONISTS AND CALCIUM SALTS FOR THE PREPARATION OF MEDICATIONS FOR THE TREATMENT OF ENDORPHINE-MEDIATED PATHOLOGICAL FORMS
US5536507A (en) * 1994-06-24 1996-07-16 Bristol-Myers Squibb Company Colonic drug delivery system
US5866154A (en) * 1994-10-07 1999-02-02 The Dupont Merck Pharmaceutical Company Stabilized naloxone formulations
US5614222A (en) * 1994-10-25 1997-03-25 Kaplan; Milton R. Stable aqueous drug suspensions and methods for preparation thereof
US5965161A (en) * 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
US5578725A (en) 1995-01-30 1996-11-26 Regents Of The University Of Minnesota Delta opioid receptor antagonists
ES2094694B1 (en) * 1995-02-01 1997-12-16 Esteve Quimica Sa NEW PHARMACEUTICALLY STABLE FORMULATION OF A COMPOUND OF BENZMIDAZOLE AND ITS PROCESS OF OBTAINING.
US6096763A (en) 1995-02-23 2000-08-01 Merck & Co., Inc. α1a adrenergic receptor antagonists
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US5714586A (en) * 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5821219A (en) 1995-08-11 1998-10-13 Oregon Health Sciences University Opioid antagonists and methods of their use
GB9517001D0 (en) 1995-08-18 1995-10-18 Denny William Enediyne compounds
US5804595A (en) * 1995-12-05 1998-09-08 Regents Of The University Of Minnesota Kappa opioid receptor agonists
EP0780372B3 (en) 1995-12-21 2013-05-22 Syngenta Participations AG 3-Amino-2-mercaptobenzoic acid derivatives and processes for their preparation
WO1997029739A2 (en) 1996-02-15 1997-08-21 Janssen Pharmaceutica N.V. Use of 5ht4 receptor antagonists for overcoming gastrointestinal effects of serotonin reuptake inhibitors
DK0914097T3 (en) 1996-03-12 2002-04-29 Alza Corp Composition and dosage form comprising opioid antagonist
US6136780A (en) 1996-03-29 2000-10-24 The Penn State Research Foundation Control of cancer growth through the interaction of [Met5 ]-enkephalin and the zeta (ζ) receptor
US20040024006A1 (en) * 1996-05-06 2004-02-05 Simon David Lew Opioid pharmaceutical compositions
DE19651551C2 (en) 1996-12-11 2000-02-03 Klinge Co Chem Pharm Fab Opioid antagonist-containing galenic formulation
US20010036469A1 (en) 1997-01-13 2001-11-01 Gooberman Lance L. Opiate antagonist implant and process for preparation therefor
WO1998035679A1 (en) 1997-02-14 1998-08-20 Gödecke Aktiengesellschaft Stabilization of naloxonhydrochlorid
US6884879B1 (en) * 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
US6329516B1 (en) 1997-04-28 2001-12-11 Fmc Corporation Lepidopteran GABA-gated chloride channels
GB9801231D0 (en) * 1997-06-05 1998-03-18 Merck & Co Inc A method of treating cancer
HU9701081D0 (en) * 1997-06-23 1997-08-28 Gene Research Lab Inc N Pharmaceutical composition of antitumoral activity
US6525038B1 (en) * 1997-06-24 2003-02-25 Werner Kreutz Synergistic compositions for the selective control of tumor tissue
US6353004B1 (en) * 1997-07-14 2002-03-05 Adolor Coporation Peripherally acting anti-pruritic opiates
US6096764A (en) 1997-08-21 2000-08-01 Eli Lilly And Company Methods for inhibiting detrimental side-effects due to GnRH of GnRH agonist administration
US6099853A (en) 1997-09-04 2000-08-08 Protein Express Vaginal suppository vaccine for urogenital infections
EP0913152B1 (en) 1997-11-03 2001-12-19 Stada Arzneimittel Ag Stabilised combination of drugs comprising naloxone and an opioid analgesic
US6559158B1 (en) * 1997-11-03 2003-05-06 Ur Labs, Inc. Use of methylnaltrexone and related compounds to treat chronic opioid use side affects
US20030158220A1 (en) * 1997-11-03 2003-08-21 Foss Joseph F. Use of methylnaltrexone and related compounds to treat chronic opioid use side effects
US5972954A (en) * 1997-11-03 1999-10-26 Arch Development Corporation Use of methylnaltrexone and related compounds
AU2003204844B2 (en) 1997-11-03 2007-06-07 Arch Development Corporation Use of methylnaltrexone and related compounds
US6274591B1 (en) 1997-11-03 2001-08-14 Joseph F. Foss Use of methylnaltrexone and related compounds
US6777534B1 (en) 1997-12-09 2004-08-17 Children's Medical Center Corporation Peptide antagonists of vascular endothelial growth factor
ES2415876T3 (en) * 1997-12-22 2013-07-29 Euro-Celtique S.A. Oral pharmaceutical dosage form comprising a combination of an opioid agonist and an opioid antagonist
EP0930334A1 (en) 1998-01-16 1999-07-21 Quest International B.V. Polysaccharide conjugate capable of binding cellulose
EP1051493A2 (en) 1998-01-26 2000-11-15 Unilever Plc Method for producing antibody fragments
GB9802251D0 (en) 1998-02-03 1998-04-01 Ciba Geigy Ag Organic compounds
JP4470321B2 (en) * 1998-04-03 2010-06-02 味の素株式会社 Antitumor agent
DE19818802A1 (en) 1998-04-27 1999-10-28 Dresden Arzneimittel Stable mitoxantron solutions useful for cancer therapy
US6359111B1 (en) * 1998-05-28 2002-03-19 Neorx Corporation Opioid receptor targeting
HN1999000149A (en) * 1998-09-09 2000-01-12 Pfizer Prod Inc DERIVATIVES OF 4,4-BIARILPIPERIDINA
US20010010919A1 (en) 1998-10-13 2001-08-02 David K. Grandy Opioid antagonists and methods of their use
BR9916765A (en) 1999-01-05 2001-09-25 Unilever Nv Process for producing an immunoadsorbent material, use of a protein that is linked via a covalent bond to an antibody fragment, immunosorbent material, use of a material, and, diagnostic test kit
DE60013767T3 (en) 1999-01-19 2009-07-09 Unilever N.V. PROCESS FOR THE PRODUCTION OF ANTIBODY FRAGMENTS
CA2293008A1 (en) 1999-01-28 2000-07-28 Hong Qi Premixed alatrofloxacin injectable compositions
NZ513356A (en) 1999-02-05 2004-01-30 Univ Leiden Method of modulating metabolite biosynthesis in recombinant cells using an AP2-domain transcription factor that is involved in the response of a plant cell to a jasmonate
US6194382B1 (en) * 1999-03-03 2001-02-27 Albert Einstein College Of Medicine Of Yeshiva University Method and composition for treating irritable bowel syndrome using low doses of opioid receptor antagonists
JP4049477B2 (en) 1999-03-23 2008-02-20 大鵬薬品工業株式会社 Side effect reducing agent
OA11862A (en) 1999-04-22 2006-03-02 Unilever Nv Inhibition of viral infection using monovalent antigen-binding proteins.
US7129265B2 (en) 1999-04-23 2006-10-31 Mason R Preston Synergistic effects of amlodipine and atorvastatin metabolite as a basis for combination therapy
US6171620B1 (en) 1999-04-27 2001-01-09 Health Research, Inc. Method of enhancing the efficacy of anti-tumor agents
AU4564200A (en) 1999-04-29 2000-11-17 Aventis Pharma S.A. Method for treating cancer using camptothecin derivatives and 5-fluorouracil
US6833349B2 (en) 1999-06-08 2004-12-21 Regeneron Pharmaceuticals, Inc. Methods of treating inflammatory skin diseases
US20020068712A1 (en) 1999-07-23 2002-06-06 Troy Stevens Use of decreasing levels of functional transient receptor potential gene product
US20030105121A1 (en) 1999-07-27 2003-06-05 Bernard Bihari Method of preventing lipodystrophy syndrome or reversing a pre-existing syndrome in HIV-infected patients being treated with antiretroviral agents
EP1198572A2 (en) 1999-08-02 2002-04-24 Keygene N.V. Method for generating cgmmv resistant plants, genetic constructs, and obtained cgmmv-resistant plants
WO2001013909A2 (en) 1999-08-25 2001-03-01 Cooper Barrett R Compositions and methods for treating opiate intolerance
CA2388560A1 (en) 1999-08-31 2001-03-08 Grunenthal Gmbh Sustained-release form of administration containing tramadol saccharinate
US6451806B2 (en) * 1999-09-29 2002-09-17 Adolor Corporation Methods and compositions involving opioids and antagonists thereof
EP1225897B1 (en) 1999-11-01 2004-09-08 RHODES, John Composition for treatment of constipation and irritable bowel syndrome
DE69927688T2 (en) * 1999-11-04 2006-07-06 Institut Gustave Roussy Antiviral agent in combination with radiotherapy for use in the treatment of cancer
US6469030B2 (en) * 1999-11-29 2002-10-22 Adolor Corporation Methods for the treatment and prevention of ileus
EP1244447B1 (en) 1999-11-29 2007-01-10 Adolor Corporation Novel methods and compositions involving opioids and antagonists thereof
IL149650A0 (en) 1999-11-29 2002-11-10 Adolor Corp Novel methods for the treatment and prevention of ileus
US6384044B1 (en) 1999-11-29 2002-05-07 Bernard Bihari Method of treating cancer of the prostate
WO2001041705A2 (en) * 1999-11-29 2001-06-14 Adolor Corporation Novel methods for the treatment and prevention of dizziness and pruritus
US6911455B2 (en) * 1999-12-22 2005-06-28 Smithkline Beecham Corporation Methods for preparing pharmaceutical formulations
US6545010B2 (en) 2000-03-17 2003-04-08 Aventis Pharma S.A. Composition comprising camptothecin or a camptothecin derivative and a platin derivative for the treatment of cancer
US20010046968A1 (en) 2000-03-23 2001-11-29 Zagon Ian S. Opioid growth factor modulates angiogenesis
US6967075B2 (en) * 2000-04-07 2005-11-22 Schering Corporation HCV replicase complexes
CA2408106A1 (en) 2000-05-05 2001-11-15 Pain Therapeutics, Inc. Opioid antagonist compositions and dosage forms
AU7013401A (en) * 2000-06-22 2002-01-02 Univ Iowa Res Found Methods for enhancing antibody-induced cell lysis and treating cancer
EP1175905A1 (en) 2000-07-24 2002-01-30 Societe Des Produits Nestle S.A. Nutritional Composition
FI116089B (en) * 2000-07-27 2005-09-15 Johan Tore Karlstroem Device and procedures for controls
CZ302087B6 (en) 2000-07-28 2010-10-06 F. Hoffmann-La Roche Ag Pharmaceutical composition containing orlistat along with bile acid sequestrant for use in treatment and prevention of obesity
JP4183371B2 (en) 2000-08-31 2008-11-19 日本・バイオ株式会社 Manufacturing method of fermented turmeric
NZ507152A (en) * 2000-09-27 2001-06-29 Hiltive Pty Ltd Wall cladding assembly with cladding having recesses along opposite sides to engage with flanges of support members
US6630462B2 (en) 2000-11-17 2003-10-07 Adolor Corporation Delta agonist analgesics
JP2002229183A (en) * 2000-12-01 2002-08-14 Hoya Corp Lithography mask blank and method for manufacturing the same
CN1322143C (en) 2000-12-04 2007-06-20 普里马根公司 Testing endosymbiont cellular organelles and compounds identifiable therewith
UA73619C2 (en) 2000-12-13 2005-08-15 Pfizer Prod Inc Stable pharmaceutical compositions of nmda receptor agonist (variants) and method of treatment
MXPA03005135A (en) 2000-12-13 2003-12-04 Lilly Co Eli Chronic treatment regimen using glucagon-like insulinotropic peptides.
KR100405161B1 (en) 2000-12-14 2003-11-12 신풍제약주식회사 Pharmaceutical composition for intramuscular injection containing loxoprofen
GB0100115D0 (en) 2001-01-04 2001-02-14 Alchemia Pty Ltd Delivery systems
US6693125B2 (en) * 2001-01-24 2004-02-17 Combinatorx Incorporated Combinations of drugs (e.g., a benzimidazole and pentamidine) for the treatment of neoplastic disorders
ATE450265T1 (en) 2001-03-23 2009-12-15 Shire Canada Inc PHARMACEUTICAL MIXTURE FOR THE TREATMENT OF CANCER CONTAINING DIOXOLANE NUCLEOSIDE ANALOGUES
US20040242523A1 (en) 2003-03-06 2004-12-02 Ana-Farber Cancer Institue And The Univiersity Of Chicago Chemo-inducible cancer gene therapy
WO2002080987A1 (en) 2001-04-09 2002-10-17 Progenics Pharmaceuticals, Inc. Anti-cd19 immunotoxins
WO2002087586A1 (en) * 2001-04-26 2002-11-07 Control Delivery Systems, Inc. Sustained release drug delivery system containing codrugs
DE60234183D1 (en) * 2001-06-05 2009-12-10 Univ Chicago USE OF METHYL NALTREXONE FOR THE TREATMENT OF IMMUNOSUPPRESSION
EP1432429A4 (en) 2001-08-31 2005-09-21 Univ Louisiana State Inhibition of angiogenesis and destruction of angiogenic vessels with extracts of noni juice morinda citrifolia
SI1436012T1 (en) * 2001-10-18 2018-03-30 Nektar Therapeutics Polymer conjugates of opioid antagonists
US20030144312A1 (en) 2001-10-30 2003-07-31 Schoenhard Grant L. Inhibitors of ABC drug transporters in multidrug resistant cancer cells
WO2003037365A1 (en) * 2001-11-01 2003-05-08 The Johns Hopkins University Methods and compositions for treating vascular leak using hepatocyte growth factor
WO2003059904A1 (en) * 2001-12-21 2003-07-24 Merck & Co., Inc. Heteroaryl substituted pyrrole modulators of metabotropic glutamate receptor-5
AU2002368150A1 (en) 2002-02-04 2004-02-25 Jonathan Moss Use of methylnaltrexone in treating gastrointestinal dysfunction in equines
US20050011468A1 (en) * 2002-02-04 2005-01-20 Jonathan Moss Use of methylnaltrexone in treating gastrointestinal dysfunction in equines
US7074825B2 (en) 2002-03-07 2006-07-11 Huanbiao Mo Composition and method for treating cancer
US20030229111A1 (en) 2002-03-14 2003-12-11 Benjamin Oshlack Naltrexone hydrochloride compositions
US20030191147A1 (en) * 2002-04-09 2003-10-09 Barry Sherman Opioid antagonist compositions and dosage forms
US7355081B2 (en) 2002-04-17 2008-04-08 The University Of North Carolina At Chapel Hill Curcumin analogues and uses thereof
US7329642B2 (en) 2002-05-17 2008-02-12 Board Of Regents, University Of Texas System β-2-glycoprotein is an inhibitor of angiogenesis
US7012100B1 (en) 2002-06-04 2006-03-14 Avolix Pharmaceuticals, Inc. Cell migration inhibiting compositions and methods and compositions for treating cancer
US6986901B2 (en) * 2002-07-15 2006-01-17 Warner-Lambert Company Llc Gastrointestinal compositions
US20040010997A1 (en) * 2002-07-17 2004-01-22 Oren Close Guides to align masonry walls defining apertures, and methods of use
US7160913B2 (en) * 2002-09-13 2007-01-09 Thomas Jefferson University Methods and kit for treating Parkinson's disease
US7691374B2 (en) * 2002-10-23 2010-04-06 Health Research, Inc. Method for increasing the efficacy of anti-tumor agents by anti-endoglin antibody
EP1562953B1 (en) 2002-11-08 2009-09-30 Mallinckrodt Inc. Process for the preparation of quaternary n-alkyl morphinan alkaloid salts
WO2004054511A2 (en) 2002-12-13 2004-07-01 The Regents Of The University Of California Analgesic combination comprising nalbuphine
WO2004054569A1 (en) * 2002-12-16 2004-07-01 Council Of Scientific And Industrial Research Pharmaceutical composition containing brevifoliol for use in chemotherapeutic treatment of human beings
TW200500067A (en) * 2003-01-21 2005-01-01 Control Delivery Sys Inc Salts of codrugs and uses related thereto
ATE406360T1 (en) 2003-03-07 2008-09-15 Lilly Co Eli ANTAGONISTS OF OPIOID RECEPTORS
RU2373936C2 (en) * 2003-04-08 2009-11-27 Проджиникс Фармасьютикалз, Инк. Application of methyl naltrexone for treating irritable colon syndrome
CA2521420A1 (en) * 2003-04-08 2004-10-28 Progenics Pharmaceuticals, Inc. Combination therapy for constipation comprising a laxative and a peripheral opioid antagonist
DK1615646T4 (en) 2003-04-08 2022-10-10 Progenics Pharm Inc PHARMACEUTICAL FORMULATIONS CONTAINING METHYLNALTREXONE
MXPA05011557A (en) 2003-04-29 2006-03-09 Orexigen Therapeutics Inc Compositions for affecting weight loss.
US6992193B2 (en) 2003-06-10 2006-01-31 Adolor Corporation Sulfonylamino phenylacetamide derivatives and methods of their use
US7494979B2 (en) 2003-06-13 2009-02-24 Ironwood Pharmaceuticals, Inc. Method for treating congestive heart failure and other disorders
PL1644021T3 (en) * 2003-06-13 2013-01-31 Ironwood Pharmaceuticals Inc Methods and compositions for the treatment of gastrointestinal disorders
AU2003903387A0 (en) 2003-07-02 2003-07-17 Sirtex Medical Limited Combination therapy for treatment of neoplasia
US7066888B2 (en) * 2003-10-29 2006-06-27 Allez Physionix Ltd Method and apparatus for determining an ultrasound fluid flow centerline
US8946262B2 (en) 2003-12-04 2015-02-03 Adolor Corporation Methods of preventing and treating gastrointestinal dysfunction
US6984403B2 (en) * 2003-12-04 2006-01-10 Pfizer Inc. Azithromycin dosage forms with reduced side effects
AU2004303425C1 (en) 2003-12-19 2010-02-18 Schering Corporation Pharmaceutical compositions
JP4225922B2 (en) 2004-01-15 2009-02-18 アキレス株式会社 Polyolefin resin foam sheet
US20070082044A1 (en) * 2004-03-10 2007-04-12 Trustees Of Tufts College Synergistic effect of compositions comprising carotenoids selected from lutein, beta-carotene and lycopene
TW200533339A (en) 2004-03-16 2005-10-16 Bristol Myers Squibb Co Therapeutic synergy of anti-cancer compounds
US6946556B1 (en) 2004-05-21 2005-09-20 Acura Pharmaceuticals, Inc. Preparation of opioid analgesics by a one-pot process
US7094775B2 (en) 2004-06-30 2006-08-22 Bone Care International, Llc Method of treating breast cancer using a combination of vitamin D analogues and other agents
US7388008B2 (en) * 2004-08-02 2008-06-17 Ambrilia Biopharma Inc. Lysine based compounds
US20060063792A1 (en) * 2004-09-17 2006-03-23 Adolor Corporation Substituted morphinans and methods of their use
AU2005286662B2 (en) 2004-09-23 2011-10-06 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
ES2392645T3 (en) 2004-09-30 2012-12-12 Becton,Dickinson And Company Method to reduce or eliminate waste in a glass medical container and container made in accordance with it
EP1845989A1 (en) * 2005-01-20 2007-10-24 Progenics Pharmaceuticals, Inc. Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction
US8524731B2 (en) 2005-03-07 2013-09-03 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
ES2714198T3 (en) 2005-03-07 2019-05-27 Univ Chicago Use of opioid antagonists to attenuate the proliferation and migration of endothelial cells
AR057035A1 (en) 2005-05-25 2007-11-14 Progenics Pharm Inc SYNTHESIS OF (R) -N-METHYLNTREXONE, PHARMACEUTICAL COMPOSITIONS AND USES
AR057325A1 (en) * 2005-05-25 2007-11-28 Progenics Pharm Inc SYNTHESIS OF (S) -N-METHYLNTREXONE, PHARMACEUTICAL COMPOSITIONS AND USES
CA2610951A1 (en) 2005-06-03 2006-12-14 The University Of Chicago Modulation of microbial pathogen-host cell interactions
CA2609985A1 (en) 2005-06-03 2007-05-10 The University Of Chicago Modulation of cell barrier dysfunction
US20080194611A1 (en) 2005-06-03 2008-08-14 Alverdy John C Modulation of Cell Barrier Dysfunction
CA2611105A1 (en) 2005-06-09 2006-12-21 Mallinckrodt Inc. Method for separation and purification of naltrexone by preparative chromatography
PE20070207A1 (en) * 2005-07-22 2007-03-09 Genentech Inc COMBINED TREATMENT OF TUMORS THAT EXPRESS HER
WO2007025383A1 (en) * 2005-08-30 2007-03-08 Queen's University At Kingston Potentiation of the therapeutic action of an opioid receptor agonist and/or inhibition or reversal of tolerance to an opioid receptoi agonists using an ultralow dose of an alpha-2 receptor antagonist
PL116330U1 (en) 2005-10-31 2007-04-02 Alza Corp Method for the reduction of alcohol provoked rapid increase in the released dose of the orally administered opioide with prolonged liberation
US20070259939A1 (en) 2006-05-04 2007-11-08 Accelerated Technologies Using naltrexone as a multi-purpose health supplement to improve the human condition and preventing multiple diseases and infirmities by stimulating immune system vitality and robustness
US20080075771A1 (en) * 2006-07-21 2008-03-27 Vaughn Jason M Hydrophilic opioid abuse deterrent delivery system using opioid antagonists
TW200815451A (en) * 2006-08-04 2008-04-01 Wyeth Corp 6-carboxy-normorphinan derivatives, synthesis and uses thereof
TWI489984B (en) 2006-08-04 2015-07-01 Wyeth Corp Formulations for parenteral delivery of compounds and uses thereof
WO2008021394A2 (en) 2006-08-15 2008-02-21 Theraquest Biosciences, Llc Pharmaceutical formulations of cannabinoids and method of use
TW200817048A (en) 2006-09-08 2008-04-16 Wyeth Corp Dry powder compound formulations and uses thereof
MX2009005461A (en) 2006-11-22 2009-08-28 Progenics Pharm Inc N-oxides of 4,5-epoxy-morphinanium analogs.
MX2009005463A (en) 2006-11-22 2009-08-28 Progenics Pharm Inc Processes for synthesizing quaternary 4,5-epoxy-morphinan analogs and isolating their n-stereoisomers.
JP2010510329A (en) 2006-11-22 2010-04-02 プロジェニックス ファーマスーティカルス インコーポレーテッド 7,8-saturated-4,5-epoxy-morphinanium analogs
TW200843802A (en) 2007-02-09 2008-11-16 Drugtech Corp Compositions for improving gastrointestinal nutrient and drug absorption
TWI466671B (en) 2007-03-29 2015-01-01 Progenics Pharm Inc Peripheral opioid receptor antagonists and uses thereof
CA2865389A1 (en) 2007-03-29 2008-10-09 Progenics Pharmaceuticals, Inc. Crystal forms and uses thereof
SI2565195T1 (en) 2007-03-29 2015-09-30 Wyeth Llc Peripheral opioid receptor and antagonists and uses thereof
WO2009137086A1 (en) 2008-05-07 2009-11-12 Nektar Therapeutics Oral administration of peripherally-acting opioid antagonists
CA2676881C (en) 2008-09-30 2017-04-25 Wyeth Peripheral opioid receptor antagonists and uses thereof
JP5213763B2 (en) 2009-03-03 2013-06-19 キヤノン株式会社 Lens barrel and optical apparatus having the same
US8524276B2 (en) 2010-03-11 2013-09-03 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone

Also Published As

Publication number Publication date
EP2368553B1 (en) 2014-12-31
MXPA05010817A (en) 2006-03-30
US20140235664A1 (en) 2014-08-21
EP1615646B2 (en) 2022-07-27
US8552025B2 (en) 2013-10-08
BRPI0409133A (en) 2006-05-02
CY1116372T1 (en) 2017-02-08
PL2368554T3 (en) 2015-05-29
PT1615646E (en) 2015-02-12
HK1207965A1 (en) 2016-02-19
AU2004229463B2 (en) 2010-07-22
DK2368553T3 (en) 2015-02-09
ES2527870T5 (en) 2022-10-14
IL171228A (en) 2016-05-31
AU2010202824A1 (en) 2010-07-22
US20040266806A1 (en) 2004-12-30
HRP20150037T4 (en) 2022-09-02
BRPI0409133B1 (en) 2019-09-03
PL1615646T3 (en) 2015-05-29
CA2811272C (en) 2016-12-20
EP1615646B1 (en) 2014-12-24
CY1116373T1 (en) 2017-02-08
DK1615646T4 (en) 2022-10-10
RU2005134361A (en) 2006-05-10
PT2368554E (en) 2015-02-06
BRPI0409133B8 (en) 2021-05-25
US10376584B2 (en) 2019-08-13
HRP20150038T1 (en) 2015-06-05
PL1615646T5 (en) 2022-12-19
CN1767831A (en) 2006-05-03
DK1615646T3 (en) 2015-02-02
US20100261744A1 (en) 2010-10-14
PL2368553T3 (en) 2015-05-29
US20100267758A1 (en) 2010-10-21
ES2528631T3 (en) 2015-02-11
WO2004091623A1 (en) 2004-10-28
CN104383542A (en) 2015-03-04
CA2521379C (en) 2013-07-02
AU2004229463A1 (en) 2004-10-28
JP2015147783A (en) 2015-08-20
SI1615646T1 (en) 2015-05-29
HRP20150039T1 (en) 2015-06-05
US20170258911A1 (en) 2017-09-14
JP2006522818A (en) 2006-10-05
HRP20150037T1 (en) 2015-06-05
US20100261746A1 (en) 2010-10-14
EP2368554A1 (en) 2011-09-28
SI2368553T1 (en) 2015-05-29
CN104383542B (en) 2017-09-26
US20190358328A1 (en) 2019-11-28
US9669096B2 (en) 2017-06-06
EP2368554B1 (en) 2014-12-24
HK1082181A1 (en) 2006-06-02
ES2528669T3 (en) 2015-02-11
RU2362560C2 (en) 2009-07-27
US20100261745A1 (en) 2010-10-14
AU2010202824B2 (en) 2013-05-02
JP2012184240A (en) 2012-09-27
JP6262169B2 (en) 2018-01-17
ES2527870T3 (en) 2015-02-02
EP2368553A1 (en) 2011-09-28
DK2368554T3 (en) 2015-01-26
JP5847643B2 (en) 2016-01-27
SI2368554T1 (en) 2015-05-29
PT2368553E (en) 2015-03-03
CN1767831B (en) 2014-12-10
CA2811272A1 (en) 2004-10-28
CY1116272T1 (en) 2017-02-08
EP1615646A1 (en) 2006-01-18
SI1615646T2 (en) 2022-11-30

Similar Documents

Publication Publication Date Title
US10376584B2 (en) Stable pharmaceutical formulations of methylnaltrexone
EP2046388A2 (en) Formulations for parenteral delivery of compounds and uses thereof
AU2013203378B2 (en) Pharmaceutical formulations containing methylnaltrexone
AU2013263750C1 (en) Formulations for parenteral delivery of compounds and uses thereof

Legal Events

Date Code Title Description
EEER Examination request