US20030065003A1 - Use of methylnaltrexone and related compounds - Google Patents

Use of methylnaltrexone and related compounds Download PDF

Info

Publication number
US20030065003A1
US20030065003A1 US10/278,630 US27863002A US2003065003A1 US 20030065003 A1 US20030065003 A1 US 20030065003A1 US 27863002 A US27863002 A US 27863002A US 2003065003 A1 US2003065003 A1 US 2003065003A1
Authority
US
United States
Prior art keywords
methylnaltrexone
mntx
opioid
administration
enterically coated
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/278,630
Inventor
Joseph Foss
Michael Roizen
Jonathan Moss
Chun-Su Yuan
William Drell
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.)
University of Chicago
Progenics Pharmaceuticals Nevada Inc
Original Assignee
University of Chicago
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
Priority claimed from US08/962,742 external-priority patent/US5972954A/en
Application filed by University of Chicago filed Critical University of Chicago
Priority to US10/278,630 priority Critical patent/US20030065003A1/en
Publication of US20030065003A1 publication Critical patent/US20030065003A1/en
Assigned to ARCH DEVELOPMENT CORPORATION reassignment ARCH DEVELOPMENT CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOSS, JOSEPH F., MOSS, JONATHAN, ROIZEN, MICHAEL F., YUAN, CHUN-SU
Assigned to UR LABS, INC. reassignment UR LABS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DRELL, WILLIAM
Assigned to CHICAGO, THE UNIVERSITY OF reassignment CHICAGO, THE UNIVERSITY OF ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ARCH DEVELOPMENT CORPORATION
Priority to US10/779,129 priority patent/US20040162308A1/en
Priority to US10/962,729 priority patent/US20050048117A1/en
Assigned to PROGENICS PHARMACEUTICALS NEVADA, INC. reassignment PROGENICS PHARMACEUTICALS NEVADA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UR LABS, INC.
Priority to US12/333,912 priority patent/US20090312359A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system

Definitions

  • the present invention is directed at the treatment of certain side effects associated with the use of opioids as analgesics.
  • the present invention is directed at treating opioid-induced inhibition of gastrointestinal motility and constipation.
  • Opioids are effective analgesics. However, their use is associated with a number of undesirable side effects. One such effect is constipation. Opioid-induced changes in gastrointestinal motility are almost universal when these drugs are used to treat pain, and at times may limit their use, leaving the patient in pain. Common treatments of bulking agents and laxatives have limited efficacy and may be associated with side effects such as electrolyte imbalances.
  • opioid antagonists which cross the blood-brain-barrier, or which are administered directly into the central nervous system.
  • Opioid antagonists such as naltrexone and naloxone have been administered intramuscularly or orally to treat opioid induced side effects.
  • Naltrexone and naloxone are highly lipid soluble and rapidly diffuse across biological membranes, including the blood-brain barrier.
  • naltrexone, naloxone, nalmefene, and other opioid antagonists which may reverse many opioid side effects have a narrow therapeutic window before they are observed to reverse the desired analgesic effect of the opioid being used.
  • quaternary amine opioid antagonist derivatives such as methylnaltrexone, do not reduce the analgesic effect of opioids.
  • These quaternary amine opioid antagonist derivatives which have a relatively higher polarity and reduced lipid solubility when compared to the tertiary forms of the drugs, were specifically developed to not traverse the blood-brain barrier or to traverse it at a greatly reduced rate.
  • high levels of MNTX in the plasma can lead to undesirable side effects such as orthostatic hypotension.
  • oral medications are particularly desirable for the treatment of opioid-induced side effects.
  • FIG. 1A is a graph representing plasma concentrations of MNTX following administration of 6.4 mg/kg of uncoated MNTX.
  • FIG. 1B is a graph representing plasma concentrations of MNTX following administration of 6.4 mg/kg of enterically coated MNTX.
  • FIG. 1C is a graph representing plasma concentrations of MNTX following administration of 3.2 mg/kg of enterically coated MNTX.
  • FIG. 2 illustrates the reversal of morphine's effect on oral-cecal transit time following administration of 6.4 mg/kg of uncoated MNTX.
  • the darker line represents the average of all points of a given treatment.
  • FIG. 3 illustrates the reversal of morphine's effect on oral-cecal transit time and its decrease below baseline following administration of 6.4 mg/kg of enterically coated MNTX.
  • the darker line represents the average of all points of a given treatment.
  • FIG. 4 illustrates the reversal of morphine's effect on oral-cecal transit time following administration of 3.2 mg/kg of enterically coated MNTX.
  • the darker line represents the average of all points of a given treatment.
  • the present invention is directed at methods for preventing and treating opioid-induced inhibition of gut motility and constipation.
  • the method for preventing opioid-induced decrease gut, motility and constipation comprises administering enterically coated methylnaltrexone or other quaternary derivatives of noroxymorphone as disclosed in U.S. Pat. No. 4,176,186 to Goldberg et al. (herein incorporated by reference) to a patient prior to or simultaneously with the administration of an opioid.
  • the method for treating opioid-induced inhibition of gut motility and constipation comprises administering enterically coated methylnaltrexone or other quaternary derivatives of noroxymorphone to a patient after the onset of the side effect.
  • the present invention is directed to methods for preventing and treating opioid-induced constipation and changes in gut motility via the oral administration of an enteric coated quaternary derivatives of noroxymorphone (QDNM), particularly methylnaltrexone (MNTX).
  • QDNM quaternary derivatives of noroxymorphone
  • MNTX methylnaltrexone
  • Administration of non-enterically coated MNTX results in rapid absorption of MNTX through the stomach and early peak and sustained high levels of MNTX in the plasma.
  • an enteric coating on the QDNM designed to prevent dissolution and subsequent absorption of the drug in the stomach, would be expected to produce delayed elevation of plasma levels of the QDNM, and to produce a lower peak plasma level.
  • enterically coated MNTX has been found to result in substantially lower plasma levels as compared to non-enterically coated MNTX at the same dosage level, and surprisingly and unexpectedly resulted in enhanced efficacy in the reversal of opioid-induced decreases in gastrointestinal motility.
  • a significantly lower dose e.g., less than half the amount of coated MNTX can be used if enterically coated to achieve the same levels of relief of opioid-induced constipation.
  • reduced dosage levels of MNTX administered with an enteric coating results in exceedingly low peak and sustained plasma levels of MNTX, greatly reducing the potential adverse side effects of the MNTX. This novel improvement in the clinical indication for use of MNTX has led to an increased therapeutic index for this drug.
  • MNTX When used as a treatment for the opioid- and nonopioid-induced side effects of constipation and reduction of gastrointestinal motility, orally administered, particularly if enterically coated, MNTX or other quaternary derivatives of noroxymorphone provide prolonged relief of the side effects. MNTX has been demonstrated to have the ability to block the gastrointestinal effects of opioids on motility when administered intravenously or orally. The oral administration of non-enterically coated MNTX is associated with plasma levels with an early peak (20 min) and prolonged presence (half-life of about 3 hours after single dose of 6.4 mg/kg).
  • enteric coating surprisingly allows for equal or better efficacy despite lower plasma levels.
  • Idiopathic constipation i.e., that due to causes other than exogenous administration of opioids, may be mediated by opioid sensitive mechanisms.
  • Endogenous opioid receptors have been identified in the gut, and these receptors may modulate gut motility.
  • R is allyl or a related radical such as chlorallyl, cyclopropyl-methyl or propargyl
  • X is the anion of an acid, especially a chloride, bromide, iodide or methylsulfate anion.
  • the presently preferred quaternary derivative of noroxymorphone is methylnaltrexone.
  • Methylnaltrexone is a quaternary amine derivative of naltrexone.
  • Methylnaltrexone has been found to have only 2 to 4% of the opiate antagonistic activity of naltrexone in vivo due to its inability to pass the blood-brain-barrier and bind to the opiate receptors in the central nervous system.
  • Opioids are typically administered at a morphine equivalent dosage of: 0.005 to 0.15 mg/kg body weight for intrathecal administration; 0.05 to 1.0 mg/kg body weight for intravenous administration; 0.05 to 1.0 mg/kg body weight for intramuscular administration; 0.05 to 1.0 mg/kg body weight/hour for transmucosal or transdermal administration.
  • morphine equivalent dosage is meant representative doses of other opioids which equal one milligram of morphine, for example 10 mg meperidine, 1 mg methadone, and 80 ⁇ g fentanyl.
  • methylnaltrexone is administered at a dosage of: 0.1 to 40.0 mg/kg body weight for oral administration, including enteric coated methylnaltrexone.
  • the administration of the methylnaltrexone is preferably commenced prior to administration of the opioid to prevent opioid-induced inhibition of gastrointestinal motility or constipation. It is desirable to commence internal administration of methylnaltrexone about 20 minutes prior to administration of opioids in order to prevent these opioid-induced side effects. While the prevention of symptoms is preferred, methylnaltrexone administration may also be commenced after the administration of the opioid or after the onset of opioid induced symptoms as a treatment for those symptoms.
  • Methylnaltrexone is rapidly absorbed after oral administration from the stomach and bowel. Initial plasma levels of the drug are seen within 5-10 minutes of the administration of non-enteric coated compound. Addition of an enteric coating which prevents gastric absorption is associated with lower plasma levels of the methylnaltrexone. Surprisingly, the addition of an enteric coating (i.e., a coating which will prevent degradation or release in the stomach, but will release drug in the small and large bowel) enhances the efficacy of methylnaltrexone in the prevention of decreases in gut motility by intravenously administered opioids (morphine).
  • an enteric coating i.e., a coating which will prevent degradation or release in the stomach, but will release drug in the small and large bowel
  • the QDNM or MNTX is enterically coated and administered orally.
  • the QDNM or methylnaltrexone is formulated with pharmacologically acceptable binders to make a tablet or capsule with an enteric coating.
  • An enteric coating is one which remains intact during passage through the stomach, but dissolves and releases the contents of the tablet or capsule once it reaches the small intestine.
  • Most currently used enteric coatings are those which will not dissolve in low pH environments, but readily ionize when the pH rises to about 4 or 5, for example synthetic polymers such as polyacids having a pKa of 3 to 5.
  • the enteric coating may be made of any suitable composition. Suitable enteric coatings are described, for example, in U.S. Pat. No. 4,311,833 to Namikoshi, et al.; U.S. Pat. No. 4,377,568 to Chopra; U.S. Pat. No. 4,385,078 to Onda, et al.; U.S. Pat. No. 4,457,907 to Porter; U.S. Pat. Nos. 4,462,839 to McGinley, et al.; 4,518,433 to McGinley, et al.; U.S. Pat. No. 4,556,552 to Porter, et al.; U.S. Pat. No.
  • enteric coating compositions include alkyl and hydroxyalkyl celluloses and their aliphatic esters, e.g., methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxybutylcellulose, hydroxyethylethylcellulose, hydroxyprophymethylcellulose, hydroxybutylinethylcellulose, hydroxypropylcellulose phthalate, hydroxypropylmethylcellulose phthalate and hydroxypropylmethylcellulose acetate succinate; carboxyalkylcelluloses and their salts, e.g., carboxymethylethylcellulose; cellulose acetate phthalate; cellulose acetate trimellitate, polycarboxymethylene and its salts and derivatives; polyvinyl alcohol and its esters: polyvinyl acetate phthalate; polycarboxymethylene copolymer with sodium formaldehyde carboxylate; acrylic polymers and copolymers, e.g., methacrylic acid
  • enteric coatings include polyvinylacetate esters, e.g., polyvinyl acetate phthalate; alkyleneglycolether esters of copolymers such as partial ethylene glycol monomethylether ester of ethylacrylate-maleic anhydride copolymer or diethyleneglycol monomethylether ester of methylacrylate-maleic anhydride copolymer, N-butylacrylate-maleic anhydride copolymer, isobutylacrylate-maleic anhydride copolymer or ethylacrylate-maleic anhydride copolymer; and polypeptides resistant to degradation in the gastric environment, e.g., polyarginine and polylysine.
  • the presently preferred enteric coating comprises cellulose acetate phthalate.
  • the enteric coating material may be mixed with various excipients including plasticizers such as triethyl citrate, acetyl triethyl citrate, diethyl phthalate, dibutyl phthalate, dibutyl subacute, dibutyl tartrate, dibutyl maleate, dibutyl succinate and diethyl succinate and inert fillers such as chalk or pigments.
  • plasticizers such as triethyl citrate, acetyl triethyl citrate, diethyl phthalate, dibutyl phthalate, dibutyl subacute, dibutyl tartrate, dibutyl maleate, dibutyl succinate and diethyl succinate and inert fillers such as chalk or pigments.
  • composition and thickness of the enteric coating may be selected to dissolve immediately upon contact with the digestive juice of the intestine.
  • the composition and thickness of the external coating may be selected to be a time-release coating which dissolves over a selected period of time, as is well known in the art.
  • the amount of enteric coating depends on the particular enteric coating composition used and is preferably sufficient to substantially prevent the absorption of MNTX in the stomach:.
  • Hydroxyalkyl celluloses and their aliphatic esters, carboxyalkyl celluloses and their salts, polycarboxymethylene and its salts and derivatives, polyvinyl alcohol and its esters, polycarboxymethylene copolymer with sodium formaldehyde carboxylates, poly-vinylpyrrolidone, and polyethylene glycol and its esters can be applied as enteric coatings by first dissolving the compound in a minimum amount of water. Alcohol is then added to the point of incipient cloudiness. The mixture can then be applied by conventional techniques.
  • cellulose acetate phthalate may be accomplished by simply dissolving the cellulose acetate phthalate in a minimum amount of alcohol and then applying by conventional techniques.
  • Hydrogenated vegetable oils may be applied by first dissolving the oil in a minimal amount of a non-polymer solvent, such as methylene chloride, chloroform or carbon tetrachloride, then adding alcohol to the point of incipient cloudiness and then applying by conventional techniques.
  • the MNTX is coated with Eudragit L100 or S100, a methacrylic acid copolymer enteric coating, at a 50% coating level to provide stability at gastric pH and dissolution at gut pH per a US Pharmacopeia (USP) standard for enteric coatings.
  • USP US Pharmacopeia
  • methylnaltrexone is used as an example of a particularly effective QDNM. It is apparent that other QDNM's may be used as desired.
  • Oral methylnaltrexone whether enterically coated or uncoated, was shown to reverse the inhibitory effects of opioid administration on gastrointestinal motility as measured by oral-cecal transit time. As compared to non-enterically coated MNTX, however, treatment with enterically coated MNTX enhanced the efficacy of the drug at a lower dose while producing lower plasma levels of MNTX.
  • Subjects were divided into five treatment groups A-E. With the exception of subjects in Group A, who were given a placebo in place of morphine, all were given an intravenous dose of morphine at 0.05 mg/kg. Prior to morphine administration, subjects were given either a placebo or MNTX in various doses and formulations (see Table 1). The subjects in Group A and B were given a placebo in place of MNTX. Group C received uncoated MNTX at 6.4 mg/kg, Group D received enterically coated MNTX at 6.4 mg/kg active drug, and Group E received enterically coated MNTX at 3.2 mg/kg active drug. Table 1 shows the treatments for each group. TABLE 1 Group Treatment combination FIG.
  • Plasma levels of MNTX were measured following administration of morphine and MNTX or placebo several times over the duration of the six hour monitoring period, at the times shown in FIG. 1. Measurements of plasma and urine MNTX levels were determined by high performance liquid chromatography (HPLC) using the modified method originally reported by Kim et al. (1989) Chromatographia 28:359-63, herein incorporated by reference). Methylnaltrexone was separated from plasma by solid phase extraction (SPE). Plasma samples (100-500 ⁇ l) diluted in water with the internal standard (naltrexone) were passed through SPE columns. Prior to use, the columns were conditioned by methanol and washed with water.
  • HPLC high performance liquid chromatography
  • the analytes were eluted from the columns by the mixture of n-propanol and trifluoroacetic acid (25 mM) aqueous solution prepared in 2:1 proportion.
  • the eluate was evaporated to dryness in a stream of nitrogen at 55° C.
  • the residue was reconstituted in the mobile phase, filtered through a nylon HPLC syringe filter and subjected to HPLC analysis.
  • a Shimadzu Corporation (Kyoto, Japan) HPLC system was used. It consisted of the LC-10AD pump, SCL-10A system controller, and SIL-10A auto injector equipped with sample cooler. Used HPLC Analytical Column made by Phenomenex (Prodigy C8, Torrance, Calif.).
  • the electrochemical detector (ESA Coulochem, model 5100A) worked at the following settings: detector 1, +360 mV, detector 2+600 mV, guard cell +650 mV. Data were collected with the use of EZChrom 2-2 HPLC software.
  • the mobile phase consisted of 50 mM sodium acetate, 7.5% methanol at pH 4.2. The system was calibrated daily in the range of 5-100 ng/ml (3 point calibration). Practical limit of detection for plasma samples was approximately 2 ng/ml (100 pg/injection).
  • FIG. 1 shows the plasma levels of MNTX following the treatments in Groups C, D, and E.
  • MNTX plasma levels in Group C (given 6.4 mg/kg MNTX, uncoated) peaked at about 15 min. post-MNTX administration and remained at a roughly constant level (between about 35-50 ng/ml) for the duration of the study period (6 hours).
  • Group D given 6.4 mg/kg MNTX in an enterically coated formulation, exhibited a constant low plasma level of MNTX (under 10 ng/ml) for the duration of observation (see FIG. 1B).
  • Group E given 3.2 mg/kg MNTX in an enterically coated formulation, showed plasma levels of MNTX over the course of observation that were undetectable or at the lower limit of detection of the assay (see FIG. 1C).
  • Oral-cecal transit time was used as a measure of gut motility and propensity for constipation. Oral-cecal transit time was measured by the lactulose-breath hydrogen method.
  • Group A demonstrated normal transit times as previously described in the literature (Yuan et al. (1996) Clin. Pharmacol. Ther. 59:469-475; Yuan et al. (1997) Clin. Pharmacol. Ther. 61:467-475).
  • Group B had prolongation of their oral-cecal transit times by 50-100%, while Groups C (FIG. 2) and E (FIG. 4) had their transit times return to baseline levels.
  • Group D showed an obvious decrease in oral-cecal transit time (FIG. 3).
  • enterically coated MNTX provides the therapeutic effects on gastrointestinal motility of uncoated MNTX, but requires a lower dose of active drug and results in significantly reduced plasma levels of MNTX.
  • Patients provided with a dose of 6.4 mg/kg of uncoated MNTX had gut motility return to baseline following morphine administration (FIG. 2) and showed plasma MNTX levels of over 40 ng MNTX/ml, while patients given the same dose in an enterically coated formulation showed oral-cecal transit times below baseline levels (FIG. 3) and plasma MNTX levels under 10 ng/ml.
  • Enterically coated formulations of MNTX with one half the dose of active drug (3.2 mg/kg) were required to return oral-cecal transit times to normal without increasing gut motility. At this dosage, plasma levels of MNTX were negligible.
  • MNTX As with most drugs, it is desirable to maintain the lowest possible systemic levels of MNTX which are sufficient to provide the desired therapeutic effect. For example, elevated circulating levels of MNTX can result in orthostatic hypotension.
  • the present discovery provides an unexpected means to avoid such undesirable drug side effects by lowering the dose administered and subsequently minimizing circulating levels of the drug. Since endogenous and externally supplied opioid-induced inhibition of gastrointestinal motility and constipation is thought to result from opioid receptors located within the gastrointestinal tract, enterically coated MNTX or other QDNMs may provide a local administration of the drug that does not require a circulating level for effective prevention or treatment of symptoms. Thus, the amount and/or frequency of drug administered can be reduced.

Abstract

A method for preventing or treating opioid induced side effects including changes in gastrointestinal motility. The method comprises administering methylnaltrexone or another quaternary derivative of noroxymorphone to a patient prior to the administration of an opioid or after the onset of side effects induced by the administration of an opioid, wherein the methylnaltrexone or quaternary derivative is administered orally in an enterically coated form.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of Ser. No. 08/962,742, filed Nov. 3, 1997, the disclosure of which is herein incorporated by reference.[0001]
  • FIELD OF THE INVENTION
  • The present invention is directed at the treatment of certain side effects associated with the use of opioids as analgesics. In particular the present invention is directed at treating opioid-induced inhibition of gastrointestinal motility and constipation. [0002]
  • Partial funding of the work described herein was provided under M01 RR00055 awarded by the U.S. Public Health Service General Clinical Research Center, and the U.S. Government has certain rights in the invention. [0003]
  • BACKGROUND OF THE INVENTION
  • Opioids are effective analgesics. However, their use is associated with a number of undesirable side effects. One such effect is constipation. Opioid-induced changes in gastrointestinal motility are almost universal when these drugs are used to treat pain, and at times may limit their use, leaving the patient in pain. Common treatments of bulking agents and laxatives have limited efficacy and may be associated with side effects such as electrolyte imbalances. [0004]
  • One treatment for opioid side effects is the use of opioid antagonists which cross the blood-brain-barrier, or which are administered directly into the central nervous system. Opioid antagonists such as naltrexone and naloxone have been administered intramuscularly or orally to treat opioid induced side effects. Naltrexone and naloxone are highly lipid soluble and rapidly diffuse across biological membranes, including the blood-brain barrier. However, naltrexone, naloxone, nalmefene, and other opioid antagonists which may reverse many opioid side effects have a narrow therapeutic window before they are observed to reverse the desired analgesic effect of the opioid being used. [0005]
  • Many quaternary amine opioid antagonist derivatives, such as methylnaltrexone, do not reduce the analgesic effect of opioids. These quaternary amine opioid antagonist derivatives, which have a relatively higher polarity and reduced lipid solubility when compared to the tertiary forms of the drugs, were specifically developed to not traverse the blood-brain barrier or to traverse it at a greatly reduced rate. However, high levels of MNTX in the plasma can lead to undesirable side effects such as orthostatic hypotension. [0006]
  • It is desirable in the treatment of many conditions to have oral medications with prolonged effects. Such oral medications are particularly desirable for the treatment of opioid-induced side effects. [0007]
  • It is further desirable to develop a method for the prevention of opioid-induced and inhibition of gut motility constipation which does not counteract the analgesic effects of the opioid, or risk increased levels of pain. Ideally, such a treatment has few side effects either due to low drug toxicity or because administration of small amounts are effective and/or administration results in low circulating levels of the drug.[0008]
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1A is a graph representing plasma concentrations of MNTX following administration of 6.4 mg/kg of uncoated MNTX. [0009]
  • FIG. 1B is a graph representing plasma concentrations of MNTX following administration of 6.4 mg/kg of enterically coated MNTX. [0010]
  • FIG. 1C is a graph representing plasma concentrations of MNTX following administration of 3.2 mg/kg of enterically coated MNTX. [0011]
  • FIG. 2 illustrates the reversal of morphine's effect on oral-cecal transit time following administration of 6.4 mg/kg of uncoated MNTX. The darker line represents the average of all points of a given treatment. [0012]
  • FIG. 3 illustrates the reversal of morphine's effect on oral-cecal transit time and its decrease below baseline following administration of 6.4 mg/kg of enterically coated MNTX. The darker line represents the average of all points of a given treatment. [0013]
  • FIG. 4 illustrates the reversal of morphine's effect on oral-cecal transit time following administration of 3.2 mg/kg of enterically coated MNTX. The darker line represents the average of all points of a given treatment.[0014]
  • SUMMARY OF THE INVENTION
  • The present invention is directed at methods for preventing and treating opioid-induced inhibition of gut motility and constipation. [0015]
  • The method for preventing opioid-induced decrease gut, motility and constipation comprises administering enterically coated methylnaltrexone or other quaternary derivatives of noroxymorphone as disclosed in U.S. Pat. No. 4,176,186 to Goldberg et al. (herein incorporated by reference) to a patient prior to or simultaneously with the administration of an opioid. [0016]
  • The method for treating opioid-induced inhibition of gut motility and constipation comprises administering enterically coated methylnaltrexone or other quaternary derivatives of noroxymorphone to a patient after the onset of the side effect. [0017]
  • DETAILED DESCRIPTION
  • The present invention is directed to methods for preventing and treating opioid-induced constipation and changes in gut motility via the oral administration of an enteric coated quaternary derivatives of noroxymorphone (QDNM), particularly methylnaltrexone (MNTX). Administration of non-enterically coated MNTX results in rapid absorption of MNTX through the stomach and early peak and sustained high levels of MNTX in the plasma. However, an enteric coating on the QDNM, designed to prevent dissolution and subsequent absorption of the drug in the stomach, would be expected to produce delayed elevation of plasma levels of the QDNM, and to produce a lower peak plasma level. Suprisingly, however, administration of enterically coated MNTX has been found to result in substantially lower plasma levels as compared to non-enterically coated MNTX at the same dosage level, and surprisingly and unexpectedly resulted in enhanced efficacy in the reversal of opioid-induced decreases in gastrointestinal motility. In fact, it has been found that as compared to non-enterically coated MNTX, a significantly lower dose, e.g., less than half the amount of coated MNTX can be used if enterically coated to achieve the same levels of relief of opioid-induced constipation. Moreover, such reduced dosage levels of MNTX administered with an enteric coating results in exceedingly low peak and sustained plasma levels of MNTX, greatly reducing the potential adverse side effects of the MNTX. This novel improvement in the clinical indication for use of MNTX has led to an increased therapeutic index for this drug. [0018]
  • When used as a treatment for the opioid- and nonopioid-induced side effects of constipation and reduction of gastrointestinal motility, orally administered, particularly if enterically coated, MNTX or other quaternary derivatives of noroxymorphone provide prolonged relief of the side effects. MNTX has been demonstrated to have the ability to block the gastrointestinal effects of opioids on motility when administered intravenously or orally. The oral administration of non-enterically coated MNTX is associated with plasma levels with an early peak (20 min) and prolonged presence (half-life of about 3 hours after single dose of 6.4 mg/kg). [0019]
  • Furthermore, for treatment or prevention of constipation and delayed gastrointestinal emptying, whether caused by extrinsic or endogenous opioids, enteric coating surprisingly allows for equal or better efficacy despite lower plasma levels. Idiopathic constipation, i.e., that due to causes other than exogenous administration of opioids, may be mediated by opioid sensitive mechanisms. Endogenous opioid receptors have been identified in the gut, and these receptors may modulate gut motility. Thus, administration of an opioid antagonist with peripheral action, such a methylnaltrexone or other quaternary derivatives of noroxymorphone, would block the effects of endogenous opioids. [0020]
  • Quaternary derivatives of noroxymorphone are described in full in Goldberg et al., (supra), and in general are represented by the formula: [0021]
    Figure US20030065003A1-20030403-C00001
  • wherein R is allyl or a related radical such as chlorallyl, cyclopropyl-methyl or propargyl, and X is the anion of an acid, especially a chloride, bromide, iodide or methylsulfate anion. [0022]
  • The presently preferred quaternary derivative of noroxymorphone is methylnaltrexone. Methylnaltrexone is a quaternary amine derivative of naltrexone. Methylnaltrexone has been found to have only 2 to 4% of the opiate antagonistic activity of naltrexone in vivo due to its inability to pass the blood-brain-barrier and bind to the opiate receptors in the central nervous system. [0023]
  • Opioids are typically administered at a morphine equivalent dosage of: 0.005 to 0.15 mg/kg body weight for intrathecal administration; 0.05 to 1.0 mg/kg body weight for intravenous administration; 0.05 to 1.0 mg/kg body weight for intramuscular administration; 0.05 to 1.0 mg/kg body weight/hour for transmucosal or transdermal administration. By “morphine equivalent dosage” is meant representative doses of other opioids which equal one milligram of morphine, for example 10 mg meperidine, 1 mg methadone, and 80 μg fentanyl. [0024]
  • In accordance with the present invention, methylnaltrexone is administered at a dosage of: 0.1 to 40.0 mg/kg body weight for oral administration, including enteric coated methylnaltrexone. [0025]
  • The administration of the methylnaltrexone is preferably commenced prior to administration of the opioid to prevent opioid-induced inhibition of gastrointestinal motility or constipation. It is desirable to commence internal administration of methylnaltrexone about 20 minutes prior to administration of opioids in order to prevent these opioid-induced side effects. While the prevention of symptoms is preferred, methylnaltrexone administration may also be commenced after the administration of the opioid or after the onset of opioid induced symptoms as a treatment for those symptoms. [0026]
  • Methylnaltrexone is rapidly absorbed after oral administration from the stomach and bowel. Initial plasma levels of the drug are seen within 5-10 minutes of the administration of non-enteric coated compound. Addition of an enteric coating which prevents gastric absorption is associated with lower plasma levels of the methylnaltrexone. Surprisingly, the addition of an enteric coating (i.e., a coating which will prevent degradation or release in the stomach, but will release drug in the small and large bowel) enhances the efficacy of methylnaltrexone in the prevention of decreases in gut motility by intravenously administered opioids (morphine). [0027]
  • In a preferred embodiment for the prevention and/or treatment of constipation and inhibition of gastrointestinal motility, the QDNM or MNTX is enterically coated and administered orally. For oral administration, the QDNM or methylnaltrexone is formulated with pharmacologically acceptable binders to make a tablet or capsule with an enteric coating. An enteric coating is one which remains intact during passage through the stomach, but dissolves and releases the contents of the tablet or capsule once it reaches the small intestine. Most currently used enteric coatings are those which will not dissolve in low pH environments, but readily ionize when the pH rises to about 4 or 5, for example synthetic polymers such as polyacids having a pKa of 3 to 5. [0028]
  • The enteric coating may be made of any suitable composition. Suitable enteric coatings are described, for example, in U.S. Pat. No. 4,311,833 to Namikoshi, et al.; U.S. Pat. No. 4,377,568 to Chopra; U.S. Pat. No. 4,385,078 to Onda, et al.; U.S. Pat. No. 4,457,907 to Porter; U.S. Pat. Nos. 4,462,839 to McGinley, et al.; 4,518,433 to McGinley, et al.; U.S. Pat. No. 4,556,552 to Porter, et al.; U.S. Pat. No. 4,606,909 to Bechgaard et al.; U.S. Pat. No. 4,615,885 to Nakagame, et al.; U.S. Pat. No. 4,670,287 to Tsuji; U.S. Pat. No. 5,536,507 to Abramowitz, et al.; U.S. Pat. No. 5,567,423 to Ying, et al.; U.S. Pat. No. 5,591,433 to Michael, et al.; U.S. Pat. No. 5,597,564 to Ying, et al.; U.S. Pat. No. 5,609,871 to Michael, et al.; U.S. Pat. No. 5,614,222 to Kaplan; U.S. Pat. No. 5,626,875 to Rodes, et al.; and U.S. Pat. No. 5,629,001 to Michael, et al., all of which are incorporated herein by reference. [0029]
  • Preferred enteric coating compositions include alkyl and hydroxyalkyl celluloses and their aliphatic esters, e.g., methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxybutylcellulose, hydroxyethylethylcellulose, hydroxyprophymethylcellulose, hydroxybutylinethylcellulose, hydroxypropylcellulose phthalate, hydroxypropylmethylcellulose phthalate and hydroxypropylmethylcellulose acetate succinate; carboxyalkylcelluloses and their salts, e.g., carboxymethylethylcellulose; cellulose acetate phthalate; cellulose acetate trimellitate, polycarboxymethylene and its salts and derivatives; polyvinyl alcohol and its esters: polyvinyl acetate phthalate; polycarboxymethylene copolymer with sodium formaldehyde carboxylate; acrylic polymers and copolymers, e.g., methacrylic acid-methyl methacrylic acid copolymer and methacrylic acid-methyl acrylate copolymer; edible oils such as peanut oil, palm oil, olive oil and hydrogenated vegetable oils; polyvinylpyrrolidone; polyethylene glycol and its esters: natural products such as shellac, and zein. [0030]
  • Other preferred enteric coatings include polyvinylacetate esters, e.g., polyvinyl acetate phthalate; alkyleneglycolether esters of copolymers such as partial ethylene glycol monomethylether ester of ethylacrylate-maleic anhydride copolymer or diethyleneglycol monomethylether ester of methylacrylate-maleic anhydride copolymer, N-butylacrylate-maleic anhydride copolymer, isobutylacrylate-maleic anhydride copolymer or ethylacrylate-maleic anhydride copolymer; and polypeptides resistant to degradation in the gastric environment, e.g., polyarginine and polylysine. Other suitable coatings and methods to make and use such formulations are well known to those skilled in the art (see, e.g., [0031] Remington: The Science and Practice of Pharmacy, 19th ed. (1995) Mack Publishing Company, Easton, Pa.; herein incorporated by reference).
  • Mixtures of two or more of the above compounds may be used as desired. The presently preferred enteric coating comprises cellulose acetate phthalate. [0032]
  • The enteric coating material may be mixed with various excipients including plasticizers such as triethyl citrate, acetyl triethyl citrate, diethyl phthalate, dibutyl phthalate, dibutyl subacute, dibutyl tartrate, dibutyl maleate, dibutyl succinate and diethyl succinate and inert fillers such as chalk or pigments. [0033]
  • The composition and thickness of the enteric coating may be selected to dissolve immediately upon contact with the digestive juice of the intestine. Alternatively, the composition and thickness of the external coating may be selected to be a time-release coating which dissolves over a selected period of time, as is well known in the art. [0034]
  • The amount of enteric coating depends on the particular enteric coating composition used and is preferably sufficient to substantially prevent the absorption of MNTX in the stomach:. [0035]
  • Hydroxyalkyl celluloses and their aliphatic esters, carboxyalkyl celluloses and their salts, polycarboxymethylene and its salts and derivatives, polyvinyl alcohol and its esters, polycarboxymethylene copolymer with sodium formaldehyde carboxylates, poly-vinylpyrrolidone, and polyethylene glycol and its esters can be applied as enteric coatings by first dissolving the compound in a minimum amount of water. Alcohol is then added to the point of incipient cloudiness. The mixture can then be applied by conventional techniques. [0036]
  • Application of cellulose acetate phthalate may be accomplished by simply dissolving the cellulose acetate phthalate in a minimum amount of alcohol and then applying by conventional techniques. Hydrogenated vegetable oils may be applied by first dissolving the oil in a minimal amount of a non-polymer solvent, such as methylene chloride, chloroform or carbon tetrachloride, then adding alcohol to the point of incipient cloudiness and then applying by conventional techniques. [0037]
  • In a particularly preferred embodiment, the MNTX is coated with Eudragit L100 or S100, a methacrylic acid copolymer enteric coating, at a 50% coating level to provide stability at gastric pH and dissolution at gut pH per a US Pharmacopeia (USP) standard for enteric coatings. [0038]
  • In the above description, methylnaltrexone is used as an example of a particularly effective QDNM. It is apparent that other QDNM's may be used as desired. [0039]
  • The following Examples are intended to illustrate aspects of the invention and are not to be construed as limitations upon it. The methylnaltrexone used in the following Examples was manufactured by Mallinckrodt Pharmaceuticals, St. Louis, Mo. The Enteric Coating was manufactured by Coating Place, Inc., Verona, Wis. [0040]
  • EXAMPLE 1 Effects of Enterically Coated MNTX on Oral-Cecal Transit Time and Plasma Levels of MNTX
  • Oral methylnaltrexone, whether enterically coated or uncoated, was shown to reverse the inhibitory effects of opioid administration on gastrointestinal motility as measured by oral-cecal transit time. As compared to non-enterically coated MNTX, however, treatment with enterically coated MNTX enhanced the efficacy of the drug at a lower dose while producing lower plasma levels of MNTX. [0041]
  • Subjects were divided into five treatment groups A-E. With the exception of subjects in Group A, who were given a placebo in place of morphine, all were given an intravenous dose of morphine at 0.05 mg/kg. Prior to morphine administration, subjects were given either a placebo or MNTX in various doses and formulations (see Table 1). The subjects in Group A and B were given a placebo in place of MNTX. Group C received uncoated MNTX at 6.4 mg/kg, Group D received enterically coated MNTX at 6.4 mg/kg active drug, and Group E received enterically coated MNTX at 3.2 mg/kg active drug. Table 1 shows the treatments for each group. [0042]
    TABLE 1
    Group Treatment combination FIG.
    A placebo placebo
    B morphine (0.05 mg/kg) placebo
    C morphine (0.05 mg/kg) methylnaltrexone uncoated FIG.
    (6.4 mg/kg) 2
    D morphine (0.05 mg/kg) methylnaltrexone enteric coated FIG.
    (6.4 mg/kg active drug) 3
    E morphine (0.05 mg/kg) methylnaltrexone enteric coated FIG.
    (3.2 mg/kg active drug) 4
  • Plasma levels of MNTX were measured following administration of morphine and MNTX or placebo several times over the duration of the six hour monitoring period, at the times shown in FIG. 1. Measurements of plasma and urine MNTX levels were determined by high performance liquid chromatography (HPLC) using the modified method originally reported by Kim et al. (1989) [0043] Chromatographia 28:359-63, herein incorporated by reference). Methylnaltrexone was separated from plasma by solid phase extraction (SPE). Plasma samples (100-500 μl) diluted in water with the internal standard (naltrexone) were passed through SPE columns. Prior to use, the columns were conditioned by methanol and washed with water. The analytes were eluted from the columns by the mixture of n-propanol and trifluoroacetic acid (25 mM) aqueous solution prepared in 2:1 proportion. The eluate was evaporated to dryness in a stream of nitrogen at 55° C. The residue was reconstituted in the mobile phase, filtered through a nylon HPLC syringe filter and subjected to HPLC analysis. A Shimadzu Corporation (Kyoto, Japan) HPLC system was used. It consisted of the LC-10AD pump, SCL-10A system controller, and SIL-10A auto injector equipped with sample cooler. Used HPLC Analytical Column made by Phenomenex (Prodigy C8, Torrance, Calif.). The electrochemical detector (ESA Coulochem, model 5100A) worked at the following settings: detector 1, +360 mV, detector 2+600 mV, guard cell +650 mV. Data were collected with the use of EZChrom 2-2 HPLC software. The mobile phase consisted of 50 mM sodium acetate, 7.5% methanol at pH 4.2. The system was calibrated daily in the range of 5-100 ng/ml (3 point calibration). Practical limit of detection for plasma samples was approximately 2 ng/ml (100 pg/injection).
  • FIG. 1 shows the plasma levels of MNTX following the treatments in Groups C, D, and E. In FIG. 1A, MNTX plasma levels in Group C (given 6.4 mg/kg MNTX, uncoated) peaked at about 15 min. post-MNTX administration and remained at a roughly constant level (between about 35-50 ng/ml) for the duration of the study period (6 hours). Group D, given 6.4 mg/kg MNTX in an enterically coated formulation, exhibited a constant low plasma level of MNTX (under 10 ng/ml) for the duration of observation (see FIG. 1B). Group E, given 3.2 mg/kg MNTX in an enterically coated formulation, showed plasma levels of MNTX over the course of observation that were undetectable or at the lower limit of detection of the assay (see FIG. 1C). [0044]
  • Oral-cecal transit time was used as a measure of gut motility and propensity for constipation. Oral-cecal transit time was measured by the lactulose-breath hydrogen method. Group A demonstrated normal transit times as previously described in the literature (Yuan et al. (1996) [0045] Clin. Pharmacol. Ther. 59:469-475; Yuan et al. (1997) Clin. Pharmacol. Ther. 61:467-475). Group B had prolongation of their oral-cecal transit times by 50-100%, while Groups C (FIG. 2) and E (FIG. 4) had their transit times return to baseline levels. Group D showed an obvious decrease in oral-cecal transit time (FIG. 3).
  • As demonstrated in FIGS. [0046] 1-4, enterically coated MNTX provides the therapeutic effects on gastrointestinal motility of uncoated MNTX, but requires a lower dose of active drug and results in significantly reduced plasma levels of MNTX. Patients provided with a dose of 6.4 mg/kg of uncoated MNTX had gut motility return to baseline following morphine administration (FIG. 2) and showed plasma MNTX levels of over 40 ng MNTX/ml, while patients given the same dose in an enterically coated formulation showed oral-cecal transit times below baseline levels (FIG. 3) and plasma MNTX levels under 10 ng/ml. Enterically coated formulations of MNTX with one half the dose of active drug (3.2 mg/kg) were required to return oral-cecal transit times to normal without increasing gut motility. At this dosage, plasma levels of MNTX were negligible.
  • As with most drugs, it is desirable to maintain the lowest possible systemic levels of MNTX which are sufficient to provide the desired therapeutic effect. For example, elevated circulating levels of MNTX can result in orthostatic hypotension. The present discovery provides an unexpected means to avoid such undesirable drug side effects by lowering the dose administered and subsequently minimizing circulating levels of the drug. Since endogenous and externally supplied opioid-induced inhibition of gastrointestinal motility and constipation is thought to result from opioid receptors located within the gastrointestinal tract, enterically coated MNTX or other QDNMs may provide a local administration of the drug that does not require a circulating level for effective prevention or treatment of symptoms. Thus, the amount and/or frequency of drug administered can be reduced. [0047]
  • The preceding description and Examples are intended to be illustrative. Those skilled in the art to which the invention pertains will appreciate that alterations and changes in the described protocols may be practiced without departing from the meaning, spirit, and scope of this invention. Therefore, the foregoing description should be read consistent with and as support to the following claims, which are to have their fullest and fair scope. [0048]

Claims (17)

What is claimed is:
1. A method for preventing opioid induced inhibition of gastrointestinal motility comprising orally administering an enterically coated quaternary derivative of noroxymorphone to a patient prior to the administration of an opioid.
2. The method of claim 1 wherein the quaternary derivative is methylnaltrexone.
3. The method of claim 1 wherein the inhibition of gastrointestinal motility is manifested as constipation.
4. The method of claim 2 wherein the methylnaltrexone is administered at a dosage 0.1 to 40.0 mg of active drug per kg body weight.
5. The method of claim 4 wherein the methylnaltrexone is administered orally at a dosage of about 0.1 to about 10 mg/kg body weight.
6. The method of claim 2 wherein the methylnaltrexone is administered as an enterically coated tablet or capsule.
7. The method of claim 2 wherein the patient's plasma level of methylnaltrexone remains below 25 ng/ml.
8. A method for treating opioid induced inhibition of gastrointestinal motility comprising orally administering an enterically coated quaternary derivative of noroxymorphone to a patient subsequent to the administration of an opioid.
9. The method of claim 8 wherein the quaternary derivative is methlynaltrexone.
10. The method of claim 8 wherein the inhibition of gastrointestinal motility is manifested as constipation.
11. The method of claim 9 wherein the patient's plasma level of methylnaltrexone remains below 25 ng/ml.
12. The method of claim 9 wherein the methylnaltrexone is administered at a dosage 0.1 to 40.0 mg of active drug per kg body weight.
13. The method of claim 12 wherein the methylnaltrexone is administered orally at a dosage of about 0.1 to about 10 mg/kg body weight.
14. The method of claim 9 wherein the methylnaltrexone is administered as an enterically coated tablet or capsule.
15. The method of claim 8 wherein the constipation is induced by endogenous opioids.
16. The method of claim 2 wherein the enteric coating provides time release of the methylnaltrexone.
17. The method of claim 9 wherein the enteric coating provides time release of the methylnaltrexone.
US10/278,630 1997-11-03 2002-10-23 Use of methylnaltrexone and related compounds Abandoned US20030065003A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/278,630 US20030065003A1 (en) 1997-11-03 2002-10-23 Use of methylnaltrexone and related compounds
US10/779,129 US20040162308A1 (en) 1997-11-03 2004-02-12 Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US10/962,729 US20050048117A1 (en) 1997-11-03 2004-10-12 Use of methylnaltrexone and related compounds
US12/333,912 US20090312359A1 (en) 1997-11-03 2008-12-12 Use of methylnaltrexone and related compounds for treatment of gastrointestinal dysfunction induced by endogenous opioids

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/962,742 US5972954A (en) 1997-11-03 1997-11-03 Use of methylnaltrexone and related compounds
US09/120,703 US6274591B1 (en) 1997-11-03 1998-07-22 Use of methylnaltrexone and related compounds
US09/862,169 US6608075B2 (en) 1997-11-03 2001-05-21 Use of methylnaltrexone and related compounds
US10/278,630 US20030065003A1 (en) 1997-11-03 2002-10-23 Use of methylnaltrexone and related compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/862,169 Division US6608075B2 (en) 1997-11-03 2001-05-21 Use of methylnaltrexone and related compounds

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/779,129 Continuation US20040162308A1 (en) 1997-11-03 2004-02-12 Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US10/962,729 Continuation US20050048117A1 (en) 1997-11-03 2004-10-12 Use of methylnaltrexone and related compounds

Publications (1)

Publication Number Publication Date
US20030065003A1 true US20030065003A1 (en) 2003-04-03

Family

ID=26818669

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/120,703 Expired - Lifetime US6274591B1 (en) 1997-11-03 1998-07-22 Use of methylnaltrexone and related compounds
US09/862,169 Expired - Lifetime US6608075B2 (en) 1997-11-03 2001-05-21 Use of methylnaltrexone and related compounds
US10/278,630 Abandoned US20030065003A1 (en) 1997-11-03 2002-10-23 Use of methylnaltrexone and related compounds
US10/779,129 Abandoned US20040162308A1 (en) 1997-11-03 2004-02-12 Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US10/962,729 Abandoned US20050048117A1 (en) 1997-11-03 2004-10-12 Use of methylnaltrexone and related compounds
US12/333,912 Abandoned US20090312359A1 (en) 1997-11-03 2008-12-12 Use of methylnaltrexone and related compounds for treatment of gastrointestinal dysfunction induced by endogenous opioids

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/120,703 Expired - Lifetime US6274591B1 (en) 1997-11-03 1998-07-22 Use of methylnaltrexone and related compounds
US09/862,169 Expired - Lifetime US6608075B2 (en) 1997-11-03 2001-05-21 Use of methylnaltrexone and related compounds

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/779,129 Abandoned US20040162308A1 (en) 1997-11-03 2004-02-12 Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US10/962,729 Abandoned US20050048117A1 (en) 1997-11-03 2004-10-12 Use of methylnaltrexone and related compounds
US12/333,912 Abandoned US20090312359A1 (en) 1997-11-03 2008-12-12 Use of methylnaltrexone and related compounds for treatment of gastrointestinal dysfunction induced by endogenous opioids

Country Status (5)

Country Link
US (6) US6274591B1 (en)
EP (1) EP1047426B1 (en)
AU (1) AU758416B2 (en)
CA (1) CA2312234C (en)
WO (1) WO1999022737A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040162308A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US20040162307A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds to induce laxation in chronic opioid users
US20040259899A1 (en) * 2003-04-08 2004-12-23 Sanghvi Suketu P. Combination therapy for constipation
US20040266806A1 (en) * 2003-04-08 2004-12-30 Sanghvi Suketu P. Pharmaceutical formulation
US20050004155A1 (en) * 2003-04-08 2005-01-06 Boyd Thomas A. Use of methylnaltrexone to treat irritable bowel syndrome
US20060205753A1 (en) * 2005-01-20 2006-09-14 Israel Robert J Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction
US20060258696A1 (en) * 2005-03-07 2006-11-16 Jonathan Moss Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US20070099946A1 (en) * 2005-05-25 2007-05-03 Doshan Harold D Synthesis of R-N-methylnaltrexone
US20080064743A1 (en) * 2006-09-08 2008-03-13 Wyeth Dry powder compound formulations and uses thereof
US20080194611A1 (en) * 2005-06-03 2008-08-14 Alverdy John C Modulation of Cell Barrier Dysfunction
US20080274119A1 (en) * 2005-03-07 2008-11-06 The University Of Chicago Use of Opioid Antagonists to Attenuate Endothelial Cell Proliferation and Migration
US7563899B2 (en) 2005-05-25 2009-07-21 Progenics Pharmaceuticals, Inc. (S)-N-methylnaltrexone
US20100099699A1 (en) * 2007-03-29 2010-04-22 Wyeth Peripheral opioid receptor antagonists and uses thereof
US20100120813A1 (en) * 2008-09-30 2010-05-13 Wyeth Peripheral opioid receptor antagonists and uses thereof
US20100305323A1 (en) * 2007-03-29 2010-12-02 Smolenskaya Valeriya N Crystal forms of (r)-n-methylnaltrexone bromide and uses thereof
US20110021551A1 (en) * 2008-03-21 2011-01-27 Jonathan Moss TREATMENT WITH OPIOID ANTAGONISTS AND mTOR INHIBITORS
US20110100099A1 (en) * 2008-02-06 2011-05-05 Progenics Pharmaceuticals, Inc. Preparation and use of (r),(r)-2,2'-bis-methylnaltrexone
US20110190331A1 (en) * 2007-03-29 2011-08-04 Avey Alfred A Peripheral opioid receptor antagonists and uses thereof
US8518962B2 (en) 2005-03-07 2013-08-27 The University Of Chicago Use of opioid antagonists
US9662325B2 (en) 2005-03-07 2017-05-30 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US10307417B2 (en) 2010-03-11 2019-06-04 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6559158B1 (en) * 1997-11-03 2003-05-06 Ur Labs, Inc. Use of methylnaltrexone and related compounds to treat chronic opioid use side affects
US6375957B1 (en) 1997-12-22 2002-04-23 Euro-Celtique, S.A. Opioid agonist/opioid antagonist/acetaminophen combinations
AU773642C (en) 1997-12-22 2006-04-06 Mundipharma Pty Limited Opioid agonist/antagonist combinations
CA2380524A1 (en) * 1999-08-25 2001-03-01 Barrett R. Cooper Compositions and methods for treating opiate intolerance
US6451806B2 (en) 1999-09-29 2002-09-17 Adolor Corporation Methods and compositions involving opioids and antagonists thereof
DK1225897T3 (en) * 1999-11-01 2005-01-10 John Rhodes Composition for the treatment of constipation and irritable colon
US6469030B2 (en) 1999-11-29 2002-10-22 Adolor Corporation Methods for the treatment and prevention of ileus
WO2001042207A2 (en) * 1999-11-29 2001-06-14 Adolor Corporation Novel methods for the treatment and prevention of ileus
PT2092936E (en) 2000-02-08 2013-06-20 Euro Celtique Sa Tamper-resistant oral opioid agonist formulations
DE10059415A1 (en) 2000-11-30 2002-06-06 Gruenenthal Gmbh Use of weak opioids and mixed opioid agonists / antagonists for the treatment of urinary incontinence
CA2446550C (en) 2001-05-11 2012-03-06 Endo Pharmaceuticals, Inc. Abuse-resistant controlled-release opioid dosage form
US20030022909A1 (en) * 2001-06-05 2003-01-30 University Of Chicago Use of methylnaltrexone to treat immune suppression
DE60230632D1 (en) 2001-07-18 2009-02-12 Euro Celtique Sa PHARMACEUTICAL COMBINATIONS OF OXYCODONE AND NALOXONE
DE60232417D1 (en) 2001-08-06 2009-07-02 Euro Celtique Sa OPIOID AGONIST FORMULATIONS WITH FREEZER AND SEQUESTRATED ANTAGONIST
CA2475305A1 (en) * 2002-02-04 2004-02-19 Jonathan Moss Use of methylnaltrexone in treating gastrointestinal dysfunction in equines
CN102813654A (en) 2002-04-05 2012-12-12 欧洲凯尔蒂克公司 Matrix for sustained, invariant and independent release of active compounds
SI1894562T1 (en) * 2002-08-15 2011-04-29 Euro Celtique Sa Pharmaceutical compositions comprising an opioid antagonist
MY135852A (en) 2003-04-21 2008-07-31 Euro Celtique Sa Pharmaceutical products
CA2536816A1 (en) * 2003-08-12 2005-03-03 Endo Pharmaceuticals, Inc. Method for deterring abuse of opioids by combination with non-release formulation of emetic
MXPA06003392A (en) * 2003-09-25 2006-06-08 Euro Celtique Sa Pharmaceutical combinations of hydrocodone and naltrexone.
TW200633721A (en) * 2004-12-14 2006-10-01 Shionogi & Co A pharmaceutical composition for treating constipation
EP1702558A1 (en) 2005-02-28 2006-09-20 Euro-Celtique S.A. Method and device for the assessment of bowel function
US20070185145A1 (en) * 2006-02-03 2007-08-09 Royds Robert B Pharmaceutical composition containing a central opioid agonist, a central opioid antagonist, and a peripheral opioid antagonist, and method for making the same
US20080027579A1 (en) * 2006-07-31 2008-01-31 Van Der Hoop Roland Gerritsen Dosage limiting medication dispensing method and apparatus
TW200815451A (en) * 2006-08-04 2008-04-01 Wyeth Corp 6-carboxy-normorphinan derivatives, synthesis and uses thereof
WO2009009292A2 (en) * 2007-07-11 2009-01-15 Mallinckrodt Inc. Crystalline forms of naltrexone methobromide
WO2009051824A2 (en) * 2007-10-18 2009-04-23 Aiko Biotechnology Combination analgesic employing opioid and neutral antagonist
US8748448B2 (en) 2007-10-18 2014-06-10 Aiko Biotechnology Combination analgesic employing opioid agonist and neutral antagonist
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
SI2405915T1 (en) 2009-03-10 2019-03-29 Euro-Celtique S.A. Immediate release pharmaceutical compositions comprising oxycodone and naloxone
US8987289B2 (en) 2012-12-14 2015-03-24 Trevi Therapeutics, Inc. Methods for treating pruritus
US8637538B1 (en) 2012-12-14 2014-01-28 Trevi Therapeutics, Inc. Methods for treatment of pruritis
US20140179727A1 (en) 2012-12-14 2014-06-26 Trevi Therapeutics, Inc. Methods for treating pruritus
JP2016525138A (en) 2013-07-23 2016-08-22 ユーロ−セルティーク エス.エイ. Oxycodone and naloxone combination for use in the treatment of pain in patients suffering from diseases that cause pain and intestinal dysbiosis and / or diseases that increase the risk for intestinal bacterial transfer
CA3002137A1 (en) 2014-10-17 2016-04-21 Salix Pharmaceuticals, Inc. Use of methylnaltrexone to attenuate tumor progression
CN112703000A (en) 2018-07-23 2021-04-23 特雷维治疗股份有限公司 Treatment of chronic cough, shortness of breath and dyspnea
CA3137488C (en) 2019-05-07 2023-10-31 Bausch Health Ireland Limited Liquid oral dosage formulations of methylnaltrexone
CA3177250A1 (en) 2020-05-02 2021-11-11 Bausch Health Ireland Limited Methods of reducing mortality risk in subjects suffering from an underlying disease or condition by administration of methylnaltrexone

Citations (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4311833A (en) * 1979-03-06 1982-01-19 Daicel Chemical Industries Ltd. Process for preparing ethylcarboxymethylcellulose
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
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
US4385078A (en) * 1978-09-04 1983-05-24 Shin-Etsu Chemical Co., Ltd. Method for providing enteric coating on solid dosage forms and aqueous compositions therefor
US4457907A (en) * 1982-08-05 1984-07-03 Clear Lake Development Group Composition and method for protecting a therapeutic drug
US4462839A (en) * 1983-06-16 1984-07-31 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4518433A (en) * 1982-11-08 1985-05-21 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4556552A (en) * 1983-09-19 1985-12-03 Colorcon, Inc. Enteric film-coating compositions
US4606909A (en) * 1981-11-20 1986-08-19 A/S Alfred Benzon Pharmaceutical multiple-units formulation
US4615885A (en) * 1983-11-01 1986-10-07 Terumo Kabushiki Kaisha Pharmaceutical composition containing urokinase
US4670287A (en) * 1985-07-30 1987-06-02 Washu Kirai Kogyo Kabushiki Kaisha Method of film-coating hard capsules
US4857833A (en) * 1987-08-27 1989-08-15 Teradyne, Inc. Diagnosis of faults on circuit board
US4888346A (en) * 1986-10-07 1989-12-19 Bernard Bihari Method for the treatment of persons infected with HTLV-III (AIDS) virus
US4965269A (en) * 1989-12-20 1990-10-23 Ab Hassle Therapeutically active chloro substituted benzimidazoles
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
US5202159A (en) * 1990-12-27 1993-04-13 Standard Chemical & Pharmaceutical Corp., Ltd. Preparation method of microdispersed tablet formulation of spray-dried sodium diclofenac enteric-coated microcapsules
US5391372A (en) * 1993-06-28 1995-02-21 Campbell; Elizabeth Methods of treating colic and founder in horses
US5426112A (en) * 1984-04-09 1995-06-20 Scully, Scott, Murphy & Presser, P.C. Growth regulation and related applications of opioid antagonists
US5536507A (en) * 1994-06-24 1996-07-16 Bristol-Myers Squibb Company Colonic drug delivery system
US5567423A (en) * 1986-08-28 1996-10-22 Enzacor Properties, Ltd. Animal growth promotant
US5591433A (en) * 1991-06-21 1997-01-07 University Of Cincinnati Oral administration of immunologically active biomolecules and other therapeutic proteins
US5597564A (en) * 1986-08-28 1997-01-28 Enzacor Properties Limited Method of administering a microgranular preparation to the intestinal region of animals
US5609871A (en) * 1991-06-21 1997-03-11 Michael; J. Gabriel Oral administration of therapeutic proteins for treatment of infectious disease
US5614222A (en) * 1994-10-25 1997-03-25 Kaplan; Milton R. Stable aqueous drug suspensions and methods for preparation thereof
US5614219A (en) * 1991-12-05 1997-03-25 Alfatec-Pharma Gmbh Oral administration form for peptide pharmaceutical substances, in particular insulin
US5626875A (en) * 1995-02-01 1997-05-06 Esteve Quimica, S.A. Stabilized galenic formulations comprising an acid labile benzimidazole compound and its preparation
US5656290A (en) * 1993-02-26 1997-08-12 The Procter & Gamble Company Bisacodyl dosage form with multiple enteric polymer coatings for colonic delivery
US5804595A (en) * 1995-12-05 1998-09-08 Regents Of The University Of Minnesota Kappa opioid receptor agonists
US5866154A (en) * 1994-10-07 1999-02-02 The Dupont Merck Pharmaceutical Company Stabilized naloxone formulations
US5981185A (en) * 1994-05-05 1999-11-09 Beckman Coulter, Inc. Oligonucleotide repeat arrays
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US20010036951A1 (en) * 1999-11-29 2001-11-01 Farrar John J. Novel methods for the treatment and prevention of ileus
US6353004B1 (en) * 1997-07-14 2002-03-05 Adolor Coporation Peripherally acting anti-pruritic opiates
US20020064771A1 (en) * 2000-04-07 2002-05-30 Weidong Zhong HCV replicase complexes
US6455537B1 (en) * 1999-08-25 2002-09-24 Barrett R. Cooper Methods for treating opiate intolerance
US20030026801A1 (en) * 2000-06-22 2003-02-06 George Weiner Methods for enhancing antibody-induced cell lysis and treating cancer
US20030187010A1 (en) * 1997-11-03 2003-10-02 Foss Joseph F. Use of methylnaltrexone and related compounds to treat chronic opioid use side effects
US20040162307A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds to induce laxation in chronic opioid users
US20040162308A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US20040259899A1 (en) * 2003-04-08 2004-12-23 Sanghvi Suketu P. Combination therapy for constipation
US20040266806A1 (en) * 2003-04-08 2004-12-30 Sanghvi Suketu P. Pharmaceutical formulation
US20050004155A1 (en) * 2003-04-08 2005-01-06 Boyd Thomas A. Use of methylnaltrexone to treat irritable bowel syndrome
US20050124885A1 (en) * 2003-10-29 2005-06-09 Vuesonix Sensors, Inc. Method and apparatus for determining an ultrasound fluid flow centerline
US20060205753A1 (en) * 2005-01-20 2006-09-14 Israel Robert J Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US222911A (en) * 1879-12-23 Improvement in book-cases
US2003A (en) * 1841-03-12 Improvement in horizontal windivhlls
US1047726A (en) * 1912-03-12 1912-12-17 Anthony J Yoggerst Pipe-cleaner.
US2625457A (en) * 1951-07-26 1953-01-13 Bernard J Baecher Course recorder
US4176186A (en) 1978-07-28 1979-11-27 Boehringer Ingelheim Gmbh Quaternary derivatives of noroxymorphone which relieve intestinal immobility
US4987136A (en) 1982-03-16 1991-01-22 The Rockefeller University Method for controlling gastrointestinal dysmotility
JPS59500418A (en) 1982-03-16 1984-03-15 ザ ロツクフエラ− ユニバ−シテイ How to recover from gastrointestinal dysfunction
US4861781A (en) * 1986-03-07 1989-08-29 The 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
US4719215A (en) * 1986-03-07 1988-01-12 University Of Chicago Quaternary derivatives of noroxymorphone which relieve nausea and emesis
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
CA1315689C (en) 1987-09-03 1993-04-06 Leon I. Goldberg Quarternary derivatives of noroxymorphone which relieve nausea and emesis
EP0352361A1 (en) 1988-07-29 1990-01-31 The Rockefeller University Method of treating patients suffering from chronic pain or chronic cough
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
US5270328A (en) 1991-03-29 1993-12-14 Eli Lilly And Company Peripherally selective piperidine opioid antagonists
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
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
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
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
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
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
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
US5965161A (en) 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
DE69709646T2 (en) 1996-03-12 2002-08-14 Alza Corp COMPOSITION AND DOSAGE WITH AN OPIOID ANTAGONIST
DE19651551C2 (en) 1996-12-11 2000-02-03 Klinge Co Chem Pharm Fab Opioid antagonist-containing galenic formulation
US5972954A (en) 1997-11-03 1999-10-26 Arch Development Corporation Use of methylnaltrexone and related compounds
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
US6451806B2 (en) 1999-09-29 2002-09-17 Adolor Corporation Methods and compositions involving opioids and antagonists thereof
WO2001041705A2 (en) 1999-11-29 2001-06-14 Adolor Corporation Novel methods for the treatment and prevention of dizziness and pruritus
DK1244447T3 (en) 1999-11-29 2007-04-23 Adolor Corp New methods and compositions containing opioids and their antagonists
WO2001042207A2 (en) 1999-11-29 2001-06-14 Adolor Corporation Novel methods for the treatment and prevention of ileus
CA2408106A1 (en) 2000-05-05 2001-11-15 Pain Therapeutics, Inc. Opioid antagonist compositions and dosage forms
US20030022909A1 (en) * 2001-06-05 2003-01-30 University Of Chicago Use of methylnaltrexone to treat immune suppression
PT1436012T (en) * 2001-10-18 2018-03-27 Nektar Therapeutics Polymer conjugates of opioid antagonists
US20030191147A1 (en) * 2002-04-09 2003-10-09 Barry Sherman Opioid antagonist compositions and dosage forms

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4385078A (en) * 1978-09-04 1983-05-24 Shin-Etsu Chemical Co., Ltd. Method for providing enteric coating on solid dosage forms and aqueous compositions therefor
US4311833A (en) * 1979-03-06 1982-01-19 Daicel Chemical Industries Ltd. Process for preparing ethylcarboxymethylcellulose
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
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
US4606909A (en) * 1981-11-20 1986-08-19 A/S Alfred Benzon Pharmaceutical multiple-units formulation
US4457907A (en) * 1982-08-05 1984-07-03 Clear Lake Development Group Composition and method for protecting a therapeutic drug
US4518433A (en) * 1982-11-08 1985-05-21 Fmc Corporation Enteric coating for pharmaceutical dosage forms
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
US4615885A (en) * 1983-11-01 1986-10-07 Terumo Kabushiki Kaisha Pharmaceutical composition containing urokinase
US5426112A (en) * 1984-04-09 1995-06-20 Scully, Scott, Murphy & Presser, P.C. Growth regulation and related applications of opioid antagonists
US4670287A (en) * 1985-07-30 1987-06-02 Washu Kirai Kogyo Kabushiki Kaisha Method of film-coating hard capsules
US5597564A (en) * 1986-08-28 1997-01-28 Enzacor Properties Limited Method of administering a microgranular preparation to the intestinal region of animals
US5567423A (en) * 1986-08-28 1996-10-22 Enzacor Properties, Ltd. Animal growth promotant
US4888346A (en) * 1986-10-07 1989-12-19 Bernard Bihari Method for the treatment of persons infected with HTLV-III (AIDS) virus
US4857833A (en) * 1987-08-27 1989-08-15 Teradyne, Inc. Diagnosis of faults on circuit board
US4965269A (en) * 1989-12-20 1990-10-23 Ab Hassle Therapeutically active chloro substituted benzimidazoles
US5202159A (en) * 1990-12-27 1993-04-13 Standard Chemical & Pharmaceutical Corp., Ltd. Preparation method of microdispersed tablet formulation of spray-dried sodium diclofenac enteric-coated microcapsules
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
US5609871A (en) * 1991-06-21 1997-03-11 Michael; J. Gabriel Oral administration of therapeutic proteins for treatment of infectious disease
US5591433A (en) * 1991-06-21 1997-01-07 University Of Cincinnati Oral administration of immunologically active biomolecules and other therapeutic proteins
US5629001A (en) * 1991-06-21 1997-05-13 University Of Cincinnati Oral administration of therapeutic proteins for treatment of infectious disease
US5614219A (en) * 1991-12-05 1997-03-25 Alfatec-Pharma Gmbh Oral administration form for peptide pharmaceutical substances, in particular insulin
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
US5981185A (en) * 1994-05-05 1999-11-09 Beckman Coulter, Inc. Oligonucleotide repeat arrays
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
US5626875A (en) * 1995-02-01 1997-05-06 Esteve Quimica, S.A. Stabilized galenic formulations comprising an acid labile benzimidazole compound and its preparation
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US5804595A (en) * 1995-12-05 1998-09-08 Regents Of The University Of Minnesota Kappa opioid receptor agonists
US6353004B1 (en) * 1997-07-14 2002-03-05 Adolor Coporation Peripherally acting anti-pruritic opiates
US20040162306A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnal trexone and related compounds to treat constipation in chronic opioid users
US20040167148A1 (en) * 1997-11-03 2004-08-26 Foss Joseph F. Oral use of methylnaltrexone and related compounds to treat constipation in chronic opioid users
US20040167147A1 (en) * 1997-11-03 2004-08-26 Foss Joseph F. Oral use of methylnaltrexone and related compounds to induce laxation in chronic opioid users
US20040162308A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US20050048117A1 (en) * 1997-11-03 2005-03-03 Foss Joseph F. Use of methylnaltrexone and related compounds
US20030187010A1 (en) * 1997-11-03 2003-10-02 Foss Joseph F. Use of methylnaltrexone and related compounds to treat chronic opioid use side effects
US20040162307A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds to induce laxation in chronic opioid users
US6455537B1 (en) * 1999-08-25 2002-09-24 Barrett R. Cooper Methods for treating opiate intolerance
US6469030B2 (en) * 1999-11-29 2002-10-22 Adolor Corporation Methods for the treatment and prevention of ileus
US20010036951A1 (en) * 1999-11-29 2001-11-01 Farrar John J. Novel methods for the treatment and prevention of ileus
US20020064771A1 (en) * 2000-04-07 2002-05-30 Weidong Zhong HCV replicase complexes
US20030026801A1 (en) * 2000-06-22 2003-02-06 George Weiner Methods for enhancing antibody-induced cell lysis and treating cancer
US20040259899A1 (en) * 2003-04-08 2004-12-23 Sanghvi Suketu P. Combination therapy for constipation
US20040266806A1 (en) * 2003-04-08 2004-12-30 Sanghvi Suketu P. Pharmaceutical formulation
US20050004155A1 (en) * 2003-04-08 2005-01-06 Boyd Thomas A. Use of methylnaltrexone to treat irritable bowel syndrome
US20050124885A1 (en) * 2003-10-29 2005-06-09 Vuesonix Sensors, Inc. Method and apparatus for determining an ultrasound fluid flow centerline
US20060205753A1 (en) * 2005-01-20 2006-09-14 Israel Robert J Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040162308A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds for treatment of constipation caused by endogenous opioids
US20040162307A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnaltrexone and related compounds to induce laxation in chronic opioid users
US20040162306A1 (en) * 1997-11-03 2004-08-19 Foss Joseph F. Use of methylnal trexone and related compounds to treat constipation in chronic opioid users
US20040167148A1 (en) * 1997-11-03 2004-08-26 Foss Joseph F. Oral use of methylnaltrexone and related compounds to treat constipation in chronic opioid users
US20100087472A1 (en) * 1997-11-03 2010-04-08 Foss Joseph F Use of methylnaltrexone and related compound to treat constipation in chronic opioid users
US20050048117A1 (en) * 1997-11-03 2005-03-03 Foss Joseph F. Use of methylnaltrexone and related compounds
US8552025B2 (en) 2003-04-08 2013-10-08 Progenics Pharmaceuticals, Inc. Stable methylnaltrexone preparation
US20040266806A1 (en) * 2003-04-08 2004-12-30 Sanghvi Suketu P. Pharmaceutical formulation
US20050004155A1 (en) * 2003-04-08 2005-01-06 Boyd Thomas A. Use of methylnaltrexone to treat irritable bowel syndrome
US20040259899A1 (en) * 2003-04-08 2004-12-23 Sanghvi Suketu P. Combination therapy for constipation
US9669096B2 (en) 2003-04-08 2017-06-06 Progenics Pharmaceuticals, Inc. Stable pharmaceutical formulations of methylnaltrexone
US20100261745A1 (en) * 2003-04-08 2010-10-14 Progenics Pharmaceuticals, Inc. Pharmaceutical formulation
US20100261746A1 (en) * 2003-04-08 2010-10-14 Progenics Pharmaceuticals, Inc. Pharmaceutical formulation
US20100261744A1 (en) * 2003-04-08 2010-10-14 Progenics Pharmaceuticals, Inc. Pharmaceutical formulation
US10376584B2 (en) 2003-04-08 2019-08-13 Progenics Pharmaceuticals, Inc. Stable pharmaceutical formulations of methylnaltrexone
US20060205753A1 (en) * 2005-01-20 2006-09-14 Israel Robert J Use of methylnaltrexone and related compounds to treat post-operative gastrointestinal dysfunction
US9662390B2 (en) 2005-03-07 2017-05-30 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US8524731B2 (en) 2005-03-07 2013-09-03 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US8518962B2 (en) 2005-03-07 2013-08-27 The University Of Chicago Use of opioid antagonists
US9717725B2 (en) 2005-03-07 2017-08-01 The University Of Chicago Use of opioid antagonists
US20080274119A1 (en) * 2005-03-07 2008-11-06 The University Of Chicago Use of Opioid Antagonists to Attenuate Endothelial Cell Proliferation and Migration
US20060258696A1 (en) * 2005-03-07 2006-11-16 Jonathan Moss Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US9662325B2 (en) 2005-03-07 2017-05-30 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US9675602B2 (en) 2005-03-07 2017-06-13 The University Of Chicago Use of opioid antagonists to attenuate endothelial cell proliferation and migration
US20100311781A1 (en) * 2005-05-25 2010-12-09 Progenics Pharmaceuticals, Inc. Synthesis of r-n-methylnaltrexone
US20070099946A1 (en) * 2005-05-25 2007-05-03 Doshan Harold D Synthesis of R-N-methylnaltrexone
US7674904B2 (en) 2005-05-25 2010-03-09 Progenics Pharmaceuticals, Inc. Synthesis of R-N-methylnaltrexone
US8003794B2 (en) 2005-05-25 2011-08-23 Progenics Pharmaceuticals, Inc. (S)-N-methylnaltrexone
US9597327B2 (en) 2005-05-25 2017-03-21 Progenics Pharmaceuticals, Inc. Synthesis of (R)-N-methylnaltrexone
US8343992B2 (en) 2005-05-25 2013-01-01 Progenics Pharmaceuticals, Inc. Synthesis of R-N-methylnaltrexone
US8916581B2 (en) 2005-05-25 2014-12-23 Progenics Pharmaceuticals, Inc. (S)-N-methylnaltrexone
US20100105911A1 (en) * 2005-05-25 2010-04-29 Boyd Thomas A (S)-N-methylnal trexone
US7563899B2 (en) 2005-05-25 2009-07-21 Progenics Pharmaceuticals, Inc. (S)-N-methylnaltrexone
US20080194611A1 (en) * 2005-06-03 2008-08-14 Alverdy John C Modulation of Cell Barrier Dysfunction
US20080064743A1 (en) * 2006-09-08 2008-03-13 Wyeth Dry powder compound formulations and uses thereof
US8338446B2 (en) 2007-03-29 2012-12-25 Wyeth Llc Peripheral opioid receptor antagonists and uses thereof
US8546418B2 (en) 2007-03-29 2013-10-01 Progenics Pharmaceuticals, Inc. Peripheral opioid receptor antagonists and uses thereof
US20100099699A1 (en) * 2007-03-29 2010-04-22 Wyeth Peripheral opioid receptor antagonists and uses thereof
US9879024B2 (en) 2007-03-29 2018-01-30 Progenics Pharmaceuticals., Inc. Crystal forms of (R)-N-methylnaltrexone bromide and uses thereof
US8772310B2 (en) 2007-03-29 2014-07-08 Wyeth Llc Peripheral opioid receptor antagonists and uses thereof
US20100305323A1 (en) * 2007-03-29 2010-12-02 Smolenskaya Valeriya N Crystal forms of (r)-n-methylnaltrexone bromide and uses thereof
US8853232B2 (en) 2007-03-29 2014-10-07 Wyeth Llc Peripheral opioid receptor antagonists and uses thereof
US9102680B2 (en) 2007-03-29 2015-08-11 Wyeth Llc Crystal forms of (R)-N-methylnaltrexone bromide and uses thereof
US20110190331A1 (en) * 2007-03-29 2011-08-04 Avey Alfred A Peripheral opioid receptor antagonists and uses thereof
US8471022B2 (en) 2008-02-06 2013-06-25 Progenics Pharmaceuticals, Inc. Preparation and use of (R),(R)-2,2′-bis-methylnaltrexone
US20110100099A1 (en) * 2008-02-06 2011-05-05 Progenics Pharmaceuticals, Inc. Preparation and use of (r),(r)-2,2'-bis-methylnaltrexone
US8916706B2 (en) 2008-02-06 2014-12-23 Progenics Pharmaceuticals, Inc. Preparation and use of (R),(R)-2,2′-bis-methylnaltrexone
US9526723B2 (en) 2008-03-21 2016-12-27 The University Of Chicago Treatment with opioid antagonists and mTOR inhibitors
US20110021551A1 (en) * 2008-03-21 2011-01-27 Jonathan Moss TREATMENT WITH OPIOID ANTAGONISTS AND mTOR INHIBITORS
US10383869B2 (en) 2008-03-21 2019-08-20 The University Of Chicago Treatment with opioid antagonists and mTOR inhibitors
US8685995B2 (en) 2008-03-21 2014-04-01 The University Of Chicago Treatment with opioid antagonists and mTOR inhibitors
US20100120813A1 (en) * 2008-09-30 2010-05-13 Wyeth Peripheral opioid receptor antagonists and uses thereof
US8420663B2 (en) 2008-09-30 2013-04-16 Wyeth Peripheral opioid receptor antagonists and uses thereof
US8822490B2 (en) 2008-09-30 2014-09-02 Wyeth Llc Peripheral opioid receptor antagonists and uses thereof
US9492445B2 (en) 2008-09-30 2016-11-15 Wyeth, Llc Peripheral opioid receptor antagonists and uses thereof
US9724343B2 (en) 2008-09-30 2017-08-08 Wyeth, Llc Peripheral opioid receptor antagonists and uses thereof
US9180125B2 (en) 2008-09-30 2015-11-10 Wyeth, Llc Peripheral opioid receptor antagonists and uses thereof
US8455644B2 (en) 2008-09-30 2013-06-04 Wyeth Peripheral opioid receptor antagonists and uses thereof
US8247425B2 (en) 2008-09-30 2012-08-21 Wyeth Peripheral opioid receptor antagonists and uses thereof
US10307417B2 (en) 2010-03-11 2019-06-04 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone
US10376505B2 (en) 2010-03-11 2019-08-13 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone
US10507206B2 (en) 2010-03-11 2019-12-17 Wyeth, Llc Oral formulations and lipophilic salts of methylnaltrexone

Also Published As

Publication number Publication date
US20020028825A1 (en) 2002-03-07
US20090312359A1 (en) 2009-12-17
AU758416B2 (en) 2003-03-20
EP1047426B1 (en) 2010-05-12
EP1047426A4 (en) 2004-07-14
US6608075B2 (en) 2003-08-19
WO1999022737A1 (en) 1999-05-14
US20050048117A1 (en) 2005-03-03
CA2312234A1 (en) 1999-05-14
CA2312234C (en) 2005-03-22
US6274591B1 (en) 2001-08-14
US20040162308A1 (en) 2004-08-19
EP1047426A1 (en) 2000-11-02
AU1380299A (en) 1999-05-24

Similar Documents

Publication Publication Date Title
US6608075B2 (en) Use of methylnaltrexone and related compounds
US5972954A (en) Use of methylnaltrexone and related compounds
JP4256259B2 (en) Pharmaceutical formulation of oxycodone and naloxone
Greiff et al. Pharmacokinetic drug interactions with gastrointestinal motility modifying agents
EP0377518B1 (en) Sustained release pharmaceutical composition
US6559158B1 (en) Use of methylnaltrexone and related compounds to treat chronic opioid use side affects
US20120190702A1 (en) Use of methylnaltrexone and related compounds to treat constipation in chronic opioid users
US20070190142A1 (en) Dosage forms for the delivery of drugs of abuse and related methods
KR101774676B1 (en) Pharmaceutical compositions comprising hydromorphone and naloxone
AU2002316738A1 (en) Pharmaceutical combinations of oxycodone and naloxone
US20090312358A1 (en) Method for management of diarrhea
JP2001081031A (en) Benzamide derivative-containing preparation having improved solubility and oral adsorption
MXPA05010821A (en) Combination therapy for constipation comprising a laxative and a peripheral opioid antagonist.
JP2002534374A (en) Multiparticulate oral dosage form
AU2003204844B2 (en) Use of methylnaltrexone and related compounds
EP2234630A1 (en) Method of stimulating the motility of the gastrointestinal system using ipamorelin
WO2001095941A1 (en) Solid dispersions and medicines
US20160256453A1 (en) Opioid receptor antagonist for use in treating patients with severe constipation induced by high opiate dosage regimen
CN111629758A (en) Pharmaceutical resinate compositions and methods of making and using same

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARCH DEVELOPMENT CORPORATION, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FOSS, JOSEPH F.;ROIZEN, MICHAEL F.;MOSS, JONATHAN;AND OTHERS;REEL/FRAME:014120/0085

Effective date: 19990825

AS Assignment

Owner name: UR LABS, INC., NEVADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DRELL, WILLIAM;REEL/FRAME:014120/0091

Effective date: 19990723

Owner name: CHICAGO, THE UNIVERSITY OF, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ARCH DEVELOPMENT CORPORATION;REEL/FRAME:014120/0083

Effective date: 20030219

AS Assignment

Owner name: PROGENICS PHARMACEUTICALS NEVADA, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UR LABS, INC.;REEL/FRAME:017480/0272

Effective date: 20051222

STCB Information on status: application discontinuation

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