WO2015065867A2 - Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting - Google Patents

Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting Download PDF

Info

Publication number
WO2015065867A2
WO2015065867A2 PCT/US2014/062320 US2014062320W WO2015065867A2 WO 2015065867 A2 WO2015065867 A2 WO 2015065867A2 US 2014062320 W US2014062320 W US 2014062320W WO 2015065867 A2 WO2015065867 A2 WO 2015065867A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
ring
peripherally
opioid receptor
receptor agonist
Prior art date
Application number
PCT/US2014/062320
Other languages
French (fr)
Other versions
WO2015065867A3 (en
Inventor
Derek T. Chalmers
James B. Jones
Original Assignee
Cara Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cara Therapeutics, Inc. filed Critical Cara Therapeutics, Inc.
Priority to US15/032,351 priority Critical patent/US20160250277A1/en
Publication of WO2015065867A2 publication Critical patent/WO2015065867A2/en
Publication of WO2015065867A3 publication Critical patent/WO2015065867A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1016Tetrapeptides with the first amino acid being neutral and aromatic or cycloaliphatic

Definitions

  • Strong mu opioid analgesics such as morphine, fentanyl, or hydromorphone, are mainstays of pain treatment in the immediate postoperative period, and are used as part of a multimodal analgesic approach.
  • the use of strong mu opioid analgesics is associated with an array of unwanted and serious side effects, including postoperative opioid-induced respiratory depression, or POIRD, postoperative nausea and vomiting, or PONV, and opioid- induced bowel dysfunction, or OBD, which contributes to the severity of postoperative ileus, or POL
  • POIRD postoperative opioid-induced respiratory depression
  • PONV postoperative nausea and vomiting
  • OBD opioid- induced bowel dysfunction
  • OBD opioid- induced bowel dysfunction
  • the incidence of POIRD may be as high as 29 percent, can occur unexpectedly in even the healthiest of patients, and exerts a disproportionately high toll on length of stay and hospital costs due to the significant expenses associated with the treatment of POIRD.
  • PONV occurs in approximately one-third of surgical patients overall, and is an important factor in determining length of stay after surgery, resulting in annual costs in the U.S. in the range of $1 billion.
  • acetaminophen which carry the risks attendant to these therapeutics.
  • mu opioids due to their CNS activity, mu opioids produce feelings of euphoria, which can give rise to abuse and addiction.
  • DEA United States Drug Enforcement Agency
  • Controlled Substances Act which imposes strict registration, record keeping and reporting requirements, security control and restrictions on prescriptions - all of which significantly increase the costs and the liability attendant to prescription opioid analgesics.
  • the present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject.
  • the peripherally-restricted kappa opioid receptor agonist includes a peptide.
  • the peptide includes one or more D-amino acids.
  • the present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist, wherein the peripherally restricted kappa opioid receptor agonist comprises a synthetic peptide amide having the formula:
  • ring moiety is an optionally substituted 4 to 8-membered heterocyclic ring moiety wherein all ring heteroatoms in said ring moiety are N; wherein Y and Z are each independently C or N; provided that when such ring moiety is a six, seven or eight-membered ring, Y and Z are separated by at least two ring atoms; and provided that when such ring moiety has a single ring heteroatom which is N, then such ring moiety is non-aromatic; V is CrC 6 alkyl, and e is zero or 1, wherein when e is zero, then V is null and and R 2 are directly bonded to the same or different ring atoms; wherein (i) is selected from the group consisting of -H, -OH, halo, -CF 3 , -NH 2 , - COOH, CrC 6 alkyl, CrC 6 alkoxy, amidino, CrC 6 alkyl-substituted amidino, aryl
  • R' and R" are each independently -H, Ci-C 8 alkyl, aryl, or heterocyclyl or R' and R" are combined to form a 4- to 8-membered ring, which ring is optionally singly or doubly substituted with substituents independently selected from the group consisting of Ci-C 6 alkyl, -Ci-C 6 alkoxy, -OH, -CI, -F, -NH 2 , -N0 2 , -CN, -COOH and amidino; and R 2 is selected from the group consisting of -H, amidino, singly or doubly CrC 6 alkyl-substituted amidino, -CN, -CONH 2 , -CONR'R
  • heterocyclic ring moieties comprising Ri and R 2 is optionally singly or doubly substituted with substituents independently selected from the group consisting of CrC 6 alkyl, C C 6 alkoxy, optionally substituted phenyl, oxo, -OH, -CI, -F, -NH 2 , -N0 2 , -CN, -COOH, and amidino;
  • Y and Z-containing ring moiety is a six membered ring having two ring heteroatoms, both Y and Z are N and W is null, then -(V) e RiR 2 is attached to a ring atom other than Z; and if e is zero, then Ri and R 2 are not both -H.
  • FIG. 2 Phase 2b Laparoscopic Hysterectomy - Summed Pain Intensity Difference from 0-24 Hours (SPIDo- 24 ) following postoperative treatment. *p ⁇ 0.05, **p ⁇ 0.01;
  • FIG. 3 Phase 2b Laparoscopic Hysterectomy - Pain Intensity Difference (PID) at specific times relative to postoperative baseline pain intensity. *p ⁇ 0.05, **p ⁇ 0.01 for CR845/CR845. #p ⁇ 0.05 for both Placebo/CR845 and CR845/Placebo. Values represent mean + SEM
  • FIG. 4 Phase 2b Laparoscopic Hysterectomy - Total Pain Relief Within the first 2 hours (TOTPARO-2) following postoperative treatment. *p ⁇ 0.05. Values represent mean + SEM.
  • FIG. 5 Phase 2b Laparoscopic Hysterectomy - Morphine Consumption For 2-24 hours post- treatment in patients. *p ⁇ 0.05; Values represent mean + SEM.
  • FIG. 6 Phase 2b Laparoscopic Hysterectomy - Incidence of opioid-related adverse events over 24 hours. ***p ⁇ 0.001; *p ⁇ 0.05.
  • FIG. 8a Phase 2 Bunionectomy - Summed Pain Intensity Difference from 0-24 hours (SPIDo- 24 ), 0-36 hours (p SPIDO-36) and 0-48 hours (SPIDO-48) in completer population.
  • FIG. 8b Phase 2 Bunionectomy - Summed Pain Intensity Difference from 0-24 hours (SPIDO-24), 0-36 hours (SPIDO-36) and 0-48 hours (SPIDO-48) in mITT Population (Completers plus non-completers). *p ⁇ 0.05 - One-sided ANOVA with Treatment Group as a Main Effect (mean +/- SEM).
  • FIG. 9a Phase 2 Bunionectomy - Pain Intensity Difference relative to baseline in CR845 and placebo completer treatment groups over a 48 hour period. * p ⁇ 0.05 (0-36 hours). ** p ⁇ 0.01 (0- 12 hours).
  • FIG. 9b Phase 2 Bunionectomy - Pain Intensity Difference relative to baseline in CR845 and placebo treatment Groups in mITT populations across 48 hours. *p ⁇ 0.05 (0-12 hours)
  • FIG. 10 Phase 2 Bunionectomy - CR845 Suppression of Nausea and Vomiting. *p ⁇ 0.05
  • FIG. 11 Phase la Pharmacokinetic profiles of increasing concentrations of CR845 in capsules in human subjects.
  • Nausea is an unpleasant experience in humans and probably animals. Physiologically, nausea is typically associated with decreased gastric motility and increased tone in the small intestine. Additionally, there is often reverse peristalsis in the proximal small intestine.
  • Emesis or vomiting is when gastric and often small intestinal contents are propelled up to and out of the mouth.
  • a deep breath is taken, the glottis is closed and the larynx is raised to open the upper esophageal sphincter.
  • the soft palate is elevated to close off the nasal cavity.
  • the diaphragm is contracted sharply downward to create negative pressure in the thorax, which opens the esophagus and distal esophageal sphincter. Then, simultaneously with downward movement of the diaphragm, the muscles of the abdominal walls contract vigorously, squeezing the stomach and elevating intragastric pressure.
  • the present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject such as a human, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject, wherein the moiety:
  • Ri-(V) e -R 2 is selected from any one of the following:
  • the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject, wherein the synthetic peptide amide has the structure:
  • the peripherally-restricted kappa opioid receptor agonist can be administered to the subject within 12, 24 or 36 hours prior to, during or within 12, 24 or 36 hours after undergoing a medical procedure.
  • the medical procedure causes pain, which may be soft tissue pain e.g. muscular pain or visceral pain; or hard tissue pain, e.g. bone pain Kappa opioid receptor agonists and their uses for the prophylaxis, inhibition and treatment of painful and inflammatory diseases, disorders and conditions are described in US Patent Nos.
  • the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, wherein the peripherally-restricted kappa opioid receptor agonist is administered to the subject by a route of injection chosen from the following: subcutaneous injection, intravenous injection, intraperitoneal injection, intra-articular injection, and intramuscular injection.
  • the peripherally-restricted kappa opioid receptor agonist can be any suitable peripherally-restricted kappa opioid receptor agonist, such as for instance a nonnarcotic analgesic, for example, asimadoline (N-[(lS)-2-[(3S)-3-hydroxypyrrolidin-l-yl]-l- phenylethyl]-N-methyl-2,2-diphenylacetamide), or nalfurafine ((2E)-N-[(5a,6P)-17-(cyclo- propylmethyl)-3,14-dihydroxy-4,5-epoxymorphinan-6-yl]-3-(3-furyl)-N-methylacrylamide).
  • a nonnarcotic analgesic for example, asimadoline (N-[(lS)-2-[(3S)-3-hydroxypyrrolidin-l-yl]-l- phenylethyl]-N-methyl-2,2-diphenylacet
  • the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the nausea and/or vomiting occurs within 48 hours after administration of at least one dose of a mu opioid analgesic.
  • the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the nausea and/or vomiting occurs within 24 hours after administration of at least one dose of a mu opioid analgesic.
  • the mu opioid analgesic is administered to treat, inhibit or prevent hard tissue pain, such as bone pain.
  • the hard tissue pain may be due to a medical procedure.
  • the medical procedure may be any medical procedure that causes hard tissue pain, such as, for instance and without limitation, a bunionectomy procedure.
  • the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the peripherally-restricted kappa opioid receptor agonist is administered prior to administration of a first dose of the mu opioid analgesic.
  • the peripherally-restricted kappa opioid receptor agonist is co-administered with at least one dose of the mu opioid analgesic.
  • the peripherally-restricted kappa opioid receptor agonist is administered after administration of at least one dose of the mu opioid analgesic. Kappa Receptor Agonist CR845
  • CR845 is a peripherally- acting kappa opioid receptor agonist useful for treatment of both acute and chronic pain, and also has anti-inflammatory properties.
  • the most advanced product candidate, I.V. CR845 has demonstrated significant pain relief and a favorable safety and tolerability profile in three Phase 2 clinical trials in patients with acute postoperative pain. Due to its selectivity for the kappa opioid receptor and ability to decrease mu opioid use, CR845 has demonstrated a consistent ability to decrease the acute opioid-related adverse events (AEs) of nausea and vomiting with no evidence of drug-related respiratory depression.
  • AEs acute opioid-related adverse events
  • CR845 has been administered to over 300 human subjects in Phase 1 and Phase 2 clinical trials as an intravenous infusion, short bolus or oral capsule and was safe and well tolerated in these clinical trials.
  • CR845 successfully attenuated acute and chronic visceral, inflammatory and neuropathic pain in a dose-dependent manner (see Table 1, below).
  • the analgesic effect of CR845 was recordable within 15 minutes post- administration and lasted for up to 18 hours following single-dose administration.
  • CR845 also decreased the production and release of pro-inflammatory mediators, likely due to the direct activation of kappa opioid receptors expressed on immune cells that synthesize and secrete these substances.
  • CR845 The peripheral mechanism of action of CR845 is supported preclinically by both biochemical in vitro assays and in vivo functional pharmacological studies.
  • animals administered analgesic and supra-analgesic doses of CR845 exhibited no measurable concentrations of drug in extracted brain tissue indicating that the CNS was not the site of action for CR845.
  • the analgesic action of CR845 was blocked with kappa opioid receptor antagonists administered directly to the local site of injury, indicating a peripheral site of action for CR845 ( Figure 1).
  • neuropathic pain is induced
  • CR845 in an injectable version of the most advanced kappa opioid receptor-based peripheral analgesic is designed to provide pain relief without stimulating mu opioid receptors and therefore without mu opioid-related side effects, such as nausea, vomiting, respiratory depression and euphoria.
  • Intravenous CR845 has demonstrated efficacy and tolerability in three randomized, double-blind, placebo-controlled Phase 2 clinical trials in patients undergoing soft tissue (laparoscopic hysterectomy) and hard tissue (bunionectomy) surgery. In both the laparoscopic hysterectomy and bunionectomy clinical trials, CR845 administration resulted in statistically significant reductions in pain intensity, as measured by summed pain intensity differences, or SPID, which is the FDA-recommended acute pain endpoint: See below.
  • a Phase 2 multicenter, double-randomized, double-blind, placebo-controlled clinical trial was a conducted in 203 patients at 22 sites in the United States.
  • the trial enrolled female patients, ages 21 to 65, scheduled for elective laparoscopic hysterectomy under general anesthesia.
  • patients were administered either placebo or one dose of 0.04 mg/kg I.V. CR845 preoperatively.
  • following surgery if they were medically stable and had a pain intensity score >40 on a 100 point pain scale based on the visual analog scale, or VAS, they were re- randomized to receive either placebo or one dose of 0.04 mg/kg I.V. CR845.
  • Efficacy was measured using time- specific 24 hour pain intensity differences.
  • Pain intensity was measured at various times by asking patients to rate their pain on a 100-point scale, where "0" is absence of pain and "100" is the worst possible pain.
  • ⁇ 3 is the difference between the PI measured prior to treatment and at subsequent times of measurement.
  • SPID or the summed pain intensity difference, is the time-weighted sum of all of the PID scores, from the pretreatment level to a subsequent time of measurement, such as 24 hours after the pretreatment baseline pain measurement. Both PID and SPID are FDA-recognized endpoints for acute pain clinical trials. Additional endpoints included the amount of morphine consumption over 24 hours, time-specific total pain relief and patient global evaluation of study medication. Of the 203 patients that participated in the trial, 183 received a post operative dose; however, two subjects did not record baseline pain scores and were not included in calculated PID and SPID values. Accordingly, four treatment groups resulted from pre- and post-operative randomization:
  • the CR845/CR845 group exhibited a statistically significant reduction in pain over a 24-hour time period, as indicated by an improvement in 0-24 hour mean SPID, compared to the
  • Placebo/Placebo group (p ⁇ 0.01).
  • the Placebo/CR845 group also exhibited a statistically significant improvement in 0-24 hour mean SPID compared to the Placebo/Placebo group (p ⁇ 0.05).
  • the CR845/Placebo group exhibited an improved 0-24 hour mean SPID compared to the Placebo/Placebo group, but this difference did not reach statistical significance, which we believe was due to the small number of patients.
  • Figure 2 illustrates the 0-24 hour mean SPIDs of the four treatment groups listed above.
  • Placebo/Placebo group Compared to the Placebo/Placebo group, patients in the CR845/CR845 group exhibited an approximately 60% greater reduction in pain intensity at 24 hours (p ⁇ 0.01), as well as statistically significant improvements for the 0-4, 0-8 and 0-16 hour time intervals. Patients in the CR845/Placebo and Placebo/CR845 groups also exhibited statistically significant decreases in pain intensity for the 0-8 and 0-16 hour time intervals, compared to patients in the Placebo/Placebo group.
  • Figure 3 illustrates the PID relative to postoperative baseline in patients in the four treatment groups.
  • TOTPAR total pain relief score
  • TOTPAR scores were numerically superior across all intervals for the CR845/CR845 and Placebo/CR845 groups relative to the Placebo/Placebo group.
  • the patients in the CR845/CR845 group and Placebo/CR845 exhibited statistically superior pain relief as compared to the
  • FIG. 4 depicts the mean TOTPAR scores for the first 2 hour period for each of the four treatment groups listed above.
  • Intravenous morphine was available as rescue medication to all treatment groups upon patient request. Calculations of morphine consumption per treatment group in the 2-24 hour period, after patients leave the post-anesthesia care unit, or PACU, indicated that patients in the CR845/CR845 group used approximately 45% less morphine than those in the Placebo/Placebo group (p ⁇ 0.05) and patients in the Placebo/CR845 and CR845/Placebo groups used
  • Figure 6 depicts the percentage of patients reporting opioid- related adverse events of nausea, vomiting and pruritus.
  • Bunionectomy is a surgical procedure to remove a bunion, an enlargement of the joint at the base of the big toe and includes bone and soft tissue.
  • the procedure typically results in intense pain requiring postoperative analgesic care, usually beginning with local anesthetic infusion and ongoing administration of a strong opioid, such as morphine or fentanyl, for several days after surgery during which the patient often suffers from nausea and vomiting.
  • a strong opioid such as morphine or fentanyl
  • Clinical trial was a randomized, double-blind, placebo-controlled trial conducted in 51 patients following bunionectomy surgery at a single site in the U.S.
  • the trial enrolled female and male patients, ages 18 years and older, scheduled for elective bunionectomy under regional anesthesia.
  • patients were randomized into one of two treatment groups (CR845 or Placebo, in a 2: 1 ratio) after reporting moderate-to-severe pain, defined as a pain intensity score > 40 on a 100-point pain scale.
  • Patients randomized to receive I.V. CR845 were administered an I.V.
  • the Completer analysis is indicative of the actual efficacy of I.V. CR845 under conditions where patients are exposed to the drug as specified in the protocol, while the ⁇ analysis is indicative of the actual variability that will be encountered in the mITT populations.
  • the understanding of this variability serves as the basis for determining the appropriate number of patients for enrollment in our Phase 3 clinical trials. In this trial, mean PID from baseline at each time interval was measured, and was numerically superior across the 48 hour trial period in the I.V. CR845 treatment group relative to the placebo group for both the Completer and mITT populations (see Figures 9a and 9b).
  • CR845 an attractive treatment option for postoperative patients and their physicians.
  • I.V. CR845 administered intravenous administration of I.V. CR845 at a dose of 0.005 mg/kg was safe and generally well tolerated.
  • the most frequent TEAEs (greater than 10%) observed in the CR845 treatment group were transient facial tingling and somnolence. Of the seven cases of somnolence reported, four were reported as "mild” and/or "related to drug” and three as "moderate” and/or "not related to drug”.
  • the mean plasma sodium concentration in CR845-treated patients exhibited an approximately 3% rise over 24 hours from baseline levels, but was not outside the normal physiological range at either 24 or 48 hours post-CR845 administration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Emergency Medicine (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A method for preventing, inhibiting or treating nausea and/or vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject. The nausea and/or vomiting can be associated with use of an opioid, such as morphine or fentanyl. The peripherally-restricted kappa opioid receptor agonist can be an L-amino acid-containing peptide, a D-amino acid-containing peptide, or a synthetic peptide amide, such as for instance, D-Phe-D-Phe-D-Leu-D-Lys-[ω(4-aminopiperidine-4-carboxylic acid)] -OH (CR845).

Description

PERIPHERAL KAPPA OPIOID RECEPTOR AGONISTS FOR PREVENTING, INHIBITING
OR TREATING NAUSEA AND VOMITING
Strong mu opioid analgesics, such as morphine, fentanyl, or hydromorphone, are mainstays of pain treatment in the immediate postoperative period, and are used as part of a multimodal analgesic approach. However, the use of strong mu opioid analgesics is associated with an array of unwanted and serious side effects, including postoperative opioid-induced respiratory depression, or POIRD, postoperative nausea and vomiting, or PONV, and opioid- induced bowel dysfunction, or OBD, which contributes to the severity of postoperative ileus, or POL The incidence of POIRD may be as high as 29 percent, can occur unexpectedly in even the healthiest of patients, and exerts a disproportionately high toll on length of stay and hospital costs due to the significant expenses associated with the treatment of POIRD. PONV occurs in approximately one-third of surgical patients overall, and is an important factor in determining length of stay after surgery, resulting in annual costs in the U.S. in the range of $1 billion. These mu opioid-related adverse events not only significantly increase the cost of care, but also reduce a patient's quality of care and lead to sub-optimal recovery.
In 2005, the FDA announced a requirement for boxed warnings of potential
cardiovascular risk for all NSAIDs. The FDA warning related to cardiovascular adverse events associated with NSAIDs and the increased awareness of the risk of liver toxicity associated with high doses of acetaminophen have led to increased use of mu opioid analgesics for the treatment of chronic pain. However, the use of mu opioid analgesics carries significant additional risks. Chronic opioid use causes patients to develop tolerance for the opioid, which results in the patient needing increasing opioid doses to achieve the same level of pain relief. For the most commonly prescribed analgesic combination products, the need for increasing doses to achieve the same level of pain relief means exposure to increasing amounts of NSAIDs or
acetaminophen, which carry the risks attendant to these therapeutics. Moreover, due to their CNS activity, mu opioids produce feelings of euphoria, which can give rise to abuse and addiction. Underlining the severity of this issue, in 2013, the FDA announced class-wide safety labeling changes and new post-market study requirements for all extended-release and long- acting mu opioid analgesics intended to treat pain. In addition, as a result of their potential for misuse, abuse and addiction, currently approved mu opioids are strictly regulated by the United States Drug Enforcement Agency (DEA), under the Controlled Substances Act, which imposes strict registration, record keeping and reporting requirements, security control and restrictions on prescriptions - all of which significantly increase the costs and the liability attendant to prescription opioid analgesics.
SUMMMARY
The present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject. In one embodiment, the peripherally-restricted kappa opioid receptor agonist includes a peptide. In another embodiment, the peptide includes one or more D-amino acids.
In one embodiment the present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist, wherein the peripherally restricted kappa opioid receptor agonist comprises a synthetic peptide amide having the formula:
Figure imgf000003_0001
Ri-(V)e-R2
or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof.
In one embodiment, the residue Xaai is selected from the group consisting of (A)(A')D- Phe, (A)(A')(a-Me)D-Phe, D-Tyr, D-Tic, D-tert-leucine, D-neopentylglycine, D-phenylglycine, D-homophenylalanine, and P-(E)D-Ala, wherein each (A) and each (Α') are phenyl ring substituents independently selected from the group consisting of -H, -F, -CI, -N02, -CH3, -CF3, - CN, and -CONH2, and wherein each (E) is independently selected from the group consisting of cyclobutyl, cyclopentyl, cyclohexyl, pyridyl, thienyl and thiazolyl; Xaa2 is selected from the group consisting of (A)(A')D-Phe, 3,4-dichloro-D-Phe, (A)(A')(a-Me)D-Phe, D-lNal, D-2Nal, D-Tyr, (E)D-Ala and D-Trp; Xaa3 is selected from the group consisting of D-Nle, D-Phe, (E)D- Ala, D-Leu, (a-Me)D-Leu, D-Hle, D-Val, and D-Met; Xaa4 is selected from the group consisting of (B)2D-Arg, (B)2D-Nar, (B)2D-Har, ζ-(Β)ϋ-Η1 8, D-Dap, s-(B)D-Lys, s-(B)2-D-Lys, D-Amf, amidino-D-Amf, y-(B)2D-Dbu, 5-(B)2a-(B')D-Orn, D-2-amino-3(4-piperidyl)propionic acid, D- 2-amino-3(2-aminopyrrolidyl)propionic acid, D-a-amino-P-amidinopropionic acid, a-amino-4- piperidineacetic acid, cis-a,4-diaminocyclohexane acetic acid, trans-a,4- diaminocyclohexaneacetic acid, cis-a-amino-4-methylaminocyclo-hexane acetic acid, trans-a- amino-4-methylaminocyclohexane acetic acid, a-amino-l-amidino-4-piperidineacetic acid, cis- a-amino-4-guanidinocyclohexane acetic acid, and trans-a-amino-4-guanidinocyclohexane acetic acid; wherein each (B) is independently selected from the group consisting of H and C C4 alkyl, and (Β') is H or (a- Me); W is selected from the group consisting of: Null, provided that when W is null, Y is N; -NH-(CH2)b- with b equal to zero, 1, 2, 3, 4, 5, or 6; and -NH-(CH2)c-0- with c equal to 2, or 3, provided that Y is C.
In another embodiment the moiety
Figure imgf000004_0001
is an optionally substituted 4 to 8-membered heterocyclic ring moiety wherein all ring heteroatoms in said ring moiety are N; wherein Y and Z are each independently C or N; provided that when such ring moiety is a six, seven or eight-membered ring, Y and Z are separated by at least two ring atoms; and provided that when such ring moiety has a single ring heteroatom which is N, then such ring moiety is non-aromatic; V is CrC6 alkyl, and e is zero or 1, wherein when e is zero, then V is null and and R2 are directly bonded to the same or different ring atoms; wherein (i) is selected from the group consisting of -H, -OH, halo, -CF3, -NH2, - COOH, CrC6 alkyl, CrC6 alkoxy, amidino, CrC6 alkyl-substituted amidino, aryl, optionally substituted heterocyclyl, Pro-amide, Pro, Gly, Ala, Val, Leu, He, Lys, Arg, Orn, Ser, Thr, -CN, - CONH2, -COR', -S02R', -CONR'R", -NHCOR', OR' and S02NR'R"; wherein said optionally substituted heterocyclyl is optionally singly or doubly substituted with substituents
independently selected from the group consisting of CrC6 alkyl, CrC6 alkoxy, oxo, -OH, -CI, - F, -NH2, -NO2, -CN, -COOH, and amidino; wherein R' and R" are each independently -H, Ci-C8 alkyl, aryl, or heterocyclyl or R' and R" are combined to form a 4- to 8-membered ring, which ring is optionally singly or doubly substituted with substituents independently selected from the group consisting of Ci-C6 alkyl, -Ci-C6 alkoxy, -OH, -CI, -F, -NH2, -N02, -CN, -COOH and amidino; and R2 is selected from the group consisting of -H, amidino, singly or doubly CrC6 alkyl-substituted amidino, -CN, -CONH2, -CONR'R", -NHCOR', -S02NR'R" and -COOH; or (ii) Ri and R2 taken together can form an optionally substituted 4- to 9-membered heterocyclic monocyclic or bicyclic ring moiety which is bonded to a single ring atom of the Y and Z- containing ring moiety; or (iii) Ri and R2 taken together with a single ring atom of the Y and Z- containing ring moiety can form an optionally substituted 4- to 8-membered heterocyclic ring moiety to form a spiro structure; or (iv) Ri and R2 taken together with two or more adjacent ring atoms of the Y and Z-containing ring moiety can form an optionally substituted 4- to 9- membered heterocyclic monocyclic or bicyclic ring moiety fused to the Y and Z-containing ring moiety; wherein each of said optionally substituted 4-, 5-, 6,-, 7-, 8- and 9-membered
heterocyclic ring moieties comprising Ri and R2 is optionally singly or doubly substituted with substituents independently selected from the group consisting of CrC6 alkyl, C C6 alkoxy, optionally substituted phenyl, oxo, -OH, -CI, -F, -NH2, -N02, -CN, -COOH, and amidino;
provided that when the Y and Z-containing ring moiety is a six or seven membered ring having a single ring heteroatom and e is zero, then Ri is not -OH, and Ri and R2 are not both -H;
and provided further that when the Y and Z-containing ring moiety is a six membered ring having two ring heteroatoms, both Y and Z are N and W is null, then -(V)eRiR2 is attached to a ring atom other than Z; and if e is zero, then Ri and R2 are not both -H.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1: Efficacy of CR845 in "Chung Model" of neuropathic pain is blocked with Peripheral (Intrapaw) administration of a kappa antagonist (norBNI) in rats. *** denotes p<0.001 compared to vehicle-treated controls (two-way ANOVA). Vehicle or Nor-BNI was administered intraplantarly (0.2 mg) 15 min prior to CR845. Injection (1 mg/kg). N=6 male rats/group, mean + SEM.
FIG. 2: Phase 2b Laparoscopic Hysterectomy - Summed Pain Intensity Difference from 0-24 Hours (SPIDo-24) following postoperative treatment. *p<0.05, **p<0.01;
FIG. 3: Phase 2b Laparoscopic Hysterectomy - Pain Intensity Difference (PID) at specific times relative to postoperative baseline pain intensity. *p<0.05, **p<0.01 for CR845/CR845. #p<0.05 for both Placebo/CR845 and CR845/Placebo. Values represent mean + SEM
FIG. 4: Phase 2b Laparoscopic Hysterectomy - Total Pain Relief Within the first 2 hours (TOTPARO-2) following postoperative treatment. *p<0.05. Values represent mean + SEM.
FIG. 5: Phase 2b Laparoscopic Hysterectomy - Morphine Consumption For 2-24 hours post- treatment in patients. *p<0.05; Values represent mean + SEM. FIG. 6: Phase 2b Laparoscopic Hysterectomy - Incidence of opioid-related adverse events over 24 hours. ***p<0.001; *p<0.05.
FIG. 7: Phase 2b Laparoscopic Hysterectomy - Responder analysis of global evaluation of study medication. **p=0.001
FIG. 8a: Phase 2 Bunionectomy - Summed Pain Intensity Difference from 0-24 hours (SPIDo- 24), 0-36 hours (p SPIDO-36) and 0-48 hours (SPIDO-48) in completer population.
FIG. 8b: Phase 2 Bunionectomy - Summed Pain Intensity Difference from 0-24 hours (SPIDO-24), 0-36 hours (SPIDO-36) and 0-48 hours (SPIDO-48) in mITT Population (Completers plus non-completers). *p<0.05 - One-sided ANOVA with Treatment Group as a Main Effect (mean +/- SEM).
FIG. 9a: Phase 2 Bunionectomy - Pain Intensity Difference relative to baseline in CR845 and placebo completer treatment groups over a 48 hour period. * p<0.05 (0-36 hours). ** p<0.01 (0- 12 hours).
FIG. 9b: Phase 2 Bunionectomy - Pain Intensity Difference relative to baseline in CR845 and placebo treatment Groups in mITT populations across 48 hours. *p<0.05 (0-12 hours)
FIG. 10: Phase 2 Bunionectomy - CR845 Suppression of Nausea and Vomiting. *p<0.05 FIG. 11: Phase la Pharmacokinetic profiles of increasing concentrations of CR845 in capsules in human subjects.
DETAILED DESCRIPTION
Nausea is an unpleasant experience in humans and probably animals. Physiologically, nausea is typically associated with decreased gastric motility and increased tone in the small intestine. Additionally, there is often reverse peristalsis in the proximal small intestine.
Emesis or vomiting is when gastric and often small intestinal contents are propelled up to and out of the mouth. Usually, a deep breath is taken, the glottis is closed and the larynx is raised to open the upper esophageal sphincter. Also, the soft palate is elevated to close off the nasal cavity. The diaphragm is contracted sharply downward to create negative pressure in the thorax, which opens the esophagus and distal esophageal sphincter. Then, simultaneously with downward movement of the diaphragm, the muscles of the abdominal walls contract vigorously, squeezing the stomach and elevating intragastric pressure. The pylorus closes and the esophagus is open and the vomitus is forced out. In one embodiment the present invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject such as a human, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject, wherein the moiety:
Figure imgf000007_0001
Ri-(V)e-R2 is selected from any one of the following:
Figure imgf000007_0002
Figure imgf000008_0001
Figure imgf000008_0002
In another embodiment, the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the subject, wherein the synthetic peptide amide has the structure:
Figure imgf000008_0003
D-Phe-D-Phe-D-Leu-D-Lys-[ro(4-aminopiperidine-4-carboxylic acid)] -OH (also called CR845). The peripherally-restricted kappa opioid receptor agonist can be administered to the subject within 12, 24 or 36 hours prior to, during or within 12, 24 or 36 hours after undergoing a medical procedure. In one embodiment, the medical procedure causes pain, which may be soft tissue pain e.g. muscular pain or visceral pain; or hard tissue pain, e.g. bone pain Kappa opioid receptor agonists and their uses for the prophylaxis, inhibition and treatment of painful and inflammatory diseases, disorders and conditions are described in US Patent Nos. 7,402,564; 7,713,937; 7,727,963; 7,842,662; 8,217,007; 8,486,894; and 8,536,131, the disclosures of which are hereby incorporated by reference herein in their entireties. In another embodiment, the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a mammalian subject, wherein the peripherally-restricted kappa opioid receptor agonist is administered to the subject by a route of injection chosen from the following: subcutaneous injection, intravenous injection, intraperitoneal injection, intra-articular injection, and intramuscular injection.
In another embodiment, the peripherally-restricted kappa opioid receptor agonist can be any suitable peripherally-restricted kappa opioid receptor agonist, such as for instance a nonnarcotic analgesic, for example, asimadoline (N-[(lS)-2-[(3S)-3-hydroxypyrrolidin-l-yl]-l- phenylethyl]-N-methyl-2,2-diphenylacetamide), or nalfurafine ((2E)-N-[(5a,6P)-17-(cyclo- propylmethyl)-3,14-dihydroxy-4,5-epoxymorphinan-6-yl]-3-(3-furyl)-N-methylacrylamide).
In another embodiment, the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the nausea and/or vomiting occurs within 48 hours after administration of at least one dose of a mu opioid analgesic. In another embodiment, the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the nausea and/or vomiting occurs within 24 hours after administration of at least one dose of a mu opioid analgesic. In a particular embodiment, the mu opioid analgesic is administered to treat, inhibit or prevent hard tissue pain, such as bone pain. The hard tissue pain may be due to a medical procedure. The medical procedure may be any medical procedure that causes hard tissue pain, such as, for instance and without limitation, a bunionectomy procedure.
In another embodiment, the invention provides a method for preventing, inhibiting or treating nausea and vomiting in a human, by administering a peripherally-restricted kappa opioid receptor agonist to the human, wherein the peripherally-restricted kappa opioid receptor agonist is administered prior to administration of a first dose of the mu opioid analgesic. In still another embodiment, the peripherally-restricted kappa opioid receptor agonist is co-administered with at least one dose of the mu opioid analgesic. In yet another embodiment, the peripherally-restricted kappa opioid receptor agonist is administered after administration of at least one dose of the mu opioid analgesic. Kappa Receptor Agonist CR845
CR845 is a peripherally- acting kappa opioid receptor agonist useful for treatment of both acute and chronic pain, and also has anti-inflammatory properties. The most advanced product candidate, I.V. CR845, has demonstrated significant pain relief and a favorable safety and tolerability profile in three Phase 2 clinical trials in patients with acute postoperative pain. Due to its selectivity for the kappa opioid receptor and ability to decrease mu opioid use, CR845 has demonstrated a consistent ability to decrease the acute opioid-related adverse events (AEs) of nausea and vomiting with no evidence of drug-related respiratory depression. CR845 has been administered to over 300 human subjects in Phase 1 and Phase 2 clinical trials as an intravenous infusion, short bolus or oral capsule and was safe and well tolerated in these clinical trials.
In standard preclinical pain models, CR845 successfully attenuated acute and chronic visceral, inflammatory and neuropathic pain in a dose-dependent manner (see Table 1, below). The analgesic effect of CR845 was recordable within 15 minutes post- administration and lasted for up to 18 hours following single-dose administration. CR845 also decreased the production and release of pro-inflammatory mediators, likely due to the direct activation of kappa opioid receptors expressed on immune cells that synthesize and secrete these substances.
The peripheral mechanism of action of CR845 is supported preclinically by both biochemical in vitro assays and in vivo functional pharmacological studies. In pharmacokinetic studies, animals administered analgesic and supra-analgesic doses of CR845 exhibited no measurable concentrations of drug in extracted brain tissue indicating that the CNS was not the site of action for CR845. Moreover, in standard preclinical pain models, such as the "Chung Model" of neuropathic pain, the analgesic action of CR845 was blocked with kappa opioid receptor antagonists administered directly to the local site of injury, indicating a peripheral site of action for CR845 (Figure 1). In the "Chung Model", neuropathic pain is induced
experimentally by ligating spinal nerves mediating sensation for a hind limb. This results in a type of neuropathic pain, referred to as allodynia. Experimental animals with allodynia exhibit a "paw withdrawal reflex" upon contact with a relatively thin filament on the injured site. Sets of different thickness filaments are used to test sensitivity, each of which is designed to produce a given force (in grams) upon bending after contact. By testing with these filaments, the minimum force to evoke a withdrawal response defines the paw withdrawal threshold. The nerve injury produces a marked reduction in paw withdrawal thresholds (increased sensitivity to force) in response to probing with the filaments. I.V. administration of CR845 reduces this neuropathic pain as demonstrated by a subsequent increase in the withdrawal threshold (see Figure 1).
Administration of a low dose of the selective peripherally-acting kappa opioid receptor antagonist nor-binaltorphamine, or nor-BNI, into the plantar surface of the injured paw significantly reduces the effect of CR845, whereas injection of saline had no effect on the efficacy of CR845. Since nor-BNI was only able to block local peripheral kappa opioid receptors in this experiment, these results show that the effect of CR845 is a result of activation of kappa opioid receptors located at the peripheral site of injury rather than in the CNS.
Intravenous CR845
CR845, in an injectable version of the most advanced kappa opioid receptor-based peripheral analgesic is designed to provide pain relief without stimulating mu opioid receptors and therefore without mu opioid-related side effects, such as nausea, vomiting, respiratory depression and euphoria. Intravenous CR845 has demonstrated efficacy and tolerability in three randomized, double-blind, placebo-controlled Phase 2 clinical trials in patients undergoing soft tissue (laparoscopic hysterectomy) and hard tissue (bunionectomy) surgery. In both the laparoscopic hysterectomy and bunionectomy clinical trials, CR845 administration resulted in statistically significant reductions in pain intensity, as measured by summed pain intensity differences, or SPID, which is the FDA-recommended acute pain endpoint: See below.
A Phase 2 multicenter, double-randomized, double-blind, placebo-controlled clinical trial (CLIN2002) was a conducted in 203 patients at 22 sites in the United States. The trial enrolled female patients, ages 21 to 65, scheduled for elective laparoscopic hysterectomy under general anesthesia. In this trial, patients were administered either placebo or one dose of 0.04 mg/kg I.V. CR845 preoperatively. Following surgery, if they were medically stable and had a pain intensity score >40 on a 100 point pain scale based on the visual analog scale, or VAS, they were re- randomized to receive either placebo or one dose of 0.04 mg/kg I.V. CR845. Efficacy was measured using time- specific 24 hour pain intensity differences. Pain intensity, or PI, was measured at various times by asking patients to rate their pain on a 100-point scale, where "0" is absence of pain and "100" is the worst possible pain. ΡΠ3, or pain intensity difference, is the difference between the PI measured prior to treatment and at subsequent times of measurement. SPID, or the summed pain intensity difference, is the time-weighted sum of all of the PID scores, from the pretreatment level to a subsequent time of measurement, such as 24 hours after the pretreatment baseline pain measurement. Both PID and SPID are FDA-recognized endpoints for acute pain clinical trials. Additional endpoints included the amount of morphine consumption over 24 hours, time-specific total pain relief and patient global evaluation of study medication. Of the 203 patients that participated in the trial, 183 received a post operative dose; however, two subjects did not record baseline pain scores and were not included in calculated PID and SPID values. Accordingly, four treatment groups resulted from pre- and post-operative randomization:
(1) I.V. CR845 administered both preoperatively and postoperatively (CR845/CR845);
(2) placebo administered preoperatively and I.V. CR845 administered postoperatively
(Placebo/CR845);
(3) I.V. CR845 administered preoperatively and placebo administered postoperatively
(CR845/Placebo); and
(4) placebo administered both preoperatively and postoperatively (Placebo/Placebo).
The CR845/CR845 group exhibited a statistically significant reduction in pain over a 24-hour time period, as indicated by an improvement in 0-24 hour mean SPID, compared to the
Placebo/Placebo group (p<0.01). The Placebo/CR845 group also exhibited a statistically significant improvement in 0-24 hour mean SPID compared to the Placebo/Placebo group (p<0.05). The CR845/Placebo group exhibited an improved 0-24 hour mean SPID compared to the Placebo/Placebo group, but this difference did not reach statistical significance, which we believe was due to the small number of patients. Figure 2 illustrates the 0-24 hour mean SPIDs of the four treatment groups listed above.
Similar observations were made for different time periods after treatment. For example, over the 0-4 hour time period, in the CR845/CR845 group, there was a statistically significant 3.5-fold improvement in mean SPID values compared to the Placebo/Placebo group (p<0.05). In addition, over the 0-8, 0-12 and 0-16 time periods, patients in the Placebo/CR845 group also exhibited reduced pain intensity compared to the Placebo/Placebo group in a statistically significant manner (p<0.05), based on improved SPID values. The mean PID from baseline at each time interval was numerically superior across all groups that received I.V. CR845 preoperatively and/or postoperatively relative to the
Placebo/Placebo group. Compared to the Placebo/Placebo group, patients in the CR845/CR845 group exhibited an approximately 60% greater reduction in pain intensity at 24 hours (p<0.01), as well as statistically significant improvements for the 0-4, 0-8 and 0-16 hour time intervals. Patients in the CR845/Placebo and Placebo/CR845 groups also exhibited statistically significant decreases in pain intensity for the 0-8 and 0-16 hour time intervals, compared to patients in the Placebo/Placebo group. Figure 3 illustrates the PID relative to postoperative baseline in patients in the four treatment groups.
At the same time points at which pain intensity measurements were taken, patients' perceived pain relief scores were recorded using a 5 point subjective Likert scale (0-4), where zero corresponds to no relief and a score of four represents total relief. The "TOTPAR" score is calculated as the "total pain relief score", which is a time-weighted sum of pain relief scores over any given time period following post operative treatment with CR845 or placebo. Mean
TOTPAR scores were numerically superior across all intervals for the CR845/CR845 and Placebo/CR845 groups relative to the Placebo/Placebo group. The patients in the CR845/CR845 group and Placebo/CR845 exhibited statistically superior pain relief as compared to the
Placebo/Placebo group within the first 2 hours following postoperative randomization, as indicated by increased mean TOTPAR0_2 values (p<0.05). Figure 4 depicts the mean TOTPAR scores for the first 2 hour period for each of the four treatment groups listed above.
In the CR845/CR845 and Placebo/CR845 groups, there were also statistically significant improvements in reported pain relief for the 0-4, 2-4 and 0-8 hour time periods. In addition, the improvement in mean TOTPAR also reached statistical significance for the 0-12 hour interval for the CR845/CR845 group relative to the Placebo/Placebo group.
Intravenous morphine was available as rescue medication to all treatment groups upon patient request. Calculations of morphine consumption per treatment group in the 2-24 hour period, after patients leave the post-anesthesia care unit, or PACU, indicated that patients in the CR845/CR845 group used approximately 45% less morphine than those in the Placebo/Placebo group (p<0.05) and patients in the Placebo/CR845 and CR845/Placebo groups used
approximately 23% less morphine than those in the Placebo/Placebo group. Figure 5 depicts the morphine usage in each of the treatment groups between hours 2-24. Concurrently with the observed reduction in morphine use, patients treated with I. V. CR845 exhibited a statistically significant lower incidence of opioid-related AEs through 24 hours after the start of the first infusion compared to patients who received only placebo. The incidence of nausea was reduced by approximately 50% (only 26.1% of patients administered CR845 experienced nausea as compared to 51.2% for placebo, p<0.001) and the incidence of vomiting was reduced nearly 80% (only 1.7% of patients administered CR845 experienced vomiting, as compared to 8.3% for placebo, p=0.035). There was also less pruritus, or itching sensation, reported in patients treated with CR845 compared to placebo. Figure 6 depicts the percentage of patients reporting opioid- related adverse events of nausea, vomiting and pruritus.
In addition to the reduction of opioid-related adverse events, a standard responder analysis indicated that a higher percentage of patients who received I. V. CR845 were
characterized as "Responders" as compared to those receiving placebo (p=0.001). Responders included patients who rated their medication "Excellent" or "Very Good" and Non-Responders as those who rated their medication "Fair" or "Poor". The lower overall pain intensity scores at the end of the study period for CR845-treated patients and the significant reduction in nausea and vomiting reported in these patients contributed to patients' greater satisfaction with I.V. CR845 treatment compared to placebo. Figure 7 shows the number of Responder or Non-Responder patients in the I.V. CR845-treated patients compared to the patients receiving only placebo.
In this trial, intravenous administration of 0.04 mg/kg of I.V. CR845 pre- and/or postoperatively was safe and generally well tolerated. The placebo and CR845 treatment patient groups showed a similar overall incidence of treatment-emergent adverse events (TEAEs), the majority of which were mild to moderate in severity. The most frequent TEAEs, reported in 10% or more of total patients, were nausea, hypotension, flatulence, blood sodium increase, or hypernatremia, and headache. There were no apparent consistent differences between CR845 and placebo groups in clinical laboratory results, vital signs, electrocardiogram, or oxygen saturation results, with the exception of blood sodium increase, which was evident only in CR845 treatment groups (14% of total patients). The increase in blood sodium levels (hypernatremia), observed in CR845 treatment groups was likely a result of the aquaretic effect of I.V. CR845 at this dose and the replacement of fluid loss with sodium-containing I.V. solutions, rather than water or low/no sodium-containing fluids. In subsequent trials, fluid replacement with water or I.V. solutions with low or no sodium was used with no evidence of hypernatremia. CR845 for Bunionectomy
Bunionectomy is a surgical procedure to remove a bunion, an enlargement of the joint at the base of the big toe and includes bone and soft tissue. The procedure typically results in intense pain requiring postoperative analgesic care, usually beginning with local anesthetic infusion and ongoing administration of a strong opioid, such as morphine or fentanyl, for several days after surgery during which the patient often suffers from nausea and vomiting.
Clinical trial (CLIN2003) was a randomized, double-blind, placebo-controlled trial conducted in 51 patients following bunionectomy surgery at a single site in the U.S. The trial enrolled female and male patients, ages 18 years and older, scheduled for elective bunionectomy under regional anesthesia. Using a standard clinical trial protocol in which local anesthetic infusion was terminated on the day after surgery, patients were randomized into one of two treatment groups (CR845 or Placebo, in a 2: 1 ratio) after reporting moderate-to-severe pain, defined as a pain intensity score > 40 on a 100-point pain scale. Patients randomized to receive I.V. CR845 were administered an I.V. injection at a dose of 0.005 mg/kg, and additional doses on an as-needed basis 30-60 minutes later, and then no more frequently than every 8 hours through a 48-hour dosing period. The results were analyzed separately for the per protocol population, or "Completers", which includes only patients who completed the trial, and the modified Intent-to-Treat, or mITT, population, which includes Completers and all patients who discontinued the trial, or "non-Completers". In the Completer group, CR845 treatment resulted in a statistically significant reduction in pain intensity compared to placebo, as measured by the SPID score over the initial 24 hour time period (SPID0-24; p<0.05). This reduction in pain intensity after CR845 dosing was also statistically significant over a 36 hour time period (SPID0_ 36, p<0.03), as well as over the entire two-day dosing period (SPIDo-48, p<0.03), compared to placebo-treated patients (see Figure 8a). Numerical improvements in SPID scores in the CR845 group as compared to placebo were also evident across the same time periods when analyzing the ιηΓΓΤ population of Completers together with non-Completers (see Figure 8b).
The Completer analysis is indicative of the actual efficacy of I.V. CR845 under conditions where patients are exposed to the drug as specified in the protocol, while the ιηΓΓΤ analysis is indicative of the actual variability that will be encountered in the mITT populations. The understanding of this variability serves as the basis for determining the appropriate number of patients for enrollment in our Phase 3 clinical trials. In this trial, mean PID from baseline at each time interval was measured, and was numerically superior across the 48 hour trial period in the I.V. CR845 treatment group relative to the placebo group for both the Completer and mITT populations (see Figures 9a and 9b). Statistically significant reductions in pain intensity differences in the CR845 group versus placebo were evident in the 0-12 hour time interval for both the Completer and mITT populations (p<0.01 and p<0.05 respectively) and for the 0-36 hour time interval for the Completer populations (p<0.05), consistent with the findings with the primary SPID endpoints.
Fentanyl was available to both CR845 and placebo treatment groups upon patient request. While there was no difference in mean fentanyl use between the placebo and CR845 groups, the incidence of opioid-related AEs of nausea and vomiting was significantly reduced (by 60% and 80%, respectively; p<0.05) in patients who received CR845 compared to placebo during the 48 hour period after randomization (see Figure 10). Without wishing to be bound by theory, the ability of I.V. CR845 to reduce nausea and vomiting despite not meaningfully reducing fentanyl usage is believed to be due to a direct antiemetic effect resulting from its kappa opioid agonist mechanism of action. The ability to provide postsurgical analgesia and simultaneously reduce opioid-related side effects makes I.V. CR845 an attractive treatment option for postoperative patients and their physicians. In this bunionectomy trial, repeated intravenous administration of I.V. CR845 at a dose of 0.005 mg/kg was safe and generally well tolerated. The most frequent TEAEs (greater than 10%) observed in the CR845 treatment group were transient facial tingling and somnolence. Of the seven cases of somnolence reported, four were reported as "mild" and/or "related to drug" and three as "moderate" and/or "not related to drug". The mean plasma sodium concentration in CR845-treated patients exhibited an approximately 3% rise over 24 hours from baseline levels, but was not outside the normal physiological range at either 24 or 48 hours post-CR845 administration. This lack of clinically significant hypernatremia was likely a result of both utilizing a lower dose of I.V. CR845 and replacing transient fluid loss with oral water or sodium-free intravenous fluid. In addition, consistent with our prior studies, there was no evidence of acute psychiatric side effects that were observed with prior- generation CNS- active kappa opioid agonists.

Claims

CLAIMS:
1. A method for preventing, inhibiting or treating nausea and/or vomiting in a mammalian subject, the method comprising administering an effective amount of a peripherally-restricted kappa opioid receptor agonist to the mammalian subject.
2. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist comprises a peptide.
3. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist comprises one or more D-amino acids.
4. The method according to claim 1, wherein the peripherally restricted kappa opioid receptor agonist comprises a synthetic peptide amide having the formula:
XaaiXaaoXaa Xaa^i-W-Y . Z
Figure imgf000017_0001
Rr(V)e-R2
or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof;
wherein
Xaai is selected from the group consisting of (A)(A')D-Phe, (A)(A')(a-Me)D-Phe, D-Tyr, D-Tic, D-tert-leucine, D-neopentylglycine, D-phenylglycine, D-homophenylalanine, and P-(E)D-Ala, wherein each (A) and each (Α') are phenyl ring substituents independently selected from the group consisting of -H, -F, -CI, -N02, -CH3, -CF3, -CN, and -CONH2, and wherein each (E) is independently selected from the group consisting of cyclobutyl, cyclopentyl, cyclohexyl, pyridyl, thienyl and thiazolyl;
Xaa2 is selected from the group consisting of (A)(A')D-Phe, 3,4-dichloro-D-Phe, (A)(A')(a-Me)D-Phe, D-lNal, D-2Nal, D-Tyr, (E)D-Ala and D-Trp;
Xaa3 is selected from the group consisting of D-Nle, D-Phe, (E)D-Ala, D-Leu, (a-Me)D-Leu, D-Hle, D-Val, and D-Met; Xaa.4 is selected from the group consisting of (B)2D-Arg, (B)2D-Nar, (B)2D-Har,
Figure imgf000018_0001
D-Dap, s-(B)D-Lys, s-(B)2-D-Lys, D-Amf, amidino-D-Amf, y-(B)2D-Dbu, δ-(Β)2α- (B')D-Orn, D-2-amino-3(4-piperidyl)propionic acid, D-2-amino-3(2- aminopyrrolidyl)propionic acid, D-a-amino-P-amidinopropionic acid, a-amino-4- piperidineacetic acid, cis-a,4-diaminocyclohexane acetic acid, trans-a,4- diaminocyclohexaneacetic acid, cis-a-amino-4-methylaminocyclo-hexane acetic acid, trans-a-amino-4-methylaminocyclohexane acetic acid, a-amino-l-amidino-4- piperidineacetic acid, cis-a-amino-4-guanidinocyclohexane acetic acid, and trans-a- amino-4-guanidinocyclohexane acetic acid, wherein each (B) is independently selected from the group consisting of H and Ci-C4 alkyl, and (Β') is H or (a-Me);
W is selected from the group consisting of:
Null, provided that when W is null, Y is N;
-NH-(CH2)b- with b equal to zero, 1, 2, 3, 4, 5, or 6; and
-NH-(CH2)c-0- with c equal to 2, or 3, provided that Y is C;
the moiety
Figure imgf000018_0002
is an optionally substituted 4 to 8-membered heterocyclic ring moiety wherein all ring heteroatoms in said ring moiety are N; wherein Y and Z are each independently C or N; provided that when such ring moiety is a six, seven or eight-membered ring, Y and Z are separated by at least two ring atoms; and provided that when such ring moiety has a single ring heteroatom which is N, then such ring moiety is non-aromatic;
V is Q-C6 alkyl, and e is zero or 1, wherein when e is zero, then V is null and Ri and R2 are directly bonded to the same or different ring atoms;
wherein (i) Ri is selected from the group consisting of -H, -OH, halo, -CF3, -NH2, -COOH, Ci-C6 alkyl, C C6 alkoxy, amidino, C C6 alkyl-substituted amidino, aryl, optionally substituted heterocyclyl, Pro-amide, Pro, Gly, Ala, Val, Leu, He, Lys, Arg, Orn, Ser, Thr, -CN, -CONH2, -COR', -S02R', -CONR'R", -NHCOR', OR' and S02NR'R"; wherein said optionally substituted heterocyclyl is optionally singly or doubly substituted with substituents independently selected from the group consisting of C C6 alkyl, C C6 alkoxy, oxo, -OH, -CI, -F, -NH2, -N02, -CN, -COOH, and amidino; wherein R' and R" are each independently -H, Ci-C% alkyl, aryl, or heterocyclyl or R' and R" are combined to form a 4- to 8-membered ring, which ring is optionally singly or doubly substituted with substituents independently selected from the group consisting of C -C alkyl, -C1-C6 alkoxy, -OH, -CI, -F, -NH2, -N02, -CN, -COOH and amidino; and R2 is selected from the group consisting of -H, amidino, singly or doubly C1-C6 alkyl-substituted amidino, -CN, - CONH2, -CONR'R", -NHCOR', -S02NR'R" and -COOH; or
(ii) Ri and R2 taken together can form an optionally substituted 4- to 9-membered heterocyclic monocyclic or bicyclic ring moiety which is bonded to a single ring atom of the Y and Z-containing ring moiety; or
(iii) Ri and R2 taken together with a single ring atom of the Y and Z-containing ring moiety can form an optionally substituted 4- to 8-membered heterocyclic ring moiety to form a spiro structure; or
(iv) Ri and R2 taken together with two or more adjacent ring atoms of the Y and Z- containing ring moiety can form an optionally substituted 4- to 9-membered heterocyclic monocyclic or bicyclic ring moiety fused to the Y and Z-containing ring moiety;
wherein each of said optionally substituted 4-,
5-, 6,-, 7-, 8- and 9-membered heterocyclic ring moieties comprising Ri and R2 is optionally singly or doubly substituted with substituents independently selected from the group consisting of Ci-C6 alkyl, C - C6 alkoxy, optionally substituted phenyl, oxo, -OH, -CI, -F, -NH2, -N02, -CN, - COOH, and amidino;
provided that when the Y and Z-containing ring moiety is a six or seven membered ring having a single ring heteroatom and e is zero, then Ri is not -OH, and Ri and R2 are not both -H; and
provided further that when the Y and Z-containing ring moiety is a six membered ring having two ring heteroatoms, both Y and Z are N and W is null, then -(V)eRiR2 is attached to a ring atom other than Z; and if e is zero, then Ri and R2 are not both -H. e method of claim 4, wherein the moiety:
Figure imgf000020_0001
Ri-(V)e-R2 selected from the group consisting
Figure imgf000020_0002
Figure imgf000021_0001
Figure imgf000021_0002
6. The method of claim 4, wherein the synthetic peptide amide has the structure:
Figure imgf000021_0003
D-Phe-D-Phe-D-Leu-D-Lys-[ro(4-aminopiperidine-4-carboxylic acid)] -OH.
7. The method of claim 6, wherein the mammalian subject is a human.
8. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist is administered to the subject within 24 hours prior to, during, or within 24 hours after undergoing a medical procedure.
9. The method according to claim 8, wherein the medical procedure causes pain.
10. The method according to claim 6, wherein the peripherally-restricted kappa opioid receptor agonist is administered to the subject by a route of injection selected from the group consisting of subcutaneous injection, intravenous injection, intraperitoneal injection, intraarticular injection, and intramuscular injection.
11. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist is a non-narcotic analgesic.
12. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist is asimadoline (N-[(lS)-2-[(3S)-3-hydroxypyrrolidin-l-yl]-l-phenylethyl]-N- methyl-2,2-diphenylacetamide).
13. The method according to claim 1, wherein the peripherally-restricted kappa opioid receptor agonist is nalfurafine ((2E)-N-[(5a,6P)-17-(cyclopropylmethyl)-3,14-dihydroxy-4,5- epoxymorphinan- 6-yl]-3-(3-furyl)-N-methylacrylamide).
14. The method according to claim 1, wherein the mammal is a human.
15. The method according to claim 14, wherein the nausea and/or vomiting in the human occurs within 48 hours after administration of at least one dose of a mu opioid analgesic.
16. The method according to claim 15, wherein the mu opioid analgesic is administered to treat, inhibit or prevent hard tissue pain.
17. The method according to claim 16, wherein the hard tissue pain is bone pain.
18. The method according to claim 17, wherein the hard tissue pain is due to a medical procedure.
19. The method according to claim 15, wherein the peripherally-restricted kappa opioid receptor agonist is administered prior to administration of a first dose of the mu opioid analgesic.
20. The method according to claim 15, wherein the peripherally-restricted kappa opioid receptor agonist is administered after administration of at least one dose of the mu opioid analgesic.
PCT/US2014/062320 2013-10-28 2014-10-27 Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting WO2015065867A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/032,351 US20160250277A1 (en) 2013-10-28 2014-10-27 Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361896469P 2013-10-28 2013-10-28
US61/896,469 2013-10-28

Publications (2)

Publication Number Publication Date
WO2015065867A2 true WO2015065867A2 (en) 2015-05-07
WO2015065867A3 WO2015065867A3 (en) 2015-10-15

Family

ID=53005359

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/062320 WO2015065867A2 (en) 2013-10-28 2014-10-27 Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting

Country Status (3)

Country Link
US (1) US20160250277A1 (en)
TW (1) TW201601743A (en)
WO (1) WO2015065867A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016181408A2 (en) 2015-05-11 2016-11-17 Cadila Healthcare Limited NOVEL SHORT-CHAIN PEPTIDES AS KAPPA (κ) OPIOID RECEPTORS (KOR) AGONIST
CN107098876A (en) * 2016-02-23 2017-08-29 江苏恒瑞医药股份有限公司 Phenylpropionyl amine derivant, its preparation method and its in application pharmaceutically
WO2017211272A1 (en) * 2016-06-07 2017-12-14 江苏恒瑞医药股份有限公司 Phenyl propanamide derivative, and manufacturing method and pharmaceutical application thereof
WO2018188641A1 (en) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 Pharmaceutical composition containing mor agonist and kor agonist, and uses thereof
CN109422798A (en) * 2017-08-22 2019-03-05 江苏恒瑞医药股份有限公司 A kind of free alkali crystal form of phenylpropionyl amine derivant and preparation method thereof
WO2019109937A1 (en) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Use of kor agonist in combination with mor agonist in preparing drug for treating pain
WO2019109934A1 (en) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Salt of phenylpropionamide derivative and preparation method therefor
WO2019219019A1 (en) 2018-05-16 2019-11-21 江苏恒瑞医药股份有限公司 Pharmaceutical composition of kor receptor agonist
EP3521301A4 (en) * 2016-09-27 2020-05-27 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Polyamide compound and use thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230321265A1 (en) * 2020-05-29 2023-10-12 Cara Therapeutics, Inc. Kappa opioid receptor agonist-therapeutic antigen binding protein conjugate (ktac) compounds
AU2020454871A1 (en) 2020-06-25 2023-01-19 Humanwell Pharmaceutical US Peptides for treatment of medical disorders
CN115043904A (en) * 2021-03-08 2022-09-13 成都奥达生物科技有限公司 Long-acting K opioid receptor agonist

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL371596A1 (en) * 2002-05-17 2005-06-27 Merck Patent Gmbh Use of compounds that are effective as selective opiate receptor modulators

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016181408A2 (en) 2015-05-11 2016-11-17 Cadila Healthcare Limited NOVEL SHORT-CHAIN PEPTIDES AS KAPPA (κ) OPIOID RECEPTORS (KOR) AGONIST
CN107098876A (en) * 2016-02-23 2017-08-29 江苏恒瑞医药股份有限公司 Phenylpropionyl amine derivant, its preparation method and its in application pharmaceutically
CN107098876B (en) * 2016-02-23 2021-04-06 江苏恒瑞医药股份有限公司 Phenyl propionamide derivative, preparation method and medical application thereof
EP3466962A4 (en) * 2016-06-07 2020-01-08 Jiangsu Hengrui Medicine Co., Ltd. Phenyl propanamide derivative, and manufacturing method and pharmaceutical application thereof
WO2017211272A1 (en) * 2016-06-07 2017-12-14 江苏恒瑞医药股份有限公司 Phenyl propanamide derivative, and manufacturing method and pharmaceutical application thereof
CN108290926A (en) * 2016-06-07 2018-07-17 江苏恒瑞医药股份有限公司 Phenylpropionyl amine derivant, preparation method and its application in medicine
AU2017277003B2 (en) * 2016-06-07 2021-01-07 Jiangsu Hengrui Medicine Co., Ltd. Phenyl propanamide derivative, and manufacturing method and pharmaceutical application thereof
US11084847B2 (en) 2016-09-27 2021-08-10 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Polyamide compound and use thereof
EP3521301A4 (en) * 2016-09-27 2020-05-27 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Polyamide compound and use thereof
WO2018188641A1 (en) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 Pharmaceutical composition containing mor agonist and kor agonist, and uses thereof
CN109422798A (en) * 2017-08-22 2019-03-05 江苏恒瑞医药股份有限公司 A kind of free alkali crystal form of phenylpropionyl amine derivant and preparation method thereof
WO2019109934A1 (en) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Salt of phenylpropionamide derivative and preparation method therefor
WO2019109937A1 (en) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Use of kor agonist in combination with mor agonist in preparing drug for treating pain
US11180530B2 (en) 2017-12-06 2021-11-23 Jiangsu Hengrui Medicine Co., Ltd. Salt of phenylpropionamide derivative and preparation method therefor
US11471503B2 (en) 2017-12-06 2022-10-18 Jiangsu Hengrui Medicine Co., Ltd. Use of KOR agonist in combination with MOR agonist in preparing drug for treating pain
WO2019219019A1 (en) 2018-05-16 2019-11-21 江苏恒瑞医药股份有限公司 Pharmaceutical composition of kor receptor agonist

Also Published As

Publication number Publication date
TW201601743A (en) 2016-01-16
US20160250277A1 (en) 2016-09-01
WO2015065867A3 (en) 2015-10-15

Similar Documents

Publication Publication Date Title
US20160250277A1 (en) Peripheral kappa opioid receptor agonists for preventing, inhibiting or treating nausea and vomiting
Schmidt Alvimopan (ADL 8-2698) is a novel peripheral opioid antagonist
US8268821B2 (en) Methods and compositions
AU756120B2 (en) Compositions and methods for reducing respiratory depression and attendant side effects of mu opioid compounds
TWI263496B (en) Pharmaceutical combinations and their use in treating gastrointestinal disorders
CA2678481C (en) Improved medicinal compositions comprising buprenorphine and naltrexone
DK2574167T3 (en) LIQUID NOSE SPRAY CONTAINING low-dose naltrexone
US20050038062A1 (en) Methods and materials for the treatment of pain comprising opioid antagonists
JP2011173931A (en) Method for reducing pain
CA2678582A1 (en) Improvements in and relating to medicinal compositions
WO2007120485A2 (en) Methods of treating pain with alkylxanthines and antiepileptics and compositions for use therefor
TW201200521A (en) Therapeutic or prophylactic agent for bile duct disease
US20180028594A1 (en) Peripheral kappa opioid receptor agonists for hard tissue pain
JP5577102B2 (en) Improved pharmaceutical composition comprising buprenorphine and nalmefene
CN1668294A (en) Use of nefopam for the treatment of nausea or emesis
JP2013040206A (en) Method for stimulating motion of digestive system by using ipamorelin
TW201118084A (en) The use of an opioid receptor antagonist for the treatment or prevention of gastrointestinal tract disorders
JP2004525096A (en) Novel medical uses of thienylcyclohexylamine derivatives
RU2622980C1 (en) Means for effective cupping of acute and/or chronic pain syndrome and method of its application
TW202216145A (en) Ameliorating agent or prophylactic agent for muscle weakness symptom in disease or syndrome associated with metabolic disorder
KR20130092470A (en) Composition for preventing or curing neuropathic pain comprising substance p
AU2004298288B2 (en) Methods and compositions
WO2019200061A1 (en) Anesthetic compounds
Lee et al. Management of Surgical Pain
AU2014201782A1 (en) Improved medicinal compositions comprising buprenorphine and naltrexone

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14857434

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 15032351

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14857434

Country of ref document: EP

Kind code of ref document: A2