Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Pain Symptom Manage ; 67(1): 39-49.e5, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37757956

ABSTRACT

CONTEXT: µ-opioid receptor gene (OPRM1) A118G polymorphism (rs1799971) causes loss of N-glycosylation sites at the extracellular domain of µ-opioid receptors. G-allele carriers show a limited response to morphine; however, studies investigating the impact of A118G polymorphism on the efficacy of opioids other than morphine are limited. OBJECTIVE: To compare the impact of A118G polymorphism on the efficacy of various opioids. METHODS: This prospective cohort study enrolled 222 in-patients administered one of the following opioid therapies for cancer pain as part of an opioid introduction or rotation strategy: tapentadol extended-release tablets, methadone tablets, hydromorphone controlled-release tablets, oxycodone controlled-release tablets, or transdermal fentanyl patches. The impact of A118G polymorphism on the difference in the Brief Pain Inventory-Short Form score on days three, seven, and 14 from baseline was compared among the groups. RESULTS: Overall, 81, 74, and 67 patients had the AA, AG, and GG genotypes, respectively, with an OPRM1 A118G G-allele variant frequency of 0.47. The reduction in the Brief Pain Inventory-Short Form score after opioid therapy initiation did not differ significantly among the patients with the three A118G genotypes treated with tapentadol (p = 0.84) or methadone (p = 0.97), whereas it was significantly smaller in G-allele carriers than that in AA homozygous patients treated with hydromorphone (p < 0.001), oxycodone (p = 0.031), or fentanyl (p < 0.001). CONCLUSION: Tapentadol and methadone may be more suitable than hydromorphone, oxycodone, and fentanyl for G-allele carriers due to their dual mechanism of action and low susceptibility to OPRM1 A118G polymorphism.


Subject(s)
Analgesics, Opioid , Cancer Pain , Humans , Analgesics, Opioid/therapeutic use , Cancer Pain/drug therapy , Delayed-Action Preparations , Fentanyl/therapeutic use , Hydromorphone/therapeutic use , Methadone/therapeutic use , Oxycodone/therapeutic use , Polymorphism, Single Nucleotide/genetics , Prospective Studies , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/therapeutic use , Tapentadol/therapeutic use
2.
Free Radic Biol Med ; 212: 94-116, 2024 02 20.
Article in English | MEDLINE | ID: mdl-38142953

ABSTRACT

Opioids regulate various physiological and pathophysiological functions, including cell proliferation, immune function, obesity, and neurodegenerative disorders. They have been used for centuries as a treatment for severe pain, binding to opioid receptors a specific G protein-coupled receptor. Common opioids, like ß-endorphin, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO), and dynorphins, have analgesic effects. The use of a potent antagonist, like naltrexone hydrochloride, to block the effects of mu Opioid Receptor (µOR) may result in the withdrawal of physiological effects and could potentially impact immune responses in many diseases including respiratory disease. Asthma is a respiratory disease characterized by airway hyperresponsiveness, inflammation, bronchoconstriction, chest tightness, stress generation and release of various cytokines. Airway inflammation leads recruitment and activation of immune cells releasing mediators, including opioids, which may modulate inflammatory response by binding to their respective receptors. The study aims to explore the role of µOR antagonist (naltrexone) in regulating asthma pathophysiology, as the regulation of immune and inflammatory responses in asthma remains unclear. Balb/c mice were sensitized intranasally by 1% TDI and challenged with 2.5% TDI. Naltrexone hydrochloride (1 mg/kg body weight) was administered through intraperitoneal route 1 h before TDI induction. Blocking µOR by naltrexone exacerbates airway inflammation by recruiting inflammatory cells (lymphocytes and neutrophils), enhancing intracellular Reactive oxygen species in bronchoalveolar lavage fluid (BALF), and inflammatory mediator (histamine, Eosinophil peroxidase and neutrophil elastase) in lungs. Naltrexone administration modulated inflammatory cytokines (TNF-α, IL-4, IL-5, IL-6, IL-10, and IL-17A), and enhanced IgE and CRP levels. Naltrexone administration also increased the expression of NF-κB, and phosphorylated p-P38, p-Erk, p-JNK and NF-κB by inhibiting the µOR. Docking study revealed good binding affinity of naltrexone with µOR compared to δ and κ receptors. In future it might elucidate potential therapeutic against many respiratory pathological disorders. In conclusion, µOR blocking by naltrexone regulates and implicates inflammation, bronchoconstriction, and lung physiology.


Subject(s)
Asthma , Naltrexone , Animals , Mice , Naltrexone/pharmacology , Naltrexone/therapeutic use , NF-kappa B/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/therapeutic use , Disease Models, Animal , Asthma/drug therapy , Inflammation/pathology , Lung/metabolism , Cytokines/metabolism , Oxidative Stress , Mice, Inbred BALB C , Ovalbumin
3.
Reprod Toxicol ; 119: 108403, 2023 08.
Article in English | MEDLINE | ID: mdl-37196679

ABSTRACT

Opioids remain the most powerful analgesics for moderate to severe pain but their clinical use, misuse and abuse has been an alarming medical problem, especially for those users at child-bearing age. Mu-opioid receptor (MOR) biased agonists have been suggested as superior alternatives with better therapeutic ratios. We recently discovered and characterized a novel MOR biased agonist, LPM3480392, which demonstrates robust analgesic effect, favorable pharmacokinetic performance, and mild respiratory suppression in vivo. To understand the safety profile of LPM3480392 on the reproductive system and embryonic development, this study evaluated the effects of LPM3480392 on the fertility and early embryonic development, embryo-fetal development, and pre- and postnatal development in rats. Results showed that LPM3480392 had mild effects on parental male and female animals, accompanied by subtle early embryonic loss and delayed ossification of fetal development during organogenesis period. In addition, although minor effects were found on normal developmental milestones and behaviors in the pups, there was no evidence of malformed effect. In conclusion, these results suggest that LPM3480392 has a favorable safety profile with only minor effects on the reproductive and developmental outcomes in animals, which support the development of LPM3480392 as a novel analgesic.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Pregnancy , Rats , Male , Animals , Female , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/therapeutic use , Analgesics, Opioid/toxicity , Analgesics/therapeutic use , Pain/drug therapy , Reproduction
4.
J Exp Anal Behav ; 119(2): 392-406, 2023 03.
Article in English | MEDLINE | ID: mdl-36759567

ABSTRACT

Opioid overdose and opioid use disorder continue to be significant public health challenges despite the availability of effective medications and significant efforts at all levels of society. The emergence of highly potent and efficacious opioids such as fentanyl and its derivatives over the last decade has only exacerbated what was already a substantial problem. Behavioral pharmacology research has proven invaluable for understanding the effects of drugs as well as developing and evaluating pharmacotherapies for disorders involving the central nervous system, including substance abuse disorders. This paper describes a program of research characterizing a potent, selective, and long-lasting mu opioid receptor antagonist, methocinnamox, and evaluating its potential for treating opioid overdose and opioid use disorder. Studies in rodents and nonhuman primates demonstrate that methocinnamox prevents and reverses opioid-induced ventilatory depression and selectively blocks opioid self-administration. This work, taken together with rigorous in vitro and ex vivo studies investigating methocinnamox neuropharmacology, lays a solid foundation for the therapeutic utility of this potentially life-saving medication. Moreover, these studies demonstrate how rigorous behavioral pharmacological studies can be integrated in a broader drug discovery and development research program.


Subject(s)
Opiate Overdose , Opioid-Related Disorders , Animals , Analgesics, Opioid/pharmacology , Opiate Overdose/drug therapy , Opioid-Related Disorders/drug therapy , Morphine Derivatives/pharmacology , Morphine Derivatives/therapeutic use , Receptors, Opioid, mu/therapeutic use
5.
Environ Toxicol Pharmacol ; 95: 103979, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36174909

ABSTRACT

The study aimed to examine the genetic contribution to buprenorphine (BUP) treatment in individuals with opioid use disorder (OUD), with a specific focus on BDNF and OPRM1 genes. A total of 113 controls and 111 OUD patients receiving sublingual BUP/naloxone were enrolled. OPRM1 A118G and BDNF Val66Met polymorphisms were investigated by PCR-FRLP. Plasma BDNF and beta-endorphin levels were assessed by ELISA kits in both groups. Blood BUP levels were measured by LC-MS/MS and normalized with daily BUP dose (BUP/D). OPRM1 A118G and BDNF Val66Met polymorphisms didn't have an effect on plasma beta-endorphin and BDNF levels in OUD patients, respectively. Interestingly, OUD patients had significantly higher plasma BDNF and lower beta-endorphin levels compared to the controls (p < 0.001). A negative and significant correlation between plasma BUP/D and BDNF levels was found. Age onset of first use was associated with OPRM1 A118G polymorphism. The findings indicated that sublingual BUP/naloxone may increase plasma BDNF levels, but may decrease beta-endorphin levels in individuals with OUD. Plasma BDNF level seemed to be decreased in a BUP/D concentration-dependent manner.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Brain-Derived Neurotrophic Factor/genetics , Buprenorphine/therapeutic use , Buprenorphine, Naloxone Drug Combination/therapeutic use , Chromatography, Liquid , Humans , Opioid-Related Disorders/complications , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/therapeutic use , Tandem Mass Spectrometry , beta-Endorphin/genetics , beta-Endorphin/therapeutic use
6.
CNS Drugs ; 36(6): 617-632, 2022 06.
Article in English | MEDLINE | ID: mdl-35616826

ABSTRACT

Opioids are widely used in chronic pain management, despite major concerns about their risk of adverse events, particularly abuse, misuse, and respiratory depression from overdose. Multi-mechanistic opioids, such as tapentadol and buprenorphine, have been widely studied as a valid alternative to traditional opioids for their safer profile. Special interest was focused on the role of the nociceptin opioid peptide (NOP) receptor in terms of analgesia and improved tolerability. Nociceptin opioid peptide receptor agonists were shown to reinforce the antinociceptive effect of mu opioid receptor (MOR) agonists and modulate some of their adverse effects. Therefore, multi-mechanistic opioids involving both MOR and NOP receptor activation became a major field of pharmaceutical and clinical investigations. Buprenorphine was re-discovered in a new perspective, as an atypical analgesic and as a substitution therapy for opioid use disorders; and buprenorphine derivatives have been tested in animal models of nociceptive and neuropathic pain. Similarly, cebranopadol, a full MOR/NOP receptor agonist, has been clinically evaluated for its potent analgesic efficacy and better tolerability profile, compared with traditional opioids. This review overviews pharmacological mechanisms of the NOP receptor system, including its role in pain management and in the development of opioid tolerance. Clinical data on buprenorphine suggest its role as a safer alternative to traditional opioids, particularly in patients with non-cancer pain; while data on cebranopadol still require phase III study results to approve its introduction on the market. Other bifunctional MOR/NOP receptor ligands, such as BU08028, BU10038, and AT-121, are currently under pharmacological investigations and could represent promising analgesic agents for the future.


Subject(s)
Analgesics, Opioid , Buprenorphine , Analgesics, Opioid/adverse effects , Animals , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Drug Tolerance , Humans , Isoquinolines , Naltrexone/analogs & derivatives , Opioid Peptides/therapeutic use , Pain/drug therapy , Phenylpropionates , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/therapeutic use , Nociceptin
7.
Article in English | MEDLINE | ID: mdl-35206575

ABSTRACT

Humans have used opioids to suppress moderate to severe pain for thousands of years. However, the long-term use of opioids has several adverse effects, such as opioid tolerance, opioid-induced hyperalgesia, and addiction. In addition, the low efficiency of opioids in controlling neuropathic pain limits their clinical applications. Combining nonopioid analgesics with opioids to target multiple sites along the nociceptive pathway may alleviate the side effects of opioids. This study reviews the feasibility of reducing opioid side effects by regulating the transient receptor potential vanilloid 1 (TRPV1) receptors and summarizes the possible underlying mechanisms. Blocking and activating TRPV1 receptors can improve the therapeutic profile of opioids in different manners. TRPV1 and µ-opioid receptors are bidirectionally regulated by ß-arrestin2. Thus, drug combinations or developing dual-acting drugs simultaneously targeting µ-opioid and TRPV1 receptors may mitigate opioid tolerance and opioid-induced hyperalgesia. In addition, TRPV1 receptors, especially expressed in the dorsal striatum and nucleus accumbens, participate in mediating opioid reward, and its regulation can reduce the risk of opioid-induced addiction. Finally, co-administration of TRPV1 antagonists and opioids in the primary action sites of the periphery can significantly relieve neuropathic pain. In general, the regulation of TRPV1 may potentially ameliorate the side effects of opioids and enhance their analgesic efficacy in neuropathic pain.


Subject(s)
Analgesics, Opioid , Neuralgia , Analgesics, Opioid/adverse effects , Drug Tolerance , Humans , Neuralgia/chemically induced , Neuralgia/drug therapy , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/therapeutic use , TRPV Cation Channels/metabolism
8.
Article in English | MEDLINE | ID: mdl-33252118

ABSTRACT

AIMS: The aim of this study was to compare the efficacy, consumption and safety after piritramide administered either intramuscularly (IM) on demand or via patient-controlled intravenous analgesia (PCA) and to examine the impact of OPRM1 and ABCB1 gene polymorphisms on the drug efficacy/safety in both regimens. METHODS: One hundred and four patients scheduled for elective inguinal hernioplasty received piritramide with PCA or IM for postoperative pain management. We evaluated piritramide consumption, pain intensity using visual analogue scale (VAS) and adverse effects. RESULTS: Median (IQR) piritramide consumption was 18.5 (13.5-31.2) and 15.0 (15.0-15.0) mg in the PCA and IM groups, respectively (P=0.0092). The respective values of area under the VAS2-16-time curve were 40 and 280 mm.h (P=0.0027). Opioid-induced adverse effects were more frequent in the PCA than in the IM group. Variant OPRM1 allele was associated with decreased pain relief, increased opioid consumption and increased incidence of adverse effects, while ABCB1 polymorphisms showed no impact on the observed parameters. CONCLUSIONS: We observed higher piritramide consumption, better pain relief and slightly worse safety profile in the PCA group compared with IM administration. Variant OPRM1 118G allele carriers required higher opioid dosing and suffered from more adverse effects, however, the differences between genotypes have been less pronounced in the PCA patients likely due to improved pain management via PCA.


Subject(s)
Analgesia, Patient-Controlled , Pirinitramide , ATP Binding Cassette Transporter, Subfamily B/genetics , Analgesics, Opioid , Humans , Pain, Postoperative/drug therapy , Pirinitramide/therapeutic use , Polymorphism, Genetic , Receptors, Opioid, mu/therapeutic use
9.
J Nat Prod ; 84(1): 71-80, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33326237

ABSTRACT

The seeds of the akuamma tree (Picralima nitida) have been used as a traditional treatment for pain and fever. Previous studies have attributed these effects to a series of indole alkaloids found within the seed extracts; however, these pharmacological studies were significantly limited in scope. Herein, an isolation protocol employing pH-zone-refining countercurrent chromatography was developed to provide six of the akuamma alkaloids in high purity and quantities sufficient for more extensive biological evaluation. Five of these alkaloids, akuammine (1), pseudo-akuammigine (3), akuammicine (4), akuammiline (5), and picraline (6), were evaluated against a panel of >40 central nervous system receptors to identify that their primary targets are the opioid receptors. Detailed in vitro investigations revealed 4 to be a potent kappa opioid receptor agonist, and three alkaloids (1-3) were shown to have micromolar activity at the mu opioid receptor. The mu opioid receptor agonists were further evaluated for analgesic properties but demonstrated limited efficacy in assays of thermal nociception. These findings contradict previous reports of the antinociceptive properties of the P. nitida alkaloids and the traditional use of akuamma seeds as analgesics. Nevertheless, their opioid-preferring activity does suggest the akuamma alkaloids provide distinct scaffolds from which novel opioids with unique pharmacologic properties and therapeutic utility can be developed.


Subject(s)
Alkaloids/pharmacology , Analgesics/therapeutic use , Apocynaceae/chemistry , Indoles/pharmacology , Receptors, Opioid, mu/therapeutic use , Terpenes/pharmacology , Alkaloids/chemistry , Alkaloids/isolation & purification , Analgesics/chemistry , Animals , Indoles/chemistry , Indoles/isolation & purification , Receptors, Opioid, kappa , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/analysis , Secologanin Tryptamine Alkaloids/chemistry , Seeds/chemistry , Terpenes/chemistry , Terpenes/isolation & purification
11.
Curr Opin Nephrol Hypertens ; 29(6): 671-680, 2020 11.
Article in English | MEDLINE | ID: mdl-32941189

ABSTRACT

PURPOSE OF REVIEW: This review evaluates current recommendations for pain management in chronic kidney disease (CKD) and end-stage kidney disease (ESKD) with a specific focus on evidence for opioid analgesia, including the partial agonist, buprenorphine. RECENT FINDINGS: Recent evidence supports the use of physical activity and other nonpharmacologic therapies, either alone or with pharmacological therapies, for pain management. Nonopioid analgesics, including acetaminophen, topical analgesics, gabapentinoids, serotonin-norepinephrine reuptake inhibitors, and TCA may be considered based on pain cause and type, with careful dose considerations in kidney disease. NSAIDs may be used in CKD and ESKD for short durations with careful monitoring. Opioid use should be minimized and reserved for patients who have failed other therapies. Opioids have been associated with increased adverse events in this population, and thus should be used cautiously after risk/benefit discussion with the patient. Opioids that are safer to use in kidney disease include oxycodone, hydromorphone, fentanyl, methadone, and buprenorphine. Buprenorphine appears to be a promising and safer option due to its partial agonism at the mu opioid receptor. SUMMARY: Pain is poorly managed in patients with kidney disease. Nonpharmacological and nonopioid analgesics should be first-line approaches for pain management. Opioid use should be minimized with careful monitoring and dose adjustment.


Subject(s)
Pain Management , Analgesics, Opioid/therapeutic use , Buprenorphine/administration & dosage , Fentanyl/administration & dosage , Fentanyl/adverse effects , Humans , Hydromorphone/administration & dosage , Kidney Failure, Chronic/drug therapy , Oxycodone/administration & dosage , Pain/drug therapy , Receptors, Opioid, mu/therapeutic use , Renal Insufficiency, Chronic/drug therapy
12.
Pain Med ; 21(5): 992-1004, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31165885

ABSTRACT

OBJECTIVE: Although mu-opioid receptor agonists have been the mainstay of analgesic regimens for moderate to severe pain, they are associated with serious side effects, risks, and limitations. We evaluate the most serious risks associated with conventional opioids and compare these with the pharmacology of CYT-1010, a prototypical endomorphin and mu-opioid receptor agonist. RESULTS: Addiction and respiratory depression are serious risks of traditional mu-opioid analgesics. Mitigation strategies have been inadequate at addressing the opioid crisis and may interfere with the effective treatment of pain. Improved understanding of mu-opioid receptor biology and the discovery in 1997 of an additional and unique family of endogenous opioid peptides (endomorphins) have provided a pathway for dissociating analgesia from opioid-related adverse events and developing new classes of mu-opioid receptor agonists that use biased signaling and/or target novel sites to produce analgesia with reduced side effect liability. Endomorphin-1 and -2 are endogenous opioid peptides highly selective for mu-opioid receptors that exhibit potent analgesia with reduced side effects. CYT-1010 is a cyclized, D-lysine-containing analog of endomorphin-1 with a novel mechanism of action targeting traditional mu- and exon 11/truncated mu-opioid receptor 6TM variants. CYT-1010 preclinical data have demonstrated reduced abuse potential and analgesic potency exceeding that of morphine. In an initial phase 1 clinical study, CYT-1010 demonstrated significant analgesia vs baseline and no respiratory depression at the dose levels tested. CONCLUSIONS: CYT-1010 and other novel mu-opioid receptor agonists in clinical development are promising alternatives to conventional opioids that may offer the possibility of safer treatment of moderate to severe pain.


Subject(s)
Receptors, Opioid, mu , Respiratory Insufficiency , Analgesics/therapeutic use , Analgesics, Opioid/therapeutic use , Humans , Morphine/therapeutic use , Pain/drug therapy , Receptors, Opioid, mu/therapeutic use , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy
13.
Bioorg Med Chem Lett ; 29(15): 1938-1942, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31147104

ABSTRACT

We discovered a novel compound, 5-methyl-1,4,5,7-tetrahydro-2,5-ethanoazocino[4,3-b]indol-6(3H)-one sulfuric acid salt (DS39201083), which was formed by derivatization of a natural product, conolidine. DS39201083 had a unique bicyclic skeleton and was a more potent analgesic than conolidine, as revealed in the acetic acid-induced writhing test and formalin test in ddY mice. The compound showed no agonist activity at the mu opioid receptor.


Subject(s)
Analgesics, Opioid/therapeutic use , Indole Alkaloids/therapeutic use , Receptors, Opioid, mu/therapeutic use , Analgesics, Opioid/pharmacology , Animals , Indole Alkaloids/pharmacology , Mice , Receptors, Opioid, mu/agonists
14.
Sci Rep ; 5: 17543, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633001

ABSTRACT

Cell-derived exosomes have been demonstrated to be efficient carriers of small RNAs to neighbouring or distant cells, highlighting the preponderance of exosomes as carriers for gene therapy over other artificial delivery tools. In the present study, we employed modified exosomes expressing the neuron-specific rabies viral glycoprotein (RVG) peptide on the membrane surface to deliver opioid receptor mu (MOR) siRNA into the brain to treat morphine addiction. We found that MOR siRNA could be efficiently packaged into RVG exosomes and was associated with argonaute 2 (AGO2) in exosomes. These exosomes efficiently and specifically delivered MOR siRNA into Neuro2A cells and the mouse brain. Functionally, siRNA-loaded RVG exosomes significantly reduced MOR mRNA and protein levels. Surprisingly, MOR siRNA delivered by the RVG exosomes strongly inhibited morphine relapse via the down-regulation of MOR expression levels. In conclusion, our results demonstrate that targeted RVG exosomes can efficiently transfer siRNA to the central nervous system and mediate the treatment of morphine relapse by down-regulating MOR expression levels. Our study provides a brand new strategy to treat drug relapse and diseases of the central nervous system.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Glycoproteins/genetics , Morphine Dependence/therapy , Peptide Fragments/genetics , Receptors, Opioid, mu/genetics , Viral Proteins/genetics , Animals , Exosomes/genetics , Gene Expression Regulation/genetics , Glycoproteins/administration & dosage , Humans , Mice , Morphine/metabolism , Morphine Dependence/genetics , Morphine Dependence/pathology , Neurons/metabolism , Neurons/pathology , Peptide Fragments/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Receptors, Opioid, mu/therapeutic use , Viral Proteins/administration & dosage
15.
Cochrane Database Syst Rev ; (9): CD011460, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26403220

ABSTRACT

BACKGROUND: A large proportion of people with advanced cancer will experience moderate to severe pain. Tapentadol is a novel, centrally acting analgesic medicine acting at the µ-opioid receptor and inhibiting noradrenaline reuptake. The efficacy of tapentadol is stated to be comparable to morphine and oxycodone. OBJECTIVES: To assess the analgesic efficacy of tapentadol for the relief of cancer pain in adults, and the adverse events associated with its use in clinical trials. SEARCH METHODS: We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, and EMBASE from January 2005 to July 2015, together with reference lists of retrieved papers and review articles, and two clinical trial registries. Searches started from 2005 because this covered the period during which clinical trials were conducted. We contacted the manufacturer of tapentadol in the UK to find additional trials not identified by electronic searches. We did not restrict searches by language. SELECTION CRITERIA: We included randomised controlled trials (RCTs) of tapentadol compared with placebo or active controls in adults with moderate to severe cancer pain. Pain had to be measured using a validated assessment tool, and studies had to include at least 10 participants per treatment arm. DATA COLLECTION AND ANALYSIS: Two review authors independently extracted data using a standard form and assessed risk of bias. We extracted available data on study design, participant details, interventions, and outcomes, including analgesic outcome measures, withdrawals, and adverse events. MAIN RESULTS: We included four studies with 1029 participants. All the studies used a parallel-group design, and included an initial titration phase to determine the maximum effective and tolerated dose, followed by a maintenance phase. Tapentadol medication was taken twice daily and doses ranged from 50 to 500 mg per day. Rescue medication (morphine or oxycodone immediate-release) was available to participants in all studies.Overall, 440 participants were randomised in classically designed RCTs, and 589 participants were enrolled in enriched-enrolment, randomised-withdrawal (EERW) trials. A total of 476 participants were randomised to titration with tapentadol and 338 participants took tapentadol throughout the maintenance phase of their trial.All studies used numerical rating scores, Patient Global Impression of Change scores, and use of rescue medication as measures of efficacy, and all reported on adverse events and withdrawals.All studies enrolled fewer than 200 participants per treatment arm and were therefore at risk of overestimating efficacy. One study was terminated early due to problems with supply of rescue medication, with fewer than 20 participants enrolled per treatment arm in the maintenance phase of the trial. We judged another study at high risk of bias due to an open-label design.There were insufficient data for pooling and statistical analysis. Response rates for pain intensity were comparable across treatment groups in each study. In one EERW study, response rates were high across both treatment and placebo arms during the maintenance phase (62% tapentadol, 69% morphine, 50% placebo). For pain relief, tapentadol is no more and no less effective than oxycodone or morphine (low quality evidence).Treatment emergent adverse event rates were high, approximately 50% to 90%. The most common adverse events were gastrointestinal (nausea, vomiting, constipation) (low quality evidence). There was no advantage of tapentadol over morphine or oxycodone in terms of serious adverse events. The number of people experiencing effects on consciousness, appetite, or thirst was low. AUTHORS' CONCLUSIONS: Information from RCTs on the effectiveness and tolerability of tapentadol was limited. The available studies were of moderate or small size and used different designs, which prevented pooling of data. Pain relief and adverse events were comparable between the tapentadol and morphine and oxycodone groups.


Subject(s)
Neoplasms/complications , Pain/drug therapy , Phenols/therapeutic use , Receptors, Opioid, mu/therapeutic use , Adult , Humans , Randomized Controlled Trials as Topic , Tapentadol
16.
Drug Ther Bull ; 50(3): 30-3, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22419754

ABSTRACT

Tapentadol (Palexia - Grünenthal) is a recently introduced strong analgesic with µ-agonistic opioid and additional noradrenaline reuptake inhibition properties. The summary of product characteristics (SPC) states that it is indicated for the relief of moderate to severe acute pain in adults, which can be adequately managed only with opioid analgesics.1 Here we review the place of tapentadol in the treatment of patients with moderate to severe acute pain.


Subject(s)
Acute Pain/prevention & control , Analgesics, Opioid/therapeutic use , Phenols/therapeutic use , Receptors, Opioid, mu/therapeutic use , Delayed-Action Preparations , Drug Approval , Drug Costs , Humans , Phenols/adverse effects , Phenols/economics , Practice Guidelines as Topic , Randomized Controlled Trials as Topic , Tapentadol
17.
Aliment Pharmacol Ther ; 32(7): 884-93, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20839388

ABSTRACT

BACKGROUND: The short-term effects of methylnaltrexone (MNTX), a peripherally acting mu-opioid receptor antagonist, on gastrointestinal and colonic transit remain unclear. AIM: To compare the effects of placebo, codeine, subcutaneous (s.c.) MNTX and codeine with s.c. MNTX on gastrointestinal and colonic transit of solids in healthy humans. METHODS: In a randomized, parallel-group, double-blind, placebo-controlled trial of 48 healthy volunteers, effects of 6 consecutive days of placebo [s.c. and p.o. (orally), n = 8], codeine (p.o. 30 mg q.d.s., n = 8), MNTX (s.c. 0.30 mg/kg, n = 16) and combined MNTX and codeine (same doses and routes, n = 16) on gastrointestinal and colonic transit were assessed. A validated scintigraphic method was used to measure transit during the last 48 h of treatment. Bowel function was estimated during treatment as well as 1 week preceding treatment using standard diaries. Analysis of covariance was used to assess treatment effects. RESULTS: Codeine delayed colonic transit [geometric centre at 24 h (P = 0.04) and ascending colon t(1/2) (P = 0.02)] and reduced stool frequency (P = 0.002), but had no effect on stool form. MNTX did not affect transit, stool frequency or stool form, either alone or with codeine (P > 0.3). No drug interaction effects were detected (P > 0.15). CONCLUSION: Methylnaltrexone does not alter gastrointestinal or colonic transit and does not reverse acute codeine-associated delayed gut transit in health.


Subject(s)
Analgesics, Opioid/therapeutic use , Codeine/therapeutic use , Gastrointestinal Transit/drug effects , Naltrexone/analogs & derivatives , Narcotic Antagonists/therapeutic use , Receptors, Opioid, mu/antagonists & inhibitors , Adolescent , Adult , Double-Blind Method , Drug Combinations , Female , Gastrointestinal Tract/drug effects , Humans , Male , Middle Aged , Naltrexone/therapeutic use , Quaternary Ammonium Compounds/therapeutic use , Receptors, Opioid, mu/therapeutic use , Young Adult
18.
J Am Acad Dermatol ; 63(4): 680-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20462660

ABSTRACT

During the past two decades, systemic µ-opioid receptor antagonists (MORA) have been used in the treatment of various forms of chronic pruritus. In a number of case reports, case series, and controlled trials, treatment with MORA has demonstrated considerable antipruritic effects. In double-blind controlled studies, significant antipruritic relief has been achieved by MORA in cholestatic pruritus, chronic urticaria, and atopic dermatitis. In case reports and case series, antipruritic efficacy of MORA has been reported in prurigo nodularis, mycosis fungoides, postburn pruritus, aquagenic pruritus, hydroxyethyl starch-induced pruritus, and pruritus of unknown origin. However, most of the evidence remains anecdotal, the design of these trials varies, and comparison of results is difficult. In this review we aim to present an overview of these reports and to assess the evidence for the antipruritic action of the drugs naloxone, nalmefene, and naltrexone, which are currently in use for the treatment of chronic pruritus of different origins. We will also evaluate recommendations for the use of MORA in daily medical practice.


Subject(s)
Antipruritics/therapeutic use , Pruritus/drug therapy , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/therapeutic use , Administration, Oral , Antipruritics/adverse effects , Chronic Disease , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Male , Naloxone/adverse effects , Naloxone/therapeutic use , Naltrexone/adverse effects , Naltrexone/analogs & derivatives , Naltrexone/therapeutic use , Pruritus/diagnosis , Randomized Controlled Trials as Topic , Risk Assessment , Severity of Illness Index , Treatment Outcome
19.
Arthritis Rheum ; 56(6): 2038-48, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17530644

ABSTRACT

OBJECTIVE: To evaluate opioid receptor function as a basis for novel antinociceptive therapy in arthritis. METHODS: We induced human mu-opioid receptor (HuMOR) expression in arthritic joints of mice, using the feline immunodeficiency virus (FIV) vector, which is capable of stably transducing dividing, growth-arrested, and terminally differentiated cells. Male and female Col1-IL-1beta(XAT)-transgenic mice developed on a C57BL/6J background and wild-type littermates were studied. RESULTS: A single injection of FIV(HuMOR) into the temporomandibular joints of Col1-IL-1beta(XAT)-transgenic mice 1 week prior to induction of arthritis prevented the development of orofacial pain and joint dysfunction, and reduced the degree of histopathologic abnormality in the joint. In addition, FIV(HuMOR) prevented the attendant sensitization of trigeminal sensory neurons and activation of astroglia in brainstem trigeminal sensory nuclei. These effects were mediated by the transduction of primary sensory neurons via transport of FIV vectors from peripheral nerve endings to sensory ganglia, as evidenced by HuMOR expression in neuronal cell bodies located in the trigeminal ganglia, as well as in their proximal and distal nerve branches located in the main sensory and subnucleus caudalis of the brainstem and joints, respectively. The presence of MOR ligands predominantly in the descending trigeminal nucleus suggested that the observed antinociception occurred at the subnucleus caudalis. Articular chondrocytes and meniscal tissue were also infected by FIV(HuMOR), which presumably exerted an antiinflammatory effect on cartilage. CONCLUSION: Our results indicate that prophylactic therapy with MOR overexpression in joints can successfully prevent the development of pain, dysfunction, and histopathologic abnormalities in the joints in arthritis. These findings may provide a basis for the future development of spatiotemporally controlled antinociceptive and antiinflammatory therapy for arthritis.


Subject(s)
Interleukin-1beta/physiology , Matrix Metalloproteinase 2/physiology , Osteoarthritis/physiopathology , Pain/prevention & control , Peptide Fragments/physiology , Receptors, Opioid, mu/metabolism , Temporomandibular Joint Disorders/prevention & control , Animals , Disease Models, Animal , Female , Humans , Immunodeficiency Virus, Feline , Injections, Intra-Articular , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 2/genetics , Mice , Mice, Transgenic , Neurons, Afferent/physiology , Osteoarthritis/complications , Osteoarthritis/genetics , Pain/drug therapy , Pain/etiology , Peptide Fragments/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/therapeutic use , Temporomandibular Joint/metabolism , Temporomandibular Joint/physiopathology , Temporomandibular Joint Disorders/drug therapy , Temporomandibular Joint Disorders/etiology , Transduction, Genetic , Trigeminal Nuclei/pathology , Trigeminal Nuclei/physiopathology
20.
Alcohol ; 18(2-3): 225-33, 1999.
Article in English | MEDLINE | ID: mdl-10456575

ABSTRACT

Numerous pharmacological and other studies have implicated both Mmu and dopamine receptor subtypes in alcohol consumption. In the genetic drinking rat as well as those chemically induced to drink, evidence has accrued that the abnormal intake of alcohol is underpined by these receptors in the brain. The purpose of this investigation was to demonstrate unequivocally that a biological impairment by antisense oligodeoxynucleotide (ODN) targeted specifically to these two receptor subtypes would disrupt ongoing alcohol drinking. In this project, a new strain of female and male high-ethanol preferring (HEP) rats was used that had free access to preferred concentrations of alcohol over water in a two choice paradigm. A guide cannula for a microinjection needle was first implanted bilaterally above the nucleus accumbens (NAC) of each rat. Following recovery, a dose of either 250 or 500 ng of the Mmu ODN or 500 ng D2ODN was microinjected into the NAC of the rat in a volume of 0.8-1.0 microl. A standard temporal sequence was used in which microinjections were given four times at successive 12-h intervals over a 2-day interval. The control mismatch ODNs corresponding to both the Mmu or D2 receptor antisense were microinjected identically at homologous sites in the NAC. Following the experiments, the brain of each rat was removed and sectioned in the coronal plane for histological analysis so that each microinjection site was identified. The results showed that the Mmu receptor antisense caused a significant dose dependent fall in free access alcohol drinking within 12 to 24 h following the initial microinjection. This decline often persisted for 1 to 2 days in terms of both g/kg intake and proportion of alcohol to water consumed. Similarly, the D2 receptor ODN likewise induced an intense and significant decline in both g/kg and proportion measures of alcohol intake. Since the corresponding mismatch ODN for both Mmu and D2 receptors exerted no effect on either of these measures of alcohol consumption, the specificity of molecular action of the respective antisense molecules on drinking behavior of the HEP rats was confirmed. Thus, these results provide the first unequivocal evidence that the genes for D2 and Mmu receptors are fundamentally involved in abnormal alcohol drinking in the genetically predisposed individual. Finally, important new anatomical evidence is introduced for the critical role of the NAC in the genetic basis of aberrant drinking of alcohol.


Subject(s)
Alcohol Drinking/drug therapy , Alcoholism/genetics , Oligonucleotides, Antisense/therapeutic use , Receptors, Dopamine D2/therapeutic use , Receptors, Opioid, mu/therapeutic use , Alcoholism/drug therapy , Animals , Breeding , Female , Male , Nucleus Accumbens/drug effects , Oligonucleotides, Antisense/genetics , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/genetics , Receptors, Opioid, mu/genetics
SELECTION OF CITATIONS
SEARCH DETAIL