1
|
Miura R, Utano T, Miura Y, Chiba K, Hasegawa A, Takafuji Y, Takahashi H, Tanzawa A, Iwahashi K, Yamatani A, Yotani N. Efficacy and Safety of Naldemedine for Opioid-Induced Constipation in Children. J Palliat Med 2024; 27:1354-1358. [PMID: 39122251 DOI: 10.1089/jpm.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024] Open
Abstract
Background: Naldemedine, a peripherally acting opioid μ receptor antagonist, is effective for prevention of opioid-induced constipation (OIC); however, evidence on its use in children is limited. Objective: To evaluate the efficacy and safety of naldemedine in pediatric patients with OIC. Design, Setting/Subjects: Retrospective analysis of 32 pediatric patients with OIC treated with naldemedine in a single institution in Japan from June 2017 to March 2021. Measurements: Efficacy was evaluated in 13 evaluable patients with bowel movement (BM) response, defined as those with at least three BMs in the first 7 days after naldemedine initiation and an increase of at least one BM from baseline. Safety was evaluated by examining adverse events (AEs) based on the Common Terminology Criteria for AEs (v5.0). Results: BM response was recorded in 11 of the 13 patients (85%), and the number BMs per day significantly increased from 0.43 before naldemedine to 1.00 after naldemedine (p = 0.025). The most common AE was diarrhea, observed in 16 of the 32 patients (50%), and all instances were grade 1 or 2. In three of the 16 patients, naldemedine was discontinued owing to worsening diarrhea. Conclusions: In pediatric patients, naldemedine resulted in a high rate of BM response and increased the BM frequency, indicating its efficacy. In some patients, grade 2 diarrhea required naldemedine discontinuation, suggesting that it should be used with caution in pediatric patients. Further studies are warranted to determine the optimal naldemedine dose in pediatric patients.
Collapse
Affiliation(s)
- Rina Miura
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Tomoyuki Utano
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Yoriko Miura
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Kyoko Chiba
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Ayaka Hasegawa
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Yukiko Takafuji
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Hayato Takahashi
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Ayano Tanzawa
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Kana Iwahashi
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Akimasa Yamatani
- Department of Pharmaceuticals, National Center for Child Health and Development, Tokyo, Japan
| | - Nobuyuki Yotani
- Division of Palliative Medicine, National Center of Child Health and Development, Tokyo, Japan
| |
Collapse
|
2
|
Imai H, Fujita Y, Hiruta E, Masuno T, Yamazaki S, Tanaka H, Kamiya T, Sandoh M, Takei S, Arai K, Nishiba H, Mogi J, Koizuka S, Saito T, Obayashi K, Kaira K, Minato K. Efficacy and safety of naldemedine for opioid-induced constipation in older patients with cancer: a retrospective study. Eur J Gastroenterol Hepatol 2024; 36:571-577. [PMID: 38477855 DOI: 10.1097/meg.0000000000002746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
BACKGROUND Opioids are pain relievers that are often associated with opioid-induced constipation (OIC) that worsens with age. We performed a multicenter, retrospective analysis on the efficacy and safety of naldemedine, an opioid receptor antagonist, in treating OIC in patients with cancer (age >75 years). METHODS The electronic medical records of cancer patients who received naldemedine at 10 Japanese institutions between 7 June 2017 and August 31, 2019, were retrieved. Patients aged ≥75 years who were treated with naldemedine for the first time and hospitalized for at least 7 days before and after initiating naldemedine therapy were included in this analysis. RESULTS Sixty patients were observed for at least 7 days before and after starting naldemedine. The response rate was 68.3%, and the frequency of bowel movements increased significantly after naldemedine administration in the overall population ( P < 0.0001) and among those who defecated <3 times/week before naldemedine administration ( P < 0.0001). Diarrhea was the most frequent adverse event in all grades, observed in 45% of patients, of which 92.6% were Grade 1 or 2. Grade 4 or higher adverse events, including death, were not observed. CONCLUSION Naldemedine exhibits significant efficacy and safety in OIC treatment in older patients with cancer.
Collapse
Affiliation(s)
- Hisao Imai
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center
| | | | - Eriko Hiruta
- Division of Pharmacy, Gunma Prefectural Cancer Center, Ota
| | | | | | - Hajime Tanaka
- Division of Pharmacy, Haramachi Red Cross Hospital, Agatsuma-gun
| | - Teruhiko Kamiya
- Department of Pharmacy, Tatebayashi Kosei General Hospital, Tatebayashi
| | | | | | - Kazuya Arai
- Division of Pharmacy, Gunma Saiseikai Maebashi Hospital, Maebashi
| | - Hiromi Nishiba
- Division of Pharmacy, Japan Community Health Care Organization (JCHO) Gunma Chuo Hospital, Maebashi
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare
| | - Junnosuke Mogi
- Division of Pharmacy, Hidaka Hospital. 886 Nakao, Takasaki
| | - Shiro Koizuka
- Division of Palliative Care, Gunma Prefectural Cancer Center, Ota, Gunma, Japan
| | - Taeko Saito
- Division of Pharmacy, Gunma Prefectural Cancer Center, Ota
| | - Kyoko Obayashi
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama
| | - Koichi Minato
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center
| |
Collapse
|
3
|
Wang L, Gharibani P, Yang Y, Guo Y, Yin J. Regulation of enteric nervous system via sacral nerve stimulation in opioid-induced constipated rats. Front Neurosci 2023; 17:1146883. [PMID: 37332864 PMCID: PMC10272359 DOI: 10.3389/fnins.2023.1146883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Objectives Sacral nerve stimulation (SNS) has been employed for treating constipation. However, its mechanisms involving enteric nervous system (ENS) and motility are largely unknown. In this study, we investigated the possible ENS involvement of SNS in treating Loperamide-induced constipation in rats. Methods Experiment-1 was designed to study the effects of acute SNS on whole colon transit time (CTT). In experiment-2, we induced constipation by Loperamide and then applied daily SNS or sham-SNS for 1 week. Choline acetyltransferase (ChAT), nitric oxide synthase (nNOS), and PGP9.5 in colon tissue were examined at the end of the study. Moreover, survival factors such as phosphorylated AKT (p-AKT) and Glial cell-derived neurotrophic factor (GDNF) were measures by immunohistochemistry (IHC) and western blot (WB). Key results (1) SNS with one set of parameters shortened CTT starting at 90 min after phenol red administration (p < 0.05). (2) While Loperamide induced slow transit constipation with a significant reduction in fecal pellet number and feces wet weight, daily SNS for a week resolved constipation. (3) Moreover, SNS was able to shorten whole gut transit time comparing to sham-SNS (p = 0.01). (4) Loperamide reduced the number of PGP9.5 and ChAT positive cells, and downregulated ChAT protein expression and upregulated nNOS protein expression, whereas these detrimental effects were significantly reversed by SNS. (5) Furthermore, SNS increased expressions of both GDNF and p-AKT in colon tissue. (6) Vagal activity was reduced following Loperamide (p < 0.01); yet SNS normalized vagal activity. Conclusion SNS with appropriate parameters improves opioid-induced constipation and reversed the detrimental effects of Loperamide on enteric neurons possibly via the GDNF-PI3K/Akt pathway.GRAPHICAL ABSTRACT.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Payam Gharibani
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yi Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Guo
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jieyun Yin
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Yamada M, Jimaru Y, Torii S, Mitsuba N, Takahashi K. A Retrospective Observational Study of Factors Affecting the Efficacy of Concurrent Prescription of Naldemedine for Opioid-Induced Constipation Caused by Oxycodone Tablets. Biol Pharm Bull 2023; 46:1826-1831. [PMID: 38044102 DOI: 10.1248/bpb.b23-00487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Naldemedine is indicated for the treatment of opioid-induced constipation (OIC), but reports on its efficacy in preventing OIC are few. Therefore, we retrospectively investigated factors affecting the efficacy of concurrent prescription of naldemedine on OIC. Outpatients with cancer who were started on oxycodone 10 mg/d were included in the study. The eligible patients were classified by their physicians into the following three groups: Group A used regular laxatives before the introduction of oxycodone and initiated naldemedine treatment simultaneously with oxycodone administration, Group B did not take laxatives before the introduction of oxycodone and started naldemedine simultaneously with oxycodone administration, and Group C had been administering regular laxatives before the introduction of oxycodone and were not prescribed naldemedine simultaneously with oxycodone treatment. The Support Team Assessment Schedule Japanese edition score for constipation, frequency of defecation, Bristol Stool Form Scale, sense of incomplete rectal evacuation, and development or worsening of straining to pass bowel movements were compared among the three groups before and after oxycodone administration. In Group B, there was significant worsening of the four parameters except for the sense of incomplete rectal evacuation, whereas Groups A and C did not present any changes. In logistic regression analysis, body weight ≥51.8 kg was a factor significantly decreasing the preventive effect of naldemedine on OIC, and regular use of laxatives was a factor significantly increasing the preventive effect of naldemedine on OIC. Thus, the initiation of naldemedine should be considered depending on the body weight and regular laxative use.
Collapse
Affiliation(s)
- Masami Yamada
- Department of Pharmacy, Osaka-fu, Saiseikai Noe Hospital
| | - Yumi Jimaru
- Department of Pharmacy, Osaka-fu, Saiseikai Noe Hospital
| | - Sari Torii
- Department of Pharmacy, Osaka-fu, Saiseikai Noe Hospital
| | - Naoko Mitsuba
- Department of Pharmacy, Osaka-fu, Saiseikai Noe Hospital
| | | |
Collapse
|
5
|
Candy B, Jones L, Vickerstaff V, Larkin PJ, Stone P. Mu-opioid antagonists for opioid-induced bowel dysfunction in people with cancer and people receiving palliative care. Cochrane Database Syst Rev 2022; 9:CD006332. [PMID: 36106667 PMCID: PMC9476137 DOI: 10.1002/14651858.cd006332.pub4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Opioid-induced bowel dysfunction (OIBD) is characterised by constipation, incomplete evacuation, bloating, and gastric reflux. It is one of the major adverse events (AEs) of treatment for pain in cancer and palliative care, resulting in increased morbidity and reduced quality of life. This review is a partial update of a 2008 review, and critiques as previous update (2018) trials only for people with cancer and people receiving palliative care. OBJECTIVES To assess for OIBD in people with cancer and people receiving palliative care the effectiveness and safety of mu-opioid antagonists (MOAs) versus different doses of MOAs, alternative pharmacological/non-pharmacological interventions, placebo, or no treatment. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, CINAHL, and Web of Science (December 2021), clinical trial registries and regulatory websites. We sought contact with MOA manufacturers for further data. SELECTION CRITERIA Randomised controlled trials (RCTs) assessing the effectiveness and safety of MOAs for OIBD in people with cancer and people at a palliative stage irrespective of the type of terminal disease. DATA COLLECTION AND ANALYSIS Two review authors assessed risk of bias and extracted data. The appropriateness of combining data from the trials depended upon sufficient homogeneity across trials. Our primary outcomes were laxation response, effect on analgesia, and AEs. We assessed the certainty of evidence using GRADE and created summary of findings tables. MAIN RESULTS We included 10 studies (two new trials) randomising in-total 1343 adults with cancer irrespective of stage, or at palliative care stage of any disease. The MOAs were oral naldemedine and naloxone (alone or in combination with oxycodone), and subcutaneous methylnaltrexone. The trials compared MOAs with placebo, MOAs at different doses, or in combination with other drugs. Two trials of naldemedine and three of naloxone with oxycodone were in people with cancer irrespective of disease stage. The trial on naloxone alone was in people with advanced cancer. Four trials on methylnaltrexone were in palliative care where most participants had advanced cancer. All trials were vulnerable to biases; most commonly, blinding of the outcome assessor was not reported. Oral naldemedine versus placebo Risk (i.e. chance) of spontaneous laxations in the medium term (over two weeks) for naldemedine was over threefold greater risk ratio (RR) 2.00, 95% confidence interval (CI) 1.59 to 2.52, 2 trials, 418 participants, I² = 0%. Number needed to treat for an additional beneficial outcome (NNTB) 3, 95% CI 3 to 4; moderate-certainty evidence). Earlier risk of spontaneous laxations and patient assessment of bowel change was not reported. Very low-certainty evidence showed naldemedine had little to no effect on opioid withdrawal symptoms. There was little to no difference in the risk of serious (non-fatal) AEs (RR 3.34, 95% CI 0.85 to 13.15: low-certainty evidence). Over double the risk of AEs (non-serious) reported with naldemedine (moderate-certainty evidence). Low-dose oral naldemedine versus higher dose Risk of spontaneous laxations was lower for the lower dose (medium term, 0.1 mg versus 0.4 mg: RR 0.69, 95% CI 0.53 to 0.89, 1 trial, 111 participants (low-certainty evidence)). Earlier risk of spontaneous laxations and patient assessment of bowel change not reported. Low-certainty evidence showed little to no difference on opioid withdrawal symptoms (0.1 mg versus 0.4 mg mean difference (MD) -0.30, 95% CI -0.85 to 0.25), and occurrences of serious AEs (0.1 mg versus 0.4 mg RR 0.25, 95% CI 0.03 to 2.17). Low-certainty evidence showed little to no difference on non-serious AEs. Oral naloxone versus placebo Risk of spontaneous laxations and AEs not reported. Little to no difference in pain intensity (very low-certainty evidence). Full data not given. The trial reported that no serious AEs occurred. Oral naloxone + oxycodone versus oxycodone Risk of spontaneous laxations within 24 hours and in the medium term not reported. Low-certainty evidence showed naloxone with oxycodone reduced the risk of opioid withdrawal symptoms. There was little to no difference in the risk of serious (non-fatal) AEs (RR 0.68, 95% CI 0.44 to 1.06), 3 trials, 362 participants, I² = 55%: very low-certainty evidence). There was little to no difference in risk of AEs (low-certainty evidence). Subcutaneous methylnaltrexone versus placebo Risk of spontaneous laxations within 24 hours with methylnaltrexone was fourfold greater than placebo (RR 2.97, 95% CI 2.13 to 4.13. 2 trials, 287 participants, I² = 31%. NNTB 3, 95% CI 2 to 3; low-certainty evidence). Risk of spontaneous laxations in the medium term was over tenfold greater with methylnaltrexone (RR 8.15, 95% CI 4.76 to 13.95, 2 trials, 305 participants, I² = 47%. NNTB 2, 95% CI 2 to 2; moderate-certainty evidence). Low-certainty evidence showed methylnaltrexone reduced the risk of opioid withdrawal symptoms, and did not increase risk of a serious AE (RR 0.59, 95% CI 0.38 to 0.93. I² = 0%; 2 trials, 364 participants). The risk of AEs was higher for methylnaltrexone (low-certainty evidence). Lower-dose subcutaneous methylnaltrexone versus higher dose There was little to no difference in risk of spontaneous laxations in the medium-term (1 mg versus 5 mg or greater: RR 2.91, 95% CI 0.82 to 10.39; 1 trial, 26 participants very low-certainty evidence), or in patient assessment of improvement in bowel status (RR 0.98, 95% CI 0.71 to 1.35, 1 trial, 102 participants; low-certainty evidence). Medium-term assessment of spontaneous laxations and serious AEs not reported. There was little to no difference in symptoms of opioid withdrawal (MD -0.25, 95% CI -0.84 to 0.34, 1 trial, 102 participants) or occurrence of AEs (low-certainty evidence). AUTHORS' CONCLUSIONS This update's findings for naldemedine and naloxone with oxycodone have been strengthened with two new trials, but conclusions have not changed. Moderate-certainty evidence for oral naldemedine on risk of spontaneous laxations and non-serious AEs suggests in people with cancer that naldemedine may improve bowel function over two weeks and increase the risk of AEs. There was low-certainty evidence on serious AEs. Moderate-certainty evidence for methylnaltrexone on spontaneous laxations over two weeks suggests subcutaneous methylnaltrexone may improve bowel function in people receiving palliative care, but certainty of evidence for AEs was low. More trials are needed, more evaluation of AEs, outcomes patients rate as important, and in children.
Collapse
Affiliation(s)
- Bridget Candy
- Marie Curie Palliative Care Research Department, Division of Psychiatry, University College London, London, UK
| | - Louise Jones
- Marie Curie Palliative Care Research Department, Division of Psychiatry, University College London, London, UK
| | - Victoria Vickerstaff
- Marie Curie Palliative Care Research Department, Division of Psychiatry, University College London, London, UK
| | - Philip J Larkin
- UCD School of Nursing, Midwifery and Health Systems and Our Lady's Hospice and Care Services, UCD College of Health Sciences, Dublin, Ireland
| | - Patrick Stone
- Marie Curie Palliative Care Research Department, Division of Psychiatry, University College London, London, UK
| |
Collapse
|
6
|
Inagaki M, Kanemasa T, Yokota T. Naldemedine: Peripherally Acting Opioid Receptor Antagonist for Treating Opioid-induced Adverse Effects. Curr Top Med Chem 2020; 20:2830-2842. [PMID: 32648846 DOI: 10.2174/1568026620666200710105953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/31/2020] [Accepted: 06/19/2020] [Indexed: 11/22/2022]
Abstract
Opioids are widely used for pain management in moderate-to-severe pain. However, opioids are associated with adverse events, such as constipation and emesis/vomiting. To reduce these undesired effects, a structure-activity relationship study of morphinan derivatives was conducted, and a promising lead compound with inhibitory effects on opioid receptors was obtained. Further improvement in the potency and pharmacokinetic profiles of the lead compound led to the discovery of naldemedine, which showed anti-constipation and anti-emetic effects against these adverse events that were induced by morphine without influencing morphine's analgesic effect. Naldemedine was launched in Japan and the USA in 2017 and in the EU in 2019, for treating opioid-induced constipation.
Collapse
Affiliation(s)
- Masanao Inagaki
- Medicinal Chemistry Research Laboratory, Shionogi Co., Ltd. Osaka, Japan
| | - Toshiyuki Kanemasa
- Corporate Strategy Division, Corporate Planning Department, Shionogi Co., Ltd. Osaka, Japan
| | - Takaaki Yokota
- Project Management Department, Shionogi Co., Ltd. Global Development Division, Osaka, Japan
| |
Collapse
|
7
|
Pergolizzi JV, Christo PJ, LeQuang JA, Magnusson P. The Use of Peripheral μ-Opioid Receptor Antagonists (PAMORA) in the Management of Opioid-Induced Constipation: An Update on Their Efficacy and Safety. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1009-1025. [PMID: 32210534 PMCID: PMC7075239 DOI: 10.2147/dddt.s221278] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Peripherally acting μ-opioid receptor antagonists (PAMORAs) constitute a class of drugs which reverse opioid-induced constipation (OIC) with similar opioid analgesic effects. OIC differs from other forms of constipation in that it is an iatrogenic condition that occurs when an opioid acts on the dense network of μ-opioid receptors in the enteric system, which affect a variety of functions including gastrointestinal motility, secretion, and other factors that can cause bowel dysfunction. Unfortunately, laxative products, bowel regimens, dietary changes, and lifestyle modifications have limited effectiveness in preventing OIC, Opioid-associated adverse effect which occurs in 40% to 80% of opioid patients and may led to cessation of the treatment. PAMORAs are μ-receptor opioid antagonists specifically developed so that they have very limited ability to cross the blood-brain barrier and thus they are able to antagonize peripheral but not central μ-opioid receptors. PAMORAs are designed to have no effect on the analgesic benefits of opioid pain relievers but to relieve but antagonizing the effects of the opioid in the gastrointestinal system. The three main PAMORAS are methyltrexone (oral or parenteral), naldemedine (oral only), and naloxegol (oral only). Clinical studies demonstrate the safety and efficacy of these agents for alleviating constipation without diminishing the analgesic effect of opioid therapy. The aim of this narrative review to update the current status of PAMORAs for treating OIC in terms of safety and efficacy.
Collapse
Affiliation(s)
| | - Paul J Christo
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medicine, Baltimore, ML, USA
| | | | - Peter Magnusson
- Cardiology Research Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden.,Centre for Research and Development, Uppsala University/Region, Gävleborg, Sweden
| |
Collapse
|
8
|
Long DD, Armstrong SR, Beattie DT, Campbell CB, Church TJ, Colson PJ, Dalziel SM, Jacobsen JR, Jiang L, Obedencio GP, Rapta M, Saito D, Stergiades I, Tsuruda PR, Van Dyke PM, Vickery RG. Discovery of Axelopran (TD-1211): A Peripherally Restricted μ-Opioid Receptor Antagonist. ACS Med Chem Lett 2019; 10:1641-1647. [PMID: 31857840 DOI: 10.1021/acsmedchemlett.9b00406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
The effects of opioids in the central nervous system (CNS) provide significant benefit in the treatment of pain but can also lead to physical dependence and addiction, which has contributed to a growing opioid epidemic in the United States. Gastrointestinal dysfunction is an additional serious consequence of opioid use, and this can be treated with a localized drug distribution of a non-CNS penetrant, peripherally restricted opioid receptor antagonist. Herein, we describe the application of Theravance's multivalent approach to drug discovery coupled with a physicochemical property design strategy by which the N-substituted-endo-3-(8-aza-bicyclo[3.2.1]oct-3-yl)-phenyl carboxamide series of μ-opioid receptor antagonists was optimized to afford the orally absorbed, non-CNS penetrant, Phase 3 ready clinical compound axelopran (TD-1211) 19i as a potential treatment for opioid-induced constipation.
Collapse
Affiliation(s)
- Daniel D. Long
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Scott R. Armstrong
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - David T. Beattie
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Christina B. Campbell
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Timothy J. Church
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Pierre-Jean Colson
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Sean M. Dalziel
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - John R. Jacobsen
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Lan Jiang
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Glenmar P. Obedencio
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Miroslav Rapta
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Daisuke Saito
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Ioanna Stergiades
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Pamela R. Tsuruda
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Priscilla M. Van Dyke
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| | - Ross G. Vickery
- Theravance Biopharma US, Inc., 901 Gateway Blvd., South San Francisco, California 94080, United States
| |
Collapse
|
9
|
Inagaki M, Kume M, Tamura Y, Hara S, Goto Y, Haga N, Hasegawa T, Nakamura T, Koike K, Oonishi S, Kanemasa T, Kai H. Discovery of naldemedine: A potent and orally available opioid receptor antagonist for treatment of opioid-induced adverse effects. Bioorg Med Chem Lett 2019; 29:73-77. [DOI: 10.1016/j.bmcl.2018.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/22/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
|
10
|
Suzuki T, Sawada T, Kawai K, Ishihara Y. Pharmacological profile of TAN-452, a novel peripherally acting opioid receptor antagonist for the treatment of opioid-induced bowel syndromes. Life Sci 2018; 215:246-252. [DOI: 10.1016/j.lfs.2018.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023]
|
11
|
Could Perioperative Opioid Use Increase the Risk of Cancer Progression and Metastases? Int Anesthesiol Clin 2018; 54:e1-e16. [PMID: 27602710 DOI: 10.1097/aia.0000000000000112] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
12
|
Qi S, Lai H, Zhang Y, Dong Q, Zhu X. Chinese herbal medicine for opioid induced constipation in cancer patients: Protocol for a systematic review. Medicine (Baltimore) 2018; 97:e12594. [PMID: 30278572 PMCID: PMC6181624 DOI: 10.1097/md.0000000000012594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Opioid induced constipation (OIC) is a symptom that is commonly encountered in cancer pain management. Chinese herbal medicine (CHM) has been widely used to improve OIC in China. Many clinical trials indicate that CHM could improve OIC. In this systematic review, we aim to evaluate the effectiveness and safety of CHM for OIC in cancer patients. METHODS We will search the following electronic databases for randomized controlled trials to evaluate the effectiveness and safety of CHM for OIC in cancer patients: CENTRAL, EMBASE, MEDILINE, CINAHL and China National Knowledge Infrastructure. Each database will be searched from inception to June 2018. The entire process will include study selection, data extraction, risk of bias assessment and meta-analyses. RESULTS This proposed study will evaluate the effectiveness and safety of CHM for OIC. The outcomes will include change in bowel movements, quality of life and adverse events. CONCLUSIONS This proposed systematic review will evaluate the existing evidence on the effectiveness and safety of CHM for OIC in cancer patients. DISSEMINATION AND ETHICS The results of this review will be disseminated through peer-reviewed publication. Because all of the data used in this systematic review and meta-analysis has been published, this review does not require ethical approval. Furthermore, all data will be analyzed anonymously during the review process.
Collapse
Affiliation(s)
- Shuo Qi
- Dongzhimen Hospital Affiliate to Beijing University of Chinese Medicine, Beijing, China
- The Chinese Medicine Center, Collaboration Between Beijing University of Chinese Medicine and Western Sydney University
| | - Hezheng Lai
- School of Health and Science, Western Sydney University, Campbelltown, New South Wales
- The Chinese Medicine Center, Collaboration Between Beijing University of Chinese Medicine and Western Sydney University
| | - Yayue Zhang
- Dongzhimen Hospital Affiliate to Beijing University of Chinese Medicine, Beijing, China
| | - Qing Dong
- Dongzhimen Hospital Affiliate to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshu Zhu
- School of Health and Science, Western Sydney University, Campbelltown, New South Wales
- The Chinese Medicine Center, Collaboration Between Beijing University of Chinese Medicine and Western Sydney University
| |
Collapse
|
13
|
Candy B, Jones L, Vickerstaff V, Larkin PJ, Stone P. Mu-opioid antagonists for opioid-induced bowel dysfunction in people with cancer and people receiving palliative care. Cochrane Database Syst Rev 2018; 6:CD006332. [PMID: 29869799 PMCID: PMC6513061 DOI: 10.1002/14651858.cd006332.pub3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Opioid-induced bowel dysfunction (OIBD) is characterised by constipation, incomplete evacuation, bloating, and gastric reflux. It is one of the major adverse events of treatment for pain in cancer and in palliative care, resulting in increased morbidity and reduced quality of life.This is an update of two Cochrane reviews. One was published in 2011, Issue 1 on laxatives and methylnaltrexone for the management of constipation in people receiving palliative care; this was updated in 2015 and excluded methylnaltrexone. The other was published in 2008, Issue 4 on mu-opioid antagonists (MOA) for OIBD. In this updated review, we only included trials on MOA (including methylnaltrexone) for OIBD in people with cancer and people receiving palliative care. OBJECTIVES To assess the effectiveness and safety of MOA for OIBD in people with cancer and people receiving palliative care. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, CINAHL, and Web of Science to August 2017. We also searched clinical trial registries and regulatory websites. We contacted manufacturers of MOA to identify further data. SELECTION CRITERIA We included randomised controlled trials (RCTs) that assessed the effectiveness and safety of MOA for OIBD in people with cancer and people at a palliative stage irrespective of the type of terminal disease they experienced. DATA COLLECTION AND ANALYSIS Two review authors assessed risk of bias and extracted data. The appropriateness of combining data from the trials depended upon sufficient homogeneity across the trials. Our primary outcomes were laxation, impact on pain relief, and adverse events. Impact on pain relief was a primary outcome because a possible adverse effect of MOAs is a reduction in pain relief from opioids. We assessed the evidence on these outcomes using GRADE. MAIN RESULTS We identified four new trials for this update, bringing the total number included in this review to eight. In total, 1022 men and women with cancer irrespective of stage or at a palliative care stage of any disease were randomised across the trials. The MOAs evaluated were oral naldemedine and naloxone (alone or in combination with oxycodone), and subcutaneous methylnaltrexone. The trials compared with MOA with a placebo or with the active intervention administered at different doses or in combination with other drugs. The trial of naldemedine and the two of naloxone in combination with oxycodone were in people with cancer irrespective of disease stage. The trial on naloxone alone was in people with advanced cancer. The four trials on methylnaltrexone were undertaken in palliative care where most participants had cancer. All trials were vulnerable to biases; four were at a high risk as they involved a sample of fewer than 50 participants per arm.In the trial of naldemedine compared to placebo in 225 participants, there were more spontaneous laxations over the two-week treatment for the intervention group (risk ratio (RR) 1.93, 95% confidence intervals (CI) 1.36 to 2.74; moderate-quality evidence). In comparison with higher doses, lower doses resulted in fewer spontaneous laxations (0.1 mg versus 0.2 mg: RR 0.73, 95% CI 0.55 to 0.95; 0.1 mg versus 0.4 mg: RR 0.69, 95% CI 0.53 to 0.89; moderate-quality evidence). There was moderate-quality evidence that naldemedine had no effect on opiate withdrawal. There were five serious adverse events. All were in people taking naldemedine (low-quality evidence). There was an increase in the occurrence of other (non-serious) adverse events in the naldemedine groups (RR 1.36, 95% CI 1.04 to 1.79, moderate-quality evidence). The most common adverse event was diarrhoea.The trials on naloxone taken either on its own, or in combination with oxycodone (an opioid) compared to oxycodone only did not evaluate laxation response over the first two weeks of administration. There was very low-quality evidence that naloxone alone, and moderate-quality evidence that oxycodone/naloxone, had no effect on analgesia. There was low-quality evidence that oxycodone/naloxone did not increase the risk of serious adverse events and moderate-quality evidence that it did not increase risk of adverse events.In combined analysis of two trials of 287 participants, we found methylnaltrexone compared to placebo induced more laxations within 24 hours (RR 2.77, 95% CI 1.91 to 4.04. I² = 0%; moderate-quality evidence). In combined analysis, we found methylnaltrexone induced more laxation responses over two weeks (RR 9.98, 95% CI 4.96 to 20.09. I² = 0%; moderate-quality evidence). The proportion of participants who had a rescue-free laxation response within 24 hours of the first dose was 59.1% in the methylnaltrexone arms and 19.1% in the placebo arm. There was moderate-quality evidence that the rate of opioid withdrawal was not affected. Methylnaltrexone did not increase the likelihood of a serious adverse event; there were fewer in the intervention arm (RR 0.59, 95% CI 0.38 to 0.93; I² = 0%; moderate-quality evidence). There was no difference in the proportion of participants experiencing an adverse event (RR 1.17, 95% CI 0.94 to 1.45; I² = 74%; low-quality evidence). Methylnaltrexone increased the likelihood of abdominal pain and flatulence.Two trials compared differing methylnaltrexone schedules of higher doses with lower doses. For early laxation, there was low-quality evidence of no clear difference between doses on analgesia and adverse events. Both trials measured laxation response within 24 hours of first dose (trial one: RR 0.82, 95% CI 0.41 to 1.66; trial two: RR 1.07, 95% CI 0.81 to 1.42). AUTHORS' CONCLUSIONS In this update, the conclusions for naldemedine are new. There is moderate-quality evidence to suggest that, taken orally, naldemedine improves bowel function over two weeks in people with cancer and OIBD but increases the risk of adverse events. The conclusions on naloxone and methylnaltrexone have not changed. The trials on naloxone did not assess laxation at 24 hours or over two weeks. There is moderate-quality evidence that methylnaltrexone improves bowel function in people receiving palliative care in the short term and over two weeks, and low-quality evidence that it does not increase adverse events. There is a need for more trials including more evaluation of adverse events. None of the current trials evaluated effects in children.
Collapse
Affiliation(s)
- Bridget Candy
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry6th Floor, Maple House149 Tottenham Court RoadLondonUKW1T 7NF
| | - Louise Jones
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry6th Floor, Maple House149 Tottenham Court RoadLondonUKW1T 7NF
| | - Victoria Vickerstaff
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry6th Floor, Maple House149 Tottenham Court RoadLondonUKW1T 7NF
| | - Philip J Larkin
- UCD College of Health SciencesUCD School of Nursing, Midwifery and Health Systems and Our Lady's Hospice and Care ServicesDublinIreland
| | - Patrick Stone
- Marie Curie Palliative Care Research Department, UCL Division of PsychiatryDivision of PsychiatryRoom 119, First Floor, Charles Bell House67‐73 Riding House StreetLondonUKW1W 7EJ
| | | |
Collapse
|
14
|
Katakami N, Harada T, Murata T, Shinozaki K, Tsutsumi M, Yokota T, Arai M, Tada Y, Narabayashi M, Boku N. Randomized phase III and extension studies: efficacy and impacts on quality of life of naldemedine in subjects with opioid-induced constipation and cancer. Ann Oncol 2018; 29:1461-1467. [PMID: 32151367 PMCID: PMC6005145 DOI: 10.1093/annonc/mdy118] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The efficacy and safety of naldemedine (a peripherally acting µ-opioid receptor antagonist) for opioid-induced constipation (OIC) in subjects with cancer was demonstrated in the primary report of a phase III, double-blind study (COMPOSE-4) and its open-label extension (COMPOSE-5). The primary end point, the proportion of spontaneous bowel movement (SBM) responders, was met. Here, we report results from secondary end points, including quality of life (QOL) assessments from these studies. PATIENTS AND METHODS In COMPOSE-4, eligible adults with OIC and cancer were randomly assigned 1:1 to receive once-daily oral naldemedine 0.2 mg (n = 97) or placebo (n = 96) for 2 weeks, and those who continued on to COMPOSE-5 received naldemedine for 12 weeks (n = 131). Secondary assessments in COMPOSE-4 included the proportion of complete SBM (CSBM) responders, SBM or CSBM responders by week, and subjects with ≥1 SBM or CSBM within 24 h postinitial dose. Changes from baseline in the frequency of SBMs or CSBMs per week were assessed at weeks 1 and 2. Time to the first SBM or CSBM postinitial dose was also evaluated. In both studies, QOL impact was evaluated by Patient Assessment of Constipation-Symptoms (PAC-SYM) and PAC-QOL questionnaires. RESULTS Naldemedine improved bowel function for all secondary efficacy assessments versus placebo (all P ≤ 0.0002). The timely onset of naldemedine activity versus placebo was evidenced by median time to the first SBM (4.7 h versus 26.6 h) and CSBM (24.0 h versus 218.5 h) postinitial dose (all P < 0.0001). In COMPOSE-4, significant differences between groups were observed with the PAC-SYM stool domain (P = 0.045) and PAC-QOL dissatisfaction domain (P = 0.015). In COMPOSE-5, significant improvements from baseline were observed for overall and individual domain scores of PAC-SYM and PAC-QOL. CONCLUSIONS Naldemedine provided effective and timely symptomatic relief from OIC and improved the QOL of subjects with OIC and cancer. TRIAL REGISTRATION ID: www.ClinicalTrials.jp: JAPIC-CTI-132340 (COMPOSE-4) and JAPIC-CTI-132342 (COMPOSE-5).
Collapse
Affiliation(s)
- N Katakami
- Kobe City Medical Center General Hospital, Kobe, Japan.
| | - T Harada
- Center for Respiratory Diseases, JCHO Hokkaido Hospital, Sapporo, Japan
| | - T Murata
- Department of Breast Oncology, Aichi Cancer Center Aichi Hospital, Okazaki, Japan
| | - K Shinozaki
- Department of Clinical Oncology, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - M Tsutsumi
- Department of Urology, Hitachi General Hospital, Hitachi, Japan
| | - T Yokota
- Global Development, Shionogi & Co., Ltd, Osaka, Japan
| | - M Arai
- Global Development, Shionogi & Co., Ltd, Osaka, Japan
| | - Y Tada
- Global Development, Shionogi & Co., Ltd, Osaka, Japan
| | - M Narabayashi
- Department of Palliative Therapy, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - N Boku
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
15
|
O'Brien T, Christrup LL, Drewes AM, Fallon MT, Kress HG, McQuay HJ, Mikus G, Morlion BJ, Perez-Cajaraville J, Pogatzki-Zahn E, Varrassi G, Wells JCD. European Pain Federation position paper on appropriate opioid use in chronic pain management. Eur J Pain 2018; 21:3-19. [PMID: 27991730 PMCID: PMC6680203 DOI: 10.1002/ejp.970] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2016] [Indexed: 02/06/2023]
Abstract
Poorly controlled pain is a global public health issue. The personal, familial and societal costs are immeasurable. Only a minority of European patients have access to a comprehensive specialist pain clinic. More commonly the responsibility for chronic pain management and initiating opioid therapy rests with the primary care physician and other non‐specialist opioid prescribers. There is much confusing and conflicting information available to non‐specialist prescribers regarding opioid therapy and a great deal of unjustified fear is generated. Opioid therapy should only be initiated by competent clinicians as part of a multi‐faceted treatment programme in circumstances where more simple measures have failed. Throughout, all patients must be kept under close clinical surveillance. As with any other medical therapy, if the treatment fails to yield the desired results and/or the patient is additionally burdened by an unacceptable level of adverse effects, the overall management strategy must be reviewed and revised. No responsible clinician will wish to pursue a failed treatment strategy or persist with an ineffective and burdensome treatment. In a considered attempt to empower and inform non‐specialist opioid prescribers, EFIC convened a European group of experts, drawn from a diverse range of basic science and relevant clinical disciplines, to prepare a position paper on appropriate opioid use in chronic pain. The expert panel reviewed the available literature and harnessed the experience of many years of clinical practice to produce these series of recommendations. Its success will be judged on the extent to which it contributes to an improved pain management experience for chronic pain patients across Europe. Significance This position paper provides expert recommendations for primary care physicians and other non‐ specialist healthcare professionals in Europe, particularly those who do not have ready access to specialists in pain medicine, on the safe and appropriate use of opioid medications as part of a multi‐faceted approach to pain management, in properly selected and supervised patients.
Collapse
Affiliation(s)
- T O'Brien
- Marymount University Hospital & Hospice, Curraheen, Cork, Ireland.,Cork University Hospital, Wilton, Cork and College of Medicine and Health, University College, Cork, Ireland
| | - L L Christrup
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - A M Drewes
- Mech-Sense, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Denmark
| | - M T Fallon
- Edinburgh Cancer Research Centre, University of Edinburgh, UK
| | - H G Kress
- Department of Special Anaesthesia and Pain Therapy, Medical University of Vienna/AKH, Austria
| | | | - G Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital, Heidelberg, Germany
| | - B J Morlion
- Leuven Centre for Algology & Pain Management, University Hospital Leuven, Belgium
| | | | - E Pogatzki-Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Müenster, Germany
| | | | | |
Collapse
|
16
|
Koopmans-Klein G, Van Op den Bosch J, van Megen Y, Prenen H, Huygen F, Mancini I. Prolonged release oxycodone and naloxone treatment counteracts opioid-induced constipation in patients with severe pain compared to previous analgesic treatment. Curr Med Res Opin 2017; 33:2217-2227. [PMID: 28805471 DOI: 10.1080/03007995.2017.1367276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Treatment with prolonged-release oxycodone/naloxone (PR OXN) has been shown to improve opioid induced constipation (OIC) in constipated patients. This publication reports on a real-life observational study investigating the efficacy of PR OXN with regard to bowel function in patients switching to PR OXN from WHO step 1, step 2 and step 3 opioids. METHODS Patients with chronic pain experiencing insufficient pain relief and/or unacceptable side effects were switched to PR OXN and monitored in this observational study with respect to efficacy regarding bowel function and efficacy regarding pain relief in comparison with previous analgesic therapy. A patient was considered a responder with respect to efficacy if this assessment was "slightly better", "better" or "much better" compared with previous therapy. Bowel function index, pain intensity, quality of life, laxative medication use, and safety analgesic were also evaluated. RESULTS A total of 1338 patients (mean [SD] age 64.3 [14.9], 63% female) were observed for 43 [3-166] days (median [range]) during treatment with PR OXN. Overall response rate regarding bowel function efficacy was 82.5%. Patients with symptoms of constipation at study entry obtained a clinically relevant improvement of the bowel function index (BFI) within the first 2 weeks of PR OXN treatment. Non-constipated patients at study entry maintained normal bowel function despite switching to treatment with the opioid PR OXN. CONCLUSION In conclusion, treatment with PR OXN results in a significant and clinically relevant improvement of bowel function. During the observation of the treatment with PR OXN patients reported an improvement of quality of life (QoL). More interestingly, non-constipated patients maintained a normal bowel function, showing prevention of constipation despite the use of an opioid.
Collapse
Affiliation(s)
- Gineke Koopmans-Klein
- a Mundipharma Pharmaceuticals BV , Medical Department , Leusden , The Netherlands
- b Center for Pain Medicine, Erasmus MC , Rotterdam , The Netherlands
| | | | - Yvonne van Megen
- a Mundipharma Pharmaceuticals BV , Medical Department , Leusden , The Netherlands
| | - Hans Prenen
- d Digestive Oncology Unit , University Hospital Leuven , Leuven , Belgium
| | - Frank Huygen
- b Center for Pain Medicine, Erasmus MC , Rotterdam , The Netherlands
| | | |
Collapse
|
17
|
Webster L, Diva U, Tummala R, Sostek M. Treatment with Naloxegol Versus Placebo: Pain Assessment in Patients with Noncancer Pain and Opioid-Induced Constipation. Pain Pract 2017; 18:505-514. [PMID: 28898536 DOI: 10.1111/papr.12640] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/18/2017] [Accepted: 09/06/2017] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To summarize results from pain and opioid use assessments with naloxegol in adults with opioid-induced constipation (OIC) and chronic noncancer pain. METHODS Two phase 3 randomized, double-blind, 12-week studies evaluated the efficacy and safety of oral naloxegol (12.5 or 25 mg daily) in adults (18 to < 85 years) with confirmed OIC and chronic noncancer pain: KODIAC-04 (NCT01309841) and KODIAC-05 (NCT01323790). Pain level was assessed daily (11-point numeric rating scale [NRS]; 0 = no pain, 10 = worst imaginable pain). Changes from baseline in mean weekly pain scores and opioid dose (weeks 1 through 12) were analyzed using mixed-model repeated measures. RESULTS At baseline, mean daily NRS average pain scores ranged from 4.5 to 4.8 for all groups in KODIAC-04 (N = 652) and were 4.6 for each group in KODIAC-05 (N = 700). Respective mean ± SD changes from baseline average pain for placebo, naloxegol 12.5 mg, and naloxegol 25 mg were -0.2 ± 1.07, -0.3 ± 1.05 (P = 0.773 vs. placebo), and 0.2 ± 0.95 (P = 0.837 vs. placebo; KODIAC-04) and -0.1 ± 0.94, -0.1 ± 0.87 (P = 0.744), and 0.0 ± 1.18 (P = 0.572; KODIAC-05). At baseline, mean daily opioid doses ranged from 135.6 to 143.2 morphine equivalent units (MEUs)/day in KODIAC-04, and from 119.9 to 151.7 MEUs/day in KODIAC-05. Respective mean ± SD changes from baseline dose were -1.8 ± 30.19, -2.3 ± 20.52 (P = 0.724 vs. placebo), and 0.4 ± 13.01 (P = 0.188 vs. placebo; KODIAC-04) and -0.3 ± 17.14, -1.3 ± 17.11 (P = 0.669 vs. placebo), and 0.1 ± 8.54 (P = 0.863 vs. placebo; KODIAC-05). Changes in maintenance opioid dose were few; reasons for such changes were similar across treatment groups. CONCLUSION Centrally mediated opioid analgesia was maintained during treatment with naloxegol in patients with noncancer pain and OIC.
Collapse
Affiliation(s)
- Lynn Webster
- PRA Health Sciences, Salt Lake City, Utah, U.S.A
| | - Ulysses Diva
- Bioinformatics and Information Sciences, AstraZeneca Pharmaceuticals LP, Gaithersburg, Maryland, U.S.A
| | - Raj Tummala
- Global Medicines Development, AstraZeneca Pharmaceuticals LP, Gaithersburg, Maryland, U.S.A
| | - Mark Sostek
- Global Medicines Development, AstraZeneca Pharmaceuticals LP, Gaithersburg, Maryland, U.S.A
| |
Collapse
|
18
|
Katakami N, Harada T, Murata T, Shinozaki K, Tsutsumi M, Yokota T, Arai M, Tada Y, Narabayashi M, Boku N. Randomized Phase III and Extension Studies of Naldemedine in Patients With Opioid-Induced Constipation and Cancer. J Clin Oncol 2017; 35:3859-3866. [PMID: 28968171 DOI: 10.1200/jco.2017.73.0853] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose Opioid-induced constipation (OIC) is a frequent and debilitating adverse effect (AE) of opioids-common analgesics for cancer pain. We investigated the efficacy and safety of a peripherally acting μ-opioid receptor antagonist, naldemedine (S-297995), for OIC, specifically in patients with cancer. Patients and Methods This phase III trial consisted of a 2-week, randomized, double-blind, placebo-controlled study (COMPOSE-4) and an open-label, 12-week extension study (COMPOSE-5). In COMPOSE-4, eligible adults with OIC and cancer were randomly assigned on a 1:1 basis to receive once-daily oral naldemedine 0.2 mg or placebo. The primary end point was the proportion of spontaneous bowel movement (SBM) responders (≥ 3 SBMs/week and an increase of ≥ 1 SBM/week from baseline). The primary end point of COMPOSE-5 was safety. Results In COMPOSE-4, 193 eligible patients were randomly assigned to naldemedine (n = 97) or placebo (n = 96). The proportion of SBM responders in COMPOSE-4 was significantly greater with naldemedine than with placebo (71.1% [69 of 97 patients] v 34.4% [33 of 96 patients]; P < .0001). A greater change from baseline was observed with naldemedine than with placebo in the frequency of SBMs/week (5.16 v 1.54; P < .0001), SBMs with complete bowel evacuation/week (2.76 v 0.71; P < .0001), and SBMs without straining/week (3.85 v 1.17; P = .0005). In COMPOSE-4, more patients treated with naldemedine than with placebo reported treatment-emergent AEs (TEAEs) (44.3% [43 of 97 patients] v 26.0% [25 of 96 patients]; P = .01); in COMPOSE-5, 105 (80.2%) of 131 of patients reported TEAEs. Diarrhea was the most frequently reported TEAE in COMPOSE-4 (19.6% [19 of 97 patients] v 7.3% [seven of 96 patients] with naldemedine v placebo) and COMPOSE-5 (18.3% [24 of 131 patients] with naldemedine). Naldemedine was not associated with signs or symptoms of opioid withdrawal and had no notable impact on opioid-mediated analgesia. Conclusion Once-daily oral naldemedine 0.2 mg effectively treated OIC and was generally well tolerated in patients with OIC and cancer.
Collapse
Affiliation(s)
- Nobuyuki Katakami
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Toshiyuki Harada
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Toru Murata
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Katsunori Shinozaki
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Masakazu Tsutsumi
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Takaaki Yokota
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Masatsugu Arai
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Yukio Tada
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Masaru Narabayashi
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| | - Narikazu Boku
- Nobuyuki Katakami, Institute of Biomedical Research and Innovation, Kobe; Toshiyuki Harada, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo; Toru Murata, Aichi Hospital, Okazaki; Katsunori Shinozaki, Hiroshima Prefectural Hospital, Hiroshima; Masakazu Tsutsumi, Hitachi General Hospital, Hitachi; Takaaki Yokota, Masatsugu Arai, and Yukio Tada, Shionogi & Co Ltd, Osaka; Masaru Narabayashi, Cancer Institute Hospital of Japanese Foundation for Cancer Research; and Narikazu Boku, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Jiang L, Beattie DT, Jacobsen JR, Kintz S, Obedencio GP, Saito D, Stergiades I, Vickery RG, Long DD. Discovery of N -substituted- endo -3-(8-aza-bicyclo[3.2.1]oct-3-yl)-phenol and -phenyl carboxamide series of μ-opioid receptor antagonists. Bioorg Med Chem Lett 2017; 27:2926-2930. [DOI: 10.1016/j.bmcl.2017.04.092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/29/2017] [Accepted: 04/30/2017] [Indexed: 01/23/2023]
|
20
|
Pergolizzi JV, LeQuang JA, Berger GK, Raffa RB. The Basic Pharmacology of Opioids Informs the Opioid Discourse about Misuse and Abuse: A Review. Pain Ther 2017; 6:1-16. [PMID: 28341939 PMCID: PMC5447545 DOI: 10.1007/s40122-017-0068-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Indexed: 02/01/2023] Open
Abstract
Morphine and other opioids are widely used to manage moderate to severe acute pain syndromes, such as pain associated with trauma or postoperative pain, and they have been used to manage chronic pain, even chronic nonmalignant pain. However, recent years have seen a renewed recognition of the potential for overuse, misuse, and abuse of opioids. Therefore, prescribing opioids is challenging for healthcare providers in that clinical effectiveness must be balanced against negative outcomes-with the possibility that neither are achieved perfectly. The current discourse about the dual 'epidemics' of under-treatment of legitimate pain and the over-prescription of opioids is clouded by inadequate or inaccurate understanding of opioid drugs and the endogenous pain pathways with which they interact. An understanding of the basic pharmacology of opioids helps inform the clinician and other stakeholders about these simultaneously under- and over-used agents.
Collapse
Affiliation(s)
| | | | | | - Robert B Raffa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
21
|
Al-Huniti N, Zhou D, Xu H, Aksenov S, Bui KH, Fox R, Helmlinger G, Stanski D. Pharmacometric Modeling of Naloxegol Efficacy and Safety: Impact on Dose and Label. Clin Pharmacol Ther 2017; 102:741-744. [PMID: 28548207 DOI: 10.1002/cpt.719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/27/2017] [Accepted: 04/17/2017] [Indexed: 11/06/2022]
Abstract
Naloxegol is a peripherally acting μ-opioid receptor antagonist that was developed for the treatment of opioid-induced constipation. Modeling and simulation of naloxegol efficacy and tolerability informed selection of doses for phase III studies and provided comprehensive dosage recommendations for the naloxegol US package insert.
Collapse
Affiliation(s)
- N Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - D Zhou
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - H Xu
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - S Aksenov
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - K H Bui
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - R Fox
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - G Helmlinger
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| | - D Stanski
- Quantitative Clinical Pharmacology, AstraZeneca LP, Waltham, Massachusetts, USA
| |
Collapse
|
22
|
Abstract
Aim: Naloxegol is an oral peripherally acting μ-opioid receptor antagonist approved for the treatment of opioid-induced constipation. Sensitive, robust, bioanalytical methods were required to quantitate naloxegol in human biological matrices as part of the clinical development program. Methodology/results: Analytical plasma samples were prepared using Solid Phase Extraction (SPE) coupled with concentration. The method’s linearity was established at 0.1–50 ng/ml with up to 100-fold dilution. Urine samples were analyzed directly postdilution; dialysate samples were extracted by supported liquid extraction. Sensitive liquid chromatography/mass spectrometry (LC–MS/MS) assays were developed and validated, and demonstrated acceptable precision, accuracy and selectivity for naloxegol in the appropriate matrices. Conclusion: Methods for quantifying naloxegol in human biological matrices have been successfully validated.
Collapse
|
23
|
Koopmans-Klein G, Wagemans MFM, Wartenberg HCH, Van Megen YJB, Huygen FJPM. The efficacy of standard laxative use for the prevention and treatment of opioid induced constipation during oxycodone use: a small Dutch observational pilot study. Expert Rev Gastroenterol Hepatol 2016; 10:547-53. [PMID: 26641540 DOI: 10.1586/17474124.2016.1129275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Dutch clinical guidelines recommend that a standard laxative treatment (SLT) should be prescribed concomitantly when starting opioid treatment to prevent opioid-induced constipation (OIC). METHODS Clinical evidence for SLT in the treatment of OIC is lacking, therefore an observational pilot study was performed to explore the efficacy and tolerability of SLT on OIC in patients treated with the opioid oxycodone. RESULTS Twenty-four patients (58% female, median (range) age 65 (39-92)) were included in this pilot study. The analysis showed that 9 out of 21 patients (43%) were non-responders to SLT. When also taking into consideration patients tending to develop diarrhea 75% of patients are non-responsive to SLT. CONCLUSION This pilot study indicates that optimal laxative therapy (SLT) might not be effective and feasible for the prevention and treatment of OIC.
Collapse
Affiliation(s)
- Gineke Koopmans-Klein
- a Medical Department , Mundipharma Pharmaceuticals BV , Hoevelaken , The Netherlands.,b Center for Pain Medicine , Erasmus MC , Rotterdam , The Netherlands
| | - Michel F M Wagemans
- c Department of Anesthesiology and Pain Medicine , Reinier de Graaf , Delft , The Netherlands
| | - Hans C H Wartenberg
- d Department of Anesthesiology, Academisch Medisch Centrum , University of Amsterdam , Amsterdam , the Netherlands
| | - Yvonne J B Van Megen
- a Medical Department , Mundipharma Pharmaceuticals BV , Hoevelaken , The Netherlands
| | | |
Collapse
|
24
|
Al-Huniti N, Nielsen JC, Hutmacher MM, Lappalainen J, Cantagallo K, Sostek M. Population Exposure-Response Modeling of Naloxegol in Patients With Noncancer-Related Pain and Opioid-Induced Constipation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:359-66. [PMID: 27435972 PMCID: PMC4961079 DOI: 10.1002/psp4.12099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/18/2016] [Accepted: 06/25/2016] [Indexed: 01/18/2023]
Abstract
Naloxegol is a polyethylene glycol derivative of naloxone approved in the US as a once-daily oral treatment for opioid-induced constipation (OIC) in adults with chronic noncancer pain. Population exposure-response models were constructed based on data from two phase III studies comprising 1,331 adults with noncancer pain and OIC. In order to characterize the protocol-defined naloxegol responder rate, the number of daily spontaneous bowel movements (SBMs) was characterized by a longitudinal ordinal nonlinear mixed-effects logistic regression dose-response model, and the incidence of diary entry discontinuation was described by a time-to-event model. The mean number of SBMs per week increased with increasing naloxegol dose. The predicted placebo-adjusted responder rates (90% confidence interval) were 10.4% (4.6-13.4%) and 11.1% (4.8-14.4%) for naloxegol 12.5 and 25 mg/day, respectively. Model-predicted response to naloxegol was influenced by the baseline SBM frequency and characteristics of the opioid treatment.
Collapse
Affiliation(s)
- N Al-Huniti
- AstraZeneca Pharmaceuticals, Waltham, Massachusetts, USA
| | - J C Nielsen
- Ann Arbor Pharmacometrics Group, Ann Arbor, Michigan, USA
| | - M M Hutmacher
- Ann Arbor Pharmacometrics Group, Ann Arbor, Michigan, USA
| | - J Lappalainen
- AstraZeneca Pharmaceuticals, Waltham, Massachusetts, USA
| | - K Cantagallo
- AstraZeneca Pharmaceuticals, Waltham, Massachusetts, USA
| | - M Sostek
- AstraZeneca Pharmaceuticals, Waltham, Massachusetts, USA
| |
Collapse
|
25
|
Holder RM, Rhee D. Novel Oral Therapies for Opioid-induced Bowel Dysfunction in Patients with Chronic Noncancer Pain. Pharmacotherapy 2016; 36:287-99. [PMID: 26945548 DOI: 10.1002/phar.1711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Opioid analgesics are frequently prescribed and play an important role in chronic pain management. Opioid-induced bowel dysfunction, which includes constipation, hardened stool, incomplete evacuation, gas, and nausea and vomiting, is the most common adverse event associated with opioid use. Mu-opioid receptors are specifically responsible for opioid-induced bowel dysfunction, resulting in reduced peristaltic and secretory actions. Agents that reverse these actions in the bowel without reversing pain control in the central nervous system may be preferred over traditional laxatives. The efficacy and safety of these agents in chronic noncancer pain were assessed from publications identified through Ovid and PubMed database searches. Trials that evaluated the safety and efficacy of oral agents for opioid-induced constipation or opioid-induced bowel dysfunction, excluding laxatives, were reviewed. Lubiprostone and naloxegol are approved in the United States by the Food and Drug Administration for use in opioid-induced constipation. Axelopran (TD-1211) and sustained-release naloxone have undergone phase 2 and phase 1 studies, respectively, for the same indication. Naloxegol and axelopran are peripherally acting μ-opioid receptor antagonists. Naloxone essentially functions as a peripherally acting μ-opioid receptor antagonist when administered orally in a sustained-release formulation. Lubiprostone is a locally acting chloride channel (CIC-2) activator that increases secretions and peristalsis. All agents increase spontaneous bowel movements and reduce other bowel symptoms compared with placebo in patients with noncancer pain who are chronic opioid users. The most common adverse events were gastrointestinal in nature, and none of the drugs were associated with severe adverse or cardiovascular events. Investigations comparing these agents to regimens using standard laxative and combination therapy and trials in special populations and patients with active cancer are needed to further define their role in therapy.
Collapse
Affiliation(s)
- Renee M Holder
- Department of Medicine Section of Palliative Care, MedStar Washington Hospital Center, Washington, District of Columbia
| | - Diane Rhee
- College of Pharmacy, Roseman University of Health Sciences, Henderson, Nevada
| |
Collapse
|
26
|
Bui K, Zhou D, Sostek M, She F, Al-Huniti N. Effects of CYP3A Modulators on the Pharmacokinetics of Naloxegol. J Clin Pharmacol 2016; 56:1019-27. [DOI: 10.1002/jcph.693] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/11/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Khanh Bui
- AstraZeneca Pharmaceuticals; Waltham MA USA
| | | | - Mark Sostek
- AstraZeneca Pharmaceuticals; Gaithersburg MD USA
| | - Fahua She
- AstraZeneca Pharmaceuticals; Gaithersburg MD USA
| | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Opioid-based management of noncancer pain has become much more prevalent over the last 2 decades and is responsible for a wide range of side-effects, particularly affecting the intestinal tract causing opioid-induced constipation (OIC). This review will consider results of recent clinical trials that have provided evidence of new pharmacological management options for the treatment of OIC. RECENT FINDINGS Supportive use of conventional agents, such as stool softeners, osmotic laxatives, and stimulating laxatives in OIC has limited efficacy. The peripheral μ-opioid receptor antagonist (PAMORA) methylnaltrexone (MNTX) was first FDA approved for OIC in patients with advanced illness and later also for OIC in noncancer pain patients; clinical trial results indicated MNTX did not reverse opioid analgesia and did not trigger central opioid withdrawal. Another PAMORA, the orally available naloxegol, has also gained recent FDA approval for the treatment of OIC in adults with chronic, noncancer pain. Lubiprostone, a bicyclical fatty acid acting via activation of intestinal chloride channel-2 (ClC-2), was also approved for OIC treatment in patients with noncancer pain. SUMMARY PAMORA MNTX and naloxegol and the intestinal chloride channel-2 (ClC-2) activator lubiprostone represent additional possible therapeutic options for the management of OIC in patients with chronic noncancer pain.
Collapse
Affiliation(s)
- H Christian Weber
- Section of Gastroenterology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Goeree R, Goeree J. Cost-effectiveness analysis of oxycodone with naloxone versus oxycodone alone for the management of moderate-to-severe pain in patients with opioid-induced constipation in Canada. J Med Econ 2016; 19:277-91. [PMID: 26535790 DOI: 10.3111/13696998.2015.1116992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Approximately 20-30% of Canadians suffer from chronic pain. Guidelines for the management of chronic pain support the use of controlled-release (CR) opioids to treat chronic pain. Although effective in managing chronic pain, oxycodone is associated with high rates of opioid-induced constipation (OIC). The cost-effectiveness of a combination of oxycodone for the management of pain and naloxone for the relief of OIC has not previously been evaluated for Canada. METHODS A decision analytic model was developed to estimate the cost-utility of combination oxycodone/naloxone compared to oxycodone alone in four populations. Drug costs for managing pain and healthcare costs related to managing OIC were included in the analysis and the primary measure of effectiveness was quality adjusted life years (QALYs) derived from OIC rates observed in clinical trials. The analysis was conducted from a healthcare system perspective, used a 1-year time horizon, and results were expressed in 2015 Canadian dollars. RESULTS In all four patient populations, there was a trade-off between slightly higher total expected costs for Targin treated patients compared to oxycodone treated patients, but also improved clinical benefits in terms of reduced OIC, which resulted in higher QALYs for patients. Although analgesic costs were found to be slightly higher for Targin treated patients, Targin also resulted in cost offsets to the healthcare system in terms of less rescue laxative drug use and other resources required for the management of OIC. The resulting 1-year cost-utility of Targin compared to oxycodone ranged from $2178-$7732 per QALY gained in the base case analysis, and it was found that these cost-utility results remained robust and at low values throughout a series of one-way deterministic analyses of uncertainty. CONCLUSION The clinical effectiveness of oxycodone/naloxone in managing pain and OIC compared to CR oxycodone alone resulted in low cost-utility estimates.
Collapse
Affiliation(s)
- Ron Goeree
- a a Goeree Consulting Limited , Hamilton, Ontario , Canada
- b bProfessor Emeritus , McMaster University , Hamilton, Ontario , Canada
| | - Jeff Goeree
- a a Goeree Consulting Limited , Hamilton, Ontario , Canada
| |
Collapse
|
29
|
Bui K, She F, Zhou D, Butler K, Al-Huniti N, Sostek M. The effect of quinidine, a strong P-glycoprotein inhibitor, on the pharmacokinetics and central nervous system distribution of naloxegol. J Clin Pharmacol 2015; 56:497-505. [PMID: 26248047 DOI: 10.1002/jcph.613] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/31/2015] [Indexed: 12/31/2022]
Abstract
Naloxegol is a PEGylated, oral, peripherally acting μ-opioid receptor antagonist approved in the United States for treatment of opioid-induced constipation in patients with noncancer pain. Naloxegol is metabolized by CYP3A, and its properties as a substrate for the P-glycoprotein (PGP) transporter limit its central nervous system (CNS) permeability. This double-blind, randomized, 2-part, crossover study in healthy volunteers evaluated the effect of quinidine (600 mg PO), a CYP3A/PGP transporter inhibitor, on the pharmacokinetics and CNS distribution of naloxegol (25 mg PO). In addition, the effects of quinidine on morphine (5 mg/70 kg IV)-induced miosis and exposure to naloxegol were assessed. Coadministration of quinidine and naloxegol increased naloxegol's AUC 1.4-fold and Cmax 2.5-fold but did not antagonize morphine-induced miosis, suggesting that PGP inhibition does not increase the CNS penetration of naloxegol. Naloxegol pharmacokinetics was unaltered by coadministration of morphine and either quinidine or placebo; conversely, pharmacokinetics of morphine and its metabolites (in the presence of quinidine) were unaltered by coadministration of naloxegol. Naloxegol was safe and well tolerated, alone or in combination with quinidine, morphine, or both. The observed increase in exposure to naloxegol in the presence of quinidine is primarily attributed to quinidine's properties as a weak CYP3A inhibitor.
Collapse
Affiliation(s)
- Khanh Bui
- AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | - Fahua She
- AstraZeneca Pharmaceuticals, Gaithersburg, MD, USA
| | | | | | | | - Mark Sostek
- AstraZeneca Pharmaceuticals, Gaithersburg, MD, USA
| |
Collapse
|
30
|
Poulsen JL, Brock C, Olesen AE, Nilsson M, Drewes AM. Evolving paradigms in the treatment of opioid-induced bowel dysfunction. Therap Adv Gastroenterol 2015; 8:360-72. [PMID: 26557892 PMCID: PMC4622283 DOI: 10.1177/1756283x15589526] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years prescription of opioids has increased significantly. Although effective in pain management, bothersome gastrointestinal adverse effects are experienced by a substantial proportion of opioid-treated patients. This can lead to difficulties with therapy and subsequently inadequate pain relief. Collectively referred to as opioid-induced bowel dysfunction, these adverse effects are the result of binding of exogenous opioids to opioid receptors in the gastrointestinal tract. This leads to disturbance of three important gastrointestinal functions: motility, coordination of sphincter function and secretion. In the clinic this manifests in a wide range of symptoms such as reflux, bloating, abdominal cramping, hard, dry stools, and incomplete evacuation, although the most known adverse effect is opioid-induced constipation. Traditional treatment with laxatives is often insufficient, but in recent years a number of novel pharmacological approaches have been introduced. In this review the pathophysiology, symptomatology and prevalence of opioid-induced bowel dysfunction is presented along with the benefits and caveats of a suggested consensus definition for opioid-induced constipation. Finally, traditional treatment is appraised and compared with the latest pharmacological developments. In conclusion, opioid antagonists restricted to the periphery show promising results, but use of different definitions and outcome measures complicate comparison. However, an international working group has recently suggested a consensus definition for opioid-induced constipation and relevant outcome measures have also been proposed. If investigators within this field adapt the suggested consensus and include symptoms related to dysfunction of the upper gut, it will ease comparison and be a step forward in future research.
Collapse
Affiliation(s)
- Jakob Lykke Poulsen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Christina Brock
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Anne Estrup Olesen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matias Nilsson
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Mølleparkvej 4, DK-9000 Aalborg, Denmark
| |
Collapse
|
31
|
Al-Huniti N, Chapel S, Xu H, Bui KH, Sostek M. Population pharmacokinetics of naloxegol in a population of 1247 healthy subjects and patients. Br J Clin Pharmacol 2015; 81:89-100. [PMID: 26317320 DOI: 10.1111/bcp.12756] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 07/28/2015] [Accepted: 08/25/2015] [Indexed: 12/17/2022] Open
Abstract
AIMS Naloxegol, a polyethylene glycol conjugated derivative of the opioid antagonist naloxone, is in clinical development for treatment of opioid-induced constipation (OIC). The aim of the study was to develop a population pharmacokinetic model describing the concentration vs. time profile of orally administered naloxegol, and determine the impact of pre-specified demographic and clinical factors and concomitant medication on population estimates of apparent clearance (CL/F) and apparent central compartment volume of distribution (Vc /F). METHODS Analysis included 12,844 naloxegol plasma concentrations obtained from 1247 healthy subjects, patients with non-OIC and patients with OIC in 14 phase 1, 2b and 3 clinical studies. Pharmacokinetic analysis used the non-linear mixed effects modelling program. Goodness of fit plots and posterior predictive checks were conducted to confirm concordance with observed data. RESULTS The final model was a two compartment disposition model with dual absorptions, comprising one first order absorption (ka1 4.56 h(-1) ) and one more complex absorption with a transit compartment (ktr 2.78 h(-1) ). Mean (SE) parameter estimates for CL/F and Vc /F, the parameters assessed for covariate effects, were 115 (3.41) l h(-1) and 160 (27.4) l, respectively. Inter-individual variability was 48% and 51%, respectively. Phase of study, gender, race, concomitant strong or moderate CYP3A4 inhibitors, strong CYP3A4 inducers, P-glycoprotein inhibitors or inducers, naloxegol formulation, baseline creatinine clearance and baseline opioid dose had a significant effect on at least one pharmacokinetic parameter. Simulations indicated concomitant strong CYP3A4 inhibitors or inducers had relevant effects on naloxegol exposure. CONCLUSIONS Administration of strong CYP3A4 inhibitors or inducers had a clinically relevant influence on naloxegol pharmacokinetics.
Collapse
Affiliation(s)
- Nidal Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts
| | - Sunny Chapel
- Ann Arbor Pharmacometrics Group (A2PG), Ann Arbor, Michigan, USA
| | - Hongmei Xu
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts
| | - Khanh H Bui
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts
| | - Mark Sostek
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts
| |
Collapse
|
32
|
Ryu HS, Choi SC. Recent Updates on the Treatment of Constipation. Intest Res 2015; 13:297-305. [PMID: 26576134 PMCID: PMC4641855 DOI: 10.5217/ir.2015.13.4.297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022] Open
Abstract
The treatment of constipation aims to regulate the frequency and quantity of stool in order to promote successful defecation. Numerous studies on pharmacologic treatments and non-pharmacologic therapies for constipation have attempted to overcome limitations such as temporary and insufficient efficacy. Conventional laxatives have less adverse effects and are inexpensive, but often have limited efficacy. Recently developed enterokinetic agents and intestinal secretagogues have received attention owing to their high efficacies and low incidences of adverse events. Studies on biofeedback and surgical treatment have focused on improving symptoms as well as quality of life for patients with refractory constipation.
Collapse
Affiliation(s)
- Han Seung Ryu
- Division of Gastroenterology, Department of Internal Medicine, Digestive Disease Research Institute, Wonkwang University Hospital, Iksan, Korea
| | - Suck Chei Choi
- Division of Gastroenterology, Department of Internal Medicine, Digestive Disease Research Institute, Wonkwang University Hospital, Iksan, Korea
| |
Collapse
|
33
|
Quality of life and healthcare resource in patients receiving opioids for chronic pain: a review of the place of oxycodone/naloxone. Clin Drug Investig 2015; 35:1-11. [PMID: 25479959 PMCID: PMC4281369 DOI: 10.1007/s40261-014-0254-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In patients managed with opioids for chronic pain, opioid-induced bowel dysfunction—specifically, opioid-induced constipation (OIC)—is a common side effect, which has a significant impact on quality of life (QoL). The most recent developments for management of OIC are opioid antagonists, including naloxone, a competitive antagonist of peripheral opioid receptors that reverses opioid-induced peripheral gastrointestinal (GI) effects. A prolonged-release formulation of naloxone is available in combination with oxycodone (OXN PR). To review the specific role of OXN PR in the management of chronic pain and OIC and its impact on QoL and healthcare costs, a review of available relevant literature was conducted. Healthcare costs can be up to ten times higher for patients with GI events than for those without. Assessment of QoL in patients with OIC is essential, and multiple tools for its evaluation are available. The Bowel Function Index (BFI), a tool that was specifically developed and validated to measure bowel function in patients with OIC, can be an indication of QoL. In patients with moderate-to-severe chronic pain, randomized trials have demonstrated that OXN PR has equal analgesic efficacy and safety, but results in improved bowel function, compared with prolonged-release oxycodone (Oxy PR) alone. In conclusion, randomized studies using the BFI, as well as real-world clinical practice observations, have demonstrated improved QoL for patients taking OXN PR. This combination should allow more patients to benefit from the analgesic efficacy of opioid therapy and should minimize the side effects of constipation that correspond to improvements in QoL and healthcare offsets.
Collapse
|
34
|
Abstract
Opioids are the oldest and most potent drugs for the treatment of severe pain. Their clinical application is undisputed in acute (e.g., postoperative) and cancer pain, but their long-term use in chronic pain has met increasing scrutiny. This article reviews mechanisms underlying opioid analgesia and other opioid actions. It discusses the structure, function, and plasticity of opioid receptors; the central and peripheral sites of analgesic actions and side effects; endogenous and exogenous opioid receptor ligands; and conventional and novel opioid compounds. Challenging clinical situations, such as the tension between chronic pain and addiction, are also illustrated.
Collapse
Affiliation(s)
- Christoph Stein
- Department of Anesthesiology and Critical Care Medicine, Freie Universität Berlin, Charité Campus Benjamin Franklin, 12200 Berlin, Germany; .,Helmholtz Virtual Institute, Multifunctional Biomaterials for Medicine, 14513 Teltow, Germany
| |
Collapse
|
35
|
Leppert W. Emerging therapies for patients with symptoms of opioid-induced bowel dysfunction. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2215-31. [PMID: 25931815 PMCID: PMC4404965 DOI: 10.2147/dddt.s32684] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Opioid-induced bowel dysfunction (OIBD) comprises gastrointestinal (GI) symptoms, including dry mouth, nausea, vomiting, gastric stasis, bloating, abdominal pain, and opioid-induced constipation, which significantly impair patients’ quality of life and may lead to undertreatment of pain. Traditional laxatives are often prescribed for OIBD symptoms, although they display limited efficacy and exert adverse effects. Other strategies include prokinetics and change of opioids or their administration route. However, these approaches do not address underlying causes of OIBD associated with opioid effects on mostly peripheral opioid receptors located in the GI tract. Targeted management of OIBD comprises purely peripherally acting opioid receptor antagonists and a combination of opioid receptor agonist and antagonist. Methylnaltrexone induces laxation in 50%–60% of patients with advanced diseases and OIBD who do not respond to traditional oral laxatives without inducing opioid withdrawal symptoms with similar response (45%–50%) after an oral administration of naloxegol. A combination of prolonged-release oxycodone with prolonged-release naloxone (OXN) in one tablet (a ratio of 2:1) provides analgesia with limited negative effect on the bowel function, as oxycodone displays high oral bioavailability and naloxone demonstrates local antagonist effect on opioid receptors in the GI tract and is totally inactivated in the liver. OXN in daily doses of up to 80 mg/40 mg provides equally effective analgesia with improved bowel function compared to oxycodone administered alone in patients with chronic non-malignant and cancer-related pain. OIBD is a common complication of long-term opioid therapy and may lead to quality of life deterioration and undertreatment of pain. Thus, a complex assessment and management that addresses underlying causes and patomechanisms of OIBD is recommended. Newer strategies comprise methylnaltrexone or OXN administration in the management of OIBD, and OXN may be also considered as a preventive measure of OIBD development in patients who require opioid administration.
Collapse
Affiliation(s)
- Wojciech Leppert
- Chair and Department of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
36
|
Li JP, Wang XY, Gao CJ, Liao YH, Qu J, He ZY, Zhang T, Wang GD, Li YQ. Neurochemical phenotype and function of endomorphin 2-immunopositive neurons in the myenteric plexus of the rat colon. Front Neuroanat 2014; 8:149. [PMID: 25565974 PMCID: PMC4267282 DOI: 10.3389/fnana.2014.00149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/22/2014] [Indexed: 12/20/2022] Open
Abstract
The distribution and activity of endomorphins (EMs), which are endogenous μ-opioid receptor (MOR) ligands in the gastrointestinal tract (GI), are yet to be elucidated. The current study aimed to shed light on this topic. EM2 was expressed in the enteric neurons in the myenteric plexus of the mid-colon. Of the EM2-immunoreactive (EM2-IR) neurons, 53 ± 4.6%, 26 ± 4.5%, 26 ± 2.8% and 49 ± 4.2% displayed immunopositive staining for choline acetyl transferase (ChAT), substance P (SP), vasoactive intestinal peptide (VIP) and nitric oxide synthetase (NOS), respectively. A bath application of EM2 (2 μM) enhanced spontaneous contractile amplitude and tension, which were reversed by β-FNA (an antagonist of MOR) but not NG-nitro-L-arginine methyl ether (L-NAME, a non-selective inhibitor of NOS) or VIP6-28 (an antagonist of the VIP receptor) in the colonic strips. EM2 significantly suppressed inhibitory junction potentials (IJPs) in 14 of the 17 examined circular muscle cells, and this effect was not antagonized by preincubation in L-NAME. EM2 was widely expressed in interneurons and motor neurons in the myenteric plexus and presynaptically inhibited fast IJPs, thereby enhancing spontaneous contraction and tension in the colonic smooth muscle.
Collapse
Affiliation(s)
- Jun-Ping Li
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
- Department of Anatomy, Histology and Embryology, Ningxia Medical UniversityYinchuan, China
| | - Xi-Yu Wang
- Department of Physiology and Cell Biology, Medical Center, Ohio State UniversityColumbus, OH, USA
| | - Chang-Jun Gao
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
| | - Yong-Hui Liao
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
| | - Juan Qu
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
| | - Zhong-Yi He
- Department of Anatomy, Histology and Embryology, Ningxia Medical UniversityYinchuan, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
| | - Guo-Du Wang
- Department of Physiology and Cell Biology, Medical Center, Ohio State UniversityColumbus, OH, USA
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi’an, China
| |
Collapse
|
37
|
Use of methylnaltrexone to induce laxation in acutely injured patients with burns and necrotizing soft-tissue infections. J Burn Care Res 2014; 35:e106-11. [PMID: 23877147 DOI: 10.1097/bcr.0b013e31829b399d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The routine use of high-dose opioids for analgesia in patients with acute burns and soft-tissue injuries often leads to the development of opioid-induced constipation. The opioid antagonist methylnaltrexone (MLTX) reverses narcotic-related ileus without affecting systemic pain treatment. The authors' burn center developed a bowel protocol that included administration of MLTX for relief of opioid-induced constipation after other methods failed. The authors performed a retrospective review of patients with acute burns or necrotizing soft-tissue infections, who had been given subcutaneous MLTX to induce laxation. All patients who received MLTX were included and all administrations of the drug were included in the analysis. The primary outcome examined was time to laxation from drug administration. Forty-eight patients received MLTX a total of 112 times. Six patients were admitted with soft-tissue injuries and the rest suffered burns with an average TBSA of 17%. The median patient age was 41 years and the majority (75%) were men. Administration of a single dose of MLTX resulted in laxation within 4 hours in 38% of cases, and within 24 hours in 68%. Patients given MLTX received an average of 174 mg morphine equivalents daily for pain control. MLTX was given after an average of 52 hours since the last bowel movement. As this experience has evolved, it has been incorporated into an organized bowel protocol, which includes MLTX administration after other laxatives have failed. MLTX is an effective laxation agent in patients with burn and soft-tissue injuries, who have failed conventional agents.
Collapse
|
38
|
Laliberté F, Moore Y, Dea K, LaMori JC, Mody SH, Jones JL, Arledge MD, Damaraju CV, Schein JR, Lefebvre P. Gastrointestinal comorbidities associated with atrial fibrillation. SPRINGERPLUS 2014; 3:603. [PMID: 25392776 PMCID: PMC4210454 DOI: 10.1186/2193-1801-3-603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/03/2014] [Indexed: 11/29/2022]
Abstract
This observational study was conducted to describe the risk of gastrointestinal (GI) events among patients with atrial fibrillation (AF). We analyzed Thomson Reuters MarketScan® data from 2005 to 2009. Subjects aged ≥18 years with ≥ 1 AF diagnosis were selected. GI events were identified from claims with a primary or secondary diagnosis code for any GI condition. The risk of GI events was assessed using cumulative incidence (new GI events/patients with AF without GI condition at baseline) and incidence rates (IRs), calculated as the number of patients with new GI events divided by patient-years of observation. In addition, the CHADS2 score was evaluated at baseline to determine the patient’s risk of stroke. A total of 557,123 AF patients were identified. The mean (median) AF patient age was 68.2 years (70); 45% were female. The cumulative incidences of any GI event and dyspepsia were 40% and 19%, respectively. The corresponding IRs were 38.8 and 14.7 events per 100 patient–years. IRs of any GI events for female and male patients were 43.6 and 35.5; for patients in the age groups <65, 65–74, 75–84, and ≥85 years, IRs were 32.3, 38.9, 44.6, and 52.7; for patients with a CHADS2 score of 0, 1–2, 3–4, and 5–6, IRs were 30.3, 41.6, 56.9, and 74.5, respectively. In this large claims database, 40% of AF patients experienced a GI event, predominantly dyspepsia. Physicians should take age and comorbidities into consideration when managing AF patients.
Collapse
Affiliation(s)
- François Laliberté
- Groupe d'analyse, Ltée, 1000 rue de la Gauchetière Ouest, Bureau 1200, Montréal, Québec H3B 4 W5 Canada
| | - Yuliya Moore
- Groupe d'analyse, Ltée, 1000 rue de la Gauchetière Ouest, Bureau 1200, Montréal, Québec H3B 4 W5 Canada
| | - Katherine Dea
- Groupe d'analyse, Ltée, 1000 rue de la Gauchetière Ouest, Bureau 1200, Montréal, Québec H3B 4 W5 Canada
| | | | - Samir H Mody
- Janssen Scientific Affairs, LLC, Raritan, NJ USA
| | | | | | - C V Damaraju
- Janssen Scientific Affairs, LLC, Raritan, NJ USA
| | | | - Patrick Lefebvre
- Groupe d'analyse, Ltée, 1000 rue de la Gauchetière Ouest, Bureau 1200, Montréal, Québec H3B 4 W5 Canada
| |
Collapse
|
39
|
Webster L, Chey WD, Tack J, Lappalainen J, Diva U, Sostek M. Randomised clinical trial: the long-term safety and tolerability of naloxegol in patients with pain and opioid-induced constipation. Aliment Pharmacol Ther 2014; 40:771-9. [PMID: 25112584 DOI: 10.1111/apt.12899] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/12/2014] [Accepted: 07/14/2014] [Indexed: 12/08/2022]
Abstract
BACKGROUND Opioid-induced constipation (OIC) is a common adverse effect of opioid therapy. AIM To evaluate the long-term safety and tolerability of naloxegol, an oral, peripherally acting μ-opioid receptor antagonist (PAMORA), in patients with noncancer pain and OIC. METHODS A 52-week, multicenter, open-label, randomised, parallel-group phase 3 study was conducted in out-patients taking 30-1000 morphine-equivalent units per day for ≥4 weeks. Patients were randomised 2:1 to receive naloxegol 25 mg/day or usual-care (UC; investigator-chosen laxative regimen) treatment for OIC. RESULTS The safety set comprised 804 patients (naloxegol, n = 534; UC, n = 270). Mean exposure duration was 268 days with naloxegol and 297 days with UC. Frequency of adverse events (AEs) was 81.8% with naloxegol and 72.2% with UC. Treatment-emergent AEs occurring more frequently for naloxegol vs. UC were abdominal pain (17.8% vs. 3.3%), diarrhoea (12.9% vs. 5.9%), nausea (9.4% vs. 4.1%), headache (9.0% vs. 4.8%), flatulence (6.9% vs. 1.1%) and upper abdominal pain (5.1% vs. 1.1%). Most naloxegol-emergent gastrointestinal AEs occurred early, resolving during or after naloxegol discontinuation and were mild or moderate in severity; 11 patients discontinued due to diarrhoea and nine patients owing to abdominal pain. Pain scores and mean daily opioid doses remained stable throughout the study; no attributable opioid withdrawal AEs were observed. Two patients in each group had an adjudicated major adverse cardiovascular event unrelated to study drug; no AEs were reported nor adjudicated as bowel perforations. CONCLUSION In patients with noncancer pain and opioid-induced constipation, naloxegol 25 mg/day up to 52 weeks was generally safe and well tolerated.
Collapse
Affiliation(s)
- L Webster
- PRA Health Sciences, Salt Lake City, UT, USA
| | | | | | | | | | | |
Collapse
|
40
|
Poulsen JL, Brock C, Olesen AE, Nilsson M, Drewes AM. Clinical potential of naloxegol in the management of opioid-induced bowel dysfunction. Clin Exp Gastroenterol 2014; 7:345-58. [PMID: 25278772 PMCID: PMC4179399 DOI: 10.2147/ceg.s52097] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Opioid-induced bowel dysfunction (OIBD) is a burdensome condition which limits the therapeutic benefit of analgesia. It affects the entire gastrointestinal tract, predominantly by activating opioid receptors in the enteric nervous system, resulting in a wide range of symptoms, such as reflux, bloating, abdominal cramping, hard, dry stools, and incomplete evacuation. The majority of studies evaluating OIBD focus on constipation experienced in approximately 60% of patients. Nevertheless, other presentations of OIBD seem to be equally frequent. Furthermore, laxative treatment is often insufficient, which in many patients results in decreased quality of life and discontinuation of opioid treatment. Novel mechanism-based pharmacological approaches targeting the gastrointestinal opioid receptors have been marketed recently and even more are in the pipeline. One strategy is prolonged release formulation of the opioid antagonist naloxone (which has limited systemic absorption) and oxycodone in a combined tablet. Another approach is peripherally acting, μ-opioid receptor antagonists (PAMORAs) that selectively target μ-opioid receptors in the gastrointestinal tract. However, in Europe the only PAMORA approved for OIBD is the subcutaneously administered methylnaltrexone. Alvimopan is an oral PAMORA, but only approved in the US for postoperative ileus in hospitalized patients. Finally, naloxegol is a novel, oral PAMORA expected to be approved soon. In this review, the prevalence and pathophysiology of OIBD is presented. As PAMORAs seem to be a promising approach, their potential effect is reviewed with special focus on naloxegol's pharmacological properties, data on safety, efficacy, and patient-focused perspectives. In conclusion, as naloxegol is administered orally once daily, has proven efficacious compared to placebo, has an acceptable safety profile, and can be used as add-on to existing pain treatment, it is a welcoming addition to the targeted treatment possibilities for OIBD.
Collapse
Affiliation(s)
- Jakob Lykke Poulsen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Christina Brock
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark ; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Anne Estrup Olesen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark ; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matias Nilsson
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark ; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
41
|
Jagla C, Martus P, Stein C. Peripheral opioid receptor blockade increases postoperative morphine demands--a randomized, double-blind, placebo-controlled trial. Pain 2014; 155:2056-62. [PMID: 25046272 DOI: 10.1016/j.pain.2014.07.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/11/2014] [Accepted: 07/15/2014] [Indexed: 12/11/2022]
Abstract
Experimental studies suggest that a large proportion of opioid analgesia can be mediated by peripheral opioid receptors. This trial examined the contribution of such receptors to clinical analgesia induced by intravenous morphine. We hypothesized that the selective blockade of peripheral opioid receptors by methylnaltrexone (MNX) would increase the patients' demand for morphine to achieve satisfactory postoperative pain relief. In a double-blind, placebo-controlled, sequential 2-center trial, 50 patients undergoing knee replacement surgery were randomized (1:1) to receive either subcutaneous MNX (0.9 mg/kg) (hospital I: n=14; hospital II: n=11) or saline (hospital I: n=13; hospital II: n=12) at the end of surgery. The primary endpoint was the cumulative amount of intravenous morphine administered during the first 8 hours. Secondary endpoints were pain scores at rest and during movement (by numerical rating scale and McGill Questionnaire), vital signs, adverse side effects, and withdrawal symptoms. After MNX, demands for morphine were strongly (by about 40%) increased (hospital I: 35.31 ± 12.99 mg vs 25.51 ± 7.92 mg, P=0.03; hospital II: 35.42 ± 11.73 mg vs 24.80 ± 7.84 mg, P=0.02; pooled data: P<.001; means ± SD). Secondary endpoints were similar in all groups (P>.05). Thus, a significant proportion of analgesia produced by systemically administered morphine is mediated by peripheral opioid receptors. Drugs that selectively activate such receptors should have the potential to produce powerful clinical pain relief.
Collapse
Affiliation(s)
- Christina Jagla
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Charité, Campus Benjamin Franklin, Freie Universität Berlin, Berlin, Germany; Helmholtz Virtual Institute Multifunctional Biomaterials for Medicine, Teltow, Germany
| | - Peter Martus
- Institut für Klinische Epidemiologie und Angewandte Biometrie, Universität Tübingen, Tübingen, Germany
| | - Christoph Stein
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Charité, Campus Benjamin Franklin, Freie Universität Berlin, Berlin, Germany; Helmholtz Virtual Institute Multifunctional Biomaterials for Medicine, Teltow, Germany.
| |
Collapse
|
42
|
Bui K, She F, Sostek M. The effects of renal impairment on the pharmacokinetics, safety, and tolerability of naloxegol. J Clin Pharmacol 2014; 54:1375-82. [PMID: 24946021 DOI: 10.1002/jcph.349] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/17/2014] [Indexed: 01/28/2023]
Abstract
The impact of renal impairment on the pharmacokinetics of a 25-mg oral dose of naloxegol was examined in patients with renal impairment classified as moderate, severe, or end-stage renal disease (ESRD) and compared with healthy subjects (n = 8/group). Geometric mean area under the plasma concentration-time curve (AUC) was increased in patients with moderate (1.7-fold) or severe (2.2-fold) impairment, and maximum plasma concentrations (Cmax ) were elevated in patients with moderate (1.1-fold) or severe (1.8-fold) impairment. These findings were driven by higher exposures in two patients in each of the moderate and severe impairment groups; exposures in all other patients were similar to the control group. Overall exposures in ESRD patients were similar and Cmax was 29% lower versus normal subjects. Renal impairment minimally affected other plasma pharmacokinetic parameters. As renal clearance was a minor component of total clearance, exposure to naloxegol was unaffected by the degree of renal impairment, with no correlation between either AUC or Cmax and estimated glomerular filtration rate (eGFR). Hemodialysis was an ineffective means to remove naloxegol. Naloxegol was generally well tolerated in all groups. Renal impairment could adversely affect clearance by hepatic and gut metabolism, resulting in the increased exposures observed in outliers of the moderate and severe renal impairment groups.
Collapse
Affiliation(s)
- Khanh Bui
- AstraZeneca Pharmaceuticals, Wilmington, DE, USA
| | | | | |
Collapse
|
43
|
Chey WD, Webster L, Sostek M, Lappalainen J, Barker PN, Tack J. Naloxegol for opioid-induced constipation in patients with noncancer pain. N Engl J Med 2014; 370:2387-96. [PMID: 24896818 DOI: 10.1056/nejmoa1310246] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Opioid-induced constipation is common and debilitating. We investigated the efficacy and safety of naloxegol, an oral, peripherally acting, μ-opioid receptor antagonist, for the treatment of opioid-induced constipation. METHODS In two identical phase 3, double-blind studies (study 04, 652 participants; study 05, 700 participants), outpatients with noncancer pain and opioid-induced constipation were randomly assigned to receive a daily dose of 12.5 or 25 mg of naloxegol or placebo. The primary end point was the 12-week response rate (≥3 spontaneous bowel movements per week and an increase from baseline of ≥1 spontaneous bowel movements for ≥9 of 12 weeks and for ≥3 of the final 4 weeks) in the intention-to-treat population. The key secondary end points were the response rate in the subpopulation of patients with an inadequate response to laxatives before enrollment, time to first postdose spontaneous bowel movement, and mean number of days per week with one or more spontaneous bowel movements. RESULTS Response rates were significantly higher with 25 mg of naloxegol than with placebo (intention-to-treat population: study 04, 44.4% vs. 29.4%, P=0.001; study 05, 39.7% vs. 29.3%, P=0.02; patients with an inadequate response to laxatives: study 04, 48.7% vs. 28.8%, P=0.002; study 05, 46.8% vs. 31.4%, P=0.01); in study 04, response rates were also higher in the group treated with 12.5 mg of naloxegol (intention-to-treat population, 40.8% vs. 29.4%, P=0.02; patients with an inadequate response to laxatives, 42.6% vs. 28.8%, P=0.03). A shorter time to the first postdose spontaneous bowel movement and a higher mean number of days per week with one or more spontaneous bowel movements were observed with 25 mg of naloxegol versus placebo in both studies (P<0.001) and with 12.5 mg of naloxegol in study 04 (P<0.001). Pain scores and daily opioid dose were similar among the three groups. Adverse events (primarily gastrointestinal) occurred most frequently in the groups treated with 25 mg of naloxegol. CONCLUSIONS Treatment with naloxegol, as compared with placebo, resulted in a significantly higher rate of treatment response, without reducing opioid-mediated analgesia. (Funded by AstraZeneca; KODIAC-04 and KODIAC-05 ClinicalTrials.gov numbers, NCT01309841 and NCT01323790, respectively.).
Collapse
Affiliation(s)
- William D Chey
- From the Department of Internal Medicine, University of Michigan Health System, Ann Arbor (W.D.C.); PRA International, Salt Lake City (L.W.); AstraZeneca Pharmaceuticals, Wilmington, DE (M.S., J.L., P.N.B.); and the Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium (J.T.)
| | | | | | | | | | | |
Collapse
|
44
|
Bassotti G, Blandizzi C. Understanding and treating refractory constipation. World J Gastrointest Pharmacol Ther 2014; 5:77-85. [PMID: 24868488 PMCID: PMC4023327 DOI: 10.4292/wjgpt.v5.i2.77] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/20/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic constipation is a frequently encountered disorder in clinical practice. Most constipated patients benefit from standard medical approaches. However, current therapies may fail in a proportion of patients. These patients deserve better evaluation and thorough investigations before their labeling as refractory to treatment. Indeed, several cases of apparent refractoriness are actually due to misconceptions about constipation, poor basal evaluation (inability to recognize secondary causes of constipation, use of constipating drugs) or inadequate therapeutic regimens. After a careful re-evaluation that takes into account the above factors, a certain percentage of patients can be defined as being actually resistant to first-line medical treatments. These subjects should firstly undergo specific diagnostic examination to ascertain the subtype of constipation. The subsequent therapeutic approach should be then tailored according to their underlying dysfunction. Slow transit patients could benefit from a more robust medical treatment, based on stimulant laxatives (or their combination with osmotic laxatives, particularly over the short-term), enterokinetics (such as prucalopride) or secretagogues (such as lubiprostone or linaclotide). Patients complaining of obstructed defecation are less likely to show a response to medical treatment and might benefit from biofeedback, when available. When all medical treatments prove to be unsatisfactory, other approaches may be attempted in selected patients (sacral neuromodulation, local injection of botulinum toxin, anterograde continence enemas), although with largely unpredictable outcomes. A further although irreversible step is surgery (subtotal colectomy with ileorectal anastomosis or stapled transanal rectal resection), which may confer some benefit to a few patients with refractoriness to medical treatments.
Collapse
|
45
|
Leppert W. New treatment possibilities for opioid-induced bowel dysfunction. Pain 2014; 154:1491-1492. [PMID: 23953125 DOI: 10.1016/j.pain.2013.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/25/2013] [Accepted: 06/06/2013] [Indexed: 01/18/2023]
Affiliation(s)
- Wojciech Leppert
- Chair and Department of Palliative Medicine, Poznan University of Medical Sciences, Osiedle Rusa 25 A, 61-245 Poznan, Poland
| |
Collapse
|
46
|
Rauck RL. Treatment of opioid-induced constipation: focus on the peripheral μ-opioid receptor antagonist methylnaltrexone. Drugs 2014; 73:1297-306. [PMID: 23881667 DOI: 10.1007/s40265-013-0084-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Most prescribed opioids exert their analgesic effects via activation of central μ-opioid receptors. However, μ-opioid receptors are also located in the gastrointestinal (GI) tract, and activation of these receptors by opioids can lead to GI-related adverse effects, in particular opioid-induced constipation (OIC). OIC has been associated with increased use of healthcare resources, increased healthcare costs, and decreased quality of life for patients. Nonpharmacologic (e.g., increased fiber uptake) and pharmacologic agents (e.g., laxatives) may be considered for the treatment and prevention of OIC. However, many interventions, such as laxatives alone, are generally insufficient to reverse OIC because they do not target the underlying cause of OIC, opioid activation of μ-opioid receptors in the GI tract. Therefore, there has been keen interest in antagonism of the μ-opioid receptor in the periphery to inhibit the effects of opioids in the GI tract. In this review, currently available pharmacologic therapies for the treatment and prevention of OIC are summarized briefly, with a primary focus on the administration of the peripheral μ-opioid receptor antagonist methylnaltrexone bromide in patients with OIC and advanced illness who are receiving palliative care. Also, clinical trial data of methylnaltrexone treatment in patients with OIC and other pain conditions (i.e., chronic noncancer pain and pain after orthopedic surgery) are reviewed. Data support that methylnaltrexone is efficacious for the treatment of OIC and has a favorable tolerability profile.
Collapse
|
47
|
Abstract
Opiates are among the oldest medications available to manage a number of medical problems. Although pain is the current focus, early use initially focused upon the treatment of dysentery. Opium contains high concentrations of both morphine and codeine, along with thebaine, which is used in the synthesis of a number of semisynthetic opioid analgesics. Thus, it is not surprising that new agents were initially based upon the morphine scaffold. The concept of multiple opioid receptors was first suggested almost 50 years ago (Martin, 1967), opening the possibility of new classes of drugs, but the morphine-like agents have remained the mainstay in the medical management of pain. Termed mu, our understanding of these morphine-like agents and their receptors has undergone an evolution in thinking over the past 35 years. Early pharmacological studies identified three major classes of receptors, helped by the discovery of endogenous opioid peptides and receptor subtypes-primarily through the synthesis of novel agents. These chemical biologic approaches were then eclipsed by the molecular biology revolution, which now reveals a complexity of the morphine-like agents and their receptors that had not been previously appreciated.
Collapse
Affiliation(s)
- Gavril W Pasternak
- Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065.
| | | |
Collapse
|
48
|
Labianca R, Sarzi-Puttini P, Zuccaro SM, Cherubino P, Vellucci R, Fornasari D. Adverse effects associated with non-opioid and opioid treatment in patients with chronic pain. Clin Drug Investig 2013; 32 Suppl 1:53-63. [PMID: 23389876 DOI: 10.2165/11630080-000000000-00000] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic pain is a debilitating condition that is associated with many common diseases; this places a major burden on the healthcare system. There are currently numerous analgesic agents available for the treatment of chronic pain. In general, the oral non-opioid analgesic, paracetamol, is recommended for the initial treatment of mild to moderate pain. Therapeutic doses of paracetamol do not appear to result in hepatotoxicity, although overdose may lead to acute liver failure. Current data suggest that paracetamol has acceptable gastrointestinal tolerability. Another class of non-opioid analgesic with confirmed efficacy for the treatment of chronic mild to moderate pain are non-steroidal anti-inflammatory drugs (NSAIDs), although this efficacy is offset by the potential of adverse gastrointestinal events. In particular, non-selective NSAIDs, also known as cyclooxygenase (COX) inhibitors, carry an increased risk of serious upper gastrointestinal complications, including ulcers, perforation and bleeding. The introduction of COX-2 inhibitors provided a NSAID-based option with improved gastrointestinal safety, but increased risk of cardiovascular effects. Opioids are powerful analgesic agents used to treat moderate to severe chronic pain. However, treatment with opioids is associated with a number of common adverse effects, including constipation, nausea or vomiting, pruritus, somnolence or cognitive impairment, dry mouth, tolerance or dependence and urinary retention. Although there are multiple strategies in place to manage adverse events that arise from both non-opioid and opioid analgesic therapy, a better understanding of the mechanisms involved in the development of specific drug-related adverse effects is required along with proper prescribing practices and adequate physician/patient education. Balanced against the adverse effects of pain management medications, there is a need to be mindful of the widespread, often serious, adverse consequences of poorly managed pain itself.
Collapse
|
49
|
Affiliation(s)
- Bill H McCarberg
- Adjunct Assistant Clinical Professor, University of California at San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
50
|
Webster L, Dhar S, Eldon M, Masuoka L, Lappalainen J, Sostek M. A phase 2, double-blind, randomized, placebo-controlled, dose-escalation study to evaluate the efficacy, safety, and tolerability of naloxegol in patients with opioid-induced constipation. Pain 2013; 154:1542-1550. [PMID: 23726675 DOI: 10.1016/j.pain.2013.04.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/04/2013] [Accepted: 04/09/2013] [Indexed: 12/24/2022]
Abstract
Naloxegol (previously known as NKTR-118) is a peripherally acting μ-opioid receptor antagonist engineered using polymer conjugate technology in development as an oral, once-daily agent for the treatment of opioid-induced constipation (OIC). Eligible patients with OIC (n=207), defined as <3 spontaneous bowel movements (SBMs) per week with accompanying symptoms, on a stable opioid regimen of 30-1000 mg/day morphine equivalents for ≥ 2 weeks were randomized to receive 4 weeks of double-blind placebo or naloxegol (5, 25, or 50mg) once daily in sequential cohorts after a 1-week placebo run-in. The primary end point, median change from baseline in SBMs per week after week 1 of drug administration, was statistically significant for the 25- and 50-mg naloxegol cohorts vs placebo (2.9 vs 1.0 [P=0.0020] and 3.3 vs 0.5 [P=0.0001], respectively). The increase in SBMs vs placebo was maintained over 4 weeks for naloxegol 25mg (3.0 vs 0.8 [P=0.0022]) and 50mg (3.5 vs 1.0 [P<0.0001]). Naloxegol was generally well tolerated across all dosages. The most frequent adverse events (AEs) were abdominal pain, diarrhea, and nausea. Most AEs at 5 and 25mg/day were mild and transient. Similar AEs occurred with increased frequency and severity in the 50-mg cohort. There was no evidence of a statistically significant increase from baseline in pain, opioid use for the 25- and 50-mg cohorts, or centrally mediated opioid withdrawal signs and/or symptoms with naloxegol. These data demonstrate that once-daily oral naloxegol improves the frequency of SBMs compared with placebo and is generally well tolerated in this population of patients with OIC.
Collapse
Affiliation(s)
- Lynn Webster
- CRI Lifetree Research, Salt Lake City, UT, USA Genentech Inc., San Francisco, CA, USA Nektar Therapeutics, San Francisco, CA, USA Oakland, CA, USA AstraZeneca Pharmaceuticals, Wilmington, DE, USA
| | | | | | | | | | | |
Collapse
|