1
|
Seidmohammadi K, Haghshenas H, Moghaddam S, Kargar Jahromi H, Delam H. The Effectiveness of Tramadol in Pain Relief in Chronic Diseases: A Review Based on Clinical Trials. J Pain Palliat Care Pharmacother 2025; 39:139-161. [PMID: 39440803 DOI: 10.1080/15360288.2024.2411239] [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] [Received: 06/14/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Tramadol is a synthetic opioid with a central effect from the aminocyclohexanol group, which has two main mechanisms of action, including as a weak agonist of opioid receptors and as a norepinephrine and serotonin reuptake inhibitor. The present study presents a review based on clinical trials designed in 2023. In July 2023, six international databases, including Medline/PubMed, ProQuest, Scopus, EMBASE, Google Scholar, and ISI (Web of Science), were searched and 58 articles were included in the study. The results of most studies showed that tramadol can be used as an analgesic drug, although in some studies it was shown that tramadol is not therapeutically superior in reducing pain compared to other treatments. Also, complications related to this treatment have been reported in some studies. Physicians should consider these factors to prevent drug toxicity, poor pain relief, use disorder in patients, and unpredictable complications. It should be noted that there is not enough evidence to support the long-term effectiveness of tramadol, but this argument also extends to nonopioid and other types of opioid analgesics, and the lack of long-term trials is due to regulatory and ethical issues. Although opioids can cause addiction when used for a long time, tramadol has a reasonable safety profile. According to the patient's condition and the clinical judgment of the medical professional, tramadol can be prescribed for patients, but the consequences of its use must be considered and a personalized treatment algorithm should be selected if the benefits outweigh the risks of the drug.
Collapse
Affiliation(s)
- Kosar Seidmohammadi
- Kosar Seidmohammadi, Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran and Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Hoda Haghshenas
- Hoda Haghshenas, MD, Student Research Committee, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Sara Moghaddam
- Sara Moghaddam, Instructor, Department of Nursing, Faculty of Nursing, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Hossein Kargar Jahromi
- Hossein Kargar Jahromi, PhD, Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Hamed Delam
- Hamed Delam, PhD Student of Epidemiology, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Sato T, Fukutomi A, Kawamura T, Kawakami K, Sato T, Kamo Y, Suzuki T, Hagiya S, Tanaka R. Low-dose add-on methadone for cancer pain management: a retrospective analysis of 102 Japanese patients. Jpn J Clin Oncol 2025; 55:123-130. [PMID: 39498764 DOI: 10.1093/jjco/hyae156] [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] [Received: 07/31/2024] [Accepted: 10/18/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Methadone was introduced in 2013 for the treatment of intractable cancer pain in Japan and is indicated for patients receiving opioid doses ≧60 mg/day as an oral morphine equivalent. Low-dose (≦10 mg/day) add-on methadone to prior opioids has been reported from European countries to successfully relieve various types of intractable cancer pain; however, there are few reports of such use in Japan. The aim of this study was to analyze more than a hundred cases with low-dose add-on methadone to treat intractable pain in Japanese cancer patients. METHODS All cases in which 5 or 10 mg/day of methadone was added to prior opioids by the Palliative Care Team or Division of Palliative Medicine in our hospital during the period between April 2016 and September 2023 were extracted and analyzed retrospectively on electrical medical charts. RESULTS AND CONCLUSIONS A total of 102 cases were extracted with a male-to-female ratio of 60:42, and the age (mean ± SD) was 62.8 ± 14.7 years old. Methadone was introduced in an inpatient setting to 86 patients. The major pathologies that caused intractable pain were spinal metastases in 48, pelvis or pelvic floor lesions in 29 and pleural and/or chest wall lesions in 16. The most common mechanism of pain was the mixture of somatic and neuropathic components. The major opioids administered prior to methadone included tapentadol in 46 patients, hydromorphone in 36 and oxycodone in 19. The dose of the prior opioids [median, (interquartile range: IQR)] was 97, (62.8-167.3) (range: 15-1313) mg/day of oral morphine equivalent. Radiotherapy, chemotherapy and nerve blocks were performed as concomitant therapies in 48, 22 and 11 patients, respectively (with some overlap). The number of rescue doses [median (IQR)] was significantly decreased from three (two to five) on the day before methadone to one (zero to four) after seven days from methadone initiation. The side effects leading to discontinuation of methadone were drowsiness in three cases, nausea in three cases and dizziness in one case (with some overlap). Compared with complete switching from other opioids, low-dose add-on methadone can reduce the possibility of major dose discrepancies and can be quickly adjusted by combined opioid reduction/increase. Low-dose add-on methadone can be an effective and safe method for intractable cancer pain.
Collapse
Affiliation(s)
- Tetsumi Sato
- Division of Palliative Medicine, Shizuoka Cancer Center, Nagaizumi-cho, Shiuzoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
| | - Akira Fukutomi
- Division of Palliative Medicine, Shizuoka Cancer Center, Nagaizumi-cho, Shiuzoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
| | - Taiichi Kawamura
- Division of Palliative Medicine, Shizuoka Cancer Center, Nagaizumi-cho, Shiuzoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
| | - Kyohei Kawakami
- Division of Palliative Medicine, Shizuoka Cancer Center, Nagaizumi-cho, Shiuzoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
| | - Tetsu Sato
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
- Department of Pharmacy, Shizuoka Cancer Center, Nagaizumi-cho, Japan
| | - Yoshiko Kamo
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
- Department of Pharmacy, Shizuoka Cancer Center, Nagaizumi-cho, Japan
| | - Tomomi Suzuki
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
- Patient and Family Support Center, Shizuoka Cancer Center, Nagaizuim-cho, Japan
| | - Shota Hagiya
- Palliative Care Team, Shizuoka Cancer Center, Nagaizumi-cho, Shizuoka, Japan
- Patient and Family Support Center, Shizuoka Cancer Center, Nagaizuim-cho, Japan
| | - Rei Tanaka
- Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
Zapata-Morales JR, Alonso-Castro AJ, González-Rivera ML, González Prado HI, Barragán-Gálvez JC, Hernández-Flores A, Juárez-Vázquez MDC, Domínguez F, Carranza-Álvarez C, de Jesús Pozos-Guillén A, López-Rodríguez JF, Aguirre-Bañuelos P, Ramírez-Morales MA. Synergistic Interaction Between Justicia spicigera Extract and Analgesics on the Formalin Test in Rats. Pharmaceuticals (Basel) 2025; 18:187. [PMID: 40006001 PMCID: PMC11859130 DOI: 10.3390/ph18020187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Combining antinociceptive drugs with different mechanisms of action can reduce the doses and the adverse effects, with a possible increase in the antinociceptive effect. This work evaluated the antinociceptive effect of the combination of an ethanol extract of Justicia spicigera (JSE) with naproxen (NPX) or tramadol (TML) using the formalin test in rats. Methods: Rats received JSE (30-200 mg/kg p.o.), NPX (50-300 mg/kg p.o.), or TML (5-50 mg/kg p.o.) 60 min before paw administration with formalin (5%). Different proportions of the combination between NPX and JSE, as well as TML and JSE, were used in the formalin test to obtain the dose-response curve of each drug and the experimental effective dose 50 (ED50). The levels of IL-1β and COX2 were assessed using a Western blot analysis as a possible mechanism of action for the combination of JSE and analgesics. A pharmacokinetic study was conducted to evaluate the effect of JSE on the pharmacokinetic parameters of NPX. Results: The ED50 values for the proportions NPX:JSE were 107.09 mg/kg (1:1), 102.44 mg/kg (3:1), and 73.82 mg/kg (1:3). The ED50 values for the proportions TML:JSE were 66 mg/kg (1:1), 29.5 mg/kg (1:3), and 78 mg/kg (3:1). The combination NPX:JSE (1:3) showed the best synergistic interaction index (0.501). The pharmacokinetic study revealed that there were no significant changes in the pharmacokinetic parameters of NPX administered individually and the combination NPX:JSE. Conclusions: In this preclinical study, the combination NPX:JSE showed antinociceptive effects by decreasing the levels of COX2 and IL-1β without affecting NPX's pharmacokinetics.
Collapse
Affiliation(s)
- Juan Ramón Zapata-Morales
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - Angel Josabad Alonso-Castro
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - María Leonor González-Rivera
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - Hugo Israel González Prado
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - Juan Carlos Barragán-Gálvez
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - Araceli Hernández-Flores
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - María del Carmen Juárez-Vázquez
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| | - Fabiola Domínguez
- Eastern Biomedical Research Center of Mexican Social Security Institute (IMSS), Metepec 74360, Mexico;
| | - Candy Carranza-Álvarez
- School of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosí, Ciudad Valles 79060, Mexico;
| | - Amaury de Jesús Pozos-Guillén
- Basics Sciences Laboratory, School of Dentistry, Autonomous University of San Luis Potosi (UASLP), San Luis Potosí 78290, Mexico;
| | - Juan F. López-Rodríguez
- Animal Laboratory, School of Medicine Autonomous University of San Luis Potosi, San Luis Potosi 78290, Mexico;
| | - Patricia Aguirre-Bañuelos
- Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, Manuel Nava Martínez 6 Avenue, San Luis Potosí 78210, Mexico;
| | - Marco Antonio Ramírez-Morales
- Department of Pharmacy, Natural and Exact Sciences Division, University of Guanajuato (UG), Guanajuato 36050, Mexico; (A.J.A.-C.); (M.L.G.-R.); (H.I.G.P.); (J.C.B.-G.); (A.H.-F.); (M.d.C.J.-V.); (M.A.R.-M.)
| |
Collapse
|
4
|
Liu D, Wang J, Hou T, Zhang Y, Zhou H, Zhao Y, Zhou L, Cao C, Liu Y, Liang X. Dihydrokoumine, a dual-target analgesic with reduced side effects isolated from a traditional Chinese medicine. J Adv Res 2024:S2090-1232(24)00465-X. [PMID: 39461422 DOI: 10.1016/j.jare.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
INTRODUCTION Opioids are the most common antinociceptive drugs, but long-term administration causes serious adverse side effects. Gelsemium elegans Benth. is traditionally used as an analgesic agent and mainly contains indole alkaloids with structures different from those in common opioids, indicating distinct pharmacological properties. This work aims to find a new analgesic from Gelsemium elegans Benth. and evaluate it in vitro and in vivo. METHODS Dihydrokoumine was purified from Gelsemium elegans Benth. Binding to mu opioid receptor (MOR), M3 receptor (M3R) and other 15 G protein-coupled receptors were evaluated in vitro combined with molecular docking analysis. Analgesic efficacy and side effects were measured in vivo using hot-plate, formalin paw, and rotarod tests in mice. Cytotoxicity, acute toxicity in mice and pharmacokinetics were assessed. RESULTS A MOR agonist, dihydrokoumine, was first identified from Gelsemium elegans Benth. Further investigations showed that dihydrokoumine exhibited selective partial agonist action on the MOR and antagonist action on the M3R among other 15 GPCRs. In in vivo mouse models, dihydrokoumine could relieve acute pain and chronic inflammatory pain without drug tolerance and sedative side effects. Additionally, we observed a good safety profile and favorable pharmacokinetic properties. CONCLUSION A MOR partial agonist/M3R antagonist analgesic with reduced side effects was isolated from a traditional Chinese medicine. This study bestows dihydrokoumine as a new dual-target analgesic and as a potential lead compound in long-term pain management.
Collapse
Affiliation(s)
- Dian Liu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Jixia Wang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Tao Hou
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Zhou
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yaopeng Zhao
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Liangliang Zhou
- Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Cuiyan Cao
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Yanfang Liu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| | - Xinmiao Liang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| |
Collapse
|
5
|
Dibaei M, Hosseini A, Lavasani H, Kiani-Dehkordi B, Rouini M. Assessment of metabolic interaction between curcumin and tramadol using the isolated perfused rat liver. Heliyon 2024; 10:e35070. [PMID: 39170468 PMCID: PMC11336359 DOI: 10.1016/j.heliyon.2024.e35070] [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: 03/20/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction The presence of phytochemicals in herbal medicines can lead to herb-drug interactions, altering the levels of these compounds and conventional drugs in the bloodstream by influencing CYP450 activity. Considering curcumin's effect on the CYP enzymes responsible for tramadol metabolism, it is essential to assess the potential interaction between curcumin and tramadol when administered together. Materials and methods The pharmacokinetics of tramadol were examined in rats receiving either single or multiple doses of curcumin (80 mg/kg) compared to rats without curcumin treatment. Tramadol liver perfusion was conducted on all rat groups and perfusate samples were collected at specified intervals. Tramadol and its main metabolite were detected using an HPLC system coupled with a fluorescence detector. Results Tramadol concentrations were notably higher in the co-administered group compared to both the control and treatment groups. Conversely, lower concentrations of M1 were observed in the co-administered and treatment groups compared to the control group. The AUC0-60 parameters for tramadol were as follows: 32944.8 ± 1355.5, 22925.7 ± 1650.1, and 36548.0 ± 2808.4 ng⋅min/ml for the control, treatment, and co-administered groups, respectively. Both the co-administered and treatment groups exhibited a lower AUC0-60 of M1 compared to the control group. The lack of significant difference in Cmax and AUC0-60 of M1 between the treatment and co-administered groups suggests that single and multiple doses of curcumin have comparable effects on CYP2D6. Conclusions These results indicate a potential for drug interactions when curcumin and tramadol are taken together. Furthermore, the influence of curcumin on tramadol metabolism varied between single and multiple oral administrations of curcumin. Hence, it is vital to highlight this interaction in clinical settings and conduct additional research to fully understand the clinical implications of combining curcumin and tramadol.
Collapse
Affiliation(s)
- Maryam Dibaei
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hoda Lavasani
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Banafsheh Kiani-Dehkordi
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Rouini
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Sato T, Ono S, Sato T, Tanaka R, Kamo Y, Suzuki T. Safety and Efficacy of Combined Injection of Pure-μ-Opioid Agonist with Tramadol as an Opioid Induction Agent for Opioid-Naïve Cancer Patients. Palliat Med Rep 2024; 5:340-349. [PMID: 39144134 PMCID: PMC11319850 DOI: 10.1089/pmr.2023.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 08/16/2024] Open
Abstract
Background Tramadol is known to provide synergistic analgesia when used in combination with morphine. Objectives The aims of this study were: (1) to introduce an opioid combination therapy using pure-μ-opioid receptor agonist (OPI) + tramadol injections (OPI + tramadol) and (2) to elucidate safety and efficacy of this combination therapy for opioid-naïve cancer pain patients. Methods Opioid-naïve patients referred to our palliative care team (in Japan) who were unable to take oral medications and received OPI + tramadol as opioid induction agents were retrospectively investigated on the electric medical chart. OPI + tramadol dosage was adjusted to achieve the patient's pain as Numerical Rating Scale ≤4/10 or Support Team Assessment Schedule-Japanese ≤1. Patients' demography, doses of OPI and tramadol administered, and adverse events were analyzed. Results A total of 44 patients were included. The primary organs of malignancy were pancreas (11), stomach (5), lung (4), breast (4), liver (4), and others (13). OPI injections administered were hydromorphone (39), morphine (6), oxycodone (1), and fentanyl (1). The starting doses of OPI (morphine equivalent) and tramadol were 6.05 ± 1.63 and 67.8 ± 13.6 mg/day, respectively, and the final doses of OPI (morphine equivalent) and tramadol were 8.14 ± 3.85 and 80.0 ± 28.5 mg/day, respectively. Treatment goals were achieved in all patients. There were three patients in whom OPI was switched owing to inadequate analgesia and no new side effects other than those known to occur when OPI or tramadol is administered appeared. Conclusion The results suggest that this innovative and unique opioid therapy can be safely and effectively introduced to opioid-naïve cancer patients who are relatively close to the end of life.
Collapse
Affiliation(s)
- Tetsumi Sato
- Division of Palliative Medicine, Shizuoka Cancer Center, Shizuoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shigeki Ono
- Division of Palliative Medicine, Shizuoka Cancer Center, Shizuoka, Japan
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
| | - Tetsu Sato
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
- Department of Pharmacy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Rei Tanaka
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
- Department of Pharmacy, Shizuoka Cancer Center, Shizuoka, Japan
- Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama, Japan
| | - Yoshiko Kamo
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
- Department of Pharmacy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Tomomi Suzuki
- Palliative Care Team, Shizuoka Cancer Center, Shizuoka, Japan
- Patient and Family Support Center, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
7
|
Soares-Santos RR, Machado DP, Romero TL, Duarte IDG. Nitric oxide and potassium channels but not opioid and cannabinoid receptors mediate tramadol-induced peripheral antinociception in rat model of paw pressure withdrawal. Can J Physiol Pharmacol 2024; 102:218-227. [PMID: 37976474 DOI: 10.1139/cjpp-2023-0314] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Tramadol, an analgesic classified as an "atypical opioid", exhibits both opioid and non-opioid mechanisms of action. This study aimed to explore these mechanisms, specifically the opioid-, cannabinoid-, nitric oxide-, and potassium channel-based mechanisms, which contribute to the peripheral antinociception effect of tramadol, in an experimental rat model. The nociceptive threshold was determined using paw pressure withdrawal. To examine the mechanisms of action, several substances were administered intraplantarly: naloxone, a non-selective opioid antagonist (50 µg/paw); AM251 (80 µg/paw) and AM630 (100 µg/paw) as the selective antagonists for types 1 and 2 cannabinoid receptors, respectively; nitric oxide synthase inhibitors L-NOArg, L-NIO, L-NPA, and L-NIL (24 µg/paw); and the enzyme inhibitors of guanylatocyclase and phosphodiesterase of cGMP, ODQ, and zaprinast. Additionally, potassium channel blockers glibenclamide, tetraethylammonium, dequalinium, and paxillin were used. The results showed that opioid and cannabinoid receptor antagonists did not reverse tramadol's effects. L-NOarg, L-NIO, and L-NPA partially reversed antinociception, while ODQ completely reversed, and zaprinast enhanced tramadol's antinociception effect. Notably, glibenclamide blocked tramadol's antinociception in a dose-dependent manner. These findings suggest that tramadol's peripheral antinociception effect is likely mediated by the nitrergic pathway and sensitive ATP potassium channels, rather than the opioid and cannabinoid pathways.
Collapse
Affiliation(s)
- Raquel R Soares-Santos
- Department of Pharmacology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Daniel P Machado
- Department of Pharmacology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thiago L Romero
- Department of Pharmacology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Igor D G Duarte
- Department of Pharmacology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
8
|
Boccella S, De Filippis L, Giorgio C, Brandolini L, Jones M, Novelli R, Amorizzo E, Leoni MLG, Terranova G, Maione S, Luongo L, Leone M, Allegretti M, Minnella EM, Aramini A. Combination Drug Therapy for the Management of Chronic Neuropathic Pain. Biomolecules 2023; 13:1802. [PMID: 38136672 PMCID: PMC10741625 DOI: 10.3390/biom13121802] [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] [Received: 11/01/2023] [Revised: 12/01/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic neuropathic pain (NP) is an increasingly prevalent disease and leading cause of disability which is challenging to treat. Several distinct classes of drugs are currently used for the treatment of chronic NP, but each drug targets only narrow components of the underlying pathophysiological mechanisms, bears limited efficacy, and comes with dose-limiting side effects. Multimodal therapies have been increasingly proposed as potential therapeutic approaches to target the multiple mechanisms underlying nociceptive transmission and modulation. However, while preclinical studies with combination therapies showed promise to improve efficacy over monotherapy, clinical trial data on their efficacy in specific populations are lacking and increased risk for adverse effects should be carefully considered. Drug-drug co-crystallization has emerged as an innovative pharmacological approach which can combine two or more different active pharmaceutical ingredients in a single crystal, optimizing pharmacokinetic and physicochemical characteristics of the native molecules, thus potentially capitalizing on the synergistic efficacy between classes of drugs while simplifying adherence and minimizing the risk of side effects by reducing the doses. In this work, we review the current pharmacological options for the treatment of chronic NP, focusing on combination therapies and their ongoing developing programs and highlighting the potential of co-crystals as novel approaches to chronic NP management.
Collapse
Affiliation(s)
- Serena Boccella
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via De Amicis, 80131 Naples, Italy; (S.B.); (C.G.)
| | - Lidia De Filippis
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via S. Lucia, 20122 Milan, Italy; (L.D.F.); (R.N.); (M.L.); (E.M.M.)
| | - Cristina Giorgio
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via De Amicis, 80131 Naples, Italy; (S.B.); (C.G.)
| | - Laura Brandolini
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via Campo di Pile, 67100 L’Aquila, Italy; (L.B.); (M.A.)
| | - Meghan Jones
- Research & Early Development (R&D), Dompé US, 181 2nd Avenue, STE 600, San Mateo, CA 94401, USA;
| | - Rubina Novelli
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via S. Lucia, 20122 Milan, Italy; (L.D.F.); (R.N.); (M.L.); (E.M.M.)
| | - Ezio Amorizzo
- Pain Unit, San Paolo Hospital, 00053 Civitavecchia, Italy;
- Pain Clinic Roma, 00191 Rome, Italy
| | - Matteo Luigi Giuseppe Leoni
- Azienda USL di Piacenza, 29121 Piacenza, Italy;
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza University, 00185 Rome, Italy
| | | | - Sabatino Maione
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.M.); (L.L.)
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.M.); (L.L.)
| | - Manuela Leone
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via S. Lucia, 20122 Milan, Italy; (L.D.F.); (R.N.); (M.L.); (E.M.M.)
| | - Marcello Allegretti
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via Campo di Pile, 67100 L’Aquila, Italy; (L.B.); (M.A.)
| | - Enrico Maria Minnella
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via S. Lucia, 20122 Milan, Italy; (L.D.F.); (R.N.); (M.L.); (E.M.M.)
| | - Andrea Aramini
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via Campo di Pile, 67100 L’Aquila, Italy; (L.B.); (M.A.)
| |
Collapse
|
9
|
He Y, Su Q, Zhao L, Zhang L, Yu L, Shi J. Historical perspectives and recent advances in small molecule ligands of selective/biased/multi-targeted μ/δ/κ opioid receptor (2019-2022). Bioorg Chem 2023; 141:106869. [PMID: 37797454 DOI: 10.1016/j.bioorg.2023.106869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
The opioids have been used for more than a thousand years and are not only the most widely prescribed drugs for moderate to severe pain and acute pain, but also the preferred drugs. However, their non-analgesic effects, especially respiratory depression and potential addiction, are important factors that plague the safety of clinical use and are an urgent problem for pharmacological researchers to address. Current research on analgesic drugs has evolved into different directions: de-opioidization; application of pharmacogenomics to individualize the use of opioids; development of new opioids with less adverse effects. The development of new opioid drugs remains a hot research topic, and with the in-depth study of opioid receptors and intracellular signal transduction mechanisms, new research ideas have been provided for the development of new opioid analgesics with less side effects and stronger analgesic effects. The development of novel opioid drugs in turn includes selective opioid receptor ligands, biased opioid receptor ligands, and multi-target opioid receptor ligands and positive allosteric modulators (PAMs) or antagonists and the single compound as multi-targeted agnoists/antagonists for different receptors. PAMs strategies are also getting newer and are the current research hotspots, including the BMS series of compounds and others, which are extensive and beyond the scope of this review. This review mainly focuses on the selective/biased/multi-targeted MOR/DOR/KOR (mu opioid receptor/delta opioid receptor/kappa opioid receptor) small molecule ligands and involves some cryo-electron microscopy (cryoEM) and structure-based approaches as well as the single compound as multi-targeted agnoists/antagonists for different receptors from 2019 to 2022, including discovery history, activities in vitro and vivo, and clinical studies, in an attempt to provide ideas for the development of novel opioid analgesics with fewer side effects.
Collapse
Affiliation(s)
- Ye He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qian Su
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lijuan Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Lu Yu
- Department of Respiratory Medicine, Sichuan Academy of Medical Sciences and Sichuan provincial People's Hospital, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
10
|
Spalinger M, Schwarzfischer M, Niechcial A, Atrott K, Laimbacher A, Jirkof P, Scharl M. Evaluation of the effect of tramadol, paracetamol and metamizole on the severity of experimental colitis. Lab Anim 2023; 57:529-540. [PMID: 36960681 DOI: 10.1177/00236772231163957] [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: 03/25/2023]
Abstract
Application of dextran sodium sulfate (DSS) is often used to induce experimental colitis. Current state of the art is to refrain from the use of analgesics due to their possible interaction with the model. However, the use of analgesics would be beneficial to reduce the overall constraint imposed on the animals. Here, we analyzed the effect of the analgesics Dafalgan (paracetamol), Tramal (tramadol) and Novalgin (metamizole) on DSS-induced colitis. To study the effect of those analgesics in colitis mouse models, acute and chronic colitis was induced in female C57BL6 mice by DSS administration in the drinking water. Analgesics were added to the drinking water on days four to seven (acute colitis) or on days six to nine of each DSS cycle (chronic colitis). Tramadol and paracetamol had minor effects on colitis severity. Tramadol reduced water uptake and activity levels slightly, while mice receiving paracetamol presented with a better overall appearance. Metamizole, however, significantly reduced water uptake, resulting in pronounced weight loss. In conclusion, our experiments show that tramadol and paracetamol are viable options for the use in DSS-induced colitis models. However, paracetamol seems to be slightly more favorable since it promoted the overall wellbeing of the animals upon DSS administration without interfering with typical readouts of colitis severity.
Collapse
Affiliation(s)
- Marianne Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Marlene Schwarzfischer
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Anna Niechcial
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Kirstin Atrott
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Andrea Laimbacher
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Paulin Jirkof
- Institute of Animal Welfare and 3R, University of Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Switzerland
| |
Collapse
|
11
|
Ghorab D, Abu-El-Rub EM, Gharaibeh MH, Yehya A, Khasawneh RR, Matalqah LM, Helaly AM. Neurological Effects of Combining Low Toxic Dose of Tramadol and Nicotine: An Animal Model Evidence of Endoplasmic Reticulum Stress. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1953356. [PMID: 37593523 PMCID: PMC10432102 DOI: 10.1155/2023/1953356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/19/2023]
Abstract
Tramadol abuse is a common problem in the Middle East in conjunction with smoking. The current study applied immunohistochemistry, western blot, real-time PCR, and ELISA to test the combination toxicity. Low toxic doses of tramadol induced animal brain cortex inflammation and hippocampus injury. Adding nicotine reverted hippocampus pathological changes without triggering marked brain injury. The expression of CHOP protein with real-time PCR showed mild endoplasmic reticulum stress (ER) in rat's brain. Histological, immunohistochemical, and western blotting analysis of CHOP (CCAAT-enhancer-binding protein homologous protein) and BIP (immunoglobulin heavy chain-binding protein) chaperones demonstrated endoplasmic reticulum stress in the brains of animals. Furthermore, the levels of apoptosis and autophagy markers demonstrated a mild reaction. The blood level of serotonin was high in all study groups, with a marked increase in the combined one. The high serotonin levels in the blood can be critical and associated with a high risk of serious withdrawal and pathological consequences. Serotonin receptor blockers such as olanzapine may increase systemic serotonin levels and need further investigation to utterly pinpoint their roles in managing mood disorders. In conclusion, the combination of tramadol and nicotine is less harmful than expected. However, serious withdrawal effects can occur as a result of high systemic serotonin effects.
Collapse
Affiliation(s)
- Doaa Ghorab
- Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Yarmouk, Irbid, Jordan
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ejlal M. Abu-El-Rub
- Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Yarmouk, Irbid, Jordan
| | - Mohamed Hamdi Gharaibeh
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary, Jordan University of Science and Technology, Irbid, Jordan
| | - Alaa Yehya
- Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Ramada R. Khasawneh
- Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Yarmouk, Irbid, Jordan
| | - Laila M. Matalqah
- Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Yarmouk, Irbid, Jordan
| | - Ahmed Mohamed Helaly
- Forensic Medicine and Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Clinical Sciences Department, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| |
Collapse
|
12
|
Vullo C, Falcone A, Marino G, Monti S, Tambella AM, Bufalari A, Catone G. Comparison between the effects of epidural lidocaine, tramadol, and lidocaine-tramadol on postoperative pain in cats undergoing elective orchiectomy. Acta Vet Scand 2023; 65:33. [PMID: 37434222 DOI: 10.1186/s13028-023-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 07/05/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND In veterinary clinical practice, orchiectomy is one of the most common surgical procedures for cats and is performed mainly in young animals. The purpose of this study was to compare three different epidural (EP) analgesic protocols used in cats undergoing orchiectomy in order to determine which protocol resulted in superior outcomes in terms of perioperative analgesia. Twenty-one client-owned male cats were premedicated with a combination of dexmedetomidine (10 µg/kg) and midazolam (0.2 mg/kg) injected intramuscularly. Anesthesia was induced intravenously with propofol. Cats were randomly divided in three treatment groups of seven animals each: Group L received EP lidocaine (2 mg/kg), Group T received EP tramadol (1 mg/kg), and Group LT received EP lidocaine (2 mg/kg) plus tramadol (1 mg/kg). The post-operative pain level was assessed using two different scales: the Glasgow Composite Measure Pain Scale-Feline (CMPS-F) and the Feline Grimace Scale (FGS). Rescue analgesia was administered when the CMPS-F total score was ≥5 or the FGS total score was ≥4. RESULTS No adverse effects related to tramadol or lidocaine were observed. Based on post-operative pain assessments, significant differences between groups were observed according to both pain scoring systems. In particular, in Group LT, the CMPS-F and FGS scores decreased significantly in the first six hours following castration. CONCLUSIONS Based on our results, EP lidocaine plus tramadol provided the best post-operative analgesic effects in cats submitted to orchiectomy lasting 6 h and could also be a choice to consider for longer surgical procedures.
Collapse
Affiliation(s)
- Cecilia Vullo
- Department of ChiBioFarAm, University of Messina, Viale Ferdinando Stagno d'Alcontres, Messina, 98166, Italy.
- Veterinary Teaching Hospital, University of Messina, Via Palatucci, Messina, 98168, Italy.
| | - Annastella Falcone
- Veterinary Teaching Hospital, University of Messina, Via Palatucci, Messina, 98168, Italy
| | - Gabriele Marino
- Veterinary Teaching Hospital, University of Messina, Via Palatucci, Messina, 98168, Italy
- Department of Veterinary Sciences, University of Messina, Via Palatucci, Messina, 98168, Italy
| | - Salvatore Monti
- Veterinary Teaching Hospital, University of Messina, Via Palatucci, Messina, 98168, Italy
- Department of Veterinary Sciences, University of Messina, Via Palatucci, Messina, 98168, Italy
| | - Adolfo Maria Tambella
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione, Matelica, 62022, Italy
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, Perugia, 06126, Italy
| | - Giuseppe Catone
- Veterinary Teaching Hospital, University of Messina, Via Palatucci, Messina, 98168, Italy
- Department of Veterinary Sciences, University of Messina, Via Palatucci, Messina, 98168, Italy
| |
Collapse
|
13
|
Michel MC, Cardozo L, Chermansky CJ, Cruz F, Igawa Y, Lee KS, Sahai A, Wein AJ, Andersson KE. Current and Emerging Pharmacological Targets and Treatments of Urinary Incontinence and Related Disorders. Pharmacol Rev 2023; 75:554-674. [PMID: 36918261 DOI: 10.1124/pharmrev.121.000523] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 03/16/2023] Open
Abstract
Overactive bladder syndrome with and without urinary incontinence and related conditions, signs, and disorders such as detrusor overactivity, neurogenic lower urinary tract dysfunction, underactive bladder, stress urinary incontinence, and nocturia are common in the general population and have a major impact on the quality of life of the affected patients and their partners. Based on the deliberations of the subcommittee on pharmacological treatments of the 7th International Consultation on Incontinence, we present a comprehensive review of established drug targets in the treatment of overactive bladder syndrome and the aforementioned related conditions and the approved drugs used in its treatment. Investigational drug targets and compounds are also reviewed. We conclude that, despite a range of available medical treatment options, a considerable medical need continues to exist. This is largely because the existing treatments are symptomatic and have limited efficacy and/or tolerability, which leads to poor long-term adherence. SIGNIFICANCE STATEMENT: Urinary incontinence and related disorders are prevalent in the general population. While many treatments have been approved, few patients stay on long-term treatment despite none of them being curative. This paper provides a comprehensive discussion of existing and emerging treatment options for various types of incontinence and related disorders.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Linda Cardozo
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Christopher J Chermansky
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Francisco Cruz
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Yasuhiko Igawa
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Kyu-Sung Lee
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Arun Sahai
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Alan J Wein
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Karl-Erik Andersson
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| |
Collapse
|
14
|
Zafar K, Wasim M, Fatima B, Hussain D, Mehmood R, Najam-Ul-Haq M. Quantification of tramadol and serotonin by cobalt nickel tungstate in real biological samples to evaluate the effect of analgesic drugs on neurotransmitters. Sci Rep 2023; 13:10239. [PMID: 37353529 PMCID: PMC10290146 DOI: 10.1038/s41598-023-37053-9] [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] [Received: 01/17/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023] Open
Abstract
In this work, CoNiWO4 nanocomposite was used as an electrochemical sensor for the simultaneous electrochemical detection of tramadol and serotonin. The nanocomposite was synthesized using a hydrothermal method and characterized via XRD, SEM, TGA, Zeta, UV, and FTIR. The sensor was developed by depositing CoNiWO4-NPs onto the glassy carbon electrode surface. Tramadol and serotonin were detected by employing cyclic voltammetry (CV), differential pulse voltammetry (DPV), electrochemical impedance spectroscopy (EIS), and chronoamperometry. Analytes were detected at different pH, concentrations, and scan rates. The prepared sensor showed a 0-60 µM linear range, with a LOD of 0.71 µM and 4.29 µM and LOQ of 14.3 µM and 2.3 µM for serotonin and tramadol, respectively. Finally, the modified electrode (CoNiWO4-GCE) was applied to determine tramadol and serotonin in biological samples.
Collapse
Affiliation(s)
- Komal Zafar
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muhammad Wasim
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Batool Fatima
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Dilshad Hussain
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | | | - Muhammad Najam-Ul-Haq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| |
Collapse
|
15
|
Influence of cerebral infarction on both bladder and urethral activities and changes after tramadol administration in rats. Neurourol Urodyn 2022; 41:1679-1691. [DOI: 10.1002/nau.25043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/07/2022]
|
16
|
Hosseindoost S, Akbarabadi A, Sadat-Shirazi MS, Mousavi SM, Khalifeh S, Mokri A, Hadjighassem M, Zarrindast MR. Effect of tramadol on apoptosis and synaptogenesis in hippocampal neurons: The possible role of µ-opioid receptor. Drug Dev Res 2022; 83:1425-1433. [PMID: 35808942 DOI: 10.1002/ddr.21973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/23/2022] [Accepted: 04/22/2022] [Indexed: 11/07/2022]
Abstract
Tramadol is a synthetic opioid with centrally acting analgesic activity that alleviates moderate to severe pain and treats withdrawal symptoms of the other opioids. Like other opioid drugs, tramadol abuse has adverse effects on central nervous system components. Chronic administration of tramadol induces maladaptive plasticity in brain structures responsible for cognitive function, such as the hippocampus. However, the mechanisms by which tramadol induces these alternations are not entirely understood. Here, we examine the effect of tramadol on apoptosis and synaptogenesis of hippocampal neuronal in vitro. First, the primary culture of hippocampal neurons from neonatal rats was established, and the purity of the neuronal cells was verified by immunofluorescent staining. To evaluate the effect of tramadol on neuronal cell viability MTT assay was carried out. The western blot analysis technique was performed for the assessment of apoptosis and synaptogenesis markers. Results show that chronic exposure to tramadol reduces cell viability of neuronal cells and naloxone reverses this effect. Also, the level of caspase-3 significantly increased in tramadol-exposed hippocampal neurons. Moreover, tramadol downregulates protein levels of synaptophysin and stathmin as synaptogenesis markers. Interestingly, the effects of tramadol were abrogated by naloxone treatment. These findings suggest that tramadol can induce neurotoxicity in hippocampal neuronal cells, and this effect was partly mediated through opioid receptors.
Collapse
Affiliation(s)
- Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Ardeshir Akbarabadi
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed M Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran
| | - Azarakhsh Mokri
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran.,Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Park S, Lee H, Kim JH, Jeon HL, Shin JY. Association between tramadol use and seizures: A nationwide case-case-time-control study. Pharmacoepidemiol Drug Saf 2022; 31:614-622. [PMID: 35141978 DOI: 10.1002/pds.5417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Tramadol may lower the seizure threshold; however, there is no conclusive evidence to confirm this. This study aimed to determine whether the use of tramadol is associated with the occurrence of seizures. METHODS We conducted a case-case-time-control (CCTC) study by identifying patients who had received tramadol and seizure diagnosis in a nationwide healthcare database in South Korea between 2003 and 2015. Each case was matched for age and sex to one future case to adjust for time trends in exposure without selection bias from the use of an external control group. The use of tramadol was assessed during a risk period of 1-30 days, and two reference periods, 61-90 days and 91-120 days, preceding the first diagnosis of seizures. We calculated the adjusted odds ratio (aOR) by dividing the OR in cases (case-crossover) by the OR in future cases (control-crossover). We performed a dose-response analysis using the average daily dose. RESULTS We identified 2,523 incident cases with matched future cases (mean age, 45.4 years; 50% men). The aOR for seizure with tramadol use was 0.94 (95% confidence interval [CI], 0.98-1.43) in the CCTC analysis, with a case-crossover OR of 1.19 (0.98-1.43) and control-crossover OR of 1.27 (1.03-1.56). The dose-response analysis showed a similar trend in the main analysis: a low-dose aOR of 0.80 (0.50-1.28) and a high-dose aOR of 0.92 (0.41-2.11). CONCLUSION We could not identify a significant association between transient use of tramadol and incidence of seizures in clinical practice.
Collapse
Affiliation(s)
- Sohee Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyesung Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ju Hwan Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ha-Lim Jeon
- School of Pharmacy, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Ju-Young Shin
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.,Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
18
|
Kim HC, Yoon DY, Lee S, Jang IJ, Hong JH, Kim J. Comparative pharmacokinetics between two tablets of tramadol 37.5 mg/acetaminophen 325 mg and one tablet of tramadol 75 mg/acetaminophen 650 mg for extended-release fixed-dose combination. Transl Clin Pharmacol 2022; 30:112-119. [PMID: 35800670 PMCID: PMC9253448 DOI: 10.12793/tcp.2022.30.e12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022] Open
Abstract
An extended-release (ER) fixed-dose combination (FDC) of tramadol 37.5 mg/acetaminophen 325 mg was developed due to the demand for varying dosages. This study aimed to evaluate the pharmacokinetics (PKs) for two tablets of the new developed tramadol 37.5 mg/acetaminophen 325 mg ER FDC (DW-0920, Wontran Semi ER®) as test formulation compared to one tablet of the tramadol 75 mg/acetaminophen 650 mg ER FDC (DW-0919, Wontran ER®) as reference formulation. A randomized, open-label, 2-way crossover study was conducted in 30 healthy subjects. Subjects were orally administered one of 2 formulations followed by an alternate formulation with a 7-day washout period. Blood samples were collected up to 36 hours post-dose. Plasma concentrations of tramadol and acetaminophen were determined using a validated high-performance liquid chromatography with tandem mass spectrometric method. The geometric mean ratios (GMRs) and their 90% confidence intervals (90% CIs) of test formulation to reference formulation were calculated for the maximum plasma concentration (Cmax) and the area under the plasma concentration-time curve from zero to the last measurable time point (AUClast). The PK profiles of 2 formulations were comparable. The GMRs (90% CI) of Cmax and AUClast for tramadol were 1.086 (1.047–1.127) and 1.008 (0.975–1.042), respectively. The corresponding values for acetaminophen were 0.956 (0.897–1.019) and 0.986 (0.961–1.011), respectively. All the values were within the bioequivalence range of 0.80–1.25. Two tablets of DW-0920 were comparable to one tablet of DW-0919. The DW-0920 may be used for optimal pharmacotherapy for pain control with a lower dose.
Collapse
Affiliation(s)
- Hyun Chul Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - Deok Yong Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - Jang Hee Hong
- Clinial Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 34134, Korea
| | - JaeWoo Kim
- H Plus Yangji Hospital, Seoul 08779, Korea
| |
Collapse
|
19
|
Yasufuku K, Koike K, Kobayashi M, Chiba H, Kitaura M, Takenouchi S, Hasegawa M, Morioka Y, Mishima H, Suzuki T, Fujita M. Involvement of the Peripheral μ-Opioid Receptor in Tramadol-Induced Constipation in Rodents. Biol Pharm Bull 2021; 44:1746-1751. [PMID: 34719650 DOI: 10.1248/bpb.b21-00474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tramadol is a weak opioid that produces analgesic effect via both the μ-opioid receptor (MOR) and non-opioid targets. Constipation is the most common opioid-related side effect in patients with cancer and non-cancer pain. However, the contribution of MOR to tramadol-induced constipation is unclear. Therefore, we used naldemedine, a peripherally acting MOR antagonist, and MOR-knockout mice to investigate the involvement of peripheral MOR in tramadol-induced constipation using a small intestinal transit model. A single dose of tramadol (3-100 mg/kg, per os (p.o.)) inhibited small intestinal transit dose-dependently in rats. Naldemedine (0.01-10 mg/kg, p.o.) blocked the inhibition of small intestinal transit induced by tramadol (30 mg/kg, p.o.) in rats. The transition rate increased dose-dependently over the range of naldemedine 0.01-0.3 mg/kg, and complete recovery was observed at 0.3-10 m/kg. Additionally, tramadol (30 and 100 mg/kg, subcutaneously (s.c.)) inhibited small intestinal transit in wild-type mice but not in MOR-knockout mice. These results suggest that peripheral MOR participates in tramadol-induced constipation.
Collapse
Affiliation(s)
- Kana Yasufuku
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd
| | - Katsumi Koike
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd
| | - Mika Kobayashi
- Research Area for Pharmacological Evaluation, Shionogi TechnoAdvance Research Co., Ltd
| | - Hiroki Chiba
- Research Area for Pharmacological Evaluation, Shionogi TechnoAdvance Research Co., Ltd
| | - Motoji Kitaura
- General Administration Division, Shionogi Administration Service Co., Ltd
| | - Shino Takenouchi
- Transgenic and Breeding Technology, Shionogi TechnoAdvance Research Co., Ltd
| | - Minoru Hasegawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd
| | | | | | - Tsutomu Suzuki
- Department of Pharmacology, School of Pharmacy, Shonan University of Medical Sciences
| | - Masahide Fujita
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd
| |
Collapse
|
20
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
21
|
Huang H, Li X, Xie P, Li X, Xu X, Qian Y, Yuan C, Meng X, Chai J, Chen J, Liu J, Wang W, Li W, Wang Y, Fu W, Liu J. Discovery, Structure-Activity Relationship, and Mechanistic Studies of 1-((3 R,4 S)-3-((Dimethylamino)methyl)-4-hydroxy-4-(3-methoxyphenyl)piperidin-1-yl)-2-(2,4,5-trifluorophenyl)ethan-1-one as a Novel Potent Analgesic. J Med Chem 2021; 64:9458-9483. [PMID: 34152138 DOI: 10.1021/acs.jmedchem.1c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Management of moderate to severe pain relies heavily on opioid analgesics such as morphine, oxycodone, and fentanyl in clinics. However, their prolonged use was associated with undesirable side effects. Many new strategies to reduce side effects have been proposed, but not without disadvantages. Using a hot plate model as a phenotypic screening method, our studies identified (3R,4S)-9d with a new scaffold as a potent analgesic with ED50 values of 0.54 mg/kg and 0.021 mg/kg in hot plate and antiwrithing models, respectively. Mechanistic studies showed that it elicited its analgesic effect via the active metabolite (3R,4S)-10a. The mechanism of (3R,4S)-10a-induced activation of the μ opioid receptor (MOR) was proposed by means of molecular dynamics (MD) simulation.
Collapse
Affiliation(s)
- Huoming Huang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xueping Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Peng Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xinwei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - XueJun Xu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuanyuan Qian
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Congmin Yuan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiangguo Meng
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - JingRui Chai
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Chen
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Wenli Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - YuJun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jinggen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
22
|
EEG and Sleep Effects of Tramadol Suggest Potential Antidepressant Effects with Different Mechanisms of Action. Pharmaceuticals (Basel) 2021; 14:ph14050431. [PMID: 34064349 PMCID: PMC8147808 DOI: 10.3390/ph14050431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/18/2023] Open
Abstract
Tramadol is a widely used, centrally acting, opioid analgesic compound, with additional inhibitory effects on the synaptic reuptake of serotonin and noradrenaline, as well as on the 5-HT2 and NMDA receptors. Preclinical and clinical evidence also suggests its therapeutic potential in the treatment of depression and anxiety. The effects of most widely used antidepressants on sleep and quantitative electroencephalogram (qEEG) are well characterized; however, such studies of tramadol are scarce. Our aim was to characterize the effects of tramadol on sleep architecture and qEEG in different sleep–wake stages. EEG-equipped Wistar rats were treated with tramadol (0, 5, 15 and 45 mg/kg) at the beginning of the passive phase, and EEG, electromyogram and motor activity were recorded. Tramadol dose-dependently reduced the time spent in rapid eye movement (REM) sleep and increased the REM onset latency. Lower doses of tramadol had wake-promoting effects in the first hours, while 45 mg/kg of tramadol promoted sleep first, but induced wakefulness thereafter. During non-REM sleep, tramadol (15 and 45 mg/kg) increased delta and decreased alpha power, while all doses increased gamma power. In conclusion, the sleep-related and qEEG effects of tramadol suggest antidepressant-like properties, including specific beneficial effects in selected patient groups, and raise the possibility of a faster acting antidepressant action.
Collapse
|
23
|
Abdalla HB, Napimoga MH, de Macedo Maganin AG, Lopes AH, Cunha TM, Gill HS, Clemente-Napimoga JT. The role of adenosine A 1 receptor in the peripheral tramadol's effect in the temporomandibular joint of rats. Int Immunopharmacol 2021; 97:107680. [PMID: 33932698 DOI: 10.1016/j.intimp.2021.107680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/20/2021] [Accepted: 04/12/2021] [Indexed: 01/06/2023]
Abstract
Peripheral tramadol's delivery in the temporomandibular joint (TMJ) leads to significant analgesic outcomes and inflammatory process's resolvent actions. Mechanistically, these properties are apart from the opioid system. Nevertheless, the molecular mechanisms behind these effects are still unclear. Therefore, the present study investigated the hypothesis that adenosine A1 receptors are involved in the tramadol-induced analgesic and anti-inflammatory effects in the TMJ. Animals were pretreated with an intra-TMJ injection of DPCPX (antagonist of A1 receptor) or tramadol and subsequent nociceptive challenge with an intra-TMJ injection of 1.5% formalin. For over 45 min, the nociceptive behavior was quantitated, and by the end of this assessment, the animals were euthanized, and the periarticular tissue was collected. Lastly, an in vitro assay of BMDM (Bone Marrow-Derived Macrophages) was performed to investigate tramadol activity in macrophages. The intra-TMJ injection of tramadol ameliorates formalin-induced hypernociception along with inhibiting leukocyte migration. The tramadol's peripheral anti-inflammatory effect was mediated by the adenosine A1 receptor and was associated with increased protein expression of α2a-adrenoceptor in the periarticular tissues (p < 0.05: ANOVA, Tukey's test). Also, tramadol inhibits formalin-induced leukocyte migration and protein expression of P2X7 receptors in the periarticular tissue (p < 0.05); however, DPCPX did not alter this effect (p > 0.05). Moreover, DPCPX significantly reduced the protein expression of the M2 macrophage marker, MRC1. In BMDM, tramadol significantly reduces inflammatory cytokines release, and DPCPX abrogated this effect (p < 0.05). We identify tramadol's peripheral effect is mediated by adenosine A1 receptor, possibly expressed in macrophages in the TMJ tissue. We also determined an important discovery related to the activation of A1R/α2a receptors in the tramadol action.
Collapse
Affiliation(s)
- Henrique Ballassini Abdalla
- Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil; Laboratory of Neuroimmune Interface of Pain Research, Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Campinas, SP, Brazil
| | - Marcelo Henrique Napimoga
- Laboratory of Neuroimmune Interface of Pain Research, Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Campinas, SP, Brazil
| | - Alexandre Gomes de Macedo Maganin
- Center for Research in Inflammatory Diseases, Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Hashimoto Lopes
- Center for Research in Inflammatory Diseases, Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Juliana Trindade Clemente-Napimoga
- Laboratory of Neuroimmune Interface of Pain Research, Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Campinas, SP, Brazil.
| |
Collapse
|
24
|
Lelescu CA, Dumitras DA, Iurian S, Staffieri F, Muresan C. Effects of topical application of tramadol with/without dexmedetomidine and proparacaine on corneal sensitivity in rats. Int Ophthalmol 2021; 41:465-473. [PMID: 33095346 DOI: 10.1007/s10792-020-01596-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/01/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To evaluate the corneal anesthetic effect following topical application of tramadol alone and in combination with dexmedetomidine, and compare it to proparacaine, in clinically healthy rats. METHODS A randomized, crossover study was performed. Twenty Wistar albino rats (n = 40 eyes) were used. Corneal touch threshold (CTT) measurements (in mm) were obtained using a Cochet-Bonnet aesthesiometer. CTT measurements were obtained at baseline, 1-min following application of the topical anesthetic agent, and repeated at 5-min intervals up to 75 min. The topical protocol involved 3 treatment conditions, separated by a 2-week washout period: proparacaine, tramadol alone, and tramadol in combination with dexmedetomidine. RESULTS CTT values were significantly decreased compared to baseline at each timepoint until completion of the 75-min evaluation in all treated eyes, regardless of the assigned treatment (p < 0.0083). With tramadol, complete corneal anesthesia (CTT = 0) was achieved within 1-5 min in 18 eyes and ranged from 5 to 25 min. Co-administration of dexmedetomidine to tramadol resulted in significantly increased CTT values from 5 to 20 min following topical application, compared to tramadol alone (p < 0.0083), and complete corneal anesthesia was achieved in only 14 out of 20 treated eyes. CONCLUSION Tramadol might be a useful alternative to topical anesthetic agents, providing a dose-related corneal anesthetic effect. Co-administration of dexmedetomidine does not potentiate its anesthetic effect. The underlying mechanism(s) of drug antagonism between tramadol and dexmedetomidine remains to be determined.
Collapse
Affiliation(s)
- Cristina A Lelescu
- Modis Competence Center, Modis Life Sciences Belgium, Mureșului 9, 400000, Cluj-Napoca, Romania
| | - Daria A Dumitras
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania
| | - Sonia Iurian
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hațieganu", V. Babeș 41, 400012, Cluj-Napoca, Romania
| | - Francesco Staffieri
- Division of Veterinary Surgery, Department of Emergency and Organ Transplantation, Faculty of Veterinary Medicine, University "Aldo Moro" of Bari, SP Per Casamassima Km. 3, 70010, Valenzano, Bari, Italy
| | - Cosmin Muresan
- Department of Surgery, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania.
| |
Collapse
|
25
|
Huang H, Li X, Guo W, Zhu C, Qian Y, Shen Q, Xu X, Li W, Wang Y, Fu W. Design, Synthesis, and Structure-Activity Relationship Exploration of Alkyl/Phenylalkyl Piperidine Analogues as Novel Highly Potent and Selective μ Opioid Receptor Agonists. ACS Chem Neurosci 2021; 12:285-299. [PMID: 32852933 DOI: 10.1021/acschemneuro.0c00487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pain was implicated in many diseases. Despite effectiveness to treat moderate to severe pain, opioid analgesics elicited many side effects, greatly limiting their prescription in clinics. Based on M1, an active metabolite of tramadol, 3-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-4-ol analogues were designed, synthesized, and evaluated in vitro. Among all the compounds tested, compound 23 was found to be a novel, highly selective, and potent MOR agonist (Ki MOR = 0.0034 nM, EC50 MOR = 0.68 nM, Emax = 206.5%; Ki DOR = 41.67 nM; Ki KOR = 7.9 nM). Structure-activity relationship exploration showed that the linker between the piperidine ring and the phenyl ring as well as substituent pattern of the phenyl ring played a pivotal role in binding affinity and selectivity. (3R, 4S)-23 (Ki MOR = 0.0021 ± 0.0001 nM, EC50 MOR = 0.0013 ± 0.0001 nM, Emax = 209.1 ± 1.4%; Ki DOR = 18.4 ± 0.7 nM, EC50 DOR = 74.5 ± 2.8 nM, Emax = 267.1 ± 1.4%; Ki KOR = 25.8 ± 0.2 nM, EC50 DOR = 116.2 ± 4.4 nM, Emax = 209.5 ± 1.4%) had more potent activity for opioid receptors than its enantiomer (3S, 4R)-23 and was found to be a potent, highly selective MOR agonist with novel scaffold. High binding affinity and selectivity of (3R, 4S)-23 for MOR over KOR and DOR and its mechanism of activating MOR were proposed by docking and molecular dynamics simulations, respectively.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuejun Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | | | - Yujun Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | | |
Collapse
|
26
|
Kamin HS, Bhatt SS, Mulligan CJ, Kertes DA. Dehydroepiandrosterone at birth: Response to stress and relation to demographic, pregnancy and delivery factors. J Neuroendocrinol 2020; 32:e12906. [PMID: 33006172 DOI: 10.1111/jne.12906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 11/27/2022]
Abstract
Enhanced production of dehydroepiandrosterone (DHEA) by the foetal hypothalamic-pituitary-adrenal (HPA) axis enables maturational events critical for labour induction and neonatal adaptation. Despite knowledge of the interconnected nature of maternal and foetal physiology and dramatic changes in DHEA production after birth, few studies have examined DHEA levels in newborns and none have examined DHEA's response to acute stress. Understanding normative patterns of early DHEA activity is needed to accurately assess functioning of the biological stress system with relevance for health and development. The present study analysed DHEA concentrations and change after stress among 93 newborns and associations with pregnancy, delivery and demographic risk factors. Three saliva samples, collected prior to and following a blood draw stressor, were used to determine baseline and stress reactive DHEA levels. Mothers self-reported on health behaviours during pregnancy. Data on obstetric factors were obtained from medical records. DHEA levels declined from pre- to post-stressor assessments. Results also showed that post-stressor DHEA change was significantly associated with administration of medications used to treat pain and accelerate labour. However, there was no significant variation in DHEA pre-stress levels or change after stress as a function of time after birth. By capturing DHEA levels after birth, the present study provides a window into prenatal health of the HPA system. The study also advances knowledge of DHEA in newborns by providing data on reference levels and important covariates. This information on basic adrenal physiology provides a foundation that can be expanded on to enhance understanding of early hypothalamic-pituitary-adrenal axis activity.
Collapse
Affiliation(s)
- Hayley S Kamin
- Department of Psychology, University of Florida, Gainesville, FL, USA
| | - Samarth S Bhatt
- Department of Psychology, University of Florida, Gainesville, FL, USA
| | - Connie J Mulligan
- Department of Anthropology, University of Florida, Gainesville, FL, USA
- Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Darlene A Kertes
- Department of Psychology, University of Florida, Gainesville, FL, USA
- Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
27
|
Affiliation(s)
- Samuel A. Krug
- Forensic Science Arcadia University Glenside Pennsylvania USA
| | - Karen S. Scott
- Forensic Science Arcadia University Glenside Pennsylvania USA
| |
Collapse
|
28
|
Holmstedt A, Olsson M, Håkansson A. Clinical characteristics distinguishing tramadol-using adolescents from other substance-using adolescents in an out-patient treatment setting. Addict Behav Rep 2020; 11:100272. [PMID: 32322659 PMCID: PMC7160423 DOI: 10.1016/j.abrep.2020.100272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Non-medical Prescription Opioid Use (NMPOU) has increased worldwide during the last decades, and specifically, tramadol misuse may represent a novel pattern of substance use among adolescents. The present study aims to analyze characteristics distinguishing tramadol-using adolescents from other substance-using adolescents seeking out-patient treatment. METHODS This is a cross-sectional study of treatment-seeking patients between 13 and 24 years of age in an out-patient facility for substance use problems in Malmö, Sweden. A total of 526 treatment-seeking adolescents at an out-patient treatment center were included. Data on substance use, treatment history and socio-demographic variables were extracted through a semi-structured interview method aimed specifically for adolescents with alcohol or drug problems (Ung-DOK). Lifetime tramadol users were compared to non-users, and also, primary tramadol users were compared to remaining subjects. RESULTS Thirty-one percent (n = 162) were tramadol users (lifetime prevalence). In logistic regression, the tramadol group showed a significantly increased risk of tobacco use, problematic lifetime cocaine, benzodiazepine and amphetamine use, and were more likely to report contacts with the judicial system, and less likely to report contacts with child or adult psychiatry, and more likely to have parents born outside the Scandinavian countries. In logistic regression, primary tramadol use was negatively associated with frequent cannabis use. CONCLUSIONS Tramadol use appears to be a novel pattern among treatment-seeking adolescents. They showed a significantly increased risk of initiation of other illicit drugs and criminal behaviour, despite less contact with psychiatric care. More attention may be needed to this relatively novel pattern of opioid use.
Collapse
Affiliation(s)
- A. Holmstedt
- Lund University, Faculty of Medicine, Dept of Clinical Sciences Lund, Psychiatry, Lund, Sweden
- Malmö Addiction Center, Region Skåne, Malmö, Sweden
| | - M.O. Olsson
- Stockholm Center for Dependency Disorders, Region Stockholm, Centre for Psychiatric Research, Karolinska Institute, Stockholm, Sweden
| | - A. Håkansson
- Lund University, Faculty of Medicine, Dept of Clinical Sciences Lund, Psychiatry, Lund, Sweden
- Malmö Addiction Center, Region Skåne, Malmö, Sweden
| |
Collapse
|
29
|
Zafar MI. Suitability of APINCH high-risk medications use in diabetes mellitus. Eur J Pharmacol 2020; 867:172845. [DOI: 10.1016/j.ejphar.2019.172845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
|
30
|
Tramadol use is associated with enhanced postoperative outcomes in breast cancer patients: a retrospective clinical study with in vitro confirmation. Br J Anaesth 2019; 123:865-876. [DOI: 10.1016/j.bja.2019.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022] Open
|
31
|
Lucena F, Callado de Oliveira DMM, Norões MM, Mujica EMM, Melleu FF, Benedet PDO, Stein T, Ribeiro LFC, Tonussi CR. Intrathecally injected tramadol reduces articular incapacitation and edema in a rat model of lipopolysaccharide (LPS)-induced reactive arthritis. Life Sci 2019; 236:116860. [PMID: 31518605 DOI: 10.1016/j.lfs.2019.116860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/29/2019] [Accepted: 09/08/2019] [Indexed: 10/26/2022]
Abstract
AIMS Intrathecal injection of morphine presents analgesic and antiedematogenic effects in rats. However, it is unknown whether tramadol, which possess a mixed mechanism of action, can also produce analgesic and antiedematogenic effects similarly. MAIN METHODS Male Wistar rats received carrageenan and LPS in the right knee joint. Tramadol (10 μg) was injected intrathecally 20 min before articular LPS injection. Incapacitation and articular edema were measured 5 h after LPS stimulation. Synovial fluid was collected for leukocyte counting and western blot analysis. Whole joint and lumbar spinal cord were also collected for histology and immunohistochemistry, respectively. Intrathecal pretreatments groups were with the NKCC1 blocker bumetanide, TRPV1 agonist resiniferatoxin, μ-opioid receptor antagonist CTOP and serotonergic neurotoxin 5,7-DHT, all previously to tramadol. KEY FINDINGS Tramadol treatment caused the reduction of incapacitation and edema. It also reduced c-Fos protein expression in the spinal cord dorsal horn and slightly reduced TNF-α levels in synovial fluid, but neither reduced cell migration nor tissue damage. Bumetanide and resiniferatoxin prevented the analgesic and antiedematogenic effects of tramadol. CTOP prevented the analgesic and the antiedematogenic effects, but 5,7-DHT prevented only tramadol-induced analgesia. SIGNIFICANCE Spinal NKCC1 cotransporter and peptidergic peripheral afferents seem to be important for the analgesic and antiedematogenic effects of tramadol, as well as μ-opioid receptor. However, the monoamine uptake inhibition effect of tramadol seems to be important only to the analgesic effect.
Collapse
Affiliation(s)
- Flora Lucena
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | | | - Maíra M Norões
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Elba M M Mujica
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Fernando F Melleu
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Patrícia de O Benedet
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Taciane Stein
- Program in Biosciences and Health, State University of Western Parana, Cascavel, PR 85819-110, Brazil
| | - Lucineia F C Ribeiro
- Program in Biosciences and Health, State University of Western Parana, Cascavel, PR 85819-110, Brazil
| | - Carlos R Tonussi
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil.
| |
Collapse
|
32
|
Abstract
On October 26, 2017, US president, Donald J. Trump, declared the opioid epidemic a national public health emergency. This declaration opened the door to government funding for programs geared toward studying and treating opioid addiction; however, part of the responsibility to slow the epidemic falls on physicians. Orthopaedic surgeons prescribed the third-highest number of narcotic prescriptions among all physician groups, and therefore, they have a major role to play in decreasing the use of opioids. Although restricting prescriptions is part of the solution, orthopaedic surgeries are often painful, and opioid medications, along with other multimodal nonopioid medications, allow patients to control pain and improve function, especially in the short term. Therefore, to successfully manage pain and opioid prescriptions, orthopaedic surgeons should know the basic science behind these powerful medications. Understanding the basic mechanism of action of opioid and nonopioid medication classes, knowing what oral morphine equivalent are, and knowing which nonopioid drugs can amplify or weaken the potency of opioids will help orthopaedic surgeons become more informed prescribers and leaders in helping control the opioid epidemic.
Collapse
|
33
|
Samadi M, Shaki F, Bameri B, Fallah M, Ahangar N, Mohammadi H. Caffeine attenuates seizure and brain mitochondrial disruption induced by Tramadol: the role of adenosinergic pathway. Drug Chem Toxicol 2019; 44:613-619. [PMID: 31368376 DOI: 10.1080/01480545.2019.1643874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Tramadol (TR) is an analgesic drug used to treat moderate-to-severe pain but it induces seizure even at therapeutic doses. The exact mechanism of TR-inducing seizure is not clear but inhibition of the serotonin, GABA, and nitrous oxide (NOS) pathways are the commonly proposed mechanisms. Adenosinergic system has a crucial function in the modulation of seizure. Also, oxidative damage is an unavoidable effect of the seizure. This study was conducted to evaluate the role of the adenosinergic system on the seizure and oxidative stress biomarkers induced by TR using antagonist of the adenosinergic receptors in the Albino mice. For that purpose, generated clonic seizure, as seizure threshold, was evaluated by TR. Caffeine (CAF; 8 mg/kg, i.p.), a nonselective antagonist of adenosine receptors, was administered 1 hour before the seizure induction. The seizure threshold significantly increased by CAF-treated group when compared to TR group (p < 0.001). Oxidative stress biomarkers such as reactive oxygen species, protein carbonyl content, and lipid peroxidation significantly decreased and glutathione content significantly increased by CAF in brain mitochondria compared to the TR group, whereas oxidative biomarkers significantly increased in the TR group compared to the control group. The results of the present study suggested that the adenosinergic system is involved in seizure induced by TR and meanwhile, inhibition of adenosine receptors can decrease the TR seizure threshold and also decrease the induced oxidative damage in the brain mitochondria.
Collapse
Affiliation(s)
- Mahedeh Samadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Shaki
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.,Pharmacutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Behnaz Bameri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Marjan Fallah
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nematollah Ahangar
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.,Pharmacutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamidreza Mohammadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.,Pharmacutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
34
|
Addition of 100 mg of Tramadol to 40 mL of 0.5% Ropivacaine for Interscalene Brachial Plexus Block Improves Postoperative Analgesia in Patients Undergoing Shoulder Surgeries as Compared to Ropivacaine Alone-A Randomized Controlled Study. ACTA ACUST UNITED AC 2019; 55:medicina55070399. [PMID: 31340565 PMCID: PMC6681507 DOI: 10.3390/medicina55070399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
Background and objectives: Brachial plexus block is commonly used in shoulder surgery, as it provides satisfactory surgical conditions and adequate postoperative pain control. However, there are contradictory reports regarding the addition of tramadol to the injected regional anesthetic solution. We performed a prospective randomized study to evaluate the effectiveness of tramadol as an adjuvant to ropivacaine during interscalene brachial plexus block and assess its impact on the opioid consumption and the early postoperative pain in patients that underwent shoulder surgery. Materials and Methods: Eighty patients scheduled for elective shoulder surgery and anesthesia via interscalene brachial plexus block were randomly divided into two groups. In group A (n = 40), a solution of 40 mL of ropivacaine 0.5% and 2 mL (100 mg) of tramadol was administered during the brachial plexus block, while in group B (n = 40), a solution of 40 mL of ropivacaine 0.5% and 2 mL NaCl 0.9% (placebo) was administered. The effectiveness and duration of sensory and motor blocks were recorded in both groups. The sensory block was assessed recording the loss of sensation to pin prick test over the skin distribution of the axillary, radial, and musculocutaneous nerves. The motor block was assessed using the modified 3-point Bromage score (0–2 points). Cumulative morphine consumption and pain, using the Visual Analog Scale (VAS), were evaluated in both groups at 2, 4, 8, and 24 h after surgery. Results: Sensory block onset was achieved earlier in group A than in group B (5.21 ± 3.15 minutes (min) vs. 7.1 ± 4.51 min, p = 0.029). The motor block onset was similar between the two groups (13.08 ± 6.23 min vs. 13.28 ± 6.59 min; p = 0.932). The duration of the sensory block was longer in group A as compared to group B (13 ± 2.3 h vs. 12 ± 2.8 h; p = 0.013). The duration of the motor block did not present any difference between the groups (10 ± 2.2 h vs. 10 ± 2.8 h; p = 0.308). Differences in morphine administration were not significant at 2, 4, and 8 h, however, morphine consumption was found to be decreased in group A 24 h postoperatively A (p = 0.04). The values of VAS were similar at 2, 4, and 8 h, however, they were lower in group A at 24 h (p < 0.013). Conclusions: Combined regional administration of tramadol and ropivacaine during interscalene brachial plexus block improves the time of onset and the duration of the sensory block, while it is associated with reduced morphine consumption during the first 24 h after shoulder surgery.
Collapse
|
35
|
Monteiro BP, Lambert C, Bianchi E, Genevois JP, Soldani G, Troncy E. Safety and efficacy of reduced dosage ketoprofen with or without tramadol for long-term treatment of osteoarthritis in dogs: a randomized clinical trial. BMC Vet Res 2019; 15:213. [PMID: 31238912 PMCID: PMC6591828 DOI: 10.1186/s12917-019-1960-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 06/13/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the safety and efficacy of reduced-dosage ketoprofen with or without tramadol in dogs. Five healthy dogs receiving standard-dosage ketoprofen (2 mg/kg SC, then 1 mg/kg PO daily) comprised Group A. Twenty dogs with osteoarthritis were randomized to receive reduced-dosage ketoprofen (0.5 mg/kg SC once; 0.25 mg/kg PO daily) alone (Group B) or in combination with tramadol (5 mg/kg/day PO) (Group C). Treatments were administered for 28 days. Platelet aggregation time (PAT), gastrointestinal (GI) endoscopy and glomerular filtration rate (GFR) were performed up to 60 days after treatment initiation. Pain was scored using a validated clinical metrology instrument up to D120. Data were analyzed with general linear mixed model for repeated measures (α = 0.05). RESULTS PAT was not different between groups but was increased with time for all groups. GI lesion scores were higher in Group A than Groups B and C (day 28; P = 0.005) and were increased with time for Group A (P = 0.005). GFR was lower in Group A than Groups B and C (day 28; P < 0.01) and were decreased with time for group A (P < 0.001). Standard-dosage ketoprofen administration resulted in clinically relevant adverse effects. Pain score decreased in both treated groups (B and C) from D0 to D28. Need of rescue analgesia from D29 to D120 was higher in Group B than in Group C (P = 0.039). CONCLUSIONS The long-term safety profile of reduced-dosage ketoprofen is similar whether the drug is administered alone or in combination with tramadol to dogs with osteoarthritis. Analgesic efficacy of the combination looks attractive.
Collapse
Affiliation(s)
- Beatriz P Monteiro
- Department of Biomedical Sciences, GREPAQ (Groupe de recherche en pharmacologie animale du Québec), Faculty of Veterinary Medicine - Université de Montréal, 3200 Sicotte Street, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Cedric Lambert
- Department of Companion Animals, Vet-Agro Sup, Lyon, France
| | - Elena Bianchi
- Department of Pharmacology, College of Veterinary Medicine, University of Pisa, Pisa, Italy
| | | | - Giulio Soldani
- Department of Pharmacology, College of Veterinary Medicine, University of Pisa, Pisa, Italy
| | - Eric Troncy
- Department of Biomedical Sciences, GREPAQ (Groupe de recherche en pharmacologie animale du Québec), Faculty of Veterinary Medicine - Université de Montréal, 3200 Sicotte Street, Saint-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
36
|
Koga A, Fujita T, Piao LH, Nakatsuka T, Kumamoto E. Inhibition by O-desmethyltramadol of glutamatergic excitatory transmission in adult rat spinal substantia gelatinosa neurons. Mol Pain 2019; 15:1744806918824243. [PMID: 30799694 PMCID: PMC6348506 DOI: 10.1177/1744806918824243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
To reveal cellular mechanisms for antinociception produced by clinically used tramadol, we investigated the effect of its metabolite O-desmethyltramadol (M1) on glutamatergic excitatory transmission in spinal dorsal horn lamina II (substantia gelatinosa; SG) neurons. The whole-cell patch-clamp technique was applied at a holding potential of −70 mV to SG neurons of an adult rat spinal cord slice with an attached dorsal root. Under the condition where a postsynaptic action of M1 was inhibited, M1 superfused for 2 min reduced the frequency of spontaneous excitatory postsynaptic current in a manner sensitive to a μ-opioid receptor antagonist CTAP; its amplitude and also a response of SG neurons to bath-applied AMPA were hardly affected. The presynaptic effect of M1 was different from that of noradrenaline or serotonin which was examined in the same neuron. M1 also reduced by almost the same extent the peak amplitudes of monosynaptic primary-afferent Aδ-fiber and C-fiber excitatory postsynaptic currents evoked by stimulating the dorsal root. These actions of M1 persisted for >10 min after its washout. These results indicate that M1 inhibits the quantal release of L-glutamate from nerve terminals by activating μ-opioid but not noradrenaline and serotonin receptors; this inhibition is comparable in extent between monosynaptic primary-afferent Aδ-fiber and C-fiber transmissions. Considering that the SG plays a pivotal role in regulating nociceptive transmission, the present findings could contribute to at least a part of the inhibitory action of tramadol on nociceptive transmission together with its hyperpolarizing effect as reported previously.
Collapse
Affiliation(s)
- Akiko Koga
- 1 Department of Physiology, Saga Medical School, Saga, Japan.,2 Department of Anesthesiology & Critical Care Medicine, Saga Medical School, Saga, Japan
| | - Tsugumi Fujita
- 1 Department of Physiology, Saga Medical School, Saga, Japan
| | - Lian-Hua Piao
- 1 Department of Physiology, Saga Medical School, Saga, Japan
| | | | - Eiichi Kumamoto
- 1 Department of Physiology, Saga Medical School, Saga, Japan
| |
Collapse
|
37
|
Khodayari S, Ghaderi Pakdel F, Shahabi P, Naderi S. Acute Tramadol-Induced Cellular Tolerance and Dependence of Ventral Tegmental Area Dopaminergic Neurons: An In Vivo Electrophysiological Study. Basic Clin Neurosci 2019; 10:209-224. [PMID: 31462976 PMCID: PMC6712631 DOI: 10.32598/bcn.9.10.180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/25/2017] [Accepted: 04/30/2018] [Indexed: 01/28/2023] Open
Abstract
Introduction Ventral Tegmental Area (VTA) is a core region of the brainstem that contributes to different vital bio-responses such as pain and addiction. The Dopaminergic (DA) cellular content of VTA has major roles in different functions. This study aims to evaluate the cellular effect of tramadol on the putative VTA-DA neurons. Methods Wistar rats were assigned into three groups of control, sham, and tramadol-treated. The animals were anesthetized and their VTA-DA neuronal activity was obtained under controlled stereotaxic operation. The firing rate of the neurons was extracted according to principal component analysis by Igor Pro software and analyzed statistically considering P<0.05 as significant. Tramadol (20 mg/kg) was infused intraperitoneally. Results Overall, 121 putative VTA-DA neurons were isolated from all groups. In tramadol-treated rats, the inhibition of the neuronal firing was proposed as tolerance and the excitation period as dependence or withdrawal. The Mean±SD inhibition time lasted up to 50.34±10.17 minutes and 31% of neurons stopped firing and silenced after 24±3 min on average but the remaining neurons lowered their firing up to 43% to 67% of their baseline firing. All neurons showed the excitation period, lasted about 56.12±15.30 min, and the firing of neurons increased from 176% to 244% of their baseline or pre-injection period. Conclusion The tolerance and dependence effects of tramadol are related to the changes in the neuronal firing rate at the putative VTA-DA neurons. The acute injection of tramadol can initiate neuroadaptation on the opioid and non-opioid neurotransmission to mediate these effects.
Collapse
Affiliation(s)
- Shabnam Khodayari
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Firouz Ghaderi Pakdel
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parviz Shahabi
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayyeh Naderi
- Danesh Pey Hadi Co., Health Technology Incubator Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
38
|
Abstract
OBJECTIVE Pain management is unsatisfactory worldwide, particularly in developing countries where access to opioids is restricted and cost is an issue. It is a major concern in West Africa and we therefore undertook a survey of pain experts to obtain a better understanding of the problems in the region. METHODS Medical practitioners involved in pain management, identified via professional networks, were interviewed to share their experience in treating moderate-to-severe pain in West Africa. The questionnaire was based on an existing version modified to meet African conditions. Additionally, informal focus group meetings with palliative care physicians and pharmacists were conducted. RESULTS A total of 11 questionnaires were returned. All respondents were physicians who reported availability of opioids in their clinics, but access to morphine was challenging and not possible in some rural settings. Obligatory maintenance of detailed records was considered a burden. The main concern raised was the risk of misuse/addiction. Seven of 11 respondents reported that they would use tramadol as an alternative to strong opioids if required and, interestingly, 9 of 11 considered it to be an essential medicine. Based on personal experience, the respondents noted that some properties of tramadol make it a key pain treatment option in West Africa (strong/fast-acting, better tolerated than NSAIDs, less addictive than other opioids and relatively inexpensive). CONCLUSIONS Most stakeholders who completed the survey indicated that tramadol was essential to provide optimal pain management in the absence of access to strong opioids.
Collapse
Affiliation(s)
- Ernest Yorke
- a Department of Medicine & Therapeutics School of Medicine & Dentistry , College of Health Sciences, University of Ghana , Accra , Ghana
| | - Folaju O Oyebola
- b Pain and Palliative Medicine Department , Federal Medical Centre , Abeokuta , Nigeria
| | - Samuel Anaja Otene
- c Department of Radiotherapy and Oncology , Ahmadu Bello University Teaching Hospital , Zaria , Nigeria
| | - Axel Klein
- d Global Drug Policy Observatory , University of Swansea , Swansea , UK
| |
Collapse
|
39
|
Hong GS, Stein K, Lysson M, Kalff J, Wehner S. A comparative study about the immunomodulatory effects of tramadol and metamizole in a murine model of postoperative ileus. Lab Anim 2019; 53:610-618. [PMID: 30907232 DOI: 10.1177/0023677219832919] [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] [Indexed: 11/15/2022]
Abstract
Postoperative ileus (POI) is a common complication after abdominal surgery characterized by motility disturbances leading to increased morbidity and mortality in surgical patients. Intestinal manipulation of the murine small bowel is an established animal model resulting in an increased postsurgical inflammation within the intestinal muscular externa and a delayed gastrointestinal transit. Some analgesics have been shown to affect inflammation. In this study, we compared the immunomodulatory effects of two different analgesics. Mice were treated with tramadol, metamizole or saline as a control in our established POI model. The postoperative inflammatory response was assessed by gene expression of pro-inflammatory cytokines at different time points and immunocytes extravasation into the muscularis externa. Functional motility analyses were performed by a gastrointestinal transit measurement. Metamizole application reduced the pro-inflammatory response after surgery and improved gastrointestinal motility, while tramadol showed no alteration in cytokine gene expression, influx of immunocytes and gastrointestinal transit compared with the controls. In conclusion. we suggest tramadol as analgesia in immunological studies on POI in mice as it does not affect the underlying inflammation of POI.
Collapse
Affiliation(s)
- Gun-Soo Hong
- Department of Surgery, University of Bonn, Bonn Germany
| | - Kathy Stein
- Department of Surgery, University of Bonn, Bonn Germany
| | | | - Joerg Kalff
- Department of Surgery, University of Bonn, Bonn Germany
| | - Sven Wehner
- Department of Surgery, University of Bonn, Bonn Germany
| |
Collapse
|
40
|
McGarry K, Graham Y, McDonald S, Rashid A. RESKO: Repositioning drugs by using side effects and knowledge from ontologies. Knowl Based Syst 2018. [DOI: 10.1016/j.knosys.2018.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
41
|
Asari Y, Ikeda Y, Tateno A, Okubo Y, Iijima T, Suzuki H. Acute tramadol enhances brain activity associated with reward anticipation in the nucleus accumbens. Psychopharmacology (Berl) 2018; 235:2631-2642. [PMID: 29951769 DOI: 10.1007/s00213-018-4955-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/19/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tramadol is an analgesic with monoamine reuptake inhibition and μ-opioid receptor activation. Although tramadol has been widely used for treatment of various pain conditions, there is controversy over the risk of abuse potential. We examined the effects of tramadol on the reward system in humans using functional magnetic resonance imaging (fMRI) to assess the potential of tramadol for drug abuse or dependence. METHODS A randomized, double-blind, placebo-controlled, crossover study was conducted for 19 healthy adults under tramadol or placebo. In association with subjective mood questionnaires, monetary incentive delay (MID) task was performed to assess the neural response to reward anticipation during fMRI. Subjective mood measures and blood oxygenation level-dependent (BOLD) signal during gain and loss anticipation were compared between tramadol and placebo. RESULTS Tramadol significantly reduced anxiety (Z = - 2.513, p = 0.012) and enhanced vigor (Z = - 2.725, p = 0.006) compared with placebo. By Mood Rating Scale, tramadol provoked contented (Z = - 2.316, p = 0.021), relaxed (Z = - 2.236, p = 0.025), and amicable feelings (Z = - 2.015, p = 0.044) as well as increased alertness (Z = - 1.972, p = 0.049) and contentedness domains (Z = - 2.174, p = 0.030) compared with placebo. Several brain regions including nucleus accumbens (NAc) were activated during gain anticipation in the MID task under both tramadol and placebo. Tramadol increased the %BOLD signal change in NAc at +¥500 cue significantly more than the placebo (Z = - 2.295, p = 0.022). CONCLUSION Tramadol enhances the reward system and thereby may have abuse potential or precipitate drug abuse in human.
Collapse
Affiliation(s)
- Yuki Asari
- Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Yumiko Ikeda
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Amane Tateno
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Yoshiro Okubo
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Takehiko Iijima
- Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
42
|
Miranda HF, Sierralta F, Aranda N, Poblete P, Noriega V, Prieto JC. Synergism between gabapentin-tramadol in experimental diabetic neuropathic pain. Fundam Clin Pharmacol 2018; 32:581-588. [DOI: 10.1111/fcp.12400] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 06/10/2018] [Accepted: 07/04/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Hugo F. Miranda
- Department of Neuroscience; Faculty of Medicine; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| | - Fernando Sierralta
- ICBM; Pharmacology Program; Faculty of Medicine; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| | - Nicolas Aranda
- ICBM; Pharmacology Program; Faculty of Medicine; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| | - Paula Poblete
- ICBM; Pharmacology Program; Faculty of Medicine; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| | - Viviana Noriega
- Cardiovascular Department; Clinical Hospital; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| | - Juan C. Prieto
- ICBM; Pharmacology Program; Faculty of Medicine; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
- Cardiovascular Department; Clinical Hospital; University of Chile; Indepedencia 1027 Independencia Santiago Chile P.O. 8380453
| |
Collapse
|
43
|
Abstract
In modern times, all procedures in oral surgery need to be painless and management of postoperative pain needs to be adequate. The surgical extraction of the third molar or alveolectomy of the wisdom tooth is one of the most common surgical procedures carried out in oral surgery and it includes rising a flap, bone removal and suturing. These surgical procedures usually cause swelling, trismus and moderate to severe pain. Third molar surgery is often used as a model in clinical trials that are directed toward reducing postoperative pain and improving its management. Tramadol is a well-known central acting opioid analgesic that produces analgesia against multiple pain conditions such as postsurgical pain, obstetric pain, terminal cancer pain, pain of coronary origin and neuropathic pain. Tramadol is an atypical opioid. When administered locally, it has both analgesic and anesthetic properties. The aim of this paper was to present new findings on local effects of tramadol in oral surgery.
Collapse
Affiliation(s)
- Petar Đanić
- Department of Oral and Maxillofacial Surgery, University Hospital Dubrava, Zagreb
| | - Ivan Salarić
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine University of Zagreb
| | - Darko Macan
- Department of Oral and Maxillofacial Surgery, University Hospital Dubrava, Zagreb.,Department of Oral and Maxillofacial Surgery, School of Dental Medicine University of Zagreb
| |
Collapse
|
44
|
Mehdizadeh H, Pourahmad J, Taghizadeh G, Vousooghi N, Yoonessi A, Naserzadeh P, Behzadfar L, Rouini MR, Sharifzadeh M. Mitochondrial impairments contribute to spatial learning and memory dysfunction induced by chronic tramadol administration in rat: Protective effect of physical exercise. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:426-433. [PMID: 28757160 DOI: 10.1016/j.pnpbp.2017.07.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/06/2017] [Accepted: 07/26/2017] [Indexed: 01/15/2023]
Abstract
Despite the worldwide use of tramadol, few studies have been conducted about its effects on memory and mitochondrial function, and controversial results have been reported. Recently, there has been an increasing interest in physical exercise as a protective approach to neuronal and cognitive impairments. Therefore, the aim of this study was to investigate the effects of physical exercise on spatial learning and memory and brain mitochondrial function in tramadol-treated rats. After completion of 2-week (short-term) and 4-week (long-term) treadmill exercise regimens, male Wistar rats received tramadol (20, 40, 80mg/kg/day) intraperitoneally for 30days. Then spatial learning and memory was assessed by Morris water maze test (MWM). Moreover, brain mitochondrial function was evaluated by determination of mitochondrial reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP), mitochondrial swelling and cytochrome c release from mitochondria. Chronic administration of tramadol impaired spatial learning and memory as well as brain mitochondrial function as indicated by increased ROS level, MMP collapse, increased mitochondrial swelling and cytochrome c release from mitochondria. Conversely, treadmill exercise significantly attenuated the impairments of spatial learning and memory and brain mitochondrial dysfunction induced by tramadol. The results revealed that chronic tramadol treatment caused memory impairments through induction of brain mitochondrial dysfunction. Furthermore, pre-exposure to physical exercise markedly mitigated these impairments through its positive effects on brain mitochondrial function.
Collapse
Affiliation(s)
- Hajar Mehdizadeh
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, Faculty of Rehabilitation, Iran University of Medical Sciences, Tehran, Iran; Rehabilitation Research Center, Faculty of Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Nasim Vousooghi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Yoonessi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ladan Behzadfar
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rouini
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Nimmo SM, Foo ITH, Paterson HM. Enhanced recovery after surgery: Pain management. J Surg Oncol 2017; 116:583-591. [PMID: 28873505 DOI: 10.1002/jso.24814] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Effective pain management is fundamental to enhanced recovery after surgery. Selection of strategies should be tailored to patient and operation. As well as improving the quality of recovery, effective analgesia reduces the host stress response, facilitates mobilization and allows resumption of oral intake. Multi-modal regimens combining paracetamol, non-steroidal anti-inflammatory agents where indicated, a potent opioid and a local anaesthetic technique achieve effective analgesia while limiting the dose and thereby side effects of any one agent.
Collapse
Affiliation(s)
- Susan M Nimmo
- Department of Anaesthesia, Critical Care and Pain Medicine, Western General Hospital, Edinburgh, Scotland
| | - Irwin T H Foo
- Department of Anaesthesia, Critical Care and Pain Medicine, Western General Hospital, Edinburgh, Scotland
| | - Hugh M Paterson
- Colorectal Surgery Unit, Western General Hospital, University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
46
|
Golightly LK, Simendinger BA, Barber GR, Stolpman NM, Kick SD, McDermott MT. Hypoglycemic effects of tramadol analgesia in hospitalized patients: a case-control study. J Diabetes Metab Disord 2017; 16:30. [PMID: 28748177 PMCID: PMC5525300 DOI: 10.1186/s40200-017-0311-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND In outpatient populations, hypoglycemia has been associated with tramadol. We sought to determine the magnitude of risk for hypoglycemia associated with tramadol use in hospitalized patients. METHODS During a 2-year period of observation, adult inpatients who received ≥1 dose of tramadol were identified and their medical records were reviewed. Patients were included if they had blood or plasma glucose (BG) concentrations measured on at least two occasions within five days after the initial administration of tramadol. A contemporary comparator group of hospitalized oxycodone recipients was similarly reviewed. RESULTS Tramadol was administered to 2927 patients who met inclusion criteria. Among these, hypoglycemia (BG ≤70 mg/dL) was documented in 22 (46.8%) of 47 patients with type 1 diabetes, 113 (16.8%) of 673 patients with type 2 diabetes, and 103 (4.7%) of 2207 patients who did not have a diabetes mellitus diagnosis. In those without a diabetes diagnosis, the causality association between hypoglycemia and tramadol use was probable in 77 patients (3.5%). By comparison, hypoglycemia was documented in 8 (1.1%) of 716 matched oxycodone recipients without diabetes (p = 0.002). As compared with tramadol recipients who did not develop low BG concentrations, those who experienced tramadol-related hypoglycemia were relatively young (mean age 52.0 versus 59.8 years; p = 0.027) and predominantly female (74.0% versus 59.8%; p = 0.012). CONCLUSIONS Tramadol use was causally associated with hypoglycemia in hospitalized patients. The proportion of patients without diabetes who developed hypoglycemia was higher among those who received tramadol than among those who received oxycodone. TRIAL REGISTRATION Colorado Multiple Institutional Review Board Protocol № 15-2215. Registered/approved 8 December 2015.
Collapse
Affiliation(s)
- Larry K. Golightly
- University of Colorado Hospital, Aurora, CO USA
- Division of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO USA
- Health Sciences Library/Center for Drug Information, Education, and Evaluation, University of Colorado Hospital, Anschutz Medical Campus Box A-003, 12950 East Montview Boulevard, Aurora, CO 80045-2515 USA
| | - Bonita A. Simendinger
- University of Colorado Hospital, Aurora, CO USA
- Division of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO USA
- Health Sciences Library/Center for Drug Information, Education, and Evaluation, University of Colorado Hospital, Anschutz Medical Campus Box A-003, 12950 East Montview Boulevard, Aurora, CO 80045-2515 USA
| | - Gerard R. Barber
- University of Colorado Hospital, Aurora, CO USA
- Division of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO USA
| | - Nancy M. Stolpman
- University of Colorado Hospital, Aurora, CO USA
- Division of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO USA
| | - Steven D. Kick
- University of Colorado Hospital, Aurora, CO USA
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, CO USA
| | - Michael T. McDermott
- University of Colorado Hospital, Aurora, CO USA
- Division of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO USA
- Division of Endocrinology Diabetes and Metabolism, University of Colorado School of Medicine, Aurora, CO USA
| |
Collapse
|
47
|
Wiffen PJ, Derry S, Moore RA, Cochrane Pain, Palliative and Supportive Care Group. Tramadol with or without paracetamol (acetaminophen) for cancer pain. Cochrane Database Syst Rev 2017; 5:CD012508. [PMID: 28510996 PMCID: PMC6481722 DOI: 10.1002/14651858.cd012508.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tramadol is an opioid analgesic licensed for use in moderate to severe pain. It is considered as a low risk for abuse, so control regulations are not as stringent as for 'strong' opioids such as morphine. It has a potential role as a step 2 option of the World Health Organization (WHO) analgesic ladder. OBJECTIVES To assess the benefits and adverse effects of tramadol with or without paracetamol (acetaminophen) for cancer-related pain. SEARCH METHODS We searched the following databases using a wide range of search terms: the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and LILACS. We also searched three clinical trials registry databases. The date of the last search was 2 November 2016. SELECTION CRITERIA We selected studies that were randomised, with placebo or active controls, or both, and included a minimum of 10 participants per treatment arm. We were interested particularly in blinded studies, but also included open studies.We excluded non-randomised studies, studies of experimental pain, case reports, and clinical observations. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data using a standard form and checked for agreement before entry into Review Manager 5. We included information about the number of participants treated and demographic details, type of cancer, drug and dosing regimen, study design (placebo or active control) and methods, study duration and follow-up, analgesic outcome measures and results, withdrawals, and adverse events. We collated multiple reports of the same study, so that each study, rather than each report, was the unit of interest in the review. We assessed the evidence using GRADE and created a 'Summary of findings' table.The main outcomes of interest for benefit were pain reduction of 30% or greater and 50% or greater from baseline, participants with pain no worse than mild, and participants feeling much improved or very much improved. MAIN RESULTS We included 10 studies (12 reports) with 958 adult participants. All the studies enrolled participants with chronic malignant tumour-related pain who were experiencing pain intensities described as moderate to severe, with most experiencing at least 4/10 with current treatment. The mean ages were 59 to 70 years, with participants aged between 24 and 87 years. Study length ranged from one day to six months. Five studies used a cross-over design. Tramadol doses ranged from 50 mg as single dose to 600 mg per day; doses of 300 mg per day to 400 mg per day were most common.Nine studies were at high risk of bias for one to four criteria (only one high risk of bias for size). We judged all the results to be very low quality evidence because of widespread lack of blinding of outcome assessment, inadequately described sequence generation, allocation concealment, and small numbers of participants and events. Important outcomes were poorly reported. There were eight different active comparators and one comparison with placebo. There was little information available for any comparison and no firm conclusions could be drawn for any outcome.Single comparisons of oral tramadol with codeine plus paracetamol, of dihydrocodeine, and of rectal versus oral tramadol provided no data for key outcomes. One study used tramadol combined with paracetamol; four participants received this intervention. One study compared tramadol with flupirtine - a drug that is no longer available. One study compared tramadol with placebo and a combination of cobrotoxin, tramadol, and ibuprofen, but the dosing schedule poorly explained.Two studies (191 participants) compared tramadol with buprenorphine. One study (131 participants) reported a similar proportion of no or mild pain at 14 days.Three studies (300 participants) compared tramadol with morphine. Only one study, combining tramadol, tramadol plus paracetamol, and paracetamol plus codeine as a single weak-opioid group reported results. Weak opioid produced reduction in pain of at least 30% from baseline in 55/117 (47%) participants, compared with 91/110 (82%) participants with morphine. Weak opioid produced reduction in pain of at least 50% in 49/117 (42%) participants, compared with 83/110 (75%) participants with morphine.There was no useful information for any other outcome of benefit or harm. AUTHORS' CONCLUSIONS There is limited, very low quality, evidence from randomised controlled trials that tramadol produced pain relief in some adults with pain due to cancer and no evidence at all for children. There is very low quality evidence that it is not as effective as morphine. This review does not provide a reliable indication of the likely effect. The likelihood that the effect will be substantially different is very high. The place of tramadol in managing cancer pain and its role as step 2 of the WHO analgesic ladder is unclear.
Collapse
|
48
|
Fornasari D, Allegri M, Gerboni S, Fanelli G. A "novel" association to treat pain: tramadol/dexketoprofen. The first drug of a "new pharmacological class". ACTA BIO-MEDICA : ATENEI PARMENSIS 2017; 88:17-24. [PMID: 28467329 PMCID: PMC6166198 DOI: 10.23750/abm.v88i1.6361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 04/11/2017] [Indexed: 01/21/2023]
Abstract
Acute and chronic pain have an important socio-economical impact. In order to help physicians to choose the appropriate drug, especially for cancer pain, in 1986 WHO has developed a three-step analgesic “ladder” for cancer pain relief in adults. Later it has also been used for acute pain and chronic non-cancer pain. In step I nonsteroidal anti-inflammatory drugs (NSAIDs) are considered with or without adjuvants, in step II the use of weak opioids for mild-moderate pain, with or without NSAIDs and adjuvant, is suggested, while the step III is reserved to strong opioids for moderate-severe pain with or without non-opioids or adjuvants. In the last two decades, a better pathophysiology knowledge has improved pain management shifting our view from the pain ladder to a modern pain pyramid, in which drugs are selected not only on the basis of pain intensity, but mainly according to mechanisms underlying pain, including peripheral and spinal sensitization which is the main trigger of chronic pain. The best pharmacological approach has become multimodal, in which drugs belonging to different steps should be combined, matching the mechanisms of action with the type of pain. An important corollary of combining analgesic drugs with different mechanism of action is that proper matching achieves the same effect with lower doses, better outcome and fewer adverse effects. In this new perspective, fixed-dose pharmaceutical combinations of different drugs are very useful to fulfil pharmacodynamics, pharmacokinetics and adherence criteria, enriching the pain pyramid of half-steps between the first and second step and between the second and third step. Hence, a new fixed combination of a NSAID with peripheral and central anti-infilammatory activities, such as dexketoprofen, and a weak opioid, such as tramadol, with double analgesic activity in the spinal cord as an opioid and, at the same time, on the descending modulatory pathways, is expected to cover a wide range of acute and recurrent painful conditions, ranging from nociceptive inflammatory pain to neuropathic pain of moderate/severe intensity. In this review we evaluate the rationale that justifies its use as new class of pharmacological modality to treat pain accordingly also to a more update view of WHO pain ladder. (www.actabiomedica.it)
Collapse
|
49
|
Monteiro BP, Klinck MP, Moreau M, Guillot M, Steagall PVM, Pelletier JP, Martel-Pelletier J, Gauvin D, del Castillo JRE, Troncy E. Analgesic efficacy of tramadol in cats with naturally occurring osteoarthritis. PLoS One 2017; 12:e0175565. [PMID: 28403198 PMCID: PMC5389833 DOI: 10.1371/journal.pone.0175565] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/28/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study aimed to (1) compare outcome assessments in normal and osteoarthritic cats and (2) evaluate the analgesic efficacy of tramadol in feline osteoarthritis (OA), in a prospective, randomised, blinded, placebo-controlled, crossover design. METHODS Twenty cats were included after clinical examination, blood work and full body radiographs were performed. In Phase 1, outcome assessments aimed to differentiate normal (n = 5; i.e. exempt of any radiographic and clinical sign of OA) from OA (n = 15) cats. In Phase 2, OA cats were treated twice daily with a placebo (PG: cornstarch 15 mg) or tramadol (TG: 3 mg/kg) orally for 19 days, with a 3-month washout period between treatments. Evaluations were performed in normal and OA cats at baseline and consisted of: 1) peak vertical force (PVF) after staircase exercise; 2) telemetered night-time motor activity (NMA); and 3) response to mechanical temporal summation (RMTS). After treatment, PVF, NMA and RMTS evaluations were repeated in OA cats. Data were analysed with mixed model methods with an alpha-threshold of 5%. RESULTS Phase 1: 1) PVF (% of body weight; mean ± SD) was higher in normal (59 ± 10.5) than in OA cats (50.6 ± 5.7) (p = 0.005); 2) NMA (no unit) was not different between groups; 3) RMTS (number of stimuli; median (range)) was higher in normal [29.5 (23.5-30)] than in OA cats [14 (8.5-28)] (p < 0.0001). Phase 2: PVF, NMA and RMTS presented a treatment effect (p = 0.024, p = 0.008 and p = 0.018, respectively). No clinically important adverse-effects were observed. CONCLUSION Outcome assessments such as kinetics (PVF) and evaluation of central sensitisation (RMTS) are discriminant of OA status. Mobility measured by NMA was not discriminant of OA status, however it increased in OA cats with tramadol treatment. Nociceptive hypersensitivity quantified by RMTS was evident in OA cats and was responsive to tramadol treatment.
Collapse
Affiliation(s)
- Beatriz P. Monteiro
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Mary P. Klinck
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| | - Maxim Moreau
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| | - Martin Guillot
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| | - Paulo V. M. Steagall
- Department of Clinical Sciences, Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| | - Dominique Gauvin
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Jérôme R. E. del Castillo
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Eric Troncy
- GREPAQ (Animal Pharmacology Research Group of Quebec), Faculty of Veterinary Medicine–Université de Montréal, Saint-Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, Research Center of the University of Montreal Hospital Centre, Montreal, QC, Canada
| |
Collapse
|
50
|
Lamana SMS, Napimoga MH, Nascimento APC, Freitas FF, de Araujo DR, Quinteiro MS, Macedo CG, Fogaça CL, Clemente-Napimoga JT. The anti-inflammatory effect of tramadol in the temporomandibular joint of rats. Eur J Pharmacol 2017; 807:82-90. [PMID: 28412371 DOI: 10.1016/j.ejphar.2017.04.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/05/2017] [Accepted: 04/11/2017] [Indexed: 12/27/2022]
Abstract
Tramadol is a centrally acting analgesic drug able to prevent nociceptor sensitization when administered into the temporomandibular joint (TMJ) of rats. The mechanism underlying the peripheral anti-inflammatory effect of tramadol remains unknown. This study demonstrated that intra-TMJ injection of tramadol (500µg/TMJ) was able to inhibit the nociceptive response induced by 1.5% formalin or 1.5% capsaicin, suggesting that tramadol has an antinociceptive effect, acting directly on the primary nociceptive neurons activating the nitric oxide/cyclic guanosine monophosphate signaling pathway. Tramadol also inhibited the nociceptive response induced by carrageenan (100µg/TMJ) or 5-hydroxytryptamine (225µg/TMJ) along with inhibition of inflammatory cytokines levels, leukocytes migration and plasma extravasation. In conclusion, the results demonstrate that peripheral administration of tramadol has a potential antinociceptive and anti-inflammatory effect. The antinociceptive effect is mediated by activation of the intracellular nitric oxide/cyclic guanosine monophosphate pathway, at least in part, independently from the opioid system.
Collapse
Affiliation(s)
- Simone Monaliza S Lamana
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil
| | - Marcelo H Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil
| | - Ana Paula Camatta Nascimento
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil
| | - Fabiana F Freitas
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil
| | - Daniele R de Araujo
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, SP, Brazil
| | - Mariana S Quinteiro
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil
| | - Cristina G Macedo
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil; Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil
| | - Carlos L Fogaça
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil
| | - Juliana T Clemente-Napimoga
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, SP, Brazil; Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil.
| |
Collapse
|