1
|
de Moura FB, Kohut SJ. Alternative Reinforcers Enhance the Effects of Opioid Antagonists, but Not Agonists, on Oxycodone Choice Self-Administration in Nonhuman Primates. J Pharmacol Exp Ther 2024; 391:301-307. [PMID: 39284627 PMCID: PMC11493445 DOI: 10.1124/jpet.123.001911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/06/2024] [Indexed: 10/20/2024] Open
Abstract
Clinical reports suggest that the most effective strategies for managing opioid use disorder comprise a comprehensive treatment program of both pharmacological and nonpharmacological approaches. However, the conditions under which these combinations are most effective are not well characterized. This study examined whether the presence of an alternative reinforcer could alter the efficacy of Food and Drug Administration-approved opioid antagonist or agonist medications, as well as the nonopioid flumazenil, in decreasing oxycodone choice self-administration in nonhuman primates. Adult squirrel monkeys (n = 7; four females) responded under concurrent second-order fixed-ratio (FR)-3(FR5:S);TO45s schedules of reinforcement for intravenous oxycodone (0.1 mg/kg) or saline on one lever and 30% sweetened condensed milk or water on the other. Doses of naltrexone (0.00032-1.0 mg/kg), nalbuphine (0.32-10 mg/kg), buprenorphine (0.0032-0.032 mg/kg), methadone (0.32-1.0 mg/kg), or flumazenil (1-3.2 mg/kg) were administered intramuscularly prior to oxycodone self-administration sessions that occurred with either milk or water as the alternative. Naltrexone, a μ-opioid receptor antagonist, was >30-fold more potent when milk was available compared with water and abolished oxycodone intake (injections/session) while concomitantly increasing milk deliveries at the highest dose tested. Pretreatment with the low-efficacy μ-agonist nalbuphine was most effective in the presence of milk compared with water, decreasing oxycodone preference to <50% of control values. The higher efficacy μ-agonists, methadone and buprenorphine, and the benzodiazepine antagonist flumazenil did not appreciably alter the reinforcing potency of oxycodone under either condition. These results suggest that antagonist medications used in combination with alternative reinforcers may be an effective strategy to curtail opioid abuse-related behaviors. SIGNIFICANCE STATEMENT: Clinical treatment programs for opioid use disorder use a combination of pharmacological and nonpharmacological approaches. However, the conditions under which these combinations are most effective have not been fully characterized. This study examined whether the effectiveness of μ-opioid medications to decrease oxycodone self-administration is altered in the presence of an alternative reinforcer. The results suggest that alternative reinforcers enhance the effects of antagonist or low-efficacy partial agonists, suggesting they may be a more effective strategy to curtail opioid use.
Collapse
Affiliation(s)
- Fernando B de Moura
- Behavioral Neuroimaging Laboratory, McLean Hospital, Belmont, Massachusetts (F.B.d.M., S.J.K.) and Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (F.B.d.M., S.J.K.)
| | - Stephen J Kohut
- Behavioral Neuroimaging Laboratory, McLean Hospital, Belmont, Massachusetts (F.B.d.M., S.J.K.) and Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (F.B.d.M., S.J.K.)
| |
Collapse
|
2
|
Fithian J, Doden G, Brandão J. Effective Dose of Dexmedetomidine with Nalbuphine Hydrochloride or Butorphanol Tartrate for Sedation in Buff Orpington Hens ( Gallus gallus domesticus). J Avian Med Surg 2024; 38:133-140. [PMID: 39405212 DOI: 10.1647/avianms-d-23-00054] [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: 03/20/2025]
Abstract
Chickens (Gallus gallus domesticus) are commonly used for research, food production, show, and companionship. Sedation is often necessary for sample collection, imaging, or treatment. Dexmedetomidine has been previously used to sedate birds, often with other sedatives. Butorphanol tartrate, a Schedule IV controlled substance, is commonly used but presents regulatory challenges. Nalbuphine hydrochloride, an opioid with similar receptor affinity to butorphanol, has potential as a noncontrolled alternative. Although information regarding nalbuphine use in birds is limited, its noncontrolled status makes it more accessible. The purpose of this study was to determine the effective dose to produce sedation in 50% (ED50) of patients and to estimate the calculated effective dose of dexmedetomidine in combination with either butorphanol (DexBut) or nalbuphine (DexNal) in domestic hens to sedate 99% of patients (ED99). Eighteen 33-week-old laying Buff Orpington hens were divided into 2 groups: one receiving DexBut (n = 9) and the second receiving DexNal (n = 9). Each hen was sedated with varying doses of intramuscular dexmedetomidine with a constant dose of either 2 mg/kg IM butorphanol or 12.5 mg/kg IM nalbuphine by an up-and-down design. Sedation was determined using a clinically applicable scoring system. The ED50 values of dexmedetomidine with 2 mg/kg IM of butorphanol, calculated by both the up-and-down method and logistic regression, were 38 and 49 µg/kg, respectively, while the ED50 values of dexmedetomidine in combination with 12.5 mg/kg IM of nalbuphine were 19 and 18 µg/kg, respectively. The estimated dexmedetomidine ED99 values with butorphanol or nalbuphine were 51 and 19 µg/kg, respectively. Multiple chickens in both groups exhibited open-mouth breathing and comb pallor but no lasting morbidity or mortality occurred. Combinations of DexBut or DexNal should be considered for sedation of domestic chickens.
Collapse
Affiliation(s)
- Julie Fithian
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Greta Doden
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - João Brandão
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA,
| |
Collapse
|
3
|
Korzekwa K, Nagar S, Clark D, Sciascia T, Hawi A. A Continuous Intestinal Absorption Model to Predict Drug Enterohepatic Recirculation in Healthy Humans: Nalbuphine as a Model Substrate. Mol Pharm 2024; 21:4510-4523. [PMID: 38956965 PMCID: PMC11999028 DOI: 10.1021/acs.molpharmaceut.4c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Nalbuphine (NAL) is a κ-agonist/μ-antagonist opioid being developed as an oral extended formulation (ER) for the treatment of chronic cough in idiopathic pulmonary fibrosis and itch in prurigo nodularis. NAL is extensively glucuronidated and likely undergoes enterohepatic recirculation (EHR). The purpose of this work is to develop pharmacokinetic models for NAL absorption and enterohepatic recirculation (EHR). Clinical pharmacokinetic (PK) data sets in healthy subjects from three trials that included IV, oral solution, and ER tablets in fed and fasted state and two published trials were used to parametrize a novel partial differential equation (PDE)-based model, termed "PDE-EHR" model. Experimental inputs included in vitro dissolution and permeability data. The model incorporates a continuous intestinal absorption framework, explicit liver and gall bladder compartments, and compartments for systemic drug disposition. The model was fully PDE-based with well-stirred compartments achieved by rapid diffusion. The PDE-EHR model accurately reproduces NAL concentration-time profiles for all clinical data sets. NAL disposition simulations required inclusion of both parent and glucuronide recirculation. Inclusion of intestinal P-glycoprotein efflux in the simulations suggests that NAL is not expected to be a victim or perpetrator of P-glycoprotein-mediated drug interactions. The PDE-EHR model is a novel tool to predict EHR and food/formulation effects on drug PK. The results strongly suggest that even intravenous dosing studies be conducted in fasted subjects when EHR is suspected. The modeling effort is expected to aid in improved prediction of dosing regimens and drug disposition in patient populations.
Collapse
Affiliation(s)
- Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania 19140, United States
| | - Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania 19140, United States
| | - David Clark
- Trevi Therapeutics, New Haven, Connecticut 06510, United States
| | - Thomas Sciascia
- Trevi Therapeutics, New Haven, Connecticut 06510, United States
| | - Amale Hawi
- A. Hawi Consulting, Ridgefield, Connecticut 06877, United States
| |
Collapse
|
4
|
Cernea M, Nikonov G, Ataiants J, Ştefănuţ C, Abernethy J, Voronkov M. Nalbuphine Potentiates Reversal of Fentanyl Overdose by Naloxone. Pharmaceuticals (Basel) 2024; 17:866. [PMID: 39065717 PMCID: PMC11279547 DOI: 10.3390/ph17070866] [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: 06/06/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Developing an effective antidote for fentanyl-induced overdose to achieve timely reversal is an unmet public health need. Previously, we found that naloxone derivative NX90 with mild κ-opioid agonistic properties was three-fold more effective than the parent naloxone in reversing a fentanyl overdose in rats. To investigate whether κ-agonistic properties could indeed augment the robustness of overdose reversal, we evaluated a κ-agonist/µ-antagonist nalbuphine (NB) as well as its combinations with naloxone (NX) in a fentanyl overdose model in rodents. An administration of either NB or NX as single agents at 0.1 mg/kg doses produced a full recovery in 90 ± 9.9 min and 11.4 ± 2.7 min, respectively. A higher dose of NX at 0.2 mg/kg reversed an overdose within 4.8 ± 1.0 min. In contrast to that, the coadministration of NB and NX at 0.1 mg/kg each produced a synergistic effect, with overdose reversal in 3.4 ± 0.2 min. The coadministration of NX and NB at sub-therapeutic doses of 0.05 mg/kg each was also 1.2-fold more effective than NX at 0.2 mg/kg. We further found that co-administration of NB at different doses (0.025, 0.05, 0.1 mg/kg) and ratios (1:4 and 1:1) with NX had differential effects on overdose reversal, cardiorespiratory liabilities, and analgesia.
Collapse
Affiliation(s)
- Mihai Cernea
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.C.)
| | | | - Janna Ataiants
- Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, USA
| | - Cristina Ştefănuţ
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.C.)
| | | | | |
Collapse
|
5
|
Huang P, Ho CK, Cao D, Inan S, Rawls SM, Li M, Huang B, Pagare PP, Townsend EA, Poklis JL, Halquist MS, Banks M, Zhang Y, Liu-Chen LY. NCP, a Dual Kappa and Mu Opioid Receptor Agonist, Is a Potent Analgesic Against Inflammatory Pain without Reinforcing or Aversive Properties. J Pharmacol Exp Ther 2024; 389:106-117. [PMID: 38409113 PMCID: PMC10949162 DOI: 10.1124/jpet.123.001870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
While agonists of μ (MOR) and κ (KOR) opioid receptors have analgesic effects, they produce euphoria and dysphoria, respectively. Other side effects include respiratory depression and addiction for MOR agonists and sedation for KOR agonists. We reported that 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-{[4'-(2'-cyanopyridyl)]carboxamido}morphinan (NCP) displayed potent KOR full agonist and MOR partial agonist activities (58%) with 6.5x KOR-over-MOR selectivity in vitro. Herein, we characterized pharmacological effects of NCP in rodents. In mice, NCP exerted analgesic effects against inflammatory pain in both the formalin test and the acetic acid writhing test, with A50 values of 47.6 and 14.4 μg/kg (s.c.), respectively. The analgesic effects in the acetic acid writhing test were mediated by the KOR. NCP at doses much higher than those effective in reducing inflammatory pain did not produce antinociception in the hot plate and tail flick tests, inhibit compound 48/80-induced scratching, cause conditioned place aversion (CPA) or preference, impair rotarod performance, inhibit locomotor activity, cause respiratory depression, or precipitate morphine withdrawal. However, NCP (10∼100 μg/kg) inhibited gastrointestinal transit with a maximum of ∼40% inhibition. In MOR knockout mice, NCP caused CPA, demonstrating that its lack of CPA is due to combined actions on the MOR and KOR. Following subcutanous injection, NCP penetrated into the mouse brain. In rats trained to self-administer heroin, NCP (1∼320 μg/kg/infusion) did not function as a reinforcer. Thus, NCP produces potent analgesic effects via KOR without side effects except constipation. Therefore, dual full KOR/partial MOR agonists with moderate KOR-over-MOR selectivity may be promising as nonaddictive analgesics for inflammatory pain. SIGNIFICANCE STATEMENT: Developing nonaddictive analgesics is crucial for reducing opioid overdose deaths, minimizing drug misuse, and promoting safer pain management practices. Herein, the pharmacology of a potential nonaddictive analgesic, NCP, is reported. NCP has full KOR agonist/partial MOR agonist activities with a 6.5x selectivity for KOR over MOR. Unlike MOR agonists, analgesic doses of NCP do not lead to self-administration or respiratory depression. Furthermore, NCP does not produce aversion, hypolocomotion, or motor incoordination, side effects typically associated with KOR activation.
Collapse
Affiliation(s)
- Peng Huang
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Conrad K Ho
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Danni Cao
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Saadet Inan
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Scott M Rawls
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Mengchu Li
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Boshi Huang
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Piyusha P Pagare
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - E Andrew Townsend
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Justin L Poklis
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Matthew S Halquist
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Matthew Banks
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Yan Zhang
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania (P.H., C.K.H., D.C., S.I., S.M.R., L.-Y.L.-C.); Departments of Medicinal Chemistry (M.L., B.H., P.P.P., Y.Z.), Pharmacology and Toxicology (E.A.T., J.L.P., M.B., Y.Z.), and Pharmaceutics (M.S.H.) and Institute for Drug and Alcohol Studies (Y.Z.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
6
|
Chen YL, Lai YH, Huang EYK, Wang HJ, Hung HY. Nalbuphine-6-glucuronide is a potent analgesic with superior safety profiles by altering binding affinity and selectivity for mu-/kappa-opioid receptors. Life Sci 2024; 340:122441. [PMID: 38253309 DOI: 10.1016/j.lfs.2024.122441] [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: 09/28/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024]
Abstract
Although nalbuphine, a semi-synthetic analgesic compound, is less potent than morphine in terms of alleviating severe pain, our recent findings have revealed that nalbuphine-6-glucuronide (N6G), one of the glucuronide metabolites of nalbuphine, promotes a significantly more robust analgesic effect than its parent drug. Nevertheless, despite these promising observations, the precise mechanisms underlying the analgesic effects of nalbuphine glucuronides have yet to be determined. In this study, we aim to elucidate the mechanisms associated with the analgesic effects of nalbuphine glucuronides. Pharmacokinetic and pharmacodynamic studies were conducted to investigate the relationship between the central and peripheral compartments of nalbuphine and its derivatives. The analgesic responses of these compounds were evaluated based on multiple behavioral tests involving thermal and mechanical stimuli. Radioligand binding assays were also performed to determine the binding affinity and selectivity of these compounds for different opioid receptors. The results of these tests consistently confirmed that the heightened analgesic effects of N6G are mediated through its enhanced binding affinity for both mu- and kappa-opioid receptors, even comparable to those of morphine. Notably, N6G exhibited fewer side effects and did not induce sudden death, thereby highlighting its superior safety profile. Additionally, pharmacokinetic studies indicated that N6G could cross the blood-brain barrier when administered peripherally, offering pain relief. Overall, N6G provides great analgesic efficacy and enhanced safety. These findings highlight the potential value of nalbuphine glucuronides, particularly N6G, as promising candidates for the development of novel analgesic drugs.
Collapse
Affiliation(s)
- Yen-Lun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yen-Hsun Lai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Hong-Jaan Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| | - Hao-Yuan Hung
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China; Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China; Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| |
Collapse
|
7
|
Chen GY, Kuo KK, Chuang SC, Tseng KY, Wang SN, Chang WT, Cheng KI. Optimal Post-Operative Nalbuphine Dose Regimen: A Randomized Controlled Trial in Patients with Laparoscopic Cholecystectomy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:195. [PMID: 38399483 PMCID: PMC10890534 DOI: 10.3390/medicina60020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024]
Abstract
Background and Objectives: Optimal opioid analgesia is an excellent analgesia that does not present unexpected adverse effects. Nalbuphine, acting on the opioid receptor as a partial mu antagonist and kappa agonist, is considered a suitable option for patients undergoing laparoscopic surgery. Therefore, we aim to investigate the appropriate dosage of nalbuphine for post-operative pain management in patients with laparoscopic cholecystectomy. Materials and Methods: Patients were randomly categorized into low, medium, and high nalbuphine groups. In each group, a patient control device for post-operative pain control was programed with a low (0.05 mg/kg), medium (0.10 mg/kg), or high (0.20 mg/kg) nalbuphine dose as a loading dose and each bolus dose with a lockout interval of 7 min and without background infusion. Primary and secondary outcomes included the post-operative pain scale and nalbuphine consumption, and episodes of post-operative opioid-related adverse events and satisfactory scores. Results: The low-dosage group presented a higher initial self-reported pain score in comparison to the other two groups for the two hours post-op (p = 0.039) but presented lower nalbuphine consumption than the other two groups for four hours post-op (p = 0.047). There was no significant difference in the analysis of the satisfactory score and adverse events. Conclusions: An appropriate administration of nalbuphine could be 0.1 to 0.2 mg/kg at the initial four hours; this formula could be modified to a lower dosage (0.05 mg/kg) in the post-operative management of laparoscopic cholecystectomy.
Collapse
Affiliation(s)
- Guan-Yu Chen
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (G.-Y.C.); (K.-Y.T.)
- Department of Anesthesiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kung-Kai Kuo
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-K.K.); (S.-C.C.); (S.-N.W.); (W.-T.C.)
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-K.K.); (S.-C.C.); (S.-N.W.); (W.-T.C.)
| | - Kuang-Yi Tseng
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (G.-Y.C.); (K.-Y.T.)
- Department of Anesthesiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shen-Nien Wang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-K.K.); (S.-C.C.); (S.-N.W.); (W.-T.C.)
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-K.K.); (S.-C.C.); (S.-N.W.); (W.-T.C.)
| | - Kuang-I Cheng
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (G.-Y.C.); (K.-Y.T.)
- Department of Anesthesiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Singh S, Sri Krishna V, Cherian Ambooken G, Peter DK. Nalbuphine: an underrecognized battlefield analgesic and its utilization in combat care and peripheral areas. Med J Armed Forces India 2024; 80:41-45. [PMID: 38239600 PMCID: PMC10793233 DOI: 10.1016/j.mjafi.2023.03.003] [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: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 01/22/2024] Open
Abstract
Battlefield injuries result in acute and severe uncontrolled pain, which can be reduced with the early use of analgesia. Apart from pain, battlefield injuries may also cause significant morbidity and a prolonged period of absence from active duty. Traditionally available opioids are known to cause various undesirable side effects such as respiratory depression that may worsen the condition of an already injured combatant. Nalbuphine is an opioid agonist-antagonist and has been increasingly used for postoperative analgesia over the last decade. In India, it is the only opioid analgesic that does not come under the Controlled Substances Act at the time of this publication. In today's world, where nalbuphine is being recommended for acute pain worldwide, its use in the Indian combat scenario needs to be conceptualized at the medical officer level (primary caregiver). This conceptualization will be discussed in detail in this review article.
Collapse
Affiliation(s)
- Shalendra Singh
- Senior Advisor (Anaesthesia & Neuro Anaesthesia), Command Hospital (Northern Command), Udhampur, India
| | | | | | - Deepu K. Peter
- Graded Specialist (Respiratory Medicine), Command Hospital (Northern Command), Udhampur, India
| |
Collapse
|
9
|
Negus SS, Akbarali HI, Kang M, Lee YK, Marsh SA, Santos EJ, Zhang Y. Role of mu opioid receptor (MOR) agonist efficacy as a determinant of opioid antinociception in a novel assay of pain-depressed behavior in female and male mice. FRONTIERS IN PAIN RESEARCH 2023; 4:1281698. [PMID: 37886350 PMCID: PMC10598607 DOI: 10.3389/fpain.2023.1281698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Introduction Intermediate efficacy mu opioid receptor (MOR) agonists have potential to retain analgesic effectiveness while improving safety, but the optimal MOR efficacy for effective and safe opioid analgesia is unknown. Preclinical assays of pain-depressed behavior can assess effects of opioids and other candidate analgesics on pain-related behavioral depression, which is a common manifestation of clinically relevant pain and target of pain treatment. Accordingly, the present study goal was to validate a novel assay of pain-depressed locomotor behavior in mice and evaluate the role of MOR efficacy as a determinant of opioid analgesic effects and related safety measures. Methods Male and female ICR mice were tested in a locomotor chamber consisting of 2 compartments connected by a doorway that contained a 1-inch-tall barrier. Dependent measures during 15-min behavioral sessions included crosses between compartments (which required vertical activity to surmount the barrier) and total movement counts (which required horizontal activity to break photobeams in each compartment). Results and Discussion Intraperitoneal injection of lactic acid (IP acid) produced a concentration- and time-dependent depression of both endpoints. Optimal blockade of IP acid-induced behavioral depression with minimal motor impairment was achieved with intermediate-efficacy MOR treatments that also produced less gastrointestinal-transit inhibition and respiratory depression than the high-efficacy MOR agonist fentanyl. Sex differences in treatment effects were rare. Overall, these findings validate a novel procedure for evaluating opioids and other candidate analgesic effects on pain-related behavioral depression in mice and support continued research with intermediate-efficacy MOR agonists as a strategy to retain opioid analgesic effectiveness with improved safety.
Collapse
Affiliation(s)
- S. Stevens Negus
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Hamid I. Akbarali
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Minho Kang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Young K. Lee
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Samuel A. Marsh
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Edna J. Santos
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
10
|
Agrawal H, Chaudhary S, Salhotra R. Comparison of Nalbuphine Versus Clonidine as an Adjuvant to Intrathecal Hyperbaric Bupivacaine in Orthopedic Lower Limb Surgeries: A Randomized Controlled Double-Blind Study. Cureus 2023; 15:e42857. [PMID: 37664267 PMCID: PMC10473549 DOI: 10.7759/cureus.42857] [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: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Nalbuphine and clonidine are used as adjuvants to intrathecal local anesthetics, but studies on their comparative efficacy have shown inconsistent results. This study aimed to compare nalbuphine, clonidine, or normal saline as adjuvants to intrathecal hyperbaric bupivacaine in orthopedic lower limb surgeries. Method: Sixty-three American Society of Anesthesiologists (ASA) I/II patients between 18 and 60 years old with lower limb fractures were randomized into three groups (n=21 each). Patients with contraindications to spinal block, bilateral lower limb fractures, or long-term opioid therapy were excluded. The subarachnoid block was given in L3-L4 interspace. Group N received 1 mg of nalbuphine, group Cl received 30 mcg of clonidine, and group C received 0.5 ml of normal saline with 15 mg (0.5%) of hyperbaric bupivacaine. Sensory and motor block characteristics, hemodynamic variables and side effects were noted, and the data were analyzed using Student's t-test, Mann-Whitney test, Chi-square test, and ANOVA followed by Tukey's test. RESULTS Patients receiving intrathecal nalbuphine (group N) and clonidine (group Cl) had a faster onset of the sensory and motor block than controls (group C) (p=0.000). The time to two-segment regression was more prolonged in group Cl when compared to group N (p=0.000). Duration of spinal analgesia was 216.75 ± 25.96 minutes, 292.86 ± 24.92 minutes, and 178.50 ± 16.06 minutes in groups N, Cl, and C, respectively (p=0.000). The 24-hour rescue analgesic requirement was maximum in group C and least in group Cl (p=0.000). The three groups were comparable to each other in terms of side effects. CONCLUSION Clonidine was found to be superior to nalbuphine as an intrathecal adjuvant with no significant side effects.
Collapse
Affiliation(s)
- Heena Agrawal
- Department of Anesthesia, Late Bisahu Das Mahant Memorial Medical College, Korba, Jhagarha, IND
| | - Sujata Chaudhary
- Department of Anesthesia, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, IND
| | - Rashmi Salhotra
- Department of Anesthesia, University College of Medical Sciences, New Delhi, IND
| |
Collapse
|
11
|
Varga B, Streicher JM, Majumdar S. Strategies towards safer opioid analgesics-A review of old and upcoming targets. Br J Pharmacol 2023; 180:975-993. [PMID: 34826881 PMCID: PMC9133275 DOI: 10.1111/bph.15760] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 10/08/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
Opioids continue to be of use for the treatment of pain. Most clinically used analgesics target the μ opioid receptor whose activation results in adverse effects like respiratory depression, addiction and abuse liability. Various approaches have been used by the field to separate receptor-mediated analgesic actions from adverse effects. These include biased agonism, opioids targeting multiple receptors, allosteric modulators, heteromers and splice variants of the μ receptor. This review will focus on the current status of the field and some upcoming targets of interest that may lead to a safer next generation of analgesics. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Balazs Varga
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St Louis and Washington University School of Medicine, St Louis, MO, USA
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St Louis and Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
12
|
Nie X, Gao X, Gao J, Heng T, Zhang Y, Sun Y, Feng Z, Jia L, Wang M. Population pharmacokinetics of nalbuphine in patients undergoing general anesthesia surgery. Front Pharmacol 2023; 14:1130287. [PMID: 37025491 PMCID: PMC10070753 DOI: 10.3389/fphar.2023.1130287] [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: 12/23/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Purpose: The aim of this study was to build a population pharmacokinetics (PopPK) model of nalbuphine and to estimate the suitability of bodyweight or fixed dosage regimen. Method: Adult patients who were undergoing general anesthetic surgery using nalbuphine for induction of anesthesia were included. Plasma concentrations and covariates information were analyzed by non-linear mixed-effects modeling approach. Goodness-of-fit (GOF), non-parametric bootstrap, visual predictive check (VPC) and external evaluation were applied for the final PopPK model evaluation. Monte Carlo simulation was conducted to assess impact of covariates and dosage regimens on the plasma concentration to nalbuphine. Results: 47 patients aged 21-78 years with a body weight of 48-86 kg were included in the study. Among them, liver resection accounted for 14.8%, cholecystectomy for 12.8%, pancreatic resection for 36.2% and other surgeries for 36.2%. 353 samples from 27 patients were enrolled in model building group; 100 samples from 20 patients were enrolled in external validation group. The results of model evaluation showed that the pharmacokinetics of nalbuphine was adequately described by a two-compartment model. The hourly net fluid volume infused (HNF) was identified as a significant covariate about the intercompartmental clearance (Q) of nalbuphine with objective function value (OFV) decreasing by 9.643 (p < 0.005, df = 1). Simulation results demonstrated no need to adjust dosage based on HNF, and the biases of two dosage methods were less than 6%. The fixed dosage regimen had lower PK variability than the bodyweight regimen. Conclusion: A two-compartment PopPK model adequately described the concentration profile of nalbuphine intravenous injection for anesthesia induction. While HNF can affect the Q of nalbuphine, the magnitude of the effect was limited. Dosage adjustment based on HNF was not recommended. Furthermore, fixed dosage regimen might be better than body weight dosage regimen.
Collapse
Affiliation(s)
- Xuyang Nie
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaonan Gao
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinglin Gao
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tianfang Heng
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuqi Zhang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaqi Sun
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhangying Feng
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Mingxia Wang, ; Li Jia,
| | - Mingxia Wang
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Mingxia Wang, ; Li Jia,
| |
Collapse
|
13
|
Gettelman TE, Nielsen CK, Scimeca JM, Schauber EM. River otter chemical immobilization and field surgery using nonscheduled drugs. WILDLIFE SOC B 2022. [DOI: 10.1002/wsb.1354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tatiana E. Gettelman
- Department of Zoology and Cooperative Wildlife Research Laboratory Southern Illinois University Carbondale IL 62901 USA
| | - Clayton K. Nielsen
- Department of Forestry and Cooperative Wildlife Research Laboratory Southern Illinois University Carbondale IL 62901 USA
| | - Joseph M. Scimeca
- Department of Animal Science, Food and Nutrition Southern Illinois University Carbondale IL 62901 USA
| | - Eric M. Schauber
- Illinois Natural History Survey, Prairie Research Institute, University of Illinois Champaign IL 61820 USA
| |
Collapse
|
14
|
McLaughlin JP, Rayala R, Bunnell AJ, Tantak MP, Eans SO, Nefzi K, Ganno ML, Dooley CT, Nefzi A. Bis-Cyclic Guanidine Heterocyclic Peptidomimetics as Opioid Ligands with Mixed μ-, κ- and δ-Opioid Receptor Interactions: A Potential Approach to Novel Analgesics. Int J Mol Sci 2022; 23:9623. [PMID: 36077029 PMCID: PMC9455983 DOI: 10.3390/ijms23179623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022] Open
Abstract
The design and development of analgesics with mixed-opioid receptor interactions has been reported to decrease side effects, minimizing respiratory depression and reinforcing properties to generate safer analgesic therapeutics. We synthesized bis-cyclic guanidine heterocyclic peptidomimetics from reduced tripeptides. In vitro screening with radioligand competition binding assays demonstrated variable affinity for the mu-opioid receptor (MOR), delta-opioid receptor (DOR), and kappa-opioid receptor (KOR) across the series, with compound 1968-22 displaying good affinity for all three receptors. Central intracerebroventricular (i.c.v.) administration of 1968-22 produced dose-dependent, opioid receptor-mediated antinociception in the mouse 55 °C warm-water tail-withdrawal assay, and 1968-22 also produced significant antinociception up to 80 min after oral administration (10 mg/kg, p.o.). Compound 1968-22 was detected in the brain 5 min after intravenous administration and was shown to be stable in the blood for at least 30 min. Central administration of 1968-22 did not produce significant respiratory depression, locomotor effects or conditioned place preference or aversion. The data suggest these bis-cyclic guanidine heterocyclic peptidomimetics with multifunctional opioid receptor activity may hold potential as new analgesics with fewer liabilities of use.
Collapse
Affiliation(s)
- Jay P. McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Ramanjaneyulu Rayala
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Ashley J. Bunnell
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Mukund P. Tantak
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Khadija Nefzi
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Colette T. Dooley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Adel Nefzi
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| |
Collapse
|
15
|
Bello M. Molecular Basis of Inhibitory Mechanism of Naltrexone and Its Metabolites through Structural and Energetic Analyses. Molecules 2022; 27:molecules27154919. [PMID: 35956868 PMCID: PMC9369988 DOI: 10.3390/molecules27154919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Naltrexone is a potent opioid antagonist with good blood–brain barrier permeability, targeting different endogenous opioid receptors, particularly the mu-opioid receptor (MOR). Therefore, it represents a promising candidate for drug development against drug addiction. However, the details of the molecular interactions of naltrexone and its derivatives with MOR are not fully understood, hindering ligand-based drug discovery. In the present study, taking advantage of the high-resolution X-ray crystal structure of the murine MOR (mMOR), we constructed a homology model of the human MOR (hMOR). A solvated phospholipid bilayer was built around the hMOR and submitted to microsecond (µs) molecular dynamics (MD) simulations to obtain an optimized hMOR model. Naltrexone and its derivatives were docked into the optimized hMOR model and submitted to µs MD simulations in an aqueous membrane system. The MD simulation results were submitted to the molecular mechanics–generalized Born surface area (MMGBSA) binding free energy calculations and principal component analysis. Our results revealed that naltrexone and its derivatives showed differences in protein–ligand interactions; however, they shared contacts with residues at TM2, TM3, H6, and TM7. The binding free energy and principal component analysis revealed the structural and energetic effects responsible for the higher potency of naltrexone compared to its derivatives.
Collapse
Affiliation(s)
- Martiniano Bello
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón, s/n, Col. Casco de Santo Tomas, Mexico City 11340, Mexico
| |
Collapse
|
16
|
Gao XN, Nie XY, Gao JL, Heng TF, Zhang YQ, Hua L, Sun YQ, Feng ZY, Wang MX, Jia L. Pharmacokinetic Study of Nalbuphine in Surgical Patients Undergoing General Anesthesia with Varying Degrees of Liver Dysfunction. Drug Des Devel Ther 2022; 16:2383-2393. [PMID: 35923933 PMCID: PMC9341258 DOI: 10.2147/dddt.s371596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/09/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aimed to characterize the pharmacokinetics of nalbuphine in patients undergoing general anesthesia with varying degrees of liver dysfunction. Patients and Methods Twenty-four patients were enrolled and divided into three cohorts based on liver function: normal liver function (n = 13), mild liver dysfunction (n = 5), and moderate/severe liver dysfunction (n = 6). During the induction of anesthesia, they received 15 mg of nalbuphine intravenously. Venous blood samples were collected from each patient. The plasma concentration of nalbuphine was determined using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The pharmacokinetic parameters of nalbuphine were calculated by non-compartmental analysis (NCA) using Phoenix WinNonlin software. Results Compared with the normal liver function group, the plasma elimination half-life (T1/2) of nalbuphine was increased by approximately 33% in the moderate/severe liver dysfunction group (2.66 h vs 3.54 h, P<0.05), and the volume of distribution (Vd) increased by approximately 85% (100.08 L vs 184.95 L, P<0.05). Multivariate analysis revealed that weight and platelet were associated with clearance (CL); total bilirubin as an independent factor was associated with T1/2, and weight associated with area under the curve (AUC(0→∞)) independently. Conclusion The T1/2, mean residence time, and Vd of nalbuphine in patients with moderate/severe liver dysfunction were prolonged or increased significantly compared with those in the normal liver function group. These data suggest that it may need to be used with caution when nalbuphine is administered to patients with moderate or severe liver dysfunction.
Collapse
Affiliation(s)
- Xiao-nan Gao
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xu-yang Nie
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Jing-lin Gao
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Tian-fang Heng
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yu-qi Zhang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Li Hua
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Ya-qi Sun
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Zhang-ying Feng
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Ming-xia Wang
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Correspondence: Ming-xia Wang, Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, People’s Republic of China, Tel +86 311-66696233, Email
| | - Li Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Li Jia, Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, People’s Republic of China, Email
| |
Collapse
|
17
|
Modulation of the kappa and mu opioid axis for the treatment of chronic pruritus: A review of basic science and clinical implications. JAAD Int 2022; 7:156-163. [PMID: 35497636 PMCID: PMC9046882 DOI: 10.1016/j.jdin.2022.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 11/11/2022] Open
Abstract
Introduction Treating chronic pruritus is challenging for dermatologists due to the lack of therapeutic options. We review the effects of κ-opioid receptor (KOR) and μ-opioid receptor (MOR) in the modulation of itch, summarize evidence supporting the efficacy and safety of opioid receptor–targeting agents in chronic pruritus, and address clinical considerations. Results Preclinical studies have found neural pathways underlying detection, transmission, and modulation of itch signaling and spotlighted the importance of neuronal KOR and MOR in itch perception. Clinical reports suggest that opioid axis modulation may be the basis for the successful treatment of chronic itch. Several agents (MOR antagonist naltrexone; KOR agonists nalfurafine and difelikefalin; dual-acting KOR agonists/MOR antagonists butorphanol and nalbuphine) have been evaluated for treating chronic pruritus in case series, small studies, and clinical trials; nalbuphine has progressed through preliminary (phase II/III) studies in uremic pruritus and prurigo nodularis. The antipruritic efficacy of these agents has been observed across multiple disorders with disparate etiologies, suggesting the potential utility of this class to provide a unified approach to chronic pruritus treatment. Conclusions The relative safety of these agents, including a reduced potential for dependence versus MOR-agonist analgesics, should help overcome resistance to the use of opioid receptor–targeting agents in chronic pruritus treatment.
Collapse
|
18
|
Ruan D, Wang Y, Li S, Zhang C, Zheng W, Yu C. Nalbuphine alleviates inflammation by down-regulating NF-κB in an acute inflammatory visceral pain rat model. BMC Pharmacol Toxicol 2022; 23:34. [PMID: 35642022 PMCID: PMC9158276 DOI: 10.1186/s40360-022-00573-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Nalbuphine can relieve patients’ inflammation response after surgery compared to other opioid drugs. However, its molecular mechanism has not been clear. Activation of NF-κB signaling pathway under oxidative stress and inflammation can maintain pain escalation. Methods We firstly investigated the effect of nalbuphine on writhing test and mechanical allodynia using a rat model of inflammatory visceral pain (acetic acid (AA) administrated). Cytokines (including tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, IL-2, and IL-6 in plasma were tested with ELISA technology. Expression levels of TNF-α, IκBα and p-NF-κB p65 at the spinal cord (L3–5) were measured by western blot or RT-qPCR. Results We found that the paw withdrawal threshold (PWT) values of rats were reduced in the model group, while the numbers of writhing, levels of IL-1β, IL-2, IL-6, and TNF-α in plasma, and p-NF-κB protein and its gene expressions in the lumbar spinal cord were up-regulated. Subcutaneously injection of nalbuphine (10 μg/kg) or PDTC (NF-κB inhibitor) attenuated acetic acid-induced inflammatory pain, and this was associated with reversal of up-regulated IL-1β, IL-2, IL-6, and TNF-α in both plasma and spinal cord. Furthermore, acetic acid increased p-NF-κB and TNF-α protein levels in the white matter of the spinal cord, which was attenuated by nalbuphine. These results suggested that nalbuphine can significantly ameliorate inflammatory pain via modulating the expression of NF-κB p65 as well as inflammation factors level in the spinal cord. Conclusion In conclusion, nalbuphine inhibits inflammation through down-regulating NF-κB pathway at the spinal cord in a rat model of inflammatory visceral pain. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00573-7.
Collapse
Affiliation(s)
- Dijiao Ruan
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuanyuan Wang
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Sisi Li
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Chao Zhang
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Wenwen Zheng
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Cong Yu
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, 426 Songs North Road, Yubei District, Chongqing, China. .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China. .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
19
|
Pokotylo PB, Fedevych YM, Denysenko NV, Logash MV, Genyk ID. CHANGES IN THE MITOCHONDRIAL APPARATUS OF CARDIOMYOCYTES UNDER THE INFLUENCE OF OPIOID IN THE EXPERIMENT. BULLETIN OF PROBLEMS BIOLOGY AND MEDICINE 2022. [DOI: 10.29254/2077-4214-2022-3-166-436-441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | | | - M. V. Logash
- Danylo Halytsky Lviv National Medical University
| | - I. D. Genyk
- Danylo Halytsky Lviv National Medical University
| |
Collapse
|
20
|
Ayad AE, Salman OH, Ibrahim AMF, Al-Taher WAM, Mishriky AM, Pergolizzi JV, Viswanath O, Urits I, Rekatsina M, Peppin JF, Paladini A, Varrassi G. A Response to: Letter to the Editor regarding "Influences of Gender on Intravenous Nalbuphine Actions After Major Abdominal Surgery: A Multicenter Study". Pain Ther 2021; 10:1783-1786. [PMID: 34431072 PMCID: PMC8586306 DOI: 10.1007/s40122-021-00305-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Amany E Ayad
- Department of Anesthesia ICU and Pain, Cairo University, Cairo, 11566, Egypt
| | - Ossama H Salman
- Department of Anesthesiology, South Valley University, Qena, 83511, Egypt
| | | | | | - Adel M Mishriky
- Department of Community Medicine, Suez Canal University, Ismailia, 41511, Egypt
| | | | - Omar Viswanath
- Department of Anesthesiology, University of Arizona, Phoenix, AZ, 85003, USA
| | - Ivan Urits
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MS, USA
| | | | - John F Peppin
- College of Osteopathic Medicine, Pikeville University, c, KT, USA
| | | | | |
Collapse
|
21
|
Chaudhary S, Chaudhary S, Kumar M, Salhotra R. Fentanyl versus nalbuphine for intubating conditions during awake fiberoptic bronchoscopy: A randomized double-blind comparative study. J Anaesthesiol Clin Pharmacol 2021; 37:378-382. [PMID: 34759547 PMCID: PMC8562462 DOI: 10.4103/joacp.joacp_359_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background and Aims: Patient cooperation, sedation, anxiolysis, and topicalization are important prerequisites for the successful and safe conduct of awake intubation. Because of the pharmacological properties, opioids can facilitate this process. Fentanyl is an opioid agonist and nalbuphine is an agonist-antagonist. This study aims to compare these two opioids for their effect on sedation and intubating conditions during awake fiberoptic intubation. Material and Methods: This randomized double-blind controlled study was conducted on 62 ASA I/II patients of either sex between the age of 20 and 60 years, weight between 40 and 80 kg, with MP class I/II airways requiring general anesthesia with endotracheal intubation. All patients received standard airway topicalization and nebulization. Patients were randomly allocated to one of the two groups according to a computer-generated random number table. Group F (n = 31) received fentanyl 2 μg/kg i.v. and group N (n = 31) received nalbuphine 0.2 mg/kg i.v. over 10 min before intubation. Fiberoptic intubation was attempted and lignocaine spray and propofol boluses were administered as and when required. Hemodynamic responses and intubating conditions were recorded. Repeated measure ANOVA, McNemar test, and Chi-square test or Fischer's exact test were used for data analysis. A P < 0.05 was considered significant. Results: Cough score (P = 0.458), post-intubation score (P = 1.000), and sedation score (P = 1.000) were comparable among the two groups. Hemodynamic responses and propofol and lignocaine requirements were also comparable. Conclusion: Both fentanyl and nalbuphine provide comparable intubating conditions when used before awake fiberoptic intubation with minimal adverse effects on hemodynamic profile.
Collapse
Affiliation(s)
- Sujata Chaudhary
- Department of Anesthesiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Dilshad Garden, Delhi, India
| | - Sachin Chaudhary
- Department of Anesthesiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Dilshad Garden, Delhi, India
| | - Mahendra Kumar
- Department of Anesthesiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Dilshad Garden, Delhi, India
| | - Rashmi Salhotra
- Department of Anesthesiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Dilshad Garden, Delhi, India
| |
Collapse
|
22
|
Antipruritic Effect of Nalbuphine, a Kappa Opioid Receptor Agonist, in Mice: A Pan Antipruritic. Molecules 2021; 26:molecules26185517. [PMID: 34576988 PMCID: PMC8466557 DOI: 10.3390/molecules26185517] [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: 08/13/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Antipruritic effects of kappa opioid receptor (KOR) agonists have been shown in rodent models of acute and chronic scratching (itchlike behavior). Three KOR agonists, nalfurafine, difelikefalin, and nalbuphine, are in clinical studies for antipruritic effects in chronic itch of systemic and skin diseases. Nalfurafine (in Japan) and difelikefalin (in the USA) were approved to be used in the treatment of chronic itch in hemodialysis patients. The FDA-approved nalbuphine has been used in clinic for over 40 years, and it is the only narcotic agonist that is not scheduled. We aimed to study (a) antiscratch activity of nalbuphine against TAT-HIV-1 protein (controls HIV transcription)-, deoxycholic acid (DCA, bile acid)-, and chloroquine (CQ)-induced scratching in a mouse model of acute itch; and (b) whether the effect of nalbuphine is produced via KORs. First, dose-responses were developed for pruritogens. Mice were pretreated with nalbuphine (0.3-10 mg/kg) and then a submaximal dose of pruritogens were administered and the number of scratching bouts was counted. To study if the antiscratch effect of nalbuphine is produced via KOR, we used KOR knock out mice and pharmacologic inhibition of KORs using nor-binaltorphimine, a KOR antagonist. For this aim, we used CQ as a pruritogen. We found that: (a) TAT-HIV-1 protein elicits scratching in a dose-dependent manner; (b) nalbuphine inhibits scratching induced by TAT-HIV-1, DCA, and CQ dose-dependently; and (c) nalbuphine inhibits scratching induced by CQ through KORs. In conclusion, nalbuphine inhibits scratching elicited by multiple pruritogens.
Collapse
|
23
|
Co-administration of nalbuphine attenuates the morphine-induced anxiety and dopaminergic alterations in morphine-withdrawn rats. Psychopharmacology (Berl) 2021; 238:1193-1211. [PMID: 33655408 DOI: 10.1007/s00213-021-05765-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The classical effects of exogenous opioids, such as morphine, are predominantly mediated through μ-opioid receptors. The chronic use of morphine induces anxiety-like behavior causing functional changes in the mesolimbic dopaminergic system. The mixed μ/κ-agonist, nalbuphine, used either as an analgesic or as an adjuvant with morphine, produces different and opposite effects. However, whether nalbuphine can be used to antagonize morphine-induced anxiety and dopaminergic alterations is not fully known. OBJECTIVE This study aimed to compare acute and chronic effects of nalbuphine on morphine-induced anxiety and dopaminergic alterations in rats. METHODS Male adult Wistar albino rats were made opioid-dependent by administering increasing doses of morphine (5-25 mg/kg; i.p.; b.i.d.). Withdrawal was induced by naloxone (1 mg/kg, i.p.), 4 h after the last morphine injection. Anxiety-like behavior was measured using Activity Monitor (Coulbourn Instruments, Inc. USA). Thereafter, the animals were sacrificed and the brain dissected out and the level of cAMP and the transcriptional and translational expression of TH was measured. Nalbuphine was co-administered with morphine, acutely and chronically, at various doses (0.1, 0.3, 1.0, 3.0 mg/kg, i.p.). RESULTS Morphine-dependent rats showed a significant higher anxiety and cAMP levels and a significant decrease in the expression of TH. Co-administration of chronic doses of nalbuphine attenuates the higher anxiety, cAMP levels, and upregulates the TH expressions; however, the acute nalbuphine treatment does not attenuate the morphine-induced side effects. CONCLUSION Therefore, nalbuphine might have an important role in attenuating the anxiety and the effects of the dopaminergic pathway and may have potential in the treatment of opioid addiction.
Collapse
|
24
|
Khanna K, Sharma N, Rawat S, Khan N, Karwasra R, Hasan N, Kumar A, Jain GK, Nishad DK, Khanna S, Popli H, Bhatnagar A. Intranasal solid lipid nanoparticles for management of pain: A full factorial design approach, characterization & Gamma Scintigraphy. Chem Phys Lipids 2021; 236:105060. [PMID: 33582127 DOI: 10.1016/j.chemphyslip.2021.105060] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 01/03/2023]
Abstract
Pain is a noxious stimulus caused due to tissue damage and varies from mild to severe. Nalbuphine (NLB) is an approved, inexpensive, non-controlled, opioid agonist/antagonist analgesic used worldwide in various clinical settings for pain management. The current study aims to formulate NLB loaded solid lipid nanoparticles (SLNs) using solvent injection technology. The morphological and chemical structure of the developed SLNs were characterized using Field Emission Scanning Electron Microscopy (FESEM), Transmission Electron Microscopy (TEM) and Fourier Transformation Infrared Spectroscopy (FTIR). The results revealed from the point prediction confirmation in design expert software was the formulation of NLB-SLNs with an average particle size of (170.07 ± 25.1 nm), encapsulation efficiency (93.6 ± 1.5%) & loading capacity of 26.67%. The in-vitro permeation of developed NLB-SLNs was observed to be 94.18% at 8 h when compared with NLB solution whose maximum permeation was seen within 3 h of application. Efficacy of the formulation was also evaluated using eddy's hot plate method, where the onset of action started within 10 min of administration, and the maximum effect was observed at 1 h. The NLB-SLNs was screened for cytotoxicity in human embryonic kidney cells (HEK-293), and the dosage was considered safe when administered intranasally in animal since no detectable effect to the brain was observed. Biodistribution and gamma scintigraphy study of NLB-SLNs showed the prepared formulation reaching the target site, i.e. brain and was retained. Conclusively, the prepared NLB-SLNs formulation was safe and effective in producing an analgesic effect in vivo.
Collapse
Affiliation(s)
- Kushagra Khanna
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Delhi, India; Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Nitin Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, NH-58, Delhi Roorkee Highway, Baghpat Bypass Crossing, Meerut, 250005, India
| | - Sonalika Rawat
- Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Nazia Khan
- School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, India
| | - Ritu Karwasra
- Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Nazeer Hasan
- School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, India
| | - Abhishek Kumar
- Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Delhi, India
| | - Dhruv Kumar Nishad
- Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Sakshum Khanna
- Department of Solar Energy, Pandit Deendayal Energy University, Gandhinagar, Gujarat, India
| | - Harvinder Popli
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Delhi, India.
| | - Aseem Bhatnagar
- Institute of Nuclear Medicine and Allied Sciences (INMAS) Defence Research and Development Organisation, Timarpur, Delhi, India.
| |
Collapse
|
25
|
Uprety R, Che T, Zaidi SA, Grinnell SG, Varga BR, Faouzi A, Slocum ST, Allaoa A, Varadi A, Nelson M, Bernhard SM, Kulko E, Le Rouzic V, Eans SO, Simons CA, Hunkele A, Subrath J, Pan YX, Javitch JA, McLaughlin JP, Roth BL, Pasternak GW, Katritch V, Majumdar S. Controlling opioid receptor functional selectivity by targeting distinct subpockets of the orthosteric site. eLife 2021; 10:e56519. [PMID: 33555255 PMCID: PMC7909954 DOI: 10.7554/elife.56519] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 02/07/2021] [Indexed: 12/12/2022] Open
Abstract
Controlling receptor functional selectivity profiles for opioid receptors is a promising approach for discovering safer analgesics; however, the structural determinants conferring functional selectivity are not well understood. Here, we used crystal structures of opioid receptors, including the recently solved active state kappa opioid complex with MP1104, to rationally design novel mixed mu (MOR) and kappa (KOR) opioid receptor agonists with reduced arrestin signaling. Analysis of structure-activity relationships for new MP1104 analogs points to a region between transmembrane 5 (TM5) and extracellular loop (ECL2) as key for modulation of arrestin recruitment to both MOR and KOR. The lead compounds, MP1207 and MP1208, displayed MOR/KOR Gi-partial agonism with diminished arrestin signaling, showed efficient analgesia with attenuated liabilities, including respiratory depression and conditioned place preference and aversion in mice. The findings validate a novel structure-inspired paradigm for achieving beneficial in vivo profiles for analgesia through different mechanisms that include bias, partial agonism, and dual MOR/KOR agonism.
Collapse
Affiliation(s)
- Rajendra Uprety
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Tao Che
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of MedicineSt. LouisUnited States
- Department of Anesthesiology, Washington University in St. Louis School of MedicineSt. LouisUnited States
| | - Saheem A Zaidi
- Department of Quantitative and Computational Biology, Department of Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern CaliforniaLos AngelesUnited States
| | - Steven G Grinnell
- Division of Molecular Therapeutics, New York State Psychiatric Institute and Departments of Psychiatry, Pharmacology, Columbia University Vagelos College of Physicians & SurgeonsNew YorkUnited States
| | - Balázs R Varga
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of MedicineSt. LouisUnited States
- Department of Anesthesiology, Washington University in St. Louis School of MedicineSt. LouisUnited States
| | - Abdelfattah Faouzi
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of MedicineSt. LouisUnited States
- Department of Anesthesiology, Washington University in St. Louis School of MedicineSt. LouisUnited States
| | - Samuel T Slocum
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
| | - Abdullah Allaoa
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - András Varadi
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Melissa Nelson
- Division of Molecular Therapeutics, New York State Psychiatric Institute and Departments of Psychiatry, Pharmacology, Columbia University Vagelos College of Physicians & SurgeonsNew YorkUnited States
| | - Sarah M Bernhard
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of MedicineSt. LouisUnited States
| | - Elizaveta Kulko
- Division of Molecular Therapeutics, New York State Psychiatric Institute and Departments of Psychiatry, Pharmacology, Columbia University Vagelos College of Physicians & SurgeonsNew YorkUnited States
| | - Valerie Le Rouzic
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Shainnel O Eans
- Department of Pharmacodynamics, University of FloridaGainesvilleUnited States
| | - Chloe A Simons
- Department of Pharmacodynamics, University of FloridaGainesvilleUnited States
| | - Amanda Hunkele
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Joan Subrath
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Ying Xian Pan
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Department of Anesthesiology, Rutgers New Jersey Medical School, New JerseyNewarkUnited States
| | - Jonathan A Javitch
- Division of Molecular Therapeutics, New York State Psychiatric Institute and Departments of Psychiatry, Pharmacology, Columbia University Vagelos College of Physicians & SurgeonsNew YorkUnited States
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of FloridaGainesvilleUnited States
| | - Bryan L Roth
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
| | - Gavril W Pasternak
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, Department of Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern CaliforniaLos AngelesUnited States
| | - Susruta Majumdar
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of MedicineSt. LouisUnited States
- Department of Anesthesiology, Washington University in St. Louis School of MedicineSt. LouisUnited States
| |
Collapse
|
26
|
Chen JW, Tian L, Zhang L, Hu JY, Huang SQ, Jiao J. Nalbuphine on postoperative gastrointestinal tract dysfunction after laparoscopic surgery for gynaecological malignancies: A randomized controlled trial. Clin Exp Pharmacol Physiol 2021; 48:170-176. [PMID: 33063882 DOI: 10.1111/1440-1681.13421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 11/30/2022]
Abstract
The aim of this study was to compare the effects of nalbuphine and sufentanil on the gastrointestinal (GI) tract after laparoscopic surgery for gynaecological malignancies. A total of 100 patients with American Society of Anesthesiologists (ASA) physical status I-II undergoing laparoscopic radical hysterectomy under general anaesthesia were enrolled. The patients were randomized to receive sufentanil (Group S) or nalbuphine (Group N) intraoperatively and postoperatively. The time to first passage of flatus, the time to first defaecation, the time to toleration of diet, the serum gastrin level, and the duration of hospital stay of the groups were compared. The Visual Analogue Scale (VAS) score for postoperative pain, the number of dispensed patient-controlled analgaesics (PCAs), and the prevalence of postoperative nausea, vomiting, and dizziness of the groups were also compared. The time to first passage of flatus (P = .551), time to first defaecation (P = .310), time to toleration of diet (P = .182), serum gastrin level (P = .397), prevalence of postoperative nausea (P = .920) and vomiting (P = .334), number of dispensed PCAs (P = .167), and the duration of hospital stay (P = .482) of the two groups were not significantly different. The VAS scores at 6 hours (P = .008), 12 hours (P = .002), and 24 hours (P = .013) postoperatively were lower in Group N than in Group S. In conclusion, nalbuphine was not associated with improved postoperative GI dysfunction after laparoscopic surgery for gynaecological malignancies, but it was associated with reduced postoperative pain.
Collapse
Affiliation(s)
- Jia-Wei Chen
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lin Tian
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Le Zhang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jian-Ying Hu
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jing Jiao
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Lipman ZM, Yosipovitch G. An evaluation of difelikefalin as a treatment option for moderate-to-severe pruritus in end stage renal disease. Expert Opin Pharmacother 2020; 22:549-555. [PMID: 33190563 DOI: 10.1080/14656566.2020.1849142] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Introduction: Chronic kidney disease-associated pruritus (CKD-aP), or uremic pruritus, is a severely distressing condition that occurs in greater than 60% of patients undergoing dialysis. However, there are currently no FDA approved treatments for CKD-aP in the United States or Europe. Difelikefalin (DFK) is a kappa opioid receptor agonist with limited central nervous system (CNS) penetration that aims to fill this void by effectively and safely reducing itch in these patients.Areas covered: Through a review of the current literature (using PubMed and Google Scholar keyword searches of difelikefalin, CR845, pruritus, itch, opioids, hemodialysis, chronic kidney disease, uremic pruritus), the authors review DFK's mechanism of action and use published clinical trial data to evaluate its effectiveness in treating CKD-aP both individually and comparatively to other treatment alternatives.Expert opinion: DFK's IV formulation seems to provide safe, rapid-acting and effective itch reduction in hemodialysis patients without many of the negative mu opioid receptor (MOR)- or CNS- related side effects or drug-drug interactions of other currently available opioids. Its administration through IV bolus immediately after dialysis sessions at dialysis centers also increases availability to and ease of drug scheduling for this target population.
Collapse
Affiliation(s)
- Zoe M Lipman
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| | - Gil Yosipovitch
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| |
Collapse
|
28
|
Chen F, Wang CY, Zhang J, Wang F, Zhang M, Gu H, Song X, Chen J, Li Y, Cai YH, Li J, Lian QQ, Wu J, Liu HC. Comparison of Postoperative Analgesic Effects Between Nalbuphine and Fentanyl in Children Undergoing Adenotonsillectomy: A Prospective, Randomized, Double-Blind, Multicenter Study. Front Pharmacol 2020; 11:597550. [PMID: 33536911 PMCID: PMC7849154 DOI: 10.3389/fphar.2020.597550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Objective: There is no universal agreement on optimal pharmacological regimens for pain management during surgeries. The aim of this study to compare the postoperative analgesic effects of nalbuphine with fentanyl in children undergoing adenotonsillectomy. Design, Setting, Participants: We conducted a prospective, randomized, double-blind, non-inferiority and multicenter trial in 311 patients admitted to four different medical facilities in China from October 2017 to November 2018. Main Outcome Measure: The primary outcome was postoperative pain score. The secondary outcomes were as follows: the numbers of patients who developed moderate or severe pain (FLACC ≥4 points); time to first rescue analgesic top up and the actual number of rescue pain medicine given in pain control in post-anesthesia care unit (PACU), and additional analgesics requirement (received ≥2 rescue analgesics or/and other analgesics except study medications administered in PACU and ward); emergence and extubation time; Waking up time; time of PACU stay, and other side effects (desaturation, nausea/vomiting etc.). Results: A total of 356 children were screened and 322 patients were randomized. The mean age was 5.8 (5.5, 6.1) in the nalbuphine group and 5.6 (5.3, 5.8) in the fentanyl group (p = 0.2132). FLACC score of nalbuphine group was lower than that of fentanyl group upon patients' arrival at PACU (p < 0.05). The time to first required rescue dose of pain drug for nalbuphine group was longer than for the fentanyl group (2.5 vs 1.2 h, p < 0.0001). Only one patient (0.6%) in nalbuphine group presented a slow respiratory rate (RR) at 9/min while 29 patients (18.5%) in fentanyl group developed slow RR ≤10/min in PACU. Meanwhile, SpO2 was lower in the fentanyl group at 10 min after patients’ arrival in PACU (p < 0.05). The other profiles observed from these two drug groups were similar. Conclusion: Nalbuphine provided better pain relief with minimal respiration depression than fentanyl in children undergoing Adenotonsillectomy.
Collapse
Affiliation(s)
- Fang Chen
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Cheng-Yu Wang
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jianmin Zhang
- Department of Anesthesiology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Fang Wang
- Department of Anesthesiology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Mazhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, China
| | - Hongbin Gu
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Jia Chen
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yang Li
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yu-Hang Cai
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jun Li
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qing-Quan Lian
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Junzheng Wu
- Department of Anesthesiology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Hua-Cheng Liu
- Key Laboratory of Anesthesiology of Zhejiang Province, Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
29
|
Paton KF, Atigari DV, Kaska S, Prisinzano T, Kivell BM. Strategies for Developing κ Opioid Receptor Agonists for the Treatment of Pain with Fewer Side Effects. J Pharmacol Exp Ther 2020; 375:332-348. [PMID: 32913006 PMCID: PMC7589957 DOI: 10.1124/jpet.120.000134] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
There is significant need to find effective, nonaddictive pain medications. κ Opioid receptor (KOPr) agonists have been studied for decades but have recently received increased attention because of their analgesic effects and lack of abuse potential. However, a range of side effects have limited the clinical development of these drugs. There are several strategies currently used to develop safer and more effective KOPr agonists. These strategies include identifying G-protein-biased agonists, developing peripherally restricted KOPr agonists without centrally mediated side effects, and developing mixed opioid agonists, which target multiple receptors at specific ratios to balance side-effect profiles and reduce tolerance. Here, we review the latest developments in research related to KOPr agonists for the treatment of pain. SIGNIFICANCE STATEMENT: This review discusses strategies for developing safer κ opioid receptor (KOPr) agonists with therapeutic potential for the treatment of pain. Although one strategy is to modify selective KOPr agonists to create peripherally restricted or G-protein-biased structures, another approach is to combine KOPr agonists with μ, δ, or nociceptin opioid receptor activation to obtain mixed opioid receptor agonists, therefore negating the adverse effects and retaining the therapeutic effect.
Collapse
Affiliation(s)
- Kelly F Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Diana V Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Sophia Kaska
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Thomas Prisinzano
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| |
Collapse
|
30
|
Tavana T, Rezvani AR, Karimi-Maleh H. Pt-Pd-doped NiO nanoparticle decorated at single-wall carbon nanotubes: An excellent, powerful electrocatalyst for the fabrication of An electrochemical sensor to determine nalbuphine in the presence of tramadol as two opioid analgesic drugs. J Pharm Biomed Anal 2020; 189:113397. [PMID: 32563934 DOI: 10.1016/j.jpba.2020.113397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/12/2023]
Abstract
In this study, a Pt-Pd-doped NiO nanoparticle decorated at the surface of single-wall carbon nanotubes (Pt-Pd/NiO-NPs/SWCNTs) was synthesized using a simple chemical precipitation method and characterized by XRD, TEM, and EDS methods. The results confirmed that Pt-Pd/NiO-NPs/SWCNTs were synthesized with good purity and at the nanoscale size. Moreover, a highly sensitive electroanalytical sensor was fabricated by incorporating synthesized Pt-Pd/NiO-NPs/SWCNT nanocomposites into a carbon paste electrode (CPE) in the presence of 1-ethyl-3-methylimidazolium methanesulfonate (EMICH3SO3-) as binder. The Pt-Pd/NiO-NPs/SWCNTs/EMICH3SO3-/CPE showed a powerful electro-catalytic activity for electro-oxidation of nalbuphine, and the results confirmed that the oxidation of nalbuphine was improved 6.34 times and relative oxidation potential was decreased about 110 mV compared to unmodified electrodes. The Pt-Pd/NiO-NPs/SWCNTs/EMICH3SO3-/CPE also showed good catalytic activity for the determination of nalbuphine in the presence of tramadol and the oxidation potential of these opioid analgesic drugs separated with ΔE =460 mV. In the final step, the Pt-Pd/NiO-NPs/SWCNTs/EMICH3SO3-/CPE was used to determine nalbuphine with a detection limit of 0.9 nM and tramadol with a detection limit of 50.0 nM in drug samples. The results confirmed the powerful and interesting ability of the sensor in the analysis of a real sample.
Collapse
Affiliation(s)
- Toktam Tavana
- Department of Chemistry, University of Sistan and Baluchestan, Iran
| | - Ali Reza Rezvani
- Department of Chemistry, University of Sistan and Baluchestan, Iran.
| | - Hassan Karimi-Maleh
- Nanostructure Based Biosensors Research Group, Ton Duc Thang University, Ho Chi Minh City, Vietnam; Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
31
|
Brice‐Tutt AC, Wilson LL, Eans SO, Stacy HM, Simons CA, Simpson GG, Coleman JS, Ferracane MJ, Aldrich JV, McLaughlin JP. Multifunctional opioid receptor agonism and antagonism by a novel macrocyclic tetrapeptide prevents reinstatement of morphine-seeking behaviour. Br J Pharmacol 2020; 177:4209-4222. [PMID: 32562259 PMCID: PMC7443475 DOI: 10.1111/bph.15165] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The macrocyclic tetrapeptide natural product CJ-15,208 (cyclo[Phe-d-Pro-Phe-Trp]) is a multifunctional μ-opioid receptor and κ-opioid receptor agonist and κ-opioid receptor antagonist that produces antinociception and prevents stress-induced reinstatement of extinguished cocaine-conditioned place preference (CPP). We hypothesized that an analogue of CJ-15,208, cyclo[Pro-Sar-Phe-d-Phe], would demonstrate multifunctional μ-opioid receptor and κ-opioid receptor ligand activity, producing potent antinociception with fewer liabilities than selective μ-opioid receptor agonists, while preventing both drug- and stress-induced reinstatement of morphine-induced CPP. EXPERIMENTAL APPROACH The opioid receptor agonist and antagonist activity of cyclo[Pro-Sar-Phe-d-Phe] was characterized after i.c.v. and i.p. administration to C57BL/6J or transgenic opioid receptor "knockout" mice using the 55°C warm-water tail-withdrawal assay. Liabilities of locomotor coordination, respiration and spontaneous ambulation, and direct rewarding or aversive properties were assessed. Finally, the ability of cyclo[Pro-Sar-Phe-d-Phe] to block morphine- and stress-induced reinstatement of extinguished CPP was determined. KEY RESULTS cyclo[Pro-Sar-Phe-d-Phe] demonstrated dose-dependent, short-lasting antinociception, with an ED50 (and 95% confidence interval) of 0.15 (0.05-0.21) nmol i.c.v. and 1.91 (0.40-3.54) mg·kg-1 i.p., mediated by μ- and κ-opioid receptors. The macrocyclic tetrapeptide also demonstrated potent dose-dependent κ-opioid receptor antagonist-like activity at 2.5, but not at 4.5, h after administration. cyclo[Pro-Sar-Phe-d-Phe] displayed reduced liabiities compared with morphine, attributed to its additional activity at κ-receptors. Pretreatment with cyclo[Pro-Sar-Phe-d-Phe] prevented stress- and drug-induced reinstatement of extinguished morphine-place preference responses in a time-dependent manner. CONCLUSIONS AND IMPLICATIONS These data suggest that cyclo[Pro-Sar-Phe-d-Phe] is a promising lead compound for both the treatment of pain with reduced sideeffects and preventing both drug- and stress-induced relapse in morphine-abstinent subjects.
Collapse
Affiliation(s)
- Ariana C. Brice‐Tutt
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Lisa L. Wilson
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Chloe A. Simons
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Grant G. Simpson
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jeremy S. Coleman
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Michael J. Ferracane
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
32
|
Wu L, Yu J, Wang Q, Lu Y. Effects of nalbuphine on the cardioprotective effect of morphine in rats. Int J Cardiol 2020; 322:207-210. [PMID: 32828962 DOI: 10.1016/j.ijcard.2020.08.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/03/2020] [Accepted: 08/17/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND When combined with morphine, nalbuphine not only does not affect the analgesic effect but also prevents opioid-induced side effects. The authors investigated whether nalbuphine interferes with morphine-induced cardioprotection in rats. METHODS Anesthetized male Sprague-Dawley rats were randomly assigned to 1 of 4 treatment groups. Nalbuphine (NAL, 1 mg/kg) and morphine (MOR, 0.3 mg/kg) were administered 10 and 5 min prior to myocardial ischemia, respectively. Additionally, the NAL + MOR group received the combination of NAL and MOR prior to myocardial ischemia. An in vivo animal model was established by occluding the left anterior descending artery for 30 min and reperfusing it for 2 h. After 2 h of reperfusion, the infarcted area of heart was measured by Evans blue/triphenyl tetrazolium staining, and the levels of creatine kinase isoenzymes (CK-MB) in serum were detected by enzyme-linked immunosorbent assay. RESULTS Nalbuphine had no protective effect against the infarct area compared with the control treatment (NAL, 52.5 ± 5% versus CON, 52.6 ± 4%; *P < 0.01), and the infarct size-sparing effects of morphine were not affected by nalbuphine (NAL + MOR, 42.6 ± 7% versus MOR, 40.4 ± 3%; P > 0.05). The nalbuphine group did not show a change the levels of serum CK-MB compared with the control group, and nalbuphine did not affect the levels of serum CK-MB in the MOR group. CONCLUSIONS Nalbuphine does not interfere with the cardioprotective effect of morphine in vivo. Therefore, nalbuphine could be safely used or combined with morphine in patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Lining Wu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230061, PR China; Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Junma Yu
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230061, PR China
| | - Qiuyue Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230061, PR China
| | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230061, PR China.
| |
Collapse
|
33
|
Hu J, Chen S, Zhu M, Wu Y, Wang P, Chen J, Zhang Y. Preemptive Nalbuphine Attenuates Remifentanil-Induced Postoperative Hyperalgesia After Laparoscopic Cholecystectomy: A Prospective Randomized Double-Blind Clinical Trial. J Pain Res 2020; 13:1915-1924. [PMID: 32801849 PMCID: PMC7395683 DOI: 10.2147/jpr.s257018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
Background Remifentanil-induced hyperalgesia (RIH) is a paradoxical phenomenon that may increase sensitivity to painful stimuli. Nalbuphine, which is both a μ-receptor antagonist and κ-receptor agonist, may affect RIH. The aim of this study was to evaluate the effects of nalbuphine on RIH during laparoscopic cholecystectomy. Methods A total of 96 patients were divided into the following four groups: 0.4 μg/kg/min of remifentanil with 0.2 mg/kg of nalbuphine (HRNA), 0.4 μg/kg/min of remifentanil with saline (HRSA), 0.1 μg/kg/min of remifentanil with 0.2 mg/kg of nalbuphine (LRNA), and 0.1 μg/kg/min of remifentanil with saline (LRSA). The pain thresholds of postoperative mechanical hyperalgesia were measured with von Frey filaments. Pain intensity and analgesic consumption were recorded up to 48 h after surgery. Results Pain thresholds on the inner forearm decreased in the HRSA group compared with the HRNA (P = 0.0167), LRNA (P = 0.0027), and LRSA (P = 0.0318) groups at 24 h after surgery. Pain thresholds on the peri-incisional area decreased in the HRSA group compared with HRNA, LRNA, and LRSA (all P < 0.0001) groups at 24 h after surgery. Patients in the HRNA group showed lower numeric rating scale scores at 1 h (P = 0.0159), 3 h (P = 0.0118), 6 h (P = 0.0213), and 12 h (P = 0.0118) than those in the HRSA group. Postoperative requirement for sufentanil was greater in the HRSA group than the HRNA group during the first 3 h (P = 0.0321) and second 3 h (P = 0.0040). Postoperative sufentanil consumption was also greater in the LRSA group than in the LRNA group during the first 3 h (P = 0.0321) and second 3 h (P = 0.0416). Conclusion Preemptive nalbuphine can ameliorate postoperative hyperalgesia induced by high-dose remifentanil in patients undergoing laparoscopic cholecystectomy.
Collapse
Affiliation(s)
- Jun Hu
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Shuangshuang Chen
- Department of Anesthesiology, Tongling People's Hospital of Anhui Medical University, Tongling, Anhui, People's Republic of China
| | - Mudan Zhu
- Department of Anesthesiology, Tongling People's Hospital of Anhui Medical University, Tongling, Anhui, People's Republic of China
| | - Yun Wu
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ping Wang
- Department of Anesthesiology, Tongling People's Hospital of Anhui Medical University, Tongling, Anhui, People's Republic of China
| | - Jinbao Chen
- Department of Anesthesiology, Tongling People's Hospital of Anhui Medical University, Tongling, Anhui, People's Republic of China
| | - Ye Zhang
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
34
|
Blomqvist KJ, Dudek KA, Viisanen H, Mätlik K, Ahlström FHG, Laitila J, Kalso EA, Rauhala PV, Lilius TO. Antagonism of peripheral opioid receptors by methylnaltrexone does not prevent morphine tolerance in rats. J Neurosci Res 2020; 100:329-338. [PMID: 32459013 DOI: 10.1002/jnr.24638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 11/10/2022]
Abstract
Opioids are effective analgesics in the management of severe pain. However, tolerance, leading to dose escalation and adverse effects are significant limiting factors in their use. The role of peripheral opioid receptors in analgesia has been discussed especially under inflammatory conditions. The results from pharmacological and conditional knockout studies together do not provide a clear picture of the contribution of peripheral opioid receptors on antinociceptive tolerance and this needs to be evaluated. Therefore, we studied whether the peripherally restricted opioid receptor antagonist, methylnaltrexone (MNTX), could prevent morphine tolerance without attenuating the antinociceptive effect of morphine. Male Sprague-Dawley rats were treated for 7 days with increasing subcutaneous doses of morphine (5-30 mg/kg) and were coadministered saline, MNTX (0.5 or 2 mg/kg), or naltrexone (NTX; 2 mg/kg). Nociception was assessed with tail-flick, hotplate, and von Frey tests. Morphine, MNTX, and NTX concentrations in the plasma, brain, and spinal cord were measured by liquid chromatography-tandem mass spectrometry. In acute coadministration, NTX, but not MNTX, abolished the acute antinociceptive effects of morphine in all nociceptive tests. The antinociceptive tolerance after repeated morphine administration was also prevented by NTX but not by MNTX. MNTX penetrated to the spinal cord and the brain to some extent after repeated administration. The results do not support the use of MNTX for preventing opioid tolerance and also suggest that morphine tolerance is mediated by central rather than peripheral opioid receptors in the rat.
Collapse
Affiliation(s)
- Kim Juhani Blomqvist
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katarzyna Anna Dudek
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanna Viisanen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kert Mätlik
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fredrik Harry Gustav Ahlström
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jouko Laitila
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Finland
| | - Eija Anneli Kalso
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Anaesthesiology, Intensive Care Medicine, and Pain Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Pekka Veli Rauhala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuomas Olavi Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Finland.,Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Medland NA, Banys P, Baton VP, Nadela J, Maher L, Tac-An I. Responding to expanding HIV epidemics in Cebu, Philippines. Lancet HIV 2020; 7:e380-e381. [PMID: 31954437 DOI: 10.1016/s2352-3018(19)30405-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/27/2019] [Accepted: 11/22/2019] [Indexed: 11/17/2022]
Affiliation(s)
| | - Peter Banys
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | | | | | - Lisa Maher
- Kirby Institute, University of NSW, Sydney, 2052 NSW, Australia
| | - Ilya Tac-An
- Cebu City Health Department, Cebu, Philippines
| |
Collapse
|
36
|
Liang RJ, Shih YN, Chen YL, Liu WY, Yang WL, Lee SY, Wang HJ. A dual system platform for drug metabolism: Nalbuphine as a model compound. Eur J Pharm Sci 2020; 141:105093. [DOI: 10.1016/j.ejps.2019.105093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/26/2019] [Accepted: 09/28/2019] [Indexed: 01/26/2023]
|
37
|
Antipruritic Effects of Kappa Opioid Receptor Agonists: Evidence from Rodents to Humans. Handb Exp Pharmacol 2020; 271:275-292. [PMID: 33296031 DOI: 10.1007/164_2020_420] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Centrally administered bombesin induces scratching and grooming in rats. These behaviors were blocked by early benzomorphan kappa opioid receptor (KOR) agonists as reported by Gmerek and Cowan in 1984. This was the first evidence that KORs may be involved in the sensation of itch-like behaviors. Subsequent development of additional animal models for acute and chronic itch has led to important discoveries since then. For example, it was found that (a) gastrin-releasing peptide (GRP), natriuretic polypeptide b and their cognate receptors are keys for the transmission of itch sensation at the spinal cord level, (b) dynorphins (Dyns), the endogenous KOR agonists, work as inhibitory neuromodulators of itch at the spinal cord level, (c) in a mouse model for acute itch, certain KOR antagonists elicit scratching, (d) in mouse models of acute or chronic itch, KOR agonists (e.g., U50,488, nalfurafine, CR 845, nalbuphine) suppress scratching induced by different pruritogens, and (e) nalfurafine, CR 845, and nalbuphine are in the clinic or in clinical trials for pruritus associated with chronic kidney disease and chronic liver disease, as well as pruritus in chronic skin diseases.
Collapse
|
38
|
|
39
|
Nalbuphine, a kappa opioid receptor agonist and mu opioid receptor antagonist attenuates pruritus, decreases IL-31, and increases IL-10 in mice with contact dermatitis. Eur J Pharmacol 2019; 864:172702. [PMID: 31568781 DOI: 10.1016/j.ejphar.2019.172702] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
Chronic itch is one of the disturbing symptoms of inflammatory skin diseases. Kappa opioid receptor agonists are effective in suppressing scratching in mice against different pruritogens. Nalbuphine, a nonscheduled kappa opioid receptor agonist and mu opioid receptor antagonist, has been in clinical use for post-operative pain management since the 1980s and recently has been in clinical trials for chronic itch of prurigo nodularis (https://www.trevitherapeutics.com/nalbuphine). We studied whether nalbuphine is effective against chronic scratching induced by rostral neck application of 1-fluoro-2,4-dinitrobenzene (DNFB), an accepted mouse model of contact dermatitis to study pruritoceptive itch. Mice were treated once a week with either saline or nalbuphine 20 min before the third, fifth, seventh, and ninth sensitizations with DNFB and the number of scratching bouts was counted for 30 min. Skin samples from the neck of mice at week 4 were used to measure protein levels and mRNA expressions of chemokines and cytokines. Different sets of mice were used to study sedation and anhedonic-like behavior of nalbuphine. We found that: nalbuphine (a) antagonized scratching in a dose- and time-dependent manner without affecting locomotion, b) decreased IL-31, and increased anti-inflammatory IL-10, and c) induced more elevations in the levels of CCL2, CCL3, CCL12, CXCL1, CXCL2, CXCL9, CXCL10, IL-1β, IL-16, TIMP-1, M-CSF, TREM-1 and M1-type macrophages compared to saline. Increases in chemokines and cytokines and M1 macrophages by nalbuphine suggest an inflammatory phase of healing in damaged skin due to scratching. Our data indicate that nalbuphine is an effective antipruritic in murine model of pruritoceptive itch.
Collapse
|
40
|
Liang RJ, Lai YH, Kao YT, Yang TH, Chen YL, Wang HJ. A novel finding of nalbuphine-6-glucuronide, an active opiate metabolite, possessing potent antinociceptive effects: Synthesis and biological evaluation. Eur J Med Chem 2019; 178:544-551. [PMID: 31212133 DOI: 10.1016/j.ejmech.2019.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/22/2019] [Accepted: 06/10/2019] [Indexed: 10/26/2022]
Abstract
Nalbuphine, a partial agonist/antagonist opioid analgesic, is structurally related to morphine. It is equipotent to morphine and has no serious side effects. In the past few decades, studies focusing on morphine metabolism have indicated that one of its sugar-conjugated metabolites, morphine-6-glucuronide, exerts a higher analgesic effect than its parent drug. Considering that nalbuphine is a morphine analog that follows a similar metabolic scheme, nalbuphine glucuronides were synthesized in this study and their potential analgesic effects were assessed. Nalbuphine-3-glucuronide (N3G) and nalbuphine-6-glucuronide (N6G) were synthesized based on Schmidt's glycosylation with OPiv protections on the glycosyl donor. In a pharmacodynamic study, paw pressure and cold-ethanol tail-flick tests were conducted in rats to evaluate the analgesic response after intracisternal and intraperitoneal administrations of nalbuphine, N3G, or N6G. The antinociceptive response was evaluated for each compound by calculating the area under the curve and the duration spent at greater than 50% maximum possible analgesia. In conclusion, intracisternal administration of N6G exhibited a stronger analgesic response than nalbuphine in the pain tests after both cold and mechanical stimuli, but N3G had no obvious effect. Similar to that of morphine, the glucuronide metabolite of nalbuphine at the 6-O-position exerted at least three-fold higher antinociceptive potency and five-fold longer analgesic duration than nalbuphine.
Collapse
Affiliation(s)
- Ren-Jong Liang
- Medical Supplies and Maintenance, Hualien Armed Forces General Hospital, Hualien, Taiwan, ROC
| | - Yen-Hsun Lai
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Yu-Ting Kao
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ting-Hsuan Yang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yen-Lun Chen
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hong-Jaan Wang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
41
|
Gomes VH, Barcellos MC, Lima VC, de Moura RA, de Freitas JB, da Silva MF. Effect of three doses of nalbuphine on reversal of sedation and cardiopulmonary effects of morphine-acepromazine in healthy dogs. Vet Anaesth Analg 2019; 46:429-434. [PMID: 31160169 DOI: 10.1016/j.vaa.2019.03.001] [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: 07/10/2018] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate the efficacy of three doses of nalbuphine in reversing sedative and cardiopulmonary effects of morphine-acepromazine in dogs. STUDY DESIGN Prospective, randomized experimental trial. ANIMALS A group of eight healthy Beagle dogs, aged 5-6 years and weighing 12.5 ± 2.1 kg. METHODS Dogs were administered morphine (0.5 mg kg-1) and acepromazine (0.05 mg kg-1) intravenously (IV). After 20 minutes, dogs were administered one of four treatments IV: saline (control); or nalbuphine (0.3 mg kg-1; treatment N0.3), (0.6 mg kg-1; treatment N0.6) or (1.0 mg kg-1; treatment N1.0), in random order separated by 1 week. Sedation was scored using a numeric descriptive scale (NDS) and simple numerical scale (SNS). Heart rate, systolic arterial pressure (SAP), respiratory rate (fR) and rectal temperature (RT) were recorded before (BL), 20 minutes after morphine-acepromazine (T0), then 10 (T10), 30, 60 and 90 minutes after saline or nalbuphine. Arterial blood gases were measured at BL, T0 and T10. Values were compared with BL, T0 and among treatments using anova (p < 0.05) and the Bonferroni correction (p < 0.008). RESULTS NDS for N0.6 and SNS for N0.6 and N1.0 at T30, and both scores for all nalbuphine treatments at T60-T90 were lower compared with T0 (p < 0.05). Sedation scores were not different among nalbuphine treatments. SNS scores were lower than control at T10 for N0.3 and N0.6 (p < 0.05). SAP and fR were lower than BL for all treatments at some time points (p < 0.05). RT was higher than control at T60 in the nalbuphine treatments (p < 0.001). PaO2 was lower in N0.3 at T0 compared with BL (p = 0.036). CONCLUSIONS AND CLINICAL RELEVANCE All nalbuphine doses decreased the degree of sedation, without differences among them. Administration of nalbuphine resulted in minimal changes in measured cardiopulmonary variables.
Collapse
Affiliation(s)
- Viviane H Gomes
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil.
| | - Myla Cb Barcellos
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil
| | - Virgínia Ct Lima
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil
| | - Raiane A de Moura
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil
| | - Jacqueline B de Freitas
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil
| | - Marta Fa da Silva
- Department of Veterinary Medicine and Surgery, Universidade Federal Rural do Rio de Janeiro, UFRRJ, Seropédica, RJ, Brazil
| |
Collapse
|
42
|
Walentiny DM, Moisa LT, Beardsley PM. Oxycodone-like discriminative stimulus effects of fentanyl-related emerging drugs of abuse in mice. Neuropharmacology 2019; 150:210-216. [PMID: 30735691 DOI: 10.1016/j.neuropharm.2019.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/15/2019] [Accepted: 02/04/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Fentanyl and its structurally related compounds have emerged as the most significant contributors to opioid overdose fatalities in recent years. While there is abundant information about the pharmacological effects of fentanyl, far less is known of its more recently abused analogs. The objective of this study was to determine whether fentanyl and several fentanyl-related substances would engender oxycodone-like responding in a mouse model of oxycodone discrimination. Oxycodone was selected as the training drug due to its high selectivity for mu opioid receptors. Compounds that elicited oxycodone-like responding in this procedure would likely evoke overlapping subjective experiences. METHODS Adult male C57BL/6 mice were trained to discriminate 1.3 mg/kg oxycodone from vehicle in a food-reinforced, two-lever choice procedure. Generalization tests were conducted with fentanyl and the following fentanyl-related compounds: ocfentanil, 3-furanyl fentanyl, crotonylfentanyl, and valerylfentanyl. RESULTS Fentanyl and each of its analogs completely generalized to the 1.3 mg/kg oxycodone discriminative stimulus and naltrexone pretreatment significantly decreased oxycodone-like responding for each compound. Rank order potency for engendering oxycodone-appropriate responding was ocfentanil > fentanyl > 3-furanyl fentanyl ≈ crotonylfentanyl > oxycodone > valerylfentanyl. Drug doses that evoked full substitution also significantly suppressed response rates compared to vehicle. CONCLUSIONS These results indicate that the discriminative stimulus, and by extension, the interoceptive and subjective effects of the tested fentanyl analogs, overlap with those of oxycodone. These observations consequentially support the prediction that they would also engender the likelihood for abuse similar to oxycodone. This article is part of the Special Issue entitled 'Opioid Neuropharmacology: Advances in treating pain and opioid addiction'.
Collapse
Affiliation(s)
- D Matthew Walentiny
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 1217 E. Marshall Street, Richmond, VA, 23298-0613, USA.
| | - Léa T Moisa
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 1217 E. Marshall Street, Richmond, VA, 23298-0613, USA
| | - Patrick M Beardsley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 1217 E. Marshall Street, Richmond, VA, 23298-0613, USA; Institute for Drug and Alcohol Studies & Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, 410 N. 12th Street, PO Box 980613, Richmond, VA, 23298-0613, USA
| |
Collapse
|
43
|
Chronic co-administration of nalbuphine attenuates the development of opioid dependence. Pharmacol Biochem Behav 2018; 175:130-138. [DOI: 10.1016/j.pbb.2018.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 09/29/2018] [Accepted: 10/01/2018] [Indexed: 01/21/2023]
|
44
|
Borsook D, Youssef AM, Simons L, Elman I, Eccleston C. When pain gets stuck: the evolution of pain chronification and treatment resistance. Pain 2018; 159:2421-2436. [PMID: 30234696 PMCID: PMC6240430 DOI: 10.1097/j.pain.0000000000001401] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
It is well-recognized that, despite similar pain characteristics, some people with chronic pain recover, whereas others do not. In this review, we discuss possible contributions and interactions of biological, social, and psychological perturbations that underlie the evolution of treatment-resistant chronic pain. Behavior and brain are intimately implicated in the production and maintenance of perception. Our understandings of potential mechanisms that produce or exacerbate persistent pain remain relatively unclear. We provide an overview of these interactions and how differences in relative contribution of dimensions such as stress, age, genetics, environment, and immune responsivity may produce different risk profiles for disease development, pain severity, and chronicity. We propose the concept of "stickiness" as a soubriquet for capturing the multiple influences on the persistence of pain and pain behavior, and their stubborn resistance to therapeutic intervention. We then focus on the neurobiology of reward and aversion to address how alterations in synaptic complexity, neural networks, and systems (eg, opioidergic and dopaminergic) may contribute to pain stickiness. Finally, we propose an integration of the neurobiological with what is known about environmental and social demands on pain behavior and explore treatment approaches based on the nature of the individual's vulnerability to or protection from allostatic load.
Collapse
Affiliation(s)
- David Borsook
- Center for Pain and the Brain, Boston Children’s (BCH), McLean and Massachusetts Hospitals (MGH), Boston MA
- Departments of Anesthesia (BCH), Psychiatry (MGH, McLean) and Radiology (MGH)
| | - Andrew M Youssef
- Center for Pain and the Brain, Boston Children’s (BCH), McLean and Massachusetts Hospitals (MGH), Boston MA
| | - Laura Simons
- Department of Anesthesia, Stanford University, Palo Alto, CA
| | | | - Christopher Eccleston
- Centre for Pain Research, University of Bath, UK
- Department of Clinical and Health Psychology, Ghent University, Belgium
| |
Collapse
|
45
|
Withey SL, Paronis CA, Bergman J. Concurrent Assessment of the Antinociceptive and Behaviorally Disruptive Effects of Opioids in Squirrel Monkeys. THE JOURNAL OF PAIN 2018; 19:728-740. [PMID: 29477761 DOI: 10.1016/j.jpain.2018.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 10/17/2022]
Abstract
Although the clinical application of opioids for pain management is often hindered by undesired behavioral impairment, preclinical assays of antinociception typically do not provide information regarding the behaviorally disruptive effects of opioids that may accompany their antinociceptive effects. To address this, we modified a warm water tail withdrawal procedure to determine concurrently the effects of opioids on tail withdrawal latency (antinociception) and indices of food-maintained operant behavior (rates of responding and reinforcement density) in squirrel monkeys. Six opioid agonists were tested, and all produced dose-dependent antinociception and impairment of operant behavior. The ratio of median effective dose (ED50) values for both measures (behavioral impairment:antinociception) was used as a quantitative measure of therapeutic index. Nalbuphine had the highest ED50 ratio (4.88), reflecting antinociception with minimal behavioral disruption. Oxycodone, heroin, buprenorphine, and methadone all produced similar ED50 ratios (.82-1.14), whereas butorphanol yielded a significantly lower ED50 ratio (.17) reflecting behavioral disruption at doses producing only minimal antinociception. The antinociceptive and behaviorally disruptive effects of oxycodone and buprenorphine were further characterized using Schild analysis to calculate apparent pA2 values for antagonism of the 2 drugs by naltrexone. These analyses suggest that µ-receptor mechanisms likely mediate the antinociceptive as well as behaviorally disruptive effects of oxycodone (pA2 values: 8.13 and 8.57) and buprenorphine (pA2 values: 8.6 and 7.9). PERSPECTIVE This article presents an assay that allows for the concurrent assessment of the antinociceptive and behaviorally disruptive effects of opioids. Our results show that the tail withdrawal assay in squirrel monkeys can provide a useful index of the behavioral selectivity with which opioids produce antinociception.
Collapse
Affiliation(s)
- Sarah L Withey
- Preclinical Pharmacology Laboratory, McLean Hospital/Harvard Medical School, Belmont, Massachusetts.
| | - Carol A Paronis
- Preclinical Pharmacology Laboratory, McLean Hospital/Harvard Medical School, Belmont, Massachusetts
| | - Jack Bergman
- Preclinical Pharmacology Laboratory, McLean Hospital/Harvard Medical School, Belmont, Massachusetts
| |
Collapse
|
46
|
Morris LS, Baek K, Tait R, Elliott R, Ersche KD, Flechais R, McGonigle J, Murphy A, Nestor LJ, Orban C, Passetti F, Paterson LM, Rabiner I, Reed L, Smith D, Suckling J, Taylor EM, Bullmore ET, Lingford-Hughes AR, Deakin B, Nutt DJ, Sahakian BJ, Robbins TW, Voon V. Naltrexone ameliorates functional network abnormalities in alcohol-dependent individuals. Addict Biol 2018; 23:425-436. [PMID: 28247526 PMCID: PMC5811832 DOI: 10.1111/adb.12503] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/03/2017] [Accepted: 02/05/2017] [Indexed: 01/30/2023]
Abstract
Naltrexone, an opioid receptor antagonist, is commonly used as a relapse prevention medication in alcohol and opiate addiction, but its efficacy and the mechanisms underpinning its clinical usefulness are not well characterized. In the current study, we examined the effects of 50‐mg naltrexone compared with placebo on neural network changes associated with substance dependence in 21 alcohol and 36 poly‐drug‐dependent individuals compared with 36 healthy volunteers. Graph theoretic and network‐based statistical analysis of resting‐state functional magnetic resonance imaging (MRI) data revealed that alcohol‐dependent subjects had reduced functional connectivity of a dispersed network compared with both poly‐drug‐dependent and healthy subjects. Higher local efficiency was observed in both patient groups, indicating clustered and segregated network topology and information processing. Naltrexone normalized heightened local efficiency of the neural network in alcohol‐dependent individuals, to the same levels as healthy volunteers. Naltrexone failed to have an effect on the local efficiency in abstinent poly‐substance‐dependent individuals. Across groups, local efficiency was associated with substance, but no alcohol exposure implicating local efficiency as a potential premorbid risk factor in alcohol use disorders that can be ameliorated by naltrexone. These findings suggest one possible mechanism for the clinical effects of naltrexone, namely, the amelioration of disrupted network topology.
Collapse
Affiliation(s)
- Laurel S. Morris
- Department of Psychology; University of Cambridge; UK
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
| | | | - Roger Tait
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | - Rebecca Elliott
- Neuroscience and Psychiatry Unit; University of Manchester; UK
| | - Karen D. Ersche
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | - Remy Flechais
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - John McGonigle
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Anna Murphy
- Neuroscience and Psychiatry Unit; University of Manchester; UK
| | - Liam J. Nestor
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Csaba Orban
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Filippo Passetti
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Louise M. Paterson
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | | | - Laurence Reed
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Dana Smith
- Department of Psychology; University of Cambridge; UK
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | - John Suckling
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | | | - Edward T. Bullmore
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | | | - Bill Deakin
- Neuroscience and Psychiatry Unit; University of Manchester; UK
| | - David J. Nutt
- Centre for Neuropsychopharmacology, Division of Brain Sciences; Imperial College London; UK
| | - Barbara J. Sahakian
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | - Trevor W. Robbins
- Department of Psychology; University of Cambridge; UK
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | - Valerie Voon
- Behavioural and Clinical Neuroscience Institute; University of Cambridge; UK
- Department of Psychiatry; University of Cambridge; UK
| | | |
Collapse
|
47
|
Sadafule NN, Karhade SS. Comparative Study of Efficacy of Preoperative Nalbuphine Hydrochloride and Pentazocine Lactate on Hemodynamic Response to Tracheal Intubation and Postoperative Analgesia. Anesth Essays Res 2018; 12:218-222. [PMID: 29628585 PMCID: PMC5872867 DOI: 10.4103/aer.aer_168_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: For General anaesthesia perioperative administration of opioids are choice of drugs to attenuate intubation response and to provide good intraoperative and postoperative analgesia. Mixed agonist-antagonist opioids produces powerful analgesia without undesirable side effects. Aim: Aim of present study to compare efficacy of Nalbuphine and Pentazocine on hemodynamic response to tracheal intubation and postoperative analgesia. Settings: Present study carried out in operation theater. Design: This is prospective randomized double blind controlled trial. Material and Methods: Patients of ASA physical status I and II, age 20-60 years undergoing abdominal surgical procedure of 1-2 hour duration were randomly divided into two groups of 30 patients each (Group N) receiving nalbuphine 0.3 mg.kg-1 and (Group P) receiving pentazocine 0.6 mg.kg-1 intravenously before induction. Baseline vital parameters were noted and then noted till 15 minutes after intubation to see hemodynamic response. Time of rescue analgesia required was noted postoperatively. Stastical Analysis: Chi-square test, paired and unpaired t-test were used for statistical analysis. Results: There is rise in heart rate, blood pressure after intubation in both groups but rise was more with Pentazocine as compared to Nalbuphine (P < 0.05). Postoperatively within 30 minutes 60% of patient from Pentazocine group require rescue analgesia compare to only 16.60% of patient of Nalbuphine group (P = 0.05) which is highly significant. Conclusion: Nalbuphine is potent mixed opioid analgesic which can be used for attenuation of pressor response of tracheal intubation and for perioperative analgesia with minimal side effects.
Collapse
Affiliation(s)
- Namrata Natraj Sadafule
- Department of Anaesthesiology and Critical care, SMT Kashibai Navale Medical College and General Hospital, Sinhgad Institute, Pune, Maharashtra, India
| | - Seema Shripad Karhade
- Department of Anaesthesiology and Critical care, SMT Kashibai Navale Medical College and General Hospital, Sinhgad Institute, Pune, Maharashtra, India
| |
Collapse
|
48
|
Li CJ, Ku MY, Lu CY, Tien YE, Chern WH, Huang JD. In vitro and in vivo release of dinalbuphine sebacate extended release formulation: Effect of the oil ratio on drug release. Int J Pharm 2017; 531:306-312. [DOI: 10.1016/j.ijpharm.2017.08.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/28/2017] [Accepted: 08/15/2017] [Indexed: 11/17/2022]
|
49
|
Tien YE, Huang WC, Kuo HY, Tai L, Uang YS, Chern WH, Huang JD. Pharmacokinetics of dinalbuphine sebacate and nalbuphine in human after intramuscular injection of dinalbuphine sebacate in an extended-release formulation. Biopharm Drug Dispos 2017; 38:494-497. [PMID: 28741675 DOI: 10.1002/bdd.2088] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/22/2017] [Accepted: 06/26/2017] [Indexed: 11/10/2022]
Abstract
Nalbuphine is a semi-synthetic opioid indicated for the relief of moderate to severe pain. Its short half-life requires frequent injections in clinical practice, resulting in a greater incidence of adverse events. A prodrug of nalbuphine has been developed, dinalbuphine sebacate (DNS), dissolved in a simple oil-based injectable formulation, which could deliver and maintain an effective blood level of nalbuphine. An open-label, prospective, two-period study was performed in healthy volunteers to verify the extended blood concentration profile of nalbuphine. Twelve healthy Taiwanese were randomized to receive an intramuscular injection of 20 mg nalbuphine HCl and 150 mg DNS sequentially with a washout period of 5 days. To prevent DNS hydrolysis during sample analysis, the effect of four esterase inhibitors was evaluated in the quantitation of DNS in human whole blood and thenoyltrifluoroacetone was chosen. The bioavailability of nalbuphine from intramuscularly injected DNS relative to that from nalbuphine HCl was 85.4%. The mean absorption time of nalbuphine from DNS was 145.2 h. It took approximately 6 days for the complete release of DNS into the blood stream where DNS was rapidly hydrolysed to nalbuphine; suggesting a single injection of 150 mg DNS in our extended-release formulation could provide long-lasting pain relief.
Collapse
Affiliation(s)
- Yu En Tien
- National Cheng Kung University, Clinical Pharmacy and Pharmaceutical Sciences, Tainan, Taiwan
| | | | | | - Lily Tai
- Rosetta Pharmamate Co., Ltd, Taipei, Taiwan
| | | | | | - Jin-Ding Huang
- National Cheng Kung University, Clinical Pharmacy and Pharmaceutical Sciences, Tainan, Taiwan
| |
Collapse
|
50
|
Li W, Long JD, Qian YY, Long Y, Xu XJ, Wang YJ, Shen Q, Wang ZN, Yang XC, Xiao L, Sun HP, Xu YL, Chen YY, Xie Q, Wang YH, Shao LM, Liu JG, Qiu ZB, Fu W. The Pharmacological Heterogeneity of Nepenthone Analogs in Conferring Highly Selective and Potent κ-Opioid Agonistic Activities. ACS Chem Neurosci 2017; 8:766-776. [PMID: 28033462 DOI: 10.1021/acschemneuro.6b00321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To develop novel analgesics with no side effects or less side effects than traditional opioids is highly demanded to treat opioid receptor mediated pain and addiction issues. Recently, κ-opioid receptor (KOR) has been established as an attractive target, although its selective agonists could bear heterogeneous pharmacological activities. In this study, we designed and synthesized two new series of nepenthone derivatives by inserting a spacer (carbonyl) between 6α,14α-endo-ethenylthebaine and the 7α-phenyl substitution of the skeleton and by substituting the 17-N-methyl group with a cyclopropylmethyl group. We performed in vitro tests (binding and functional assays) and molecular docking operations on our newly designed compounds. The results of wet-experimental measures and modeled binding structures demonstrate that these new compounds are selective KOR agonists with nanomolar level affinities. Compound 4 from these new derivatives showed the highest affinity (Ki = 0.4 ± 0.1 nM) and the highest selectivity (μ/κ = 339, δ/κ = 2034) toward KOR. The in vivo tests revealed that compound 4 is able to induce stronger (ED50 = 2.1 mg/kg) and much longer antinociceptive effect than that of the typical KOR agonist U50488H (ED50 = 4.4 mg/kg). Therefore, compound 4 can be used as a perfect lead compound for future design of potent analgesics acting through KOR.
Collapse
Affiliation(s)
- Wei Li
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jian-Dong Long
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, Shanghai 201203, China
| | - Yuan-Yuan Qian
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yu Long
- Department of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xue-Jun Xu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, Shanghai 201203, China
| | - Yu-Jun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, Shanghai 201203, China
| | - Qing Shen
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zuo-Neng Wang
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xi-Cheng Yang
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li Xiao
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hong-Peng Sun
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yu-Long Xu
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi-Yi Chen
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yong-Hui Wang
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li-Ming Shao
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jing-Gen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, Shanghai 201203, China
| | - Zhui-Bai Qiu
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Fu
- Department of Medicinal
Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|