1
|
Liu Y, Xu B, Cheng S, Wang Y, Ding J, Shen X, Wu B, Xu L, Wei J. Novel chimeric peptides of endomorphin-2 and the active fragments of ghrelin exhibit blood-brain barrier permeability and central antinociceptive effects with reduced opioid-related side effects. Neuropharmacology 2025; 269:110324. [PMID: 39904410 DOI: 10.1016/j.neuropharm.2025.110324] [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: 08/11/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Pharmacological research has showed that multi-targeted drug therapies offer superior efficacy and reduced side effects compared to single-target drug therapies. In this study, we designed and characterized four novel chimeric peptides G (1-5)-EM2, EM2-G (1-5), G (1-9)-EM2 and EM2-G (1-9) which incorporate endomorphin-2 (EM-2) and the active fragments of ghrelin. Calcium mobilization assays revealed that these four chimeric peptides acted as weak mixed agonists for the μ-opioid receptor (MOR), κ-opioid receptor (KOR), and growth hormone secretagogue receptor 1α (GHS-R1α). The results of fluorescence imaging experiments indicated that G (1-5)-EM2 and G (1-9)-EM2 could penetrate the blood-brain barrier (BBB) following intravenous (i.v.) injection. All chimeric peptides induced almost equal antinociceptive effects compared with EM-2 or better antinociceptive effects than EM-2 after intracerebroventricular (i.c.v.) injection in the acute pain in mice. Among them, G (1-5)-EM2 could cross the BBB and enter the brain to induce antinociceptive effect through central opioid receptors after i. v. Injection. Our findings demonstrated that the chimeric peptides produced significant antinociception mainly via MOR, DOR and GHS-R1α without inducing antinociceptive tolerance, or with a lower tendency for antinociceptive tolerance after i. c.v. Injection in the acute pain in mice. Furthermore, the chimeric peptides mitigated or eliminated the digestive side effects associated with EM-2. The collective results highlight G (1-5)-EM2 as the most promising candidate among the chimeric peptides. The chimeric peptides represent a promising class of potential analgesics for clinical pain management. However, further optimization is necessary to maximize their therapeutic potential.
Collapse
Affiliation(s)
- Yongling Liu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, PR China
| | - Songxia Cheng
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Yan Wang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Jiali Ding
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiaoyu Shen
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Bing Wu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Liangquan Xu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Jie Wei
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China; Jiangxi Province Key Laboratory of Brain Science and Brian Health, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
2
|
Malik A, Hosztafi S, Vincze A, Marton A, Kraszni M, Balogh GT, Noszál B, Mazák K. Characterization of Opioid Agonist Morphine Derivatives with Emphasis on Medicinal Chemistry. ChemMedChem 2025; 20:e202400654. [PMID: 39479891 PMCID: PMC11831872 DOI: 10.1002/cmdc.202400654] [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: 08/21/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/02/2024]
Abstract
Relieving severe pains is an unmet medical need, in which opioids are of prime importance and in the focus of pharmaceutical companies. The objective of this study is to characterize the physicochemical properties, namely basicity, lipophilicity and permeability of thirty opioid ligands, which include eleven newly synthesized compounds. pH-potentiometry and the shake-flask method were used for the characterization of species-specific basicity and lipophilicity. The effective permeability was determined using a brain-specific parallel artificial membrane permeability assay. Structural modifications, namely O-methylation in position 3, isomerization in position 6, saturation of the double bond in position 7, 14-hydroxylation, and the substitution of N-(β-phenylethyl) and N-cyclobutylmethyl side chains all have various effects on these physicochemical properties, and these are explained and compared to computationally predicted values. Computational predictions inadequately capture hydrogen bond formation with the tertiary amino group in case of 14-hydroxylation, just as the effects of hydroxy oxidation at position 6 and N-methyl substitution with N-(β-phenylethyl). The relationship between lipophilicity, permeability and potency is presented by lipophilic efficiency plots that reveal the most promising compounds. This study emphasizes the importance of experimental determination of these essential physicochemical parameters, furthermore, it can contribute to a more thorough understanding of the pharmacokinetic properties.
Collapse
Affiliation(s)
- Alaa Malik
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| | - Sándor Hosztafi
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| | - Anna Vincze
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| | - András Marton
- Department of Chemical and Environmental Process EngineeringFaculty of Chemical Technology and BiotechnologyBudapest University of Technology and EconomicsMűegyetem rkp. 3.H-1111BudapestHungary
| | - Márta Kraszni
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| | - György T. Balogh
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
- Department of Chemical and Environmental Process EngineeringFaculty of Chemical Technology and BiotechnologyBudapest University of Technology and EconomicsMűegyetem rkp. 3.H-1111BudapestHungary
| | - Béla Noszál
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| | - Károly Mazák
- Semmelweis UniversityDepartment of Pharmaceutical ChemistryHőgyes E. u. 9.H-1092BudapestHungary
- Center for Pharmacology and Drug Research & DevelopmentSemmelweis UniversityÜllői út 26H-1085BudapestHungary
| |
Collapse
|
3
|
Zhen G, Do N, Manh NV, Ha HJ, Kim H, Kim H, Choi K, Ann J, Lee J. Discovery of a Novel Multitarget Analgesic Through an In Vivo-Guided Approach. Pharmaceuticals (Basel) 2025; 18:205. [PMID: 40006019 PMCID: PMC11859264 DOI: 10.3390/ph18020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Pain is a complex condition influenced by peripheral, central, immune, and psychological factors. Multitarget approaches offer a more effective and safer alternative to single-target analgesics by enhancing efficacy, reducing side effects, and minimizing tolerance. This study aimed to identify a novel multitarget analgesic with improved pharmacological properties. Methods: An in vivo-guided screening approach was used to discover a new analgesic compound. Compound 29, derived from a novel scaffold inspired by opiranserin and vilazodone pharmacophores, was identified through analog screening in the formalin test. Its efficacy was further evaluated in the spinal nerve ligation (SNL) model of neuropathic pain. Mechanistic studies explored its interaction with neurotransmitter transporters and receptors, while pharmacokinetic and safety assessments were conducted to determine its stability, brain penetration, and potential toxicity. Results: Compound 29 demonstrated high potency in the formalin test, with an ED50 of 0.78 mg/kg in the second phase and a concentration-dependent effect in the first phase. In the SNL model, it produced dose-dependent analgesic effects, increasing withdrawal thresholds by 24% and 45% maximum possible effect (MPE) at 50 and 100 mg/kg, respectively. Mechanistic studies revealed strong triple uptake inhibition, particularly at dopamine (DAT) and serotonin (SERT) transporters, alongside high-affinity 5-HT2A receptor antagonism. Pharmacokinetic analysis indicated enhanced stability and blood-brain barrier permeability. In vitro studies confirmed its nontoxicity to HT-22 cells but revealed potential hERG inhibition and strong CYP3A4 inhibition. Conclusions: Compound 29 is a promising multitarget analgesic with potent efficacy and favorable pharmacokinetics. Ongoing optimization efforts aim to mitigate side effects and enhance its therapeutic profile for clinical application.
Collapse
Affiliation(s)
- Guo Zhen
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Nayeon Do
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Nguyen Van Manh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Hee-Jin Ha
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Hee Kim
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Hyunsoo Kim
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Kwanghyun Choi
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Jihyae Ann
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Jeewoo Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| |
Collapse
|
4
|
Wu B, Cheng S, Liu F, Wei J, Liu Y, Qian T, Ding J, Xu B, Wei J. Novel chimeric peptides based on endomorphins and ghrelin receptor antagonist produced supraspinal antinociceptive effects with reduced acute tolerance in mice. Biochimie 2025; 228:58-70. [PMID: 39147011 DOI: 10.1016/j.biochi.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
It is widely recognized that developing bi- or multifunctional opioid compounds could offer a valuable approach to pain management with fewer side effects compared to single-target compounds. In this study, we designed and characterized two novel chimeric peptides, EM-1-DLS and EM-2-DLS, incorporating endomorphins (EMs) and the ghrelin receptor antagonist [D-Lys3]-GHRP-6 (DLS). Functional assays demonstrated that EM-1-DLS and EM-2-DLS acted as κ-opioid receptor (κ-OR)-preferring agonists, weak μ-opioid receptors (μ-OR) and ghrelin receptor (GHSR) agonists. Upon intracerebroventricular (i.c.v.) administration in mice, both EM-1-DLS and EM-2-DLS exhibited dose- and time-dependent antinociceptive effects in the tail withdrawal test. EM-1-DLS demonstrated the highest antinociceptive potency among the peptides, with an ED50 approximately 8-fold greater than EM-1, while EM-2-DLS showed comparable effects to EM-2. The antinociceptive actions of EM-1-DLS involved activation of GHS-R1α, μ-OR, and κ-OR, whereas EM-2-DLS acted via GHS-R1α, δ-OR, and κ-OR pathways. Additionally, acute antinociceptive tolerance was investigated, revealing that EM-1-DLS induced a tolerance ratio of 2.33-fold, significantly lower than the 5.19-fold ratio induced by EM-1. Cross-tolerance ratios between the chimeric peptides and EMs ranged from 0.92 to 1.76, indicating reduced tolerance compared to EMs alone. These findings highlight the potential of these chimeric peptides to mitigate pain with diminished tolerance development, suggesting a promising strategy for the development of new analgesic therapies with improved safety profiles.
Collapse
Affiliation(s)
- Bing Wu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Songxia Cheng
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Fuyan Liu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China; Department of Anatomy, Basic Medical Teaching and Research Section of Nanchang Health School, Nanchang, Jiangxi Province, 330006, PR China
| | - Jia Wei
- Department of Gynaecology and Obstetrics, Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Cheng Du, Sichuan Province, 610017, PR China
| | - Yongling Liu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Teng Qian
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Jiali Ding
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, PR China.
| | - Jie Wei
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
5
|
Breault É, Desgagné M, Neve JD, Côté J, Barlow TMA, Ballet S, Sarret P. Multitarget ligands that comprise opioid/nonopioid pharmacophores for pain management: Current state of the science. Pharmacol Res 2024; 209:107408. [PMID: 39307212 DOI: 10.1016/j.phrs.2024.107408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Chronic pain, which affects more than one-third of the world's population, represents one of the greatest medical challenges of the 21st century, yet its effective management remains sub-optimal. The 'gold standard' for the treatment of moderate to severe pain consists of opioid ligands, such as morphine and fentanyl, that target the µ-opioid receptor (MOP). Paradoxically, these opioids also cause serious side effects, including constipation, respiratory depression, tolerance, and addiction. In addition, the development of opioid-use disorders, such as opioid diversion, misuse, and abuse, has led to the current opioid crisis, with dramatic increases in addiction, overdoses, and ultimately deaths. As pain is a complex, multidimensional experience involving a variety of pathways and mediators, dual or multitarget ligands that can bind to more than one receptor and exert complementary analgesic effects, represent a promising avenue for pain relief. Indeed, unlike monomodal therapeutic approaches, the modulation of several endogenous nociceptive systems can often result in an additive or even synergistic effect, thereby improving the analgesic-to-side-effect ratio. Here, we provide a comprehensive overview of research efforts towards the development of dual- or multi-targeting opioid/nonopioid hybrid ligands for effective and safer pain management. We reflect on the underpinning discovery rationale by discussing the design, medicinal chemistry, and in vivo pharmacological effects of multitarget antinociceptive compounds.
Collapse
Affiliation(s)
- Émile Breault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Michael Desgagné
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Jérôme Côté
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
6
|
Vieira WF, Coelho DRA, Litwiler ST, McEachern KM, Clancy JA, Morales-Quezada L, Cassano P. Neuropathic pain, mood, and stress-related disorders: A literature review of comorbidity and co-pathogenesis. Neurosci Biobehav Rev 2024; 161:105673. [PMID: 38614452 DOI: 10.1016/j.neubiorev.2024.105673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neuropathic pain can be caused by multiple factors, and its prevalence can reach 10% of the global population. It is becoming increasingly evident that limited or short-lasting response to treatments for neuropathic pain is associated with psychological factors, which include psychiatric comorbidities known to affect quality of life. It is estimated that 60% of patients with neuropathic pain also experience depression, anxiety, and stress symptoms. Altered mood, including stress, can be a consequence of several painful conditions but can also favor pain chronicization when preexisting. Despite the apparent tight connection between clinical pain and mood/stress disorders, the exact physiological mechanisms remain unclear. This review aims to provide an overview of state-of-the-art research on the mechanisms of pain related to the pathophysiology of depression, anxiety, and stress disorders.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil.
| | - David Richer Araujo Coelho
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Harvard T. H. Chan School of Public Health (HSPH), Boston, USA
| | - Scott Thomas Litwiler
- Center for Computational and Integrative Biology (CCIB), Massachusetts General Hospital (MGH), Boston, USA
| | - Kayla Marie McEachern
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Julie A Clancy
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, USA
| | - Paolo Cassano
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA
| |
Collapse
|
7
|
Hochrainer N, Serafin P, D’Ingiullo S, Mollica A, Granica S, Brytan M, Kleczkowska P, Spetea M. In Vitro and In Vivo Pharmacological Profiles of LENART01, a Dermorphin-Ranatensin Hybrid Peptide. Int J Mol Sci 2024; 25:4007. [PMID: 38612817 PMCID: PMC11012005 DOI: 10.3390/ijms25074007] [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] [Received: 03/08/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Diverse chemical and pharmacological strategies are currently being explored to minimize the unwanted side effects of currently used opioid analgesics while achieving effective pain relief. The use of multitarget ligands with activity at more than one receptor represents a promising therapeutic approach. We recently reported a bifunctional peptide-based hybrid LENART01 combining dermorphin and ranatensin pharmacophores, which displays activity to the mu-opioid receptor (MOR) and dopamine D2 receptor (D2R) in rat brains and spinal cords. In this study, we investigated the in vitro binding and functional activities to the human MOR and the in vivo pharmacology of LENART01 in mice after subcutaneous administration. In vitro binding assays showed LENART01 to bind and be selective to the human MOR over the other opioid receptor subtypes and delta, kappa and nociceptin receptors. In the [35S]GTPγS binding assay, LENART01 acted as a potent and full agonist to the human MOR. In mice, LENART01 produced dose-dependent antinociceptive effects in formalin-induced inflammatory pain, with increased potency than morphine. Antinociceptive effects were reversed by naloxone, indicating MOR activation in vivo. Behavioral studies also demonstrated LENART01's properties to induce less adverse effects without locomotor dysfunction and withdrawal syndrome compared to conventional opioid analgesics, such as morphine. LENART01 is the first peptide-based MOR-D2R ligand known to date and the first dual MOR-dopamine D2R ligand for which in vivo pharmacology is reported with antinociceptive efficacy and reduced opioid-related side effects. Our current findings may pave the way to new pain therapeutics with limited side effects in acute and chronic use.
Collapse
Affiliation(s)
- Nadine Hochrainer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Pawel Serafin
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (P.S.); (M.B.)
| | - Sara D’Ingiullo
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.D.); (A.M.)
| | - Adriano Mollica
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.D.); (A.M.)
| | - Sebastian Granica
- Microbiota Lab, Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Marek Brytan
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (P.S.); (M.B.)
| | | | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
8
|
He Y, Su Q, Zhao L, Zhang L, Yu L, Shi J. Historical perspectives and recent advances in small molecule ligands of selective/biased/multi-targeted μ/δ/κ opioid receptor (2019-2022). Bioorg Chem 2023; 141:106869. [PMID: 37797454 DOI: 10.1016/j.bioorg.2023.106869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
The opioids have been used for more than a thousand years and are not only the most widely prescribed drugs for moderate to severe pain and acute pain, but also the preferred drugs. However, their non-analgesic effects, especially respiratory depression and potential addiction, are important factors that plague the safety of clinical use and are an urgent problem for pharmacological researchers to address. Current research on analgesic drugs has evolved into different directions: de-opioidization; application of pharmacogenomics to individualize the use of opioids; development of new opioids with less adverse effects. The development of new opioid drugs remains a hot research topic, and with the in-depth study of opioid receptors and intracellular signal transduction mechanisms, new research ideas have been provided for the development of new opioid analgesics with less side effects and stronger analgesic effects. The development of novel opioid drugs in turn includes selective opioid receptor ligands, biased opioid receptor ligands, and multi-target opioid receptor ligands and positive allosteric modulators (PAMs) or antagonists and the single compound as multi-targeted agnoists/antagonists for different receptors. PAMs strategies are also getting newer and are the current research hotspots, including the BMS series of compounds and others, which are extensive and beyond the scope of this review. This review mainly focuses on the selective/biased/multi-targeted MOR/DOR/KOR (mu opioid receptor/delta opioid receptor/kappa opioid receptor) small molecule ligands and involves some cryo-electron microscopy (cryoEM) and structure-based approaches as well as the single compound as multi-targeted agnoists/antagonists for different receptors from 2019 to 2022, including discovery history, activities in vitro and vivo, and clinical studies, in an attempt to provide ideas for the development of novel opioid analgesics with fewer side effects.
Collapse
Affiliation(s)
- Ye He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qian Su
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lijuan Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Lu Yu
- Department of Respiratory Medicine, Sichuan Academy of Medical Sciences and Sichuan provincial People's Hospital, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
9
|
Turnaturi R, Piana S, Spoto S, Costanzo G, Reina L, Pasquinucci L, Parenti C. From Plant to Chemistry: Sources of Active Opioid Antinociceptive Principles for Medicinal Chemistry and Drug Design. Molecules 2023; 28:7089. [PMID: 37894567 PMCID: PMC10609244 DOI: 10.3390/molecules28207089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Pain continues to be an enormous global health challenge, with millions of new untreated or inadequately treated patients reported annually. With respect to current clinical applications, opioids remain the mainstay for the treatment of pain, although they are often associated with serious side effects. To optimize their tolerability profiles, medicinal chemistry continues to study novel ligands and innovative approaches. Among them, natural products are known to be a rich source of lead compounds for drug discovery, and they hold potential for pain management. Traditional medicine has had a long history in clinical practice due to the fact that nature provides a rich source of active principles. For instance, opium had been used for pain management until the 19th century when its individual components, such as morphine, were purified and identified. In this review article, we conducted a literature survey aimed at identifying natural products interacting either directly with opioid receptors or indirectly through other mechanisms controlling opioid receptor signaling, whose structures could be interesting from a drug design perspective.
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Silvia Piana
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Salvatore Spoto
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125 Catania, Italy; (S.S.); (C.P.)
| | - Giuliana Costanzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy;
| | - Lorena Reina
- Postgraduate School of Clinical Pharmacology, Toxicology University of Catania, Via Santa Sofia n. 97, 95100 Catania, Italy;
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Carmela Parenti
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125 Catania, Italy; (S.S.); (C.P.)
| |
Collapse
|
10
|
Imam MZ, Kuo A, Ghassabian S, Cai Y, Qin Y, Li T, Smith MT. CYX-5, a G-protein biassed MOP receptor agonist, DOP receptor antagonist and KOP receptor agonist, evokes constipation but not respiratory depression relative to morphine in rats. Pharmacol Rep 2023; 75:634-646. [PMID: 36637684 PMCID: PMC10227131 DOI: 10.1007/s43440-023-00446-8] [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] [Received: 10/05/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Strong opioid analgesics such as morphine alleviate moderate to severe acute nociceptive pain (e.g. post-surgical or post-trauma pain) as well as chronic cancer pain. However, they evoke many adverse effects and so there is an unmet need for opioid analgesics with improved tolerability. Recently, a prominent hypothesis has been that opioid-related adverse effects are mediated by β-arrestin2 recruitment at the µ-opioid (MOP) receptor and this stimulated research on discovery of G-protein biassed opioid analgesics. In other efforts, opioids with MOP agonist and δ-opioid (DOP) receptor antagonist profiles are promising for reducing side effects c.f. morphine. Herein, we report on the in vivo pharmacology of a novel opioid peptide (CYX-5) that is a G-protein biassed MOP receptor agonist, DOP receptor antagonist and kappa opioid (KOP) receptor agonist. METHODS Male Sprague-Dawley received intracerebroventricular bolus doses of CYX-5 (3, 10, 20 nmol), morphine (100 nmol) or vehicle, and antinociception (tail flick) was assessed relative to constipation (charcoal meal and castor oil-induced diarrhoea tests) and respiratory depression (whole body plethysmography). RESULTS CYX-5 evoked naloxone-sensitive, moderate antinociception, at the highest dose tested. Although CYX-5 did not inhibit gastrointestinal motility, it reduced stool output markedly in the castor oil-induced diarrhoea test. In contrast to morphine that evoked respiratory depression, CYX-5 increased tidal volume, thereby stimulating respiration. CONCLUSION Despite its lack of recruitment of β-arrestin2 at MOP, DOP and KOP receptors, CYX-5 evoked constipation, implicating a mechanism other than β-arrestin2 recruitment at MOP, DOP and KOP receptors, mediating constipation evoked by CYX-5 and potentially other opioid ligands.
Collapse
Affiliation(s)
- Mohammad Zafar Imam
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Andy Kuo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Sussan Ghassabian
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Yunxin Cai
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yajuan Qin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingyou Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
11
|
Smith MT, Kong D, Kuo A, Imam MZ, Williams CM. Multitargeted Opioid Ligand Discovery as a Strategy to Retain Analgesia and Reduce Opioid-Related Adverse Effects. J Med Chem 2023; 66:3746-3784. [PMID: 36856340 DOI: 10.1021/acs.jmedchem.2c01695] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The global "opioid crisis" has placed enormous pressure on the opioid ligand discovery community to produce novel opioid analgesics with superior opioid-related adverse-effect profiles compared with morphine. In this Perspective, the multitargeted opioid ligand strategy for the discovery of opioid analgesics with superior preclinical therapeutic indices relative to morphine is reviewed and discussed. Dual-targeted μ-opioid (MOP)/δ-opioid (DOP) ligands in which the in vitro DOP antagonist potency at least equals that of the MOP agonist activity, and are devoid of DOP or κ-opioid (KOP) agonist activity, are sufficiently promising candidates to warrant further investigation. Dual-targeted MOP/NOP partial agonists have superior preclinical therapeutic indices to morphine and/or fentanyl in nonhuman primates and are also considered promising. Based on the poor preclinical and clinical therapeutic indices of cebranopadol, which is a full agonist at MOP, DOP, and NOP receptors and a partial agonist at the KOP receptor, this pharmacologic template should be avoided.
Collapse
|
12
|
Fu K, Xu W, Yang R, Zhao H, Xu H, Wei Y, Liu H, Qiu Y, Chen D, Guo D, Xiong B. 2,6-diazaspiro[3.4]octan-7-one derivatives as potent sigma-1 receptor antagonists that enhanced the antinociceptive effect of morphine and rescued morphine tolerance. Eur J Med Chem 2023; 249:115178. [PMID: 36753922 DOI: 10.1016/j.ejmech.2023.115178] [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: 12/13/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Opioids are efficacious analgesics for pain treatments. However, their repeated use in large doses often leads to analgesic tolerance, which limits the clinical application. Sigma-1 receptor (σ1R) antagonists were reported to synergistically enhance the analgesic effect of mu opioid receptor (MOR) agonists without amplifying the adverse effects. Therefore, the σ1R is considered a promising drug target for pain management. Based on the recently elucidated co-crystal structure of σ1R with 4-IBP, we designed and developed a series of σ1R antagonists harboring the 2,6-diazaspiro[3.4]octan-7-one scaffold. Through a detailed structure-activity relationship study, we identified compound 32 as a potent σ1R antagonist, which significantly enhanced the antinociceptive effect of morphine and rescued morphine-induced analgesic tolerance. Our results support σ1R antagonism as a promising strategy to develop novel analgesics and highlight the therapeutic potential of compound 32 to prevent morphine tolerance.
Collapse
Affiliation(s)
- Kequan Fu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Wen Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China; School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Ruicong Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China; School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Huimin Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Huanyu Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqin Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Hongli Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yinli Qiu
- Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd., 1 Yunhe Road, Xuzhou, 221135, Jiangsu, China
| | - Danqi Chen
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Bing Xiong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Dumitrascuta M, Martin C, Ballet S, Spetea M. Bifunctional Peptidomimetic G Protein-Biased Mu-Opioid Receptor Agonist and Neuropeptide FF Receptor Antagonist KGFF09 Shows Efficacy in Visceral Pain without Rewarding Effects after Subcutaneous Administration in Mice. Molecules 2022; 27:8785. [PMID: 36557917 PMCID: PMC9780937 DOI: 10.3390/molecules27248785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
There is still an unmet clinical need to develop new pharmaceuticals for effective and safe pain management. Current pharmacotherapy offers unsatisfactory solutions due to serious side effects related to the chronic use of opioid drugs. Prescription opioids produce analgesia through activation of the mu-opioid receptor (MOR) and are major contributors to the current opioid crisis. Multifunctional ligands possessing activity at more than one receptor represent a prominent therapeutic approach for the treatment of pain with fewer adverse effects. We recently reported on the design of a bifunctional MOR agonist/neuropeptide FF receptor (NPFFR) antagonist peptididomimetic, KGFF09 (H-Dmt-DArg-Aba-βAla-Bpa-Phe-NH2), and its antinociceptive effects after subcutaneous (s.c.) administration in acute and persistent pain in mice with reduced propensity for unwanted side effects. In this study, we further investigated the antinociceptive properties of KGFF09 in a mouse model of visceral pain after s.c. administration and the potential for opioid-related liabilities of rewarding and sedation/locomotor dysfunction following chronic treatment. KGFF09 produced a significant dose-dependent inhibition of the writhing behavior in the acetic acid-induced writhing assay with increased potency when compared to morphine. We also demonstrated the absence of harmful effects caused by typical MOR agonists, i.e., rewarding effects (conditioned-place preference test) and sedation/locomotor impairment (open-field test), at a dose shown to be highly effective in inhibiting pain behavior. Consequently, KGFF09 displayed a favorable benefit/side effect ratio regarding these opioid-related side effects compared to conventional opioid analgesics, such as morphine, underlining the development of dual MOR agonists/NPFFR antagonists as improved treatments for various pain conditions.
Collapse
Affiliation(s)
- Maria Dumitrascuta
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Charlotte Martin
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Steven Ballet
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| |
Collapse
|
14
|
Mu and Delta Opioid Receptor Targeting Reduces Connexin 43-Based Heterocellular Coupling during Neuropathic Pain. Int J Mol Sci 2022; 23:ijms23115864. [PMID: 35682543 PMCID: PMC9180638 DOI: 10.3390/ijms23115864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Chronic neuropathic pain emerges from either central or peripheral lesions inducing spontaneous or amplified responses to non-noxious stimuli. Despite different pharmacological approaches to treat such a chronic disease, neuropathic pain still represents an unmet clinical need, due to long-term therapeutic regimens and severe side effects that limit application of currently available drugs. A critical phenomenon involved in central sensitization is the exchange of signalling molecules and cytokines, between glia and neurons, driving the chronicization process. Herein, using a chronic constriction injury (CCI) model of neuropathic pain, we evaluated the efficacy of the mu (M-) and delta (D-) opioid receptor (-OR) targeting agent LP2 in modulating connexin-based heterocellular coupling and cytokine levels. We found that long-term efficacy of LP2 is consequent to MOR-DOR targeting resulting in the reduction of CCI-induced astrocyte-to-microglia heterocellular coupling mediated by connexin 43. We also found that single targeting of DOR reduces TNF and IL-6 levels in the chronic phase of the disease, but the peripheral and central discharge as the primary source of excitotoxic stimulation in the spinal cord requires a simultaneous MOR-DOR targeting to reduce CCI-induced neuropathic pain.
Collapse
|
15
|
Lipiński PFJ, Matalińska J. Fentanyl Structure as a Scaffold for Opioid/Non-Opioid Multitarget Analgesics. Int J Mol Sci 2022; 23:ijms23052766. [PMID: 35269909 PMCID: PMC8910985 DOI: 10.3390/ijms23052766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
One of the strategies in the search for safe and effective analgesic drugs is the design of multitarget analgesics. Such compounds are intended to have high affinity and activity at more than one molecular target involved in pain modulation. In the present contribution we summarize the attempts in which fentanyl or its substructures were used as a μ-opioid receptor pharmacophoric fragment and a scaffold to which fragments related to non-opioid receptors were attached. The non-opioid ‘second’ targets included proteins as diverse as imidazoline I2 binding sites, CB1 cannabinoid receptor, NK1 tachykinin receptor, D2 dopamine receptor, cyclooxygenases, fatty acid amide hydrolase and monoacylglycerol lipase and σ1 receptor. Reviewing the individual attempts, we outline the chemistry, the obtained pharmacological properties and structure-activity relationships. Finally, we discuss the possible directions for future work.
Collapse
|
16
|
Paul AK, Smith CM, Rahmatullah M, Nissapatorn V, Wilairatana P, Spetea M, Gueven N, Dietis N. Opioid Analgesia and Opioid-Induced Adverse Effects: A Review. Pharmaceuticals (Basel) 2021; 14:1091. [PMID: 34832873 PMCID: PMC8620360 DOI: 10.3390/ph14111091] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
Opioids are widely used as therapeutic agents against moderate to severe acute and chronic pain. Still, these classes of analgesic drugs have many potential limitations as they induce analgesic tolerance, addiction and numerous behavioural adverse effects that often result in patient non-compliance. As opium and opioids have been traditionally used as painkillers, the exact mechanisms of their adverse reactions over repeated use are multifactorial and not fully understood. Older adults suffer from cancer and non-cancer chronic pain more than younger adults, due to the physiological changes related to ageing and their reduced metabolic capabilities and thus show an increased number of adverse reactions to opioid drugs. All clinically used opioids are μ-opioid receptor agonists, and the major adverse effects are directly or potentially connected to this receptor. Multifunctional opioid ligands or peripherally restricted opioids may elicit fewer adverse effects, as shown in preclinical studies, but these results need reproducibility from further extensive clinical trials. The current review aims to overview various mechanisms involved in the adverse effects induced by opioids, to provide a better understanding of the underlying pathophysiology and, ultimately, to help develop an effective therapeutic strategy to better manage pain.
Collapse
Affiliation(s)
- Alok K. Paul
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Craig M. Smith
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC 3216, Australia;
| | - Mohammed Rahmatullah
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Dhanmondi, Dhaka 1207, Bangladesh;
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, World Union for Herbal Drug Discovery (WUHeDD) and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80–82, 6020 Innsbruck, Austria;
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Nikolas Dietis
- Medical School, University of Cyprus, Nicosia 1678, Cyprus;
| |
Collapse
|
17
|
Pasquinucci L, Parenti C, Georgoussi Z, Reina L, Tomarchio E, Turnaturi R. LP1 and LP2: Dual-Target MOPr/DOPr Ligands as Drug Candidates for Persistent Pain Relief. Molecules 2021; 26:molecules26144168. [PMID: 34299443 PMCID: PMC8305117 DOI: 10.3390/molecules26144168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022] Open
Abstract
Although persistent pain is estimated to affect about 20% of the adult population, current treatments have poor results. Polypharmacology, which is the administration of more than one drug targeting on two or more different sites of action, represents a prominent therapeutic approach for the clinical management of persistent pain. Thus, in the drug discovery process the "one-molecule-multiple targets" strategy nowadays is highly recognized. Indeed, multitarget ligands displaying a better antinociceptive activity with fewer side effects, combined with favorable pharmacokinetic and pharmacodynamic characteristics, have already been shown. Multitarget ligands possessing non-opioid/opioid and opioid/opioid mechanisms of action are considered as potential drug candidates for the management of various pain conditions. In particular, dual-target MOPr (mu opioid peptide receptor)/DOPr (delta opioid peptide receptor) ligands exhibit an improved antinociceptive profile associated with a reduced tolerance-inducing capability. The benzomorphan-based compounds LP1 and LP2 belong to this class of dual-target MOPr/DOPr ligands. In the present manuscript, the structure-activity relationships and the pharmacological fingerprint of LP1 and LP2 compounds as suitable drug candidates for persistent pain relief is described.
Collapse
Affiliation(s)
- Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence: (L.P.); (R.T.); Tel.: +39-095-738-4273 (L.P. & R.T.)
| | - Carmela Parenti
- Department of Drug and Health Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos” Ag. Paraskevi-Attikis, 15310 Athens, Greece;
| | - Lorena Reina
- Postgraduate School of Clinical Pharmacology, Toxicology University of Catania, via S. Sofia n. 97, 95100 Catania, Italy;
| | - Emilia Tomarchio
- Postgraduate School of Anesthesiology and Intensive Care, University of Milan, Via Francesco Sforza, 35, 20122 Milan, Italy;
| | - Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence: (L.P.); (R.T.); Tel.: +39-095-738-4273 (L.P. & R.T.)
| |
Collapse
|
18
|
Dumitrascuta M, Bermudez M, Trovato O, De Neve J, Ballet S, Wolber G, Spetea M. Antinociceptive Efficacy of the µ-Opioid/Nociceptin Peptide-Based Hybrid KGNOP1 in Inflammatory Pain without Rewarding Effects in Mice: An Experimental Assessment and Molecular Docking. Molecules 2021; 26:3267. [PMID: 34071603 PMCID: PMC8198056 DOI: 10.3390/molecules26113267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 01/09/2023] Open
Abstract
Opioids are the most effective analgesics, with most clinically available opioids being agonists to the µ-opioid receptor (MOR). The MOR is also responsible for their unwanted effects, including reward and opioid misuse leading to the current public health crisis. The imperative need for safer, non-addictive pain therapies drives the search for novel leads and new treatment strategies. In this study, the recently discovered MOR/nociceptin (NOP) receptor peptide hybrid KGNOP1 (H-Dmt-D-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) was evaluated following subcutaneous administration in mouse models of acute (formalin test) and chronic inflammatory pain (Complete Freund's adjuvant-induced paw hyperalgesia), liabilities of spontaneous locomotion, conditioned place preference, and the withdrawal syndrome. KGNOP1 demonstrated dose-dependent antinociceptive effects in the formalin test, and efficacy in attenuating thermal hyperalgesia with prolonged duration of action. Antinociceptive effects of KGNOP1 were reversed by naltrexone and SB-612111, indicating the involvement of both MOR and NOP receptor agonism. In comparison with morphine, KGNOP1 was more potent and effective in mouse models of inflammatory pain. Unlike morphine, KGNOP1 displayed reduced detrimental liabilities, as no locomotor impairment nor rewarding and withdrawal effects were observed. Docking of KGNOP1 to the MOR and NOP receptors and subsequent 3D interaction pattern analyses provided valuable insights into its binding mode. The mixed MOR/NOP receptor peptide KGNOP1 holds promise in the effort to develop new analgesics for the treatment of various pain states with fewer MOR-mediated side effects, particularly abuse and dependence liabilities.
Collapse
Affiliation(s)
- Maria Dumitrascuta
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| | - Marcel Bermudez
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany; (M.B.); (G.W.)
| | - Olga Trovato
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| | - Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium; (J.D.N.); (S.B.)
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium; (J.D.N.); (S.B.)
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany; (M.B.); (G.W.)
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| |
Collapse
|
19
|
Abstract
The management of pain, particularly chronic pain, is still an area of medical need. In this context, opioids remain a gold standard for the treatment of pain. However, significant side effects, mainly of central origin, limit their clinical use. Here, we review recent progress to improve the therapeutic and safety profiles of opioids for pain management. Characterization of peripheral opioid-mediated pain mechanisms have been a key component of this process. Several studies identified peripheral µ, δ, and κ opioid receptors (MOR, DOR, and KOR, respectively) and nociceptin/orphanin FQ (NOP) receptors as significant players of opioid-mediated antinociception, able to achieve clinically significant effects independently of any central action. Following this, particularly from a medicinal chemistry point of view, main efforts have been directed towards the peripheralization of opioid receptor agonists with the objective of optimizing receptor activity and minimizing central exposure and the associated undesired effects. These activities have allowed the characterization of a great variety of compounds and investigational drugs that show low central nervous system (CNS) penetration (and therefore a reduced side effect profile) yet maintaining the desired opioid-related peripheral antinociceptive activity. These include highly hydrophilic/amphiphilic and massive molecules unable to easily cross lipid membranes, substrates of glycoprotein P (a extrusion pump that avoids CNS penetration), nanocarriers that release the analgesic agent at the site of inflammation and pain, and pH-sensitive opioid agonists that selectively activate at those sites (and represent a new pharmacodynamic paradigm). Hopefully, patients with pain will benefit soon from the incorporation of these new entities.
Collapse
|
20
|
Matalińska J, Lipiński PFJ, Kosson P, Kosińska K, Misicka A. In Vivo, In Vitro and In Silico Studies of the Hybrid Compound AA3266, an Opioid Agonist/NK1R Antagonist with Selective Cytotoxicity. Int J Mol Sci 2020; 21:E7738. [PMID: 33086743 PMCID: PMC7588979 DOI: 10.3390/ijms21207738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/14/2020] [Accepted: 10/17/2020] [Indexed: 12/14/2022] Open
Abstract
AA3266 is a hybrid compound consisting of opioid receptor agonist and neurokinin-1 receptor (NK1R) antagonist pharmacophores. It was designed with the desire to have an analgesic molecule with improved properties and auxiliary anticancer activity. Previously, the compound was found to exhibit high affinity for μ- and δ-opioid receptors, while moderate binding to NK1R. In the presented contribution, we report on a deeper investigation of this hybrid. In vivo, we have established that AA3266 has potent antinociceptive activity in acute pain model, comparable to that of morphine. Desirably, with prolonged administration, our hybrid induces less tolerance than morphine does. AA3266, contrary to morphine, does not cause development of constipation, which is one of the main undesirable effects of opioid use. In vitro, we have confirmed relatively strong cytotoxic activity on a few selected cancer cell lines, similar to or greater than that of a reference NK1R antagonist, aprepitant. Importantly, our compound affects normal cells to smaller extent what makes our compound more selective against cancer cells. In silico methods, including molecular docking, molecular dynamics simulations and fragment molecular orbital calculations, have been used to investigate the interactions of AA3266 with MOR and NK1R. Insights from these will guide structural optimization of opioid/antitachykinin hybrid compounds.
Collapse
Affiliation(s)
- Joanna Matalińska
- Department of Neuropeptides, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (K.K.); (A.M.)
| | - Piotr F. J. Lipiński
- Department of Neuropeptides, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (K.K.); (A.M.)
| | - Piotr Kosson
- Toxicology Research Laboratory, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Katarzyna Kosińska
- Department of Neuropeptides, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (K.K.); (A.M.)
| | - Aleksandra Misicka
- Department of Neuropeptides, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (K.K.); (A.M.)
| |
Collapse
|
21
|
Synthesis, Docking, 3-D-Qsar, and Biological Assays of Novel Indole Derivatives Targeting Serotonin Transporter, Dopamine D2 Receptor, and Mao-A Enzyme: In the Pursuit for Potential Multitarget Directed Ligands. MOLECULES (BASEL, SWITZERLAND) 2020; 25:molecules25204614. [PMID: 33050524 PMCID: PMC7594025 DOI: 10.3390/molecules25204614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022]
Abstract
A series of 27 compounds of general structure 2,3-dihydro-benzo[1,4]oxazin-4-yl)-2-{4-[3-(1H-3indolyl)-propyl]-1-piperazinyl}-ethanamides, Series I: 7(a-o) and (2-{4-[3-(1H-3-indolyl)-propyl]-1-piperazinyl}-acetylamine)-N-(2-morfolin-4-yl-ethyl)-fluorinated benzamides Series II: 13(a-l) were synthesized and evaluated as novel multitarget ligands towards dopamine D2 receptor, serotonin transporter (SERT), and monoamine oxidase-A (MAO-A) directed to the management of major depressive disorder (MDD). All the assayed compounds showed affinity for SERT in the nanomolar range, with five of them displaying Ki values from 5 to 10 nM. Compounds 7k, Ki = 5.63 ± 0.82 nM, and 13c, Ki = 6.85 ± 0.19 nM, showed the highest potencies. The affinities for D2 ranged from micro to nanomolar, while MAO-A inhibition was more discrete. Nevertheless, compounds 7m and 7n showed affinities for the D2 receptor in the nanomolar range (7n: Ki = 307 ± 6 nM and 7m: Ki = 593 ± 62 nM). Compound 7n was the only derivative displaying comparable affinities for SERT and D2 receptor (D2/SERT ratio = 3.6) and could be considered as a multitarget lead for further optimization. In addition, docking studies aimed to rationalize the molecular interactions and binding modes of the designed compounds in the most relevant protein targets were carried out. Furthermore, in order to obtain information on the structure-activity relationship of the synthesized series, a 3-D-QSAR CoMFA and CoMSIA study was conducted and validated internally and externally (q2 = 0.625, 0.523 for CoMFA and CoMSIA and r2ncv = 0.967, 0.959 for CoMFA and CoMSIA, respectively).
Collapse
|
22
|
Design, Synthesis and Functional Analysis of Cyclic Opioid Peptides with Dmt-Tic Pharmacophore. Molecules 2020; 25:molecules25184260. [PMID: 32957550 PMCID: PMC7570497 DOI: 10.3390/molecules25184260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 11/17/2022] Open
Abstract
The opioid receptors are members of the G-protein-coupled receptor (GPCR) family and are known to modulate a variety of biological functions, including pain perception. Despite considerable advances, the mechanisms by which opioid agonists and antagonists interact with their receptors and exert their effect are still not completely understood. In this report, six new hybrids of the Dmt-Tic pharmacophore and cyclic peptides, which were shown before to have a high affinity for the µ-opioid receptor (MOR) were synthesized and characterized pharmacologically in calcium mobilization functional assays. All obtained ligands turned out to be selective antagonists of the δ-opioid receptor (DOR) and did not activate or block the MOR. The three-dimensional structural determinants responsible for the DOR antagonist properties of these analogs were further investigated by docking studies. The results indicate that these compounds attach to the DOR in a slightly different orientation with respect to the Dmt-Tic pharmacophore than Dmt-TicΨ[CH2-NH]Phe-Phe-NH2 (DIPP-NH2[Ψ]), a prototypical DOR antagonist peptide. Key pharmacophoric contacts between the DOR and the ligands were maintained through an analogous spatial arrangement of pharmacophores, which could provide an explanation for the predicted high-affinity binding and the experimentally observed functional properties of the novel synthetic ligands.
Collapse
|
23
|
Ma H, Huang B, Zhang Y. Recent advances in multitarget-directed ligands targeting G-protein-coupled receptors. Drug Discov Today 2020; 25:1682-1692. [PMID: 32652312 PMCID: PMC7572774 DOI: 10.1016/j.drudis.2020.07.004] [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: 05/14/2020] [Revised: 06/19/2020] [Accepted: 07/03/2020] [Indexed: 01/13/2023]
Abstract
Mounting evidence indicates that single-target drugs might be inadequate to achieve satisfactory therapeutic effects on complex diseases. Recently, increasing attention has been paid to developing drugs that can manipulate multiple targets to generate beneficial effects through potential synergy. G-protein-coupled receptors (GPCRs) become desirable targets for developing multitarget-directed ligands (MTDLs) because of their crucial roles in the pathophysiology of various human diseases and the accessibility of druggable sites at the cell surface. Herein, we review the most recent advances in the development of GPCR-targeted MTDLs in treating complex diseases, and discuss their potential therapeutic strategies to reveal current trends and shed insights into the utility of GPCR-targeted MTDLs for future drug design and development.
Collapse
Affiliation(s)
- Hongguang Ma
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA
| | - Boshi Huang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA.
| |
Collapse
|
24
|
Design and Applications of Bifunctional Small Molecules in Biology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140534. [PMID: 32871274 DOI: 10.1016/j.bbapap.2020.140534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
|
25
|
Wtorek K, Piekielna-Ciesielska J, Janecki T, Janecka A. The search for opioid analgesics with limited tolerance liability. Peptides 2020; 130:170331. [PMID: 32497566 DOI: 10.1016/j.peptides.2020.170331] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/03/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Reducing the well-known side effects of opioids prescribed to treat chronic pain remains unresolved, despite extensive research in this field. Among several options to tackle this problem the synthesis of multifunctional compounds containing hybridized structures gained a lot of interest. Recently, extensively investigated are combinations of opioid agonist and antagonist pharmacophores embodied in a single molecule. To this end, agonism at the μ opioid receptor (MOR) with simultaneous antagonism at the δ opioid receptor (DOR) emerged as a promising avenue to obtaining novel analogs devoid of serious adverse effects associated with morphine-based analgesics. In this review we covered up-to-date research on the synthesis of peptide-based ligands with MOR agonist/DOR antagonist profile.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Lodz, Poland
| | | | - Tomasz Janecki
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
26
|
Krüll J, Fehler SK, Hofmann L, Nebel N, Maschauer S, Prante O, Gmeiner P, Lanig H, Hübner H, Heinrich MR. Synthesis, Radiosynthesis and Biological Evaluation of Buprenorphine-Derived Phenylazocarboxamides as Novel μ-Opioid Receptor Ligands. ChemMedChem 2020; 15:1175-1186. [PMID: 32378310 PMCID: PMC7383964 DOI: 10.1002/cmdc.202000180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Targeted structural modifications have led to a novel type of buprenorphine-derived opioid receptor ligand displaying an improved selectivity profile for the μ-OR subtype. On this basis, it is shown that phenylazocarboxamides may serve as useful bioisosteric replacements for the widely occurring cinnamide units, without loss of OR binding affinity or subtype selectivity. This study further includes functional experiments pointing to weak partial agonist properties of the novel μ-OR ligands, as well as docking and metabolism experiments. Finally, the unique bifunctional character of phenylazocarboxylates, herein serving as precursors for the azocarboxamide subunit, was exploited to demonstrate the accessibility of an 18 F-fluorinated analogue.
Collapse
Affiliation(s)
- Jasmin Krüll
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Stefanie K. Fehler
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Laura Hofmann
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Natascha Nebel
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Simone Maschauer
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Olaf Prante
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Peter Gmeiner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Harald Lanig
- Central Institute for Scientific Computing (ZISC)Friedrich-Alexander-Universität Erlangen-NürnbergMartensstr. 5a91058ErlangenGermany
| | - Harald Hübner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Markus R. Heinrich
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| |
Collapse
|
27
|
Pasquinucci L, Parenti C, Ruiz-Cantero MC, Georgoussi Z, Pallaki P, Cobos EJ, Amata E, Marrazzo A, Prezzavento O, Arena E, Dichiara M, Salerno L, Turnaturi R. Novel N-Substituted Benzomorphan-Based Compounds: From MOR-Agonist/DOR-Antagonist to Biased/Unbiased MOR Agonists. ACS Med Chem Lett 2020; 11:678-685. [PMID: 32435370 PMCID: PMC7236032 DOI: 10.1021/acsmedchemlett.9b00549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 12/21/2022] Open
Abstract
Modifications at the basic nitrogen of the benzomorphan scaffold allowed the development of compounds able to segregate physiological responses downstream of the receptor signaling, opening new possibilities in opioid drug development. Alkylation of the phenyl ring in the N-substituent of the MOR-agonist/DOR-antagonist LP1 resulted in retention of MOR affinity. Moreover, derivatives 7a, 7c, and 7d were biased MOR agonists toward ERK1,2 activity stimulation, whereas derivative 7e was a low potency MOR agonist on adenylate cyclase inhibition. They were further screened in the mouse tail flick test and PGE2-induced hyperalgesia and drug-induced gastrointestinal transit.
Collapse
Affiliation(s)
- Lorella Pasquinucci
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department
of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - M. Carmen Ruiz-Cantero
- Department
of Pharmacology, Faculty of Medicine and Institute of Neuroscience,
Biomedical Research Center, University of
Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
- Teófilo Hernando
Institute for Drug Discovery, 28029 Madrid, Spain
| | - Zafiroula Georgoussi
- Laboratory
of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences
and Applications, National Center for Scientific
Research “Demokritos″, Ag. Paraskevi 15310, Athens, Greece
| | - Paschalina Pallaki
- Laboratory
of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences
and Applications, National Center for Scientific
Research “Demokritos″, Ag. Paraskevi 15310, Athens, Greece
| | - Enrique J. Cobos
- Department
of Pharmacology, Faculty of Medicine and Institute of Neuroscience,
Biomedical Research Center, University of
Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
- Teófilo Hernando
Institute for Drug Discovery, 28029 Madrid, Spain
| | - Emanuele Amata
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rita Turnaturi
- Department
of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
28
|
Nie C, Li Q, Qiao Y, Hu J, Gao M, Wang Y, Qiao Z, Wang Q, Yan L, Qian H. Study on chemical modification and analgesic activity of N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl) piperazine-1-carboxamide. Eur J Med Chem 2020; 194:112236. [DOI: 10.1016/j.ejmech.2020.112236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 01/14/2023]
|
29
|
Henry S, Anand JP, Twarozynski JJ, Brinkel AC, Pogozheva ID, Sears BF, Jutkiewicz EM, Traynor JR, Mosberg HI. Aromatic-Amine Pendants Produce Highly Potent and Efficacious Mixed Efficacy μ-Opioid Receptor (MOR)/δ-Opioid Receptor (DOR) Peptidomimetics with Enhanced Metabolic Stability. J Med Chem 2020; 63:1671-1683. [PMID: 31986033 DOI: 10.1021/acs.jmedchem.9b01818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We previously reported a novel SAR campaign that converted a metabolically unstable series of μ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist bicyclic core peptidomimetics with promising analgesic activity and reduced abuse liabilities into a more stable series of benzylic core analogues. Herein, we expanded the SAR of that campaign and determined that the incorporation of amines into the benzylic pendant produces enhanced MOR-efficacy in this series, whereas the reincorporation of an aromatic ring into the pendant enhanced MOR-potency. Two compounds, which contain a piperidine (14) or an isoindoline (17) pendant, retained the desired opioid profile in vitro, possessed metabolic half-lives of greater than 1 h in mouse liver microsomes (MLMs), and were active antinociceptive agents in the acetic acid stretch assay (AASA) at subcutaneous doses of 1 mg/kg.
Collapse
Affiliation(s)
- Sean Henry
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Jessica P Anand
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Jack J Twarozynski
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Ashley C Brinkel
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Bryan F Sears
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Emily M Jutkiewicz
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - John R Traynor
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States.,Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Henry I Mosberg
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
30
|
Wtorek K, Adamska-Bartłomiejczyk A, Piekielna-Ciesielska J, Ferrari F, Ruzza C, Kluczyk A, Piasecka-Zelga J, Calo’ G, Janecka A. Synthesis and Pharmacological Evaluation of Hybrids Targeting Opioid and Neurokinin Receptors. Molecules 2019; 24:molecules24244460. [PMID: 31817441 PMCID: PMC6943619 DOI: 10.3390/molecules24244460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 11/16/2022] Open
Abstract
Morphine, which acts through opioid receptors, is one of the most efficient analgesics for the alleviation of severe pain. However, its usefulness is limited by serious side effects, including analgesic tolerance, constipation, and dependence liability. The growing awareness that multifunctional ligands which simultaneously activate two or more targets may produce a more desirable drug profile than selectively targeted compounds has created an opportunity for a new approach to developing more effective medications. Here, in order to better understand the role of the neurokinin system in opioid-induced antinociception, we report the synthesis, structure–activity relationship, and pharmacological characterization of a series of hybrids combining opioid pharmacophores with either substance P (SP) fragments or neurokinin receptor (NK1) antagonist fragments. On the bases of the in vitro biological activities of the hybrids, two analogs, opioid agonist/NK1 antagonist Tyr-[d-Lys-Phe-Phe-Asp]-Asn-d-Trp-Phe-d-Trp-Leu-Nle-NH2 (2) and opioid agonist/NK1 agonist Tyr-[d-Lys-Phe-Phe-Asp]-Gln-Phe-Phe-Gly-Leu-Met-NH2 (4), were selected for in vivo tests. In the writhing test, both hybrids showed significant an antinociceptive effect in mice, while neither of them triggered the development of tolerance, nor did they produce constipation. No statistically significant differences in in vivo activity profiles were observed between opioid/NK1 agonist and opioid/NK1 antagonist hybrids.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.W.); (A.A.-B.); (J.P.-C.)
| | - Anna Adamska-Bartłomiejczyk
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.W.); (A.A.-B.); (J.P.-C.)
| | - Justyna Piekielna-Ciesielska
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.W.); (A.A.-B.); (J.P.-C.)
| | - Federica Ferrari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (F.F.); (C.R.); (G.C.)
| | - Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (F.F.); (C.R.); (G.C.)
| | - Alicja Kluczyk
- Faculty of Chemistry, University of Wroclaw, 50-383 Wroclaw, Poland;
| | - Joanna Piasecka-Zelga
- Institute of Occupational Medicine, Research Laboratory for Medicine and Veterinary Products in the GMP Head of Research Laboratory for Medicine and Veterinary Products, 91-348 Lodz, Poland;
| | - Girolamo Calo’
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (F.F.); (C.R.); (G.C.)
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.W.); (A.A.-B.); (J.P.-C.)
- Correspondence:
| |
Collapse
|
31
|
Structure-Activity Relationships of 7-Substituted Dimethyltyrosine-Tetrahydroisoquinoline Opioid Peptidomimetics. Molecules 2019; 24:molecules24234302. [PMID: 31779072 PMCID: PMC6930500 DOI: 10.3390/molecules24234302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
The opioid receptors modulate a variety of biological functions, including pain, mood, and reward. As a result, opioid ligands are being explored as potential therapeutics for a variety of indications. Multifunctional opioid ligands, which act simultaneously at more than one type of opioid receptor, show promise for use in the treatment of addiction, pain, and other conditions. Previously, we reported the creation of bifunctional kappa opioid receptor (KOR) agonist/mu opioid receptor (MOR) partial agonist ligands from the classically delta opioid receptor (DOR) antagonist selective dimethyltyrosine-tetrahydroisoquinoline (Dmt-Tiq) scaffold through the addition of a 7-benzyl pendant on the tetrahydroisoquinoline ring. This study further explores the structure–activity relationships surrounding 7-position pendants on the Dmt-Tiq scaffold. Some analogues maintain a KOR agonist/MOR partial agonist profile, which is being explored in the development of a treatment for cocaine addiction. Others display a MOR agonist/DOR antagonist profile, which has potential to be used in the creation of a less addictive pain medication. Ultimately, we report the synthesis and in vitro evaluation of novel opioid ligands with a variety of multifunctional profiles.
Collapse
|
32
|
Turnaturi R, Chiechio S, Salerno L, Rescifina A, Pittalà V, Cantarella G, Tomarchio E, Parenti C, Pasquinucci L. Progress in the development of more effective and safer analgesics for pain management. Eur J Med Chem 2019; 183:111701. [PMID: 31550662 DOI: 10.1016/j.ejmech.2019.111701] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/26/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
Opioid analgesics have been used for thousands of years in the treatment of pain and related disorders, and have become among the most widely prescribed medications. Among opioid analgesics, mu opioid receptor (MOR) agonists are the most commonly used and are indicated for acute and chronic pain management. However, their use results in a plethora of well-described side-effects. From selective delta opioid receptor (DOR) and kappa opioid receptor (KOR) agonists to multitarget MOR/DOR and MOR/KOR ligands, medicinal chemistry provided different approaches aimed at the development of opioid analgesics with an improved pharmacological and tolerability fingerprint. The emergent medicinal chemistry strategy to develop ameliorated opioid analgesics is based upon the concept that functional selectivity for G-protein signalling is necessary for the therapeutic effect, whether β-arrestin recruitment is mainly responsible for the manifestation of side effects, including the development of tolerance after repeated administrations. This review summarises most relevant biased MOR, DOR, KOR and multitarget MOR/DOR ligands synthesised in the last decade and their pharmacological profile in "in vitro" and "in vivo" studies. Such biased ligands could have a significant impact on modern drug discovery and represent a new strategy for the development of better-tolerated drug candidates.
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy.
| | - Santina Chiechio
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Troina, Italy
| | - Loredana Salerno
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, Chemistry Section, University of Catania, Viale A. Doria, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Pharmacology Section, University of Catania, Catania, Italy
| | | | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
33
|
Montgomery D, Anand JP, Griggs NW, Fernandez TJ, Hartman JG, Sánchez-Santiago AA, Pogozheva ID, Traynor JR, Mosberg HI. Novel Dimethyltyrosine-Tetrahydroisoquinoline Peptidomimetics with Aromatic Tetrahydroisoquinoline Substitutions Show in Vitro Kappa and Mu Opioid Receptor Agonism. ACS Chem Neurosci 2019; 10:3682-3689. [PMID: 31199621 DOI: 10.1021/acschemneuro.9b00250] [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] [Indexed: 11/28/2022] Open
Abstract
The dimethyltyrosine-tetrahydroisoquinoline (Dmt-Tiq) scaffold was originally developed in the production of selective delta opioid receptor (DOR) antagonists. Installation of a 7-benzyl pendant on the tetrahydroisoquinoline core of this classic opioid scaffold introduced kappa opioid receptor (KOR) agonism. Further modification of this pendant resulted in retention of KOR agonism and the addition of mu opioid receptor (MOR) partial agonism, a bifunctional profile with potential to be used in the treatment of cocaine addiction.
Collapse
Affiliation(s)
- Deanna Montgomery
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica P. Anand
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nicholas W. Griggs
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Thomas J. Fernandez
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Joshua G. Hartman
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ashley A. Sánchez-Santiago
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Irina D. Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I. Mosberg
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
34
|
Beck TC, Hapstack MA, Beck KR, Dix TA. Therapeutic Potential of Kappa Opioid Agonists. Pharmaceuticals (Basel) 2019; 12:ph12020095. [PMID: 31226764 PMCID: PMC6631266 DOI: 10.3390/ph12020095] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
Many original research articles have been published that describe findings and outline areas for the development of kappa-opioid agonists (KOAs) as novel drugs; however, a single review article that summarizes the broad potential for KOAs in drug development does not exist. It is well-established that KOAs demonstrate efficacy in pain attenuation; however, KOAs also have proven to be beneficial in treating a variety of novel but often overlapping conditions including cardiovascular disease, pruritus, nausea, inflammatory diseases, spinal anesthesia, stroke, hypoxic pulmonary hypertension, multiple sclerosis, addiction, and post-traumatic cartilage degeneration. This article summarizes key findings of KOAs and discusses the untapped therapeutic potential of KOAs in the treatment of many human diseases.
Collapse
Affiliation(s)
- Tyler C Beck
- Drug Discovery & Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, QF204, Charleston, SC 29424-2303, USA.
- College of Medicine, 173 Ashley Ave., Charleston, SC 29424-2303, USA.
| | | | - Kyle R Beck
- College of Pharmacy, The Ohio State University, 500 W 12th Ave, Columbus, OH 43210-9998, USA.
| | - Thomas A Dix
- Drug Discovery & Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, QF204, Charleston, SC 29424-2303, USA.
- JT Pharmaceuticals, Inc., 300 West Coleman Blvd., Suite 203, Mount Pleasant, SC 29464-2303, USA.
| |
Collapse
|
35
|
Henry SP, Fernandez TJ, Anand JP, Griggs NW, Traynor JR, Mosberg HI. Structural Simplification of a Tetrahydroquinoline-Core Peptidomimetic μ-Opioid Receptor (MOR) Agonist/δ-Opioid Receptor (DOR) Antagonist Produces Improved Metabolic Stability. J Med Chem 2019; 62:4142-4157. [PMID: 30924650 DOI: 10.1021/acs.jmedchem.9b00219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously reported a series of μ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist ligands to serve as potential nonaddictive opioid analgesics. These ligands have been shown to be active in vivo, do not manifest withdrawal syndromes or reward behavior in conditioned-place preference assays in mice, and do not produce dependence. Although these attributes are promising, these analogues exhibit poor metabolic stability in mouse liver microsomes, likely due to the central tetrahydroquinoline scaffold in this series. As such, a structure-activity relationship (SAR) campaign was pursued to improve their metabolic stability. This resulted in a shift from our original bicyclic tetrahydroquinoline core to a monocyclic benzylic-core system. By eliminating one of the rings in this scaffold and exploring the SAR of this new core, two promising analogues were discovered. These analogues (5l and 5m) had potency and efficacy values at MOR better or comparable to morphine, retained their DOR-antagonist properties, and showed a 10-fold improvement in metabolic stability.
Collapse
Affiliation(s)
- Sean P Henry
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | | | | | | | - John R Traynor
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Henry I Mosberg
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
36
|
Nastase AF, Anand JP, Bender AM, Montgomery D, Griggs NW, Fernandez TJ, Jutkiewicz EM, Traynor JR, Mosberg HI. Dual Pharmacophores Explored via Structure-Activity Relationship (SAR) Matrix: Insights into Potent, Bifunctional Opioid Ligand Design. J Med Chem 2019; 62:4193-4203. [PMID: 30916966 DOI: 10.1021/acs.jmedchem.9b00378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Short-acting μ-opioid receptor (MOR) agonists have long been used for the treatment of severe, breakthrough pain. However, selective MOR agonists including fentanyl and morphine derivatives are limited clinically due to high risks of dependence, tolerance, and respiratory depression. We recently reported the development of a long-acting, bifunctional MOR agonist/δ-opioid receptor (DOR) antagonist analgesic devoid of tolerance or dependence in mice (AAH8, henceforth referred to as 2B). To address the need for short-acting treatments for breakthrough pain, we present a series of novel, short-acting, high-potency MOR agonist/DOR antagonist ligands with antinociceptive activity in vivo. In this study, we utilized a two-dimensional structure-activity relationship matrix to identify pharmacological trends attributable to combinations of two key pharmacophore elements within the chemotype. This work enhances our ability to modulate efficacy at MOR and DOR, accessing a variety of bifunctional profiles while maintaining high affinity and potency at both receptors.
Collapse
|
37
|
(2S)-N-2-methoxy-2-phenylethyl-6,7-benzomorphan compound (2S-LP2): Discovery of a biased mu/delta opioid receptor agonist. Eur J Med Chem 2019; 168:189-198. [PMID: 30822708 DOI: 10.1016/j.ejmech.2019.02.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
The pivotal role of the stereocenter at the N-substituent of the 6,7-benzomorphan scaffold was investigated combining synthetic and pharmacological approaches. 2R- and 2S-diastereoisomers of the multitarget MOR/DOR antinociceptive ligand LP2 (1) were synthesized and their pharmacological profile was evaluated in in vitro and vivo assays. From our results, 2S-LP2 (5) showed an improved pharmacological profile in comparison to LP2 (1) and 2R-LP2 (4). 2S-LP2 (5) elicited an antinociceptive effect with a 1.5- and 3-times higher potency than LP2 (1) and R-antipode (4), respectively. In vivo effect of 2S-LP2 (5) was consistent with the improved MOR/DOR efficacy profile assessed by radioligand binding assay, to evaluate the opioid receptor affinity, and BRET assay, to evaluate the capability to promote receptor/G-protein and receptor/β-arrestin 2 interaction. 2S-LP2 (5) was able to activate, with different efficacy, G-protein pathway over β-arrestin 2, behaving as biased agonist at MOR and mainly at DOR. Considering the therapeutic potential of both multitarget MOR/DOR agonism and functional selectivity over G-protein, the 2S-LP2 (5) biased multitarget MOR/DOR agonist could provide a safer treatment opportunity.
Collapse
|
38
|
Pasquinucci L, Turnaturi R, Montenegro L, Caraci F, Chiechio S, Parenti C. Simultaneous targeting of MOR/DOR: A useful strategy for inflammatory pain modulation. Eur J Pharmacol 2019; 847:97-102. [PMID: 30690004 DOI: 10.1016/j.ejphar.2019.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 02/03/2023]
Abstract
Development of new analgesics endowed with mu/delta opioid receptor (MOR/DOR) activity represents a promising alternative to MOR selective compounds because of their better therapeutic and tolerability profile. Lately, we have synthetized the MOR/DOR agonist LP2 that showed a long lasting antinociceptive activity in the tail flick test, an acute pain model. Here, we investigate whether LP2 is also effective in the mouse formalin test, a model of inflammatory pain sustained by mechanisms of central sensitization. Moreover, we evaluated a possible peripheral component of LP2 analgesic activity. Different doses of LP2 were tested after either intraperitoneal (i.p.) or intraplantar (i.pl.) administration. LP2 (0.75-1.00 mg/kg, i.p.), dose-dependently, counteracted both phases of the formalin test after i.p. administration. The analgesic activity of LP2 (0.75-1.00 mg/kg) was completely blocked by a pretreatment with the opioid antagonist naloxone (3 mg/kg, i.p.). Differently, the pretreatment with naloxone methiodide (5 mg/kg, i.p.), a peripherally restricted opioid antagonist, completely blocked the lower analgesic dose of LP2 (0.75 mg/kg) but only partially relieved the antinociceptive effects of LP2 at the dose of 1.00 mg/kg, thus revealing a peripheral analgesic component of LP2. I.pl. injections of LP2 (10-20 μg/10 μl) were also performed to investigate a possible effect of LP2 on peripheral nerve terminals. Nociceptive sensitization, which occur both at peripheral and central level, is a fundamental step for pain chronicization, thus LP2 is a promising drug for pain conditions characterized by nociceptive sensitization.
Collapse
Affiliation(s)
- Lorella Pasquinucci
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Lucia Montenegro
- Department of Drug Sciences, Pharmaceutical Technology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Filippo Caraci
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy; Oasi Research Institute-IRCCS, Troina, Italy
| | - Santina Chiechio
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy; Oasi Research Institute-IRCCS, Troina, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
39
|
Tosh D, Ciancetta A, Mannes P, Warnick E, Janowsky A, Eshleman AJ, Gizewski E, Brust TF, Bohn LM, Auchampach JA, Gao ZG, Jacobson KA. Repurposing of a Nucleoside Scaffold from Adenosine Receptor Agonists to Opioid Receptor Antagonists. ACS OMEGA 2018; 3:12658-12678. [PMID: 30411015 PMCID: PMC6210068 DOI: 10.1021/acsomega.8b01237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/19/2018] [Indexed: 06/08/2023]
Abstract
While screening off-target effects of rigid (N)-methanocarba-adenosine 5'-methylamides as A3 adenosine receptor (AR) agonists, we discovered μM binding hits at the δ-opioid receptor (DOR) and translocator protein (TSPO). In an effort to increase OR and decrease AR affinity by structure activity analysis of this series, antagonist activity at κ-(K)OR appeared in 5'-esters (ethyl 24 and propyl 30), which retained TSPO interaction (μM). 7-Deaza modification of C2-(arylethynyl)-5'-esters but not 4'-truncation enhanced KOR affinity (MRS7299 28 and 29, K i ≈ 40 nM), revealed μ-OR and DOR binding, and reduced AR affinity. Molecular docking and dynamics simulations located a putative KOR binding mode consistent with the observed affinities, placing C7 in a hydrophobic region. 3-Deaza modification permitted TSPO but not OR binding, and 1-deaza was permissive to both; ribose-restored analogues were inactive at both. Thus, we have repurposed a known AR nucleoside scaffold for OR antagonism, with a detailed hypothesis for KOR recognition.
Collapse
Affiliation(s)
- Dilip
K. Tosh
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Antonella Ciancetta
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Philip Mannes
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Eugene Warnick
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Aaron Janowsky
- VA
Portland Health Care System, Research Service (R&D-22), and Departments
of Psychiatry and Behavioral Neuroscience, Oregon Health and Science University, 3710 S.W. U.S. Veterans Hospital Blvd., Portland, Oregon 97239, United States
| | - Amy J. Eshleman
- VA
Portland Health Care System, Research Service (R&D-22), and Departments
of Psychiatry and Behavioral Neuroscience, Oregon Health and Science University, 3710 S.W. U.S. Veterans Hospital Blvd., Portland, Oregon 97239, United States
| | - Elizabeth Gizewski
- Department
of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Tarsis F. Brust
- Departments
of Molecular Medicine and Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United
States
| | - Laura M. Bohn
- Departments
of Molecular Medicine and Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United
States
| | - John A. Auchampach
- Department
of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Zhan-Guo Gao
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Kenneth A. Jacobson
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| |
Collapse
|
40
|
Bivalent ligand that activates mu opioid receptor and antagonizes mGluR5 receptor reduces neuropathic pain in mice. Pain 2018; 158:2431-2441. [PMID: 28891868 DOI: 10.1097/j.pain.0000000000001050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The mu opioid receptor (MOR) and metabotropic glutamate receptor 5 (mGluR5) are well-established pharmacological targets in the management of chronic pain. Both receptors are expressed in the spinal cord. MMG22, a bivalent ligand containing 2 pharmacophores separated by 22 atoms, which simultaneously activates MOR and antagonizes mGluR5, has been shown to produce potent reversal of tactile hypersensitivity in rodent models of lipopolysaccharide (LPS)-and bone cancer-induced chronic pain. This study assessed whether intrathecal MMG22 also is effective in reducing pain of neuropathic origin. Furthermore, we theorized that MMG22 should reduce hyperalgesia in nerve-injured mice in a manner consistent with a synergistic interaction between MOR and mGluR5. Several weeks after spared nerve injury, tactile hypersensitivity was reversed in mice by the intrathecal injection of MMG22 (0.01-10 nmol) but also by its shorter spacer analog, MMG10, with similar potency. The potencies of the bivalent ligands were 10- to 14-fold higher than those of the compounds upon which the bivalent structure was based, the MOR agonist oxymorphone and the mGluR5 antagonist MPEP. Coadministration of oxymorphone and MPEP demonstrated analgesic synergism, an interaction confirmed by isobolographic analysis. This study indicates that in the spared nerve injury-induced model of neuropathic pain, the 2 pharmacophores of the bivalent ligands MMG22 and MMG10 target MOR and mGluR5 as separate receptor monomers. The observed increase in the potency of MMG22 and MMG10, compared with oxymorphone and MPEP, may reflect the synergistic interaction of the 2 pharmacophores of the bivalent ligand acting at their respective separate receptor monomers.
Collapse
|
41
|
Nastase AF, Griggs NW, Anand JP, Fernandez TJ, Harland AA, Trask TJ, Jutkiewicz EM, Traynor JR, Mosberg HI. Synthesis and Pharmacological Evaluation of Novel C-8 Substituted Tetrahydroquinolines as Balanced-Affinity Mu/Delta Opioid Ligands for the Treatment of Pain. ACS Chem Neurosci 2018; 9:1840-1848. [PMID: 29677442 PMCID: PMC9976708 DOI: 10.1021/acschemneuro.8b00139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The use of opioids for the treatment of pain, while largely effective, is limited by detrimental side effects including analgesic tolerance, physical dependence, and euphoria, which may lead to opioid abuse. Studies have shown that compounds with a μ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist profile reduce or eliminate some of these side effects including the development of tolerance and dependence. Herein we report the synthesis and pharmacological evaluation of a series of tetrahydroquinoline-based peptidomimetics with substitutions at the C-8 position. Relative to our lead peptidomimetic with no C-8 substitution, this series affords an increase in DOR affinity and provides greater balance in MOR and DOR binding affinities. Moreover, compounds with carbonyl moieties at C-8 display the desired MOR agonist/DOR antagonist profile whereas alkyl substitutions elicit modest DOR agonism. Several compounds in this series produce a robust antinociceptive effect in vivo and show antinociceptive activity for greater than 2 h after intraperitoneal administration in mice.
Collapse
Affiliation(s)
- Anthony F. Nastase
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States,Interdepartmental Program in Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor Michigan 48109, United States
| | - Nicholas W. Griggs
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica P. Anand
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States,Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Thomas J. Fernandez
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aubrie A. Harland
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States,Interdepartmental Program in Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor Michigan 48109, United States
| | - Tyler J. Trask
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Emily M. Jutkiewicz
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States,Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States,Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I. Mosberg
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States,Interdepartmental Program in Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor Michigan 48109, United States,Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Corresponding Author Phone: 734-764-8117. Fax: 734-763-5595.
| |
Collapse
|
42
|
Willemse T, Eiselt E, Hollanders K, Schepens W, van Vlijmen HWT, Chung NN, Blais V, Holleran B, Longpré JM, Schiller PW, Maes BUW, Sarret P, Gendron L, Ballet S. Chemical space screening around Phe 3 in opioid peptides: Modulating µ versus δ agonism by Suzuki-Miyaura cross-couplings. Bioorg Med Chem Lett 2018; 28:2320-2323. [PMID: 29853330 PMCID: PMC6005765 DOI: 10.1016/j.bmcl.2018.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 01/10/2023]
Abstract
In this study, affinities and activities of derivatized analogues of Dmt-dermorphin[1-4] (i.e. Dmt-d-Ala-Phe-GlyNH2, Dmt = 2',6'-dimethyl-(S)-tyrosine) for the µ opioid receptor (MOP) and δ opioid receptor (DOP) were evaluated using radioligand binding studies, functional cell-based assays and isolated organ bath experiments. By means of solid-phase or solution-phase Suzuki-Miyaura cross-couplings, various substituted regioisomers of the phenylalanine moiety in position 3 of the sequence were prepared. An 18-membered library of opioid tetrapeptides was generated via screening of the chemical space around the Phe3 side chain. These substitutions modulated bioactivity, receptor subtype selectivity and highly effective ligands with subnanomolar binding affinities, contributed to higher functional activities and potent analgesic actions. In search of selective peptidic ligands, we show here that the Suzuki-Miyaura reaction is a versatile and robust tool which could also be deployed elsewhere.
Collapse
Affiliation(s)
- Tom Willemse
- Research Group of Organic Chemistry, Departments of Bioengineering Sciences and Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Organic Synthesis Division, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Emilie Eiselt
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Karlijn Hollanders
- Research Group of Organic Chemistry, Departments of Bioengineering Sciences and Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Organic Synthesis Division, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Wim Schepens
- Discovery Sciences, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Herman W T van Vlijmen
- Discovery Sciences, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Nga N Chung
- Laboratory of Chemical Biology and Peptide Research, Montreal Clinical Research Institute, 110 avenue des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Véronique Blais
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Brain Holleran
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Peter W Schiller
- Laboratory of Chemical Biology and Peptide Research, Montreal Clinical Research Institute, 110 avenue des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Bert U W Maes
- Organic Synthesis Division, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| | - Louis Gendron
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, 3001 12(e)Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Bioengineering Sciences and Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
43
|
Turnaturi R, Montenegro L, Marrazzo A, Parenti R, Pasquinucci L, Parenti C. Benzomorphan skeleton, a versatile scaffold for different targets: A comprehensive review. Eur J Med Chem 2018; 155:492-502. [PMID: 29908442 DOI: 10.1016/j.ejmech.2018.06.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/23/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022]
Abstract
Despite the fact that the benzomorphan skeleton has mainly been employed in medicinal chemistry for the development of opioid analgesics, it is a versatile structure. Its stereochemistry, as well as opportune modifications at the phenolic hydroxyl group and at the basic nitrogen, play a pivotal role addressing the benzomorphan-based compounds to a specific target. In this review, we describe the structure activity-relationships (SARs) of benzomorphan-based compounds acting at sigma 1 receptor (σ1R), sigma 2 receptor (σ2R), voltage-dependent sodium channel, N-Methyl-d-Aspartate (NMDA) receptor-channel complex and other targets. Collectively, the SARs data have highlighted that the benzomorphan nucleus could be regarded as a useful template for the synthesis of drug candidates for different targets.
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 6, 95100, Catania, Italy.
| | - Lucia Montenegro
- Department of Drug Sciences, Pharmaceutical Technology Section, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 6, 95100, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 6, 95100, Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology Section, University of Catania, Viale A. Doria, 6, 95100, Catania, Italy
| |
Collapse
|
44
|
Synthesis and Structure-Activity Relationships of (-)- cis- N-Normetazocine-Based LP1 Derivatives. Pharmaceuticals (Basel) 2018; 11:ph11020040. [PMID: 29734749 PMCID: PMC6027146 DOI: 10.3390/ph11020040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/25/2022] Open
Abstract
(−)-cis-N-Normetazocine represents a rigid scaffold able to mimic the tyramine moiety of endogenous opioid peptides, and the introduction of different N-substituents influences affinity and efficacy of respective ligands at MOR (mu opioid receptor), DOR (delta opioid receptor), and KOR (kappa opioid receptor). We have previously identified LP1, a MOR/DOR multitarget opioid ligand, with an N-phenylpropanamido substituent linked to (−)-cis-N-Normetazocine scaffold. Herein, we report the synthesis, competition binding and calcium mobilization assays of new compounds 10⁻16 that differ from LP1 by the nature of the N-substituent. In radioligand binding experiments, the compounds 10⁻13, featured by an electron-withdrawing or electron-donating group in the para position of phenyl ring, displayed improved affinity for KOR (Ki = 0.85⁻4.80 μM) in comparison to LP1 (7.5 μM). On the contrary, their MOR and DOR affinities were worse (Ki = 0.18⁻0.28 μM and Ki = 0.38⁻1.10 μM, respectively) with respect to LP1 values (Ki = 0.049 and 0.033 μM). Analogous trends was recorded for the compounds 14⁻16, featured by indoline, tetrahydroquinoline, and diphenylamine functionalities in the N-substituent. In calcium mobilization assays, the compound 10 with a p-fluorophenyl in the N-substituent shared the functional profile of LP1 (pEC50MOR = 7.01), although it was less active. Moreover, the p-methyl- (11) and p-cyano- (12) substituted compounds resulted in MOR partial agonists and DOR/KOR antagonists. By contrast, the derivatives 13⁻15 resulted as MOR antagonists, and the derivative 16 as a MOR/KOR antagonist (pKBMOR = 6.12 and pKBKOR = 6.11). Collectively, these data corroborated the critical role of the N-substituent in (−)-cis-N-Normetazocine scaffold. Thus, the new synthesized compounds could represent a template to achieve a specific agonist, antagonist, or mixed agonist/antagonist functional profile.
Collapse
|
45
|
Chemical Diversity in the G Protein-Coupled Receptor Superfamily. Trends Pharmacol Sci 2018; 39:494-512. [PMID: 29576399 DOI: 10.1016/j.tips.2018.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell signaling transmembrane proteins that can be modulated by a plethora of chemical compounds. Systematic cheminformatics analysis of structurally and pharmacologically characterized GPCR ligands shows that cocrystallized GPCR ligands cover a significant part of chemical ligand space, despite their limited number. Many GPCR ligands and substructures interact with multiple receptors, providing a basis for polypharmacological ligand design. Experimentally determined GPCR structures represent a variety of binding sites and receptor-ligand interactions that can be translated to chemically similar ligands for which structural data are lacking. This integration of structural, pharmacological, and chemical information on GPCR-ligand interactions enables the extension of the structural GPCR-ligand interactome and the structure-based design of novel modulators of GPCR function.
Collapse
|
46
|
Development of novel LP1-based analogues with enhanced delta opioid receptor profile. Bioorg Med Chem 2017; 25:4745-4752. [PMID: 28734666 DOI: 10.1016/j.bmc.2017.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022]
Abstract
Pain relief achieved by co-administration of drugs acting at different targets is more effective than that obtained with conventional MOR selective agonists usually associated to relevant side effects. It has been demonstrated that simultaneously targeting different opioid receptors is a more effective therapeutic strategy. Giving the promising role for DOR in pain management, novel LP1-based analogues with different N-substituents were designed and synthesized with the aim to improve DOR profile. For this purpose, we maintained the phenyl ring in the N-substituent of 6,7-benzomorphan scaffold linked to an ethyl spacer bearing a hydroxyl/methyl or methoxyl group at carbon 2 or including it in a 1,4-benzodioxane ring. LP1 analogues were tested by competition binding assays. Compounds 6 (KiMOR=2.47nM, KiDOR=9.6nM), 7 (KiMOR=0.5nM and KiDOR=0.8nM) and 9 (KiMOR=1.08nM, KiDOR=6.6nM) retained MOR affinity but displayed an improved DOR binding capacity as compared to LP1 (KiMOR=0.83nM, KiDOR=29.1nM). Moreover, GPI and MVD functional assays indicated that compounds 6 (IC50=49.2 and IC50=10.8nM), 7 (IC50=9.9 and IC50=11.8nM) and 9 (IC50=21.5 and IC50=4.4nM) showed a MOR/DOR agonist profile, unlike LP1 that was a MOR agonist/DOR antagonist (IC50=1.9 and IC50=1240nM). Measurements of their antinociceptive effect was evaluated by mice radiant tail flick test displaying for compounds 6, 7 and 9 ED50 values of 1.3, 1.0 and 0.9mg/kg, i.p., respectively. Moreover, the antinociceptive effect of compound 9 was longer lasting with respect to LP1. In conclusion the N-substituent nature of compounds 6, 7 and 9 shifts the DOR profile of LP1 from antagonism to agonism.
Collapse
|
47
|
He C, Li H, Zhang J, Kang Y, Jia F, Dong S, Zhou L. Supraspinal inhibitory effects of chimeric peptide MCRT on gastrointestinal motility in mice. J Pharm Pharmacol 2017. [DOI: 10.1111/jphp.12761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Objectives
Chimeric peptide MCRT, based on morphiceptin and PFRTic-NH2, was a bifunctional ligand of μ- and δ-opioid receptors (MOR-DOR) and produced potent analgesia in tail-withdrawal test. The study focused on the supraspinal effects of morphiceptin, PFRTic-NH2 and MCRT on gastrointestinal motility. Moreover, opioid receptor antagonists, naloxone (non-selective), cyprodime (MOR selective) and naltrindole (DOR selective) were utilized to explore the mechanisms.
Methods
Intracerebroventricular administration was achieved via the implanted cannula. Gastric emptying and intestinal transit were measured to evaluate gastrointestinal motility.
Key findings
(1) At supraspinal level, morphiceptin, PFRTic-NH2 and MCRT significantly decreased gastric emptying and intestinal transit; (2) MCRT at 1 nmol/mouse, far higher than its analgesic dose (ED50 = 29.8 pmol/mouse), failed to regulate the gastrointestinal motility; (3) MCRT-induced gastrointestinal dysfunction could be completely blocked by naloxone and naltrindole, but not affected by cyprodime.
Conclusions
(1) Morphiceptin and PFRTic-NH2 played important roles in the regulation of gastrointestinal motility; (2) MCRT possessed higher bioactivity of pain relief than gastrointestinal regulation, suggesting its promising analgesic property; (3) MCRT-induced motility disorders were sensitive to DOR but not to MOR blockade, indicating the pain-relieving specificity of speculated MOR subtype or splice variant or MOR-DOR heterodimer.
Collapse
Affiliation(s)
- Chunbo He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hailan Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jing Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yanping Kang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Fang Jia
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shouliang Dong
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China
| | - Lanxia Zhou
- The Core Laboratory of the First Affiliated Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou, China
| |
Collapse
|
48
|
Yu F, Zhao ZY, He T, Yu YQ, Li Z, Chen J. Temporal and spatial dynamics of peripheral afferent-evoked activity in the dorsal horn recorded in rat spinal cord slices. Brain Res Bull 2017; 131:183-191. [PMID: 28458040 DOI: 10.1016/j.brainresbull.2017.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 02/13/2017] [Accepted: 04/21/2017] [Indexed: 01/07/2023]
Abstract
In the present study, multi-electrode array recording was used to examine dorsal horn activity following stimulation of primary afferents in a rat dorsal root attached-spinal cord slice preparation. The multi-electrode array probe was placed under the dorsal horn slice and local field potentials evoked by stimulation on the dorsal root were analyzed. Three kinds of dorsal root-evoked responses were identified. In lamina IIo, local field potentials exhibited P1 (peak latency 1.46±0.08ms), N1 (2.77±0.18ms, n=12), N2 (7.31±0.48ms), N3 (12.12±0.73ms) and P2(18.30±0.80ms) waves. In lamina IIi local field potentials exhibited P (1.99±0.10ms), N1 (3.35±0.17ms) and N2 (8.58±0.44ms) waves. In laminae III-VI, local field potentials exhibited P1 (3.01±0.07ms), P2 (7.02±0.21ms) and N waves (22.57±0.79ms). Sweep spread was calculated by two dimensional current source density (2D-CSD) analysis. Both α-amino-3-hydroxy-5-methylisoxazole-4-propionic a/kainate and N-methyl-d-aspartate-type glutamate receptors participated in this neuronal circuitry. Morphine diminished local field potentials. Gabapentin diminished the negative components in lamina II and P2 component in lamina IIo, but increased the positive components in lamina IIi and laminae III-VI. The present study revealed that functional dorsal horn activity was preserved in the spinal cord slice preparation. Glutamatergic synapses were crucially involved in information processing. Opioid interneurons and gabapentin may play a modulatory role in regulating signal flows in the dorsal horn. Taken together, these results identify a spatio-temporal profile of dorsal horn activity evoked by dorsal root stimulation, and implicate glutamatergic and opioidergic receptors and gabapentin in this activity.
Collapse
Affiliation(s)
- Fang Yu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China; Jinan Military General Hospital, Jinan, Shandong Province, 250031, PR China
| | - Zhen-Yu Zhao
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China; Jinan Military General Hospital, Jinan, Shandong Province, 250031, PR China
| | - Ting He
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China
| | - Yao-Qing Yu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, PR China.
| |
Collapse
|
49
|
Wang ZL, Pan JX, Song JJ, Tang HH, Yu HP, Li XH, Li N, Zhang T, Zhang R, Zhang MN, Xu B, Fang Q, Wang R. Structure-Based Optimization of Multifunctional Agonists for Opioid and Neuropeptide FF Receptors with Potent Nontolerance Forming Analgesic Activities. J Med Chem 2016; 59:10198-10208. [PMID: 27798836 DOI: 10.1021/acs.jmedchem.6b01181] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zi-Long Wang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Jia-Xin Pan
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Jing-Jing Song
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Hong-Hai Tang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Hong-Ping Yu
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Xu-Hui Li
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Ning Li
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Ting Zhang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Meng-Na Zhang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| | - Rui Wang
- Key Laboratory of Preclinical
Study for New Drugs of Gansu Province, and Institute of Physiology,
School of Basic Medical Sciences, Lanzhou University, 199 Donggang
West Road, Lanzhou, 730000, PR China
| |
Collapse
|
50
|
Vicario N, Parenti R, Arico' G, Turnaturi R, Scoto GM, Chiechio S, Parenti C. Repeated activation of delta opiod receptors counteracts nerve injury-induced TNF-α up-regulation in the sciatic nerve of rats with neuropathic pain: A possible correlation with delta opiod receptors-mediated antiallodinic effect. Mol Pain 2016; 12:12/0/1744806916667949. [PMID: 27590071 PMCID: PMC5024981 DOI: 10.1177/1744806916667949] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite mu opioid receptor agonists are the cornerstones of moderate-to-severe acute pain treatment, their effectiveness in chronic pain conditions is controversial. In contrast to mu opioid receptor agonists, a number of studies have reported the effectiveness of delta opioid receptor agonists on neuropathic pain strengthening the idea that delta opioid receptors gain importance when chronic pain develops. Among other effects, it has been shown that delta opioid receptor activation in optic nerve astrocytes inhibits tumor necrosis factor-α-mediated inflammation in response to severe hypoxia. Considering the involvement of tumor necrosis factor-α in the development and maintenance of neuropathic pain, with this study we sought to correlate the effect of delta opioid receptor agonist on the development of mechanical allodynia to tumor necrosis factor-α expression at the site of nerve injury in rats subjected to chronic constriction injury of the sciatic nerve. To this aim, we measured the levels of tumor necrosis factor-α in the sciatic nerve of rats with neuropathic pain after repeated injections with a delta opioid receptor agonist. Results obtained demonstrated that repeated administrations of the delta opioid receptor agonist SNC80 (10 mg/kg, i.p. for seven consecutive days) significantly inhibited the development of mechanical allodynia in rats with neuropathic pain and that the improvement of neuropathic symptom was timely related to the reduced expression of tumor necrosis factor-α in the rat sciatic nerve. We demonstrated also that when treatment with the delta opioid receptor agonist was suspended both allodynia and tumor necrosis factor-α up-regulation in the sciatic nerve of rats with neuropathic pain were restored. These results show that persistent delta opioid receptor activation significantly attenuates neuropathic pain and negatively regulates sciatic nerve tumor necrosis factor-α expression in chronic constriction injury rats.
Collapse
Affiliation(s)
| | | | | | | | | | - Santina Chiechio
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| | - Carmela Parenti
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| |
Collapse
|