scholarly journals pKa of opioid ligands as a discriminating factor for side effects

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Giovanna Del Vecchio ◽  
Dominika Labuz ◽  
Julia Temp ◽  
Viola Seitz ◽  
Michael Kloner ◽  
...  

AbstractThe non-selective activation of central and peripheral opioid receptors is a major shortcoming of currently available opioids. Targeting peripheral opioid receptors is a promising strategy to preclude side effects. Recently, we showed that fentanyl-derived μ-opioid receptor (MOR) agonists with reduced acid dissociation constants (pKa) due to introducing single fluorine atoms produced injury-restricted antinociception in rat models of inflammatory, postoperative and neuropathic pain. Here, we report that a new double-fluorinated compound (FF6) and fentanyl show similar pKa, MOR affinity and [35S]-GTPγS binding at low and physiological pH values. In vivo, FF6 produced antinociception in injured and non-injured tissue, and induced sedation and constipation. The comparison of several fentanyl derivatives revealed a correlation between pKa values and pH-dependent MOR activation, antinociception and side effects. An opioid ligand’s pKa value may be used as discriminating factor to design safer analgesics.

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 463.1-463
Author(s):  
Y. M. S. Pires ◽  
M. C. Leal de Moura ◽  
W. Amorim Dias ◽  
V. D. Pimentel

Background:The pharmacological approaches of chronic pain are a challenge in the clinical context. Currently, only palliative treatments are performed. The α-phellandrene (α-phel) is a cyclic monoterpene found in essential oils of aromatic plants, which presents several biological activities, such as antinociceptive, antihyperalgesic and immunostimulant.1,2Objectives:This study aimed to investigate the action of α-phellandrene in chronic pain throughin silicoandin vivoapproaches, aiming to develop a new therapeutic option for painful conditions, reducing analgesic doses and side effects.Methods:The pharmacokinetic analysis of α-phel was performed by PreADMET online server. The software ACD/ChemSketch 14.0 was used to optimize the 3D structure of α-phel. Molecular docking was performed with the software AutoDock Tools 1.5.6 to evaluate the pharmacodynamics interactions of α-phel and opioid receptors.The mechanism of action of α-phel in chronic pain was analyzedin vivo. Ethics Committee of UFPI approved this project (protocol n° 305/17). Female Swiss mice (25-30 g) underwent partial sciatic nerve ligation surgery to induce neuropathy. The neuropathic mice (N=6) were pre-treated with Naloxone (2 mg/kg, i.p.) or Saline (10 mL/kg, p.o.). After 20 minutes, they were treated with α-phel (6,25 mg/kg, p.o.) or morphine (5 mg/kg, i.p.) and evaluated by Von Frey test.Results:The predicted pharmacokinetic parameters (Table 1) suggest good intestinal absorption and good permeability. Plasma protein binding is elevated, however, it is reversible and technological alternatives, such as carrier systems, can improve it. The α-phel does not inhibit CYP3A4, it indicates a minimal possibility of interactions with others drugs and adverse reactions.Table 1.Pharmacokinetic parameters of α-phelIDVALUEBBB7.17054Buffer_solubility_mg_L1227.08Caco223.4164CYP_2C19 and 2C9_inhibitionInhibitorCYP_2D6_inhibition_substrateNonCYP_3A4_inhibitionNonCYP_3A4_substrateWeaklyHIA100.00000MDCK267.707Pgp_inhibitionNonPlasma_Protein_Binding90.00000Pure_water_solubility141.466The structure of α-phel binding opioid receptors is shown in Figure 1. The lowest ligand-receptor binding energies were, respectively: -6.0 kca/mol, -6.6 kcal/mol and -7.4 kcal/mol for the interaction of α-phel with Mu, Kappa and Delta receptors. It indicates that α-phel has high affinity for all three opioid receptors, binding in a strong and stable way.Figure 1.Graphical 3D representation of the binding modes of α-phellandrene with opioid receptors: A - Mu; B - Kappa; C – DeltaThe analgesic potential of the substance was testedin vivoas well. It was observed that Naloxone, an opioid antagonist, significantly reversed the effect of α-phel, indicating that it displays antinociceptive and antihyperalgesic activity through opioid system.Conclusion:The monoterpene α-phel presents antinociceptive activity and reduces the sensitivity in chronic pain through the activation of opioid receptors.Thus,in vivoandin silicoresults indicate that α-phel is an analgesic opioid agonist. This work may guide further preclinical studies, since α-phel may be an important strategy to treat chronic pain, with fewer side effects, dependence and tolerance than conventional drugs.References:[1]Nascimento AF, Camara CA, Moraes MM, Ramos CS. Essential oil composition and acaricidal activity of Schinusterebinthifolius from Atlantic Forest of Pernambuco, Brazil against Tetranychusurticae. Natural product communications. 2012 Jan:7(1):129-132.[2]Piccinelli AC, Santos JA, Konkiewitz EC, et al. Antihyperalgesic and antidepressive actions of (R)-(+)-limonene, α-phellandrene, and essential oil from Schinusterebinthifolius fruits in a neuropathic pain model. Nutritional neuroscience. 2015 Jul 1;18(5):217-24.Disclosure of Interests: :None declared


eLife ◽  
2017 ◽  
Vol 6 ◽  
Author(s):  
Sara González-Rodríguez ◽  
Mohiuddin A Quadir ◽  
Shilpi Gupta ◽  
Karolina A Walker ◽  
Xuejiao Zhang ◽  
...  

Novel painkillers are urgently needed. The activation of opioid receptors in peripheral inflamed tissue can reduce pain without central adverse effects such as sedation, apnoea, or addiction. Here, we use an unprecedented strategy and report the synthesis and analgesic efficacy of the standard opioid morphine covalently attached to hyperbranched polyglycerol (PG-M) by a cleavable linker. With its high-molecular weight and hydrophilicity, this conjugate is designed to selectively release morphine in injured tissue and to prevent blood-brain barrier permeation. In contrast to conventional morphine, intravenous PG-M exclusively activated peripheral opioid receptors to produce analgesia in inflamed rat paws without major side effects such as sedation or constipation. Concentrations of morphine in the brain, blood, paw tissue, and in vitro confirmed the selective release of morphine in the inflamed milieu. Thus, PG-M may serve as prototype of a peripherally restricted opioid formulation designed to forego central and intestinal side effects.


2020 ◽  
Vol 20 (31) ◽  
pp. 2866-2877
Author(s):  
Hirokazu Mizoguchi ◽  
Hideaki Fujii

Since a μ-opioid receptor gene containing multiple exons has been identified, the variety of splice variants for μ-opioid receptors have been reported in various species. Amidino-TAPA and IBNtxA have been discovered as new analgesics with different pharmacological profiles from morphine. These new analgesics show a very potent analgesic effect but do not have dependence liability. Interestingly, these analgesics show the selectivity to the morphine-insensitive μ-opioid receptor splice variants. The splice variants, sensitive to these new analgesics but insensitive to morphine, may be a better molecular target to develop the analgesics without side effects.


2016 ◽  
Vol 124 (3) ◽  
pp. 706-720 ◽  
Author(s):  
Vinod Tiwari ◽  
Fei Yang ◽  
Shao-Qiu He ◽  
Ronen Shechter ◽  
Chen Zhang ◽  
...  

Abstract Background Opioids have long been regarded as the most effective drugs for the treatment of severe acute and chronic pain. Unfortunately, their therapeutic efficacy and clinical utility have been limited because of central and peripheral side effects. Methods To determine the therapeutic value of peripheral μ-opioid receptors as a target for neuropathic pain treatment, the authors examined the effects of dermorphin [d-Arg2, Lys4] (1–4) amide (DALDA), a hydrophilic, peripherally acting μ-opioid receptor agonist, in male and female rats with spinal nerve ligation–induced neuropathic pain. The authors also utilized behavioral, pharmacologic, electrophysiologic, and molecular biologic tools to characterize DALDA’s possible mechanisms of action in male rats. Results DALDA, administered subcutaneously, had 70 times greater efficacy for inhibiting thermal (n = 8 to 11/group) than mechanical hypersensitivity (n = 6 to 8/group) in male rats. The pain inhibitory effects of DALDA on mechanical and heat hypersensitivity were abolished in animals pretreated with systemic methylnaltrexone (n = 7 to 9/group), a peripheral μ-opioid receptor antagonist. In the spinal wide-dynamic range neurons, systemic DALDA inhibited C-fiber–mediated, but not A-fiber–mediated, response in neuropathic male rats (n = 13). In primary sensory neurons, DALDA inhibited the capsaicin-induced [Ca2+] increase more than the β-alanine–induced [Ca2+] increase (n = 300); capsaicin and β-alanine activate subpopulations of neurons involved in the signaling of heat and mechanical pain, respectively. DALDA-treated rats (n = 5 to 8/group) did not exhibit motor deficits and locomotor impairment suggesting that it does not induce central side effects. Conclusions These findings suggest that DALDA may represent a potential alternative to current opioid therapy for the treatment of neuropathic pain and is likely to be associated with minimal adverse effects.


1977 ◽  
Vol 37 (01) ◽  
pp. 154-161 ◽  
Author(s):  
B. A Janik ◽  
S. E Papaioannou

SummaryUrokinase, streptokinase, Brinase, trypsin, and SN 687, a bacterial exoprotease, have been evaluated in an ex vivo assay system. These enzymes were injected into rabbits and the fibrinolytic activity as well as other coagulation parameters were measured by in vitro techniques. Dose-response correlations have been made using the euglobulin lysis time as a measure of fibrinolytic activity and the 50% effective dose has been determined for each enzyme. Loading doses, equal to four times the 50% effective dose, were administered to monitor potential toxicity revealing that Brinase, trypsin, and SN 687 were very toxic at this concentration.Having established the 50% effective dose for each enzyme, further testing was conducted where relevant fibrinolytic and coagulation parameters were measured for up to two days following a 50% effective dose bolus injection of each enzyme. Our results have demonstrated that urokinase and streptokinase are plasminogen activators specifically activating the rabbit fibrinolytic system while Brinase, trypsin and SN 687 increase the general proteolytic activity in vivo.The advantages of this ex vivo assay system for evaluating relative fibrinolytic potencies and side effects for plasminogen activators and fibrinolytic proteases have been discussed.


1984 ◽  
Vol 51 (02) ◽  
pp. 248-253 ◽  
Author(s):  
R J Dupe ◽  
P D English ◽  
R A G Smith ◽  
J Green

SummaryA quantitative model of venous thrombosis in the beagle dog is described. The model was adapted to permit ageing of isolated experimental clots in vivo. A model of acute pulmonary embolism in this species is also described. In the venous thrombosis model, infusion of streptokinase (SK) or SK-activated human plasmin gave significant lysis but bolus doses of SK. plasmin complex were ineffective. Active site anisoylated derivatives of SK. plasminogen complex, SK-activated plasmin and activator-free plasmin were all active when given as bolus doses in both models. At lytic doses, the acyl-enzymes caused fewer side-effects attributable to plasminaemia than the corresponding unmodified enzymes.


2019 ◽  
Author(s):  
Hamilton Lee ◽  
Jenica Lumata ◽  
Michael A. Luzuriaga ◽  
Candace Benjamin ◽  
Olivia Brohlin ◽  
...  

<div><div><div><p>Many contrast agents for magnetic resonance imaging are based on gadolinium, however side effects limit their use in some patients. Organic radical contrast agents (ORCAs) are potential alternatives, but are reduced rapidly in physiological conditions and have low relaxivities as single molecule contrast agents. Herein, we use a supramolecular strategy where cucurbit[8]uril binds with nanomolar affinities to ORCAs and protects them against biological reductants to create a stable radical in vivo. We further over came the weak contrast by conjugating this complex on the surface of a self-assembled biomacromolecule derived from the tobacco mosaic virus.</p></div></div></div>


2018 ◽  
Vol 25 (21) ◽  
pp. 2503-2519 ◽  
Author(s):  
Anne Kokel ◽  
Marianna Torok

Background: Since the first isolation of antimicrobial peptides (AMPs) they have attracted extensive interest in medicinal chemistry. However, only a few AMP-based drugs are currently available on the market. Despite their effectiveness, biodegradability, and versatile mode of action that is less likely to induce resistance compared to conventional antibiotics, AMPs suffer from major issues that need to be addressed to broaden their use. Notably, AMPs can lack selectivity leading to side effects and cytotoxicity, and also exhibit in vivo instability. Several strategies are being actively considered to overcome the limitations that restrain the success of AMPs. Methods: In the current work, recent strategies reported for improving AMPs in the context of drug design and delivery were surveyed, and also their possible impact on patients and the environment was assessed. Results: As a major advantage AMPs possess an easily tunable skeleton offering opportunities to improve their properties. Strategic structural modifications and the beneficial properties of cyclic or branched AMPs in term of stability have been reported. The conjugation of AMPs with nanoparticles has also been explored to increase their in vivo stability. Other techniques such as the coupling of AMPs with specific antibodies aim to increase the selectivity of the potential drug towards the target. These strategies were evaluated for their effect on the environment highlighting green technologies. Conclusion: Although further research is needed taking into account both environmental and human health consequences of novel AMPs, several of these compounds are promising drug candidates for use in sustainable medicine.


2019 ◽  
Vol 20 (12) ◽  
pp. 1227-1243
Author(s):  
Hina Qamar ◽  
Sumbul Rehman ◽  
D.K. Chauhan

Cancer is the second leading cause of morbidity and mortality worldwide. Although chemotherapy and radiotherapy enhance the survival rate of cancerous patients but they have several acute toxic effects. Therefore, there is a need to search for new anticancer agents having better efficacy and lesser side effects. In this regard, herbal treatment is found to be a safe method for treating and preventing cancer. Here, an attempt has been made to screen some less explored medicinal plants like Ammania baccifera, Asclepias curassavica, Azadarichta indica, Butea monosperma, Croton tiglium, Hedera nepalensis, Jatropha curcas, Momordica charantia, Moringa oleifera, Psidium guajava, etc. having potent anticancer activity with minimum cytotoxic value (IC50 >3μM) and lesser or negligible toxicity. They are rich in active phytochemicals with a wide range of drug targets. In this study, these medicinal plants were evaluated for dose-dependent cytotoxicological studies via in vitro MTT assay and in vivo tumor models along with some more plants which are reported to have IC50 value in the range of 0.019-0.528 mg/ml. The findings indicate that these plants inhibit tumor growth by their antiproliferative, pro-apoptotic, anti-metastatic and anti-angiogenic molecular targets. They are widely used because of their easy availability, affordable price and having no or sometimes minimal side effects. This review provides a baseline for the discovery of anticancer drugs from medicinal plants having minimum cytotoxic value with minimal side effects and establishment of their analogues for the welfare of mankind.


Sign in / Sign up

Export Citation Format

Share Document