Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lorella Pasquinucci is active.

Publication


Featured researches published by Lorella Pasquinucci.


Journal of Medicinal Chemistry | 2011

Novel Potent and Selective σ Ligands: Evaluation of Their Agonist and Antagonist Properties

Agostino Marrazzo; Enrique J. Cobos; Carmela Parenti; Giuseppina Aricò; Giuseppina Marrazzo; Simone Ronsisvalle; Lorella Pasquinucci; Orazio Prezzavento; Nicola Antonio Colabufo; Marialessandra Contino; Luis Garrido González; Giovanna M. Scoto; Giuseppe Ronsisvalle

Novel enantiomers and diastereoisomers structurally related to σ ligand (+)-MR200 were synthesized to improve σ(1)/σ(2) subtype selectivity. The selective σ(1) ligand (-)-8 showed an antagonist profile determined by phenytoin differential modulation of binding affinity in vitro, confirmed in vivo by an increase of κ opioid analgesia. The σ(2) ligand (-)-9 displayed agonist properties in an in vitro isolated organ bath assay and antiproliferative effects on LNCaP and PC3 prostate cancer cell lines.


Life Sciences | 2012

Antinociceptive profile of LP1, a non-peptide multitarget opioid ligand

Carmela Parenti; Rita Turnaturi; Giuseppina Aricò; Agostino Marrazzo; Orazio Prezzavento; Simone Ronsisvalle; Giovanna M. Scoto; Giuseppe Ronsisvalle; Lorella Pasquinucci

AIMS Opioid drugs are the principal treatment option for moderate to severe pain and exert their biological effects through interactions with opioid receptors that are widely distributed throughout the CNS and peripheral tissues. Ligands capable of simultaneously targeting different receptors could be successful candidates for the treatment of chronic pain. Enhanced antinociception coupled with a low incidence of side effects has been demonstrated for ligands possessing mixed mu-opioid receptor (MOR) and delta-opioid receptor (DOR) activity. We previously reported that 3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-phenylpropanamide (LP1) acted as a MOR-DOR ligand in in vitro functional assays and moreover this drug produced a valid antinociception that was longer lasting than that of morphine. The aim of this work was to determine whether the antinociceptive effect produced by LP1 was central or peripheral and to assess which opioid receptor subtypes are involved in its effects. MAIN METHODS We explored the effects of naloxone methiodide (NX-M), a quaternary opioid antagonist, administered either intracerebroventricularly (i.c.v.) or subcutaneously (s.c.), on LP1-mediated antinociception in male Sprague-Dawley rats. In addition, we administered s.c. selective antagonists for MOR, DOR and kappa-opioid receptor (KOR) to investigate the effects of LP1. To characterise this drugs DOR profile better, we also investigated the effects of LP1 on DPDPE, a selective DOR agonist. KEY FINDINGS Data obtained by tail flick test showed that LP1 induced predominantly MOR-mediated supraspinal antinociception and was able to counteract DPDPE analgesia. SIGNIFICANCE LP1, a multitarget opioid ligand, is a supraspinal acting antinociceptive agent that is useful for the treatment of chronic pain.


Journal of Medicinal Chemistry | 2013

Design and synthesis of new bifunctional sigma-1 selective ligands with antioxidant activity.

Orazio Prezzavento; Emanuela Arena; Carmela Parenti; Lorella Pasquinucci; Giuseppina Aricò; Giovanna M. Scoto; S. Grancara; A. Toninello; Simone Ronsisvalle

Herein we report the synthesis of new bifunctional sigma-1 (σ1)-selective ligands with antioxidant activity. To achieve this goal, we combined the structure of lipoic acid, a universal antioxidant, with an appropriate sigma aminic moiety. Ligands 14 and 26 displayed high affinity and selectivity for σ1 receptors (Kiσ1 = 1.8 and 5.5 nM; Kiσ2/σ1 = 354 and 414, respectively). Compound 26 exhibited in vivo antiopioid effects on kappa opioid (KOP) receptor-mediated analgesia. In rat liver and brain mitochondria (RLM, RBM), this compound significantly reduced the swelling and the oxidation of thiol groups induced by calcium ions. Our results demonstrate that the tested compound has protective effects against oxidative stress.


Life Sciences | 2012

The benzomorphan-based LP1 ligand is a suitable MOR/DOR agonist for chronic pain treatment

Lorella Pasquinucci; Carmela Parenti; Rita Turnaturi; Giuseppina Aricò; Agostino Marrazzo; Orazio Prezzavento; Simone Ronsisvalle; Zafiroula Georgoussi; Danai-Dionysia Fourla; Giovanna M. Scoto; Giuseppe Ronsisvalle

AIMS Powerful analgesics relieve pain primarily through activating mu opioid receptor (MOR), but the long-term use of MOR agonists, such as morphine, is limited by the rapid development of tolerance. Recently, it has been observed that simultaneous stimulation of the delta opioid receptor (DOR) and MOR limits the incidence of tolerance induced by MOR agonists. 3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-phenylpropanamide (LP1) is a centrally acting agent with antinociceptive activity comparable to morphine and is able to bind and activate MOR and DOR. The aim of this work was to evaluate and compare the induction of tolerance to antinociceptive effects from treatment with LP1 and morphine. MAIN METHODS Here, we evaluated the pharmacological effects of LP1 administered at a dose of 4 mg/kg subcutaneously (s.c.) twice per day for 9 days to male Sprague-Dawley rats. In addition, the LP1 mechanism of action was assessed by measurement of LP1-induced [(35)S]GTPγS binding to the MOR and DOR. KEY FINDINGS Data obtained from the radiant heat tail flick test showed that LP1 maintained its antinociceptive profile until the ninth day, while tolerance to morphine (10mg/kg s.c. twice per day) was observed on day 3. Moreover, LP1 significantly enhanced [(35)S]GTPγS binding in the membranes of HEK293 cells expressing either the MOR or the DOR. SIGNIFICANCE LP1 is a novel analgesic agent for chronic pain treatment, and its low tolerance-inducing capability may be correlated with its ability to bind both the MOR and DOR.


Bioorganic & Medicinal Chemistry | 2000

Substituted 1-phenyl-2-cyclopropylmethylamines with high affinity and selectivity for sigma sites

Giuseppe Ronsisvalle; Agostino Marrazzo; Orazio Prezzavento; Lorella Pasquinucci; Barbara Falcucci; Rosanna Di Toro; Santi Spampinato

A series of 1-phenyl-2-cyclopropylmethylamines structurally related to (+)- and (-)-MPCB were synthesized and their binding affinities for sigma1, sigma2, opioid and dopamine (D2) receptors were evaluated. Substitution of the cis-N-normetazocine with different aminic moieties provided compounds with high affinity and selectivity for sigma binding sites with respect to opioid and dopamine (D2) receptors. The observed increase in sigma2 affinity as compared to the parent (+)-MPCB, supports the idea that the particular stereochemistry of (+)-cis-N-normetazocine affects sigma1 selectivity but does not affect sigma1 affinity. The (+/-)-cis isomers of methyl 2-[(1-adamantylamino)methyl]-1-phenylcyclopropane-1-carboxyl ate (18) displayed a higher affinity and selectivity for the sigma1 and sigma2 receptor subtypes compared to the (+/-)-trans 19. Interestingly, the enantiomer (-)-cis 18 displayed a preference for sigma1 receptor subtype whereas the (+)-cis 18 did for sigma2. These results prompt us to synthesize compounds with modification of nitrogen and carboxyl groups. The compounds obtained showed high affinities and selectivity for sigma sites. Moreover, modifications of carboxyl groups provided compounds with the highest affinities in the series. In particular, compound 25 with reverse-type ester showed a Ki of 0.6 and 4.05 nM for sigma1 and sigma2 binding sites, respectively.


European Journal of Medicinal Chemistry | 2016

Multitarget opioid ligands in pain relief: New players in an old game

Rita Turnaturi; Giuseppina Aricò; Giuseppe Ronsisvalle; Carmela Parenti; Lorella Pasquinucci

Still nowadays pain is one of the most common disabling conditions and yet it remains too often unsolved. Analgesic opioid drugs, and mainly MOR agonists such as morphine, are broadly employed for pain management. MOR activation, however, has been seen to cause not only analgesia but also undesired side effects. A potential pain treatment option is represented by the simultaneous targeting of different opioid receptors. In fact, ligands possessing multitarget capabilities led to an improved pharmacological fingerprint. This review focuses on the examination of multitarget opioid ligands which have been distinguished in peptide and non-peptide and further listed as bivalent and bifunctional ligands. Moreover, the potential of these compounds, both as analgesic drugs and pharmacological tools to explore heteromer receptors, has been stressed.


Farmaco | 2001

Synthesis and pharmacological evaluation of potent and enantioselective σ1 and σ2 ligands

Agostino Marrazzo; Orazio Prezzavento; Lorella Pasquinucci; Franco Vittorio; Giuseppe Ronsisvalle

Abstract In a previous study we found that substitutions of the (+)- cis - N -normetazocine nucleus of (+)- MPCB with 1-adamantanamine provide the compound (±)- 10 with high affinity and selectivity for σ receptors. Starting with this result we have synthesized a new series of eight 1-phenyl-2-cyclopropylmethylamines structurally related to (±)- 10 , and binding affinities, with respect to σ 1 , σ 2 , opioid and dopaminergic D 2 receptors, have been reported. All compounds showed a negligible opioid and dopaminergic affinity and high selectivity for σ receptors. Modifications on the amino moiety and methylcarboxyester group of 10 provide compounds with different σ 1 and σ 2 binding affinity and selectivity. Moreover, we have also synthesized the respective enantiomers of componds (±)- 10 and (±)- 18 in order to evaluate the enantioselectivity for σ 1 and σ 2 receptors. The binding data showed that carboxymethylester on the cyclopropane ring was more critical for enantioselectivity than the hydroxymethylenic group. In fact, the (−)- 10 enantiomer showed a preference for σ 1 whereas (+)- 10 showed a preference for σ 2 .


Life Sciences | 1995

CCB, a novel specific κ opioid agonist, which discriminates between opioid and σ1 recognition sites

Giuseppe Ronsisvalle; Orazio Prezzavento; Lorella Pasquinucci; Agostino Marrazzo; Franco Vittorio; J.A. Gomez-Vidal; Lucia Carboni; Santi Spampinato

Abstract CCB, 6,11 -Dimethyl-1,2,3,4,5,6-hexahydro-3-{[2′-methoxycarbonyl-2′-(4-chlorophenyl) cyclopropyl]methyl}-2,6-methano-3-benzazocin-8-ol, displays specificity and very high affinity for κ opioid receptor types (K 1 = 0.41 ± 0.19 nM). In contrast to other κ opioid agonists, CCB is also selective with respect to σ 1 sites (K 1 = 1,050 ± 55 nM). CCB displays antinociceptive and sedative effects in the mouse comparable to those of U50,488H and morphine. Naltrexone fully antagonizes these effects. The sedative effects of CCB are blocked in mice pretreated with naltrexone or nor-BNI. CCB and U50,488H produce a superimposable diuresis in rats. Naltrexone and nor-BNI, both are effective in antagonizing the effect. CCB does not produce any stereotyped behavior or ataxia in the behavioral assay in doses up to 40 mg/kg s.c., These findings suggest that CCB might be a useful tool to investigate the physiological role of κ opioid receptors.


Bioorganic & Medicinal Chemistry | 2017

Development of novel LP1-based analogues with enhanced delta opioid receptor profile

Lorella Pasquinucci; Rita Turnaturi; Orazio Prezzavento; Emanuela Arena; Giuseppina Aricò; Zafiroula Georgoussi; Rosalba Parenti; Giuseppina Cantarella; Carmela Parenti

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.


Bioorganic & Medicinal Chemistry | 2016

Evaluation of N-substituent structural variations in opioid receptor profile of LP1

Lorella Pasquinucci; Rita Turnaturi; Giuseppina Aricò; Carmela Parenti; Paschalina Pallaki; Zafiroula Georgoussi; Simone Ronsisvalle

The benzomorphan scaffold has great potential as lead structure and the nature of the N-substituent is able to influence affinity, potency, and efficacy at all three opioid receptors. Building upon these considerations, we synthesized a new series of LP1 analogues by introducing naphthyl or heteroaromatic rings in propanamide side chain of its N-substituent (9-15). In vitro competition-binding assays in HEK293 cells stably expressing MOR, DOR or KOR showed that in compound 9 the 1-naphthyl ring led to the retention of MOR affinity (Ki(MOR)=38±4nM) displaying good selectivity versus DOR and KOR. In the electrically stimulated GPI, compound 9 was inactive as agonist but produced an antagonist potency value (pA2) of 8.6 in presence of MOR agonist DAMGO. Moreover, subcutaneously administered it antagonized the antinociceptive effects of morphine with an AD50=2.0mg/kg in mouse-tail flick test. Modeling studies on MOR revealed that compound 9 fit very well in the binding pocket but in a different way in respect to the agonist LP1. Probably the replacement of its N-substituent on the III, IV and V TM domains reflects an antagonist behavior. Therefore, compound 9 could represent a potential lead to further develop antagonists as valid therapeutic agents and useful pharmacological tools to study opioid receptor function.

Collaboration


Dive into the Lorella Pasquinucci's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge