Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Amino Acids ; 56(1): 18, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38427104

ABSTRACT

A series of 10 cyclic, biaryl analogs of enkephalin, with Tyr or Phe residues at positions 1 and 4, were synthesized according to the Miyaura borylation and Suzuki coupling methodology. Biaryl bridges formed by side chains of the two aromatic amino acid residues are of the meta-meta, meta-para, para-meta, and para-para configuration. Conformational properties of the peptides were studied by CD and NMR. CD studies allowed only to compare conformations of individual peptides while NMR investigations followed by XPLOR calculations provided detailed information on their conformation. Reliability of the XPLOR calculations was confirmed by quantum chemical ones performed for one of the analogs. No intramolecular hydrogen bonds were found in all the peptides. They are folded and adopt the type IV ß-turn conformation. Due to a large steric strain, the aromatic carbon atoms forming the biaryl bond are distinctly pyramidalized. Seven of the peptides were tested in vitro for their affinity for the µ-opioid receptor.


Subject(s)
Enkephalins , Peptides, Cyclic , Cyclization , Reproducibility of Results , Enkephalins/chemistry , Protein Conformation , Peptides, Cyclic/chemistry
2.
ChemMedChem ; 19(7): e202300519, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38126948

ABSTRACT

Synthesis of molecular hybrids, obtained by combination of two or more pharmacophoric groups of different bioactive substances in order to produce more efficient drugs, is now a frequently used approach in medicinal chemistry. Following this strategy, we synthetized a library of 3-methylidene-1-tosyl-2,3-dihydro-1,8-naphthyridin-4(1H)-ones, combining a 1,8-naphthyridin-4-one motif with an exo-methylidene bond conjugated with a carbonyl group, pharmacophoric units that are present in many natural, biologically active compounds with anticancer potential. We reasoned that such bifunctional conjugates may have enhanced cytotoxic activity. The title compounds were synthesized in a four step reaction sequence. ß-Ketophosphonate, obtained from methyl N-tosylnicotinate and diethyl methylphosphonate, was reacted with various aldehydes giving 3-diethoxyphosphoryl-2,3-dihydro-1,8-naphthyridin-4(1H)-ones as keto-enol tautomers. Later, these compounds were transformed into 3-methylidene-1-tosyl-2,3-dihydro-1,8-naphthyridin-4(1H)-ones applying the Horner-Wadsworth-Emmons methodology. Then, the cytotoxicity of the new compounds was assessed on two cancer cell lines, promyelocytic leukemia HL-60 and breast cancer adenocarcinoma MCF-7, and for comparison, on human umbilical vein endothelial cells HUVEC. The most active and selective analog, 2-ethyl-3-methylidene-1-tosyl-2,3-dihydro-1,8-naphthyridin-4(1H)-one 4 a was chosen for more detailed studies on HL-60 cell line, to determine molecular mechanisms of its anticancer activity. It was shown that 4 a strongly inhibited proliferation and induced apoptosis which could be attributed to its ability to cause DNA damage.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Molecular Structure , Structure-Activity Relationship , Endothelial Cells , Antineoplastic Agents/chemistry , HL-60 Cells , Cell Proliferation
3.
ChemMedChem ; 18(23): e202300315, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37821725

ABSTRACT

The NK1 receptor (NK1R) is a molecular target for both approved and experimental drugs intended for a variety of conditions, including emesis, pain, and cancers. While contemplating modifications to the typical NK1R pharmacophore, we wondered whether the CF3 groups common for many NK1R ligands, could be replaced with some other moiety. Our attention was drawn by the SF5 group, and so we designed, synthesized, and tested ten novel SF5 -containing compounds for NK1R affinity. All analogues exhibit detectable NK1R binding, with the best of them, compound 5 a, (3-bromo-5-(pentafluoro-λ6 -sulfanyl)benzyl acetyl-L-tryptophanate) binding only slightly worse (IC50 =34.3 nM) than the approved NK1R-targeting drug, aprepitant (IC50 =27.7 nM). Molecular docking provided structural explanation of SAR. According to our analysis, the SF5 group in our compounds occupies a position similar to that of one of the CF3 groups of aprepitant as found in the crystal structure. Additionally, we checked whether the docking scoring function or energies derived from Fragment Molecular Orbital quantum chemical calculations may be helpful in explaining and predicting the experimental receptor affinities for our analogues. Both these methods produce moderately good results. Overall, this is the first demonstration of the utility of the SF5 group in the design of NK1R ligands.


Subject(s)
Pain , Receptors, Neurokinin-1 , Humans , Receptors, Neurokinin-1/metabolism , Aprepitant , Molecular Docking Simulation
4.
J Pept Sci ; 29(9): e3487, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36898693

ABSTRACT

The treatment of hard-to-heal chronic wounds is still a major medical problem and an economic and social burden. In this work, we examine the proregenerative potential of two peptides, G11 (a trypsin-resistant analogue of growth hormone-releasing hormone [GHRH]) and biphalin (opioid peptide), and their combination in vitro on human fibroblasts (BJ). G11, biphalin and their combination exhibited no toxicity against BJ cells. On the contrary, these treatments significantly stimulated proliferation and migration of fibroblasts. Under inflammatory conditions (LPS-induced BJ cells), we noticed that the tested peptides decreased the levels of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and interleukin 1ß (IL-1ß). This was correlated with diminished phosphorylation levels of p38 kinase, but not those of ERK1/2. We found also that G11, biphalin and their combination activated the ERK1/2 signalling pathway, which has been previously implicated in promigratory activity of some regeneration enhancers, including opioids or GHRH analogues. Potential application of their combination requires further work, in particular in vivo experiments, in which the organism-level relevance of the discussed cell-level effects would be proven and, additionally, analgesic action of the opioid ingredient could be quantified.


Subject(s)
Growth Hormone-Releasing Hormone , Opioid Peptides , Humans , Opioid Peptides/pharmacology , Growth Hormone-Releasing Hormone/pharmacology , Wound Healing , Fibroblasts
5.
Pharmacol Rep ; 75(2): 465-473, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36840824

ABSTRACT

BACKGROUND: G protein-coupled receptors (GPCRs) transduce external stimuli into the cell by G proteins via an allosteric mechanism. Agonist binding to the receptor stimulates GDP/GTP exchange within the heterotrimeric G protein complex, whereas recent structures of GPCR-G protein complexes revealed that the H5, S1 and S2 domains of Gα are involved in binding the active receptor, earlier studies showed that a short peptide analog derived from the C-terminus (H5) of the G protein transducin (Gt) is sufficient to stabilize rhodopsin in an active form. METHODS: We have used Molecular Dynamics simulations along with biological evaluation by means of radio-ligand binding assay to study the interactions between Gαi-derived peptide (G-peptide) and the µ-opioid receptor (µOR). RESULTS: Here, we show that a Gαi-derived peptide of 12 amino acids binds the µ-opioid receptor and acts as an allosteric modulator. The Gαi-derived peptide increases µOR affinity for its agonist morphine in a dose-dependent way. CONCLUSIONS: These results indicate that the GPCR-Gα peptide interaction observed so far for only rhodopsin can be extrapolated to µOR. In addition, we show that the C-terminal peptide of the Gαi subunit is sufficient to stabilize the active conformation of the receptor. Our approach opens the possibility to investigate the GPCR-G protein interface with peptide modification.


Subject(s)
Receptors, Opioid , Rhodopsin , Rhodopsin/chemistry , Rhodopsin/metabolism , Receptors, Opioid/metabolism , Peptides , Receptors, G-Protein-Coupled/metabolism , GTP-Binding Proteins/metabolism , Transducin/chemistry , Transducin/metabolism , Protein Binding
6.
Exp Mol Pathol ; 129: 104849, 2023 02.
Article in English | MEDLINE | ID: mdl-36526011

ABSTRACT

17-trifluoromethylphenyl trinor prostaglandin F2α (17-CF3PTPGF2α) was reported recently to exhibit in vitro and in vivo anticancer activity. Based solely on the results of in silico molecular docking, it was claimed that this compound is NK1 receptor (NK1R) antagonist and that its activity is through this receptor. In this contribution we show that 17-CF3PTPGF2α is only a very weak NK1R ligand (IC50 > 200 µM). In connection with that we discuss the issue of this compound's molecular target. Finally, we briefly narrate on the proper use of molecular docking in biomedical research.


Subject(s)
Dinoprost , Receptors, Neurokinin-1 , Ligands , Molecular Docking Simulation
7.
Mol Cell Neurosci ; 120: 103728, 2022 05.
Article in English | MEDLINE | ID: mdl-35421568

ABSTRACT

N-acetyl-L-tryptophan (NAT) has been often shown to have neuroprotective action and other biological activities. In many of papers devoted to this compound, NAT is referenced to as an 'NK1 receptor (NK1R) antagonist' or a 'substance P (SP) antagonist'. Some of the studies treated NAT as a tool compound to interrogate NK1R function in a particular condition. Surprisingly however, no biochemical data on NAT/NK1R interaction have been available. On testing NAT in radioligand receptor binding assay, we found that this compound displays no significant binding to either human or rat NK1R up to millimolar concentrations. In light of this, the repeated claim of NAT being NK1R antagonist is an error. The use of NAT as a tool compound in NK1R-related studies should be discontinued. Some of the conclusions regarding the involvement of SP/NK1R axis in acute CNS injury or neurodegenerative diseases may then require careful rethinking. On the other hand, given interesting neuroprotective properties of NAT, the issue whether it acts by specific interactions with some molecular target or by unspecific physicochemical mechanisms needs be investigated.


Subject(s)
Neuroprotective Agents , Receptors, Neurokinin-1 , Tryptophan , Acetylation , Animals , Neuroprotection , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Substance P/pharmacology , Tryptophan/metabolism , Tryptophan/pharmacology
8.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269909

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.


Subject(s)
Analgesics, Opioid , Fentanyl , Analgesics/pharmacology , Analgesics, Opioid/chemistry , Fentanyl/chemistry , Fentanyl/pharmacology , Receptors, Drug , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
9.
Pharmaceutics ; 14(3)2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35335981

ABSTRACT

Currently, the search for promising NK1R-positive tumor-targeting radiopharmaceuticals based on the structure of small molecular antagonists of neurokinin-1 receptor can be observed. Following this trend, we continued our evaluation of aprepitant-based 177Lu-radioconjugates in terms of future oncological applications. For this purpose, three novel aprepitant homologues were synthesized to broaden the previously obtained derivative portfolio, functionalized with the DOTA chelator and labeled with 68Ga and 177Lu. The newly evaluated radioconjugates showed the intended significant increase in lipophilicity compared to the previous ones, while maintaining stability in the human serum. Then, in a receptor binding study to the human NK1 receptor, we compared the two series of 177Lu-radioconjugates of aprepitant with each other and with the reference Substance P derivative currently used in glioblastoma therapy, clearly indicating the high affinity and better binding capacity of the novel radioconjugates. The in vitro experimental results included in the presented study, supported by labeling optimization, radioconjugate characterization and docking modeling of new aprepitant-derived radioagents, confirm our assumptions about the usefulness of aprepitant as a NK1R targeting vector and point out the perspectives for the forthcoming first in vivo trials.

10.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163139

ABSTRACT

Locoregionally administered, NK1 receptor (NK1R) targeted radionuclide therapy is a promising strategy for the treatment of glioblastoma multiforme. So far, the radiopharmaceuticals used in this approach have been based on the endogenous agonist of NK1R, Substance P or on its close analogues. Herein, we used a well-known, small molecular NK1R antagonist, L732,138, as the basis for the radiopharmaceutical vector. First, 14 analogues of this compound were evaluated to check whether extending the parent structure with linkers of different lengths would not deteriorate the NK1R binding. The tested analogues had affinity similar to or better than the parent compound, and none of the linkers had a negative impact on the binding. Next, five DOTA conjugates were synthesized and used for labelling with 68Ga and 177Lu. The obtained radioconjugates turned out to be fairly lipophilic but showed rather limited stability in human plasma. Evaluation of the receptor affinity of the (radio)conjugates showed that neither the chelator nor the metal negatively impacts the NK1R binding. The 177Lu-radioconjugates exhibited the binding characteristics towards NK1R similar or better than that of the 177Lu-labelled derivative of Substance P, which is in current clinical use. The experimental results presented herein, along with their structural rationalization provided by modelling, give insight for the further molecular design of small molecular NK1R-targeting vectors.


Subject(s)
Gallium Radioisotopes/metabolism , Glioblastoma/metabolism , Lutetium/metabolism , Radioisotopes/metabolism , Radiopharmaceuticals/metabolism , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054860

ABSTRACT

Based on the mechanism of neuropathic pain induction, a new type of bifunctional hybrid peptidomimetics was obtained for potential use in this type of pain. Hybrids consist of two types of pharmacophores that are connected by different types of linkers. The first pharmacophore is an opioid agonist, and the second pharmacophore is an antagonist of the pronociceptive system, i.e., an antagonist of the melanocortin-4 receptor. The results of tests in acute and neuropathic pain models of the obtained compounds have shown that the type of linker used to connect pharmacophores had an effect on antinociceptive activity. Peptidomimetics containing longer flexible linkers were very effective at low doses in the neuropathic pain model. To elucidate the effect of linker lengths, two hybrids showing very high activity and two hybrids with lower activity were further tested for affinity for opioid (mu, delta) and melanocortin-4 receptors. Their complexes with the target receptors were also studied by molecular modelling. Our results do not show a simple relationship between linker length and affinity for particular receptor types but suggest that activity in neuropathic pain is related to a proper balance of receptor affinity rather than maximum binding to any or all of the target receptors.


Subject(s)
Melanocortins/chemistry , Neuralgia/drug therapy , Peptidomimetics/therapeutic use , Amino Acid Sequence , Analgesics , Animals , Binding Sites , HEK293 Cells , Humans , Mice , Models, Biological , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 269: 120761, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-34954483

ABSTRACT

A novel N-acylhydrazone with pharmaceutical importance was subject of structural and IR/VCD investigations in the solid state. In the crystal structure, dimers of anion-cation pairs are stabilized by H-bonding and ionic interactions. Some less common interaction types, like C=N···C-NH3+ (σ-hole) interactions, hydrazone-aromatic interactions and dispersive contacts of the CF3 groups are also present in the crystal. Satisfactory reproduction of the solid state IR and VCD spectra required that quantum-chemical calculations be done on a tetramer (four cation-anion pairs) cut out from the crystal structure, exhibiting key intermolecular interactions. Ten DFT functionals were assessed as to the agreement between the calculated and experimental spectra. Various approaches to scaling of the calculated frequencies were applied. The best results were yielded with individual (optimized) frequency scaling factors (FSFs) and band half-widths at half maximum-(HWHM) for four separate spectral subregions. The best matching between the experimental and theoretical spectra (according to SimIR, SimVCD and SimVDF indices) was found for the B3PW91 functional, however, a few other functionals follow closely in the ranking. Based on the quantum chemical calculations, spectral assignments have been made.


Subject(s)
Trifluoroacetic Acid
14.
Int J Mol Sci ; 22(5)2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33800517

ABSTRACT

A new PSMA ligand (PSMA-D4) containing the Glu-CO-Lys pharmacophore connected with a new linker system (L-Trp-4-Amc) and chelator DOTA was developed for radiolabeling with therapeutic radionuclides. Herein we describe the synthesis, radiolabeling, and preliminary biological evaluation of the novel PSMA-D4 ligand. Synthesized PSMA-D4 was characterized using TOF-ESI-MS, NMR, and HPLC methods. The novel compound was subject to molecular modeling with GCP-II to compare its binding mode to analogous reference compounds. The radiolabeling efficiency of PSMA-D4 with 177Lu, 90Y, 47Sc, and 225Ac was chromatographically tested. In vitro studies were carried out in PSMA-positive LNCaP tumor cells membranes. The ex vivo tissue distribution profile of the radioligands and Cerenkov luminescence imaging (CLI) was studied in LNCaP tumor-bearing mice. PSMA-D4 was synthesized in 24% yield and purity >97%. The radio complexes were obtained with high yields (>97%) and molar activity ranging from 0.11 to 17.2 GBq mcmol-1, depending on the radionuclide. In vitro assays confirmed high specific binding and affinity for all radiocomplexes. Biodistribution and imaging studies revealed high accumulation in LNCaP tumor xenografts and rapid clearance of radiocomplexes from blood and non-target tissues. These render PSMA-D4 a promising ligand for targeted therapy of prostate cancer (PCa) metastases.


Subject(s)
Drug Delivery Systems , Kallikreins , Prostate-Specific Antigen , Prostatic Neoplasms , Radiopharmaceuticals , Animals , Humans , Kallikreins/chemistry , Kallikreins/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , Prostate-Specific Antigen/chemistry , Prostate-Specific Antigen/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Xenograft Model Antitumor Assays
15.
Molecules ; 27(1)2021 Dec 28.
Article in English | MEDLINE | ID: mdl-35011383

ABSTRACT

Our formerly described pentapeptide opioid analog Tyr-c[D-Lys-Phe-Phe-Asp]NH2 (designated RP-170), showing high affinity for the mu (MOR) and kappa (KOR) opioid receptors, was much more stable than endomorphine-2 (EM-2) in the rat brain homogenate and displayed remarkable antinociceptive activity after central (intracerebroventricular) and peripheral (intravenous ) administration. In this report, we describe the further modification of this analog, which includes the incorporation of a ß3-amino acid, (R)- and (S)-ß3-Lys, instead of D-Lys in position 2. The influence of such replacement on the biological properties of the obtained analogs, Tyr-c[(R)-ß3-Lys-Phe-Phe-Asp]NH2 (RP-171) and Tyr-c[(S)-ß3-Lys-Phe-Phe-Asp]NH2, (RP-172), was investigated in vitro. Receptor radiolabeled displacement and functional calcium mobilization assays were performed to measure binding affinity and receptor activation of the new analogs. The obtained data revealed that only one of the diastereoisomeric peptides, RP-171, was able to selectively bind and activate MOR. Molecular modeling (docking and molecular dynamics (MD) simulations) suggests that both compounds should be accommodated in the MOR binding site. However, in the case of the inactive isomer RP-172, fewer hydrogen bonds, as well as instability of the canonical ionic interaction to Asp147, could explain its very low MOR affinity.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Lysine/chemistry , Models, Molecular , Peptides, Cyclic/chemistry , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Analgesics, Opioid/chemical synthesis , Animals , Binding Sites , Cell Line , Chemistry Techniques, Synthetic , Chromatography, Liquid , Humans , Mass Spectrometry , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Peptidomimetics/chemical synthesis , Protein Binding , Receptors, Opioid/chemistry , Receptors, Opioid/metabolism , Structure-Activity Relationship
16.
Int J Mol Sci ; 21(20)2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33086743

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.


Subject(s)
Analgesics, Opioid/agonists , Computer Simulation , Neurokinin-1 Receptor Antagonists/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Drug Tolerance , Gastrointestinal Transit/drug effects , Humans , Male , Molecular Docking Simulation , Morphine/pharmacology , Neurokinin-1 Receptor Antagonists/chemistry , Nociception/drug effects , Rats, Wistar , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Sodium Chloride/pharmacology , Thermodynamics , Time Factors
17.
Biosens Bioelectron ; 167: 112446, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32818748

ABSTRACT

Fast, simple in use and highly effective voltammetric enantiosensor dedicated for determination of thalidomide (TD) enantiomers (especially towards the toxic (S)-enantiomer) in blood plasma is still desirable. Here we have proven that newly synthesized chiral naphthalene diimide (NDI) derivatives are excellent electroactive materials for TD enantiosensors. The recognition process relies on the specific interaction between the chiral NDI receptor and the thalidomide enantiomer of the opposite configuration. This unique specific interaction between (S)-thalidomide and (R)-NDI derivative counterparts, evident in the DPV voltammograms, was confirmed by molecular modeling. The demonstrated voltammetric enantiosensors are characterized by the low detection limit at the level of µg·L-1, wide analytical range from 5·10-4 - 10 mg·L-1, high selectivity and long lifetime. The results of the recovery rates showed a very good degree of accuracy towards the determination of (S)-thalidomide in the blood samples, so it can be successfully used in the analysis of clinical samples.


Subject(s)
Biosensing Techniques , Thalidomide , Imides , Naphthalenes , Plasma , Stereoisomerism
18.
Molecules ; 25(16)2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32824729

ABSTRACT

Aprepitant, a lipophilic and small molecular representative of neurokinin 1 receptor antagonists, is known for its anti-proliferative activity on numerous cancer cell lines that are sensitive to Substance P mitogen action. In the presented research, we developed two novel structural modifications of aprepitant to create aprepitant conjugates with different radionuclide chelators. All of them were radiolabeled with 68Ga and 177Lu radionuclides and evaluated in terms of their lipophilicity and stability in human serum. Furthermore, fully stable conjugates were examined in molecular modelling with a human neurokinin 1 receptor structure and in a competitive radioligand binding assay using rat brain homogenates in comparison to the aprepitant molecule. This initial research is in the conceptual stage to give potential theranostic-like radiopharmaceutical pairs for the imaging and therapy of neurokinin 1 receptor-overexpressing cancers.


Subject(s)
Aprepitant/chemistry , Aprepitant/pharmacology , Brain/drug effects , Neoplasms/drug therapy , Neurokinin-1 Receptor Antagonists/pharmacology , Radiopharmaceuticals/pharmacology , Receptors, Neurokinin-1/chemistry , Animals , Brain/pathology , Radiochemistry , Radiopharmaceuticals/chemical synthesis , Rats
19.
Molecules ; 25(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731576

ABSTRACT

In the present contribution, we analyze the influence that C-terminal extension of short opioid peptide sequences by organic fragments has on receptor affinity, in vivo analgesic activity, and antimelanoma properties. The considered fragments were based on either N-acylhydrazone (NAH) or N'-acylhydrazide motifs combined with the 3,5-bis(trifluoromethyl)phenyl moiety. Eleven novel compounds were synthesized and subject to biological evaluation. The analyzed compounds exhibit a diversified range of affinities for the µ opioid receptor (MOR), rather low δ opioid receptor (DOR) affinities, and no appreciable neurokinin-1 receptor binding. In three out of four pairs, N-acylhydrazone-based derivatives bind MOR better than their N'-acylhydrazide counterparts. The best of the novel derivatives have similar low nanomolar MOR binding affinity as the reference opioids, such as morphine and biphalin. The obtained order of MOR affinities was compared to the results of molecular docking. In vivo, four tested compounds turned out to be relatively strong analgesics. Finally, the NAH-based analogues reduce the number of melanoma cells in cell culture, while their N'-acylhydrazide counterparts do not. The antimelanoma properties are roughly correlated to the lipophilicity of the compounds.


Subject(s)
Analgesics , Cytotoxins , Hydrazones/chemistry , Melanoma/drug therapy , Molecular Docking Simulation , Opioid Peptides , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Cell Line, Tumor , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
20.
Molecules ; 25(10)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466333

ABSTRACT

TAPP (H-Tyr-d-Ala-Phe-Phe-NH2) is a potent, µ-selective opioid ligand. In order to gain further insights into pharmacophoric features of this tetrapeptide, we have performed a ß2-Homo-amino acid (ß2hAA) scan of the TAPP sequence. To this aim, 10 novel analogues have been synthesized and evaluated for µ-opioid and δ-opioid receptor affinity as well as for stability in human plasma. The derivatives included compounds in which a (R)- or (S)-ß2-Homo-Homologue replaced the amino acids in the TAPP sequence. The derivatives with (R)- or (S)-ß2hPhe4 turned out to bind µOR with affinities equal to that of the parent. ß2hAAs in position 1 and 3 resulted in rather large affinity decreases, but the change differed depending on the stereochemistry. ß2-Homologation in the second position gave derivatives with very poor µOR binding. According to molecular modelling, the presented α/ß-peptides adopt a variety of binding poses with their common element being an ionic interaction between a protonable amine of the first residue and Asp147. A feature required for high µOR affinity seems the ability to accommodate the ring in the fourth residue in a manner similar to that found for TAPP. Contrary to what might be expected, several compounds were significantly less stable in human plasma than the parent compound.


Subject(s)
Amino Acids/chemistry , Aspartic Acid/chemistry , Oligopeptides/chemistry , Receptors, Opioid, delta/chemistry , Receptors, Opioid, mu/chemistry , Amino Acids/metabolism , Animals , Aspartic Acid/metabolism , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Oligopeptides/blood , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Stability , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...