Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 11(5): 664-670, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435368

ABSTRACT

Gold-nanoparticle (AuNP)-conjugated drugs represent a promising and innovative antitumor therapeutic approach. In our study, we describe the design, the synthesis, the preparation, and the characterization of AuNPs conjugated with the pyrazolo[3,4-d]pyrimidine derivative SI306, a c-Src inhibitor. AuNPs-SI306 showed a good loading efficacy (65%), optimal stability in polar media and in human plasma, and a suitable morphological profile: a ζ-potential of -43.9 mV, a nanoparticle diameter of 48.6 nm, and a 0.441 PDI value. The antitumoral activity of AuNPs-SI306 was evaluated in vitro in the glioblastoma model, by the low-density growth assay, and also in combination with radiotherapy (RT). Results demonstrated that AuNPs had a basal radiosensitization ability and that AuNPs-SI306, when used in combination with RT, were more effective in inhibiting tumor cell growth with respect to AuNPs and free SI306.

2.
J Med Chem ; 60(14): 6305-6320, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28650650

ABSTRACT

Pyrazolo[3,4-d]pyrimidines are potent protein kinase inhibitors with promising antitumor activity but suboptimal aqueous solubility, consequently worth being further optimized. Herein, we present the one-pot two-step procedure for the synthesis of a set of pyrazolo[3,4-d]pyrimidine prodrugs (1a-8a and 9a-e) with higher aqueous solubility and enhanced pharmacokinetic and therapeutic properties. ADME studies demonstrated for the most promising prodrugs a better aqueous solubility, a favorable hydrolysis in human and murine serum, and an increased ability to cross cell membranes with respect to the parental drugs, explaining their better 24 h in vitro cytotoxicity against human glioblastoma U87 cell line. Finally, the 4-4a couple of drug/prodrug was also evaluated in vivo, revealing a profitable pharmacokinetic profile of the prodrug associated with a good efficacy. The application of the prodrug approach demonstrated to be a successful strategy for improving aqueous solubility of the parental drugs, determining a positive impact also in their biological efficacy.


Subject(s)
Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Prodrugs/chemistry , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Male , Membranes, Artificial , Mice, Inbred C57BL , Mice, Nude , Microsomes, Liver/metabolism , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Solubility , Structure-Activity Relationship
3.
Mol Cancer Ther ; 15(7): 1535-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27196762

ABSTRACT

Glioblastoma cells efficiently interact with and infiltrate the surrounding normal tissue, rendering surgical resection and adjuvant chemo/radiotherapy ineffective. New therapeutic targets, able to interfere with glioblastoma's capacity to synergize with normal brain tissue, are currently under investigation. The compound Si306, a pyrazolo[3,4-d]pyrimidine derivative, selected for its favorable activity against SRC, was tested in vitro and in vivo on glioblastoma cell lines. In vivo, combination treatment with Si306 and radiotherapy was strongly active in reducing U-87 xenograft growth with respect to control and single treatments. The histology revealed a significant difference in the stromal compartment of tumoral tissue derived from control or radiotherapy-treated samples with respect to Si306-treated samples, showing in the latter a reduced presence of collagen and α-SMA-positive cells. This effect was paralleled in vitro by the capacity of Si306 to interfere with myofibroblastic differentiation of normal fibroblasts induced by U-87 cells. In the presence of Si306, TGF-ß released by U-87 cells, mainly in hypoxia, was ineffective in upregulating α-SMA and ß-PDGFR in fibroblasts. Si306 efficiently reached the brain and significantly prolonged the survival of mice orthotopically injected with U-87 cells. Drugs that target SRC could represent an effective therapeutic strategy in glioblastoma, able to block positive paracrine loop with stromal cells based on the ß-PDGFR axis and the formation of a tumor-promoting microenvironment. This approach could be important in combination with conventional treatments in the effort to reduce tumor resistance to therapy. Mol Cancer Ther; 15(7); 1535-44. ©2016 AACR.


Subject(s)
Brain Neoplasms/metabolism , Cell Communication , Glioblastoma/metabolism , Signal Transduction , Stromal Cells/metabolism , src-Family Kinases/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Communication/drug effects , Cell Communication/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/radiation effects , Stromal Cells/drug effects , Stromal Cells/radiation effects , Tumor Microenvironment , Xenograft Model Antitumor Assays
4.
Sci Rep ; 6: 21509, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26898318

ABSTRACT

Pyrazolo[3,4-d]pyrimidines are a class of compounds with a good activity against several cancer cell lines. Despite the promising anticancer activity, these molecules showed a poor aqueous solubility. This issue could threat the future development of pyrazolo[3,4-d]pyrimidines as clinical drug candidates. With the aim of improving their solubility profile and consequently their pharmacokinetic properties, we have chosen four compounds (1-4) on the base of their anti-neuroblastoma activity and we have developed albumin nanoparticles and liposomes for the selected candidates. Albumin nanoparticles and liposomes were prepared and characterized regarding size and ζ-potential distribution, polidispersity index, entrapment efficiency and activity against SH-SY5Y human neuroblastoma cell line. The most promising nanosystem, namely LP-2, was chosen to perform further studies: confocal microscopy, stability and drug release in physiological conditions, and biodistribution. Altogether, the obtained data strongly indicate that the encapsulation of pyrazolo[3,4-d]pyrimidines in liposomes represent an effective method to overcome the poor water solubility.


Subject(s)
Drug Delivery Systems , Nanoparticles/administration & dosage , Neuroblastoma/drug therapy , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Cell Line, Tumor , Drug Liberation , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Nanoparticles/chemistry , Neuroblastoma/pathology , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Solubility
5.
Bioorg Med Chem Lett ; 26(3): 894-898, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26774582

ABSTRACT

BV02 is a reference inhibitor of 14-3-3 protein-protein interactions, which is currently used as chemical biology tool to understand the role of 14-3-3 proteins in pathological contexts. Due to chemical instability in certain conditions, its bioactive form has remained unclear. Here, we use NMR spectroscopy to prove for the first time the direct interaction between the molecule and 14-3-3σ, and to depict its bioactive form, namely the phthalimide derivative 9. Our work provides molecular insights to the bioactive form of the 14-3-3 PPI inhibitor and facilitates further development as candidate therapeutic agent.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , Benzamides/chemistry , Pyrazoles/chemistry , 14-3-3 Proteins/metabolism , Benzamides/metabolism , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Phthalimides/chemistry , Phthalimides/metabolism , Protein Interaction Domains and Motifs , Pyrazoles/metabolism
6.
ACS Chem Biol ; 10(9): 1964-9, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26121309

ABSTRACT

In silico target fishing is an emerging tool in drug discovery, which is mostly used for primary target or off-target prediction and drug repositioning. In this work, we developed an in silico target fishing protocol to identify the primary target of GV2-20, a false-positive hit highlighted in a cell-based screen for 14-3-3 modulators. Although GV2-20 does not bind to 14-3-3 proteins, it showed remarkable antiproliferative effects in CML cells, thus raising interest toward the identification of its primary target. Six potential targets of GV2-20 were prioritized in silico and tested in vitro. Our results show that the molecule is a potent inhibitor of carbonic anhydrase 2 (CA2), thus confirming the predictive capability of our protocol. Most notably, GV2-20 experienced a remarkable selectivity for CA2, CA7, CA9, and CA12, and its scaffold was never explored before as a chemotype for CA inhibition, thus becoming an interesting lead candidate for further development.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , 14-3-3 Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Computer Simulation , Drug Repositioning , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Molecular Docking Simulation
7.
J Med Chem ; 58(11): 4590-609, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25923950

ABSTRACT

Fyn is a member of the Src-family of nonreceptor protein-tyrosine kinases. Its abnormal activity has been shown to be related to various human cancers as well as to severe pathologies, such as Alzheimer's and Parkinson's diseases. Herein, a structure-based drug design protocol was employed aimed at identifying novel Fyn inhibitors. Two hits from commercial sources (1, 2) were found active against Fyn with K(i) of about 2 µM, while derivative 4a, derived from our internal library, showed a K(i) of 0.9 µM. A hit-to-lead optimization effort was then initiated on derivative 4a to improve its potency. Slightly modifications rapidly determine an increase in the binding affinity, with the best inhibitors 4c and 4d having K(i)s of 70 and 95 nM, respectively. Both compounds were found able to inhibit the phosphorylation of the protein Tau in an Alzheimer's model cell line and showed antiproliferative activities against different cancer cell lines.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tauopathies/drug therapy , Antineoplastic Agents/chemistry , Binding Sites , Cell Proliferation/drug effects , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Neoplasms/enzymology , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-fyn/metabolism , Pyrazoles/chemistry , Pyrimidines/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Tauopathies/enzymology , Tumor Cells, Cultured
8.
Drug Metab Pharmacokinet ; 29(6): 433-40, 2014.
Article in English | MEDLINE | ID: mdl-24850316

ABSTRACT

The aim of this study is to investigate the metabolic (cytochrome P450-dependent) behaviour of pyrazolo[3,4-d]pyrimidines 1-10 dual Abl/Src kinase inhibitors. All the compounds demonstrate good metabolic stability both in human liver (HLM) and in rat liver (RLM) microsomes. Moreover, all the tested molecules undergo the same metabolic CYP-dependent reactions, namely oxidative dechlorination and N-dealkylation. These metabolic pathways are fully characterized for compound 1. In HLM, the dehalogenated metabolite accounts for about 87% of the full 1 metabolism, while the N-dealkylated metabolite accounts for 12%. Inhibition studies performed using different CYP-inhibitors indicate that the 3A family is the isoenzyme family most involved in pyrazolo[3,4-d]pyrimidine metabolism. This observation is confirmed by studies performed by using CYP3A selective substrates. Furthermore kinetic analysis performed in RLM, HLM and cDNA CYP3A4 shows that the affinity of CYPs towards compound 1 is similar in all the tested preparations (Km = 32.7, 21.8, and 48.7 µM, respectively).


Subject(s)
Antineoplastic Agents/metabolism , Cytochrome P-450 CYP3A/metabolism , Liver/enzymology , Protein Kinase Inhibitors/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Animals , Antineoplastic Agents/chemistry , Biotransformation , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Dealkylation , Drug Stability , Humans , Kinetics , Liver/drug effects , Microsomes, Liver/enzymology , Molecular Structure , Oxidation-Reduction , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Rats , Recombinant Proteins/metabolism , Substrate Specificity
9.
ChemMedChem ; 9(5): 973-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24715717

ABSTRACT

14-3-3 is a family of highly conserved adapter proteins that is attracting much interest among medicinal chemists. Small-molecule inhibitors of 14-3-3 protein-protein interactions (PPIs) are in high demand, both as tools to increase our understanding of 14-3-3 actions in human diseases and as leads to develop innovative therapeutic agents. Herein we present the discovery of novel 14-3-3 PPI inhibitors through a multidisciplinary strategy combining molecular modeling, organic synthesis, image-based high-content analysis of reporter cells, and in vitro assays using cancer cells. Notably, the two most active compounds promoted the translocation of c-Abl and FOXO pro-apoptotic factors into the nucleus and sensitized multidrug-resistant cancer cells to apoptotic inducers such as doxorubicin and the pan-Akt inhibitor GSK690693, thus becoming valuable lead candidates for further optimization. Our results emphasize the possible role of 14-3-3 PPI inhibitors in anticancer combination therapies.


Subject(s)
14-3-3 Proteins/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Drug Discovery , Drug Resistance, Multiple/drug effects , Oxadiazoles/pharmacology , Small Molecule Libraries/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Doxorubicin/chemical synthesis , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Protein Binding/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
10.
ACS Comb Sci ; 16(4): 168-75, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24597676

ABSTRACT

A library of pyrazolo[3,4-d]pyrimidines, endowed with a high level of molecular diversity, has been developed applying a synthetic sequence that allowed C3, N1, C4, and C6 substitution. The enzymatic screening of this "privileged scaffold"-based compound collection, validated the use of a diversity-oriented approach in a field characteristically explored by target-oriented synthesis. In fact, several compounds showed high activity against the selected kinases (i.e., Src, Abl wt, and T315I mutated-form), furthermore and interestingly a new compound has emerged as an allosteric inhibitor of the T315I mutated-form of Abl, opening up new opportunities for the development of a novel class of noncompetitive inhibitors of Abl (T315I).


Subject(s)
Allosteric Regulation/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Dose-Response Relationship, Drug , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 23(19): 5351-5, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23972440

ABSTRACT

Using a phenotypic screening and SAR optimization approach, a phenyl-bis-oxazole derivative has been identified with anti-proliferative activity, optimized with the use of a panel of cancer cell lines. The lead compound was synthesized by means of a short and effective two-step synthesis using Pd-catalyzed direct arylation. The compound stabilizes several quadruplex DNA sequences including a human telomeric DNA and one from the promoter of the HSP90 gene, although the structure-activity relationships of the series are not obviously related to the quadruplex binding.


Subject(s)
G-Quadruplexes , Heterocyclic Compounds/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , Ligands , Structure-Activity Relationship
12.
J Med Chem ; 56(13): 5382-94, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23746084

ABSTRACT

Starting from our in-house library of pyrazolo[3,4-d]pyrimidines, a cross-docking simulation was conducted on Bcr-Abl T315I mutant. Among the selected compounds (2a-e), the 4-bromo derivative 2b showed the best activity against the Bcr-Abl T315I mutant. Deeper computational studies highlighted the importance of the bromine atom in the para position of the N1 side chain phenyl ring for the interaction with the T315I mutant. A series of 4-bromo derivatives was thus synthesized and biologically evaluated. Compound 2j showed a good balance of different ADME properties, high activity in cell-free assays, and a submicromolar potency against T315I Bcr-Abl expressing cells. In addition, it was converted into a water-soluble formulation by liposome encapsulation, preserving a good activity on leukemic T315I cells and avoiding the use of DMSO as solubilizing agent. In vivo studies on mice inoculated with 32D-T315I cells and treated with 2j showed a more than 50% reduction in tumor volumes.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Drug Design , Female , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Humans , Mice , Mice, Nude , Models, Chemical , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Point Mutation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Structure, Tertiary , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Transfection , Tumor Burden/drug effects
13.
Drug Discov Today Technol ; 10(4): e541-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24451646

ABSTRACT

14-3-3 is a family of highly conserved regulatory proteins which is attracting a significant interest due to its potential role as target for pharmacological intervention against cancer and neurodegenerative disorders. Although modulating protein-protein interactions (PPI) is still conceived as a challenging task in drug discovery, in past few years peptide inhibitors and small molecular modulators of 14-3-3 PPI have been described. Here we examine structural and biological features of 14-3-3 and propose an overview on techniques used for discovering small molecular inhibitors and stabilizers of 14-3-3 PPI.


Subject(s)
14-3-3 Proteins/metabolism , Drug Discovery , 14-3-3 Proteins/antagonists & inhibitors , 14-3-3 Proteins/chemistry , Humans , Protein Conformation , Protein Interaction Mapping
14.
ACS Med Chem Lett ; 4(7): 622-6, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-24900720

ABSTRACT

Design and synthesis of prodrugs of promising drug candidates represents a valid strategy to overcome the lack of favorable ADME properties, in particular aqueous solubility and bioavailability. We report herein the successful application of this strategy with two representative pyrazolo[3,4-d]pyrimidine derivatives (1 and 2), which led to the development of the corresponding and highly water-soluble antitumor prodrugs (7 and 8). In vitro studies confirmed a significant improvement of aqueous solubility and, for compound 8, good plasma stability, suggesting superior in vivo bioavailability. As expected, the uncleaved water-soluble prodrugs 7 and 8 showed no activity toward the enzymatic targets (c-Src and c-Abl) but revealed promising antiproliferative activity in myeloid cell lines, as a consequence of the in vitro hydrolysis of the selected solubilizing moiety, followed by the release of the active compounds (1 and 2).

15.
Bioorg Med Chem Lett ; 21(22): 6867-71, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21962576

ABSTRACT

Targeting the binding site of 14-3-3 proteins lets the release of partner proteins involved in cell cycle progression, apoptosis, cytoskeletal rearrangement and transcriptional regulation and may therefore be regarded as an alternative strategy to integrate conventional therapeutic approaches against cancer. In the present work, we report the identification of two new small molecule inhibitors of 14-3-3σ/c-Abl protein-protein interaction (BV01 and BV101) discovered by means of computational methods. The most interesting compound (BV01) showed a lethal dose (LD(50)) in the low micromolar range against Ba/F3 murine cell lines expressing the Imatinib (IM)-sensitive wild type Bcr-Abl construct and the IM-resistant Bcr-Abl mutation T315I. BV01 interaction with 14-3-3σ was demonstrated by NMR studies and elucidated by docking. It blocked the binding domain of 14-3-3σ, hence promoting the release of the partner protein c-Abl (the one not involved in Bcr rearrangement), and its translocation to both the nuclear compartment and mitochondrial membranes to induce a pro-apoptotic response. Our results advance BV01 as a confirmed hit compound capable of eliciting apoptotic death of Bcr-Abl-expressing cells by interfering with 14-3-3σ/c-Abl protein-protein interaction.


Subject(s)
14-3-3 Proteins/metabolism , Biomarkers, Tumor/metabolism , Drug Design , Exonucleases/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-abl/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , 14-3-3 Proteins/antagonists & inhibitors , 14-3-3 Proteins/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/chemistry , Cell Line , Exonucleases/antagonists & inhibitors , Exonucleases/chemistry , Exoribonucleases , Humans , Mice , Models, Molecular , Neoplasms/drug therapy , Protein Interaction Mapping , Proto-Oncogene Proteins c-abl/antagonists & inhibitors
16.
ChemMedChem ; 6(2): 343-52, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21246739

ABSTRACT

As a continuation of our previous work, which resulted in the identification of a new hit compound as an HIV-1 integrase inhibitor, three novel series of salicylic acid derivatives were synthesized using three versatile and practical synthetic strategies and were assayed for their capacity to inhibit the catalytic activity of HIV-1 integrase. Biological evaluations revealed that some of the synthesized compounds possess good inhibitory potency in enzymatic assays and are able to inhibit viral replication in MT-4 cells at low micromolar concentrations. Finally, docking studies were conducted to analyze the binding mode of the synthesized compounds within the DNA binding site of integrase in order to refine their structure-activity relationships.


Subject(s)
HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/pharmacology , HIV Integrase/drug effects , Cell Line , Drug Evaluation, Preclinical , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...