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










Publication year range
1.
J Med Chem ; 66(21): 14824-14842, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37902628

ABSTRACT

We synthesized new pyrrole and indole derivatives as human carbonic anhydrase (hCA) inhibitors with the potential to inhibit the Wnt/ß-catenin signaling pathway. The presence of both N1-(4-sulfonamidophenyl) and 3-(3,4,5-trimethoxyphenyl) substituents was essential for strong hCA inhibitors. The most potent hCA XII inhibitor 15 (Ki = 6.8 nM) suppressed the Wnt/ß-catenin signaling pathway and its target genes MYC, Fgf20, and Sall4 and exhibited the typical markers of apoptosis, cleaved poly(ADP-ribose)polymerase, and cleaved caspase-3. Compound 15 showed strong inhibition of viability in a panel of cancer cells, including colorectal cancer and triple-negative breast cancer cells, was effective against the NCI/ADR-RES DOX-resistant cell line, and restored the sensitivity to doxorubicin (DOX) in HT29/DX and MDCK/P-gp cells. Compound 15 is a novel dual-targeting compound with activity against hCA and Wnt/ß-catenin. It thus has a broad targeting spectrum and is an anticancer agent with specific potential in P-glycoprotein overexpressing cell lines.


Subject(s)
Carbonic Anhydrases , Neoplasms , Humans , Structure-Activity Relationship , Drug Resistance, Multiple , Wnt Signaling Pathway , Drug Resistance, Neoplasm , Carbonic Anhydrases/metabolism , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase IX , Molecular Structure , Benzenesulfonamides
2.
ACS Pharmacol Transl Sci ; 6(7): 1087-1103, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37470018

ABSTRACT

Despite intensive efforts, no inhibitors of the Wnt/ß-catenin signaling pathway have been approved so far for the clinical treatment of cancer. We synthesized novel N-(heterocyclylphenyl)benzenesulfonamides as ß-catenin inhibitors. Compounds 5-10 showed strong inhibition of the luciferase activity. Compounds 5 and 6 inhibited the MDA-MB-231, HCC1806, and HCC1937 TNBC cells. Compound 9 induced in vitro cell death in SW480 and HCT116 cells and in vivo tumorigenicity of a human colorectal cancer line HCT116. In a co-immunoprecipitation study in HCT116 cells transfected with Myc-tagged T-cell factor 4 (Tcf-4), compound 9 abrogated the association between ß-catenin and Tcf-4. The crystallographic analysis of the ß-catenin Armadillo repeats domain revealed that compound 9 and Tcf-4 share a common binding site within the hotspot binding region close to Lys508. To our knowledge, compound 9 is the first small molecule ligand of this region to be reported. These results highlight the potential of this novel class of ß-catenin inhibitors as anticancer agents.

3.
Bioorg Chem ; 138: 106607, 2023 09.
Article in English | MEDLINE | ID: mdl-37210829

ABSTRACT

Growth factor receptor bound protein 2 (Grb2) is an adaptor protein featured by a nSH3-SH2-cSH3 domains. Grb2 finely regulates important cellular pathways such as growth, proliferation and metabolism and a minor lapse of this tight control may totally change the entire pathway to the oncogenic. Indeed, Grb2 is found overexpressed in many tumours type. Consequently, Grb2 is an attractive therapeutic target for the development of new anticancer drug. Herein, we reported the synthesis and the biological evaluation of a series of Grb2 inhibitors, developed starting from a hit-compound already reported by this research unit. The newly synthesized compounds were evaluated by kinetic binding experiments, and the most promising derivatives were assayed in a short panel of cancer cells. Five of the newly synthesized derivatives proved to be able to bind the targeted protein with valuable inhibitory concentration in one-digit micromolar concentration. The most active compound of this series, derivative 12, showed an inhibitory concentration of about 6 µM for glioblastoma and ovarian cancer cells, and an IC50 of 1.67 for lung cancer cell. For derivative 12, the metabolic stability and the ROS production was also evaluated. The biological data together with the docking studies led to rationalize an early structure activity relationship.


Subject(s)
Antineoplastic Agents , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/metabolism , Amino Acid Sequence , Protein Binding , Antineoplastic Agents/pharmacology , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 81: 129123, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36608774

ABSTRACT

Trypanosoma brucei is a protozoan parasite that causes Human African Trypanosomiasis (HAT), a neglected tropical disease (NTD) that is endemic in 36 countries in sub-Saharan Africa. Only a handful drugs are available for treatment, and these have limitations, including toxicity and drug resistance. Using the natural product, curcumin, as a starting point, several curcuminoids and related analogs were evaluated against bloodstream forms of T. b. brucei. A particular subset of dibenzylideneacetone (DBA) compounds exhibited potent in vitro antitrypanosomal activity with sub-micromolar EC50 values. A structure-activity relationship study including 26 DBA analogs was initiated, and several compounds exhibited EC50 values as low as 200 nM. Cytotoxicity counter screens in HEK293 cells identified several compounds having selectivity indices above 10. These data suggest that DBAs offer starting points for a new small molecule therapy of HAT.


Subject(s)
Trypanocidal Agents , Trypanosoma brucei brucei , Trypanosomiasis, African , Animals , Humans , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Neglected Diseases/drug therapy , HEK293 Cells , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Structure-Activity Relationship
5.
Eur J Med Chem ; 246: 114997, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36502578

ABSTRACT

We synthesized a new inhibitor of tubulin polymerization, the pyrrole (1-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-1H-pyrrol-3-yl)(3,4,5-trimethoxy-phenyl)methanone 6 (RS6077). Compound 6 inhibited the growth of multiple cancer cell lines, with IC50 values in the nM range, without affecting the growth of non-transformed cells. The novel agent arrested cells in the G2/M phase of the cell cycle in both transformed and non-transformed cell lines, but single cell analysis by time-lapse video recording revealed a remarkable selectivity in cell death induction by compound 6: in RPE-1 non-transformed cells mitotic arrest induced was not necessarily followed by cell death; in contrast, in HeLa transformed and in lymphoid-derived transformed AHH1 cell lines, cell death was effectively induced during mitotic arrest in cells that fail to complete mitosis. Importantly, the agent also inhibited the growth of the lymphoma TMD8 xenograft model. Together these findings suggest that derivative 6 has a selective efficacy in transformed vs non-transformed cells and indicate that the same compound has potential as novel therapeutic agent to treat lymphomas. Compound 6 showed good metabolic stability upon incubation with human liver microsomes.


Subject(s)
Apoptosis , Lymphoma , Humans , Cell Death , Mitosis , HeLa Cells , Tubulin/metabolism , Lymphoma/drug therapy , Cell Line, Tumor , Cell Proliferation
6.
J Med Chem ; 65(23): 15805-15818, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36395526

ABSTRACT

We synthesized new aroyl diheterocyclic pyrrole (ARDHEP) 15 that exhibited the hallmarks of ferroptosis. Compound 15 strongly inhibited U-87 MG, OVCAR-3, and MCF-7 cancer cells, induced an increase of cleaved PARP, but was not toxic for normal human primary T lymphocytes at 0.1 µM. Analysis of the levels of lactoperoxidase, malondialdehyde, lactic acid, total glutathione, and ATP suggested that the in vivo inhibition of cancer cell proliferation by 15 went through stimulation of oxidative stress injury and Fe2+ accumulation. Quantitative polymerase chain reaction analysis of the mRNA expression in U-87 MG and SKOV-3 tumor tissues from 15-treated mice showed the presence of Ptgs2/Nfe2l2/Sat1/Akr1c1/Gpx4 genes correlated with ferroptosis in both groups. Immunofluorescence staining revealed significantly lower expressions of proteins Ki67, CD31, and ferroptosis negative regulation proteins glutathione peroxidase 4 (GPX4) and FTH1. Compound 15 was found to be metabolically stable when incubated with human liver microsomes.


Subject(s)
Ovarian Neoplasms , Tubulin Modulators , Humans , Animals , Female , Mice , Tubulin , Pyrroles/pharmacology , Apoptosis , Cell Line, Tumor
7.
Molecules ; 27(22)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36431838

ABSTRACT

Aberrant accumulation of ß-catenin in the cell nucleus as a result of deregulation of the Wnt/ß-catenin pathway is found in various types of cancer. Direct ß-catenin targeting agents are being researched despite obstacles; however, specific ß-catenin drugs for clinical treatments have not been approved so far. We focused on direct ß-catenin targeting of potential therapeutic value as anticancer agents. This review provides recent advances on small molecule ß-catenin agents. Structure-activity relationships and biological activities of reported inhibitors are discussed. This work provides useful knowledge in the discovery of ß-catenin agents.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , beta Catenin/metabolism , Wnt Signaling Pathway , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Nucleus/metabolism
8.
Eur J Med Chem ; 240: 114605, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35868126

ABSTRACT

Most cancer cells switch their metabolism from mitochondrial oxidative phosphorylation to aerobic glycolysis to generate ATP and precursors for the biosynthesis of key macromolecules. The aerobic conversion of pyruvate to lactate, coupled to oxidation of the nicotinamide cofactor, is a primary hallmark of cancer and is catalyzed by lactate dehydrogenase (LDH), a central effector of this pathological reprogrammed metabolism. Hence, inhibition of LDH is a potential new promising therapeutic approach for cancer. In the search for new LDH inhibitors, we carried out a structure-based virtual screening campaign. Here, we report the identification of a novel specific LDH inhibitor, the pyridazine derivative 18 (RS6212), that exhibits potent anticancer activity within the micromolar range in multiple cancer cell lines and synergizes with complex I inhibition in the suppression of tumor growth. Altogether, our data support the conclusion that compound 18 deserves to be further investigated as a starting point for the development of LDH inhibitors and for novel anticancer strategies based on the targeting of key metabolic steps.


Subject(s)
L-Lactate Dehydrogenase , Neoplasms , Cell Line , Enzyme Inhibitors/pharmacology , Glycolysis , Humans , L-Lactate Dehydrogenase/metabolism , Lactic Acid , Neoplasms/drug therapy , Neoplasms/pathology , Oxidative Phosphorylation
9.
Molecules ; 26(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34946546

ABSTRACT

There are promising new therapeutic agents for CRC patients, including novel small-molecule inhibitors and immune checkpoint blockers. We focused on emerging CRC's therapeutic agents that have shown the potential for progress in clinical practice. This review provides an overview of tyrosine kinase inhibitors targeting VEGF and KIT, BRAF and MEK inhibitors, TLR9 agonist, STAT3 inhibitors, and immune checkpoint blockers (PD1/PDL-1 inhibitors), for which recent advances have been reported. These new agents have the potential to provide benefits to CRC patients with unmet medical needs.


Subject(s)
Colorectal Neoplasms , Molecular Targeted Therapy , Neoplasm Proteins , Protein Kinase Inhibitors , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use
10.
Pharmaceuticals (Basel) ; 14(10)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34681230

ABSTRACT

Human noroviruses (HuNoVs) are the most common cause of viral gastroenteritis resulting in ~219,000 deaths annually and a societal cost of ~USD60 billion. There are no antivirals or vaccines available to treat and/or prevent HuNoV. In this study, we performed a large-scale phenotypical antiviral screening using the mouse norovirus (MNV), which included ~1000 drug-like small molecules from the Drug Design and Synthesis Centre (Sapienza University, Rome). Compound 3-((3,5-dimethylphenyl)sulfonyl)-5-chloroindole-N-(phenylmethanol-4-yl)-2.carboxamide (compound 1) was identified as an inhibitor of MNV replication with an EC50 of 0.5 ± 0.1 µM. A series of 10 analogs were synthesized of which compound 6 showed an improved potency/selectivity (EC50 0.2 ± 0.1 µM) against MNV; good activity was also observed against the HuNoV GI replicon (EC50 1.2 ± 0.6 µM). Time-of-drug-addition studies revealed that analog 6 acts at a time point that coincides with the onset of viral RNA replication. After six months of selective pressure, two compound 6res variants were independently selected, both harboring one mutation in VPg and three mutations in the RdRp. After reverse engineering S131T and Y154F as single mutations into the MNV backbone, we did not find a markedly compound 6res phenotype. In this study, we present a class of novel norovirus inhibitors with a high barrier to resistance and in vitro antiviral activity.

11.
Biol Direct ; 16(1): 15, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34641953

ABSTRACT

The interaction between proteins is a fundamental event for cellular life that is generally mediated by specialized protein domains or modules. PDZ domains are the largest class of protein-protein interaction modules, involved in several cellular pathways such as signal transduction, cell-cell junctions, cell polarity and adhesion, and protein trafficking. Because of that, dysregulation of PDZ domain function often causes the onset of pathologies, thus making this family of domains an interesting pharmaceutical target. In this review article we provide an overview of the structural and functional features of PDZ domains and their involvement in the cellular and molecular pathways at the basis of different human pathologies. We also discuss some of the strategies that have been developed with the final goal to hijack or inhibit the interaction of PDZ domains with their ligands. Because of the generally low binding selectivity of PDZ domain and the scarce efficiency of small molecules in inhibiting PDZ binding, this task resulted particularly difficult to pursue and still demands increasing experimental efforts in order to become completely feasible and successful in vivo.


Subject(s)
Neoplasms , Virus Diseases , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , PDZ Domains , Protein Binding , Proteins
12.
Eur J Med Chem ; 221: 113532, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34052717

ABSTRACT

Long-term survivors of glioblastoma multiforme (GBM) are at high risk of developing second primary neoplasms, including leukemia. For these patients, the use of classic tyrosine kinase inhibitors (TKIs), such as imatinib mesylate, is strongly discouraged, since this treatment causes a tremendous increase of tumor and stem cell migration and invasion. We aimed to develop agents useful for the treatment of patients with GBM and chronic myeloid leukemia (CML) using an alternative mechanism of action from the TKIs, specifically based on the inhibition of tubulin polymerization. Compounds 7 and 25, as planned, not only inhibited tubulin polymerization, but also inhibited the proliferation of both GMB and CML cells, including those expressing the T315I mutation, at nanomolar concentrations. In in vivo experiments in BALB/cnu/nu mice injected subcutaneously with U87MG cells, in vivo, 7 significantly inhibited GBM cancer cell proliferation, in vivo tumorigenesis, and tumor growth, tumorigenesis and angiogenesis. Compound 7 was found to block human topoisomerase II (hTopoII) selectively and completely, at a concentration of 100 µM.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Glioblastoma/drug therapy , Heterocyclic Compounds/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Methane/pharmacology , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Methane/analogs & derivatives , Methane/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polymerization/drug effects , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tumor Cells, Cultured
13.
Eur J Pharmacol ; 897: 173936, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33581134

ABSTRACT

Glioblastoma Multiforme (GBM) is a highly invasive primary brain tumour characterized by chemo- and radio-resistance and poor overall survival. GBM can present an aberrant functionality of p53, caused by the overexpression of the murine double minute 2 protein (MDM2) and its analogue MDM4, which may influence the response to conventional therapies. Moreover, tumour resistance/invasiveness has been recently attributed to an overexpression of the chemokine receptor CXCR4, identified as a pivotal mediator of glioma neovascularization. Notably, CXCR4 and MDM2-4 cooperate in promoting tumour invasion and progression. Although CXCR4 actively promotes MDM2 activation leading to p53 inactivation, MDM2-4 knockdown induces the downregulation of CXCR4 gene transcription. Our study aimed to assess if the CXCR4 signal blockade could enhance glioma cells' sensitivity to the inhibition of the p53-MDMs axis. Rationally designed inhibitors of MDM2/4 were combined with the CXCR4 antagonist, AMD3100, in human GBM cells and GBM stem-like cells (neurospheres), which are crucial for tumour recurrence and chemotherapy resistance. The dual MDM2/4 inhibitor RS3594 and the CXCR4 antagonist AMD3100 reduced GBM cell invasiveness and migration in single-agent treatment and mainly in combination. AMD3100 sensitized GBM cells to the antiproliferative activity of RS3594. It is noteworthy that these two compounds present synergic effects on cancer stem components: RS3594 inhibited the growth and formation of neurospheres, AMD3100 induced differentiation of neurospheres while enhancing RS3594 effectiveness preventing their proliferation/clonogenicity. These results confirm that blocking CXCR4/MDM2/4 represents a valuable strategy to reduce GBM proliferation and invasiveness, acting on the stem cell component too.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzylamines/pharmacology , Brain Neoplasms/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Cyclams/pharmacology , Glioblastoma/drug therapy , Indoles/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Neurogenesis/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Spheroids, Cellular , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
14.
ACS Med Chem Lett ; 11(10): 1869-1874, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33062166

ABSTRACT

Zika virus (ZIKV) infection, which initially was endemic only in Africa and Asia, is rapidly spreading throughout Europe, Oceania, and the Americas. Although there have been enormous efforts, there is still no approved drug to treat ZIKV infection. Herein, we report the synthesis and biological evaluation of agents with noncompetitive mechanism of the ZIKV NS2B/NS3 protease inhibition through the binding to an allosteric site. Compounds 1 and 2 showed potent activity in both enzymatic and cellular assays. Derivative 1 efficiently reduced the ZIKV protein synthesis and the RNA replication and prevented the mice from life-threatening infection and the brain damage caused by ZIKV infection in a ZIKV mouse model.

15.
ChemMedChem ; 15(23): 2264-2268, 2020 12 03.
Article in English | MEDLINE | ID: mdl-32946182

ABSTRACT

The Wnt/ß-catenin pathway is often found deregulated in cancer. The aberrant accumulation of ß-catenin in the cell nucleus results in the development of various malignancies. Specific drugs against this signaling pathway for clinical treatments have not been approved yet. Herein we report inhibitors of ß-catenin signaling of potential therapeutic value as anticancer agents. Ethyl 4-((4-(trifluoromethyl)phenyl)sulfonamido)benzoate (compound 14) inhibits the effect on Wnt reporter with an IC50 value of 7.0 µM, significantly reduces c-MYC levels, inhibits HCT116 colon cancer cell growth (IC50 20.2 µM), does not violate Lipinski and Veber rules, and shows predicted Caco-2 and MDCK cell permeability Papp >500 nm s-1 . Compound 14 seems to have potential for the development of new anticancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Sulfonamides/pharmacology , beta Catenin/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Proto-Oncogene Proteins c-myc/metabolism , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
16.
Eur J Med Chem ; 208: 112696, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32883642

ABSTRACT

We designed and synthesized 21 new indolylarylsulfones (IASs) as new HIV-1 NNRTIs. Among these, IAS 12 exhibited a remarkable antiviral activity against single and double mutants (K103N EC50 = <0.7 nM; Y181C EC50 = <0.7 nM; Y188L EC50 = 21.3 nM; K103N-Y181C EC50 = 6.2 nM), resulting equally or more active than previuosly reported IAS 6 and some approved anti-HIV-1 drugs. Docking and molecular dynamics simulations of compound 12 in complex with WT, Y181C, Y188L, K103N and K103N-Y181C RTs clarified a general binding mode that was consistent with biological results. Kinetic experiments disclosed that derivative 12 preferentially binds WT and K103N-Y181C RTs to binary and ternary complexes, respectively.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Indoles/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Sulfones/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/metabolism , Cell Line, Tumor , Drug Design , Drug Synergism , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , Humans , Indoles/chemical synthesis , Indoles/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mutation , Protein Binding , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/metabolism , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/metabolism , Zidovudine/analogs & derivatives , Zidovudine/pharmacology
17.
J Comput Aided Mol Des ; 34(11): 1171-1179, 2020 11.
Article in English | MEDLINE | ID: mdl-32700175

ABSTRACT

The exchange proteins activated by cAMP (EPAC) are implicated in a large variety of physiological processes and they are considered as promising targets for a wide range of therapeutic applications. Several recent reports provided evidence for the therapeutic effectiveness of the inhibiting EPAC1 activity cardiac diseases. In that context, we recently characterized a selective EPAC1 antagonist named AM-001. This compound was featured by a non-competitive mechanism of action but the localization of its allosteric site to EPAC1 structure has yet to be investigated. Therefore, we performed cosolvent molecular dynamics with the aim to identify a suitable allosteric binding site. Then, the docking and molecular dynamics were used to determine the binding of the AM-001 to the regions highlighted by cosolvent molecular dynamics for EPAC1. These analyses led us to the identification of a suitable allosteric AM-001 binding pocket at EPAC1. As a model validation, we also evaluated the binding poses of the available AM-001 analogues, with a different biological potency. Finally, the complex EPAC1 with AM-001 bound at the putative allosteric site was further refined by molecular dynamics. The principal component analysis led us to identify the protein motion that resulted in an inactive like conformation upon the allosteric inhibitor binding.


Subject(s)
Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/chemistry , Solvents/chemistry , Allosteric Site , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship
18.
ACS Med Chem Lett ; 11(5): 633-637, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435363

ABSTRACT

We report here the synthesis and human carbonic anhydrases (CA, EC 4.2.1.1) inhibitory properties of a series of 4'-substituted 1,1'-biphenyl-4-sulfonamides incorporating a 2″- or 3″-amino- or carboxyphenyl unit. Most compounds showed significant variations in their inhibition profiles against CA II and IX when compared to previously reported analogs 12-18 bearing a 4″-amino or a 4″-carboxy group. In particular, compounds 1-11 showed considerable improvement of the CA II inhibitory efficacy with K I values in the subnanomolar range (K Is spanning between 0.57 and 31.0 nM), a drop of activity against CA IX (K Is in the range 92.0 to 555.7 nM) and were as potent as 12-18 toward CA I (K Is in the range 5.9-217.7 nM). Docking and molecular dynamics were used to gain insights on the inhibition profiles. The reported inhibition data show that 1-11 have potential as novel agents to treat ocular pathologies, such as glaucoma, because of the potent and selective targeting of CA II, which is the isoform most implicated in this disease.

19.
Biomolecules ; 10(2)2020 02 12.
Article in English | MEDLINE | ID: mdl-32059524

ABSTRACT

Kynurenine (kyn) and kynurenic acid (kyna) are well-defined metabolites of tryptophan catabolism collectively known as "kynurenines", which exert regulatory functions in host-microbiome signaling, immune cell response, and neuronal excitability. Kynurenine containing peptides endowed with opioid receptor activity have been isolated from natural organisms; thus, in this work, novel opioid peptide analogs incorporating L-kynurenine (L-kyn) and kynurenic acid (kyna) in place of native amino acids have been designed and synthesized with the aim to investigate the biological effect of these modifications. The kyna-containing peptide (KA1) binds selectively the m-opioid receptor with a Ki = 1.08 ± 0.26 (selectivity ratio m/d/k = 1:514:10000), while the L-kyn-containing peptide (K6) shows a mixed binding affinity for m, d, and k-opioid receptors, with efficacy and potency (Emax = 209.7 + 3.4%; LogEC50 = -5.984 + 0.054) higher than those of the reference compound DAMGO. This novel oligopeptide exhibits a strong antinociceptive effect after i.c.v. and s.c. administrations in in vivo tests, according to good stability in human plasma (t1/2 = 47 min).


Subject(s)
Kynurenine/chemistry , Oligopeptides/chemistry , Receptors, Opioid/agonists , Animals , Brain/drug effects , Drug Evaluation, Preclinical , Esters/chemistry , Ethanolamine/chemistry , Female , Formaldehyde/chemistry , GTP-Binding Proteins/chemistry , Guinea Pigs , Humans , Kynurenic Acid/chemistry , Male , Oligopeptides/pharmacokinetics , Protein Binding , Rats , Rats, Wistar , Receptors, Opioid, mu/chemistry , Tryptophan/metabolism
20.
Eur J Med Chem ; 185: 111828, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31727471

ABSTRACT

Novel 3-aroyl-1,4-diarylpyrrole derivatives were synthesized to explore structure-activity relationships at the phenyls at positions 1 and 4 of the pyrrole. The presence of amino phenyl rings at positions 1 and 4 of the pyrrole ring were found to be a crucial requirement for potent antitumor activity. Several compounds strongly inhibited tubulin assembly through binding to the colchicine site. Compounds 42, 44, 48, 62 and 69 showed antitumor activity with low nanomolar IC50 values in several cancer cell lines. Compound 48 was generally more effective as an inhibitor of glioblastoma, colorectal and urinary bladder cancer cell lines; 69 consistently inhibited CML cell lines and demonstrated superiority in nilotinib and imatinib resistant LAMA84-R and KBM5-T315I cells. In animal models, compound 48 exhibited significant inhibition of the growth of T24 bladder carcinoma and ES-2 ovarian clear cell carcinoma tumors. Compounds 48 and 69 represent robust lead compounds for the design of new broad-spectrum anticancer agents active in different types of solid and hematological tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hematologic Neoplasms/drug therapy , Pyrroles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Glioblastoma/pathology , Hematologic Neoplasms/pathology , Humans , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Molecular Structure , Optical Imaging , Pyrroles/administration & dosage , Pyrroles/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...