Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Med Chem ; 67(13): 11168-11181, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38932616

RÉSUMÉ

ß-Glucocerebrosidase (GBA/GCase) mutations leading to misfolded protein cause Gaucher's disease and are a major genetic risk factor for Parkinson's disease and dementia with Lewy bodies. The identification of small molecule pharmacological chaperones that can stabilize the misfolded protein and increase delivery of degradation-prone mutant GCase to the lysosome is a strategy under active investigation. Here, we describe the first use of fragment-based drug discovery (FBDD) to identify pharmacological chaperones of GCase. The fragment hits were identified by using X-ray crystallography and biophysical techniques. This work led to the discovery of a series of compounds that bind GCase with nM potency and positively modulate GCase activity in cells.


Sujet(s)
Site allostérique , Découverte de médicament , Glucosylceramidase , Glucosylceramidase/métabolisme , Glucosylceramidase/antagonistes et inhibiteurs , Glucosylceramidase/composition chimique , Humains , Cristallographie aux rayons X , Relation structure-activité , Modèles moléculaires , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/métabolisme
2.
J Med Chem ; 67(6): 4655-4675, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38462716

RÉSUMÉ

The ubiquitously expressed protein tyrosine phosphatase SHP2 is required for signaling downstream of receptor tyrosine kinases (RTKs) and plays a role in regulating many cellular processes. Genetic knockdown and pharmacological inhibition of SHP2 suppresses RAS/MAPK signaling and inhibit the proliferation of RTK-driven cancer cell lines. Here, we describe the first reported fragment-to-lead campaign against SHP2, where X-ray crystallography and biophysical techniques were used to identify fragments binding to multiple sites on SHP2. Structure-guided optimization, including several computational methods, led to the discovery of two structurally distinct series of SHP2 inhibitors binding to the previously reported allosteric tunnel binding site (Tunnel Site). One of these series was advanced to a low-nanomolar lead that inhibited tumor growth when dosed orally to mice bearing HCC827 xenografts. Furthermore, a third series of SHP2 inhibitors was discovered binding to a previously unreported site, lying at the interface of the C-terminal SH2 and catalytic domains.


Sujet(s)
Tumeurs , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Humains , Souris , Animaux , Transduction du signal , Récepteurs à activité tyrosine kinase/métabolisme , Site allostérique
3.
Molecules ; 29(2)2024 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-38257336

RÉSUMÉ

Naturally occurring stilbenoids, such as the (E)-stilbenoid resveratrol and the (Z)-stilbenoid combretastatin A4, have been considered as promising lead compounds for the development of anticancer drugs. The antitumour properties of stilbenoids are known to be modulated by cytochrome P450 enzymes CYP1A1 and CYP1B1, which contribute to extrahepatic phase I xenobiotic and drug metabolism. Thirty-four methyl ether analogues of resveratrol were synthesised, and their anticancer properties were assessed, using the MTT cell proliferation assay on a panel of human breast cell lines. Breast tumour cell lines that express CYP1 were significantly more strongly affected by the resveratrol analogues than the cell lines that did not have CYP1 activity. Metabolism studies using isolated CYP1 enzymes provided further evidence that (E)-stilbenoids can be substrates for these enzymes. Structures of metabolic products were confirmed by comparison with synthetic standards and LC-MS co-elution studies. The most promising stilbenoid was (E)-4,3',4',5'-tetramethoxystilbene (DMU212). The compound itself showed low to moderate cytotoxicity, but upon CYP1-catalysed dealkylation, some highly cytotoxic metabolites were formed. Thus, DMU212 selectively affects proliferation of cells that express CYP1 enzymes.


Sujet(s)
Cytochrome P-450 CYP1A1 , Famille-1 de cytochromes P450 , Humains , Resvératrol/pharmacologie , Catalyse , Lignée cellulaire tumorale
4.
J Med Chem ; 64(16): 12286-12303, 2021 08 26.
Article de Anglais | MEDLINE | ID: mdl-34387469

RÉSUMÉ

Aberrant activation of the mitogen-activated protein kinase pathway frequently drives tumor growth, and the ERK1/2 kinases are positioned at a key node in this pathway, making them important targets for therapeutic intervention. Recently, a number of ERK1/2 inhibitors have been advanced to investigational clinical trials in patients with activating mutations in B-Raf proto-oncogene or Ras. Here, we describe the discovery of the clinical candidate ASTX029 (15) through structure-guided optimization of our previously published isoindolinone lead (7). The medicinal chemistry campaign focused on addressing CYP3A4-mediated metabolism and maintaining favorable physicochemical properties. These efforts led to the identification of ASTX029, which showed the desired pharmacological profile combining ERK1/2 inhibition with suppression of phospho-ERK1/2 (pERK) levels, and in addition, it possesses suitable preclinical pharmacokinetic properties predictive of once daily dosing in humans. ASTX029 is currently in a phase I-II clinical trial in patients with advanced solid tumors.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Indoles/usage thérapeutique , Mitogen-Activated Protein Kinase 1/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Pyrimidines/usage thérapeutique , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/métabolisme , Antinéoplasiques/pharmacocinétique , Cristallographie aux rayons X , Chiens , Humains , Indoles/synthèse chimique , Indoles/métabolisme , Indoles/pharmacocinétique , Mâle , Souris de lignée BALB C , Mitogen-Activated Protein Kinase 1/composition chimique , Mitogen-Activated Protein Kinase 1/métabolisme , Structure moléculaire , Phosphorylation/effets des médicaments et des substances chimiques , Liaison aux protéines , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacocinétique , Proto-oncogène Mas , Pyrimidines/synthèse chimique , Pyrimidines/métabolisme , Pyrimidines/pharmacocinétique , Rat Sprague-Dawley , Rat Wistar , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Mol Cancer Ther ; 20(10): 1757-1768, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34330842

RÉSUMÉ

The MAPK signaling pathway is commonly upregulated in human cancers. As the primary downstream effector of the MAPK pathway, ERK is an attractive therapeutic target for the treatment of MAPK-activated cancers and for overcoming resistance to upstream inhibition. ASTX029 is a highly potent and selective dual-mechanism ERK inhibitor, discovered using fragment-based drug design. Because of its distinctive ERK-binding mode, ASTX029 inhibits both ERK catalytic activity and the phosphorylation of ERK itself by MEK, despite not directly inhibiting MEK activity. This dual mechanism was demonstrated in cell-free systems, as well as cell lines and xenograft tumor tissue, where the phosphorylation of both ERK and its substrate, ribosomal S6 kinase (RSK), were modulated on treatment with ASTX029. Markers of sensitivity were highlighted in a large cell panel, where ASTX029 preferentially inhibited the proliferation of MAPK-activated cell lines, including those with BRAF or RAS mutations. In vivo, significant antitumor activity was observed in MAPK-activated tumor xenograft models following oral treatment. ASTX029 also demonstrated activity in both in vitro and in vivo models of acquired resistance to MAPK pathway inhibitors. Overall, these findings highlight the therapeutic potential of a dual-mechanism ERK inhibitor such as ASTX029 for the treatment of MAPK-activated cancers, including those which have acquired resistance to inhibitors of upstream components of the MAPK pathway. ASTX029 is currently being evaluated in a first in human phase I-II clinical trial in patients with advanced solid tumors (NCT03520075).


Sujet(s)
Tumeurs du côlon/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Extracellular Signal-Regulated MAP Kinases/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Indoles/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Animaux , Apoptose , Cycle cellulaire , Mouvement cellulaire , Prolifération cellulaire , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Humains , Mâle , Souris , Souris de lignée BALB C , Souris nude , Phosphorylation , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
6.
J Med Chem ; 64(7): 4071-4088, 2021 04 08.
Article de Anglais | MEDLINE | ID: mdl-33761253

RÉSUMÉ

Inhibition of murine double minute 2 (MDM2)-p53 protein-protein interaction with small molecules has been shown to reactivate p53 and inhibit tumor growth. Here, we describe rational, structure-guided, design of novel isoindolinone-based MDM2 inhibitors. MDM2 X-ray crystallography, quantum mechanics ligand-based design, and metabolite identification all contributed toward the discovery of potent in vitro and in vivo inhibitors of the MDM2-p53 interaction with representative compounds inducing cytostasis in an SJSA-1 osteosarcoma xenograft model following once-daily oral administration.


Sujet(s)
Antinéoplasiques/pharmacologie , Isoindoles/pharmacologie , Ostéosarcome/traitement médicamenteux , Multimérisation de protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-mdm2/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/métabolisme , Tumeurs osseuses/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Stabilité de médicament , Femelle , Humains , Isoindoles/synthèse chimique , Isoindoles/métabolisme , Macaca fascicularis , Mâle , Souris de lignée BALB C , Souris nude , Microsomes du foie/métabolisme , Structure moléculaire , Liaison aux protéines , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Bioorg Med Chem Lett ; 29(11): 1403-1406, 2019 06 01.
Article de Anglais | MEDLINE | ID: mdl-30935796

RÉSUMÉ

As part of a programme to develop anticancer prodrugs which are activated by cytochrome P450 (CYP)1B1, a library of 4,6-diaryl-2-pyridones was synthesised in yields of 6-60% from the corresponding chalcones. A number of these derivatives showed promising antiproliferative activities in human breast cancer cell lines which express CYP1B1 and CYP1A1, while showing little toxicity towards a non-tumour breast cell line with no CYP expression. Metabolism studies provided evidence supporting the involvement of CYP1 enzymes in the bioactivation of these compounds.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Cytochrome P-450 CYP1A1/antagonistes et inhibiteurs , Cytochrome P-450 CYP1B1/antagonistes et inhibiteurs , Pyridones/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytochrome P-450 CYP1A1/génétique , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1B1/génétique , Cytochrome P-450 CYP1B1/métabolisme , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Femelle , Humains , Cellules MCF-7 , Structure moléculaire , Pyridones/synthèse chimique , Pyridones/composition chimique , Relation structure-activité
8.
J Med Chem ; 61(16): 7314-7329, 2018 08 23.
Article de Anglais | MEDLINE | ID: mdl-30091600

RÉSUMÉ

Inhibitor of apoptosis proteins (IAPs) are promising anticancer targets, given their roles in the evasion of apoptosis. Several peptidomimetic IAP antagonists, with inherent selectivity for cellular IAP (cIAP) over X-linked IAP (XIAP), have been tested in the clinic. A fragment screening approach followed by structure-based optimization has previously been reported that resulted in a low-nanomolar cIAP1 and XIAP antagonist lead molecule with a more balanced cIAP-XIAP profile. We now report the further structure-guided optimization of the lead, with a view to improving the metabolic stability and cardiac safety profile, to give the nonpeptidomimetic antagonist clinical candidate 27 (ASTX660), currently being tested in a phase 1/2 clinical trial (NCT02503423).


Sujet(s)
Antinéoplasiques/pharmacologie , Composés hétérobicycliques/pharmacologie , Pipérazines/pharmacologie , Protéine inhibitrice de l'apoptose liée au chromosome X/antagonistes et inhibiteurs , Administration par voie orale , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Lignée cellulaire tumorale , Cristallographie aux rayons X , Canal potassique ERG1/antagonistes et inhibiteurs , Composés hétérobicycliques/composition chimique , Composés hétérobicycliques/pharmacocinétique , Humains , Protéines IAP/antagonistes et inhibiteurs , Macaca fascicularis , Mâle , Souris de lignée BALB C , Pipérazines/composition chimique , Pipérazines/pharmacocinétique , Rat Sprague-Dawley , Relation structure-activité , Protéine inhibitrice de l'apoptose liée au chromosome X/composition chimique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Med Chem ; 61(11): 4978-4992, 2018 06 14.
Article de Anglais | MEDLINE | ID: mdl-29775310

RÉSUMÉ

Aberrant activation of the MAPK pathway drives cell proliferation in multiple cancers. Inhibitors of BRAF and MEK kinases are approved for the treatment of BRAF mutant melanoma, but resistance frequently emerges, often mediated by increased signaling through ERK1/2. Here, we describe the fragment-based generation of ERK1/2 inhibitors that block catalytic phosphorylation of downstream substrates such as RSK but also modulate phosphorylation of ERK1/2 by MEK without directly inhibiting MEK. X-ray crystallographic and biophysical fragment screening followed by structure-guided optimization and growth from the hinge into a pocket proximal to the C-α helix afforded highly potent ERK1/2 inhibitors with excellent kinome selectivity. In BRAF mutant cells, the lead compound suppresses pRSK and pERK levels and inhibits proliferation at low nanomolar concentrations. The lead exhibits tumor regression upon oral dosing in BRAF mutant xenograft models, providing a promising basis for further optimization toward clinical pERK1/2 modulating ERK1/2 inhibitors.


Sujet(s)
Biocatalyse/effets des médicaments et des substances chimiques , Découverte de médicament , Mitogen-Activated Protein Kinase 1/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase 3/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Administration par voie orale , Animaux , Biodisponibilité , Lignée cellulaire tumorale , Humains , Souris , Mitogen-Activated Protein Kinase 1/composition chimique , Mitogen-Activated Protein Kinase 3/composition chimique , Modèles moléculaires , Phosphorylation/effets des médicaments et des substances chimiques , Conformation des protéines , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacocinétique
10.
Mol Cancer Ther ; 17(7): 1381-1391, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29695633

RÉSUMÉ

Because of their roles in the evasion of apoptosis, inhibitor of apoptosis proteins (IAP) are considered attractive targets for anticancer therapy. Antagonists of these proteins have the potential to switch prosurvival signaling pathways in cancer cells toward cell death. Various SMAC-peptidomimetics with inherent cIAP selectivity have been tested clinically and demonstrated minimal single-agent efficacy. ASTX660 is a potent, non-peptidomimetic antagonist of cIAP1/2 and XIAP, discovered using fragment-based drug design. The antagonism of XIAP and cIAP1 by ASTX660 was demonstrated on purified proteins, cells, and in vivo in xenograft models. The compound binds to the isolated BIR3 domains of both XIAP and cIAP1 with nanomolar potencies. In cells and xenograft tissue, direct antagonism of XIAP was demonstrated by measuring its displacement from caspase-9 or SMAC. Compound-induced proteasomal degradation of cIAP1 and 2, resulting in downstream effects of NIK stabilization and activation of noncanonical NF-κB signaling, demonstrated cIAP1/2 antagonism. Treatment with ASTX660 led to TNFα-dependent induction of apoptosis in various cancer cell lines in vitro, whereas dosing in mice bearing breast and melanoma tumor xenografts inhibited tumor growth. ASTX660 is currently being tested in a phase I-II clinical trial (NCT02503423), and we propose that its antagonism of cIAP1/2 and XIAP may offer improved efficacy over first-generation antagonists that are more cIAP1/2 selective. Mol Cancer Ther; 17(7); 1381-91. ©2018 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Protéines IAP/antagonistes et inhibiteurs , Facteur de nécrose tumorale alpha/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Humains , Protéines IAP/composition chimique , Protéines IAP/métabolisme , Souris , Mimétisme moléculaire , Motifs et domaines d'intéraction protéique/effets des médicaments et des substances chimiques , Relation structure-activité , Protéine inhibitrice de l'apoptose liée au chromosome X/composition chimique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Med Chem ; 14(4): 322-332, 2018.
Article de Anglais | MEDLINE | ID: mdl-29332599

RÉSUMÉ

BACKGROUND: Although the expression levels of many P450s differ between tumour and corresponding normal tissue, CYP1B1 is one of the few CYP subfamilies which is significantly and consistently overexpressed in tumours. CYP1B1 has been shown to be active within tumours and is capable of metabolising a structurally diverse range of anticancer drugs. Because of this, and its role in the activation of procarcinogens, CYP1B1 is seen as an important target for anticancer drug development. OBJECTIVE: To synthesise a series of chalcone derivatives based on the chemopreventative agent DMU-135 and investigate their antiproliferative activities in human breast cancer cell lines which express CYP1B1 and CYP1A1. METHOD: A series of chalcones were synthesised in yields of 43-94% using the Claisen-Schmidt condensation reaction. These were screened using a MTT assay against a panel of breast cancer cell lines which have been characterised for CYP1 expression. RESULT: A number of derivatives showed promising antiproliferative activities in human breast cancer cell lines which express CYP1B1 and CYP1A1, while showing significantly lower toxicity towards a non-tumour breast cell line with no CYP expression. Experiments using the CYP1 inhibitors acacetin and α-naphthoflavone provided supporting evidence for the involvement of CYP1 enzymes in the bioactivation of these compounds. CONCLUSION: Chalcones show promise as anticancer agents with evidence suggesting that CYP1 activation of these compounds may be involved.


Sujet(s)
Antinéoplasiques/pharmacologie , Chalcone/analogues et dérivés , Inhibiteurs des enzymes du cytochrome P-450/pharmacologie , Promédicaments/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/toxicité , Naphtoflavones/pharmacologie , Tumeurs du sein/traitement médicamenteux , Lignée cellulaire tumorale , Chalcone/synthèse chimique , Chalcone/composition chimique , Chalcone/pharmacologie , Chalcone/toxicité , Cytochrome P-450 CYP1A1/antagonistes et inhibiteurs , Cytochrome P-450 CYP1B1/antagonistes et inhibiteurs , Inhibiteurs des enzymes du cytochrome P-450/synthèse chimique , Inhibiteurs des enzymes du cytochrome P-450/composition chimique , Inhibiteurs des enzymes du cytochrome P-450/toxicité , Flavones/pharmacologie , Humains , Structure moléculaire , Promédicaments/synthèse chimique , Promédicaments/composition chimique , Promédicaments/toxicité
12.
Food Chem Toxicol ; 110: 383-394, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-29097115

RÉSUMÉ

Natural flavonoids with methoxy substitutions are metabolized by CYP1 enzymes to yield the corresponding demethylated products. The present study aimed to characterize the metabolism and further antiproliferative activity of the hydroxylated flavonoids apigenin, luteolin, scutellarein, kaempferol and quercetin in CYP1 recombinant enzymes and in the CYP1 expressing cell lines MCF7 and MDA-MB-468, respectively. Apigenin was converted to luteolin and scutellarein, whereas kaempferol was metabolized only to quercetin by recombinant CYP1 enzymes. Luteolin metabolism yielded 6 hydroxyluteolin only by recombinant CYP1B1, whereas CYP1A1 and CYP1A2 were not capable of metabolizing this compound. Molecular modeling demonstrated that CYP1B1 favored the A ring orientation of apigenin and luteolin to the heme group compared with CYP1A1. The IC50 of the compounds luteolin, scutellarein and 6 hydroxyluteolin was significantly lower in MDA-MB-468, MCF7 and MCF10A cells compared with that of apigenin. Similarly, the IC50 of quercetin in MDA-MB-468 cells was significantly lower compared with that of kaempferol. The most potent compound was luteolin in MDA-MB-468 cells (IC50 = 2 ± 0.3 µM). In the presence of the CYP1-inhibitors α-napthoflavone and/or acacetin, luteolin activation was lessened. Taken collectively, the data demonstrate that the metabolism of hydroxylated flavonoids by cytochrome P450 CYP1 enzymes, notably CYP1A1 and CYP1B1, can enhance their antiproliferative activity in breast cancer cells. In addition, this antiproliferative activity is attributed to the combined action of the parent compound and the corresponding CYP1 metabolites.


Sujet(s)
Tumeurs du sein/enzymologie , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1A2/métabolisme , Cytochrome P-450 CYP1B1/métabolisme , Flavones/métabolisme , Flavonols/métabolisme , Tumeurs du sein/métabolisme , Cytochrome P-450 CYP1A1/génétique , Cytochrome P-450 CYP1A2/génétique , Cytochrome P-450 CYP1B1/génétique , Femelle , Flavones/composition chimique , Flavonols/composition chimique , Humains , Hydroxylation , Lutéoline/composition chimique , Lutéoline/métabolisme , Cellules MCF-7
13.
J Med Chem ; 60(11): 4611-4625, 2017 06 08.
Article de Anglais | MEDLINE | ID: mdl-28492317

RÉSUMÉ

XIAP and cIAP1 are members of the inhibitor of apoptosis protein (IAP) family and are key regulators of anti-apoptotic and pro-survival signaling pathways. Overexpression of IAPs occurs in various cancers and has been associated with tumor progression and resistance to treatment. Structure-based drug design (SBDD) guided by structural information from X-ray crystallography, computational studies, and NMR solution conformational analysis was successfully applied to a fragment-derived lead resulting in AT-IAP, a potent, orally bioavailable, dual antagonist of XIAP and cIAP1 and a structurally novel chemical probe for IAP biology.


Sujet(s)
Composés hétérobicycliques/composition chimique , Composés hétérobicycliques/pharmacologie , Protéines IAP/antagonistes et inhibiteurs , Pipérazines/composition chimique , Pipérazines/pharmacologie , Protéine inhibitrice de l'apoptose liée au chromosome X/antagonistes et inhibiteurs , Animaux , Lignée cellulaire tumorale , Cristallographie aux rayons X , Découverte de médicament , Cellules HEK293 , Humains , Souris , Souris de lignée BALB C , Souris SCID , Peptidomimétiques , Bibliothèques de petites molécules , Relation structure-activité
14.
J Med Chem ; 58(16): 6574-88, 2015 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-26218264

RÉSUMÉ

Inhibitor of apoptosis proteins (IAPs) are important regulators of apoptosis and pro-survival signaling pathways whose deregulation is often associated with tumor genesis and tumor growth. IAPs have been proposed as targets for anticancer therapy, and a number of peptidomimetic IAP antagonists have entered clinical trials. Using our fragment-based screening approach, we identified nonpeptidic fragments binding with millimolar affinities to both cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP). Structure-based hit optimization together with an analysis of protein-ligand electrostatic potential complementarity allowed us to significantly increase binding affinity of the starting hits. Subsequent optimization gave a potent nonalanine IAP antagonist structurally distinct from all IAP antagonists previously reported. The lead compound had activity in cell-based assays and in a mouse xenograft efficacy model and represents a highly promising start point for further optimization.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines IAP/antagonistes et inhibiteurs , Protéines IAP/effets des médicaments et des substances chimiques , Fragments peptidiques/pharmacologie , Protéine inhibitrice de l'apoptose liée au chromosome X/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacocinétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Biologie informatique , Conception de médicament , Découverte de médicament , Tests de criblage à haut débit , Humains , Souris , Souris de lignée BALB C , Modèles moléculaires , Fragments peptidiques/synthèse chimique , Fragments peptidiques/pharmacocinétique , Pipérazines/synthèse chimique , Pipérazines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
15.
J Med Chem ; 53(14): 5213-28, 2010 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-20565112

RÉSUMÉ

Lead optimization studies using 7 as the starting point led to a new class of imidazo[4,5-b]pyridine-based inhibitors of Aurora kinases that possessed the 1-benzylpiperazinyl motif at the 7-position, and displayed favorable in vitro properties. Cocrystallization of Aurora-A with 40c (CCT137444) provided a clear understanding into the interactions of this novel class of inhibitors with the Aurora kinases. Subsequent physicochemical property refinement by the incorporation of solubilizing groups led to the identification of 3-((4-(6-bromo-2-(4-(4-methylpiperazin-1-yl)phenyl)-3H-imidazo[4,5-b]pyridin-7-yl)piperazin-1-yl)methyl)-5-methylisoxazole (51, CCT137690) which is a potent inhibitor of Aurora kinases (Aurora-A IC(50) = 0.015 +/- 0.003 muM, Aurora-B IC(50) = 0.025 muM, Aurora-C IC(50) = 0.019 muM). Compound 51 is highly orally bioavailable, and in in vivo efficacy studies it inhibited the growth of SW620 colon carcinoma xenografts following oral administration with no observed toxicities as defined by body weight loss.


Sujet(s)
Imidazoles/synthèse chimique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyridines/synthèse chimique , Administration par voie orale , Animaux , Aurora kinase A , Aurora kinase B , Aurora kinase C , Aurora kinases , Biodisponibilité , Protéines du sang/métabolisme , Lignée cellulaire tumorale , Tests de criblage d'agents antitumoraux , Canal potassique ERG1 , Canaux potassiques éther-à-go-go/antagonistes et inhibiteurs , Femelle , Humains , Imidazoles/pharmacocinétique , Imidazoles/pharmacologie , Techniques in vitro , Mâle , Souris , Souris de lignée BALB C , Souris nude , Microsomes du foie/métabolisme , Modèles moléculaires , Transplantation tumorale , Liaison aux protéines , Pyridines/pharmacocinétique , Pyridines/pharmacologie , Relation structure-activité , Transplantation hétérologue
16.
Mol Cancer Ther ; 6(12 Pt 1): 3147-57, 2007 Dec.
Article de Anglais | MEDLINE | ID: mdl-18089709

RÉSUMÉ

The Aurora family of serine/threonine kinases is important for the regulation of centrosome maturation, chromosome segregation, and cytokinesis during mitosis. Overexpression of Aurora kinases in mammalian cells leads to genetic instability and transformation. Increased levels of Aurora kinases have also been linked to a broad range of human tumors. Here, we describe the properties of CCT129202, a representative of a structurally novel series of imidazopyridine small-molecule inhibitors of Aurora kinase activity. This compound showed high selectivity for the Aurora kinases over a panel of other kinases tested and inhibits proliferation in multiple cultured human tumor cell lines. CCT129202 causes the accumulation of human tumor cells with >or=4N DNA content, leading to apoptosis. CCT120202-treated human tumor cells showed a delay in mitosis, abrogation of nocodazole-induced mitotic arrest, and spindle defects. Growth of HCT116 xenografts in nude mice was inhibited after i.p. administration of CCT129202. We show that p21, the cyclin-dependent kinase inhibitor, is induced by CCT129202. Up-regulation of p21 by CCT129202 in HCT116 cells led to Rb hypophosphorylation and E2F inhibition, contributing to a decrease in thymidine kinase 1 transcription. This has facilitated the use of 3'-deoxy-3'[(18)F]fluorothymidine-positron emission tomography to measure noninvasively the biological activity of the Aurora kinase inhibitor CCT129202 in vivo.


Sujet(s)
Antienzymes/pharmacologie , Imidazoles/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyridines/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Aurora kinases , Lignée cellulaire tumorale , Régulation négative , Antienzymes/pharmacocinétique , Test ELISA , Femelle , Histone/métabolisme , Humains , Souris , Microscopie de fluorescence , Mitose/effets des médicaments et des substances chimiques , Séquençage par oligonucléotides en batterie , Phosphorylation , Protéine du rétinoblastome/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
17.
Drug Metab Dispos ; 35(7): 1017-22, 2007 Jul.
Article de Anglais | MEDLINE | ID: mdl-17403917

RÉSUMÉ

Phosphoinositide-specific phospholipase C (PLC) is a key enzyme in the regulation of Ca(2+) release from inositol 1,4,5-triphosphate-sensitive stores. U73122 (1-(6-((17beta-3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione) has been extensively used as a pharmacological inhibitor of PLC to elucidate the importance of this enzyme family in signal transduction pathways. U73122 has an electrophilic maleimide group, which readily reacts with nucleophiles such as thiols and amines. In the current study the conjugation of U73122 to common components of cell culture medium, namely l-glutamine, glutathione, and bovine serum albumin (BSA), was demonstrated. The half-life of U73122 on incubation with phosphate-buffered saline (PBS), Hanks' buffered saline solution (with 2 mM glutamine), optimized basal nutrient medium (MCDB131, without BSA), complete medium, Dulbecco's modified Eagle's medium (with 2 mM l-glutamine) was approximately 150, 60, 32, 30, and 18 min, respectively. However, U73122 was not recoverable from medium supplemented with 0.5% BSA. U73122 underwent hydrolysis of the maleimide group when incubated with PBS. Glutamine conjugates of U73122 were identified in cell culture medium. Furthermore, the inhibition of epidermal growth factor-stimulated Ca(2+) release in a human epidermoid carcinoma cell line (A431) by U73122 was substantially reduced by the presence of BSA in a time-dependent manner. In complex cellular assays, the availability of U73122 to inhibit PLC may be limited by its chemical reactivity and lead to the misinterpretation of results in pharmacological assays.


Sujet(s)
Artéfacts , Dosage biologique , Signalisation calcique/effets des médicaments et des substances chimiques , Milieux de culture/composition chimique , Évaluation préclinique de médicament/méthodes , Antienzymes/pharmacologie , Oestrènes/pharmacologie , Pyrrolidones/pharmacologie , Type C Phospholipases/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Milieux de culture/métabolisme , Stabilité de médicament , Antienzymes/composition chimique , Antienzymes/métabolisme , Facteur de croissance épidermique/pharmacologie , Oestrènes/composition chimique , Oestrènes/métabolisme , Glutamine/composition chimique , Glutathion/composition chimique , Période , Humains , Liaison aux protéines , Pyrrolidones/composition chimique , Pyrrolidones/métabolisme , Sérumalbumine bovine/composition chimique , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE