Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 83
Filtrer
1.
Dalton Trans ; 53(33): 13871-13889, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39091221

RÉSUMÉ

Piperazine is an important functional unit of many clinically approved drugs, including chemotherapeutic agents. In the current study, methyl piperazine was incorporated and eight salicylaldehyde-derived piperazine-functionalized hydrazone ONN-donor ligands (L) and their Pt(II) complexes (L-PtCl) were prepared. The structures of all these ligands (L1-L8) and Pt(II) complexes (C1-C8) were determined using 1H and 13C NMR, UV-vis, FT-IR and HR-ESI MS analyses, whereas the structures of C1, C5, C6, C7 and C8 were determined in the solid state using single crystal X-ray diffraction analysis. Solution state stabilities of C3, C4, C5 and C6 were determined via time-dependent UV-vis spectroscopy. All these complexes (C1-C8) were studied for their anticancer effect in pancreatic ductal adenocarcinoma cells, including BxPC3, MIAPaCa-2 and PANC1 cells. C1-C8 displayed a potential cytotoxic effect in all these cancer cells, among which C5, C6 and C8 showed the strongest inhibitory effect in comparison with standard chemotherapeutic agents, including 5-fluorouracil (5-FU), cisplatin (CP), oxaliplatin and doxorubicin (DOX). C5, C6 and C8 suppressed the growth of pancreatic cancer cells in a dose-dependent manner. Moreover, C5, C6 and C8 inhibited clonogenic potential and invasion ability and induced apoptosis in PANC1 cells. Importantly, C5, C6 and C8 synergized the anticancer effect with PARP inhibitors, including olaparib, veliparib and niraparib, in pancreatic cancer cells, thus suggesting an important role of C5, C6 and C8 in induction of apoptosis in combination with PARP inhibitors. C5 combined with PARP inhibitors induced caspase3/7 activity and suppressed ATP production. Mechanistically, C5, C6 and C8 inhibited EZH2 protein expression to suppress EZH2-dependent tumorigenesis. Overall, these results highlighted the importance of these piperazine-functionalized Pt(II) complexes as potential anticancer agents to suppress pancreatic ductal adenocarcinoma tumorigenesis by targeting the EZH2-dependent pathway.


Sujet(s)
Aldéhydes , Antinéoplasiques , Apoptose , Protéine-2 homologue de l'activateur de Zeste , Hydrazones , Tumeurs du pancréas , Pipérazine , Inhibiteurs de poly(ADP-ribose) polymérases , Apoptose/effets des médicaments et des substances chimiques , Humains , Hydrazones/composition chimique , Hydrazones/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Ligands , Aldéhydes/composition chimique , Aldéhydes/pharmacologie , Pipérazine/composition chimique , Pipérazine/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Lignée cellulaire tumorale , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pipérazines/composition chimique , Tests de criblage d'agents antitumoraux , Synergie des médicaments , Complexes de coordination/pharmacologie , Complexes de coordination/composition chimique , Complexes de coordination/synthèse chimique , Composés organiques du platine/pharmacologie , Composés organiques du platine/composition chimique , Composés organiques du platine/synthèse chimique
2.
Chem Commun (Camb) ; 60(67): 8892-8895, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39086281

RÉSUMÉ

A biological nanoplatform (Gal-ANI@ZnAP NPs) was constructed based on a prodrug-skeletal metal-organic framework (MOF) using purine nucleobase analogue prodrug 6-allylthiopurine as a bioactive ligand, and functionalized with AIE fluorescent PARP inhibitor glycoconjugate for visualization therapy and synthetic lethal cancer therapy. This nanoplatform could actively target cancer cells, selectively release drugs in response to esterase/pH, and visualize drug uptake. In vitro studies revealed that Gal-ANI@ZnAP NPs increased the synthetic lethality in cancer cells by inducing DNA repair failure with the simultaneous targeting of PARP and nucleotide metabolism, thereby exhibiting a significant cancer-killing effect. The study presents a novel strategy to construct an AIE nanoplatform using pharmaceutical molecules for drug uptake visualization and boosting synthetic lethality in cancer.


Sujet(s)
Antinéoplasiques , Réseaux organométalliques , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Glycosylation , Promédicaments/composition chimique , Promédicaments/pharmacologie , Promédicaments/synthèse chimique , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Tests de criblage d'agents antitumoraux , Survie cellulaire/effets des médicaments et des substances chimiques
3.
Eur J Med Chem ; 277: 116726, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39116535

RÉSUMÉ

Structural modification based on natural privileged scaffolds has proven to be an attractive approach to generate potential antitumor candidates with high potency and specific targeting. As a continuation of our efforts to identify potent PARP-1 inhibitors, natural 3-arylcoumarin scaffold was served as the starting point for the construction of novel structural unit for PARP-1 inhibition. Herein, a series of novel 8-carbamyl-3-arylcoumarin derivatives were designed and synthesized. The antiproliferative activities of target compounds against four BRCA-mutated cancer cells (SUM149PT, HCC1937, MDA-MB-436 and Capan-1) were evaluated. Among them, compound 9b exhibited excellent antiproliferative effects against SUM149PT, HCC1937 and Capan-1 cells with IC50 values of 0.62, 1.91 and 4.26 µM, respectively. Moreover, 9b could significantly inhibit the intracellular PARP-1/2 activity in SUM149PT cells with IC50 values of 2.53 nM and 6.45 nM, respectively. Further mechanism studies revealed that 9b could aggravate DNA double-strand breaks, increase ROS production, decrease mitochondrial membrane potential, arrest cell cycle at G2/M phase and ultimately induce apoptosis in SUM149PT cells. In addition, molecular docking study demonstrated that the binding mode of 9b with PARP-1 was similar to that of niraparib, forming multiple hydrogen bond interactions with the active site of PARP-1. Taken together, these findings suggest that 8-carbamyl-3-arylcoumarin scaffold could serve as an effective structural unit for PARP-1 inhibition and offer a valuable paradigm for the structural modification of natural products.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Coumarines , Tests de criblage d'agents antitumoraux , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Coumarines/pharmacologie , Coumarines/composition chimique , Coumarines/synthèse chimique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Structure moléculaire , Relation dose-effet des médicaments , Découverte de médicament , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Produits biologiques/pharmacologie , Produits biologiques/composition chimique , Produits biologiques/synthèse chimique
4.
J Enzyme Inhib Med Chem ; 39(1): 2383886, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39072709

RÉSUMÉ

Specifically inducing the degradation of acidic nucleoplasmic DNA-binding protein 1 (And1) is a promising antitumor strategy. Our previous study identified Bazedoxifene (BZA) and CH3 as specific And1 degraders and validated their activity in reversing radiotherapy resistance in vitro and in vivo. However, unelucidated structure-activity relationships and moderate activity have limited their application. In this study, 27 novel CH3 derivatives were designed and synthesised based on the cavity topology of the WD40 domain of And1. Among them, A15 with a "V" conformation significantly induced And1 degradation in NSCLC cells. In addition, this study demonstrated a potential synthetic lethal effect of And1 degraders and PARP1 inhibitors. 1 µM of Olaparib in combination with 5 µM of A15 significantly inhibited the proliferation of A549 and H460 cells. Overall, these compounds are valuable tools for elucidating And1 biology, and their special spatial conformation make them promising candidates for future optimisation studies.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Tumeurs du poumon , Poly (ADP-Ribose) polymerase-1 , Stilbènes , Humains , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Structure moléculaire , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Stilbènes/pharmacologie , Stilbènes/composition chimique , Stilbènes/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Lignée cellulaire tumorale
5.
Bioorg Chem ; 151: 107556, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39068717

RÉSUMÉ

In recent years, poly(ADP-ribose)polymerase-1 (PARP-1) and histone deacetylase (HDAC) have emerged as significant targets in tumor therapy, garnering widespread attention. In this study, we designed and synthesized two novel phthalazinone PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors, named DLC-1-46 containing dithiocarboxylate fragments and DLC-47-63 containing hydroxamic acid fragments, and evaluated their inhibitory activities on enzymes and cells. Among the PARP-1 inhibitors, most compounds exhibited high inhibitory activity against the PARP-1 enzyme, with DLC-1-6 being particularly notable, showing IC50 values <0.2 nM. Notably, DLC-1 demonstrated significant anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells reaching 0.08, 26.39, and 1.01 µM, respectively. Further investigation revealed that DLC-1 arrested MDA-MB-231 cells in the G1 phase and induced apoptosis in a dose-dependent manner. Among the designed dual PARP-1/HDAC-1 inhibitors, several compounds exhibited potent dual-target inhibitory activity, with DLC-49 displaying IC50 values of 0.53 nM and 17 nM for PARP-1 and HDAC-1, respectively. DLC-50 demonstrated the most potent anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells at 0.30, 2.70, and 2.41 µM, respectively. Cell cycle arrest and apoptosis assays indicated that DLC-50 arrested the cell cycle in the G2 phase and induced apoptosis in HCT-116 cells. Our findings present a novel avenue for further exploration of PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Conception de médicament , Tests de criblage d'agents antitumoraux , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Phtalazines , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/synthèse chimique , Inhibiteurs de désacétylase d'histone/composition chimique , Relation structure-activité , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Phtalazines/pharmacologie , Phtalazines/synthèse chimique , Phtalazines/composition chimique , Structure moléculaire , Relation dose-effet des médicaments , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire
6.
Molecules ; 29(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38930900

RÉSUMÉ

The malignancy of breast cancer poses a global challenge, with existing treatments often falling short of desired efficacy. Extensive research has underscored the effectiveness of targeting the metabolism of nicotinamide adenine dinucleotide (NAD), a pivotal molecule crucial for cancer cell survival and growth, as a promising anticancer strategy. Within mammalian cells, sustaining optimal NAD concentrations relies on two key enzymes, namely nicotinamide phosphoribosyltransferase (NAMPT) and poly(ADP-ribose) polymer 1 (PARP1). Recent studies have accentuated the potential benefits of combining NAMPT inhibitors and PARP1 inhibitors to enhance therapeutic outcomes, particularly in breast cancer. In this study, we designed and synthesized eleven novel NAMPT/PARP1 dual-target inhibitors. Among them, compound DDY02 exhibited acceptable inhibitory activities against both NAMPT and PARP1, with IC50 values of 0.01 and 0.05 µM, respectively. Moreover, in vitro evaluations revealed that treatment with DDY02 resulted in proliferation inhibition, NAD depletion, DNA damage, apoptosis, and migration inhibition in MDA-MB-468 cells. These results posit DDY02, by targeting NAD metabolism through inhibiting both NAMPT and PARP1, as a promising lead compound for the development of breast cancer therapy.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Prolifération cellulaire , NAD , Nicotinamide phosphoribosyltransferase , Poly (ADP-Ribose) polymerase-1 , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Nicotinamide phosphoribosyltransferase/métabolisme , Humains , NAD/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Conception de médicament , Cytokines/métabolisme , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Simulation de docking moléculaire
7.
Eur J Med Chem ; 274: 116535, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38838546

RÉSUMÉ

Poly (ADP-ribose) polymerase (PARP) is considered an essential component in case of DNA (Deoxyribonucleic acid) damage, response by sensing DNA damage and engaging DNA repair proteins. Those proteins repair the damaged DNA via an aspect of posttranslational modification, known as poly (ADP-Ribosyl)ation (PARylation). Specifically, PARP inhibitors (PARPi) have shown better results when administered alone in a variety of cancer types with BRCA (Breast Cancer gene) mutation. The clinical therapeutic benefits of PARP inhibitors have been diminished by their cytotoxicity, progression of drug resistance, and limitation of indication, regardless of their tremendous clinical effectiveness. A growing number of PARP-1 inhibitors, particularly those associated with BRCA-1/2 mutations, have been identified as potential cancer treatments. Recently, several researchers have identified various promising scaffolds, which have resulted in the resuscitation of the faith in PARP inhibitors as cancer therapies. This review provided a comprehensive update on the anatomy and physiology of the PARP enzyme, the profile of FDA (Food and Drug Administration) and CFDA (China Food and Drug Administration)-approved drugs, and small-molecule inhibitors of PARP, including their synthetic routes, biological evaluation, selectivity, and structure-activity relationship.


Sujet(s)
Antinéoplasiques , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Poly(ADP-ribose) polymerases/métabolisme , Structure moléculaire , Animaux
8.
J Med Chem ; 67(13): 10848-10874, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38912753

RÉSUMÉ

Bifunctional conjugates targeting PD-L1/PARP7 were designed, synthesized, and evaluated for the first time. Compounds B3 and C6 showed potent activity against PD-1/PD-L1 interaction (IC50 = 0.426 and 0.342 µM, respectively) and PARP7 (IC50 = 2.50 and 7.05 nM, respectively). They also displayed excellent binding affinity with hPD-L1, approximately 100-200-fold better than that of hPD-1. Both compounds restored T-cell function, leading to the increase of IFN-γ secretion. In the coculture assay, B3 and C6 enhanced the killing activity of MDA-MB-231 cells by Jurkat T cells in a concentration-dependent manner. Furthermore, B3 and C6 displayed significant in vivo antitumor efficacy in a melanoma B16-F10 tumor mouse model, more than 5.3-fold better than BMS-1 (a PD-L1 inhibitor) and RBN-2397 (a PARP7i clinical candidate) at the dose of 25 mg/kg, without observable side effects. These results provide valuable insight and understanding for developing bifunctional conjugates for potential anticancer therapy.


Sujet(s)
Antinéoplasiques , Antigène CD274 , Immunothérapie , Humains , Animaux , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Antinéoplasiques/usage thérapeutique , Immunothérapie/méthodes , Lignée cellulaire tumorale , Souris de lignée C57BL , Femelle , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/anatomopathologie , Mélanome expérimental/thérapie
9.
Chin J Nat Med ; 22(5): 455-465, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38796218

RÉSUMÉ

In this study, we reported the discovery and structure-activity relationship analysis of chrysin derivatives as a new class of inhibitors targeting poly (ADP-ribose) polymerase 1 (PARP1). Among these derivatives, compound 5d emerged as the most effective chrysin-based inhibitor of PARP1, with an IC50 value of 108 nmol·L-1. This compound significantly inhibited the proliferation and migration of breast cancer cell lines HCC-1937 and MDA-MB-436 by inducing DNA damage. Furthermore, 5d induced apoptosis and caused an extended G1/S-phase in these cell lines. Molecular docking studies revealed that 5d possesses a strong binding affinity toward PARP1. In vivo, in a xenograft model, 5d effectively reduced tumor growth by downregulating PARP1 expression. Overall, compound 5d shows promise as a potential therapeutic agent for the treatment of BRCA wild-type breast cancer.


Sujet(s)
Apoptose , Tumeurs du sein , Prolifération cellulaire , Flavonoïdes , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Flavonoïdes/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Femelle , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Lignée cellulaire tumorale , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Souris , Conception de médicament , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Souris nude , Souris de lignée BALB C , Structure moléculaire
10.
Bioorg Chem ; 148: 107455, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38772289

RÉSUMÉ

Poly(ADP-ribose) polymerase-1 (PARP-1) is a crucial member of DNA repair enzymes responsible for repairing DNA single-strand breaks. Developing PARP inhibitors based on synthetic lethality strategies is an effective approach for treating breast cancer and other diseases. In this study, a series of novel piperidine-based benzamide derivatives were designed and synthesized using structure-based drug design principles. The anticancer activities of these compounds were evaluated against five human cancer cell lines (MDA-MB-436, CAPAN-1, SW-620, HepG2, SKOV3, and PC3) and the preliminary structure-activity relationships were delineated. Among the compounds, 6a and 15d demonstrated potent antiproliferative effects against MDA-MB-436 cells with IC50 values of 8.56 ± 1.07 µM and 6.99 ± 2.62 µM, respectively. Furthermore, both compounds exhibited excellent inhibitory activity against PARP-1, with IC50 values of 8.33 nM and 12.02 nM, respectively. Mechanistic investigations revealed that 6a and 15d effectively inhibited colony formation and cell migration of HCT116 cells. Moreover, they induced apoptosis by upregulating the expression of Bax and cleaved Caspase-3, while downregulating the expression of Caspase-3 and Bcl-2 in HCT116 cells. Based on its impressive pharmacodynamic data in vitro, we conducted a study to evaluate the efficacy of 15d in a xenograft tumor model in mice when used in combination with cytotoxic agents. Collectively, these findings suggest that 15d could be promising drug candidates worthy of further investigation.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Pipéridines , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Pipéridines/pharmacologie , Pipéridines/composition chimique , Pipéridines/synthèse chimique , Relation structure-activité , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Structure moléculaire , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris nude , Souris de lignée BALB C
11.
Bioorg Chem ; 148: 107480, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38772291

RÉSUMÉ

A novel series of erythrina derivatives as PARP-1/FTase inhibitors were synthesized, and evaluated for their biological activities. Compound T9 had excellent inhibitory effects on cell viability (A549: IC50 = 1.74 µM; A549/5-Fu: IC50 = 1.03 µM) and in vitro enzyme activities (PARP-1: IC50 = 0.40 µM; FTase: IC50 = 0.067 µM). Molecular docking and point mutation assays demonstrated the interaction of compound T9 with key amino acid residues. The compound T9 exhibited potent anti-proliferation and anti-migration capabilities against A549 and A549/5-Fu cells. PCR array and western blot results showed that compound T9 could effectively inhibit EMT-related proteins in A549 and A549/5-Fu cells, thereby inhibiting the development of lung cancer. Importantly, compound T9 could significantly inhibit tumor growth in the A549 xenograft tumor model (TGI = 65.3 %). In conclusion, this study was the first presentation of the concept of dual-target inhibitors of the PARP-1/FTase enzymes. It also provides the basis for further research and development of novel PARP-1/FTase inhibitors.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Transition épithélio-mésenchymateuse , Erythrina , Tumeurs du poumon , Poly (ADP-Ribose) polymerase-1 , Humains , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Relation structure-activité , Erythrina/composition chimique , Animaux , Structure moléculaire , Souris , Simulation de docking moléculaire , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Souris nude , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/anatomopathologie , Tumeurs expérimentales/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques
12.
Bioorg Chem ; 148: 107469, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38781669

RÉSUMÉ

PARP7 has been proven to play an important role in immunity. Substantial upregulation of PARP7 is observed in numerous cancerous cell types, consequently resulting in the inhibition of type Ⅰ interferon signaling pathways. Therefore, inhibiting the activity of PARP7 can enhance type Ⅰ interferon signaling to exert an anti-tumor immune response. In this study, we reported the identification of a newly found PARP7 inhibitor (XLY-1) with higher inhibitory activity (IC50 = 0.6 nM) than that of RBN-2397 (IC50 = 6.0 nM). Additionally, XYL-1 displayed weak inhibitory activity on PARP1 (IC50 > 1.0 µM). Mechanism studies showed that XYL-1 could enhance the type Ⅰ interferon signaling in vitro. Pharmacodynamic experiments showed that 50 mg/kg XYL-1 could significantly inhibit tumor growth (TGI: 76.5 %) and related experiments showed that XYL-1 could restore type Ⅰ interferon signaling and promote T cell infiltration in tumor tissues. Taken together, XYL-1 shows promise as a potential candidate for developing cancer immunotherapy agents.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Relation dose-effet des médicaments , Découverte de médicament , Tests de criblage d'agents antitumoraux , Immunothérapie , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Structure moléculaire , Animaux , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Poly(ADP-ribose) polymerases/métabolisme , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Souris de lignée BALB C
13.
J Med Chem ; 67(11): 8877-8901, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38776379

RÉSUMÉ

Designing selective PARP-1 inhibitors has become a new strategy for anticancer drug development. By sequence comparison of PARP-1 and PARP-2, we identified a possible selective site (S site) consisting of several different amino acid residues of α-5 helix and D-loop. Targeting this S site, 140 compounds were designed, synthesized, and characterized for their anticancer activities and mechanisms. Compound I16 showed the highest PARP-1 enzyme inhibitory activity (IC50 = 12.38 ± 1.33 nM) and optimal selectivity index over PARP-2 (SI = 155.74). Oral administration of I16 (25 mg/kg) showed high inhibition rates of Hela and SK-OV-3 tumor cell xenograft models, both of which were higher than those of the oral positive drug Olaparib (50 mg/kg). In addition, I16 has an excellent safety profile, without significant toxicity at high oral doses. These findings provide a novel design strategy and chemotype for the development of safe, efficient, and highly selective PARP-1 inhibitors.


Sujet(s)
Antinéoplasiques , Conception de médicament , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Souris , Relation structure-activité , Lignée cellulaire tumorale , Souris nude , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules HeLa , Simulation de docking moléculaire , Souris de lignée BALB C , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phtalazines/pharmacologie , Phtalazines/composition chimique , Phtalazines/synthèse chimique
14.
Eur J Med Chem ; 271: 116405, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38678823

RÉSUMÉ

PARPi have been explored and applied in the treatment of various cancers with remarkable efficacy, especially BRCA1/2 mutated ovarian, breast, prostate, and pancreatic cancers. However, PARPi renders inevitable drug resistance and showed high toxicity because of PARP-Trapping with long-term clinic tracking. To overcome the drug resistance and the high toxicity of PARPi, many novel methods have been developed including PROTACs. Being an event-driven technology, PROTACs needs a high affinity, low toxicity warhead with no steric hindrance in binding process. Veliparib shows the lowest PARP-Trapping effect but could hardly to be the warhead of PROTACs because of the strong steric hindrance. Other PARP1 inhibitors showed less steric hindrance but owns high PARP-Trapping effect. Thus, the development of novel warhead with high PARP1 affinity, low PARP1-Trapping, and no steric hindrance would be valuable. In this work, we reserved benzimidazole as the motif to reserve the low PARP1-Trapping effect and substituted the pyrrole by aromatic ring to avoiding the steric hindrance in PARP1 binding cave. Thus, a series of benzimidazole derivates were designed and synthesized, and some biological activities in vitro were evaluated including the inhibition for PARP1 enzyme and the PARP-Trapping effect using MDA-MB-436 cell line. Results showed that the compound 19A10 has higher PARP1 affinity(IC50 = 4.62 nM)) and similar low PARP-Trapping effect compared with Veliparib(IC50 (MDA-MB-436) >100 µM). Docking study showed that the compound 19A10 could avoiding the steric hindrance which was much better than Veliparib. So, the compound 19A10 could potentially be a perfect warhead for PARP1 degraders. Besides, because of the depletion of the PARP1 and the decreasing of the binding capability, we suppose that the PROTACs using 19A10 as the warhead would be no-PARP-Trapping effect. Furthermore, QSAR study showed that to develop novel compounds with high PARP1 binding affinity and low PARP-Trapping, we can choose the skeleton with substituent R1H, R2 = piperiazine, and R3 with large tPSA. And, if we want to develop the compounds with high PARP1 binding affinity and high PARP-Trapping which can possibly improve the lethality against tumor cells, we can choose the skeleton with substituent R1F, R2 = 3-methy-piperiazine, and R3 with large tPSA.


Sujet(s)
Antinéoplasiques , Benzimidazoles , Tests de criblage d'agents antitumoraux , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Benzimidazoles/composition chimique , Benzimidazoles/pharmacologie , Benzimidazoles/synthèse chimique , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Structure moléculaire , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Simulation de docking moléculaire
15.
ChemMedChem ; 19(11): e202400093, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38482564

RÉSUMÉ

Inhibition of poly (ADP-ribose) polymerase-1 (PARP1), a DNA repair enzyme, has proven to be a successful strategy for the treatment of various cancers. With the appropriate selection conditions and protein design, DNA-encoded library (DEL) technology provides a powerful avenue to identify small molecules with the desired mechanism of action towards a target of interest. However, DNA-binding proteins, such as PARP1, can be challenging targets for DEL screening due to non-specific protein-DNA interactions. To overcome this, we designed and screened a PARP1 catalytic domain construct without the autoinhibitory helical domain. This allowed us to interrogate an active, functionally-relevant form of the protein resulting in the discovery of novel isoindolinone PARP1 inhibitors with single-digit nanomolar potency. These inhibitors also demonstrated little to no PARP1-DNA trapping, a property that could be advantageous in the clinic.


Sujet(s)
ADN , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , ADN/composition chimique , ADN/métabolisme , Relation structure-activité , Découverte de médicament , Structure moléculaire , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/synthèse chimique , Relation dose-effet des médicaments , Isoindoles/composition chimique , Isoindoles/pharmacologie , Isoindoles/synthèse chimique , Domaine catalytique
16.
Mol Inform ; 43(4): e202300183, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38258328

RÉSUMÉ

De novo design has been a hotly pursued topic for many years. Most recent developments have involved the use of deep learning methods for generative molecular design. Despite increasing levels of algorithmic sophistication, the design of molecules that are synthetically accessible remains a major challenge. Reaction-based de novo design takes a conceptually simpler approach and aims to address synthesisability directly by mimicking synthetic chemistry and driving structural transformations by known reactions that are applied in a stepwise manner. However, the use of a small number of hand-coded transformations restricts the chemical space that can be accessed and there are few examples in the literature where molecules and their synthetic routes have been designed and executed successfully. Here we describe the application of reaction-based de novo design to the design of synthetically accessible and biologically active compounds as proof-of-concept of our reaction vector-based software. Reaction vectors are derived automatically from known reactions and allow access to a wide region of synthetically accessible chemical space. The design was aimed at producing molecules that are active against PARP1 and which have improved brain penetration properties compared to existing PARP1 inhibitors. We synthesised a selection of the designed molecules according to the provided synthetic routes and tested them experimentally. The results demonstrate that reaction vectors can be applied to the design of novel molecules of biological relevance that are also synthetically accessible.


Sujet(s)
Conception de médicament , Inhibiteurs de poly(ADP-ribose) polymérases , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Humains , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Logiciel
17.
J Enzyme Inhib Med Chem ; 37(1): 952-972, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-35317687

RÉSUMÉ

The poly (ADP-ribose) polymerase (PARP) inhibitors play a crucial role in cancer therapy. However, most approved PARP inhibitors cannot cross the blood-brain barrier, thus limiting their application in the central nervous system. Here, 55 benzodiazepines were designed and synthesised to screen brain penetrating PARP-1 inhibitors. All target compounds were evaluated for their PARP-1 inhibition activity, and compounds with better activity were selected for further assays in vitro. Among them, compounds H34, H42, H48, and H52 displayed acceptable inhibition effects on breast cancer cells. Also, computational prediction together with the permeability assays in vitro and in vivo proved that the benzodiazepine PARP-1 inhibitors we synthesised were brain permeable. Compound H52 exhibited a B/P ratio of 40 times higher than that of Rucaparib and would be selected to develop its potential use in neurodegenerative diseases. Our study provided potential lead compounds and design strategies for the development of brain penetrating PARP-1 inhibitors.HIGHLIGHTSStructural fusion was used to screen brain penetrating PARP-1 inhibitors.55 benzodiazepines were evaluated for their PARP-1 inhibition activity.Four compounds displayed acceptable inhibition effects on breast cancer cells.The benzodiazepine PARP-1 inhibitors were proved to be brain permeable.


Sujet(s)
Benzodiazépines/pharmacologie , Conception de médicament , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Benzodiazépines/synthèse chimique , Benzodiazépines/composition chimique , Relation dose-effet des médicaments , Humains , Structure moléculaire , Poly (ADP-Ribose) polymerase-1/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Relation structure-activité
18.
Eur J Med Chem ; 228: 114012, 2022 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-34864331

RÉSUMÉ

Cyclin-dependent kinase 12 (CDK12) plays a crucial role in DNA-damage response gene transcription and has recently been validated as a promising target in cancer therapy. However, existing CDK12 inhibitors potently inhibit its closest isoform CDK13, which could cause potential toxicity. Therefore, the development of CDK12 inhibitors with isoform-selectivity against CDK13 continues to be a challenge. By taking advantage of the emerging PROteolysis-TArgeting Chimeras (PROTACs) approach, we have synthesized a potent PROTAC degrader PP-C8 based on the noncovalent dual inhibitors of CDK12/13 and demonstrated its specificity for CDK12 over CDK13. Notably, PP-C8 induces profound degradation of cyclin K simultaneously and downregulates the mRNA level of DNA-damage response genes. Global proteomics profiling revealed PP-C8 is highly selective toward CDK12-cyclin K complex. Importantly, PP-C8 demonstrates profound synergistic antiproliferative effects with PARP inhibitor in triple-negative breast cancer (TNBC). The potent and selective CDK12 PROTAC degrader developed in this study could potentially be used to treat CDK12-dependent cancers as combination therapy.


Sujet(s)
Protéine-kinase CDC2/antagonistes et inhibiteurs , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Cyclines/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly(ADP-ribose) polymerases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Protéine-kinase CDC2/métabolisme , Lignée cellulaire tumorale , Kinases cyclines-dépendantes/métabolisme , Cyclines/métabolisme , Relation dose-effet des médicaments , Humains , Modèles moléculaires , Structure moléculaire , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Protéolyse/effets des médicaments et des substances chimiques , Relation structure-activité
19.
Eur J Med Chem ; 227: 113898, 2022 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-34656898

RÉSUMÉ

The poly (ADP-ribose) polymerase (PARP) inhibitors play a crucial role in cancer therapy. However, most approved PARP inhibitors have lower selectivity to PARP-1 than to PARP-2, so they will inevitably have side effects. Based on the different catalytic domains of PARP-1 and PARP-2, we developed a strategy to design and synthesize highly selective PARP-1 inhibitors. Compounds Y17, Y29, Y31 and Y49 showed excellent PARP-1 inhibition, and their IC50 values were 0.61, 0.66, 0.41 and 0.96 nM, respectively. Then, Y49 (PARP-1 IC50 = 0.96 nM, PARP-2 IC50 = 61.90 nM, selectivity PARP-2/PARP-1 = 64.5) was proved to be the most selective inhibitor of PARP-1. Compounds Y29 and Y49 showed stronger inhibitory effect on proliferation in BRCA1 mutant MX-1 cells than in other cancer cells. In the MDA-MB-436 xenotransplantation model, Y49 was well tolerated and showed remarkable single dose activity. The design strategy proposed in this paper is of far-reaching significance for the further construction of the next generation of selective PARP-1 inhibitors.


Sujet(s)
Antinéoplasiques/pharmacologie , Conception de médicament , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly(ADP-ribose) polymerases/métabolisme , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Protéine BRCA1/antagonistes et inhibiteurs , Protéine BRCA1/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Humains , Structure moléculaire , Poly (ADP-Ribose) polymerase-1/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Relation structure-activité
20.
Bioorg Med Chem ; 52: 116511, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34801828

RÉSUMÉ

The scaffold of TIQ-A, a previously known inhibitor of human poly-ADP-ribosyltransferase PARP1, was utilized to develop inhibitors against human mono-ADP-ribosyltransferases through structure-guided design and activity profiling. By supplementing the TIQ-A scaffold with small structural changes, based on a PARP10 inhibitor OUL35, selectivity changed from poly-ADP-ribosyltransferases towards mono-ADP-ribosyltransferases. Binding modes of analogs were experimentally verified by determining complex crystal structures with mono-ADP-ribosyltransferase PARP15 and with poly-ADP-ribosyltransferase TNKS2. The best analogs of the study achieved 10-20-fold selectivity towards mono-ADP-ribosyltransferases PARP10 and PARP15 while maintaining micromolar potencies. The work demonstrates a route to differentiate compound selectivity between mono- and poly-ribosyltransferases of the human ARTD family.


Sujet(s)
ADP ribose transferases/antagonistes et inhibiteurs , Isoquinoléines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Protéines proto-oncogènes/antagonistes et inhibiteurs , Thiophènes/pharmacologie , ADP ribose transferases/métabolisme , Cristallographie aux rayons X , Relation dose-effet des médicaments , Humains , Isoquinoléines/synthèse chimique , Isoquinoléines/composition chimique , Modèles moléculaires , Structure moléculaire , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Poly(ADP-ribose) polymerases/métabolisme , Protéines proto-oncogènes/métabolisme , Relation structure-activité , Thiophènes/synthèse chimique , Thiophènes/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE