Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 198
Filtrer
1.
Oncol Res ; 32(6): 1021-1030, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827321

RÉSUMÉ

Background: Apolipoprotein B mRNA editing catalytic polypeptide (APOBEC), an endogenous mutator, induces DNA damage and activates the ataxia telangiectasia and Rad3-related (ATR)-checkpoint kinase 1 (Chk1) pathway. Although cisplatin-based therapy is the mainstay for muscle-invasive bladder cancer (MIBC), it has a poor survival rate. Therefore, this study aimed to evaluate the efficacy of an ATR inhibitor combined with cisplatin in the treatment of APOBEC catalytic subunit 3B (APOBEC3B) expressing MIBC. Methods: Immunohistochemical staining was performed to analyze an association between APOBEC3B and ATR in patients with MIBC. The APOBEC3B expression in MIBC cell lines was assessed using real-time polymerase chain reaction and western blot analysis. Western blot analysis was performed to confirm differences in phosphorylated Chk1 (pChk1) expression according to the APOBEC3B expression. Cell viability and apoptosis analyses were performed to examine the anti-tumor activity of ATR inhibitors combined with cisplatin. Conclusion: There was a significant association between APOBEC3B and ATR expression in the tumor tissues obtained from patients with MIBC. Cells with higher APOBEC3B expression showed higher pChk1 expression than cells expressing low APOBEC3B levels. Combination treatment of ATR inhibitor and cisplatin inhibited cell growth in MIBC cells with a higher APOBEC3B expression. Compared to cisplatin single treatment, combination treatment induced more apoptotic cell death in the cells with higher APOBEC3B expression. Conclusion: Our study shows that APOBEC3B's higher expression status can enhance the sensitivity of MIBC to cisplatin upon ATR inhibition. This result provides new insight into appropriate patient selection for the effective application of ATR inhibitors in MIBC.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Cisplatine , Cytidine deaminase , Antigènes mineurs d'histocompatibilité , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Cytidine deaminase/génétique , Cytidine deaminase/métabolisme , Lignée cellulaire tumorale , Mâle , Antigènes mineurs d'histocompatibilité/métabolisme , Antigènes mineurs d'histocompatibilité/génétique , Adulte d'âge moyen , Femelle , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/génétique , Apoptose , Sujet âgé , Invasion tumorale , Prolifération cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques
2.
Bioorg Chem ; 149: 107471, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38823311

RÉSUMÉ

Applying various drug design strategies including ring variation, substituents variation, and ring fusion, two series of 2-(alkylthio)-5-(arylidene/heteroarylidene)imidazolones and imidazo[1,2-a]thieno[2,3-d]pyrimidines were designed and prepared as dual potential Chk1 and Chk2 inhibitors. The newly synthesized hybrids were screened in NCI 60 cell line panel where the most active derivatives 4b, d-f, and 6a were further estimated for their five dose antiproliferative activity against the most sensitive tumor cells including breast MCF-7 and MDA-MB-468 and non-small cell lung cancer EKVX as well as normal WI-38 cell. Noticeably, increasing the carbon chain attached to thiol moiety at C-2 of imidazolone scaffold elevated the cytotoxic activity. Hence, compounds 4e and 4f, containing S-butyl fragment, exhibited the most antiproliferative activity against the tested cells where 4f showed extremely potent selectivity toward them. As well, compound 6a, containing imidazothienopyrimidine core, exerted significant cytotoxic activity and selectivity toward the examined cells. The mechanistic investigation of the most active cytotoxic analogs was achieved through the evaluation of their inhibitory activity against Chk1 and Chk2. Results revealed that 4f displayed potent dual inhibition of both Chk1 and Chk2 with IC50 equal 0.137 and 0.25 µM, respectively. It also promoted its antiproliferative and Chk suppression activity via EKVX cell cycle arrest at S phase through stimulating the apoptotic approach. The apoptosis induction was also emphasized by elevating the expression of Caspase-3 and Bax, that are accompanied by Bcl-2 diminution. The in silico molecular docking and ADMET profiles of the most active analogs have been carried out to evaluate their potential as significant anticancer drug candidates.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Checkpoint kinase 1 , Checkpoint kinase 2 , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Imidazoles , Inhibiteurs de protéines kinases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/métabolisme , Checkpoint kinase 2/antagonistes et inhibiteurs , Checkpoint kinase 2/métabolisme , Imidazoles/pharmacologie , Imidazoles/composition chimique , Imidazoles/synthèse chimique , Simulation de docking moléculaire , Structure moléculaire , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Relation structure-activité , Pyrimidines/synthèse chimique , Pyrimidines/composition chimique , Pyrimidines/pharmacologie
3.
Anticancer Res ; 44(7): 2827-2836, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925844

RÉSUMÉ

BACKGROUND/AIM: A deep ultraviolet (DUV) light-emitting diode (LED) is a device that can irradiate electromagnetic waves from 250 nm to 350 nm. Tousled-like kinase 1 (TLK1) encodes a nuclear serine/threonine kinase, which is thought to influence the effects of DUV irradiation in cancer. The aim of this study was to clarify the interaction of TLK1 with DUV irradiation-induced DNA damage in cancer cells. MATERIALS AND METHODS: Pancreatic cancer cell lines were treated with or without DUV. TLK1 expression and phosphorylation in the two groups were examined. Then, these cancer cell lines were treated with thioridazine (THD), DUV or both. Thereafter, cytomorphology and apoptosis were assessed. Several proteins related to DNA damage, were analyzed in cancer cells treated with DUV and THD. Tumors in a subcutaneous xenograft model were treated with THD, DUV, or both for six weeks. RESULTS: DUV irradiation induced the phosphorylation of TLK1 in pancreatic cancer cell lines. Cytomorphology was significantly changed in pancreatic cancer cells treated with DUV and THD. TLK1 inhibition enhanced DUV irradiation-induced apoptosis in cancer cells. Interestingly, CHK1 and pCHK1 expression was suppressed after TLK1 inhibition. In addition, inhibition of MRE11 led to a decrease in the expression of CHK1 and pCHK1, accompanied by a notable increase in apoptosis. In the subcutaneous xenograft models, the tumor volume in the DUV and THD groups was lower than that in the other groups. CONCLUSION: TLK1 phosphorylation is an important event in DUV irradiation. DUV irradiation combined with TLK1 inhibition has therapeutic potential in pancreatic cancer cells.


Sujet(s)
Apoptose , Checkpoint kinase 1 , Altération de l'ADN , Tumeurs du pancréas , Protein-Serine-Threonine Kinases , Rayons ultraviolets , Tests d'activité antitumorale sur modèle de xénogreffe , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Humains , Animaux , Tumeurs du pancréas/radiothérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Phosphorylation , Altération de l'ADN/effets des radiations , Altération de l'ADN/effets des médicaments et des substances chimiques , Souris , Souris nude
4.
Sci Rep ; 14(1): 11788, 2024 05 23.
Article de Anglais | MEDLINE | ID: mdl-38783016

RÉSUMÉ

Fascaplysin is a red cytotoxic pigment with anticancer properties isolated from the marine sponge Fascaplysinopsis sp. Recently, structure-activity relationship analysis reported by our group suggested that selective cytotoxicity of fascaplysin derivatives towards tumor cells negatively correlates with their ability to intercalate into DNA. To validate this hypothesis, we synthesized 6- and 7-tert-butylfascaplysins which reveal mitigated DNA-intercalating properties. These derivatives were found to be strongly cytotoxic to drug-resistant human prostate cancer cells, albeit did not demonstrate improved selectivity towards cancer cells when compared to fascaplysin. At the same time, kinome analysis suggested an activation of CHK1/ATR axis in cancer cells shortly after the drug exposure. Further experiments revealed induction of replication stress that is eventually converted to the toxic DNA double-strand breaks, resulting in caspase-independent apoptosis-like cell death. Our observations highlight new DNA-targeting effect of some fascaplysin derivatives and indicate more complex structure-activity relationships within the fascaplysin family, suggesting that cytotoxicity and selectivity of these alkaloids are influenced by multiple factors. Furthermore, combination with clinically-approved inhibitors of ATR/CHK1 as well as testing in tumors particularly sensitive to the DNA damage should be considered in further studies.


Sujet(s)
Antinéoplasiques , Checkpoint kinase 1 , Humains , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Indoles/pharmacologie , Indoles/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Relation structure-activité , Mâle , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , ADN/métabolisme , Animaux , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Composés d'ammonium quaternaire , Carbolines , Indolizine
5.
Chem Biol Interact ; 397: 111063, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38795876

RÉSUMÉ

Coptisine (COP) has been shown to exhibit a wide range of anticancer properties, including in hepatocellular carcinoma (HCC). Nevertheless, the precise mechanism of COP in the treatment of HCC remains elusive. This study aims to investigate the potential mechanism of action of COP against HCC. By evaluating the anti-HCC activity of COP in different HCC cells lines and in xenografted nude mice, it was found that COP inhibited HCC in vitro and in vivo. Through RNA-Seq analysis, E2F7 was identified as a potential target of COP against HCC, as well as the cell cycle as a possible pathway. The overexpression of E2F7 and the inhibition of CHK1 demonstrated that COP inhibits the activity of HCC and induces G2/M phase arrest of HCC cells by down-regulating E2F7 and influencing the CHK1/CDC25A pathway. Finally, the promoter fragmentation experiments and chromatin immunoprecipitation revealed that COP down-regulated E2F7 by inhibiting the E2F4/NFYA/NFYB transcription factors. In conclusion, our study demonstrated that COP downregulates E2F7 by affecting key transcription factors, thereby inducing cell cycle arrest and inhibits HCC cell growth. This provides further evidence of the efficacy of COP in the treatment of tumors.


Sujet(s)
Berbérine , Carcinome hépatocellulaire , Régulation négative , Facteur de transcription E2F4 , Facteur de transcription E2F7 , Points de contrôle de la phase G2 du cycle cellulaire , Tumeurs du foie , Souris nude , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Berbérine/pharmacologie , Berbérine/analogues et dérivés , Animaux , Régulation négative/effets des médicaments et des substances chimiques , Souris , Facteur de transcription E2F4/métabolisme , Points de contrôle de la phase G2 du cycle cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription E2F7/métabolisme , Facteur de transcription E2F7/génétique , Lignée cellulaire tumorale , Souris de lignée BALB C , Prolifération cellulaire/effets des médicaments et des substances chimiques , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs
6.
Cancer Lett ; 596: 216993, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38801884

RÉSUMÉ

Ribonucleotide Reductase (RNR) is a rate-limiting enzyme in the production of deoxyribonucleoside triphosphates (dNTPs), which are essential substrates for DNA repair after radiation damage. We explored the radiosensitization property of RNR and investigated a selective RRM2 inhibitor, 3-AP, as a radiosensitizer in the treatment of metastatic pNETs. We investigated the role of RNR subunit, RRM2, in pancreatic neuroendocrine (pNET) cells and responses to radiation in vitro. We also evaluated the selective RRM2 subunit inhibitor, 3-AP, as a radiosensitizer to treat pNET metastases in vivo. Knockdown of RNR subunits demonstrated that RRM1 and RRM2 subunits, but not p53R3, play significant roles in cell proliferation. RRM2 inhibition activated DDR pathways through phosphorylation of ATM and DNA-PK protein kinases but not ATR. RRM2 inhibition also induced Chk1 and Chk2 phosphorylation, resulting in G1/S phase cell cycle arrest. RRM2 inhibition sensitized pNET cells to radiotherapy and induced apoptosis in vitro. In vivo, we utilized pNET subcutaneous and lung metastasis models to examine the rationale for RNR-targeted therapy and 3-AP as a radiosensitizer in treating pNETs. Combination treatment significantly increased apoptosis of BON (human pNET) xenografts and significantly reduced the burden of lung metastases. Together, our results demonstrate that selective RRM2 inhibition induced radiosensitivity of metastatic pNETs both in vitro and in vivo. Therefore, treatment with the selective RRM2 inhibitor, 3-AP, is a promising radiosensitizer in the therapeutic armamentarium for metastatic pNETs.


Sujet(s)
Apoptose , Prolifération cellulaire , Souris nude , Tumeurs du pancréas , Radiotolérance , Radiosensibilisants , Ribonucleoside diphosphate reductase , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/radiothérapie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/enzymologie , Ribonucleoside diphosphate reductase/génétique , Ribonucleoside diphosphate reductase/antagonistes et inhibiteurs , Ribonucleoside diphosphate reductase/métabolisme , Animaux , Lignée cellulaire tumorale , Radiosensibilisants/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Radiotolérance/effets des médicaments et des substances chimiques , Phosphorylation , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/radiothérapie , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/enzymologie , Tumeurs neuroendocrines/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/radiothérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/génétique , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/génétique , Souris , Checkpoint kinase 2/métabolisme , Checkpoint kinase 2/génétique , Checkpoint kinase 2/antagonistes et inhibiteurs , Femelle , Interférence par ARN , DNA-activated protein kinase
7.
Nat Commun ; 15(1): 4667, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38821952

RÉSUMÉ

Checkpoint kinase 1 (CHK1) is critical for cell survival under replication stress (RS). CHK1 inhibitors (CHK1i's) in combination with chemotherapy have shown promising results in preclinical studies but have displayed minimal efficacy with substantial toxicity in clinical trials. To explore combinatorial strategies that can overcome these limitations, we perform an unbiased high-throughput screen in a non-small cell lung cancer (NSCLC) cell line and identify thioredoxin1 (Trx1), a major component of the mammalian antioxidant-system, as a determinant of CHK1i sensitivity. We establish a role for redox recycling of RRM1, the larger subunit of ribonucleotide reductase (RNR), and a depletion of the deoxynucleotide pool in this Trx1-mediated CHK1i sensitivity. Further, the TrxR inhibitor auranofin, an approved anti-rheumatoid arthritis drug, shows a synergistic interaction with CHK1i via interruption of the deoxynucleotide pool. Together, we show a pharmacological combination to treat NSCLC that relies on a redox regulatory link between the Trx system and mammalian RNR activity.


Sujet(s)
Auranofine , Carcinome pulmonaire non à petites cellules , Checkpoint kinase 1 , Tumeurs du poumon , Oxydoréduction , Thiorédoxines , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Humains , Oxydoréduction/effets des médicaments et des substances chimiques , Thiorédoxines/métabolisme , Lignée cellulaire tumorale , Auranofine/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Inhibiteurs de protéines kinases/pharmacologie , Ribonucleoside diphosphate reductase/métabolisme , Ribonucleoside diphosphate reductase/génétique , Ribonucleotide reductases/métabolisme , Ribonucleotide reductases/antagonistes et inhibiteurs , Synergie des médicaments , Animaux
8.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38673980

RÉSUMÉ

Checkpoint kinase 1 (Chk1) is a key mediator of the DNA damage response that regulates cell cycle progression, DNA damage repair, and DNA replication. Small-molecule Chk1 inhibitors sensitize cancer cells to genotoxic agents and have shown preclinical activity as single agents in cancers characterized by high levels of replication stress. However, the underlying genetic determinants of Chk1-inhibitor sensitivity remain unclear. Although treatment options for advanced colorectal cancer are limited, radiotherapy is effective. Here, we report that exposure to a novel amidine derivative, K1586, leads to an initial reduction in the proliferative potential of colorectal cancer cells. Cell cycle analysis revealed that the length of the G2/M phase increased with K1586 exposure as a result of Chk1 instability. Exposure to K1586 enhanced the degradation of Chk1 in a time- and dose-dependent manner, increasing replication stress and sensitizing colorectal cancer cells to radiation. Taken together, the results suggest that a novel amidine derivative may have potential as a radiotherapy-sensitization agent that targets Chk1.


Sujet(s)
Amidines , Checkpoint kinase 1 , Tumeurs colorectales , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/radiothérapie , Amidines/pharmacologie , Lignée cellulaire tumorale , Rayonnement ionisant , Radiosensibilisants/pharmacologie , Réplication de l'ADN/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Cycle cellulaire/effets des médicaments et des substances chimiques
9.
Biol Chem ; 405(6): 395-406, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38452398

RÉSUMÉ

Checkpoint kinase 1 (Chk1) plays an important role in regulation of the cell cycle, DNA damage response and cell death, and represents an attractive target in anticancer therapy. Small-molecule inhibitors of Chk1 have been intensively investigated either as single agents or in combination with various chemotherapeutic drugs and they can enhance the chemosensitivity of numerous tumor types. Here we newly demonstrate that pharmacological inhibition of Chk1 using potent and selective inhibitor SCH900776, currently profiled in phase II clinical trials, significantly enhances cytotoxic effects of the combination of platinum-based drugs (cisplatin or LA-12) and TRAIL (tumor necrosis factor-related apoptosis inducing ligand) in human prostate cancer cells. The specific role of Chk1 in the drug combination-induced cytotoxicity was confirmed by siRNA-mediated silencing of this kinase. Using RNAi-based methods we also showed the importance of Bak-dependent mitochondrial apoptotic pathway in the combined anticancer action of SCH900776, cisplatin and TRAIL. The triple drug combination-induced cytotoxicity was partially enhanced by siRNA-mediated Mcl-1 silencing. Our findings suggest that targeting Chk1 may be used as an efficient strategy for sensitization of prostate cancer cells to killing action of platinum-based chemotherapeutic drugs and TRAIL.


Sujet(s)
Antinéoplasiques , Checkpoint kinase 1 , Cisplatine , Tumeurs de la prostate , Ligand TRAIL , Humains , Checkpoint kinase 1/métabolisme , Checkpoint kinase 1/antagonistes et inhibiteurs , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Ligand TRAIL/pharmacologie , Ligand TRAIL/métabolisme , Antinéoplasiques/pharmacologie , Cisplatine/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Composés organiques du platine/pharmacologie , Tests de criblage d'agents antitumoraux , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
10.
Clin Cancer Res ; 29(24): 5047-5056, 2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-37819936

RÉSUMÉ

PURPOSE: Combining gemcitabine with CHK1 inhibition has shown promise in preclinical models of pancreatic ductal adenocarcinoma (PDAC). Here, we report the findings from a phase I expansion cohort study (NCT02632448) investigating low-dose gemcitabine combined with the CHK1 inhibitor LY2880070 in patients with previously treated advanced PDAC. PATIENTS AND METHODS: Patients with metastatic PDAC were treated with gemcitabine intravenously at 100 mg/m2 on days 1, 8, and 15, and LY2880070 50 mg orally twice daily on days 2-6, 9-13, and 16-20 of each 21-day cycle. Pretreatment tumor biopsies were obtained from each patient for correlative studies and generation of organoid cultures for drug sensitivity testing and biomarker analyses. RESULTS: Eleven patients with PDAC were enrolled in the expansion cohort between August 27, 2020 and July 30, 2021. Four patients (36%) experienced drug-related grade 3 adverse events. No objective radiologic responses were observed, and all patients discontinued the trial by 3.2 months. In contrast to the lack of efficacy observed in patients, organoid cultures derived from biopsies procured from two patients demonstrated strong sensitivity to the gemcitabine/LY2880070 combination and showed treatment-induced upregulation of replication stress and DNA damage biomarkers, including pKAP1, pRPA32, and γH2AX, as well as induction of replication fork instability. CONCLUSIONS: No evidence of clinical activity was observed for combined low-dose gemcitabine and LY2880070 in this treatment-refractory PDAC cohort. However, the gemcitabine/LY2880070 combination showed in vitro efficacy, suggesting that drug sensitivity for this combination in organoid cultures may not predict clinical benefit in patients.


Sujet(s)
Adénocarcinome , Carcinome du canal pancréatique , Checkpoint kinase 1 , Tumeurs du pancréas , Humains , Adénocarcinome/traitement médicamenteux , Adénocarcinome/génétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Checkpoint kinase 1/antagonistes et inhibiteurs , Études de cohortes , Désoxycytidine , , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/usage thérapeutique , Antinéoplasiques/effets indésirables , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
11.
Stem Cell Rev Rep ; 19(8): 2980-2990, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37702917

RÉSUMÉ

Embryonic development is a continuum in vivo. Transcriptional analysis can separate established human embryonic stem cells (hESC) into at least four distinct developmental pluripotent stages, two naïve and two primed, early and late relative to the intact epiblast. In this study we primarily show that exposure of frozen human blastocysts to an inhibitor of checkpoint kinase 1 (CHK1) upon thaw greatly enhances establishment of karyotypically normal late naïve hESC cultures. These late naïve cells are plastic and can be toggled back to early naïve and forward to early primed pluripotent stages. The early primed cells are transcriptionally equivalent to the post inner cell mass intermediate (PICMI) stage seen one day following transfer of human blastocysts into in vitro culture and are stable at an earlier stage than conventional primed hESC.


Sujet(s)
Techniques de culture cellulaire , Checkpoint kinase 1 , Cellules souches embryonnaires humaines , Cellules souches embryonnaires humaines/cytologie , Cellules souches embryonnaires humaines/effets des médicaments et des substances chimiques , Humains , Checkpoint kinase 1/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Blastocyste/cytologie , Cellules souches pluripotentes/cytologie
12.
Clin Cancer Res ; 28(10): 2147-2159, 2022 05 13.
Article de Anglais | MEDLINE | ID: mdl-35302600

RÉSUMÉ

PURPOSE: Uterine leiomyosarcoma is among the most aggressive gynecological malignancies. No effective treatment strategies have been established. This study aimed to identify novel therapeutic targets for uterine leiomyosarcoma based on transcriptome analysis and assess the preclinical efficacy of novel drug candidates. EXPERIMENTAL DESIGN: Transcriptome analysis was performed using fresh-frozen samples of six uterine leiomyosarcomas and three myomas. The Ingenuity Pathway Analysis (IPA) was used to identify potential therapeutic target genes for uterine leiomyosarcoma. Afterward, our results were validated using three independent datasets, including 40 uterine leiomyosarcomas. Then, the inhibitory effects of several selective inhibitors for the candidate genes were examined using SK-UT-1, SK-LMS-1, and SKN cell lines. RESULTS: We identified 512 considerably dysregulated genes in uterine leiomyosarcoma compared with myoma. The IPA revealed that the function of several genes, including CHEK1 and PLK1, were predicted to be activated in uterine leiomyosarcoma. Through an in vitro drug screening, PLK1 or CHEK1 inhibitors (BI-2536 or prexasertib) were found to exert a superior anticancer effect against cell lines at low nanomolar concentrations and induce cell-cycle arrest. In SK-UT-1 tumor-bearing mice, BI-2536 monotherapy remarkably suppressed tumorigenicity. Moreover, the prexasertib and cisplatin combination therapy inhibited tumor proliferation and prolonged the time to tumor progression. CONCLUSIONS: We identified upregulated expressions of PLK1 and CHEK1; their kinase activity was activated in uterine leiomyosarcoma. BI-2536 and prexasertib demonstrated a significant anticancer effect. Therefore, cell-cycle-related kinases may present a promising therapeutic strategy for the treatment of uterine leiomyosarcoma.


Sujet(s)
Protéines du cycle cellulaire , Checkpoint kinase 1 , Léiomyosarcome , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Tumeurs de l'utérus , Animaux , Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/génétique , Cisplatine/usage thérapeutique , Femelle , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Souris , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/métabolisme ,
13.
Oncogene ; 41(14): 2106-2121, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35190641

RÉSUMÉ

Recurrent cytogenetic abnormalities are the main hallmark of multiple myeloma (MM) and patients having 2 or more high-risk prognostic events are associated with extremely poor outcome. 17p13(del) and 1q21(gain) are critical and independent high-risk cytogenetic markers, however, the biological significance underlying the poor outcome in MM patients having co-occurrence of both these chromosomal aberrations has never been interrogated. Herein, we identified that patients harbouring concomitant 17p13(del) with 1q21(gain) demonstrated the worst prognosis as compared to patients with single- (either 17p13(del) or 1q21(gain)) and with no chromosomal events (WT for both chromosomal loci); and they are highly enriched for genomic instability (GI) signature. We discovered that the GI feature in the patients with concomitant 17p13(del)-1q21(gain) was recapitulating the biological properties of myeloma cells with co-existing p53-deficiency and NEIL1 mRNA-hyper-editing (associated with chromosome 17p and 1q, respectively) that have inherent DNA damage response (DDR) and persistent activation of Chk1 pathway. Importantly, this became a vulnerable point for therapeutic targeting whereby the cells with this co-abnormalities demonstrated hyper-sensitivity to siRNA- and pharmacological-mediated-Chk1 inhibition, as observed at both the in vitro and in vivo levels. Mechanistically, this was attributable to the synthetic lethal relationship between p53-NEIL1-Chk1 abnormalities. The Chk1 inhibitor (AZD7762) tested showed good synergism with standard-of-care myeloma drugs, velcade and melphalan, thus further reinforcing the translational potential of this therapeutic approach. In summary, combination of NEIL1-p53 abnormalities with an ensuing Chk1 activation could serve as an Achilles heel and predispose MM cells with co-existing 1q21(gain) and 17p13(del) to therapeutic vulnerability for Chk1 inhibition.


Sujet(s)
Checkpoint kinase 1 , DNA Glycosylases , Myélome multiple , Protéine p53 suppresseur de tumeur , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/génétique , Aberrations des chromosomes , Délétion de segment de chromosome , DNA Glycosylases/génétique , Instabilité du génome , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Mutations synthétiques létales , Protéine p53 suppresseur de tumeur/génétique
14.
Acta Pharmacol Sin ; 43(1): 220-228, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-33782542

RÉSUMÉ

Checkpoint kinase 1 inhibitors (CHK1i) have shown impressive single-agent efficacy in treatment of certain tumors, as monotherapy or potentiators of chemotherapy in clinical trials, but the sensitive tumor types and downstream effectors to dictate the therapeutic responses to CHK1i remains unclear. In this study we first analyzed GDSC (Genomics of Drug Sensitivity in Cancer) and DepMap database and disclosed that hematologic malignancies (HMs) were relatively sensitive to CHK1i or CHK1 knockdown. This notion was confirmed by examining PY34, a new and potent in-house selective CHK1i, which exhibited potent anti-HM effect in vitro and in vivo, as single agent. We demonstrated that the downregulation of c-Myc and its signaling pathway was the common transcriptomic profiling response of sensitive HM cell lines to PY34, whereas overexpressing c-Myc could partially rescue the anticancer effect of PY34. Strikingly, we revealed the significant correlations between downregulation of c-Myc and cell sensitivity to PY34 in 17 HM cell lines and 39 patient-derived cell (PDC) samples. Thus, our results demonstrate that HMs are more sensitive to CHK1i than solid tumors, and c-Myc downregulation could represent the CHK1i efficacy in HMs.


Sujet(s)
Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Régulation négative/effets des médicaments et des substances chimiques , Tumeurs hématologiques/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Facteurs de transcription/antagonistes et inhibiteurs , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/déficit , Checkpoint kinase 1/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Femelle , Tumeurs hématologiques/métabolisme , Tumeurs hématologiques/anatomopathologie , Humains , Souris , Souris de lignée NOD , Souris nude , Souris SCID , Structure moléculaire , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Relation structure-activité , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
15.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119168, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34728235

RÉSUMÉ

The induction of DNA damage together with the interference with DNA repair represents a promising strategy in cancer treatment. Here we show that the PARP-1/2/3 inhibitor AZD2461 in combination with the CHK1 inhibitor UCN-01 altered the DNA damage response and reduced cell proliferation in PEL cells, an aggressive B cell lymphoma highly resistant to chemotherapies. AZD2461/UCN-01 combination activated p53/p21 and downregulated c-Myc in these cells, leading to a reduced expression level of RAD51, molecule involved in DNA repair. The effect of AZD2461/UCN-01 on c-Myc and p53/p21 was inter-dependent and, besides impairing cell proliferation, contributed to the activation of the replicative cycle of KSHV, carried in a latent state in PEL cells. Finally, we found that the pharmacological or genetic inhibition of p21 counteracted the viral lytic cycle activation and further reduced PEL cell proliferation, suggesting that it could induce a double beneficial effect in this setting. This study unveils that, therapeutic approaches, based on the induction of DNA damage and the reduction of DNA repair, could be used to successfully treat this malignant lymphoma.


Sujet(s)
Prolifération cellulaire , Altération de l'ADN , Lymphome primitif des séreuses/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Réplication virale , Lignée cellulaire , Cellules cultivées , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/métabolisme , Herpèsvirus humain de type 8/physiologie , Humains , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Lymphome primitif des séreuses/génétique , Lymphome primitif des séreuses/virologie , Phtalazines/pharmacologie , Pipéridines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/toxicité , Inhibiteurs de protéines kinases/toxicité , Staurosporine/analogues et dérivés , Staurosporine/pharmacologie
16.
Oncogene ; 41(4): 476-488, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34773074

RÉSUMÉ

We recently reported that genetic or pharmacological inhibition of insulin-like growth factor receptor (IGF-1R) slows DNA replication and induces replication stress by downregulating the regulatory subunit RRM2 of ribonucleotide reductase, perturbing deoxynucleotide triphosphate (dNTP) supply. Aiming to exploit this effect in therapy we performed a compound screen in five breast cancer cell lines with IGF neutralising antibody xentuzumab. Inhibitor of checkpoint kinase CHK1 was identified as a top screen hit. Co-inhibition of IGF and CHK1 caused synergistic suppression of cell viability, cell survival and tumour growth in 2D cell culture, 3D spheroid cultures and in vivo. Investigating the mechanism of synthetic lethality, we reveal that CHK1 inhibition in IGF-1R depleted or inhibited cells further downregulated RRM2, reduced dNTP supply and profoundly delayed replication fork progression. These effects resulted in significant accumulation of unreplicated single-stranded DNA and increased cell death, indicative of replication catastrophe. Similar phenotypes were induced by IGF:WEE1 co-inhibition, also via exacerbation of RRM2 downregulation. Exogenous RRM2 expression rescued hallmarks of replication stress induced by co-inhibiting IGF with CHK1 or WEE1, identifying RRM2 as a critical target of the functional IGF:CHK1 and IGF:WEE1 interactions. These data identify novel therapeutic vulnerabilities and may inform future trials of IGF inhibitory drugs.


Sujet(s)
Checkpoint kinase 1/antagonistes et inhibiteurs , Tests de criblage à haut débit/méthodes , Récepteur IGF de type 1/métabolisme , Lignée cellulaire tumorale , Humains , Transfection
17.
Cancer Sci ; 113(2): 587-596, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-34807483

RÉSUMÉ

Checkpoint kinase 1 (CHK1) plays a key role in genome surveillance and integrity throughout the cell cycle. Selective inhibitors of CHK1 (CHK1i) are undergoing clinical evaluation for various human malignancies, including neuroblastoma. In this study, one CHK1i-sensitive neuroblastoma cell line, CHP134, was investigated, which characteristically carries MYCN amplification and a chromosome deletion within the 10q region. Among several cancer-related genes in the chromosome 10q region, mRNA expression of fibroblast growth factor receptor 2 (FGFR2) was altered in CHP134 cells and associated with an unfavorable prognosis of patients with neuroblastoma. Induced expression of FGFR2 in CHP134 cells reactivated downstream MEK/ERK signaling and resulted in cells resistant to CHK1i-mediated cell growth inhibition. Consistently, the MEK1/2 inhibitor, trametinib, potentiated CHK1 inhibitor-mediated cell death in these cells. These results suggested that FGFR2 loss might be prone to highly effective CHK1i treatment. In conclusion, extreme cellular dependency of ERK activation may imply a possible application for the MEK1/2 inhibitor, either as a single inhibitor or in combination with CHK1i in MYCN-amplified neuroblastomas.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Checkpoint kinase 1/antagonistes et inhibiteurs , Protéine du proto-oncogène N-Myc/génétique , Inhibiteurs de protéines kinases/pharmacologie , Récepteur FGFR2/génétique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Synergie des médicaments , Amplification de gène , Humains , MAP Kinase Kinase 1/antagonistes et inhibiteurs , MAP Kinase Kinase 2/antagonistes et inhibiteurs , Système de signalisation des MAP kinases , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Pronostic , Pyridones/pharmacologie , Pyrimidinones/pharmacologie , ARN messager/génétique
18.
Cell Rep ; 37(9): 110060, 2021 11 30.
Article de Anglais | MEDLINE | ID: mdl-34852220

RÉSUMÉ

We apply genetic screens to delineate modulators of KRAS mutant pancreatic ductal adenocarcinoma (PDAC) sensitivity to ERK inhibitor treatment, and we identify components of the ATR-CHK1 DNA damage repair (DDR) pathway. Pharmacologic inhibition of CHK1 alone causes apoptotic growth suppression of both PDAC cell lines and organoids, which correlates with loss of MYC expression. CHK1 inhibition also activates ERK and AMPK and increases autophagy, providing a mechanistic basis for increased efficacy of concurrent CHK1 and ERK inhibition and/or autophagy inhibition with chloroquine. To assess how CHK1 inhibition-induced ERK activation promotes PDAC survival, we perform a CRISPR-Cas9 loss-of-function screen targeting direct/indirect ERK substrates and identify RIF1. A key component of non-homologous end joining repair, RIF1 suppression sensitizes PDAC cells to CHK1 inhibition-mediated apoptotic growth suppression. Furthermore, ERK inhibition alone decreases RIF1 expression and phenocopies RIF1 depletion. We conclude that concurrent DDR suppression enhances the efficacy of ERK and/or autophagy inhibitors in KRAS mutant PDAC.


Sujet(s)
Carcinome du canal pancréatique/traitement médicamenteux , Checkpoint kinase 1/antagonistes et inhibiteurs , Altération de l'ADN , Mutation , Tumeurs du pancréas/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes p21(ras)/génétique , Animaux , Apoptose , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Prolifération cellulaire , Checkpoint kinase 1/génétique , Checkpoint kinase 1/métabolisme , Humains , Souris , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Biochem Biophys Res Commun ; 584: 7-14, 2021 12 20.
Article de Anglais | MEDLINE | ID: mdl-34753066

RÉSUMÉ

Patients with triple-negative breast cancer have a poor prognosis as only a few efficient targeted therapies are available. Cancer cells are characterized by their unregulated proliferation and require large amounts of nucleotides to replicate their DNA. One-carbon metabolism contributes to purine and pyrimidine nucleotide synthesis by supplying one carbon atom. Although mitochondrial one-carbon metabolism has recently been focused on as an important target for cancer treatment, few specific inhibitors have been reported. In this study, we aimed to examine the effects of DS18561882 (DS18), a novel, orally active, specific inhibitor of methylenetetrahydrofolate dehydrogenase (MTHFD2), a mitochondrial enzyme involved in one-carbon metabolism. Treatment with DS18 led to a marked reduction in cancer-cell proliferation; however, it did not induce cell death. Combinatorial treatment with DS18 and inhibitors of checkpoint kinase 1 (Chk1), an activator of the S phase checkpoint pathway, efficiently induced apoptotic cell death in breast cancer cells and suppressed tumorigenesis in a triple-negative breast cancer patient-derived xenograft model. Mechanistically, MTHFD2 inhibition led to cell cycle arrest and slowed nucleotide synthesis. This finding suggests that DNA replication stress occurs due to nucleotide shortage and that the S-phase checkpoint pathway is activated, leading to cell-cycle arrest. Combinatorial treatment with both inhibitors released cell-cycle arrest, but induced accumulation of DNA double-strand breaks, leading to apoptotic cell death. Collectively, a combination of MTHFD2 and Chk1 inhibitors would be a rational treatment option for patients with triple-negative breast cancer.


Sujet(s)
Aminohydrolases/antagonistes et inhibiteurs , Checkpoint kinase 1/antagonistes et inhibiteurs , Antienzymes/usage thérapeutique , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonistes et inhibiteurs , Enzymes multifonctionnelles/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Administration par voie orale , Aminohydrolases/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Checkpoint kinase 1/métabolisme , Association de médicaments , Antienzymes/administration et posologie , Femelle , Humains , Methylenetetrahydrofolate Dehydrogenase (NADP)/métabolisme , Souris de lignée NOD , Souris knockout , Souris SCID , Enzymes multifonctionnelles/métabolisme , Points de contrôle de la phase S du cycle cellulaire/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/enzymologie , Tumeurs du sein triple-négatives/anatomopathologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
20.
J Med Chem ; 64(20): 15069-15090, 2021 10 28.
Article de Anglais | MEDLINE | ID: mdl-34665631

RÉSUMÉ

Checkpoint kinase 1 (CHK1) plays an important role in the DNA damage response pathway, being a potential anti-cancer drug target. In this study, we used a strategy for trifluoromethyl substitution to obtain orally bioavailable CHK1 inhibitors to overcome the limitations of lead compound 1, which can only be administered intravenously. After detailed investigation, we identified compound 6c as an oral CHK1 inhibitor, which demonstrated a considerably higher plasma exposure in mice. Compound 6c also showed good kinase selectivity. Moreover, it exhibited a significant antiproliferative effect in MV-4-11 cells singly and a synergistic effect in combination with gemcitabine in HT-29, A549, and RPMI-8226 cells. Additionally, compound 6c could inhibit tumor growth in the MV-4-11 xenograft mouse model. The combination of 6c and gemcitabine exhibited synergistic effect in the HT-29 xenograft mouse model. Thus, compound 6c was found to be a selective and oral potential anticancer CHK1 inhibitor.


Sujet(s)
Antinéoplasiques/pharmacologie , Checkpoint kinase 1/antagonistes et inhibiteurs , Développement de médicament , Inhibiteurs de protéines kinases/pharmacologie , Administration par voie orale , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Biodisponibilité , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Checkpoint kinase 1/métabolisme , Relation dose-effet des médicaments , Humains , Souris , Souris nude , Modèles moléculaires , Structure moléculaire , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/composition chimique , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...