Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.674
Filtrer
1.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38955419

RÉSUMÉ

BACKGROUND: Patients with mismatch repair-deficient (MMRd) endometrial cancer (EC) can derive great benefit from immune checkpoint inhibitors (ICI). However not all responses and predictors of primary resistance are lacking. METHODS: We compared the immune tumor microenvironment of MMRd EC ICI-responders (Rs) and ICI non-responders (NRs), using spatial multiplexed immune profiling and unsupervised hierarchical clustering analysis. RESULTS: Overall, NRs exhibited drastically lower CD8+, absent terminally differentiated T cells, lack of mature tertiary lymphoid structures and dendritic cells, as well as loss of human leukocyte antigen class I. However, no single marker could predict R versus NR with confidence. Clustering analysis identified a combination of four immune features that demonstrated that accurately predicted ICI response, with a discriminative power of 92%. Finally, 80% of NRs lacked programmed death-ligand 1, however, 60% exhibited another actionable immune checkpoint (T-cell immunoglobulin and mucin containing protein-3, indoleamine 2,3-dioxygenase 1, or lymphocyte activation gene 3). CONCLUSIONS: These findings underscore the potential of immune tumor microenvironment features for identifying patients with MMRd EC and primary resistance to ICI who should be oriented towards trials testing novel immunotherapeutic combinations.


Sujet(s)
Réparation de mésappariement de l'ADN , Tumeurs de l'endomètre , Inhibiteurs de points de contrôle immunitaires , Humains , Femelle , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/immunologie , Tumeurs de l'endomètre/génétique , Microenvironnement tumoral , Adulte d'âge moyen , Sujet âgé
2.
Curr Med Sci ; 44(3): 623-632, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38853192

RÉSUMÉ

OBJECTIVE: Endometrial carcinoma (EC) is a prevalent gynecological malignancy characterized by increasing incidence and mortality rates. This underscores the critical need for novel therapeutic targets. One such potential target is cell division cycle 20 (CDC20), which has been implicated in oncogenesis. This study investigated the effect of the CDC20 inhibitor Apcin on EC and elucidated the underlying mechanism involved. METHODS: The effects of Apcin on EC cell proliferation, apoptosis, and the cell cycle were evaluated using CCK8 assays and flow cytometry. RNA sequencing (RNA-seq) was subsequently conducted to explore the underlying molecular mechanism, and Western blotting and coimmunoprecipitation were subsequently performed to validate the results. Animal studies were performed to evaluate the antitumor effects in vivo. Bioinformatics analysis was also conducted to identify CDC20 as a potential therapeutic target in EC. RESULTS: Treatment with Apcin inhibited proliferation and induced apoptosis in EC cells, resulting in cell cycle arrest. Pathways associated with apoptosis and the cell cycle were activated following treatment with Apcin. Notably, Apcin treatment led to the upregulation of the cell cycle regulator p21, which was verified to interact with CDC20 and consequently decrease the expression of downstream cyclins in EC cells. In vivo experiments confirmed that Apcin treatment significantly impeded tumor growth. Higher CDC20 expression was observed in EC tissue than in nonmalignant tissue, and increased CDC20 expression in EC patients was associated with shorter overall survival and progress free interval. CONCLUSION: CDC20 is a novel molecular target in EC, and Apcin could be developed as a candidate antitumor drug for EC treatment.


Sujet(s)
Apoptose , Protéines Cdc20 , Points de contrôle du cycle cellulaire , Prolifération cellulaire , Inhibiteur p21 de kinase cycline-dépendante , Tumeurs de l'endomètre , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/métabolisme , Humains , Apoptose/effets des médicaments et des substances chimiques , Protéines Cdc20/génétique , Protéines Cdc20/métabolisme , Animaux , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Souris nude
3.
World J Surg Oncol ; 22(1): 169, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918837

RÉSUMÉ

OBJECTIVE: This study aimed to evaluate the oncological and reproductive outcomes of fertility-preserving re-treatment in progestin-resistant endometrial carcinoma (EC) and atypical endometrial hyperplasia (AEH) women who desire to maintain their fertility. METHODS: Our study included 61 progestin-resistant EC/AEH patients. These patients underwent treatment with gonadotropin-releasing hormone agonist (GnRHa) solely or a combination of GnRHa with levonorgestrel-releasing intrauterine system (LNG-IUD) or aromatase inhibitor (AI). Histological evaluations were performed every 3-4 months. Upon achieving complete remission (CR), we recommended maintenance treatments including LNG-IUD, cyclical oral contraceptives, or low-dose cyclic progestin until they began attempting conception. Regular follow-up was conducted for all patients. The chi-square method was utilized to compare oncological and fertility outcomes, while the Cox proportional hazards regression analysis helped identify risk factors for CR, recurrence, and pregnancy. RESULTS: Overall, 55 (90.2%) patients achieved CR, including 90.9% of AEH patients and 89.7% of EC patients. The median re-treatment time was 6 months (ranging from 3 to 12 months). The CR rate for GnRHa alone, GnRHa + LNG-IUD and GnRHa + AI were 80.0%, 91.7% and 93.3%, respectively. After a median follow-up period of 36 months (ranging from 3 to 96 months), 19 women (34.5%) experienced recurrence, 40.0% in AEH and 31.4% in EC patients, with the median recurrence time of 23 months (ranging from 6 to 77 months). Among the patients who achieved CR, 39 expressed a desire to conceive, 20 (51.3%) became pregnant, 11 (28.2%) had successfully deliveries, 1 (5.1%) was still pregnant, while 8 (20.5%) suffered miscarriages. CONCLUSION: GnRHa-based fertility-sparing treatment exhibited promising oncological and reproductive outcomes for progestin-resistant patients. Future larger multi-institutional studies are necessary to confirm these findings.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Hyperplasie endométriale , Tumeurs de l'endomètre , Préservation de la fertilité , Progestines , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Adulte , Études rétrospectives , Préservation de la fertilité/méthodes , Hyperplasie endométriale/traitement médicamenteux , Hyperplasie endométriale/anatomopathologie , Progestines/administration et posologie , Progestines/usage thérapeutique , Études de suivi , Grossesse , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Hormone de libération des gonadotrophines/agonistes , Lévonorgestrel/administration et posologie , Adulte d'âge moyen , Pronostic , Dispositifs intra-uterins libérant un agent contraceptif , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Taux de grossesse , Inhibiteurs de l'aromatase/usage thérapeutique , Inhibiteurs de l'aromatase/administration et posologie , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/administration et posologie
4.
Radiat Oncol ; 19(1): 76, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890652

RÉSUMÉ

OBJECTIVE: This retrospective study aimed to investigate the factors influencing the occurrence of neutropenia in patients with endometrial cancer (EC) following adjuvant chemoradiotherapy (CRT). METHODS: Retrospective analysis of EC patients who underwent adjuvant CRT from January 2012 to June 2023 in the Department of Gynecology and Oncology of the First Affiliated Hospital of Shandong First Medical University. Neutropenia was defined as an Absolute Neutrophil Count (ANC) of peripheral blood neutrophils below 2 × 109/L. Factors affecting neutropenia in EC patients treated with CRT using Generalized Estimating Equation (GEE), and Logistic regression was used to further analyze the effect of adding radiotherapy to different chemotherapy cycles on neutropenia, so that patients receive optimal adjuvant CRT while the risk of neutropenia is appropriately controlled. RESULTS: A total of 144 patients met the inclusion criteria. They underwent 330 cycles of adjuvant chemotherapy, of whom 96 (66.7%) developed neutropenia, which occurred 140 times. The results of one-way GEE analysis showed that before CRT, White Blood Cell (WBC) (OR = 0.827; 95%CI, 0.701-0.976), ANC (OR = 0.749; 95%CI, 0.586-0.957), Absolute Monocyte Count (AMC) (OR = 0.047; 95%CI, 0.008-0.283), Blood Urea Nitrogen (BUN) (OR = 0.857; 95%CI, 0.741-0.991), platinum and docetaxel (platinum/docetaxel) dosing regimen (OR = 2.284; 95%CI, 1.130-4.618) were associated with neutropenia with adjuvant CRT for EC (p < 0.05), results of multifactorial GEE analysis showed that before adjuvant CRT ANC (OR = 0.552; 95%CI, 0.973-2.231), AMC (OR = 0.047; 95%CI, 0.004-0.052), platinum/docetaxel (OR = 2.437; 95%CI, 1.087-5.464) were an independent influence on neutropenia in adjuvant CRT for EC (p < 0.05). Multifactorial Logistic regression shows addition of radiotherapy to the first cycle of chemotherapy (OR = 4.413; 95%CI, 1.238-18.891) was an independent influence of neutropenia (p < 0.05). CONCLUSIONS: Patients with low pre-CRT ANC and AMC, platinum/docetaxel dosing regimens need to be closely monitored during each cycle of CRT. Also, the concurrent addition of radiotherapy should be avoided during the first cycle of chemotherapy.


Sujet(s)
Chimioradiothérapie adjuvante , Tumeurs de l'endomètre , Neutropénie , Humains , Femelle , Études rétrospectives , Tumeurs de l'endomètre/thérapie , Tumeurs de l'endomètre/traitement médicamenteux , Neutropénie/étiologie , Adulte d'âge moyen , Sujet âgé , Chimioradiothérapie adjuvante/effets indésirables , Adulte , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pronostic , Docetaxel/administration et posologie , Docetaxel/effets indésirables , Facteurs de risque
5.
BMC Complement Med Ther ; 24(1): 227, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862934

RÉSUMÉ

OBJECTIVE: Endometrial cancer (EC) is an oestrogen-dependent tumour, the occurrence of which is closely related to an imbalance of oestrogen homeostasis. Our previous studies explored the effects of Resveratrol(Res) on oestrogen metabolism. However, systematic research on the exact mechanism of action of Res is still lacking. Based on network pharmacology, molecular docking and animal experiments, the effects and molecular mechanisms of Res on endometrial cancer were investigated. METHODS: The target of Res was obtained from the high-throughput experiment and reference-guided database of TCM (HERB) and the Encyclopedia of Traditional Chinese Medicine (ETCM) databases, and the target of endometrial cancer was obtained by using the Genecards database. Venny map was used to obtain the intersection target of Res in the treatment of endometrial cancer, and the protein interaction network of the intersection target was constructed by importing the data into the STRING database. Then, the drug-disease-target interaction network was constructed based on Cytoscape 3.9.1 software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for intersection targets using the OmicShare cloud platform. Res and core targets were analysed by molecular docking. EC model mice induced by MNNG were randomly divided into the control group, Res group, MNNG group, MNNG + Res group, and MNNG + Res + MAPK/ERKi group. The protein levels of ERK and p-ERK in the mouse uterus were detected by Western blot. The levels of E1, E2, E3, 16-epiE3, 17-epiE3, 2-MeOE1, 4-MeOE1, 2-MeOE2, 4-MeOE2, 3-MeOE1, 2-OHE1, 4-OHE1, 2-OHE2, 4-OHE2, and 16α-OHE1 in the serum and endometrial tissue of mice were measured by LC‒MS/MS. RESULTS: A total of 174 intersection targets of Res anti-endometrial cancer were obtained. The signalling pathways analysed by KEGG enrichment included the AGE-RAGE signalling pathway in diabetic complications, the PI3K-Akt signalling pathway and the MAPK signalling pathway. The top 10 core targets were MAPK3, JUN, TP53, CASP3, TNF, IL1B, AKT1, FOS, VEGFA and INS. Molecular docking showed that in addition to TNF, other targets had good affinity for Res, and the binding activity with MAPK3 was stable. Western blot results showed that Res increased the phosphorylation level of ERK and that MAPK/ERKi decreased ERK activation. In the LC-MS/MS analysis, the levels of 2-MeOE1, 2-MeOE2 and 4-MeOE1 in serum and uterine tissue showed a significantly decreasing trend in the MNNG group, while that of 4-OHE2 was increased (P < 0.05). The concentrations of 4-MeOE1 in serum and 2-MeOE1 and 2-MeOE2 in the endometrial tissue of mice were significantly increased after Res treatment, and those of 4-OHE2 in the serum and uterus of mice were significantly decreased (P < 0.05). Meanwhile, in the MAPK/ERKi intervention group, the effect of Res on the reversal of oestrogen homeostasis imbalance was obviously weakened. CONCLUSION: Res has multiple targets and multiple approaches in the treatment of endometrial cancer. In this study, it was found that Res regulates oestrogen metabolism by activating the MAPK/ERK pathway. This finding provides a new perspective for subsequent research on the treatment of endometrial cancer.


Sujet(s)
Tumeurs de l'endomètre , Oestrogènes , Système de signalisation des MAP kinases , Simulation de docking moléculaire , Resvératrol , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/métabolisme , Animaux , Resvératrol/pharmacologie , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Oestrogènes/métabolisme , Oestrogènes/pharmacologie , Humains , Souris de lignée BALB C , Pharmacologie des réseaux , Cartes d'interactions protéiques
8.
Gynecol Oncol ; 185: 202-211, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38834399

RÉSUMÉ

OBJECTIVE: To report long-term efficacy and safety of selinexor maintenance therapy in adults with TP53 wild-type (TP53wt) stage IV or recurrent endometrial cancer (EC) who achieved partial remission (PR) or complete remission (CR) following chemotherapy. METHODS: Analysis of the prespecified, exploratory subgroup of patients with TP53wt EC from the phase 3 SIENDO study was performed. Progression-free survival (PFS) benefit in patients with TP53wt EC and across other patient subgroups were exploratory endpoints. Safety and tolerability were also assessed. RESULTS: Of the 263 patients enrolled in the SIENDO trial, 113 patients had TP53wt EC; 70/113 (61.9%) had TP53wt/proficient mismatch repair (pMMR) EC, and 29/113 (25.7%) had TP53wt/deficient mismatch repair (dMMR) EC. As of April 1, 2024, the median PFS (mPFS) for TP53wt patients who received selinexor compared with placebo was 28.4 versus 5.2 months (36.8-month follow-up, HR 0.44; 95% CI 0.27-0.73). A benefit in mPFS was seen with selinexor versus placebo regardless of MMR status (patients with TP53wt/pMMR EC: 39.5 vs 4.9 months, HR 0.36; 95% CI 0.19-0.71; patients with TP53wt/dMMR EC: 13.1 vs 3.7 months, HR 0.49; 95% CI 0.18-1.34). Selinexor treatment was generally manageable, with no new safety signals identified. CONCLUSION: In the phase 3 SIENDO study, selinexor maintenance therapy showed a promising efficacy signal and a manageable safety profile in the prespecified subgroup of patients with TP53wt EC who achieved a PR or CR following chemotherapy. These results are being further evaluated in an ongoing randomized phase 3 trial (NCT05611931).


Sujet(s)
Tumeurs de l'endomètre , Hydrazines , Récidive tumorale locale , Triazoles , Protéine p53 suppresseur de tumeur , Humains , Femelle , Triazoles/administration et posologie , Triazoles/effets indésirables , Triazoles/usage thérapeutique , Adulte d'âge moyen , Hydrazines/effets indésirables , Hydrazines/administration et posologie , Hydrazines/usage thérapeutique , Sujet âgé , Protéine p53 suppresseur de tumeur/génétique , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Récidive tumorale locale/traitement médicamenteux , Adulte , Études de suivi , Survie sans progression , Sujet âgé de 80 ans ou plus , Chimiothérapie de maintenance/méthodes , Stadification tumorale
9.
Gynecol Oncol ; 186: 191-198, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38824752

RÉSUMÉ

OBJECTIVE: Retifanlimab is a humanized immunoglobulin G4 monoclonal antibody against programmed death 1 being investigated in several solid tumor types. We report final results from patients with recurrent microsatellite instability-high (MSI-H)/mismatch repair deficient (dMMR) endometrial cancer treated with retifanlimab in a POD1UM-101 expansion cohort. METHODS: Eligible patients (≥18 years; histologically proven/unresectable/recurrent, MSI-H/dMMR endometrial cancer; checkpoint inhibitor naive) received retifanlimab 500 mg intravenously every 4 weeks for ≤2 years. Primary endpoint was safety/tolerability. RESULTS: At data cutoff (May 17, 2023), 76 patients had received at least one retifanlimab dose. Median (range) age was 67 (49-88) years; 88.2% of patients had recurrent metastatic disease and 80.3% had visceral metastases. Seventy-five patients (98.7%) had received at least one prior systemic therapy. Median retifanlimab exposure was 10.0 (0.03-25.9) months; 23 patients completed treatment. 38 patients (50.0%) had grade ≥3 treatment-emergent adverse events (TEAEs), most commonly anemia (n = 10 [13.2%]). 63 patients (82.9%) had treatment-related AEs (TRAEs; grade ≥3, n = 14 [18.4%]); most common was fatigue (n = 14 [18.4%]). Two patients had TEAEs that led to death; no TRAEs were fatal. 39 patients had objective responses (51.3%; 95% CI, 39.6-63.0%); 19 patients (25.0%) had complete response and 20 (26.3%) had partial response. Median progression-free survival was 12.2 months; 30 patients (76.9%) had duration of response (DOR) ≥12 months. Median DOR was not reached after median follow-up time of 26.0 months. CONCLUSIONS: Retifanlimab was generally well tolerated and demonstrated encouraging anti-tumor activity in patients with pre-treated recurrent MSI-H/dMMR endometrial cancer.


Sujet(s)
Réparation de mésappariement de l'ADN , Tumeurs de l'endomètre , Instabilité des microsatellites , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Sujet âgé , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/administration et posologie , Survie sans progression , Études de cohortes , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
10.
Sultan Qaboos Univ Med J ; 24(2): 293-297, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38828252

RÉSUMÉ

Endometrial cancer (EC) is the most common gynaecological cancer. The combination of lenvatinib and pembrolizumab has exhibited efficacy as the second line treatment for advanced EC, with a significant benefit in terms of progression free survival (PFS) and overall survival, but the adverse events (AE) profile is complex. AEs associated with the treatment may represent a limitation to this combination. Here, we report the case of a 38-year-old female patient diagnosed with stage IV EC elsewhere, whose disease progressed after the first line of treatment and was referred to a specialised cacncer centre in Muscat, Oman, in 2021. We treated her with the combination of lenvatinib and pembrolizumab. During the course of the treatment, she developed hand-foot syndrome grade III and hypothyroidism grade II. The AEs were managed with supportive medications, dose interruptions, dose reductions and multidisciplinary care, which allowed the continuation of the treatment. The patient achieved a good partial response and an ongoing PFS of more than 12 months.


Sujet(s)
Anticorps monoclonaux humanisés , Tumeurs de l'endomètre , Phénylurées , Quinoléines , Humains , Femelle , Adulte , Tumeurs de l'endomètre/traitement médicamenteux , Quinoléines/effets indésirables , Quinoléines/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Phénylurées/usage thérapeutique , Phénylurées/effets indésirables , Oman , Antinéoplasiques immunologiques/effets indésirables , Antinéoplasiques immunologiques/usage thérapeutique , Hypothyroïdie/induit chimiquement , Syndrome mains-pieds/étiologie
11.
Biomolecules ; 14(5)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38786008

RÉSUMÉ

Epidemiological and clinical evidence have extensively documented the role of obesity in the development of endometrial cancer. However, the effect of fatty acids on cell growth in endometrial cancer has not been widely studied. Here, we reported that palmitic acid significantly inhibited cell proliferation of endometrial cancer cells and primary cultures of endometrial cancer and reduced tumor growth in a transgenic mouse model of endometrial cancer, in parallel with increased cellular stress and apoptosis and decreased cellular adhesion and invasion. Inhibition of cellular stress by N-acetyl-L-cysteine effectively reversed the effects of palmitic acid on cell proliferation, apoptosis, and invasive capacity in endometrial cancer cells. Palmitic acid increased the intracellular formation of lipid droplets in a time- and dose-dependent manner. Depletion of lipid droplets by blocking DGAT1 and DGAT2 effectively increased the ability of palmitic acid to inhibit cell proliferation and induce cleaved caspase 3 activity. Collectively, this study provides new insight into the effect of palmitic acid on cell proliferation and invasion and the formation of lipid droplets that may have potential clinical relevance in the treatment of obesity-driven endometrial cancer.


Sujet(s)
Apoptose , Prolifération cellulaire , Tumeurs de l'endomètre , Gouttelettes lipidiques , Acide palmitique , Femelle , Acide palmitique/pharmacologie , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Humains , Gouttelettes lipidiques/métabolisme , Gouttelettes lipidiques/effets des médicaments et des substances chimiques , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Diacylglycerol O-acyltransferase/métabolisme , Souris transgéniques
12.
Cell Mol Biol Lett ; 29(1): 63, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38698330

RÉSUMÉ

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological cancers. Herein, we aimed to define the role of specific myosin family members in EC because this protein family is involved in the progression of various cancers. METHODS: Bioinformatics analyses were performed to reveal EC patients' prognosis-associated genes in patients with EC. Furthermore, colony formation, immunofluorescence, cell counting kit 8, wound healing, and transwell assays as well as coimmunoprecipitation, cycloheximide chase, luciferase reporter, and cellular thermal shift assays were performed to functionally and mechanistically analyze human EC samples, cell lines, and a mouse model, respectively. RESULTS: Machine learning techniques identified MYH14, a member of the myosin family, as the prognosis-associated gene in patients with EC. Furthermore, bioinformatics analyses based on public databases showed that MYH14 was associated with EC chemoresistance. Moreover, immunohistochemistry validated MYH14 upregulation in EC cases compared with that in normal controls and confirmed that MYH14 was an independent and unfavorable prognostic indicator of EC. MYH14 impaired cell sensitivity to carboplatin, paclitaxel, and progesterone, and increased cell proliferation and metastasis in EC. The mechanistic study showed that MYH14 interacted with MYH9 and impaired GSK3ß-mediated ß-catenin ubiquitination and degradation, thus facilitating the Wnt/ß-catenin signaling pathway and epithelial-mesenchymal transition. Sesamolin, a natural compound extracted from Sesamum indicum (L.), directly targeted MYH14 and attenuated EC progression. Additionally, the compound disrupted the interplay between MYH14 and MYH9 and repressed MYH9-regulated Wnt/ß-catenin signaling. The in vivo study further verified sesamolin as a therapeutic drug without side effects. CONCLUSIONS: Herein, we identified that EC prognosis-associated MYH14 was independently responsible for poor overall survival time of patients, and it augmented EC progression by activating Wnt/ß-catenin signaling. Targeting MYH14 by sesamolin, a cytotoxicity-based approach, can be applied synergistically with chemotherapy and endocrine therapy to eventually mitigate EC development. This study emphasizes MYH14 as a potential target and sesamolin as a valuable natural drug for EC therapy.


Sujet(s)
Tumeurs de l'endomètre , Glycogen synthase kinase 3 beta , Chaînes lourdes de myosine , bêta-Caténine , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/anatomopathologie , Chaînes lourdes de myosine/métabolisme , Chaînes lourdes de myosine/génétique , Animaux , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Lignée cellulaire tumorale , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Pronostic , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Adulte d'âge moyen , Naphtoquinones/pharmacologie
13.
Gynecol Oncol ; 186: 154-160, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38691985

RÉSUMÉ

OBJECTIVE: This study aimed to evaluate the prognostic ability of mismatch repair deficiency (MMR-d) and abnormal p53 expression (p53abn) in patients with endometrial atypical hyperplasia (EAH) who underwent fertility-preserving treatment. METHODS: This retrospective study evaluated 51 patients with EAH who underwent fertility-sparing treatment. Endometrial biopsy specimens obtained before hormone therapy were collected and used for immunohistochemical staining for MMR and p53 proteins. Response, relapse, and progression rates were assessed based on age, body mass index, diabetes, polycystic ovary syndrome, reproductive history, MMR status, and p53 status. RESULTS: Overall, 11/51 (21.6%) patients had loss of MMR proteins and 6/51 (11.8%) had p53abn. Patients with MMR-d had lower complete response (CR) rates than those with normal staining patients at 12 months after initial treatment (p = 0.049). Patients with MMR-d had significantly higher relapse rates than those with MMR-p at the 1-year follow-ups after achieving CR (p = 0.035). Moreover, patients with MMR-d had a higher incidence of disease progression at 2, 3, and 4 years after fertility-sparing treatment (p = 0.001, p = 0.01 and p = 0.035, respectively). Patients with p53abn had higher relapse rates than those with p53wt at the 1- and 2-year follow-ups after achieving CR (p = 0.047 and p = 0.036, respectively). Moreover, patients with p53abn had a higher incidence of disease progression at 3 and 4 years after fertility-sparing treatment (p = 0.02 and p = 0.049, respectively). CONCLUSIONS: EAH patients with MMR-d and p53abn have a significantly higher risk of disease relapse and progression. Thus, MMR-d and p53abn may be used as predictive biomarkers of progestin resistance and endometrial tumorigenesis in EAH.


Sujet(s)
Réparation de mésappariement de l'ADN , Hyperplasie endométriale , Tumeurs de l'endomètre , Préservation de la fertilité , Protéine p53 suppresseur de tumeur , Humains , Femelle , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Adulte , Hyperplasie endométriale/métabolisme , Hyperplasie endométriale/anatomopathologie , Hyperplasie endométriale/traitement médicamenteux , Hyperplasie endométriale/génétique , Études rétrospectives , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/génétique , Préservation de la fertilité/méthodes , Progestérone , Pronostic
14.
Ann Oncol ; 35(7): 667-676, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38704093

RÉSUMÉ

BACKGROUND: Immunotherapy combined with chemotherapy significantly improves progression-free survival (PFS) compared to first-line chemotherapy alone in advanced endometrial cancer (EC), with a much larger effect size in microsatellite instability-high (MSI-H) cases. New biomarkers might help to select patients who may have benefit among those with a microsatellite-stable (MSS) tumor. PATIENTS AND METHODS: In a pre-planned translational analysis of the MITO END-3 trial, we assessed the significance of genomic abnormalities in patients randomized to standard carboplatin/paclitaxel without or with avelumab. RESULTS: Out of 125 randomized patients, 109 had samples eligible for next-generation sequencing analysis, and 102 had MSI tested. According to The Cancer Genome Atlas (TCGA), there were 29 cases with MSI-H, 26 with MSS TP53 wild type (wt), 47 with MSS TP53 mutated (mut), and 1 case with POLE mutation. Four mutated genes were present in >30% of cases: TP53, PIK3CA, ARID1A, and PTEN. Eleven patients (10%) had a BRCA1/2 mutation (five in MSI-H and six in MSS). High tumor mutational burden (≥10 muts/Mb) was observed in all MSI-H patients, in 4 out of 47 MSS/TP53 mut, and no case in the MSS/TP53 wt category. The effect of avelumab on PFS significantly varied according to TCGA categories, being favorable in MSI-H and worst in MSS/TP53 mut (P interaction = 0.003); a similar non-significant trend was seen in survival analysis. ARID1A and PTEN also showed a statistically significant interaction with treatment effect, which was better in the presence of the mutation (ARID1A P interaction = 0.01; PTEN P interaction = 0.002). CONCLUSION: The MITO END-3 trial results suggest that TP53 mutation is associated with a poor effect of avelumab, while mutations of PTEN and ARID1A are related to a positive effect of the drug in patients with advanced EC.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Tumeurs de l'endomètre , Instabilité des microsatellites , Mutation , Paclitaxel , Humains , Femelle , Anticorps monoclonaux humanisés/usage thérapeutique , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte d'âge moyen , Paclitaxel/usage thérapeutique , Paclitaxel/administration et posologie , Sujet âgé , Carboplatine/administration et posologie , Carboplatine/pharmacologie , Carboplatine/usage thérapeutique , Immunothérapie/méthodes , Phosphohydrolase PTEN/génétique , Adulte , Survie sans progression , Marqueurs biologiques tumoraux/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéines de liaison à l'ADN/génétique , Séquençage nucléotidique à haut débit , Facteurs de transcription , Phosphatidylinositol 3-kinases de classe I
15.
Cancer Res Commun ; 4(6): 1495-1504, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38767454

RÉSUMÉ

Cancer cells with DNA repair defects (e.g., BRCA1/2 mutant cells) are vulnerable to PARP inhibitors (PARPi) due to induction of synthetic lethality. However, recent clinical evidence has shown that PARPi can prevent the growth of some cancers irrespective of their BRCA1/2 status, suggesting alternative mechanisms of action. We previously discovered one such mechanism in breast cancer involving DDX21, an RNA helicase that localizes to the nucleoli of cells and is a target of PARP1. We have now extended this observation in endometrial and ovarian cancers and provided links to patient outcomes. When PARP1-mediated ADPRylation of DDX21 is inhibited by niraparib, DDX21 is mislocalized to the nucleoplasm resulting in decreased rDNA transcription, which leads to a reduction in ribosome biogenesis, protein translation, and ultimately endometrial and ovarian cancer cell growth. High PARP1 expression was associated with high nucleolar localization of DDX21 in both cancers. High nucleolar DDX21 negatively correlated with calculated IC50s for niraparib. By studying endometrial cancer patient samples, we were able to show that high DDX21 nucleolar localization was significantly associated with decreased survival. Our study suggests that the use of PARPi as a cancer therapeutic can be expanded to further types of cancers and that DDX21 localization can potentially be used as a prognostic factor and as a biomarker for response to PARPi. SIGNIFICANCE: Currently, there are no reliable biomarkers for response to PARPi outside of homologous recombination deficiency. Herein we present a unique potential biomarker, with clear functional understanding of the molecular mechanism by which DDX21 nucleolar localization can predict response to PARPi.


Sujet(s)
Nucléole , DEAD-box RNA helicases , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Femelle , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Nucléole/effets des médicaments et des substances chimiques , Nucléole/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Lignée cellulaire tumorale , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/mortalité , Tumeurs de l'ovaire/métabolisme , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/mortalité , Tumeurs de l'endomètre/métabolisme , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Pronostic , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de l'appareil génital féminin/génétique , Tumeurs de l'appareil génital féminin/anatomopathologie , Tumeurs de l'appareil génital féminin/traitement médicamenteux , Tumeurs de l'appareil génital féminin/mortalité , Tumeurs de l'appareil génital féminin/métabolisme , Indazoles
20.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 140-146, 2024 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-38678618

RÉSUMÉ

The incidence and mortality of endometrial carcinoma (EC) are increasing year by year. Although the curative effect of surgery and commonly used drugs is clear, it is accompanied by obvious side effects, and safe and effective means are urgently needed to promote the curative effect and decrease the toxicity of drugs. Traditional Chinese medicine has been passed down for thousands of years in China and has proved to be advantageous in the treatment of various cancers and the auxiliary enhancement and reduction of toxicity. This paper reviewed the role and internal mechanism of Salvia miltiorrhiza in preventing and treating endometrial carcinoma by referring to relevant literature and works, so as to more comprehensively understand and grasp the research status, effective components, curative effect and effective mechanism of S. miltiorrhiza in preventing and treating endometrial carcinoma, and provide ideas and basis for clinical use and basic research.


Sujet(s)
Médicaments issus de plantes chinoises , Tumeurs de l'endomètre , Salvia miltiorrhiza , Salvia miltiorrhiza/composition chimique , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/pharmacologie , Médecine traditionnelle chinoise/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...