Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.044
Filtrer
1.
Acta Oncol ; 63: 620-635, 2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39099323

RÉSUMÉ

BACKGROUND AND PURPOSE: Metaplastic breast carcinoma (BC-Mp) is an uncommon subtype that poses unique challenges. The limited information on patient prognosis and therapeutic strategies motivated our research initiative. We aimed to assess disease-free survival (DFS), overall survival (OS), and influential factors in patients with nonmetastatic BC-Mp. MATERIALS AND METHODS: In this multicenter retrospective cohort study, clinicopathological data for nonmetastatic BC-Mp patients treated at four oncology units in Poland (2012-2022) were gathered. RESULTS: Among 115 women (median age 61, range: 28-91), the median tumor size was 40 mm (range 20-130); 30% of patients exhibited positive local lymph nodes. The majority of patients presented with stage II (46%) and triple-negative breast cancer (TNBC) (84%). Radiotherapy was administered to 61% of patients. Surgical procedures included breast-conserving surgery in 31% of patients and mastectomy in 68%. Eighty-three per cent of patients received chemotherapy. The median estimated DFS and OS were 59 and 68 months, respectively. Multivariable analysis revealed that tumor size influenced DFS and OS (Hazard ratios [HR] = 1.02, 95%CI 0.01-0.03 for both endpoints) and taxanes application improved DFS (HR = 0.47, 95%CI 0.24-0.93), but other factors did not. For patients receiving neoadjuvant systemic therapy (N = 51), taxanes improved DFS and OS according to univariable analysis. INTERPRETATION: Our findings highlight poor DFS and OS regardless of receiving optimal treatment, emphasizing the need for tailored therapeutic strategies for BC-Mp patients. Taxanes appear promising in a neoadjuvant setting, particularly within the current standard of care for the TNBC subtype.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Études rétrospectives , Adulte d'âge moyen , Adulte , Sujet âgé , Pronostic , Tumeurs du sein/thérapie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/mortalité , Sujet âgé de 80 ans ou plus , Résultat thérapeutique , Survie sans rechute , Métaplasie/anatomopathologie , Métaplasie/thérapie , Mastectomie , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/thérapie , Tumeurs du sein triple-négatives/mortalité
2.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3868-3877, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39099360

RÉSUMÉ

The study investigated the effect of Compound Shougong Powder(CSGP) on the biological functions of triple-negative breast cancer(TNBC) cells and whether its mechanism of action was related to the epithelial-mesenchymal transition(EMT) signaling pathway. TNBC cells(MDA-MB-231 and BT-549) were treated with different concentrations of CSGP-containing serum. MTS assay was used to detect the effect of CSGP on the proliferation of TNBC cells. The EdU staining was used to detect the effect of CSGP on the proliferation of TNBC cells. Flow cytometry was used to examine the impact of CSGP on apoptosis of TNBC cells. Wound-healing and Transwell assays were used to evaluate the effects of different concentrations of CSGP on the migration and invasion capabilities of TNBC cells. RNA sequencing technology was utilized to elucidate its mechanism. Subsequently, qRT-PCR was performed to measure the mRNA expression levels of E-cadherin, N-cadherin, Slug, Snail, Vimentin, Twist, Zinc finger E-box-Binding homeobox 1(Zeb1), and Zinc finger E-box-Binding homeobox 2(Zeb2). Western blot was used to assess the protein expression levels of Slug, Vimentin, and E-cadherin. After intervention with CSGP, the proliferation of MDA-MB-231 and BT-549 cells significantly decreased, while the apoptosis rate markedly increased. The expression levels of the epithelial marker protein E-cadherin significantly increased, while the expression levels of the EMT-related transcription factors Slug and Vimentin showed a decrease. In conclusion, CSGP inhibits the EMT, thereby suppressing the malignant progression of TNBC.


Sujet(s)
Apoptose , Prolifération cellulaire , Médicaments issus de plantes chinoises , Transition épithélio-mésenchymateuse , Tumeurs du sein triple-négatives , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Médicaments issus de plantes chinoises/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Poudres/composition chimique , Cadhérines/génétique , Cadhérines/métabolisme
3.
Sci Rep ; 14(1): 18055, 2024 08 05.
Article de Anglais | MEDLINE | ID: mdl-39103475

RÉSUMÉ

The role of neoadjuvant chemotherapy and its benefits in patients with triple-negative breast cancer (TNBC) and small tumors are unclear. This study aims to compare survival differences between clinical T1 TNBC receiving neoadjuvant chemotherapy (NAC) and adjuvant chemotherapy (AC). Data for patients with clinical T1 TNBC were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. Patients were categorized according to whether they received chemotherapy before or after surgery. Propensity Score Matching (PSM) was used to minimize the influence of confounding factors. OS and BCSS were compared between the two treatment sequences using Kaplan-Meier and univariate and multivariable Cox proportional hazards regression analyses. The study included 6249 women with T1 TNBC. In multivariate analysis, compared with that in the AC group, the hazard ratio for death in the NAC group was 1.54 (95% confidence interval 1.26-1.89, p < 0.001). NAC offers no additional benefits in any age group or T, N subgroups. Our findings suggest that NAC does not provide additional benefit to patients with clinical T1 TNBC, even in the presence of lymph node metastasis, or T1c.


Sujet(s)
Traitement néoadjuvant , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/anatomopathologie , Femelle , Traitement néoadjuvant/méthodes , Adulte d'âge moyen , Adulte , Sujet âgé , Traitement médicamenteux adjuvant/méthodes , Programme SEER , Stadification tumorale , Estimation de Kaplan-Meier , Modèles des risques proportionnels
4.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126001

RÉSUMÉ

Breast cancer is the most diagnosed type of cancer worldwide and the second cause of death in women. Triple-negative breast cancer (TNBC) is the most aggressive, and due to the lack of specific targets, it is considered the most challenging subtype to treat and the subtype with the worst prognosis. The present study aims to determine the antitumor effect of beta-D-glucose-reduced silver nanoparticles (AgNPs-G) in a murine model of TNBC, as well as to study its effect on the tumor microenvironment. In an airbag model with 4T1 tumor cell implantation, the administration of AgNPs-G or doxorubicin showed antitumoral activity. Using immunohistochemistry it was demonstrated that treatment with AgNPs-G decreased the expression of PCNA, IDO, and GAL-3 and increased the expression of Caspase-3. In the tumor microenvironment, the treatment increased the percentage of memory T cells and innate effector cells and decreased CD4+ cells and regulatory T cells. There was also an increase in the levels of TNF-α, IFN-γ, and IL-6, while TNF-α was increased in serum. In conclusion, we suggest that AgNPs-G treatment has an antitumor effect that is demonstrated by its ability to remodel the tumor microenvironment in mice with TNBC.


Sujet(s)
Glucose , Nanoparticules métalliques , Argent , Tumeurs du sein triple-négatives , Microenvironnement tumoral , Animaux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Argent/composition chimique , Nanoparticules métalliques/composition chimique , Femelle , Souris , Glucose/métabolisme , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Souris de lignée BALB C , Doxorubicine/pharmacologie , Humains
5.
Sci Rep ; 14(1): 18181, 2024 08 06.
Article de Anglais | MEDLINE | ID: mdl-39107323

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks an actionable target with limited treatment options beyond conventional chemotherapy. Therapeutic failure is often encountered due to inherent or acquired resistance to chemotherapy. Previous studies implicated PI3K/Akt/mTOR signaling pathway in cancer stem cells (CSCs) enrichment and hence chemoresistance. The present study aimed at investigating the potential effect of piperine (PIP), an amide alkaloid isolated from Piper nigrum, on enhancing the sensitivity of TNBC cells to doxorubicin (DOX) in vitro on MDA-MB-231 cell line and in vivo in an animal model of Ehrlich ascites carcinoma solid tumor. Results showed a synergistic interaction between DOX and PIP on MDA-MB-231 cells. In addition, the combination elicited enhanced suppression of PI3K/Akt/mTOR signaling that paralleled an upregulation in this pathway's negative regulator, PTEN, along with a curtailment in the levels of the CSCs surrogate marker, aldehyde dehydrogenase-1 (ALDH-1). Meanwhile, in vivo investigations demonstrated the potential of the combination regimen to enhance necrosis while downregulating PTEN and curbing PI3K levels as well as p-Akt, mTOR, and ALDH-1 immunoreactivities. Notably, the combination failed to change cleaved poly-ADP ribose polymerase levels suggesting a pro-necrotic rather than pro-apoptotic mechanism. Overall, these findings suggest a potential role of PIP in decreasing the resistance to DOX in vitro and in vivo, likely by interfering with the PI3K/Akt/mTOR pathway and CSCs.


Sujet(s)
Alcaloïdes , Benzodioxoles , Doxorubicine , Cellules souches tumorales , Phosphatidylinositol 3-kinases , Pipéridines , Amides gras polyinsaturés N-alkylés , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Tumeurs du sein triple-négatives , Doxorubicine/pharmacologie , Amides gras polyinsaturés N-alkylés/pharmacologie , Pipéridines/pharmacologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Humains , Alcaloïdes/pharmacologie , Benzodioxoles/pharmacologie , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Femelle , Lignée cellulaire tumorale , Sérine-thréonine kinases TOR/métabolisme , Synergie des médicaments , Souris , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
6.
J Clin Invest ; 134(16)2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39145451

RÉSUMÉ

CD44 is associated with a high risk of metastasis, recurrence, and drug resistance in various cancers. Here we report that platelet endothelial aggregation receptor 1 (PEAR1) is a CD44 chaperone protein that protected CD44 from endocytosis-mediated degradation and enhances cleavage of the CD44 intracellular domain (CD44-ICD). Furthermore, we found that lysyl oxidase-like protein 2 (LOXL2), an endogenous ligand of PEAR1, bound to the PEAR1-EMI domain and facilitated the interaction between PEAR1 and CD44 by inducing PEAR1 Ser891 phosphorylation in a manner that was independent of its enzyme activity. Levels of PEAR1 protein and PEAR1 phosphorylation at Ser891 were increased in patients with triple-negative breast cancer (TNBC), were positively correlated with expression of LOXL2 and CD44, and were negatively correlated with overall survival. The level of PEAR1 Ser891 phosphorylation was identified as the best independent prognostic factor in TNBC patients. The prognostic efficacy of the combination of PEAR1 phosphorylation at Ser891 and CD44 expression was superior to that of PEAR1 phosphorylation at Ser891 alone. Blocking the interaction between LOXL2 and PEAR1 with monoclonal antibodies significantly inhibited TNBC metastasis, representing a promising therapeutic strategy for TNBC.


Sujet(s)
Amino-acid oxidoreductases , Antigènes CD44 , Métastase tumorale , Récepteurs de surface cellulaire , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Femelle , Phosphorylation , Récepteurs de surface cellulaire/métabolisme , Récepteurs de surface cellulaire/génétique , Amino-acid oxidoreductases/métabolisme , Amino-acid oxidoreductases/génétique , Animaux , Lignée cellulaire tumorale , Souris , Protéolyse , Protéines tumorales/métabolisme , Protéines tumorales/génétique
7.
BMC Cancer ; 24(1): 1016, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148033

RÉSUMÉ

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive subtype with poor prognosis. We aimed to determine whether circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) could predict response and long-term outcomes to neoadjuvant chemotherapy (NAC). METHODS: Patients with TNBC were enrolled between 2017-2021 at The University of Texas MD Anderson Cancer Center (Houston, TX). Serial plasma samples were collected at four timepoints: pre-NAC (baseline), 12-weeks after NAC (mid-NAC), after NAC/prior to surgery (post-NAC), and one-year after surgery. ctDNA was quantified using a tumor-informed ctDNA assay (SignateraTM, Natera, Inc.) and CTC enumeration using CellSearch. Wilcoxon and Fisher's exact tests were used for comparisons between groups and Kaplan-Meier analysis used for survival outcomes. RESULTS: In total, 37 patients were enrolled. The mean age was 50 and majority of patients had invasive ductal carcinoma (34, 91.9%) with clinical T2, (25, 67.6%) node-negative disease (21, 56.8%). Baseline ctDNA was detected in 90% (27/30) of patients, of whom 70.4% (19/27) achieved ctDNA clearance by mid-NAC. ctDNA clearance at mid-NAC was significantly associated with pathologic complete response (p = 0.02), whereas CTC clearance was not (p = 0.52). There were no differences in overall survival (OS) and recurrence-free survival (RFS) with positive baseline ctDNA and CTC. However, positive ctDNA at mid-NAC was significantly associated with worse OS and RFS (p = 0.0002 and p = 0.0034, respectively). CONCLUSIONS: Early clearance of ctDNA served as a predictive and prognostic marker in TNBC. Personalized ctDNA monitoring during NAC may help predict response and guide treatment.


Sujet(s)
ADN tumoral circulant , Traitement néoadjuvant , Cellules tumorales circulantes , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/sang , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Femelle , Traitement néoadjuvant/méthodes , Adulte d'âge moyen , Adulte , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Marqueurs biologiques tumoraux/sang , Sujet âgé , Pronostic , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Résultat thérapeutique
8.
FASEB J ; 38(15): e23872, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39126272

RÉSUMÉ

Paclitaxel is among the most active chemotherapy drugs for the aggressive triple negative breast cancer (TNBC). Unfortunately, it often induces painful peripheral neuropathy (CIPN), a major debilitating side effect. Here we demonstrate that in naive and breast tumor-bearing immunocompetent mice, a clinically relevant dose of FTY720/Fingolimod that targets sphingosine-1-phosphate receptor 1 (S1PR1), alleviated paclitaxel-induced neuropathic pain. FTY720 also significantly attenuated paclitaxel-stimulated glial fibrillary acidic protein (GFAP), a marker for activated astrocytes, and expression of the astrocyte-secreted synaptogenic protein Sparcl1/Hevin, a key regulator of synapse formation. Notably, the formation of excitatory synapses containing VGluT2 in the spinal cord dorsal horn induced by paclitaxel was also inhibited by FTY720 treatment, supporting the involvement of astrocytes and Sparcl1 in CIPN. Furthermore, in this TNBC mouse model that mimics human breast cancer, FTY720 administration also enhanced the anti-tumor effects of paclitaxel, leading to reduced tumor progression and lung metastasis. Taken together, our findings suggest that targeting the S1P/S1PR1 axis with FTY720 is a multipronged approach that holds promise as a therapeutic strategy for alleviating both CIPN and enhancing the efficacy of chemotherapy in TNBC treatment.


Sujet(s)
Chlorhydrate de fingolimod , Névralgie , Paclitaxel , Animaux , Chlorhydrate de fingolimod/pharmacologie , Paclitaxel/pharmacologie , Névralgie/induit chimiquement , Névralgie/traitement médicamenteux , Névralgie/métabolisme , Névralgie/anatomopathologie , Souris , Femelle , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Lignée cellulaire tumorale , Récepteurs de la sphingosine-1-phosphate/métabolisme , Humains , Évolution de la maladie , Antinéoplasiques d'origine végétale/pharmacologie , Protéine gliofibrillaire acide/métabolisme , Protéine gliofibrillaire acide/génétique
9.
J Med Case Rep ; 18(1): 368, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39138583

RÉSUMÉ

BACKGROUND: Breast cancer has emerged as the foremost cause of female mortality worldwide, with triple negative breast cancer accounting for approximately 10-15% of all breast cancer cases. The triple negative breast cancer family has obvious familial heritability, but no potential pathogenic variation was found in BRCA1/2. CASE PRESENTATION: The patient was a 56-year-old woman of Han ethnicity. The clinical characteristics of this patient with breast cancer were summarized, peripheral blood of one normal female and two patients with breast cancer in this family was collected, DNA was extracted, and the potential pathogenic variation was analyzed by whole exome sequencing. The normal female and two patients with breast cancer in this family shared a maternal grandmother. The proband's right breast mass was punctured, and the biopsy showed invasive carcinoma of the right breast, grade II-III, with necrosis. No mutation was found in BRCA1/2 gene test; immunohistochemical of surgical specimens showed triple negative breast cancer. Three mutation types and 17 gene mutation sites were detected through bioinformatics prediction analysis on the basis of co-segregation of genotype and phenotype within the family and whole exome sequencing results. Combined with the Cancer Genome Atlas database comprehensive analysis, the MT1E c.G107A (p.C36Y) mutation may be a potential pathogenic site. CONCLUSIONS: Through whole exome sequencing, we identified a total of 17 potential pathogenic mutation loci, none of which have been reported thus far. Therefore, our work expanded the gene mutation spectrum of familial hereditary triple negative breast cancer, which can provide more basis for family genetic counseling.


Sujet(s)
Tumeurs du sein , , Tumeurs du sein triple-négatives , Humains , Femelle , Adulte d'âge moyen , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Pedigree , Prédisposition génétique à une maladie , Mutation , Protéine BRCA1/génétique , Dépistage génétique
10.
IET Nanobiotechnol ; 2024: 3786627, 2024.
Article de Anglais | MEDLINE | ID: mdl-39144410

RÉSUMÉ

Background: Naringenin has shown great promise in the realm of cancer therapeutics, demonstrating excellent cytotoxic action toward cancer cells and the enhanced effects of radiation therapy in vitro. However, the medicinal value of naringenin is severely limited clinically by poor bioavailability. Thus, multiple drug-delivery strategies for overcoming this limitation have been developed, of which liposomes are considered the most suitable due to their amphiphilic, modifiable, and biocompatible characteristics. In this study, we investigated the role of naringenin and liposomal-delivered naringenin as adjuncts to radiotherapy in the MDA-MB-231 triple-negative breast cancer cell line in vitro. Materials and Methods: Liposomal-naringenin was synthesized by thin-film hydration and extrusion and was characterized by spectrophotometry, dynamic light scattering, and zeta potential. The effects of free-from naringenin and liposomal-naringenin were evaluated toward MDA-MB-231 cell viability when combined with varying doses of radiation. Additionally, cell growth patterns, morphology, and colony formation were evaluated. Results: The analysis demonstrated IC50 values of 387.5 and 546.6 µg/ml for naringenin and liposomal-naringenin, respectively. Naringenin and liposomal-naringenin significantly lowered cell viability, proliferation, and colony formation dose-dependently, as compared to radiation in isolation. Conclusion: The findings presented herein concur with previous accounts of the radiosensitizing potential of naringenin and further highlight the considerable biomedical application of liposomal-naringenin within the realm of radiotherapy.


Sujet(s)
Survie cellulaire , Flavanones , Liposomes , Radiosensibilisants , Tumeurs du sein triple-négatives , Flavanones/composition chimique , Flavanones/pharmacologie , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Liposomes/composition chimique , Lignée cellulaire tumorale , Radiosensibilisants/pharmacologie , Radiosensibilisants/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules MDA-MB-231
11.
Med Oncol ; 41(9): 222, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39120634

RÉSUMÉ

Breast cancer (BC) is a significant cause of cancer-related mortality, and triple-negative breast cancer (TNBC) is a particularly aggressive subtype associated with high mortality rates, especially among younger females. TNBC poses a considerable clinical challenge due to its aggressive tumor behavior and limited therapeutic options. Aberrations within the PI3K/AKT pathway are prevalent in TNBC and correlate with increased therapeutic intervention resistance and poor outcomes. MicroRNAs (miRs) have emerged as crucial PI3K/AKT pathway regulators influencing various cellular processes involved in TNBC pathogenesis. The levels of miRs, including miR-193, miR-4649-5p, and miR-449a, undergo notable changes in TNBC tumor tissues, emphasizing their significance in cancer biology. This review explored the intricate interplay between miR variants and PI3K/AKT signaling in TNBC. The review focused on the molecular mechanisms underlying miR-mediated dysregulation of this pathway and highlighted specific miRs and their targets. In addition, we explore the clinical implications of miR dysregulation in TNBC, particularly its correlation with TNBC prognosis and therapeutic resistance. Elucidating the roles of miRs in modulating the PI3K/AKT signaling pathway will enhance our understanding of TNBC biology and unveil potential therapeutic targets. This comprehensive review aims to discuss current knowledge and open promising avenues for future research, ultimately facilitating the development of precise and effective treatments for patients with TNBC.


Sujet(s)
microARN , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , microARN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Transduction du signal/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Femelle , Régulation de l'expression des gènes tumoraux
12.
Mol Biol Rep ; 51(1): 901, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39126511

RÉSUMÉ

BACKGROUND: Within the subtypes of breast cancer pathologies, triple-negative breast cancer (TNBC) exhibits the highest degree of malignancy and unfavorable outcome, which has great significance in exploring the molecular mechanisms underlying TNBC. This study especially investigated the expression and function of hsa_circ_0003528 in TNBC. METHODS: The expression changes of hsa_circ_0003528 were identified from the GEO database (GSE101123) and validated by RT-qPCR. The clinical significance of hsa_circ_0003528 was evaluated using χ2 tests and Kaplan-Meier curve analysis. Bioinformatic analysis and dual-luciferase reporter assay were used to identify the potential downstream miRNA of hsa_circ_0003528. The cellular experiments were conducted to evaluate the impact of hsa_circ_0003528 or/and miR-215 on TNBC cells. RESULTS: The hsa_circ_0003528 was selected from the circRNA profile in breast cancer obtained from the GSE101123 dataset. hsa_circ_0003528 expression levels were increased in breast cancer tissues, especially in TNBC tissues. The elevated expression of hsa_circ_0003528 was negatively associated with TNBC patients' overall survival. Silencing of hsa_circ_0003528 hindered the proliferative potential, migration abilities, and invasive capacities of TNBC cells, while downregulation of miR-215 partially diminished the effects of si-hsa_circ_0003528 on TNBC cells. CONCLUSION: hsa_circ_0003528 is upregulated in TNBC and can facilitate aggressive cellular behaviors by regulating miR-215 expression, hinting at its potential as a biomarker and therapeutic target in the treatment of TNBC.


Sujet(s)
Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , microARN , ARN circulaire , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , microARN/génétique , ARN circulaire/génétique , Femelle , Pronostic , Régulation de l'expression des gènes tumoraux/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Marqueurs biologiques tumoraux/génétique , Adulte d'âge moyen , Estimation de Kaplan-Meier
13.
Am J Case Rep ; 25: e943882, 2024 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-39127886

RÉSUMÉ

BACKGROUND Hereditary breast cancer arising in BRCA1-deficient patients is commonly diagnosed as invasive carcinoma of no special type (NST) with medullary features, while invasive lobular carcinoma (ILC) appears to be significantly under-represented in BRCA1 mutation carriers. We report a case of pleomorphic ILC arising in a 28-year-old woman harboring a germline BRCA1 c.3756_3759delGTCT p.(Ser1253Argfs*10) pathogenic variant. CASE REPORT A nulliparous 28-year-old woman with a family history of BRCA1 mutation presented to the symptomatic breast clinic with a several-week history of a left 80-mm breast lump. Core biopsy established a diagnosis of a poorly differentiated triple-negative breast cancer (TNBC) of pleomorphic lobular phenotype. Her clinical diagnosis was cT3, N0, M0, cStageIIB. The MDT recommended CT staging, MRI breast imaging and neoadjuvant chemotherapy (NACT). PET CT imaging showed no evidence of distant metastatic disease. The patient had a good radiological response to NACT with a FEC-T carboplatin regimen. Post-NACT imaging showed a residual cystic mass and the patient underwent a mastectomy and sentinel lymph node biopsy with plans for a delayed latissimus dorsi reconstruction following her adjuvant radiotherapy treatment. A complete pathological response was subsequently demonstrated without any evidence of metastatic disease. CONCLUSIONS This case is the first report of pleomorphic ILC with a triple-negative receptor status and a complete pathological response in a BRCA1 mutation carrier. Our study expands the heterogeneous spectrum of TNBC and contributes to a better understanding of the molecular genetic landscape that characterizes invasive pleomorphic lobular neoplasia.


Sujet(s)
Carcinome lobulaire , Tumeurs du sein triple-négatives , Humains , Femelle , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/thérapie , Adulte , Carcinome lobulaire/génétique , Carcinome lobulaire/anatomopathologie , Carcinome lobulaire/thérapie , Protéine BRCA1/génétique , Mutation germinale
14.
BMC Cancer ; 24(1): 997, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39135184

RÉSUMÉ

BACKGROUND: Increased level of stromal tumor-infiltrating lymphocytes (sTILs) are associated with therapeutic outcomes and prognosis in triple-negative breast cancer (TNBC). This study aimed to investigate the associations of clinicopathologic and sonographic features with sTILs level in TNBC. METHODS: This study included invasive TNBC patients with postoperative evaluation of sTILs after surgical resection. Tumor shape, margin, orientation, echo pattern, posterior features, calcification, and vascularity were retrospectively evaluated. The patients were categorized into high-sTILs (≥ 20%) and low-sTILs (< 20%) level groups. Chi-square or Fisher's exact tests were used to assess the association of clinicopathologic and sonographic features with sTILs level. RESULTS: The 171 patients (mean ± SD age, 54.7 ± 10.3 years [range, 22‒87 years]) included 58.5% (100/171) with low-sTILs level and 41.5% (71/171) with high-sTILs level. The TNBC tumors with high-sTILs level were more likely to be no special type invasive carcinoma (p = 0.008), higher histologic grade (p = 0.029), higher Ki-67 proliferation rate (all p < 0.05), and lower frequency of associated DCIS component (p = 0.026). In addition, the TNBC tumors with high-sTILs level were more likely to be an oval or round shape (p = 0.001), parallel orientation (p = 0.011), circumscribed or micro-lobulated margins (p < 0.001), complex cystic and solid echo patterns (p = 0.001), posterior enhancement (p = 0.002), and less likely to have a heterogeneous pattern (p = 0.001) and no posterior features (p = 0.002). CONCLUSIONS: This preliminary study showed that preoperative sonographic characteristics could be helpful in distinguishing high-sTILs from low-sTILs in TNBC patients.


Sujet(s)
Lymphocytes TIL , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/imagerie diagnostique , Tumeurs du sein triple-négatives/anatomopathologie , Femelle , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Adulte d'âge moyen , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études rétrospectives , Jeune adulte , Pronostic , Échographie mammaire/méthodes , Échographie/méthodes
15.
Front Immunol ; 15: 1375528, 2024.
Article de Anglais | MEDLINE | ID: mdl-39104525

RÉSUMÉ

Tissue-resident macrophages (TRMs) are an integral part of the innate immune system, but their biology is not well understood in the context of cancer. Distinctive resident macrophage populations are identified in different organs in mice using fate mapping studies. They develop from the yolk sac and self-maintain themselves lifelong in specific tissular niches. Similarly, breast-resident macrophages are part of the mammary gland microenvironment. They reside in the breast adipose tissue stroma and close to the ductal epithelium and help in morphogenesis. In breast cancer, TRMs may promote disease progression and metastasis; however, precise mechanisms have not been elucidated. TRMs interact intimately with recruited macrophages, cytotoxic T cells, and other immune cells along with cancer cells, deciding further immunosuppressive or cytotoxic pathways. Moreover, triple-negative breast cancer (TNBC), which is generally associated with poor outcomes, can harbor specific TRM phenotypes. The influence of TRMs on adipose tissue stroma of the mammary gland also contributes to tumor progression. The complex crosstalk between TRMs with T cells, stroma, and breast cancer cells can establish a cascade of downstream events, understanding which can offer new insight for drug discovery and upcoming treatment choices. This review aims to acknowledge the previous research done in this regard while exploring existing research gaps and the future therapeutic potential of TRMs as a combination or single agent in breast cancer.


Sujet(s)
Tumeurs du sein , Macrophages , Microenvironnement tumoral , Humains , Animaux , Femelle , Microenvironnement tumoral/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Tissu adipeux/immunologie , Tissu adipeux/métabolisme , Tissu adipeux/cytologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme
16.
AAPS J ; 26(5): 91, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107504

RÉSUMÉ

Repurposing drugs offers several advantages, including reduced time and cost compared to developing new drugs from scratch. It leverages existing knowledge about drug safety, dosage, and pharmacokinetics, expediting the process of clinical trials and regulatory approval. Dihydroartemisinin (DHA) is a semi-synthetic and active metabolite of all artemisinin molecules and is FDA-approved for the treatment of malaria. Apart from having anti-malarial properties, DHA also possesses anticancer properties. However, its pharmacological actions are limited by toxicity and solubility problems. To overcome these challenges and enhance its anticancer effectiveness, we designed an exosomal formulation of DHA. We isolated exosomes from bovine milk using differential ultracentrifugation and loaded DHA using sonication. Scanning and transition electron microscopy revealed a size of roughly 100 nm, with a spherical shape. Furthermore, in pH 7.4 and 5.5, the exosomes exhibited burst release followed by sustained release. Multiple in vitro cell culture tests demonstrated that Exo-DHA exhibited enhanced anticancer activity, including cytotoxicity, cellular uptake, generation of reactive oxygen species (ROS), disruption of mitochondrial membrane potential, and inhibition of colony formation. Additional evidence supporting Exo-DHA's anti-migration ability came from transwell migration and scratch assays. Based on these results, it was concluded that the anticancer efficacy of DHA was improved when loaded into bovine milk-derived exosomes. While the in vitro results are encouraging, more in vivo testing in suitable animal models and biochemical marker analysis are warranted.


Sujet(s)
Antinéoplasiques , Artémisinines , Exosomes , Lait , Tumeurs du sein triple-négatives , Artémisinines/pharmacologie , Artémisinines/administration et posologie , Artémisinines/composition chimique , Animaux , Lait/composition chimique , Bovins , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Lignée cellulaire tumorale , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Femelle , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques
17.
Luminescence ; 39(8): e4848, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39092486

RÉSUMÉ

Herein, luminous blue carbon quantum dots (CDs) employing Anisomeles indica (Catmint) were reported with imaging, self-targeting, and therapeutic effects on triple-negative breast cancer (TNBC, MDA-MB-231) cells. The salient features of CDs generated from catmint are as follows: i) optical studies confirm CDs with excitation-dependent emission; ii) high-throughput characterization authenticates the formation of CDs with near-spherical shape with diameter ranging between 5 and 15 nm; iii) CDs induce cytotoxicity (3.22 ± 0.64 µg/ml) in triple-negative breast cancer (TNBC, MDA-MB-231) cells; iv) fluorescence microscopy demonstrates that CDs promote apoptosis by increasing reactive oxygen species (ROS) and decreasing mitochondrial membrane potential; v) CDs significantly up-regulate pro-apoptotic gene expression levels such as caspases-8/9/3. Finally, our work demonstrates that catmint-derived CDs are prospective nanotheranostics that augment cancer targeting and imaging.


Sujet(s)
Apoptose , Carbone , Boîtes quantiques , Transduction du signal , Tumeurs du sein triple-négatives , Boîtes quantiques/composition chimique , Humains , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Carbone/composition chimique , Carbone/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Lamiaceae/composition chimique , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
18.
Rev Med Inst Mex Seguro Soc ; 62(1): 1-7, 2024 Jan 08.
Article de Espagnol | MEDLINE | ID: mdl-39110839

RÉSUMÉ

Background: Primary breast tumors with neuroendocrine (NE) differentiation are a heterogeneous tumor group with diversity of biological behavior, with poorly defined prevalence and prognosis. Objective: To evaluate the chromogranin, synaptophysin, CD56, INSM1 markers expression prevalence and the association between NE differentiation and tumor molecular type. Material and methods: Observational, cross-sectional study which included 110 breast tissue samples with primary invasive carcinoma. Immunohistochemistry was performed for chromogranin, synaptophysin, CD56 and INMS1 markers. NE differentiation was considered with 10-90% positive cells, and NE tumor with > 90% positive cells. Results: 26.3% showed neuroendocrine differentiation. Out of these, 48.2% were luminal-A type, 24.1% luminal-B, 11.5% HER2neu, 17.2% triple-negative; 1.8% were NE tumors. Tumors were marker positive, and out of these to chromogranin in 24.5%, synaptophysin in 28.2%, CD56 in 2.7%, INSM1 in 16.4%. Synaptophysin was expressed in 17.3% luminal-A type, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negative. NE differentiation showed association with synaptophysin expression (r = 0.586, p = 0.0001). Conclusion: The NE differentiation prevalence was 26.3% in primary invasive breast cancers, with luminal-A molecular type predominance.


Introducción: los tumores primarios de mama con diferenciación neuroendócrina (NEBC por sus siglas en inglés) son un grupo heterogéneo de tumores con diversidad de comportamiento biológico, con prevalencia y pronóstico poco definido. Objetivo: evaluar la prevalencia de la expresión los marcadores cromogranina, sinaptofisina, CD56, INSM1 y la asociación entre la diferenciación neuroendócrina y el tipo molecular del tumor. Material y métodos: estudio observacional, transversal que incluyó 110 muestras de tejido mamario con carcinoma invasor primario. Se realizó inmunohistoquímica para los marcadores cromogranina, sinaptofisina, CD56 y INMS1. La presencia 10-90% de células positivas se consideró diferenciación neuroendócrina y tumor neuroendócrino con > 90% de células positivas. Resultados: el 26.3% mostró diferenciación neuroendócrina. De estos, 48.2% fueron tipo luminal-A, 24.1% luminal-B, 11.5% HER2neu y 17.2% triple-negativo; 1.8% resultaron tumores neuroendócrinos. Los tumores presentaron marcadores positivos y de estos, 24.5% fueron a cromogranina, 28.2% a sinaptofisina, 2.7% a CD56 y 16.4% a INSM1. La sinaptofisina se expresó en 17.3% del tipo luminal-A, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negativo. La diferenciación neuroendócrina mostró asociación con la expresión de sinaptofisina (r = 0.586, p = 0.0001). Conclusión: la prevalencia de la diferenciación neuroendócrina fue del 26.3% en los cánceres invasores primarios de mama, con predominio en el tipo molecular luminal-A.


Sujet(s)
Marqueurs biologiques tumoraux , Synaptophysine , Humains , Femelle , Études transversales , Marqueurs biologiques tumoraux/métabolisme , Adulte d'âge moyen , Adulte , Synaptophysine/métabolisme , Sujet âgé , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Antigènes CD56/métabolisme , Immunohistochimie , Protéines de répression/métabolisme , Chromogranine/métabolisme , Récepteur ErbB-2/métabolisme , Sujet âgé de 80 ans ou plus
19.
Sci Adv ; 10(32): eadl4043, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39110799

RÉSUMÉ

Sequencing-based mapping of ensemble pairwise interactions among regulatory elements support the existence of topological assemblies known as promoter-enhancer hubs or cliques in cancer. Yet, prevalence, regulators, and functions of promoter-enhancer hubs in individual cancer cells remain unclear. Here, we systematically integrated functional genomics, transcription factor screening, and optical mapping of promoter-enhancer interactions to identify key promoter-enhancer hubs, examine heterogeneity of their assembly, determine their regulators, and elucidate their role in gene expression control in individual triple negative breast cancer (TNBC) cells. Optical mapping of individual SOX9 and MYC alleles revealed the existence of frequent multiway interactions among promoters and enhancers within spatial hubs. Our single-allele studies further demonstrated that lineage-determining SOX9 and signaling-dependent NOTCH1 transcription factors compact MYC and SOX9 hubs. Together, our findings suggest that promoter-enhancer hubs are dynamic and heterogeneous topological assemblies, which are controlled by oncogenic transcription factors and facilitate subtype-restricted gene expression in cancer.


Sujet(s)
Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux , Régions promotrices (génétique) , Facteur de transcription SOX-9 , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Humains , Facteur de transcription SOX-9/génétique , Facteur de transcription SOX-9/métabolisme , Lignée cellulaire tumorale , Femelle , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Oncogènes , Récepteur Notch1/génétique , Récepteur Notch1/métabolisme
20.
Cells ; 13(15)2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39120328

RÉSUMÉ

Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, with a bad prognosis and lack of targeted therapeutic options. Characterized by the absence of estrogen receptors, progesterone receptors, and HER2 expression, TNBC is often associated with a significantly lower survival rate compared to other breast cancer subtypes. Our study aimed to explore the prognostic significance of 83 immune-related genes, by using transcriptomic data from the TCGA database. Our analysis identified the Poliovirus Receptor-Like 3 protein (PVRL3) as a critical negative prognostic marker in TNBC patients. Furthermore, we found that the Enhancer of Zeste Homolog 2 (EZH2), a well-known epigenetic regulator, plays a pivotal role in modulating PVRL3 levels in TNBC cancer cell lines expressing EZH2 along with high levels of PVRL3. The elucidation of the EZH2-PVRL3 regulatory axis provides valuable insights into the molecular mechanisms underlying TNBC aggressiveness and opens up potential pathways for personalized therapeutic intervention.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste , Régulation de l'expression des gènes tumoraux , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Pronostic , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Femelle , Lignée cellulaire tumorale , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Nectines/métabolisme , Nectines/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE