Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.231
Filtrer
1.
Breast Cancer Res ; 26(1): 111, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965614

RÉSUMÉ

BACKGROUND: Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS: We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS: We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS: We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.


Sujet(s)
Tumeurs du sein , Étude d'association pangénomique , Polymorphisme de nucléotide simple , Locus de caractère quantitatif , Récepteurs aux androgènes , Humains , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , 5alpha-Dihydrotestostérone/pharmacologie , 3-Phényl-2-thiohydantoïne/pharmacologie , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Nitriles/usage thérapeutique , Génotype , Pharmacogénétique/méthodes , Variants pharmacogénomiques , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Benzamides
2.
Cancer Genomics Proteomics ; 21(4): 368-379, 2024.
Article de Anglais | MEDLINE | ID: mdl-38944420

RÉSUMÉ

BACKGROUND/AIM: Aggressive breast cancer (BC) cells show high expression of Rho GTPase activating protein 29 (ARHGAP29), a negative regulator of RhoA. In breast cancer cells in which mesenchymal transformation was induced, ARHGAP29 was the only one of 32 GTPase-activating enzymes whose expression increased significantly. Therefore, we investigated whether there is a correlation between expression of ARHGAP29 and tumor progression in BC. Since tamoxifen-resistant BC cells exhibit increased mesenchymal properties and invasiveness, we additionally investigated the relationship between ARHGAP29 and increased invasion rate in tamoxifen resistance. The question arises as to whether ARHGAP29 is a suitable prognostic marker for the progression of BC. MATERIALS AND METHODS: Tissue microarrays were used to investigate expression of ARHGAP29 in BC and adjacent normal breast tissues. Knockdown experiments using siRNA were performed to investigate the influence of ARHGAP29 and the possible downstream actors RhoC and pAKT1 on invasive growth of tamoxifen-resistant BC spheroids in vitro. RESULTS: Expression of ARHGAP29 was frequently increased in BC tissues compared to adjacent normal breast tissues. In addition, there was evidence of a correlation between high ARHGAP29 expression and advanced clinical tumor stage. Tamoxifen-resistant BC cells show a significantly higher expression of ARHGAP29 compared to their parental wild-type cells. After knockdown of ARHGAP29 in tamoxifen-resistant BC cells, expression of RhoC was significantly reduced. Further, expression of pAKT1 decreased significantly. Invasive growth of three-dimensional tamoxifen-resistant BC spheroids was reduced after knockdown of ARHGAP29. This could be partially reversed by AKT1 activator SC79. CONCLUSION: Expression of ARHGAP29 correlates with the clinical tumor parameters of BC patients. In addition, ARHGAP29 is involved in increased invasiveness of tamoxifen-resistant BC cells. ARHGAP29 alone or in combination with its downstream partners RhoC and pAKT1 could be suitable prognostic markers for BC progression.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Protéines d'activation de la GTPase , Invasion tumorale , Tamoxifène , Humains , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Protéines d'activation de la GTPase/métabolisme , Protéines d'activation de la GTPase/génétique , Femelle , Adulte d'âge moyen , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Pronostic , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Protéine rhoC liant le GTP/métabolisme , Protéine rhoC liant le GTP/génétique
3.
Biosci Rep ; 44(7)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-38864530

RÉSUMÉ

Tamoxifen (TAM) is a key player in estrogen receptor-positive (ER+) breast cancer (BC); however, ∼30% of patients experience relapse and a lower survival rate due to TAM resistance. TAM resistance was related to the over expression of SOX-2 gene, which is regulated by the E2F3 transcription factor in the Wnt signaling pathway. It was suggested that SOX-2 overexpression was suppressed by dexamethasone (DEX), a glucocorticoid commonly prescribed to BC patients. The aim of the present study is to explore the effect of combining DEX and TAM on the inhibition of TAM-resistant LCC-2 cells (TAMR-1) through modulating the E2F3/SOX-2-mediated Wnt signaling pathway. The effect of the combination therapy on MCF-7 and TAMR-1 cell viability was assessed. Drug interactions were analyzed using CompuSyn and SynergyFinder softwares. Cell cycle distribution, apoptotic protein expression, gene expression levels of SOX-2 and E2F3, and cell migration were also assessed. Combining DEX with TAM led to synergistic inhibition of TAMR-1 cell proliferation and migration, induced apoptosis, reduced SOX-2 and E2F3 expression and was also associated with S and G2-M phase arrest. Therefore, combining DEX with TAM may present an effective therapeutic option to overcome TAM resistance, by targeting the E2F3/SOX-2/Wnt signaling pathway, in addition to its anti-inflammatory effect.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Dexaméthasone , Résistance aux médicaments antinéoplasiques , Synergie des médicaments , Tamoxifène , Humains , Tamoxifène/pharmacologie , Dexaméthasone/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Cellules MCF-7 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques hormonaux/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Facteur de transcription E2F3/métabolisme , Facteur de transcription E2F3/génétique , Facteurs de transcription SOX-B1/métabolisme , Facteurs de transcription SOX-B1/génétique
4.
Cell Death Dis ; 15(6): 444, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38914552

RÉSUMÉ

Endocrine resistance poses a significant clinical challenge for patients with hormone receptor-positive and human epithelial growth factor receptor 2-negative (HR + HER2-) breast cancer. Dysregulation of estrogen receptor (ER) and ERBB signaling pathways is implicated in resistance development; however, the integration of these pathways remains unclear. While SMAD4 is known to play diverse roles in tumorigenesis, its involvement in endocrine resistance is poorly understood. Here, we investigate the role of SMAD4 in acquired endocrine resistance in HR + HER2- breast cancer. Genome-wide CRISPR screening identifies SMAD4 as a regulator of 4-hydroxytamoxifen (OHT) sensitivity in T47D cells. Clinical data analysis reveals downregulated SMAD4 expression in breast cancer tissues, correlating with poor prognosis. Following endocrine therapy, SMAD4 expression is further suppressed. Functional studies demonstrate that SMAD4 depletion induces endocrine resistance in vitro and in vivo by enhancing ER and ERBB signaling. Concomitant inhibition of ER and ERBB signaling leads to aberrant autophagy activation. Simultaneous inhibition of ER, ERBB, and autophagy pathways synergistically impacts SMAD4-depleted cells. Our findings unveil a mechanism whereby endocrine therapy-induced SMAD4 downregulation drives acquired resistance by integrating ER and ERBB signaling and suggest a rational treatment strategy for endocrine-resistant HR + HER2- breast cancer patients.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Récepteur ErbB-2 , Récepteurs des oestrogènes , Transduction du signal , Protéine Smad-4 , Humains , Protéine Smad-4/métabolisme , Protéine Smad-4/génétique , Femelle , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Récepteurs des oestrogènes/métabolisme , Lignée cellulaire tumorale , Animaux , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Tamoxifène/analogues et dérivés , Souris , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Souris nude , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique
5.
Expert Opin Ther Pat ; 34(5): 333-350, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38836316

RÉSUMÉ

INTRODUCTION: Breast cancer is the most frequently diagnosed cancer worldwide. With around 70% of breast cancers expressing the estrogen receptor (ER), molecules capable of antagonizing and degrading ER (SERDs) or covalently binding to and antagonizing ER (SERCAs) are at the forefront of efforts to bring better treatments to patients. AREAS COVERED: This review summarizes patent applications that claim estrogen receptor degraders (SERDs) and covalent antagonists (SERCAs) identified using SciFinder between the period July 2021 to December 2023. A total of 91 new patent applications from 32 different applicants are evaluated with stratification into acidic SERDs, basic SERDs, SERCAs and miscellaneous degraders. EXPERT OPINION: The widespread adoption of fulvestrant in the treatment of ER+ breast cancer continues to stimulate research into orally bioavailable SERDs and SERCAs. A number of molecules have entered clinical development and, although some have been discontinued, a cohort of potential new treatments have generated encouraging efficacy and safety data. Notably, the first example of an oral SERD, elacestrant, has now been approved by the FDA and EMA, providing further encouragement for this class of targeted therapies.


Sujet(s)
Tumeurs du sein , Développement de médicament , Brevets comme sujet , Récepteurs des oestrogènes , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Animaux , Femelle , Récepteurs des oestrogènes/métabolisme , Antagonistes des récepteurs des oestrogènes/pharmacologie , Thérapie moléculaire ciblée , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Antinéoplasiques hormonaux/pharmacologie
6.
Oncol Res ; 32(6): 1093-1107, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827320

RÉSUMÉ

Breast cancer is the leading cause of cancer-related deaths in women worldwide, with Hormone Receptor (HR)+ being the predominant subtype. Tamoxifen (TAM) serves as the primary treatment for HR+ breast cancer. However, drug resistance often leads to recurrence, underscoring the need to develop new therapies to enhance patient quality of life and reduce recurrence rates. Artemisinin (ART) has demonstrated efficacy in inhibiting the growth of drug-resistant cells, positioning art as a viable option for counteracting endocrine resistance. This study explored the interaction between artemisinin and tamoxifen through a combined approach of bioinformatics analysis and experimental validation. Five characterized genes (ar, cdkn1a, erbb2, esr1, hsp90aa1) and seven drug-disease crossover genes (cyp2e1, rorc, mapk10, glp1r, egfr, pgr, mgll) were identified using WGCNA crossover analysis. Subsequent functional enrichment analyses were conducted. Our findings confirm a significant correlation between key cluster gene expression and immune cell infiltration in tamoxifen-resistant and -sensitized patients. scRNA-seq analysis revealed high expression of key cluster genes in epithelial cells, suggesting artemisinin's specific impact on tumor cells in estrogen receptor (ER)-positive BC tissues. Molecular target docking and in vitro experiments with artemisinin on LCC9 cells demonstrated a reversal effect in reducing migratory and drug resistance of drug-resistant cells by modulating relevant drug resistance genes. These results indicate that artemisinin could potentially reverse tamoxifen resistance in ER-positive breast cancer.


Sujet(s)
Artémisinines , Tumeurs du sein , Biologie informatique , Résistance aux médicaments antinéoplasiques , Récepteurs des oestrogènes , Tamoxifène , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Humains , Artémisinines/pharmacologie , Artémisinines/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Résistance aux médicaments antinéoplasiques/génétique , Biologie informatique/méthodes , Récepteurs des oestrogènes/métabolisme , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Simulation de docking moléculaire , Prolifération cellulaire/effets des médicaments et des substances chimiques
7.
Life Sci ; 350: 122763, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823505

RÉSUMÉ

AIMS: The intricate molecular mechanisms underlying estrogen receptor-positive (ER+) breast carcinogenesis and resistance to endocrine therapy remain elusive. In this study, we elucidate the pivotal role of GPR81, a G protein-coupled receptor, in ER+ breast cancer (BC) by demonstrating low expression of GPR81 in tamoxifen (TAM)-resistant ER+ BC cell lines and tumor samples, along with the underlying molecular mechanisms. MAIN METHODS: Fatty acid oxidation (FAO) levels and lipid accumulation were explored using MDA and FAßO assay, BODIPY 493/503 staining, and Lipid TOX staining. Autophagy levels were assayed using CYTO-ID detection and Western blotting. The impact of GPR81 on TAM resistance in BC was investigated through CCK8 assay, colony formation assay and a xenograft mice model. RESULTS: Aberrantly low GPR81 expression in TAM-resistant BC cells disrupts the Rap1 pathway, leading to the upregulation of PPARα and CPT1. This elevation in PPARα/CPT1 enhances FAO, impedes lipid accumulation and lipid droplet (LD) formation, and subsequently inhibits cell autophagy, ultimately promoting TAM-resistant BC cell growth. Moreover, targeting GPR81 and FAO emerges as a promising therapeutic strategy, as the GPR81 agonist and the CPT1 inhibitor etomoxir effectively inhibit ER+ BC cell and tumor growth in vivo, re-sensitizing TAM-resistant ER+ cells to TAM treatment. CONCLUSION: Our data highlight the critical and functionally significant role of GPR81 in promoting ER+ breast tumorigenesis and resistance to endocrine therapy. GPR81 and FAO levels show potential as diagnostic biomarkers and therapeutic targets in clinical settings for TAM-resistant ER+ BC.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Acides gras , Souris nude , Oxydoréduction , Récepteur PPAR alpha , Récepteurs couplés aux protéines G , Tamoxifène , Tamoxifène/pharmacologie , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Femelle , Récepteurs couplés aux protéines G/métabolisme , Animaux , Acides gras/métabolisme , Souris , Récepteur PPAR alpha/métabolisme , Antinéoplasiques hormonaux/pharmacologie , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Autophagie/effets des médicaments et des substances chimiques , Souris de lignée BALB C
8.
Sci Rep ; 14(1): 12542, 2024 05 31.
Article de Anglais | MEDLINE | ID: mdl-38822093

RÉSUMÉ

Around 75% of breast cancer (BC) patients have tumors expressing the predictive biomarker estrogen receptor α (ER) and are offered endocrine therapy. One-third eventually develop endocrine resistance, a majority with retained ER expression. Mutations in the phosphatidylinositol bisphosphate 3-kinase (PI3K) catalytic subunit encoded by PIK3CA is a proposed resistance mechanism and a pharmacological target in the clinical setting. Here we explore the frequency of PIK3CA mutations in endocrine-resistant BC before and during treatment and correlate to clinical features. Patients with ER-positive (ER +), human epidermal growth factor receptor 2 (HER2)-negative primary BC with an ER + relapse within 5 years of ongoing endocrine therapy were retrospectively assessed. Tissue was collected from primary tumors (n = 58), relapse tumors (n = 54), and tumor-free lymph nodes (germline controls, n = 62). Extracted DNA was analyzed through panel sequencing. Somatic mutations were observed in 50% (31/62) of the patients, of which 29% occurred outside hotspot regions. The presence of PIK3CA mutations was significantly associated with nodal involvement and mutations were more frequent in relapse than primary tumors. Our study shows the different PIK3CA mutations in endocrine-resistant BC and their fluctuations during therapy. These results may aid investigations of response prediction, facilitating research deciphering the mechanisms of endocrine resistance.


Sujet(s)
Tumeurs du sein , Phosphatidylinositol 3-kinases de classe I , Résistance aux médicaments antinéoplasiques , Mutation , Humains , Phosphatidylinositol 3-kinases de classe I/génétique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Résistance aux médicaments antinéoplasiques/génétique , Adulte d'âge moyen , Sujet âgé , Adulte , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Études rétrospectives , Sujet âgé de 80 ans ou plus , Récepteur alpha des oestrogènes/génétique , Récepteur alpha des oestrogènes/métabolisme , Récidive tumorale locale/génétique , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme
9.
Aust J Gen Pract ; 53(5): 291-300, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38697060

RÉSUMÉ

BACKGROUND: Prostate cancer (PCa) is the most common malignancy after skin cancer in men in Australia. Its management varies according to tumour stage. Due to the significant dependence on androgen receptor signalling, agents that interfere with this pathway (most commonly medical castration in the form of androgen deprivation therapy [ADT]) are the mainstay treatment of advanced disease. OBJECTIVE: This review provides a contemporary update on ADT, with further discussion of emerging novel therapies for primary care. DISCUSSION: ADT is currently indicated for the treatment of metastatic prostate cancer, disease recurrence following attempted local curative therapy, as well as combined use with radiotherapy for intermediate/high-risk disease. There has been rapid development of new pharmaceuticals targeting the androgen receptor. These are reviewed historically with an emphasis placed on emerging therapies, their common side effects, and how to manage them in the general practice setting.


Sujet(s)
Antagonistes des androgènes , Tumeurs de la prostate , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Antagonistes des androgènes/usage thérapeutique , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Australie
10.
Expert Opin Drug Metab Toxicol ; 20(6): 491-502, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38778707

RÉSUMÉ

INTRODUCTION: The therapeutic scenario of metastatic hormone-sensitive prostate cancer (mHSPC) has dramatically changed in recent years, with the approval of new-generation Androgen Receptor Signaling Inhibitors (ARSIs), in combination with the androgen deprivation therapy (ADT), which was the previous standard of care. Despite showing a similar clinical efficacy, ARSIs, all of which are administered orally, are different in terms of pharmacokinetic and drug-drug interactions (DDIs). AREAS COVERED: This review covers the main pharmacokinetic characteristics of ARSIs that have been approved for the first-line therapy of mHSPC patients, underlying the differences among these molecules and focusing on the known or possible interactions with other drugs. Full-text articles and abstracts were searched in PubMed. EXPERT OPINION: Since prostate cancer occurs mainly in older age, comorbidities and the consequent polypharmacy increase the DDI risk in mHSPC patients who are candidates for ARSI. Waiting for new therapeutic options, in the absence of direct comparisons, pharmacokinetic knowledge is essential to guide clinicians in prescribing ARSI in this setting.


Sujet(s)
Antagonistes du récepteur des androgènes , Interactions médicamenteuses , Métastase tumorale , Tumeurs de la prostate , Transduction du signal , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Antagonistes du récepteur des androgènes/pharmacocinétique , Antagonistes du récepteur des androgènes/administration et posologie , Antagonistes du récepteur des androgènes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Polypharmacie , Sujet âgé , Antagonistes des androgènes/administration et posologie , Antagonistes des androgènes/pharmacocinétique , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/effets indésirables , Administration par voie orale , Antinéoplasiques hormonaux/pharmacocinétique , Antinéoplasiques hormonaux/administration et posologie , Antinéoplasiques hormonaux/pharmacologie , Animaux
11.
Biochem Pharmacol ; 225: 116256, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38729448

RÉSUMÉ

Endocrine treatment, particularly tamoxifen, has shown significant improvement in the prognosis of patients with estrogen receptor-positive (ER-positive) breast cancer. However, the clinical utility of this treatment is often hindered by the development of endocrine resistance. Therefore, a comprehensive understanding of the underlying mechanisms driving ER-positive breast cancer carcinogenesis and endocrine resistance is crucial to overcome this clinical challenge. In this study, we investigated the expression of MICAL-L2 in ER-positive breast cancer and its impact on patient prognosis. We observed a significant upregulation of MICAL-L2 expression in ER-positive breast cancer, which correlated with a poorer prognosis in these patients. Furthermore, we found that estrogen-ERß signaling promoted the expression of MICAL-L2. Functionally, our study demonstrated that MICAL-L2 not only played an oncogenic role in ER-positive breast cancer tumorigenesis but also influenced the sensitivity of ER-positive breast cancer cells to tamoxifen. Mechanistically, as an estrogen-responsive gene, MICAL-L2 facilitated the activation of the AKT/mTOR signaling pathway in ER-positive breast cancer cells. Collectively, our findings suggest that MICAL-L2 could serve as a potential prognostic marker for ER-positive breast cancer and represent a promising molecular target for improving endocrine treatment and developing therapeutic approaches for this subtype of breast cancer.


Sujet(s)
Antinéoplasiques hormonaux , Tumeurs du sein , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Tamoxifène , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Humains , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Animaux , Oestrogènes/pharmacologie , Oestrogènes/métabolisme , Souris nude , Souris , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Cellules MCF-7 , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/physiologie , Évolution de la maladie , Lignée cellulaire tumorale , Souris de lignée BALB C
12.
Cancer Lett ; 593: 216968, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38788968

RÉSUMÉ

In patients with ER + metastatic breast cancer (mBC), the first-line treatment involves the combination of endocrine therapy (ET) and CDK4/6 inhibitors (CDK4/6i). However, a significant group of patients experiences disease progression, emphasizing the urgent clinical need to identify novel anti-tumor therapies. We previously generated breast cancer cells resistant to the combination of fulvestrant (ER downregulator) and abemaciclib (CDK4/6 inhibitor) from MCF7 and T47D (MCF7-FAR and T47D-FAR). RNA-seq-based Gene Set Enrichment Analysis (GSEA) revealed hyper-activation of EGFR, HER2, and AKT signaling in both MCF7-FAR and T47D-FAR. Modulating EGFR or ERBB2 expression through loss- and gain-of-function experiments altered tumor sensitivity to fulvestrant and abemaciclib in parental and FAR spheroids, affecting ERK and AKT/S6 pathways. Cetuximab treatment overcame tumor resistance to fulvestrant and abemaciclib in FAR and EGFR-overexpressing breast cancer spheroids and xenografts. Likewise, patient-derived organoids (PDOs) from individuals with ER + mBC, progressing on palbociclib, exhibited up-regulation of EGFR and HER2 pathways. In conclusion, our findings suggest that inhibiting EGFR and HER2 pathways might overcome resistance to ET + CDK4/6i in selected patients with ER + mBC.


Sujet(s)
Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Femelle , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Animaux , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Récepteurs des oestrogènes/métabolisme , Souris , Fulvestrant/pharmacologie , Fulvestrant/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Benzimidazoles/pharmacologie , Aminopyridines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Cellules MCF-7 , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
13.
Cancer Discov ; 14(5): 704-706, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690600

RÉSUMÉ

SUMMARY: Rosano, Sofyali, Dhiman, and colleagues show that epigenetic-related changes occur in endocrine therapy (ET)-induced dormancy in estrogen receptor positive (ER+) breast cancer, as well as in its reawakening. Targeting these epigenetic changes blocks the entrance to dormancy and reduces the persister cancer cell population, enhancing the cytotoxic effects of ET in vitro. See related article by Rosano et al., p. 866 (9).


Sujet(s)
Antinéoplasiques hormonaux , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Humains , Épigenèse génétique/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Femelle , Récepteurs des oestrogènes/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
14.
Front Endocrinol (Lausanne) ; 15: 1365321, 2024.
Article de Anglais | MEDLINE | ID: mdl-38779454

RÉSUMÉ

Background: Adrenocortical carcinoma (ACC) is an aggressive endocrine malignancy with limited therapeutic options. Treating advanced ACC with mitotane, the cornerstone therapy, remains challenging, thus underscoring the significance to predict mitotane response prior to treatment and seek other effective therapeutic strategies. Objective: We aimed to determine the efficacy of mitotane via an in vitro assay using patient-derived ACC cells (PDCs), identify molecular biomarkers associated with mitotane response and preliminarily explore potential agents for ACC. Methods: In vitro mitotane sensitivity testing was performed in 17 PDCs and high-throughput screening against 40 compounds was conducted in 8 PDCs. Genetic features were evaluated in 9 samples using exomic and transcriptomic sequencing. Results: PDCs exhibited variable sensitivity to mitotane treatment. The median cell viability inhibition rate was 48.4% (IQR: 39.3-59.3%) and -1.2% (IQR: -26.4-22.1%) in responders (n=8) and non-responders (n=9), respectively. Median IC50 and AUC were remarkably lower in responders (IC50: 53.4 µM vs 74.7 µM, P<0.0001; AUC: 158.0 vs 213.5, P<0.0001). Genomic analysis revealed CTNNB1 somatic alterations were only found in responders (3/5) while ZNRF3 alterations only in non-responders (3/4). Transcriptomic profiling found pathways associated with lipid metabolism were upregulated in responder tumors whilst CYP27A1 and ABCA1 expression were positively correlated to in vitro mitotane sensitivity. Furthermore, pharmacologic analysis identified that compounds including disulfiram, niclosamide and bortezomib exhibited efficacy against PDCs. Conclusion: ACC PDCs could be useful for testing drug response, drug repurposing and guiding personalized therapies. Our results suggested response to mitotane might be associated with the dependency on lipid metabolism. CYP27A1 and ABCA1 expression could be predictive markers for mitotane response, and disulfiram, niclosamide and bortezomib could be potential therapeutics, both warranting further investigation.


Sujet(s)
Tumeurs corticosurrénaliennes , Carcinome corticosurrénalien , Antinéoplasiques hormonaux , Mitotane , Test pharmacogénomique , Humains , Mitotane/usage thérapeutique , Carcinome corticosurrénalien/traitement médicamenteux , Carcinome corticosurrénalien/génétique , Carcinome corticosurrénalien/anatomopathologie , Carcinome corticosurrénalien/métabolisme , Tumeurs corticosurrénaliennes/traitement médicamenteux , Tumeurs corticosurrénaliennes/génétique , Tumeurs corticosurrénaliennes/anatomopathologie , Tumeurs corticosurrénaliennes/métabolisme , Femelle , Mâle , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Adulte d'âge moyen , Adulte , Sujet âgé , Pharmacogénétique
15.
Sci China Life Sci ; 67(7): 1413-1426, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38565741

RÉSUMÉ

Endocrine therapy that blocks estrogen signaling is the most effective treatment for patients with estrogen receptor positive (ER+) breast cancer. However, the efficacy of agents such as tamoxifen (Tam) is often compromised by the development of resistance. Here we report that cytokines-activated nuclear IKKα confers Tam resistance to ER+ breast cancer by inducing the expression of FAT10, and that the expression of FAT10 and nuclear IKKα in primary ER+ human breast cancer was correlated with lymphotoxin ß (LTB) expression and significantly associated with relapse and metastasis in patients treated with adjuvant mono-Tam. IKKα activation or enforced FAT10 expression promotes Tam-resistance while loss of IKKα or FAT10 augments Tam sensitivity. The induction of FAT10 by IKKα is mediated by the transcription factor Pax5, and coordinated via an IKKα-p53-miR-23a circuit in which activation of IKKα attenuates p53-directed repression of FAT10. Thus, our findings establish IKKα-to-FAT10 pathway as a new therapeutic target for the treatment of Tam-resistant ER+ breast cancer.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , I-kappa B Kinase , Transduction du signal , Tamoxifène , Animaux , Femelle , Humains , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Cytokines/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , I-kappa B Kinase/métabolisme , Cellules MCF-7 , Transduction du signal/effets des médicaments et des substances chimiques , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique
16.
Clin Cancer Res ; 30(14): 2884-2894, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38656833

RÉSUMÉ

Adjuvant endocrine therapy (ET) represents the standard of care for almost all hormone receptor (HR)+/HER2- breast cancers, and different agents and durations are currently available. In this context, the tailoring and optimization of adjuvant endocrine treatment by reducing unnecessary toxic treatment while taking into account the biological heterogeneity of HR+/HER2- breast cancer represents a clinical priority. There is therefore a significant need for the integration of biological biomarkers in the choice of adjuvant ET beyond currently used clinicopathological characteristics. Several gene expression assays have been developed to identify patients with HR+/HER2- breast cancer who will not derive benefit from the addition of adjuvant chemotherapy. By enhancing risk stratification and predicting therapeutic response, genomic assays have also shown to be a promising tool for optimizing endocrine treatment decisions. In this study, we review evidence supporting the use of most common commercially available gene expression assays [Oncotype DX, MammaPrint, Breast Cancer Index (BCI), Prosigna, and EndoPredict] in tailoring adjuvant ET. Available data on the use of genomic tests to inform extended adjuvant treatment choice based on the risk of late relapse and on the estimated benefit of a prolonged ET are discussed. Moreover, preliminary evidence regarding the use of genomic assays to inform de-escalation of endocrine treatment, such as shorter durations or omission, for low-risk patients is reviewed. Overall, gene expression assays are emerging as potential tools to further personalize adjuvant treatment for patients with HR+/HER2- breast cancers.


Sujet(s)
Antinéoplasiques hormonaux , Marqueurs biologiques tumoraux , Tumeurs du sein , Récepteur ErbB-2 , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Traitement médicamenteux adjuvant/méthodes , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Marqueurs biologiques tumoraux/génétique , Analyse de profil d'expression de gènes , Récepteurs des oestrogènes/métabolisme , Récepteurs à la progestérone/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Médecine de précision/méthodes
17.
Cell Signal ; 119: 111184, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38640982

RÉSUMÉ

Estrogen receptor alpha (ERα) is expressed in approximately 70% of breast cancer cases and determines the sensitivity and effectiveness of endocrine therapy. 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase3 (PFKFB3) is a glycolytic enzyme that is highly expressed in a great many human tumors, and recent studies have shown that it plays a significant role in improving drug sensitivity. However, the role of PFKFB3 in regulating ERα expression and the underlying mechanism remains unclear. Here, we find by using immunohistochemistry (IHC) that PFKFB3 is elevated in ER-positive breast cancer and high expression of PFKFB3 resulted in a worse prognosis. In vitro and in vivo experiments verify that PFKFB3 promotes ER-positive breast cancer cell proliferation. The overexpression of PFKFB3 promotes the estrogen-independent ER-positive breast cancer growth. In an estrogen-free condition, RNA-sequencing data from MCF7 cells treated with siPFKFB3 showed enrichment of the estrogen signaling pathway, and a luciferase assay demonstrated that knockdown of PFKFB3 inhibited the ERα transcriptional activity. Mechanistically, down-regulation of PFKFB3 promotes STUB1 binding to ERα, which accelerates ERα degradation by K48-based ubiquitin linkage. Finally, growth of ER-positive breast cancer cells in vivo was more potently inhibited by fulvestrant combined with the PFKFB3 inhibitor PFK158 than for each drug alone. In conclusion, these data suggest that PFKFB3 is identified as an adverse prognosis factor for ER-positive breast cancer and plays a previously unrecognized role in the regulation of ERα stability and activity. Our results further explores an effective approach to improve fulvestrant sensitivity through the early combination with a PFKFB3 inhibitor.


Sujet(s)
Tumeurs du sein , Récepteur alpha des oestrogènes , Fulvestrant , Phosphofructokinase-2 , Humains , Phosphofructokinase-2/métabolisme , Phosphofructokinase-2/génétique , Récepteur alpha des oestrogènes/métabolisme , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Femelle , Fulvestrant/pharmacologie , Animaux , Stabilité protéique/effets des médicaments et des substances chimiques , Souris , Cellules MCF-7 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Carcinogenèse/métabolisme , Carcinogenèse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques hormonaux/pharmacologie , Lignée cellulaire tumorale
18.
Cancer Lett ; 590: 216847, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38583647

RÉSUMÉ

Tamoxifen (TAM) resistance presents a major clinical obstacle in the management of estrogen-sensitive breast cancer, highlighting the need to understand the underlying mechanisms and potential therapeutic approaches. We showed that dysregulated mitochondrial dynamics were involved in TAM resistance by protecting against mitochondrial apoptosis. The dysregulated mitochondrial dynamics were associated with increased mitochondrial fusion and decreased fission, thus preventing the release of mitochondrial cytochrome c to the cytoplasm following TAM treatment. Dynamin-related GTPase protein mitofusin 1 (MFN1), which promotes fusion, was upregulated in TAM-resistant cells, and high MFN1 expression indicated a poor prognosis in TAM-treated patients. Mitochondrial translocation of MFN1 and interaction between MFN1 and mitofusin 2 (MFN2) were enhanced to promote mitochondrial outer membrane fusion. The interaction of MFN1 and cristae-shaping protein optic atrophy 1 (OPA1) and OPA1 oligomerization were reduced due to augmented OPA1 proteolytic cleavage, and their apoptosis-promoting function was reduced due to cristae remodeling. Furthermore, the interaction of MFN1 and BAK were increased, which restrained BAK activation following TAM treatment. Knockdown or pharmacological inhibition of MFN1 blocked mitochondrial fusion, restored BAK oligomerization and cytochrome c release, and amplified activation of caspase-3/9, thus sensitizing resistant cells to apoptosis and facilitating the therapeutic effects of TAM both in vivo and in vitro. Conversely, overexpression of MFN1 alleviated TAM-induced mitochondrial apoptosis and promoted TAM resistance in sensitive cells. These results revealed that dysregulated mitochondrial dynamics contributes to the development of TAM resistance, suggesting that targeting MFN1-mediated mitochondrial fusion is a promising strategy to circumvent TAM resistance.


Sujet(s)
Apoptose , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , dGTPases , Dynamique mitochondriale , Tamoxifène , Humains , Tamoxifène/pharmacologie , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , dGTPases/génétique , dGTPases/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Animaux , Souris , Protéines de transport de la membrane mitochondriale/métabolisme , Protéines de transport de la membrane mitochondriale/génétique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Lignée cellulaire tumorale , Antinéoplasiques hormonaux/pharmacologie , Protéine Bak/métabolisme , Protéine Bak/génétique , Cellules MCF-7 , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
19.
BMC Mol Cell Biol ; 25(1): 12, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38649821

RÉSUMÉ

Hormone receptor (HR)-positive breast cancer can become aggressive after developing hormone-treatment resistance. This study elucidated the role of long non-coding RNA (lncRNA) SOX2OT in tamoxifen-resistant (TAMR) breast cancer and its potential interplay with the tumor microenvironment (TME). TAMR breast cancer cell lines TAMR-V and TAMR-H were compared with the luminal type A cell line (MCF-7). LncRNA expression was assessed via next-generation sequencing, RNA extraction, lncRNA profiling, and quantitative RT-qPCR. SOX2OT overexpression effects on cell proliferation, migration, and invasion were evaluated using various assays. SOX2OT was consistently downregulated in TAMR cell lines and TAMR breast cancer tissue. Overexpression of SOX2OT in TAMR cells increased cell proliferation and cell invasion. However, SOX2OT overexpression did not significantly alter SOX2 levels, suggesting an independent interaction within TAMR cells. Kaplan-Meier plot analysis revealed an inverse relationship between SOX2OT expression and prognosis in luminal A and B breast cancers. Our findings highlight the potential role of SOX2OT in TAMR breast cancer progression. The downregulation of SOX2OT in TAMR breast cancer indicates its involvement in resistance mechanisms. Further studies should explore the intricate interactions between SOX2OT, SOX2, and TME in breast cancer subtypes.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , ARN long non codant , Tamoxifène , Femelle , Humains , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/génétique , Résistance aux médicaments antinéoplasiques/génétique , Cellules MCF-7 , Invasion tumorale , Pronostic , ARN long non codant/génétique , ARN long non codant/métabolisme , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Microenvironnement tumoral/génétique
20.
Clin Breast Cancer ; 24(4): 368-375.e2, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38443227

RÉSUMÉ

BACKGROUND: Breast cancer, particularly the estrogen receptor positive (ER+) subtype, remains a leading cause of cancer-related death among women. Endocrine therapy is the most effective treatment for ER+ breast cancer; however, the development of resistance presents a significant challenge. This study explored the role of the breast cancer antiestrogen resistance 4 (BCAR4) gene as a potential driver of resistance and a pivotal biomarker in breast cancer. PATIENTS AND METHODS: The researchers undertook a comprehensive analysis of 1743 patients spanning 6 independent cohorts. They examined the association of BCAR4 expression with patient outcomes across all breast cancer types and the PAM50 molecular subtypes. The relationship between elevated BCAR4 expression and resistance to endocrine therapy including AIs, the prevailing standard-of-care for endocrine therapy, was also investigated. RESULTS: This meta-analysis corroborated the link between BCAR4 expression and adverse outcomes as well as resistance to endocrine therapy in breast cancer. Notably, BCAR4 expression is clinically significant in luminal A and B subtypes. Additionally, an association between BCAR4 expression and resistance to AI treatment was discerned. CONCLUSION: This study expands on previous findings by demonstrating that BCAR4 expression is associated with resistance to newer therapies. The identification of patients with intrinsic resistance to hormone therapy is crucial to avoid ineffective treatment strategies. These findings contribute to our understanding of endocrine therapy resistance in breast cancer and could potentially guide the development of more effective treatment strategies.


Sujet(s)
Antinéoplasiques hormonaux , Marqueurs biologiques tumoraux , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Résistance aux médicaments antinéoplasiques/génétique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Récepteurs des oestrogènes/métabolisme , Pronostic , Régulation de l'expression des gènes tumoraux , ARN long non codant
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...