Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.732
Filtrer
1.
Sci Rep ; 14(1): 16030, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992220

RÉSUMÉ

This study examines the biological effects of palbociclib and ribociclib in hormone receptor-positive breast cancer, pivotal to the HARMONIA prospective phase III clinical trial. We explore the downstream impacts of these CDK4/6 inhibitors, focusing on cell lines and patient-derived tumor samples. We treated HR+ breast cancer cell lines (T47D, MCF7, and BT474) with palbociclib or ribociclib (100 nM or 500 nM), alone or combined with fulvestrant (1 nM), over periods of 24, 72, or 144 h. Our assessments included PAM50 gene expression, RB1 phosphorylation, Lamin-B1 protein levels, and senescence-associated ß-galactosidase activity. We further analyzed PAM50 gene signatures from the CORALLEEN and NeoPalAna phase II trials. Both CDK4/6 inhibitors similarly inhibited proliferation across the cell lines. At 100 nM, both drugs partially reduced p-RB1, with further decreases at 500 nM over 144 h. Treatment led to reduced Lamin-B1 expression and increased senescence-associated ß-galactosidase activity. Both drugs enhanced Luminal A and reduced Luminal B and proliferation signatures at both doses. However, the HER2-enriched signature significantly diminished only at the higher dose of 500 nM. Corresponding changes were observed in tumor samples from the CORALLEEN and NeoPalAna studies. At 2 weeks of treatment, both drugs significantly reduced the HER2-enriched signature, but at surgery, this reduction was consistent only with ribociclib. Our findings suggest that while both CDK4/6 inhibitors effectively modulate key biological pathways in HR+/HER2- breast cancer, nuances in their impact, particularly on the HER2-enriched signature, are dose-dependent, influenced by the addition of fulvestrant and warrant further investigation.


Sujet(s)
Aminopyridines , Tumeurs du sein , Prolifération cellulaire , Pipérazines , Purines , Pyridines , Humains , Aminopyridines/pharmacologie , Pipérazines/pharmacologie , Purines/pharmacologie , Pyridines/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteurs des oestrogènes/métabolisme , Fulvestrant/pharmacologie , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Kinase-4 cycline-dépendante/métabolisme , Récepteurs à la progestérone/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Kinase-6 cycline-dépendante/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
2.
J Cell Biochem ; 125(7): e30610, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38860517

RÉSUMÉ

17ß-estradiol is a hormone that plays a vital role in human physiology. It acts through estrogen receptors, specifically estrogen receptor α and estrogen receptor ß, and its action is determined by the pulsatile secretion in the bloodstream. 17ß-estradiol affects cell proliferation, and dysregulation of 17ß-estradiol:estrogen receptor α signaling contribute to the development of breast cancer. Previous research on 17ß-estradiol:estrogen receptor α signaling has primarily used two-dimensional cell cultures, which do not fully recapitulate the complexity of tumors that exist in a three-dimensional environment and do not consider the pulsatile nature of this hormone. To address these limitations, we studied 17ß-estradiol:estrogen receptor α signaling in cell proliferation using both two-dimensional and three-dimensional breast cancer cell culture models under continuous and pulsatile stimulation conditions. Results revealed that breast cancer cells grown in an alginate-based three-dimensional matrix exhibited similar responsiveness to 17ß-estradiol compared with cells grown in conventional two-dimensional culture plates. 17ß-estradiol induced the expression of proteins containing estrogen response element in the three-dimensional model. The efficacy of the antiestrogen drugs fulvestrant (ICI182,280) and 4OH-tamoxifen was also demonstrated in the three-dimensional model. These results support the use of the three-dimensional culture model for studying tumor response to drugs and provide a more realistic microenvironment for such studies. Furthermore, the study revealed that a brief 5-min exposure to 17ß-estradiol triggered a physiological response comparable with continuous hormone exposure, suggesting that the cellular response to 17ß-estradiol is more important than the continuous presence of the hormone. In conclusion, the study demonstrates that the alginate-based three-dimensional culture model is suitable for studying the effects of 17ß-estradiol and antiestrogen drugs on breast cancer cells, offering a more realistic representation of tumor-microenvironment interactions. The results also highlight the importance of considering the physiological importance of the temporal dynamics in studying 17ß-estradiol signaling and cellular responses.


Sujet(s)
Prolifération cellulaire , Oestradiol , Récepteur alpha des oestrogènes , Transduction du signal , Humains , Oestradiol/pharmacologie , Récepteur alpha des oestrogènes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Cellules MCF-7 , Techniques de cultures cellulaires tridimensionnelles/méthodes , Techniques de culture cellulaire/méthodes , Fulvestrant/pharmacologie
3.
ESMO Open ; 9(6): 103465, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38833970

RÉSUMÉ

BACKGROUND: In most patients with advanced human epidermal growth factor receptor-2-positive (HER2+) breast cancer, anti-HER2 therapies fail due to the development of acquired resistance, potentially mediated through phosphoinositide-3-kinase (PI3K) signaling. We investigated adding taselisib, an α-selective potent oral inhibitor of PI3K, to different HER2-directed regimens in order to improve disease control. PATIENTS AND METHODS: Patients (n = 68) with advanced HER2+ breast cancer were enrolled to this open-label, dose-escalation phase Ib study. The primary endpoint was defining the maximal tolerated dose (MTD) for the various taselisib-containing combinations. The secondary endpoint was safety. Exploratory endpoints included circulating tumor DNA analysis. The study included four cohorts: (A) taselisib + trastuzumab emtansine (T-DM1), (C) taselisib + trastuzumab and pertuzumab (TP), (D) taselisib + TP + paclitaxel, and (E) taselisib + TP + fulvestrant. RESULTS: Following dose escalation, the taselisib MTD was defined as 4 mg once daily. Treatment was associated with significant toxicities, as 34 out of 68 patients experienced grade ≥3 adverse events (AEs) attributed to taselisib, the most common all-grade AEs being diarrhea, fatigue, and oral mucositis. At a median follow-up of 43.8 months, median progression-free survival (PFS) for the MTD-treated population in cohorts A, C, and E was 6.3 [95% confidence interval (CI) 3.2-not applicable (NA)] months, 1.7 (95% CI 1.4-NA) months, and 10.6 (95% CI 8.3-NA) months, respectively. The median PFS for patients in cohort A with prior T-DM1 use was 10.4 (95% CI 2.7-NA) months. CONCLUSIONS: PIK3CA targeting with taselisib in combination with HER2-targeted therapies was associated with both promising efficacy and substantial toxicities.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Dose maximale tolérée , Récepteur ErbB-2 , Humains , Femelle , Adulte d'âge moyen , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Récepteur ErbB-2/métabolisme , Sujet âgé , Adulte , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Oxazoles/usage thérapeutique , Oxazoles/pharmacologie , Oxazoles/administration et posologie , Quinazolines/usage thérapeutique , Quinazolines/pharmacologie , Quinazolines/administration et posologie , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Paclitaxel/administration et posologie , Uracile/analogues et dérivés , Uracile/pharmacologie , Uracile/usage thérapeutique , Uracile/administration et posologie , Ado-trastuzumab emtansine/usage thérapeutique , Ado-trastuzumab emtansine/pharmacologie , Fulvestrant/pharmacologie , Fulvestrant/usage thérapeutique , Fulvestrant/administration et posologie , Trastuzumab/usage thérapeutique , Trastuzumab/pharmacologie , Imidazoles , Oxazépines , Anticorps monoclonaux humanisés
4.
Breast Cancer Res ; 26(1): 95, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849889

RÉSUMÉ

BACKGROUND: Breast cancers treated with aromatase inhibitors (AIs) can develop AI resistance, which is often driven by estrogen receptor-alpha (ERα/ESR1) activating mutations, as well as by ER-independent signaling pathways. The breast ER antagonist lasofoxifene, alone or combined with palbociclib, elicited antitumor activities in a xenograft model of ER + metastatic breast cancer (mBC) harboring ESR1 mutations. The current study investigated the activity of LAS in a letrozole-resistant breast tumor model that does not have ESR1 mutations. METHODS: Letrozole-resistant, MCF7 LTLT cells tagged with luciferase-GFP were injected into the mammary duct inguinal glands of NSG mice (MIND model; 6 mice/group). Mice were randomized to vehicle, lasofoxifene ± palbociclib, fulvestrant ± palbociclib, or palbociclib alone 2-3 weeks after cell injections. Tumor growth and metastases were monitored with in vivo and ex vivo luminescence imaging, terminal tumor weight measurements, and histological analysis. The experiment was repeated with the same design and 8-9 mice in each treatment group. RESULTS: Western blot analysis showed that the MCF7 LTLT cells had lower ERα and higher HER2 expressions compared with normal MCF7 cells. Lasofoxifene ± palbociclib, but not fulvestrant, significantly reduced primary tumor growth versus vehicle as assessed by in vivo imaging of tumors at study ends. Percent tumor area in excised mammary glands was significantly lower for lasofoxifene plus palbociclib versus vehicle. Ki67 staining showed decreased overall tumor cell proliferation with lasofoxifene ± palbociclib. The lasofoxifene + palbociclib combination was also associated with significantly fewer bone metastases compared with vehicle. Similar results were observed in the repeat experiment. CONCLUSIONS: In a mouse model of letrozole-resistant breast cancer with no ESR1 mutations, reduced levels of ERα, and overexpression of HER2, lasofoxifene alone or combined with palbociclib inhibited primary tumor growth more effectively than fulvestrant. Lasofoxifene plus palbociclib also reduced bone metastases. These results suggest that lasofoxifene alone or combined with a CDK4/6 inhibitor may offer benefits to patients who have ER-low and HER2-positive, AI-resistant breast cancer, independent of ESR1 mutations.


Sujet(s)
Inhibiteurs de l'aromatase , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Pyrrolidines , 1,2,3,4-Tétrahydro-naphtalènes , Animaux , Femelle , Humains , Souris , Inhibiteurs de l'aromatase/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Récepteur alpha des oestrogènes/génétique , Fulvestrant/pharmacologie , Létrozole/pharmacologie , Cellules MCF-7 , Pipérazines/pharmacologie , Pyridines/pharmacologie , Pyrrolidines/pharmacologie , 1,2,3,4-Tétrahydro-naphtalènes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
J Geriatr Oncol ; 15(6): 101813, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38852379

RÉSUMÉ

INTRODUCTION: Palbociclib is a widely used treatment for advanced breast cancer in older adults. However, the existing evidence regarding its safety and tolerability in this age group is inconsistent and limited to retrospective subgroup or pooled analyses. MATERIALS AND METHODS: We conducted a prospective single-arm multicenter phase 2 study to evaluate the safety and tolerability of palbociclib in participants aged 70 years or older with advanced hormone receptor-positive breast cancer. Participants were given palbociclib in combination with their physician's choice of endocrine therapy (letrozole or fulvestrant). The primary endpoint was the incidence of grade 3+ adverse events (AEs) by six months. Secondary endpoints included AE-related dose delays, dose reductions, early discontinuations, and hospitalizations. Additionally, we compared these endpoints by age groups (70-74 and ≥ 75 years). RESULTS: Of the 90 participants (median age 74 years [70-87]) enrolled, 75.6% (95% confidence interval [CI], 65.4-84.0) had grade 3+ AEs by six months. The most frequent grade 3+ AEs were neutropenia (61%), fatigue (4%), and nausea (3%). Febrile neutropenia was uncommon (1.1%). Due to AEs, 36% had dose delays, 34% had dose reductions, 10% had early discontinuations, and 10% had hospitalizations. Compared to those aged 70-74 years, participants aged ≥75 years had higher rates of early discontinuations (5.9% vs 15.9%, a difference of 9.5% [95% CI 3.5%-22.5%]). DISCUSSION: Palbociclib has an overall favorable safety profile in adults aged ≥70 with advanced breast cancer. However, adults ≥75 years had a trend toward higher rates of AE-related early discontinuations compared to those 70-74 years. Further research is needed to evaluate tolerability and improve the delivery of palbociclib in older adults. CLINICALTRIALS: gov:NCT03633331.


Sujet(s)
Tumeurs du sein , Pipérazines , Pyridines , Humains , Pyridines/effets indésirables , Pyridines/administration et posologie , Pyridines/usage thérapeutique , Sujet âgé , Femelle , Tumeurs du sein/traitement médicamenteux , Pipérazines/effets indésirables , Pipérazines/administration et posologie , Pipérazines/usage thérapeutique , Sujet âgé de 80 ans ou plus , Études prospectives , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Fulvestrant/administration et posologie , Fulvestrant/usage thérapeutique , Létrozole/administration et posologie , Létrozole/usage thérapeutique , Facteurs âges
6.
Breast ; 76: 103761, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38880077

RÉSUMÉ

BACKGROUND: The capsule formulation of CDK4/6 inhibitor palbociclib has reduced solubility at gastric pH > 4.5 and may have decreased activity when used with proton-pump inhibitors (PPI). Herein, we report the effect of PPI on palbociclib capsule activity and safety in the PARSIFAL study. METHODS: First-line endocrine-sensitive, hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC) patients received palbociclib capsules plus fulvestrant or letrozole. The primary endpoint was progression-free survival (PFS). This post-hoc analysis compared PPI use. Patients were PPI-naïve (N-PPI) if not on PPI during the study, and either early (E-PPI) or long-term PPI (LT-PPI) if on PPI at study entry or for at least ≥⅔ of treatment, respectively. PPI groups were not mutually exclusive. RESULTS: Among 486 patients, 66.9 % were N-PPI, 13.2 % E-PPI, 18.7 % LT-PPI, and 11.5 % of the PPI users were defined as neither. Median PFS (mPFS) was 29.6 months in the study population, 28.7 months in N-PPI, 23.0 months in E-PPI (Hazard Ratio [HR] 1.5; 95%Confidence Interval [CI] 1.1-2.2; p = 0.024), and 23.0 months in LT-PPI (HR 1.4; 95%CI 1.0-1.9; p = 0.035). By landmark analysis, PPI use was associated with poorer mPFS at 3 and 12 months. Grade ≥3 hematological adverse events occurred in 71.7 % of N-PPI, 57.8 % of E-PPI (p = 0.021), and 54.9 % of LT-PPI (p = 0.003). Dose reductions and dosing delays due to hematological toxicity occurred in 70.8 % of N-PPI, 56.3 % of E-PPI (p = 0.018), and 52.7 % of LT-PPI (p = 0.002). CONCLUSIONS: PPI use may reduce palbociclib capsule toxicity, dose modifications, and clinical activity in HR+/HER2- ABC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Fulvestrant , Létrozole , Pipérazines , Inhibiteurs de la pompe à protons , Pyridines , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Pyridines/administration et posologie , Pyridines/usage thérapeutique , Femelle , Pipérazines/administration et posologie , Pipérazines/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Adulte d'âge moyen , Sujet âgé , Récepteur ErbB-2/métabolisme , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Létrozole/administration et posologie , Inhibiteurs de la pompe à protons/administration et posologie , Adulte , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/analyse , Fulvestrant/administration et posologie , Fulvestrant/usage thérapeutique , Survie sans progression , Récepteurs à la progestérone/métabolisme , Sujet âgé de 80 ans ou plus
8.
NEJM Evid ; 3(5): EVIDoa2300231, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38815172

RÉSUMÉ

BACKGROUND: In estrogen receptor-positive metastatic breast cancer, ESR1 mutations (ESR1m) are a common mechanism of acquired resistance to aromatase inhibitors (ArIh). However, the impact ESR1 alterations have on CDK4/6 inhibitor (CDK4/6i) sensitivity has not been established. Analyses of CDK4/6i trials suggest that the endocrine therapy partner and specific ESR1 allele may affect susceptibility. We analyzed a real-world data set to investigate CDK4/6i efficacy in ESR1m metastatic breast cancer and associated clinical factors. METHODS: ESR1m were identified by analysis of circulating-tumor deoxyribonucleic acid. The GuardantINFORM database contains genomic information from tumors linked with claims data. Patients who started a CDK4/6i within 30 days of sequencing were categorized as having ESR1m or non-ESR1-mutant (non-ESR1m) breast cancer. Data were analyzed to determine the real-world time-to-next-treatment, defined as the start of a breast cancer treatment to initiation of the subsequent treatment. RESULTS: One hundred forty-five patients with ESR1m and 612 with non-ESR1m metastatic breast cancer were analyzed. ESR1m and non-ESR1m tumors had similar real-world time-to-next-treatment on CDK4/6i regimens (hazard ratio, 1.02; 95% confidence interval, 0.82 to 1.23). Duration on therapy in the first-line and second-line plus treatment settings were comparable regardless of ESR1 status. We stratified treatment duration by concurrent endocrine therapy, and patients with ESR1m had worse outcomes on ArIh but comparable real-world time-to-next-treatment on fulvestrant. CONCLUSIONS: These data suggest ESR1 variants are not associated with pan-CDK4/6i resistance and are consistent with the hypothesis that CDK4/6 blockade combined with a selective estrogen receptor degrader is potentially an effective option for ESR1m metastatic breast cancer.


Sujet(s)
Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Récepteur alpha des oestrogènes , Mutation , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Récepteur alpha des oestrogènes/génétique , Adulte d'âge moyen , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Sujet âgé , Adulte , Inhibiteurs de l'aromatase/usage thérapeutique , Pipérazines/usage thérapeutique , Métastase tumorale , Fulvestrant/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique
9.
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
10.
Breast Cancer Res Treat ; 206(3): 551-559, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38703285

RÉSUMÉ

PURPOSE: Everolimus in combination with endocrine therapy (ET) was formerly approved as 2nd-line therapy in HR(+)/HER2(-) advanced breast cancer (aBC) patients (pts) progressing during or after a non-steroidal aromatase inhibitor (NSAI). Since this approval, the treatment landscape of aBC has changed dramatically, particularly with the arrival of CDK 4-6 inhibitors. Endocrine monotherapy after progression to CDK4/6 inhibitors has shown a limited progression-free survival (PFS), below 3 months. Evidence of the efficacy of everolimus plus ET after CDK4/6 inhibitors is scarce. METHODS: A retrospective observational study of patients with aBC treated with everolimus and ET beyond CDK4/6-i progression compiled from February 2015 to December 2022 in 4 Spanish hospitals was performed. Clinical and demographic data were collected from medical records. The main objective was to estimate the median progression-free survival (mPFS). Everolimus adverse events (AE) were registered. Quantitative variables were summarized with medians; qualitative variables with proportions and the Kaplan-Meier method were used for survival estimates. RESULTS: One hundred sixty-one patients received everolimus plus ET (exemestane: 96, fulvestrant: 54, tamoxifen: 10, unknown: 1) after progressing on a CDK4/6 inhibitor. The median follow-up time was 15 months (interquartile range: 1-56 months). The median age at diagnosis was 49 years (range: 35-90 years). The estimated mPFS was 6.0 months (95%CI 5.3-7.8 months). PFS was longer in patients with previous CDK4/6 inhibitor therapy lasting for > 18 months (8.7 months, 95%CI 6.6-11.3 months), in patients w/o visceral metastases (8.0 months, 95%CI 5.8-10.5 months), and chemotherapy-naïve in the metastatic setting (7.2 months, 95%CI 5.9-8.4 months). CONCLUSION: This retrospective analysis cohort of everolimus plus ET in mBC patients previously treated with a CDK4/6 inhibitor suggests a longer estimated mPFS when compared with the mPFS with ET monotherapy obtained from current randomized clinical data. Everolimus plus ET may be considered as a valid control arm in novel clinical trial designs.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Évérolimus , Récepteur ErbB-2 , Humains , Évérolimus/administration et posologie , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/mortalité , Tumeurs du sein/métabolisme , Adulte d'âge moyen , Sujet âgé , Études rétrospectives , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Adulte , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Récepteur ErbB-2/métabolisme , Récepteurs des oestrogènes/métabolisme , Sujet âgé de 80 ans ou plus , Récepteurs à la progestérone/métabolisme , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/effets indésirables , Tamoxifène/usage thérapeutique , Tamoxifène/administration et posologie , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/administration et posologie , Inhibiteurs de l'aromatase/usage thérapeutique , Inhibiteurs de l'aromatase/administration et posologie , Fulvestrant/administration et posologie , Fulvestrant/usage thérapeutique , Survie sans progression , Androstadiènes/administration et posologie , Androstadiènes/usage thérapeutique , Évolution de la maladie
11.
Sci Rep ; 14(1): 11367, 2024 05 18.
Article de Anglais | MEDLINE | ID: mdl-38762547

RÉSUMÉ

Fulvestrant, as the first selective estrogen receptor degrader, is widely used in the endocrine treatment of breast cancer. However, in the real world, there is a lack of relevant reports on adverse reaction data mining for fulvestrant. To perform data mining on adverse events (AEs) associated with fulvestrant and explore the risk factors contributing to severe AEs, providing a reference for the rational use of fulvestrant in clinical practice. Retrieved adverse event report information associated with fulvestrant from the U.S. Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database, covering the period from market introduction to September 30, 2023. Suspicious AEs were screened using the reporting odds ratio (ROR) and proportional reporting ratio methods based on disproportionality analysis. Univariate and multivariate logistic regression analyses were conducted on severe AEs to explore the risk factors associated with fulvestrant-induced severe AEs. A total of 6947 reports related to AEs associated with fulvestrant were obtained, including 5924 reports of severe AEs and 1023 reports of non-severe AEs. Using the disproportionality analysis method, a total of 210 valid AEs were identified for fulvestrant, with 45 AEs (21.43%) not listed in the product labeling, involving 11 systems and organs. The AEs associated with fulvestrant were sorted by frequency of occurrence, with neutropenia (325 cases) having the highest number of reports. By signal strength, injection site pruritus showed the strongest signal (ROR = 658.43). The results of the logistic regression analysis showed that concurrent use of medications with extremely high protein binding (≥ 98%) is an independent risk factor for severe AEs associated with fulvestrant. Age served as a protective factor for fulvestrant-related AEs. The co-administration of fulvestrant with CYP3A4 enzyme inhibitors did not show statistically significant correlation with the occurrence of severe AEs. Co-administration of drugs with extremely high protein binding (≥ 98%) may increase the risk of severe adverse reactions of fulvestrant. Meanwhile, age (60-74 years) may reduce the risk of severe AEs of fulvestrant. However, further clinical research is still needed to explore and verify whether there is interaction between fulvestrant and drugs with high protein binding through more clinical studies.


Sujet(s)
Systèmes de signalement des effets indésirables des médicaments , Fouille de données , Bases de données factuelles , Fulvestrant , Food and Drug Administration (USA) , Fulvestrant/effets indésirables , Humains , Femelle , Systèmes de signalement des effets indésirables des médicaments/statistiques et données numériques , Adulte d'âge moyen , Adulte , Sujet âgé , États-Unis , Tumeurs du sein/traitement médicamenteux , Facteurs de risque , Antinéoplasiques hormonaux/effets indésirables , Adolescent , Effets secondaires indésirables des médicaments/épidémiologie , Jeune adulte
12.
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
13.
Expert Rev Anticancer Ther ; 24(6): 397-405, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38642015

RÉSUMÉ

INTRODUCTION: Estrogen receptor positive (ER+) breast cancer is the most common breast cancer subtype, and therapeutic management relies primarily on inhibiting ER signaling. In the metastatic setting, ER signaling is typically targeted by selective estrogen receptor degraders (SERDs) or aromatase inhibitors (AIs), the latter of which prevent estrogen production. Activating ESR1 mutations are among the most common emergent breast cancer mutations and confer resistance to AIs. AREAS COVERED: Until 2023, fulvestrant was the only approved SERD; fulvestrant is administered intramuscularly, and in some cases may also have limited efficacy in the setting of certain ESR1 mutations. In 2023, the first oral SERD, elacestrant, was approved for use in ESR1-mutated, ER+/HER2- advanced breast cancer and represents a new class of therapeutic options. While the initial approval was as monotherapy, ongoing studies are evaluating elacestrant (as well as other oral SERDs) in combination with other therapies including CDK4/6 inhibitors and PI3K inhibitors, which parallels the current combination uses of fulvestrant. EXPERT OPINION: Elacestrant's recent approval sheds light on the use of biomarkers such as ESR1 to gauge a tumor's endocrine sensitivity. Ongoing therapeutic and correlative biomarker studies will offer new insight and expanding treatment options for patients with advanced breast cancer.


Sujet(s)
Tumeurs du sein , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Administration par voie orale , Inhibiteurs de l'aromatase/pharmacologie , Inhibiteurs de l'aromatase/administration et posologie , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Animaux , Mutation , Fulvestrant/administration et posologie , Fulvestrant/pharmacologie , Résistance aux médicaments antinéoplasiques , Récepteurs des oestrogènes/métabolisme , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Modulateurs sélectifs des récepteurs des oestrogènes/administration et posologie , Thérapie moléculaire ciblée , Transduction du signal/effets des médicaments et des substances chimiques
14.
J Clin Oncol ; 42(18): 2149-2160, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38537155

RÉSUMÉ

PURPOSE: To compare giredestrant and physician's choice of endocrine monotherapy (PCET) for estrogen receptor-positive, HER2-negative, advanced breast cancer (BC) in the phase II acelERA BC study (ClinicalTrials.gov identifier: NCT04576455). METHODS: Post-/pre-/perimenopausal women, or men, age 18 years or older with measurable disease/evaluable bone lesions, whose disease progressed after 1-2 lines of systemic therapy (≤1 targeted, ≤1 chemotherapy regimen, prior fulvestrant allowed) were randomly assigned 1:1 to giredestrant (30 mg oral once daily) or fulvestrant/aromatase inhibitor per local guidelines (+luteinizing hormone-releasing hormone agonist in pre-/perimenopausal women, and men) until disease progression/unacceptable toxicity. Stratification was by visceral versus nonvisceral disease, prior cyclin-dependent kinase 4/6 inhibitor, and prior fulvestrant. The primary end point was investigator-assessed progression-free survival (INV-PFS). RESULTS: At clinical cutoff (February 18, 2022; median follow-up: 7.9 months; N = 303), the INV-PFS hazard ratio (HR) was 0.81 (95% CI, 0.60 to 1.10; P = .1757). In the prespecified secondary end point analysis of INV-PFS by ESR1 mutation (m) status in circulating tumor DNA-evaluable patients (n = 232), the HR in patients with a detectable ESR1m (n = 90) was 0.60 (95% CI, 0.35 to 1.03) versus 0.88 (95% CI, 0.54 to 1.42) in patients with no ESR1m detected (n = 142). Related grade 3-4 adverse events (AEs), serious AEs, and discontinuations due to AEs were balanced across arms. CONCLUSION: Although the acelERA BC study did not reach statistical significance for its primary INV-PFS end point, there was a consistent treatment effect with giredestrant across most key subgroups and a trend toward favorable benefit among patients with ESR1-mutated tumors. Giredestrant was well tolerated, with a safety profile comparable to PCET and consistent with known endocrine therapy risks. Overall, these data support the continued investigation of giredestrant in other studies.


Sujet(s)
Tumeurs du sein , Fulvestrant , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/mortalité , Adulte d'âge moyen , Récepteur ErbB-2/métabolisme , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/analyse , Sujet âgé , Adulte , Fulvestrant/usage thérapeutique , Mâle , Inhibiteurs de l'aromatase/usage thérapeutique , Inhibiteurs de l'aromatase/effets indésirables , Antinéoplasiques hormonaux/usage thérapeutique , Survie sans progression , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables
15.
J Clin Oncol ; 42(17): 2050-2060, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38513188

RÉSUMÉ

PURPOSE: Cyclin-dependent kinase (CDK) 4/6 inhibitors (CDK4/6is) are an important component of treatment for hormone receptor-positive/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC), but it is not known if patients might derive benefit from continuation of CDK4/6i with endocrine therapy beyond initial tumor progression or if the addition of checkpoint inhibitor therapy has value in this setting. METHODS: The randomized multicenter phase II PACE trial enrolled patients with hormone receptor-positive/HER2- MBC whose disease had progressed on previous CDK4/6i and aromatase inhibitor (AI) therapy. Patients were randomly assigned 1:2:1 to receive fulvestrant (F), fulvestrant plus palbociclib (F + P), or fulvestrant plus palbociclib and avelumab (F + P + A). The primary end point was investigator-assessed progression-free survival (PFS) in patients treated with F versus F + P. RESULTS: Overall, 220 patients were randomly assigned between September 2017 and February 2022. The median age was 57 years (range, 25-83 years). Most patients were postmenopausal (80.9%), and 40% were originally diagnosed with de novo MBC. Palbociclib was the most common previous CDK4/6i (90.9%). The median PFS was 4.8 months on F and 4.6 months on F + P (hazard ratio [HR], 1.11 [90% CI, 0.79 to 1.55]; P = .62). The median PFS on F + P + A was 8.1 months (HR v F, 0.75 [90% CI, 0.50 to 1.12]; P = .23). The difference in PFS with F + P and F + P + A versus F was greater among patients with baseline ESR1 and PIK3CA alterations. CONCLUSION: The addition of palbociclib to fulvestrant did not improve PFS versus fulvestrant alone among patients with hormone receptor-positive/HER2- MBC whose disease had progressed on a previous CDK4/6i plus AI. The increased PFS seen with the addition of avelumab warrants further investigation in this patient population.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Inhibiteurs de l'aromatase , Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Fulvestrant , Pipérazines , Pyridines , Récepteur ErbB-2 , Récepteurs des oestrogènes , Récepteurs à la progestérone , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/mortalité , Pyridines/usage thérapeutique , Pipérazines/usage thérapeutique , Inhibiteurs de l'aromatase/usage thérapeutique , Adulte d'âge moyen , Fulvestrant/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Sujet âgé , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/analyse , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Adulte , Récepteur ErbB-2/métabolisme , Récepteurs à la progestérone/métabolisme , Anticorps monoclonaux humanisés/usage thérapeutique , Sujet âgé de 80 ans ou plus , Évolution de la maladie , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Inhibiteurs de protéines kinases/usage thérapeutique , Survie sans progression
16.
J Nanobiotechnology ; 22(1): 107, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38475902

RÉSUMÉ

BACKGROUND: Breast cancer is the most prevalent malignant tumor among women, with hormone receptor-positive cases constituting 70%. Fulvestrant, an antagonist for these receptors, is utilized for advanced metastatic hormone receptor-positive breast cancer. Yet, its inhibitory effect on tumor cells is not strong, and it lacks direct cytotoxicity. Consequently, there's a significant challenge in preventing recurrence and metastasis once cancer cells develop resistance to fulvestrant. METHOD: To address these challenges, we engineered tumor-targeting nanoparticles termed 131I-fulvestrant-ALA-PFP-FA-NPs. This involved labeling fulvestrant with 131I to create 131I-fulvestrant. Subsequently, we incorporated the 131I-fulvestrant and 5-aminolevulinic acid (ALA) into fluorocarbon nanoparticles with folate as the targeting agent. This design facilitates a tri-modal therapeutic approach-endocrine therapy, radiotherapy, and PDT for estrogen receptor-positive breast cancer. RESULTS: Our in vivo and in vitro tests showed that the drug-laden nanoparticles effectively zeroed in on tumors. This targeting efficiency was corroborated using SPECT-CT imaging, confocal microscopy, and small animal fluorescence imaging. The 131I-fulvestrant-ALA-PFP-FA-NPs maintained stability and showcased potent antitumor capabilities due to the synergism of endocrine therapy, radiotherapy, and CR-PDT. Throughout the treatment duration, we detected no notable irregularities in hematological, biochemical, or histological evaluations. CONCLUSION: We've pioneered a nanoparticle system loaded with radioactive isotope 131I, endocrine therapeutic agents, and a photosensitizer precursor. This system offers a combined modality of radiotherapy, endocrine treatment, and PDT for breast cancer.


Sujet(s)
Tumeurs du sein , Animaux , Humains , Femelle , Fulvestrant/pharmacologie , Fulvestrant/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Interactions médicamenteuses , Radio-isotopes de l'iode
17.
Breast Cancer Res ; 26(1): 36, 2024 03 04.
Article de Anglais | MEDLINE | ID: mdl-38439079

RÉSUMÉ

BACKGROUND: Hyperglycemia is an on-target effect of PI3Kα inhibitors. Early identification and intervention of treatment-induced hyperglycemia is important for improving management of patients receiving a PI3Kα inhibitor like alpelisib. Here, we characterize incidence of grade 3/4 alpelisib-related hyperglycemia, along with time to event, management, and outcomes using a machine learning model. METHODS: Data for the risk model were pooled from patients receiving alpelisib ± fulvestrant in the open-label, phase 1 X2101 trial and the randomized, double-blind, phase 3 SOLAR-1 trial. The pooled population (n = 505) included patients with advanced solid tumors (X2101, n = 221) or HR+/HER2- advanced breast cancer (SOLAR-1, n = 284). External validation was performed using BYLieve trial patient data (n = 340). Hyperglycemia incidence and management were analyzed for SOLAR-1. RESULTS: A random forest model identified 5 baseline characteristics most associated with risk of developing grade 3/4 hyperglycemia (fasting plasma glucose, body mass index, HbA1c, monocytes, age). This model was used to derive a score to classify patients as high or low risk for developing grade 3/4 hyperglycemia. Applying the model to patients treated with alpelisib and fulvestrant in SOLAR-1 showed higher incidence of hyperglycemia (all grade and grade 3/4), increased use of antihyperglycemic medications, and more discontinuations due to hyperglycemia (16.7% vs. 2.6% of discontinuations) in the high- versus low-risk group. Among patients in SOLAR-1 (alpelisib + fulvestrant arm) with PIK3CA mutations, median progression-free survival was similar between the high- and low-risk groups (11.0 vs. 10.9 months). For external validation, the model was applied to the BYLieve trial, for which successful classification into high- and low-risk groups with shorter time to grade 3/4 hyperglycemia in the high-risk group was observed. CONCLUSIONS: A risk model using 5 clinically relevant baseline characteristics was able to identify patients at higher or lower probability for developing alpelisib-induced hyperglycemia. Early identification of patients who may be at higher risk for hyperglycemia may improve management (including monitoring and early intervention) and potentially lead to improved outcomes. REGISTRATION: ClinicalTrials.gov: NCT01219699 (registration date: October 13, 2010; retrospectively registered), ClinicalTrials.gov: NCT02437318 (registration date: May 7, 2015); ClinicalTrials.gov: NCT03056755 (registration date: February 17, 2017).


Sujet(s)
Tumeurs du sein , Hyperglycémie , Thiazoles , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Fulvestrant/effets indésirables , Hyperglycémie/induit chimiquement , Hyperglycémie/épidémiologie , Appréciation des risques
18.
J Clin Oncol ; 42(12): 1450-1453, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38478799

RÉSUMÉ

ASCO Rapid Recommendation Updates highlight revisions to select ASCO guideline recommendations as a response to the emergence of new and practice-changing data. The rapid updates are supported by an evidence review and follow the guideline development processes outlined in the ASCO Guideline Methodology Manual. The goal of these articles is to disseminate updated recommendations, in a timely manner, to better inform health practitioners and the public on the best available cancer care options. Guidelines and updates are not intended to substitute for independent professional judgment of the treating provider and do not account for individual variation among patients. See appendix for disclaimers and other important information (Appendix 1 and Appendix 2, online only).


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Femelle , Humains , Tumeurs du sein/traitement médicamenteux , Fulvestrant/usage thérapeutique , Pyrimidines/usage thérapeutique , Pyrroles/usage thérapeutique , Récepteur ErbB-2/métabolisme , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
19.
Front Endocrinol (Lausanne) ; 15: 1362725, 2024.
Article de Anglais | MEDLINE | ID: mdl-38549762

RÉSUMÉ

Background: Early studies have revealed antagonistic effects associated with stacking chemotherapy (CT) and endocrine therapy (ET), thereby conventional wisdom does not advocate the simultaneous combination of these two treatment modalities. Limited clinical studies exist on the combined use of neoadjuvant CT (NACT) and neoadjuvant ET (NET), and there are no reported instances of concurrent neoadjuvant treatment for locally advanced breast cancer (LABC) using capecitabine and fulvestrant (FUL). Case presentation: We reported a 54-year-old woman who was diagnosed with hormone receptor-positive (HR+) LABC at our hospital. After neoadjuvant treatment involving two distinct CT regimens did not lead to tumor regression. Consequently, the patient was transitioned to concurrent capecitabine and FUL therapy. This change resulted in favorable pathological remission without any significant adverse events during treatment. Conclusions: A novel approach involving concurrent neoadjuvant therapy with CT and endocrine therapy may offer a potentially effective treatment avenue for some cases with HR+ LABC.


Sujet(s)
Tumeurs du sein , Femelle , Humains , Adulte d'âge moyen , Tumeurs du sein/anatomopathologie , Traitement néoadjuvant/méthodes , Capécitabine/usage thérapeutique , Fulvestrant/usage thérapeutique , Résultat thérapeutique
20.
J Pharm Biomed Anal ; 242: 116032, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38367520

RÉSUMÉ

INTRODUCTION: Aromatase inhibitors such as anastrozole, letrozole, exemestane and selective estrogen down-regulator (SERD) fulvestrant are used mostly to treat breast cancer estrogen receptor positive in post-menopausal women. These drugs are given either through the oral route or by intramuscular injection. They have shown great inter-individual variability with a risk of cardiometabolic disorders. Hence the importance of their therapeutic drug monitoring not only for exposure-efficacy but also exposure-toxicity. We describe here a LC-MS/MS method for the simultaneous quantification of anastrozole, letrozole, exemestane and fulvestrant in human plasma. MATERIAL AND METHODS: Plasma samples were prepared by a single-step protein precipitation. The liquid chromatography system was paired with a triple quadrupole mass spectrometer. Quantification were achieved in Multiple Reactions Monitoring mode and the electrospray ionization was in positive mode. RESULTS: The method demonstrated consistent analytical performance across various parameters, including linearity, specificity, sensitivity, matrix effect, upper and lower limits of quantification, extraction recovery, precision, accuracy, hemolysis effect, dilution integrity, and stability under different storage conditions, in accordance with established guidelines. The analysis time for each run was 4 min. Calibration curves exhibited linearity within the 1-100 ng/mL range, with correlation coefficients > 0.99 for the four analytes. Plasma concentrations from 42 patients were integrated into the selected calibration. Stability assessments indicated that the four drugs remained stable at - 20 °C for three months, 15 days under refrigeration, up to 7 days at room temperature, and after three freeze-thaw cycles. CONCLUSION: We have developed and validated this quantitative method for therapeutic drug monitoring of those four hormone therapy drugs:anastrozole, letrozole, fulvestrant and exemestane. This method can be also used for future clinical pharmacokinetics /pharmacodynamics studies.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Anastrozole/usage thérapeutique , Létrozole/usage thérapeutique , Chromatographie en phase liquide/méthodes , Fulvestrant/usage thérapeutique , , Spectrométrie de masse en tandem/méthodes , Reproductibilité des résultats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE