Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.181
Filter
1.
Med Oncol ; 41(10): 246, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312041

ABSTRACT

This study investigates the potential of 2-methoxyestradiol (2-ME) to overcome tamoxifen (TAM) resistance in MCF-7 breast cancer cells by downregulating hypoxia-inducible factor 1 alpha (HIF-1α). Through a series of in vitro experiments, the authors demonstrate that combining 2-ME with TAM enhances the cytotoxic effects on resistant cells, increases apoptosis markers, and reduces cholesterol and triglyceride levels. While the findings highlight a promising therapeutic approach, the lack of in vivo or clinical data limits direct clinical application. Future research should focus on validating these results in animal models and exploring long-term efficacy and molecular mechanisms.


Subject(s)
2-Methoxyestradiol , Antineoplastic Agents, Hormonal , Breast Neoplasms , Down-Regulation , Drug Resistance, Neoplasm , Estradiol , Hypoxia-Inducible Factor 1, alpha Subunit , Tamoxifen , Humans , Tamoxifen/pharmacology , Tamoxifen/analogs & derivatives , 2-Methoxyestradiol/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Drug Resistance, Neoplasm/drug effects , MCF-7 Cells , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estradiol/pharmacology , Estradiol/analogs & derivatives , Female , Down-Regulation/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects
2.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201247

ABSTRACT

Tamoxifen, a selective estrogen receptor modulator (SERM), exhibits dual agonist or antagonist effects contingent upon its binding to either G-protein-coupled estrogen receptor (GPER) or estrogen nuclear receptor (ESR). Estrogen signaling plays a pivotal role in initiating epigenetic alterations and regulating estrogen-responsive genes in breast cancer. Employing three distinct breast cancer cell lines-MCF-7 (ESR+; GPER+), MDA-MB-231 (ESR-; GPER-), and SkBr3 (ESR-; GPER+)-this study subjected them to treatment with two tamoxifen derivatives: 4-hydroxytamoxifen (4-HT) and endoxifen (Endox). Through 2D high-performance liquid chromatography with tandem mass spectrometry detection (HPLC-MS/MS), varying levels of 5-methylcytosine (5-mC) were found, with MCF-7 displaying the highest levels. Furthermore, TET3 mRNA expression levels varied among the cell lines, with MCF-7 exhibiting the lowest expression. Notably, treatment with 4-HT induced significant changes in TET3 expression across all cell lines, with the most pronounced increase seen in MCF-7 and the least in MDA-MB-231. These findings underscore the influence of tamoxifen derivatives on DNA methylation patterns, particularly through modulating TET3 expression, which appears to be contingent on the presence of estrogen receptors. This study highlights the potential of targeting epigenetic modifications for personalized anti-cancer therapy, offering a novel avenue to improve treatment outcomes.


Subject(s)
Breast Neoplasms , Dioxygenases , Gene Expression Regulation, Neoplastic , Selective Estrogen Receptor Modulators , Tamoxifen , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Tamoxifen/pharmacology , Tamoxifen/analogs & derivatives , Female , Dioxygenases/genetics , Dioxygenases/metabolism , Selective Estrogen Receptor Modulators/pharmacology , MCF-7 Cells , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA Methylation/drug effects , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Tandem Mass Spectrometry
3.
Expert Opin Pharmacother ; 25(11): 1541-1554, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39129457

ABSTRACT

INTRODUCTION: The demand for effective and safe treatments of genitourinary syndrome (GSM) in post-menopausal women (PMW) is growing. Published data on the efficacy and safety of ospemifene (OSP) prompt an updated literature review to enlighten possible improvements in the GSM treatment. AREA COVERED: We searched articles published in English from 2010 to 2023 through Medline (PubMed) and Embase databases with Boolean terms: OSP, PMW, GSM, endometrium, breast cancer, cardiometabolic syndrome, bone metabolism, adherence to treatment, and patient satisfaction. We selected randomized controlled trials (RCTs) and observational and cross-sectional studies and completed the search manually. EXPERT OPINION: Of the 157 retrieved records, 25 primary studies met the inclusion criteria (15 regarding efficacy and safety, two for additional effects, and four for adherence and satisfaction with the OSP treatment). Seven RCTs involved nearly 5,000 patients, 10 out of 18 prospective observational studies 563, and six retrospective analyses 356,439. Evidence of OSP treatment in PMW with GSM relies on RCTs and remarkable real-world data. The 25 primary studies showcased the high clinical response to symptoms, the favorable safety profile of OSP with very few adverse events, a neutral impact on the endometrium, breast, bone, and thrombosis, and the possible improvement of cardiovascular risk factors.


Subject(s)
Atrophy , Postmenopause , Tamoxifen , Vagina , Vulva , Humans , Female , Atrophy/drug therapy , Tamoxifen/therapeutic use , Tamoxifen/analogs & derivatives , Tamoxifen/adverse effects , Vagina/pathology , Vagina/drug effects , Vulva/pathology , Vulva/drug effects , Randomized Controlled Trials as Topic , Selective Estrogen Receptor Modulators/therapeutic use , Selective Estrogen Receptor Modulators/adverse effects , Female Urogenital Diseases/drug therapy
4.
Med Oncol ; 41(9): 232, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39167288

ABSTRACT

The clinical studies for breast cancer (BC) are now assessing the efficacy of 2-Methoxyestradiol (2-ME), a naturally occurring derivative of estradiol. Our study aimed to explore the potential of combining the 2-ME and tamoxifen (TAM) on sensitization of TAM-resistant cells using LCC2 the TAM-resistant cells as a model and comparing the results to the sensitive cells MCF-7. Sulphorhodamine-B (SRB) assay is used to examine the 2-ME chemo-sensitizing impact on the cytotoxicity of TAM on LCC2 cells. Colorimetric assay kits were used to assess the level of the apoptosis-related markers caspases 3, Bcl2, and Bax in cell lysate. Hypoxia-inducible factor 1 alpha (HIF-1α) expression was measured using western blotting. Total cholesterol and triglyceride (TG) levels were examined colorimetrically, using the BIOLABO kit. The use of 2-ME enhanced the cytotoxic effects of TAM and effectively reversed TAM resistance. This was achieved by inhibiting the expression of HIF-1α, while concurrently increasing the levels of apoptotic marker caspase-3, as well as the pro-apoptotic protein Bax. Additionally, there was a reduction in the levels of Bcl2, an anti-apoptotic protein. Furthermore, a reduction in TG and cholesterol levels was noted. Our findings show that HIF-1α plays an important role in TAM resistance and that suppression of HIF-1α by 2-ME-mediated sensitization of BC-resistant cells to TAM. Therefore, the concurrent administration of TAM/2-ME might potentially serve as a viable therapeutic approach to address TAM resistance and enhance the overall therapy efficacy for patients with BC.


Subject(s)
2-Methoxyestradiol , Breast Neoplasms , Down-Regulation , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit , Tamoxifen , Humans , 2-Methoxyestradiol/pharmacology , Tamoxifen/pharmacology , Tamoxifen/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Drug Resistance, Neoplasm/drug effects , MCF-7 Cells , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Down-Regulation/drug effects , Apoptosis/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Estradiol/pharmacology , Estradiol/analogs & derivatives
5.
Biosci Rep ; 44(8)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39034849

ABSTRACT

OBJECTIVE: Estrogen receptor-positive (ER+) breast cancer represents about 80% of cases, tamoxifen is the election neoadjuvant chemotherapy. However, a large percentage of patients develop chemoresistance, compromising recovery. Clinical evidence suggests that high plasmatic levels of low-density lipoproteins (LDL) could promote cancer progression. The present study analyzed the effect of LDL on the primary plasmatic active Tamoxifen's metabolites resistance acquisition, 4-hydroxytamoxifen (4OH-Tam) and 4-hydroxy-N-desmethyl-tamoxifen (endoxifen), in breast cancer ERα + cells (MCF-7). METHODS: Two resistant cellular variants, MCF-7Var-H and MCF-7Var-I, were generated by a novel strategy and their phenotype features were evaluated. Phenotypic assessment was performed by MTT assays, cytometry, immunofluorescence microscopy, zymography and protein expression analysis. RESULTS: MCF-7Var-H, generated only with tamoxifen metabolites, showed a critical down-regulation in hormone receptors, augmented migration capacity, metalloprotease 9 extracellular medium excretion, and a mesenchymal morphology in contrast with native MCF-7, suggesting the transition towards Triple-negative breast cancer (TNBC) phenotype. In contrast, MCF-7Var-I which was generated in a high LDL media, showed only a slight upregulation in ER and other less noticeable metabolic adaptations. Results suggest a potential role of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in phenotypic differences observed among variants. CONCLUSION: LDL high or low concentrations during Tamoxifen´s metabolites chemoresistance acquisition leads to different cellular mechanisms related to chemoresistance. A novel adaptative cellular response associated with Nrf2 activity could be implicated.


Subject(s)
Drug Resistance, Neoplasm , Estrogen Receptor alpha , Lipoproteins, LDL , Phenotype , Tamoxifen , Triple Negative Breast Neoplasms , Humans , Tamoxifen/pharmacology , Tamoxifen/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , MCF-7 Cells , Female , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Lipoproteins, LDL/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Cell Movement/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects
6.
Cell Death Dis ; 15(6): 444, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914552

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Receptor, ErbB-2 , Receptors, Estrogen , Signal Transduction , Smad4 Protein , Humans , Smad4 Protein/metabolism , Smad4 Protein/genetics , Female , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Signal Transduction/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Receptors, Estrogen/metabolism , Cell Line, Tumor , Animals , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Tamoxifen/analogs & derivatives , Mice , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Mice, Nude , Gene Expression Regulation, Neoplastic/drug effects , Autophagy/drug effects , ErbB Receptors/metabolism , ErbB Receptors/genetics
7.
Hum Psychopharmacol ; 39(4): e2899, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38683854

ABSTRACT

BACKGROUND: Endoxifen, a protein kinase C inhibitor and selective estrogen receptor modulator, primarily used in breast cancer treatment, has recently emerged as a potential therapeutic option for managing manic episodes associated with bipolar disorder (BD). This review aims to assess the existing evidence base for endoxifen in BD treatment and evaluate the strengths and limitations of current research findings. METHODS: A systematic search was conducted on Medline, Embase, and Web of Science databases. We included studies published in English that used endoxifen in BD, alongside any relevant studies identified through manual searching and conference papers with full-text availability. Information pertaining to dose, duration, clinical effects, and safety profiles was extracted from the included studies. The Cochrane Risk of Bias 2 tool was used to assess the risk of bias in clinical trials. RESULTS: The final review included seven case reports (including two conference presentations), two clinical trials, and one prospective study. Most studies administered endoxifen 8 mg and reported an improvement in manic symptoms. Several case reports included patients with comorbid substance use, and most patients received mood stabilizers concurrently. Few reports lacked any structured outcome measures. The clinical trials used divalproex 1000 mg as an active comparator, which was deemed sub-therapeutic. Despite being multicentric, the first trial lacked data on center-wise recruitment, and certain methodological concerns were observed across the included trials. There were no serious adverse effects noted, except for a significant elevation in lipid profile within a 3-week period. Limited data were available regarding endoxifen efficacy and safety in mixed episodes, depressive episodes, and maintenance treatment. CONCLUSION: There is a paucity of research on the efficacy and safety of endoxifen in BD. While existing evidence suggests short-term efficacy in manic episodes, significant limitations were identified in most of the included studies. Further research is imperative to establish the efficacy and safety of endoxifen in BD before considering its recommendation as a viable treatment option.


Subject(s)
Bipolar Disorder , Tamoxifen , Humans , Bipolar Disorder/drug therapy , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Tamoxifen/adverse effects , Tamoxifen/administration & dosage , Selective Estrogen Receptor Modulators/therapeutic use , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/administration & dosage , Treatment Outcome
8.
Clin Pharmacol Ther ; 116(3): 690-702, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38494911

ABSTRACT

Tamoxifen is widely used in patients with hormone receptor-positive breast cancer. The polymorphic enzyme CYP2D6 is primarily responsible for metabolic activation of tamoxifen, resulting in substantial interindividual variability of plasma concentrations of its most important metabolite, Z-endoxifen. The Z-endoxifen concentration thresholds below which tamoxifen treatment is less efficacious have been proposed but not validated, and prospective trials of individualized tamoxifen treatment to achieve Z-endoxifen concentration thresholds are considered infeasible. Therefore, we aim to validate the association between Z-endoxifen concentration and tamoxifen treatment outcomes, and identify a Z-endoxifen concentration threshold of tamoxifen efficacy, using pharmacometric modeling and simulation. As a first step, the CYP2D6 Endoxifen Percentage Activity Model (CEPAM) cohort was created by pooling data from 28 clinical studies (> 7,000 patients) with measured endoxifen plasma concentrations. After cleaning, data from 6,083 patients were used to develop a nonlinear mixed-effect (NLME) model for tamoxifen and Z-endoxifen pharmacokinetics that includes a conversion factor to allow inclusion of studies that measured total endoxifen but not Z-endoxifen. The final parent-metabolite NLME model confirmed the primary role of CYP2D6, and contributions from body weight, CYP2C9 phenotype, and co-medication with CYP2D6 inhibitors, on Z-endoxifen pharmacokinetics. Future work will use the model to simulate Z-endoxifen concentrations in patients receiving single agent tamoxifen treatment within large prospective clinical trials with long-term survival to identify the Z-endoxifen concentration threshold below which tamoxifen is less efficacious. Identification of this concentration threshold would allow personalized tamoxifen treatment to improve outcomes in patients with hormone receptor-positive breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal , Breast Neoplasms , Cytochrome P-450 CYP2D6 , Nonlinear Dynamics , Tamoxifen , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacokinetics , Tamoxifen/blood , Tamoxifen/therapeutic use , Humans , Female , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2D6/genetics , Breast Neoplasms/drug therapy , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/blood , Models, Biological , Middle Aged , Cohort Studies , Treatment Outcome , Computer Simulation , Aged
9.
Maturitas ; 183: 107950, 2024 May.
Article in English | MEDLINE | ID: mdl-38462385

ABSTRACT

OBJECTIVES: To assess clinical characteristics of postmenopausal women with moderate/severe vulvovaginal atrophy, as well as its impact on sexual function, well-being, and quality of life, and to provide an overview of most used treatments. STUDY DESIGN: Ongoing longitudinal, observational study conducted in 17 Italian gynecology centers, involving women already treated or initiating a local vaginal estrogen therapy or ospemifene. We report baseline data for women with and without a history of breast cancer. Participants filled in self-reported questionnaires at study entry. MAIN OUTCOME MEASURES: Severity of vulvovaginal atrophy; ongoing treatments; patient-reported outcomes, including severity of symptoms, Day-to-Day Impact of Vaginal Aging (DIVA), Female Sexual Function Index (FSFI), Female Sexual Distress Scale-Revised (FSDS-R), and SF-12® Health Survey. RESULTS: Overall, 334 women (20.4 % with a history of breast cancer) started or continued local therapy (61.1 %) or ospemifene (38.8 %) at study entry. Vulvovaginal atrophy was severe in 28.6 %, and was responsible for severe symptoms, particularly vulvar dryness with burning or irritation and pain during sexual intercourse. Both sexual dysfunction (FSFI≤26) (81.5 %) and sexual distress (FSDS-R ≥ 11) (74.4 %) were common. A reduction in the SF-12 mental component score was documented. Women with breast cancer more often had severe vulvovaginal atrophy (41.2 %), had more severe symptoms, and the impact of vaginal symptoms on emotional well-being, sexual functioning and self-concept/body image was greater. The majority of them (83.8 %) received ospemifene as a treatment. CONCLUSIONS: Moderate/severe vulvovaginal atrophy is a common, often neglected condition with an impact on QoL and sexuality, particularly in women with a history of breast cancer. It is important to alleviate the burden associated with the disease.


Subject(s)
Breast Neoplasms , Tamoxifen , Vaginal Diseases , Female , Humans , Atrophy/pathology , Breast Neoplasms/complications , Breast Neoplasms/pathology , Patient Satisfaction , Quality of Life , Tamoxifen/analogs & derivatives , Vagina/pathology , Vaginal Diseases/drug therapy , Vulva/pathology
10.
Clin Pharmacol Ther ; 116(1): 155-164, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38501904

ABSTRACT

Tamoxifen is part of the standard of care of endocrine therapy for adjuvant treatment of breast cancer. However, survival outcomes with tamoxifen are highly variable. The concentration of endoxifen, the 30-100 times more potent metabolite of tamoxifen and bioactivated by the CYP2D6 enzyme, has been described as the most relevant metabolite of tamoxifen metabolism. A genome-wide association study (GWAS) was performed with the objective to identify genetic polymorphisms associated with endoxifen serum concentration levels and clinical outcome in early-stage breast cancer patients receiving tamoxifen. A GWAS was conducted in 608 women of the CYPTAM study (NTR1509/PMID: 30120701). Germline DNA and clinical and survival characteristics were readily available. Genotyping was performed on Infinium Global Screening Array (686,082 markers) and single nucleotide polymorphism (SNP) imputation by using 1000 Genomes. Relapse-free survival during tamoxifen (RFSt) was defined the primary clinical outcome. Endoxifen serum concentration was analyzed as a continuous variable. Several genetic variants reached genome-wide significance (P value: ≤5 × 10-8). Endoxifen concentrations analysis identified 430 variants, located in TCF20 and WBP2NL genes (chromosome 22), which are in strong linkage disequilibrium with CYP2D6 variants. In the RFSt analysis, several SNP were identified (LPP gene: rs77693286, HR 18.3, 95% CI: 15.2-21.1; rs6790761, OR 18.2, 95% CI: 15.5-21.1). Endoxifen concentrations have a strong association with the chromosome 22, which contains the CYP2D6 gene.


Subject(s)
Antineoplastic Agents, Hormonal , Breast Neoplasms , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Tamoxifen , Humans , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Tamoxifen/blood , Tamoxifen/pharmacokinetics , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/blood , Middle Aged , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/blood , Aged , Cytochrome P-450 CYP2D6/genetics , Chemotherapy, Adjuvant , Adult , Neoplasm Staging , Treatment Outcome , Disease-Free Survival
11.
Article in English | MEDLINE | ID: mdl-38547701

ABSTRACT

Interindividual exposure differences have been identified in oral targeted antineoplastic drugs (OADs) owing to the pharmacogenetic background of the patients and their susceptibility to multiple factors, resulting in insufficient efficacy or adverse effects. Therapeutic drug monitoring (TDM) can prevent sub-optimal concentrations of OADs and improve their clinical treatment. This study aimed to develop and validate an LC-MS/MS method for the simultaneous quantification of 11 OADs (gefitinib, imatinib, lenvatinib, regorafenib, everolimus, osimertinib, sunitinib, tamoxifen, lapatinib, fruquintinib and sorafenib) and 2 active metabolites (N-desethyl sunitinib and Z-endoxifen) in human plasma. Protein precipitation was used to extract OADs from the plasma samples. Chromatographic separation was performed using an Eclipse XDB-C18 (4.6 × 150 mm, 5 µm) column with a gradient elution of the mobile phase composed of 2 mM ammonium acetate with 0.1 % formic acid in water (solvent A) and methanol (solvent B) at a flow rate of 0.8 mL/min. Mass analysis was performed using positive ion mode electrospray ionization in multiple-reaction monitoring mode. The developed method was validated following FDA bioanalytical guidelines. The calibration curves were linear over the range of 2-400 ng/mL for gefitinib, imatinib, lenvatinib, regorafenib, and everolimus; 1-200 ng/mL for osimertinib, sunitinib, N-desethyl sunitinib, tamoxifen, and Z-endoxifen; and 5-1000 ng/mL for lapatinib, fruquintinib, and sorafenib, with all coefficients of correlation above 0.99. The intra- and inter-day imprecision was below 12.81 %. This method was successfully applied to the routine TDM of gefitinib, lenvatinib, regorafenib, osimertinib, fruquintinib, and sorafenib to optimize the dosage regimens.


Subject(s)
Acrylamides , Aniline Compounds , Antineoplastic Agents , Indoles , Neoplasms , Phenylurea Compounds , Pyridines , Pyrimidines , Quinolines , Tamoxifen/analogs & derivatives , Humans , Sunitinib , Imatinib Mesylate , Sorafenib , Lapatinib , Chromatography, Liquid/methods , Drug Monitoring/methods , Liquid Chromatography-Mass Spectrometry , Gefitinib , Everolimus , Tandem Mass Spectrometry/methods , Antineoplastic Agents/therapeutic use , Tamoxifen/therapeutic use , Neoplasms/drug therapy , Solvents , Reproducibility of Results , Chromatography, High Pressure Liquid/methods
12.
Nat Commun ; 15(1): 1689, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402222

ABSTRACT

Point-of-care sensors, which are low-cost and user-friendly, play a crucial role in precision medicine by providing quick results for individuals. Here, we transform the conventional glucometer into a 4-hydroxytamoxifen therapeutic biosensor in which 4-hydroxytamoxifen modulates the electrical signal generated by glucose oxidation. To encode the 4-hydroxytamoxifen signal within glucose oxidation, we introduce the ligand-binding domain of estrogen receptor-alpha into pyrroloquinoline quinone-dependent glucose dehydrogenase by constructing and screening a comprehensive protein insertion library. In addition to obtaining 4-hydroxytamoxifen regulatable engineered proteins, these results unveil the significance of both secondary and quaternary protein structures in propagation of conformational signals. By constructing an effective bioelectrochemical interface, we detect 4-hydroxytamoxifen in human blood samples as changes in the electrical signal and use this to develop an electrochemical algorithm to decode the 4-hydroxytamoxifen signal from glucose. To meet the miniaturization and signal amplification requirements for point-of-care use, we harness power from glucose oxidation to create a self-powered sensor. We also amplify the 4-hydroxytamoxifen signal using an organic electrochemical transistor, resulting in milliampere-level signals. Our work demonstrates a broad interdisciplinary approach to create a biosensor that capitalizes on recent innovations in protein engineering, electrochemical sensing, and electrical engineering.


Subject(s)
Biosensing Techniques , Point-of-Care Systems , Tamoxifen/analogs & derivatives , Humans , Glucose , Biosensing Techniques/methods , Protein Engineering , Electrochemical Techniques
13.
Exp Physiol ; 109(4): 513-523, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38291801

ABSTRACT

Tamoxifen (TAM) is required for gene recombination in the inducible Cre/lox system. The TAM-enriched diet is considered safe, with negligible impact on animal wellbeing. However, studies reporting the long-term effects of the TAM diet and its potential impact on experimental outcomes are scarce. We conducted a longitudinal study on mice exposed to a 4-week dietary TAM citrate supplementation. Several parameters were recorded, such as body weight, body composition, mortality, and cardiac function. The collagen1a2 (Col1a2) transgenic mouse was used to assess TAM-induced recombination in vivo in cardiac fibroblasts followed by myocardial infarction (MI). The impact of TAM on the MI outcome was also evaluated. The recombination efficiency and cytotoxic effect of the TAM active metabolite, 4-hydroxy-tamoxifen (4-OHT), were assessed in vitro. Mice exposed to a TAM diet showed body weight loss and a 10% increase in mortality (P = 0.045). The TAM diet decreased cardiac function and induced cardiac remodeling, indicated by decreased fractional shortening from 32.23% to 19.23% (P = 0.001) and left ventricular (LV) wall thinning. All measured parameters were reversed to normal when mice were returned to a normal diet. Infarcted Col1a2-CreER mice on the TAM regimen showed gene recombination in fibroblasts, but it was associated with a substantial increase in mortality post-surgery (2.5-fold) compared to the controls. In vitro, 4-OHT induced gene editing in fibroblasts; however, cell growth arrest and cytotoxicity were observed at high concentrations. In conclusion, prolonged exposure to the TAM diet can be detrimental and necessitates careful model selection and interpretation of the results.


Subject(s)
Cardiomyopathies , Frailty , Tamoxifen/analogs & derivatives , Mice , Animals , Longitudinal Studies , Tamoxifen/pharmacology , Mice, Transgenic , Diet
14.
Molecules ; 27(19)2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36235291

ABSTRACT

Utilizing McMurry reactions of 4,4'-dihydroxybenzophenone with appropriate carbonyl compounds, a series of 4-Hydroxytamoxifen analogues were synthesized. Their cytotoxic activity was evaluated in vitro on four human malignant cell lines (MCF-7, MDA-MB 231, A2058, HT-29). It was found that some of these novel Tamoxifen analogues show marked cytotoxicity in a dose-dependent manner. The relative ROS-generating capability of the synthetized analogues was evaluated by cyclic voltammetry (CV) and DFT modeling studies. The results of cell-viability assays, CV measurements and DFT calculations suggest that the cytotoxicity of the majority of the novel compounds is mainly elicited by their interactions with cellular targets including estrogen receptors rather than triggered by redox processes. However, three novel compounds could be involved in ROS-production and subsequent formation of quinone-methide preventing proliferation and disrupting the redox balance of the treated cells. Among the cell lines studied, HT-29 proved to be the most susceptible to the treatment with compounds having ROS-generating potency.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Electrons , Female , Humans , Reactive Oxygen Species/pharmacology , Receptors, Estrogen/metabolism , Structure-Activity Relationship , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism
15.
Breast Cancer Res Treat ; 195(1): 65-74, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35842520

ABSTRACT

INTRODUCTION: Endoxifen-the principal metabolite of tamoxifen-is subject to a high inter-individual variability in serum concentration. Numerous attempts have been made to explain this, but thus far only with limited success. By applying predictive modeling, we aimed to identify factors that determine the inter-individual variability. Our purpose was to develop a prediction model for endoxifen concentrations, as a strategy to individualize tamoxifen treatment by model-informed dosing in order to prevent subtherapeutic exposure (endoxifen < 16 nmol/L) and thus potential failure of therapy. METHODS: Tamoxifen pharmacokinetics with demographic and pharmacogenetic data of 303 participants of the prospective TOTAM study were used. The inter-individual variability in endoxifen was analyzed according to multiple regression techniques in combination with multiple imputations to adjust for missing data and bootstrapping to adjust for the over-optimism of parameter estimates used for internal model validation. RESULTS: Key predictors of endoxifen concentration were CYP2D6 genotype, age and weight, explaining altogether an average-based optimism corrected 57% (95% CI 0.49-0.64) of the inter-individual variability. CYP2D6 genotype explained 54% of the variability. The remaining 3% could be explained by age and weight. Predictors of risk for subtherapeutic endoxifen (< 16 nmol/L) were CYP2D6 genotype and age. The model showed an optimism-corrected discrimination of 90% (95% CI 0.86-0.95) and sensitivity and specificity of 66% and 98%, respectively. Consecutively, there is a high probability of misclassifying patients with subtherapeutic endoxifen concentrations based on the prediction rule. CONCLUSION: The inter-individual variability of endoxifen concentration could largely be explained by CYP2D6 genotype and for a small proportion by age and weight. The model showed a sensitivity and specificity of 66 and 98%, respectively, indicating a high probability of (misclassification) error for the patients with subtherapeutic endoxifen concentrations (< 16 nmol/L). The remaining unexplained inter-individual variability is still high and therefore model-informed tamoxifen dosing should be accompanied by therapeutic drug monitoring.


Subject(s)
Breast Neoplasms , Antineoplastic Agents, Hormonal , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cytochrome P-450 CYP2D6/genetics , Female , Genotype , Humans , Prospective Studies , Tamoxifen/analogs & derivatives
16.
Gynecol Endocrinol ; 38(7): 577-582, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35670533

ABSTRACT

OBJECTIVE: To assess the improvement in vulvovaginal atrophy (VVA) of postmenopausal women treated with oral ospemifene 60 mg/day under conditions of routine clinical practice after 12 months of follow-up. METHODS: The AYSEX study is a Spanish observational, prospective, and unicentric study in which five gynecologists recruited postmenopausal women with VVA in routine clinical practice treated with oral ospemifene 60 mg/day as an appropriate therapeutic option. Vaginal health, the most bothersome symptoms, sexual health, endometrial safety, bone resorption markers, bone densitometry, mammography, treatment satisfaction, and quality of life were assessed at baseline and after 12 months using appropriate scales. RESULTS: A total of 100 postmenopausal women cytologically and clinically diagnosed with VVA were included in the study. After 3 months of treatment with ospemifene, a significant improvement was observed in all domains of Vaginal Health Index. This improvement was maintained at month 12 and only one patient remained with vaginal atrophy. In addition, a significant improvement was observed in the most bothersome symptoms, sexual function, and quality of life. There was no significant change in endometrial thickness, mammography, and bone health during the 12 months of treatment. CONCLUSIONS: This study in routine clinical practice conditions confirms the results previously reported by randomized controlled trials, including a significant improvement in VVA, sexual function, quality of life, endometrial safety, and satisfaction with the treatment.


Subject(s)
Dyspareunia , Vaginal Diseases , Atrophy/drug therapy , Atrophy/pathology , Dyspareunia/drug therapy , Female , Follow-Up Studies , Humans , Prospective Studies , Quality of Life , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/analogs & derivatives , Treatment Outcome , Vagina/pathology , Vaginal Diseases/pathology , Vulva/pathology
17.
Oncologist ; 27(7): e597-e600, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35604960

ABSTRACT

Although breast cancer incidence is increasing, there are few primary preventive initiatives. Tamoxifen can reduce breast cancer incidence but is rarely used for primary prevention due to adverse events and tolerance issues. We tested if endoxifen, a tamoxifen metabolite, applied directly to the skin of the breast, could reduce mammographic density, a proxy for therapy response. Ninety women were randomized to placebo, 10 and 20 mg of topical Z-endoxifen for 6 months. Mammographic density and symptoms were measured at baseline and study exit. Despite a high discontinuation rate, driven by skin rashes, we found a significant mammographic density decrease, a dose-dependent increase in the concentration of plasma Z-endoxifen but no systemic side effects. Topical application of tamoxifen metabolites has the potential to decrease breast cancer incidence without major systemic side effects. However, endoxifen may not be suitable for topical administration and is unlikely to be used for breast cancer prevention.


Subject(s)
Breast Density , Breast Neoplasms , Antineoplastic Agents, Hormonal/adverse effects , Breast , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cytochrome P-450 CYP2D6 , Female , Humans , Tamoxifen/adverse effects , Tamoxifen/analogs & derivatives
18.
J Clin Pharmacol ; 62(9): 1121-1131, 2022 09.
Article in English | MEDLINE | ID: mdl-35358345

ABSTRACT

The purpose of this study was to develop and validate a population pharmacokinetic model for Z-endoxifen in patients with advanced solid tumors and to identify clinical variables that influence pharmacokinetic parameters. Z-endoxifen-HCl was administered orally once a day on a 28-day cycle (±3 days) over 11 dose levels ranging from 20 to 360 mg. A total of 1256 Z-endoxifen plasma concentration samples from 80 patients were analyzed using nonlinear mixed-effects modeling to develop a population pharmacokinetic model for Z-endoxifen. A 2-compartment model with oral depot and linear elimination adequately described the data. The estimated apparent total clearance, apparent central volume of distribution, and apparent peripheral volume of distribution were 4.89 L/h, 323 L, and 39.7 L, respectively, with weight-effect exponents of 0.75, 1, and 1, respectively. This model was used to explore the effects of clinical and demographic variables on Z-endoxifen pharmacokinetics. Weight, race on clearance, and aspartate aminotransferase on the absorption rate constant were identified as significant covariates in the final model. This novel population pharmacokinetic model provides insight regarding factors that may affect the pharmacokinetics of Z-endoxifen and may assist in the design of future clinical trials.


Subject(s)
Neoplasms , Tamoxifen , Humans , Models, Biological , Neoplasms/drug therapy , Neoplasms/pathology , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use
19.
Sci Rep ; 12(1): 1643, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102224

ABSTRACT

The aim of the study was to compare 3 blood sampling methods, including capillary blood sampling, for determining Tamoxifen (TAM), Z-endoxifen (END), and 4-hydroxytamoxifen (4HT) concentrations. High performance liquid chromatography-mass spectrometry was used to quantify concentrations of TAM, END, and 4HT in plasma, venous blood, and capillary blood samples of 16 participants on TAM therapy for breast cancer. The rhelise kit was used for capillary sampling. Calibration curves using 13C-labeled analogs of TAM, END, and 4HT as internal standards were used for quantifications. A capillary sampling kit was used successfully for all participants. Mean TAM concentrations did not differ significantly in the 3 types of samples. Mean END and 4HT concentrations did differ significantly between capillary and venous blood samples, possibly related to photodegradation in the internal standards prior to use or degradation products with chromatographic retention times similar to the metabolites. TAM, END, and 4HT concentrations were relatively stable when stored for 14 days at 8 °C and 20 °C. Therapeutic drug monitoring of TAM using an innovative kit and capillary blood sampling is feasible. Preliminary data from this study will aid in developing a multicenter, randomized clinical trial of personalized TAM dose monitoring and adjustments, with the goal of enhancing the quality-of-life and outcomes of patients with breast cancer.Clinical Trial Identification: EudraCT No 2017-000641-44.


Subject(s)
Breast Neoplasms/blood , Drug Monitoring/instrumentation , Estrogen Antagonists/blood , Reagent Kits, Diagnostic , Tamoxifen/analogs & derivatives , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Capillaries , Chromatography, High Pressure Liquid , Estrogen Antagonists/therapeutic use , Feasibility Studies , Female , Humans , Mass Spectrometry , Middle Aged , Pilot Projects , Predictive Value of Tests , Reproducibility of Results , Sweden , Tamoxifen/blood , Tamoxifen/therapeutic use
20.
Gene ; 821: 146284, 2022 May 05.
Article in English | MEDLINE | ID: mdl-35143939

ABSTRACT

The basic region leucin zipper (bZIP) protein c-Fos constitutes together with other bZIP proteins the AP-1 transcription factor complex. Expression of the c-Fos gene is regulated by numerous extracellular signaling molecules including mitogens, metabolites, and ligands for receptor tyrosine kinases, G protein-coupled receptors, and cytokine receptors. Here, we analyzed the effects of the stimulus-responsive MAP kinases ERK1/2 (extracellular signal-regulated protein kinase), JNK (c-Jun N-terminal protein kinase) and p38 protein kinase on transcription of the c-Fos gene. We used chromatin-integrated c-Fos promoter-luciferase reporter genes containing inactivating point mutations of DNA binding sites for distinct transcription factors. ERK1/2, JNK, and p38 protein kinases were specifically activated following expression of either a mutant of B-Raf, a truncated version of mitogen-activated/extracellular signal responsive kinase kinase kinase-1 (MEKK1), or a mutant of MAP kinase kinase-6 (MKK6), respectively. The results show that the DNA binding sites for serum response factor (SRF) and for the ternary complex factor (TCF) are of major importance for stimulating c-Fos promoter activity by MAP kinases. ERK1/2 and p38-induced stimulation of the c-Fos promoter additionally required the DNA binding site for the transcription factor AP-1. Mutation of the DNA binding site for STAT had no or only a small effect on c-Fos promoter activity. We conclude that MAP kinases do not activate distinct transcription factors involving distinct genetic elements. Rather, these kinases mainly target SRF and TCF proteins, leading to an activation of transcription of the c-Fos gene via the serum response element.


Subject(s)
Proto-Oncogene Proteins c-fos/genetics , Serum Response Factor/metabolism , TCF Transcription Factors/metabolism , Tamoxifen/analogs & derivatives , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Mutation , Promoter Regions, Genetic , Serum Response Element , Tamoxifen/pharmacology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL