Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.196
Filtrar
1.
Biomaterials ; 313: 122763, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39180917

RESUMO

Cuproptosis is a new kind of cell death that depends on delivering copper ions into mitochondria to trigger the aggradation of tricarboxylic acid (TCA) cycle proteins and has been observed in various cancer cells. However, whether cuproptosis occurs in cancer stem cells (CSCs) is unexplored thus far, and CSCs often reside in a hypoxic tumor microenvironment (TME) of triple negative breast cancers (TNBC), which suppresses the expression of the cuproptosis protein FDX1, thereby diminishing anticancer efficacy of cuproptosis. Herein, a ROS-responsive active targeting cuproptosis-based nanomedicine CuET@PHF is developed by stabilizing copper ionophores CuET nanocrystals with polydopamine and hydroxyethyl starch to eradicate CSCs. By taking advantage of the photothermal effects of CuET@PHF, tumor hypoxia is overcome via tumor mechanics normalization, thereby leading to enhanced cuproptosis and immunogenic cell death in 4T1 CSCs. As a result, the integration of CuET@PHF and mild photothermal therapy not only significantly suppresses tumor growth but also effectively inhibits tumor recurrence and distant metastasis by eliminating CSCs and augmenting antitumor immune responses. This study presents the first evidence of cuproptosis in CSCs, reveals that disrupting hypoxia augments cuproptosis cancer therapy, and establishes a paradigm for potent cancer therapy by simultaneously eliminating CSCs and boosting antitumor immunity.


Assuntos
Cobre , Nanomedicina , Células-Tronco Neoplásicas , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/terapia , Microambiente Tumoral/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Animais , Feminino , Nanomedicina/métodos , Cobre/química , Cobre/farmacologia , Linhagem Celular Tumoral , Camundongos , Nanopartículas/química , Camundongos Endogâmicos BALB C , Terapia Fototérmica/métodos , Humanos , Polímeros/química , Indóis/farmacologia
2.
Biomaterials ; 312: 122712, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39098305

RESUMO

Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.


Assuntos
Lipossomos , MicroRNAs , Fotoquimioterapia , Fármacos Fotossensibilizantes , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Animais , Humanos , Lipossomos/química , MicroRNAs/genética , MicroRNAs/metabolismo , Fotoquimioterapia/métodos , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Feminino , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/patologia , Camundongos , Aptâmeros de Nucleotídeos/química , Preparações de Ação Retardada/química , Interferência de RNA , Peixe-Zebra
3.
West Afr J Med ; 41(7): 767-774, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39356615

RESUMO

BACKGROUND: Breast cancer constitutes a significant public health issue in most resource-constrained nations due to its high morbidity and mortality rates. There is a paucity of knowledge of the molecular subtypes of breast cancer in Nigeria primarily due to the lack of immunohistochemistry. This study aims to identify the molecular subtypes of histologically confirmed breast cancer cases at the University of Benin Teaching Hospital, Benin City, Nigeria, using ER, PR and HER2/neu as immunohistochemical biomarkers. MATERIALS AND METHOD: Breast cancer specimens received in the Histopathology department of the University of Benin Teaching Hospital between 2019 and 2021 were used for this study. Representative sections of paraffin-embedded blocks were recut for histological typing, tumour grading, and immunohistochemistry. RESULTS: A total of 330 cases were evaluated in this study. The average age was 49 years, with a M:F of 40.3:1. The most frequent histological type was invasive breast cancer (92.1%). Two hundred and forty-two (73.3%) cases were categorized as grade II tumours. The steroid hormone receptor positivity was 39.4%. Oestrogen and Progesterone receptor positivity were 39.4% and 22.1%, respectively. HER2/neu was positive in 16.4% of the cases. Triple-negative breast cancer (TNBC) was the most common molecular subtype, accounting for 49.4% of cases. Luminal A, Luminal B, and HER2/neu enriched subtypes were each found in 34.2%, 5.2%, and 11.2% of cases, respectively. CONCLUSION: Triple-negative breast cancers predominated among the study population and were more common in high-grade tumours with unfavourable histological types and among women who were younger than their Caucasian counterparts.


CONTEXTE: Le cancer du sein constitue un problème de santé publique majeur dans la plupart des nations aux ressources limitées en raison de son taux élevé de morbidité et de mortalité. Il existe une pénurie de connaissances sur les sous-types moléculaires du cancer du sein au Nigeria, principalement en raison du manque d'immunohistochimie. Cette étude vise à identifier les sous-types moléculaires des cas de cancer du sein confirmés histologiquement à l'Hôpital Universitaire de Benin, Benin City, Nigeria, en utilisant les biomarqueurs immunohistochimiques ER, PR et HER2/neu. MATÉRIAUX ET MÉTHODE: Les échantillons de cancer du sein reçus dans le département d'histopathologie de l'Hôpital Universitaire de Benin entre 2019 et 2021 ont été utilisés pour cette étude. Des sections représentatives de blocs inclus en paraffine ont été recoupées pour le typage histologique, le classement des tumeurs et l'immunohistochimie. RÉSULTATS: Un total de 330 cas ont été évalués dans cette étude. L'âge moyen était de 49 ans, avec un rapport H de 40,3:1. Le type histologique le plus fréquent était le cancer du sein invasif (92,1 %). Deux cent quarante-deux (73,3 %) cas ont été classés comme des tumeurs de grade II. La positivité des récepteurs hormonaux stéroïdiens était de 39,4 %. Les récepteurs des œstrogènes et de la progestérone étaient positifs dans 39,4 % et 22,1 % des cas, respectivement. HER2/neu était positif dans 16,4 % des cas. Le cancer du sein triple négatif (CSTN) était le sous-type moléculaire le plus courant, représentant 49,4 % des cas. Les sous-types Luminal A, Luminal B et enrichi en HER2/neu ont été trouvés dans 34,2 %, 5,2 % et 11,2 % des cas, respectivement. CONCLUSION: Les cancers du sein triple négatif prédominaient parmi la population étudiée et étaient plus fréquents dans les tumeurs de haut grade avec des types histologiques défavorables et chez les femmes plus jeunes que leurs homologues caucasiennes.


Assuntos
Neoplasias da Mama , Receptores de Progesterona , Humanos , Feminino , Nigéria/epidemiologia , Pessoa de Meia-Idade , Neoplasias da Mama/patologia , Neoplasias da Mama/epidemiologia , Adulto , Receptores de Progesterona/metabolismo , Receptor ErbB-2/metabolismo , Biomarcadores Tumorais/metabolismo , Idoso , Receptores de Estrogênio/metabolismo , Centros de Atenção Terciária , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/epidemiologia , Imuno-Histoquímica , Masculino , Gradação de Tumores , Adulto Jovem
5.
BMC Cancer ; 24(1): 1166, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300407

RESUMO

BACKGROUND: The prognosis of breast cancer patients with visceral metastasis (VM) is significantly worse than that of patients without VM. We aimed to evaluate EZH2 (enhancer of zeste homolog 2) mutation as a biomarker associated with VM. METHODS: Data from forty-nine patients with metastatic breast cancer (MBC) pathologically confirmed at our hospital between March 2016 and September 2018 were collected. Metastatic tissue samples were obtained via ultrasound-guided needle biopsy, and paired peripheral blood samples were also collected. Tissue and blood samples were subjected to targeted next-generation sequencing via a 247-gene panel. Stably transfected MDA-MB-231 cells expressing wild-type EZH2 (EZH2WT) or a mutant form of EZH2 (EZH2K515R) were generated. Cell proliferation, colony formation ability, migration and invasion abilities and apoptosis were assessed using CCK-8 assays, plate colony formation assays, Transwell chamber assays and flow cytometry. RESULTS: The incidence of EZH2 mutations in the VM subgroup was greater than that in the non-VM subgroup in the entire cohort (n = 49, 42.3% vs. 13.0%, p = 0.024) and in the triple-negative breast cancer (TNBC) subgroup (n = 20, 50.0% vs. 10.0%, p = 0.05). Patients carrying EZH2 mutations had a significantly greater risk of developing VM than did those in the non-EZH2 mutation group in the entire cohort (HR 2.9) and in the TNBC subgroup (HR 6.45). Multivariate analysis revealed that EZH2 mutation was an independent prognostic factor for VM (HR 2.99, p = 0.009) in the entire cohort and in the TNBC subgroup (HR 10.1, p = 0.006). Data from cBioPortal also showed that patients with EZH2 mutations had a significantly greater risk of developing VM (HR 3.1), and the time to develop VM was significantly earlier in the EZH2 mutation group (31.5 months vs. 109.7 months, p = 0.008). Multivariate analysis revealed that EZH2 mutation (HR 2.73, p = 0.026) was an independent factor for VM after breast cancer surgery. There was no correlation between EZH2 mutations and BRCA1/2 mutations. Most of the patients (81.8%) in our cohort who developed VM carried the "c.1544A > G (p.K515R)" mutation. Compared with EZH2WT MDA-MB-231 cells, EZH2K515R MDA-MB-231 cells had greater colony formation rates (p < 0.01), greater migration and invasion rates (p < 0.001), and lower apoptosis rates (p < 0.01). The proportion of S + G2/M phase cells in the EZH2K515R group was significantly greater than that in the EZH2WT group. CONCLUSIONS: EZH2 mutation is associated with VM development in breast cancer patients. The EZH2K515R mutation leads to VM and a poor prognosis by enhancing proliferation and invasion and inhibiting apoptosis in breast cancer cells.


Assuntos
Apoptose , Neoplasias da Mama , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste , Mutação , Invasividade Neoplásica , Humanos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Apoptose/genética , Proliferação de Células/genética , Pessoa de Meia-Idade , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Prognóstico , Linhagem Celular Tumoral , Adulto , Idoso , Biomarcadores Tumorais/genética , Movimento Celular/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
6.
Biomed Pharmacother ; 179: 117325, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39226729

RESUMO

Direct-acting antivirals ledipasvir (LDV) and daclatasvir (DCV) are widely used as part of combination therapies to treat Hepatitis C infections. Here we show that these compounds inhibit the proliferation, invasion, and colony formation of triple-negative MDA-MB-231 breast cancer cells, SRC-transduced SW620 colon cancer cells and SRC- transduced NIH3T3 fibroblasts. DCV also inhibits the expression of PDL-1, which is responsible for resistance to immunotherapy in breast cancer cells. The demonstrated low toxicity in many Hepatitis C patients suggests LDV and DCV could be used in combination therapies for cancer patients. At the molecular level, these direct-acting antivirals inhibit the phosphorylation of Akt and the ephrin type A receptor 2 (EPHA2) by destabilizing a Src-EPHA2 complex, although they do not affect the general kinase activity of Src. Thus, LDV and DCV could be effective drugs for Src-associated cancers without the inherent toxicity of classical Src inhibitors.


Assuntos
Antivirais , Benzimidazóis , Carbamatos , Neoplasias Colorretais , Regulação para Baixo , Fluorenos , Imidazóis , Proteínas Proto-Oncogênicas c-akt , Pirrolidinas , Transdução de Sinais , Neoplasias de Mama Triplo Negativas , Valina , Quinases da Família src , Humanos , Benzimidazóis/farmacologia , Animais , Pirrolidinas/farmacologia , Imidazóis/farmacologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinases da Família src/metabolismo , Fluorenos/farmacologia , Linhagem Celular Tumoral , Antivirais/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/genética , Carbamatos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Valina/análogos & derivados , Valina/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Transdução de Sinais/efeitos dos fármacos , Células NIH 3T3 , Feminino , Proliferação de Células/efeitos dos fármacos , United States Food and Drug Administration , Aprovação de Drogas , Estados Unidos
7.
BMC Cancer ; 24(1): 1214, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39350055

RESUMO

BACKGROUND: Pre-clinical data suggests a potential synergistic effect of eribulin and platinum. However, clinical data on the combination for metastatic breast cancer (mBC) is lacking. We evaluated the efficacy and safety of eribulin plus carboplatin (ErCb) in patients with mBC. PATIENTS AND METHODS: This multicenter, real-world cohort study included patients with pre-treated metastatic triple negative breast cancer (TNBC) or endocrine-refractory hormone receptor (HR) positive, HER2-negative mBC who received ErCb. Eribulin (1.4 mg/m2) and carboplatin (target AUC = 2) were administered intravenously on day 1 and 8 of 21-day cycle. Objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were evaluated. RESULTS: From March 2022 to December 2023, a cohort of 37 patients were recruited to the study. Among them, 22 patients have TNBC and 15 have HR + HER2 - mBC. Of the 22 patients with TNBC, 8 had an initial diagnosis of the HR + HER2 - subtype. The median treatment was 6 cycles (range, 2 - 8 cycles). In the full cohort, TNBC, and HR + HER2 - subgroup, the ORR were 51.4%, 54.5% and 46.7%, the DCR were 81.1%, 81.8% and 80%, and the median PFS were 5 months, 5 months, and 5.2 months, respectively. The median OS was 12.7 months in the entire cohort and 12.8 months in TNBC subgroup. The most common grade 3/4 hematological AEs were neutropenia (37.8%), leukopenia (35.1%), febrile neutropenia (10.8%), thrombocytopenia (5.4%), and anemia (2.7%). No grade 3/4 non-hematological AEs were observed. CONCLUSION: ErCb demonstrated favorable efficacy and tolerability in patients with heavily pre-treated mBC, especially TNBC. The findings of the current study warrant further investigation of the application of this combination in earlier lines of mBC treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Carboplatina , Furanos , Cetonas , Receptor ErbB-2 , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Pessoa de Meia-Idade , Cetonas/uso terapêutico , Cetonas/administração & dosagem , Cetonas/efeitos adversos , Carboplatina/administração & dosagem , Carboplatina/uso terapêutico , Furanos/uso terapêutico , Furanos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Idoso , Adulto , Receptor ErbB-2/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/mortalidade , Estudos de Coortes , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/metabolismo , Metástase Neoplásica , Policetídeos de Poliéter
8.
BMC Cancer ; 24(1): 1217, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39350059

RESUMO

OBJECTIVE: In this study, we evaluated the molecular mechanisms of HuangQiSiJunZi Decoction (HQSJZD) for treating triple-negative breast cancer (TNBC) using network pharmacology and bioinformatics analyses. METHODS: Effective chemical components together with action targets of HQSJZD were selected based on the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Meanwhile, differentially expressed genes (DEGs) were extracted from TNBC sample data in The Cancer Genome Atlas (TCGA) database. Additionally, we built a protein-protein interaction (PPI) network and acquired hub genes. Gene Expression Omnibus(GEO) datasets were utilized to verify the accuracy of hub gene expression. Additionally, enrichment analyses were conducted on key genes. Furthermore, TNBC severity-related high-risk factors were screened through univariate together with multivariate Cox regressions; next, the logistic regression prediction model was built. Moreover, differential levels of 22 immune cell types in TNBC tissues compared with normal tissues were analyzed. The hub gene levels within pan-cancer and the human body were subsequently visualized and analyzed. Finally, quantitative PCR (RT-qPCR) was used to validate the correlation of the hub genes in TNBC cells. RESULTS: The study predicted 256 targets of active ingredients and 1791 DEGs in TNBC, and obtained 16 hub genes against TNBC. The prognostic signature based on FOS, MMP9, and PGR was independent in predicting survival. A total of seven types of immune cells, such as CD4 + memory T cells, showed a significant difference in infiltration (p < 0.05), and immune cells were related to the hub genes. The HPA database was adopted for hub gene analyses, and as determined, FOS was highly expressed in most human organs. The results of RT-qPCR validation for the FOS hub gene were consistent with those of bioinformatic analyses. CONCLUSION: HQSJZD might regulate the interleukin-17 and aging pathways via FOS genes to increase immune cell infiltration in TNBC tissues, and thus, may treat TNBC and improve the prognosis. The FOS genes are likely to be a new marker for TNBC.


Assuntos
Biologia Computacional , Medicamentos de Ervas Chinesas , Regulação Neoplásica da Expressão Gênica , Farmacologia em Rede , Mapas de Interação de Proteínas , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Humanos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Biologia Computacional/métodos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Prognóstico , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo
9.
Cells ; 13(18)2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39329736

RESUMO

Epithelial-to-mesenchymal transition (EMT) is a complex cellular process that allows cells to change their phenotype from epithelial to mesenchymal-like. Type 3 EMT occurs during cancer progression. The aim of this study was to investigate the role of RNA-binding motif single-stranded interacting protein 3 (RBMS 3) in the process of EMT. To investigate the impact of RBMS 3 on EMT, we performed immunohistochemical (IHC) reactions on archived paraffin blocks of invasive ductal breast carcinoma (n = 449), allowing us to analyze the correlation in expression between RBMS 3 and common markers of EMT. The IHC results confirmed the association of RBMS 3 with EMT markers. Furthermore, we performed an in vitro study using cellular models of triple negative and HER-2-enriched breast cancer with the overexpression and silencing of RBMS 3. RT-qPCR and Western blot methods were used to detect changes at both the mRNA and protein levels. An invasion assay and confocal microscopy were used to study the migratory potential of cells depending on the RBMS 3 expression. The studies conducted suggest that RBMS 3 may potentially act as an EMT-promoting agent in the most aggressive subtype of breast cancer, triple negative breast cancer (TNBC), but as an EMT suppressor in the HER-2-enriched subtype. The results of this study indicate the complex role of RBMS 3 in regulating the EMT process and present it as a future potential target for personalized therapies and a diagnostic marker in breast cancer.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas de Ligação a RNA , Neoplasias de Mama Triplo Negativas , Transição Epitelial-Mesenquimal/genética , Humanos , Feminino , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Linhagem Celular Tumoral , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Pessoa de Meia-Idade , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Progressão da Doença
10.
Sci Rep ; 14(1): 22487, 2024 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-39341857

RESUMO

Triple negative breast cancer (TNBC) subtype is characterized with higher EMT/stemness properties and immune suppressive tumor microenvironment (TME). Women with advanced TNBC exhibit aggressive disease and have limited treatment options. Although immune suppressive TME is implicated in driving aggressive properties of basal/TNBC subtype and therapy resistance, effectively targeting it remains a challenge. Minnelide, a prodrug of triptolide currently being tested in clinical trials, has shown anti-tumorigenic activity in multiple malignancies via targeting super enhancers, Myc and anti-apoptotic pathways such as HSP70. Distinct super-enhancer landscape drives cancer stem cells (CSC) in TNBC subtype while inducing immune suppressive TME. We show that Minnelide selectively targets CSCs in human and murine TNBC cell lines compared to cell lines of luminal subtype by targeting Myc and HSP70. Minnelide in combination with cyclophosphamide significantly reduces the tumor growth and eliminates metastasis by reprogramming the tumor microenvironment and enhancing cytotoxic T cell infiltration in 4T1 tumor-bearing mice. Resection of residual tumors following the combination treatment leads to complete eradication of disseminated tumor cells as all mice are free of local and distant recurrences. All control mice showed recurrences within 3 weeks of post-resection while single Minnelide treatment delayed recurrence and one mouse was free of tumor. We provide evidence that Minnelide targets tumor intrinsic pathways and reprograms the immune suppressive microenvironment. Our studies also suggest that Minnelide in combination with cyclophosphamide may lead to durable responses in patients with basal/TNBC subtype warranting its clinical investigation.


Assuntos
Diterpenos , Compostos de Epóxi , Células-Tronco Neoplásicas , Fenantrenos , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Humanos , Animais , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/imunologia , Feminino , Camundongos , Linhagem Celular Tumoral , Compostos de Epóxi/farmacologia , Compostos de Epóxi/uso terapêutico , Fenantrenos/farmacologia , Fenantrenos/uso terapêutico , Diterpenos/farmacologia , Diterpenos/uso terapêutico , Ciclofosfamida/farmacologia , Ciclofosfamida/uso terapêutico , Camundongos Endogâmicos BALB C , Organofosfatos
11.
Sci Rep ; 14(1): 22541, 2024 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-39341960

RESUMO

Tumor-associated macrophages (TAMs) originating from monocytes are crucial for cancer progression; however, the mechanism of TAM differentiation is unclear. We investigated factors involved in the differentiation of monocytes into TAMs within the tumor microenvironment of triple-negative breast cancer (TNBC). We screened 172 compounds and found that a heat shock protein 90 (HSP90) inhibitor blocked TNBC-induced monocyte-to-TAM differentiation in human monocytes THP-1. TNBC-derived conditional medium (CM) activated cell signaling pathways, including MAP kinase, AKT and STAT3, and increased the expression of TAM-related genes and proteins. These inductions were suppressed by HSP90 inhibition or by knockdown of HSP90 in TNBC. Additionally, we confirmed that TNBC secreted HSP90 extracellularly and that HSP90 itself promoted TAM differentiation. In a mouse tumor model, treatment with an HSP90 inhibitor suppressed tumor growth and reduced TAMs in the tumor microenvironment. Our findings demonstrate the role of HSP90 in TAM differentiation, suggesting HSP90 as a potential target for TNBC immunotherapy due to its regulatory role in monocyte-to-TAM differentiation.


Assuntos
Diferenciação Celular , Proteínas de Choque Térmico HSP90 , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Macrófagos Associados a Tumor , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Humanos , Animais , Diferenciação Celular/efeitos dos fármacos , Feminino , Camundongos , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/imunologia , Progressão da Doença , Linhagem Celular Tumoral , Monócitos/metabolismo , Transdução de Sinais
12.
Zhonghua Zhong Liu Za Zhi ; 46(9): 878-888, 2024 Sep 23.
Artigo em Chinês | MEDLINE | ID: mdl-39293991

RESUMO

Objectives: To observe the mitochondrial morphology of normal and triple-negative breast cancer cells, extract mitochondria from normal cells, and investigate the effects of mitochondrial transplantation on proliferation, apoptosis, and stemness of triple-negative breast cancer cells. Methods: The morphology of mitochondria was observed by transmission electron microscope. Mitochondria were extracted by mitochondrial extraction kit, mitochondrial protein was identified by western blot, and mitochondrial activity was detected by mitochondrial membrane potential detection kit. MitoTracker Green or MitoTracker Deep Red fluorescent probes were used to label the mitochondria of living cells, and the degree of mitochondria entering LTT cells was observed by confocal laser microscopy at 12, 24, and 96 hours. The effects of mitochondrial transplantation on proliferation, apoptosis, and stemness of breast cancer cells were examined by CCK8, colony formation assay, flow cytometry, and sphere formation assay after 24 hours of mitochondrial transplantation. Results: The mitochondria of normal cells were rod-shaped or elongated, while the mitochondria of triple-negative breast cancer cells were swollen and vacuolated. Western blot results showed that cytochrome c oxidase subunit I (MT-CO1) protein encoded by mitochondria was present in the isolated mitochondria. The content of heat shock protein 60 (HSP60) was higher in mitochondria than that in cytoplasm. The result of the multi-mode microplate reader showed that the content of mitochondrial J-aggregates/monomer was 1.67±0.06, which was significantly higher than 0.35±0.04 of the control group (P<0.001). Exogenous mitochondria were observed in LTT cells at 12, 24, and 96 hours after mitochondrial transplantation. The results of the CCK8 experiment showed that OD450 of LTT cells was 0.27±0.13 after 48 hours transplantation, which was lower than 0.62±0.36 of the control group (P=0.023). The OD450 of MDA-MB-468 cells was 0.30±0.03, which was lower than 0.65±0.10 of the control group (P=0.004). After 120 hours of mitochondrial transplantation, OD450 in both groups was still significantly lower than that in the control group (P<0.01). The number of clones formed by mitochondrial transplantation of LTT cells was 21.33±7.31, which was lower than 35.22±13.59 of the control group (P=0.016). Flow cytometry showed that the early apoptosis rate of LTT cells was (30.07±2.15)% after 24 hours of mitochondrial transplantation, which was higher than 2.07±1.58 of the control group (P<0.001). The proportion of early apoptosis in MDA-MB-468 cells was 24.47%±5.22%, which was higher than (7.83±2.06)% in the control group (P=0.007). In addition, the number of mitochondria transplanted LTT cells into the cell sphere was 46.25±5.40, which was significantly lower than 62.58±6.43 of the control group (P<0.001). Conclusion: Normal mitochondria can enter triple-negative breast cancer cells by co-culture, inhibit the proliferation and stemness of triple-negative breast cancer cells, and promote the apoptosis of triple-negative breast cancer cells.


Assuntos
Apoptose , Proliferação de Células , Potencial da Membrana Mitocondrial , Mitocôndrias , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Mitocôndrias/metabolismo , Humanos , Linhagem Celular Tumoral , Feminino
13.
Cancer Lett ; 603: 217200, 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-39222677

RESUMO

Triple-negative breast cancer (TNBC) is difficult to treat breast cancer subtype due to lack or insignificant expressions of targetable estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2). Therefore, finding a targetable protein or signaling pathway in TNBC would impact patient care. Here, we report that a member of the Mixed Lineage Kinase (MLK) family, MLK3, is an effector of G-protein-coupled protease-activated receptors 1 (PAR1) and targeting MLK3 by a small-molecule inhibitor prevented PAR1-mediated TNBC tumorigenesis. In silico and immunohistochemistry analysis of human breast tumors showed overexpression of PAR1 and MLK3 in TNBC tumors. Treating α-thrombin and PAR1 agonist increased MLK3 and JNK activities and induced cell migration in TNBC cells. The PAR1 positive/high (PAR1+/hi) population of TNBC cells showed aggressive tumor phenotype with increased MLK3 signaling. Moreover, combined inhibition of the PAR1 and MLK3 mitigated the TNBC tumor burden in preclinical TNBC models. Our data suggests that activation of the PAR1-MLK3 axis promotes TNBC tumorigenesis. Therefore, combinatorial therapy targeting MLK3 and PAR1 could effectively reduce TNBC tumor burden.


Assuntos
MAP Quinase Quinase Quinases , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno , Receptor PAR-1 , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Humanos , Receptor PAR-1/metabolismo , Receptor PAR-1/genética , Feminino , Animais , Linhagem Celular Tumoral , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/genética , Movimento Celular , Transdução de Sinais , Inibidores de Proteínas Quinases/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Carcinogênese/metabolismo , Carcinogênese/genética , Camundongos , Proliferação de Células
14.
Diagn Pathol ; 19(1): 121, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39243111

RESUMO

OBJECTIVES: We explored Trichorhinophalangeal syndrome type 1 (TRPS1) expression in special types of breast carcinoma, and analyzed the correlation between TRPS1 and androgen receptor (AR) expression in triple-negative breast cancer (TNBC). METHODS: TRPS1 expression was analyzed in 801 patients with special types of breast carcinoma. A total of 969 TNBC were used to analyze the correlation between the expression of TRPS1 and AR. TRPS1 expression was evaluated in 1975 cases of breast cancer with different molecular types. RESULTS: A total of 801 special types of breast cancers were stained with TRPS1.TRPS1 was positive in 100% (63/63) of mucinous carcinoma, 100% (7/7) adenoid cystic carcinomas (4 classic adenoid cystic carcinomas and 3 solid-basaloid adenoid cystic carcinomas), 100% (4/4) tubular carcinomas, 100% (2/2) secretory carcinomas, and 99.59% (243/244) invasive lobular carcinomas, 99.26% (267/269) invasive micropapillary carcinomas, 97.44% (38/39) ER-positive neuroendocrine tumors, 94.44% (34/36) metaplastic breast carcinomas (MBCs), 63.73% (65/102) apocrine carcinomas. TRPS1 was negative in all triple-negative neuroendocrine carcinomas (0/7).TRPS1 was positive in 92.86% (26/28) of metastatic special types of breast cancer. TRPS1 and AR expression were analyzed in 969 cases of TNBC. 90.40% were positive for TRPS1, and 42.41% were positive for AR. A significant inverse correlation between TRPS1 and AR expression was shown in TNBC (p < .001). TRPS1 showed a higher positive rate (93.13%) in TNBC compared to GATA binding protein 3 (GATA3), gross cystic disease fluid protein 15 (GCDFP-15) and forkhead box transcription Factor C 1 (FOXC1). CONCLUSIONS: In conclusion, our study demonstrated that TRPS1 is a highly sensitive marker for most special types of breast carcinoma. TRPS1 was positive in 63.73% of apocrine carcinomas. TRPS1 and AR expression was inversely correlated in TNBC.


Assuntos
Biomarcadores Tumorais , Proteínas de Ligação a DNA , Receptores Androgênicos , Proteínas Repressoras , Fatores de Transcrição , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Biomarcadores Tumorais/análise , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/genética , Proteínas Repressoras/análise , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/análise , Receptores Androgênicos/análise , Receptores Androgênicos/genética , Imuno-Histoquímica , Pessoa de Meia-Idade , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Adulto
15.
Cell Rep ; 43(9): 114751, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39276346

RESUMO

Triple negative breast cancer (TNBC) is an aggressive type of breast cancer. While most TNBCs are initially sensitive to chemotherapy, a substantial fraction acquires resistance to treatments and progresses to more advanced stages. Here, we identify the spliceosome U2 small nuclear ribonucleoprotein particle (snRNP) complex as a modulator of chemotherapy efficacy in TNBC. Transient U2 snRNP inhibition induces persistent DNA damage in TNBC cells and organoids, regardless of their homologous recombination proficiency. U2 snRNP inhibition pervasively deregulates genes involved in the DNA damage response (DDR), an effect relying on their genomic structure characterized by a high number of small exons. Furthermore, a pulse of splicing inhibition elicits long-lasting repression of DDR proteins and enhances the cytotoxic effect of platinum-based drugs and poly(ADP-ribose) polymerase inhibitors (PARPis) in multiple TNBC models. These findings identify the U2 snRNP as an actionable target that can be exploited to enhance chemotherapy efficacy in TNBCs.


Assuntos
Dano ao DNA , Splicing de RNA , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Humanos , Feminino , Splicing de RNA/efeitos dos fármacos , Splicing de RNA/genética , Linhagem Celular Tumoral , Animais , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Camundongos , Spliceossomos/metabolismo , Spliceossomos/efeitos dos fármacos
16.
Biomolecules ; 14(9)2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39334853

RESUMO

Breast cancer (BC) is the most frequently diagnosed cancer and the primary cause of cancer-related mortality in women. Treatment of triple-negative breast cancer (TNBC) remains particularly challenging due to its resistance to chemotherapy and poor prognosis. Extensive research efforts in BC screening and therapy have improved clinical outcomes for BC patients. Therefore, identifying reliable biomarkers for TNBC is of great clinical importance. Here, we found that tyrosine aminotransferase (TAT) expression was significantly reduced in BC and strongly correlated with the poor prognosis of BC patients, which distinguished BC patients from normal individuals, indicating that TAT is a valuable biomarker for early BC diagnosis. Mechanistically, we uncovered that methylation of the TAT promoter was significantly increased by DNA methyltransferase 3 (DNMT3A/3B). In addition, reduced TAT contributes to DNA replication and cell cycle activation by regulating homologous recombination repair and mismatch repair to ensure genomic stability, which may be one of the reasons for TNBC resistance to chemotherapy. Furthermore, we demonstrated that Diazinon increases TAT expression as an inhibitor of DNMT3A/3B and inhibits the growth of BC by blocking downstream pathways. Taken together, we revealed that TAT is silenced by DNMT3A/3B in BC, especially in TNBC, which promotes the proliferation of tumor cells by supporting DNA replication, activating cell cycle, and enhancing DNA damage repair. These results provide fresh insights and a theoretical foundation for the clinical diagnosis and treatment of BC.


Assuntos
Biomarcadores Tumorais , Ciclo Celular , DNA Metiltransferase 3A , Reparo do DNA , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Reparo do DNA/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , DNA Metiltransferase 3A/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , DNA Metiltransferase 3B , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Regiões Promotoras Genéticas/genética , Prognóstico
17.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39337269

RESUMO

Previously, we have demonstrated that amiodarone (AM), a widely used antiarrhythmic drug, and its major metabolite desethylamiodarone (DEA) both affect several mitochondrial processes in isolated heart and liver mitochondria. Also, we have established DEA's antitumor properties in various cancer cell lines and in a rodent metastasis model. In the present study, we compared AM's and DEA's mitochondrial and antineoplastic effects in a human triple-negative breast cancer (TNBC) cell line. Both compounds reduced viability in monolayer and sphere cultures and the invasive growth of the MDA-MB-231 TNBC line by inducing apoptosis. They lowered mitochondrial trans-membrane potential, increased Ca2+ influx, induced mitochondrial permeability transition, and promoted mitochondrial fragmentation. In accordance with their mitochondrial effects, both substances massively decreased overall, and even to a greater extent, mitochondrial ATP production decreased, as determined using a Seahorse live cell respirometer. In all these effects, DEA was more effective than AM, indicating that DEA may have higher potential in the therapy of TNBC than its parent compound.


Assuntos
Amiodarona , Antineoplásicos , Apoptose , Mitocôndrias , Neoplasias de Mama Triplo Negativas , Amiodarona/farmacologia , Amiodarona/análogos & derivados , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Feminino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos
18.
Mol Cancer ; 23(1): 188, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39243032

RESUMO

Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFß, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.


Assuntos
Proliferação de Células , Células Supressoras Mieloides , Células-Tronco Neoplásicas , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Humanos , Animais , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Feminino , Camundongos , Células Supressoras Mieloides/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral , Prognóstico , Movimento Celular , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/imunologia , Proteínas de Neoplasias
19.
ACS Appl Mater Interfaces ; 16(39): 51783-51797, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39291812

RESUMO

Copper plays a vital role in cellular metabolism and oxidative stress regulation. Visualizing and controlling the copper level in mitochondrion have been proven to be promising and efficient strategies for the diagnosis and treatment of triple-negative breast cancer (TNBC). However, developing an advanced probe for simultaneous visualization and depletion of mitochondrial copper remains a huge challenge. Herein, we for the first time report a mitochondria-anchorable, copper-responsive, and depleting probe d-IR-DPA and evaluate its potential for quantitative visualization of intratumoral copper(II) and anti-TNBC in vivo. Taking advantage of the mitochondrion-targeting and sulfenated-protein-mediated covalent immobilization characteristics, this probe not only enables the quantitative detection of Cu2+ levels in various types of tumors through ratiometric photoacoustic (PA680 nm/PA800 nm) imaging but also scavenges the mitochondrial Cu2+, simultaneously igniting increased oxidative stress and mitochondrial membrane damage and eventually leading to severe TNBC cell apoptosis. More notably, the depletion of Cu2+ by d-IR-DPA can alter the cellular metabolic pathway from oxidative phosphorylation to glycolysis, inducing energy deprivation and significant suppression of TNBC tumor in living mice. Our probe may provide a valuable and powerful means for the effective treatment of TNBC as well as other copper-associated diseases.


Assuntos
Cobre , Mitocôndrias , Neoplasias de Mama Triplo Negativas , Cobre/química , Animais , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Humanos , Camundongos , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Feminino , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos
20.
Molecules ; 29(18)2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39339370

RESUMO

In this study, novel ergosterol peroxide (EP) derivatives were synthesized and evaluated to assess their antiproliferative activity against four human cancer cell lines (A549, HepG2, MCF-7, and MDA-MB-231). Compound 3g exhibited the most potent antiproliferative activity, with an IC50 value of 3.20 µM against MDA-MB-231. This value was 5.4-fold higher than that of the parental EP. Bioassay optimization further identified 3g as a novel glutaminase 1 (GLS1) inhibitor (IC50 = 3.77 µM). In MDA-MB-231 cells, 3g reduced the cellular glutamate levels by blocking the glutamine hydrolysis pathway, which triggered reactive oxygen species production and induced caspase-dependent apoptosis. Molecular docking indicated that 3g interacts with the reaction site of the variable binding pocket by forming multiple interactions with GLS1. In a mouse model of breast cancer, 3g showed remarkable therapeutic effects at a dose of 50 mg/kg, with no apparent toxicity. Based on these results, 3g could be further evaluated as a novel GLS1 inhibitor for triple-negative breast cancer (TNBC) therapy.


Assuntos
Antineoplásicos , Proliferação de Células , Ergosterol , Glutaminase , Simulação de Acoplamento Molecular , Neoplasias de Mama Triplo Negativas , Humanos , Glutaminase/antagonistas & inibidores , Glutaminase/metabolismo , Animais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Camundongos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Ergosterol/análogos & derivados , Ergosterol/química , Ergosterol/farmacologia , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/síntese química , Ensaios Antitumorais Modelo de Xenoenxerto , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA