Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.869
Filtrer
1.
Biomaterials ; 313: 122763, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39180917

RÉSUMÉ

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.


Sujet(s)
Cuivre , Nanomédecine , Cellules souches tumorales , Tumeurs du sein triple-négatives , Microenvironnement tumoral , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/thérapie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Animaux , Femelle , Nanomédecine/méthodes , Cuivre/composition chimique , Cuivre/pharmacologie , Lignée cellulaire tumorale , Souris , Nanoparticules/composition chimique , Souris de lignée BALB C , Thérapie photothermique/méthodes , Humains , Polymères/composition chimique , Indoles/pharmacologie
2.
Biomaterials ; 313: 122796, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39226654

RÉSUMÉ

Chemotherapy-induced cellular senescence leads to an increased proportion of cancer stem cells (CSCs) in breast cancer (BC), contributing to recurrence and metastasis, while effective means to clear them are currently lacking. Herein, we aim to develop new approaches for selectively killing senescent-escape CSCs. High CD276 (95.60%) expression in multidrug-resistant BC cells, facilitates immune evasion by low-immunogenic senescent escape CSCs. CALD1, upregulated in ADR-resistant BC, promoting senescent-escape of CSCs with an anti-apoptosis state and upregulating CD276, PD-L1 to promote chemoresistance and immune escape. We have developed a controlled-released thermosensitive hydrogel containing pH- responsive anti-CD276 scFV engineered biomimetic nanovesicles to overcome BC in primary, recurrent, metastatic and abscopal humanized mice models. Nanovesicles coated anti-CD276 scFV selectively fuses with cell membrane of senescent-escape CSCs, then sequentially delivers siCALD1 and ADR due to pH-responsive MnP shell. siCALD1 together with ADR effectively induce apoptosis of CSCs, decrease expression of CD276 and PD-L1, and upregulate MHC I combined with Mn2+ to overcome chemoresistance and promote CD8+T cells infiltration. This combined therapeutic approach reveals insights into immune surveillance evasion by senescent-escape CSCs, offering a promising strategy to immunotherapy effectiveness in cancer therapy.


Sujet(s)
Tumeurs du sein , Vieillissement de la cellule , Résistance aux médicaments antinéoplasiques , Cellules souches tumorales , Humains , Animaux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/thérapie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Génie génétique/méthodes , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Nanoparticules/composition chimique , Anticorps à chaîne unique/composition chimique , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Biomimétique/méthodes , Antigènes B7
4.
Cell Mol Life Sci ; 81(1): 417, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39367978

RÉSUMÉ

The existence of cancer stem cells (CSCs) in pancreatic ductal adenocarcinoma (PDAC) is considered to be the key factor for metastasis and chemoresistance. Thus, novel therapeutic strategies for eradicating CSCs are urgently needed. Here we aimed to explore the role of KLF15 in stemness and the feasibility of using KLF15 to inhibit CSCs and improve chemotherapy sensitivity in PDAC. In this study, we report that KLF15 is negatively associated with poor survival and advanced pathological staging of PDAC. Moreover, tumorous KLF15 suppresses the stemness of PDAC by promoting the degradation of Nanog, and KLF15 directly interacts with Nanog, inhibiting interaction between Nanog with USP21. We also demonstrate that the KLF15/Nanog complex inhibit the stemness in vivo and in PDX cells. Tazemetostat suppresses stemness and sensitizes PDAC cells to gemcitabine by promoting KLF15 expression in PDAC. In summary, the findings of our study confirm the value of KLF15 level in diagnosis and prognosis of PDAC, it is the first time to explore the inhibition role of KLF15 in stemness of PDAC and the regulation mechanism of Nanog, contributing to provide a new therapeutic strategy that using Tazemetostat sensitizes PDAC cells to gemcitabine by promoting KLF15 expression for PDAC.


Sujet(s)
Carcinome du canal pancréatique , Facteurs de transcription Krüppel-like , Protéine homéotique Nanog , Cellules souches tumorales , Tumeurs du pancréas , Ubiquitin thiolesterase , Humains , Protéine homéotique Nanog/métabolisme , Protéine homéotique Nanog/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Animaux , Souris , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Lignée cellulaire tumorale , Femelle , Mâle , Régulation de l'expression des gènes tumoraux , Souris nude , Gemcitabine , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Pronostic
5.
Biomolecules ; 14(9)2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39334933

RÉSUMÉ

Inhalation anesthesia stands as a pivotal modality within clinical anesthesia practices. Beyond its primary anesthetic effects, inhaled anesthetics have non-anesthetic effects, exerting bidirectional influences on the physiological state of the body and disease progression. These effects encompass impaired cognitive function, inhibition of embryonic development, influence on tumor progression, and so forth. For many years, inhaled anesthetics were viewed as inhibitors of stem cell fate regulation. However, there is now a growing appreciation that inhaled anesthetics promote stem cell biological functions and thus are now regarded as a double-edged sword affecting stem cell fate. In this review, the effects of inhaled anesthetics on self-renewal and differentiation of neural stem cells (NSCs), embryonic stem cells (ESCs), and cancer stem cells (CSCs) were summarized. The mechanisms of inhaled anesthetics involving cell cycle, metabolism, stemness, and niche of stem cells were also discussed. A comprehensive understanding of these effects will enhance our comprehension of how inhaled anesthetics impact the human body, thus promising breakthroughs in the development of novel strategies for innovative stem cell therapy approaches.


Sujet(s)
Anesthésiques par inhalation , Différenciation cellulaire , Cellules souches neurales , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Anesthésiques par inhalation/pharmacologie , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Animaux , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Auto-renouvellement cellulaire/effets des médicaments et des substances chimiques
6.
BMC Cancer ; 24(1): 1164, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39300412

RÉSUMÉ

The intricate interplay of cancer stem cell plasticity, along with the bidirectional transformation between epithelial-mesenchymal states, introduces further intricacy to offer insights into newer therapeutic approaches. Differentiation therapy, while successful in targeting leukemic stem cells, has shown limited overall success, with only a few promising instances. Using colon carcinoma cell strains with sequential p53/p73 knockdowns, our study underscores the association between p53/p73 and the maintenance of cellular plasticity. Morphological alterations corresponding with cell surface marker expressions, transcriptome analysis and functional assays were performed to access stemness and EMT (Epithelial-Mesenchymal Transition) characteristics in the spectrum of cells exhibiting sequential p53 and p73 knockdowns. Notably, our investigation explores the effectiveness of esculetin in reversing the shift from an epithelial to a mesenchymal phenotype, characterized by stem cell-like traits. Esculetin significantly induces enterocyte differentiation and promotes epithelial cell polarity by altering Wnt axes in Cancer Stem Cell-like cells characterized by high mesenchymal features. These results align with our previous findings in leukemic blast cells, establishing esculetin as an effective differentiating agent in both Acute Myeloid Leukemia (AML) and solid tumor cells.


Sujet(s)
Différenciation cellulaire , Plasticité cellulaire , Transition épithélio-mésenchymateuse , Techniques de knock-down de gènes , Cellules souches tumorales , Protéine tumorale p73 , Protéine p53 suppresseur de tumeur , Ombelliférones , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Humains , Ombelliférones/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéine tumorale p73/métabolisme , Protéine tumorale p73/génétique , Plasticité cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Phénotype , Transformation cellulaire néoplasique/génétique , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/génétique , Tumeurs du côlon/métabolisme
7.
Signal Transduct Target Ther ; 9(1): 249, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39300073

RÉSUMÉ

Lenvatinib is a targeted drug used for first-line treatment of hepatocellular carcinoma (HCC). A deeper insight into the resistance mechanism of HCC against lenvatinib is urgently needed. In this study, we aimed to dissect the underlying mechanism of lenvatinib resistance (LR) and provide effective treatment strategies. We established an HCC model of acquired LR. Cell counting, migration, self-renewal ability, chemoresistance and expression of stemness genes were used to detect the stemness of HCC cells. Molecular and biochemical strategies such as RNA-sequencing, immunoprecipitation, mass spectrometry and ubiquitination assays were used to explore the underlying mechanisms. Patient-derived HCC models and HCC samples from patients were used to demonstrate clinical significance. We identified that increased cancer stemness driven by the hypoxia-inducible factor-1α (HIF-1α) pathway activation is responsible for acquired LR in HCC. Phosphorylated non-muscle myosin heavy chain 9 (MYH9) at Ser1943, p-MYH9 (Ser1943), could recruit ubiquitin-specific protease 22 (USP22) to deubiquitinate and stabilize HIF-1α in lenvatinib-resistant HCC. Clinically, p-MYH9 (Ser1943) expression was upregulated in HCC samples, which predicted poor prognosis and LR. A casein kinase-2 (CK2) inhibitor and a USP22 inhibitor effectively reversed LR in vivo and in vitro. Therefore, the p-MYH9 (Ser1943)/USP22/HIF-1α axis is critical for LR and cancer stemness. For the diagnosis and treatment of LR in HCC, p-MYH9 (Ser1943), USP22, and HIF-1α might be valuable as novel biomarkers and targets.


Sujet(s)
Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Sous-unité alpha du facteur-1 induit par l'hypoxie , Tumeurs du foie , Cellules souches tumorales , Phénylurées , Quinoléines , Ubiquitin thiolesterase , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Humains , Quinoléines/pharmacologie , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Phénylurées/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Souris , Lignée cellulaire tumorale , Animaux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/génétique , Mâle
8.
Cells ; 13(17)2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39273033

RÉSUMÉ

Bladder cancer (BCa) is one of the most lethal genitourinary malignancies owing to its propensity for recurrence and poor survival. The biochemical pathway, signal transducer and activator of transcription 3 (STAT3), has gained significance as a molecular pathway that promotes proliferation, invasion, and chemoresistance. In this study, we explored the targeting of STAT3 with TTI-101 and SH5-07 in BCa and elucidated the mechanisms in three-dimensional (3D) spheroid and organoid models. We optimized the growth of spheroids from human, rat, and mouse BCa cell lines (J82, NBT-II, and MB49 respectively) and organoids from BBN (N-butyl-N-(4-hydroxybutyl)-nitrosamine)-induced rat bladder tumors. Cell viability was assessed using MTT and trypan blue assays. Intracellular ATP production, ROS production, and calcium AM (CA)/EtBr staining were used to measure the spheroid and organoid inhibition and mitochondrial function. Western blot analysis was performed to evaluate the pharmacodynamic markers involved in cell proliferation, apoptosis, cancer stem cells (CSCs), and STAT3 signaling in BCa. We found that targeting STAT3 (using TTI-101 and SH5-07) significantly reduced the proliferation of BCa spheroids and organoids, which was accompanied by decreased expression of pSTAT3, Cyclin D1, and PCNA. Our data also demonstrated that treatment with STAT3 inhibitors induced ROS production and cell death in BCa spheroids and organoids. STAT3 inhibition-induced cell death was associated with the activation of caspase 3/7 and PARP cleavage. Moreover, TTI-101 and SH5-07 target cancer stem cells by downregulating the expression of CD44 and CD133 in 3D models. This study provides the first evidence for the prevention of BCa with small-molecule inhibitors TTI-101 and SH5-07 via suppression of CSCs and STAT3 signaling.


Sujet(s)
Survie cellulaire , Facteur de transcription STAT-3 , Sphéroïdes de cellules , Tumeurs de la vessie urinaire , Facteur de transcription STAT-3/métabolisme , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/métabolisme , Humains , Animaux , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Rats , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Organoïdes/anatomopathologie
9.
Cell Commun Signal ; 22(1): 434, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251990

RÉSUMÉ

BACKGROUND: Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle. METHODS: We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo. RESULTS: We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav. CONCLUSIONS: This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide treatment.


Sujet(s)
Différenciation cellulaire , Glioblastome , Cellules souches tumorales , Canaux sodiques voltage-dépendants , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Canaux sodiques voltage-dépendants/métabolisme , Canaux sodiques voltage-dépendants/génétique , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Animaux , Témozolomide/pharmacologie , Lignée cellulaire tumorale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris
10.
AAPS PharmSciTech ; 25(7): 211, 2024 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-39242397

RÉSUMÉ

Pirarubicin attracted considerable attention in clinical studies because of its high therapeutic efficacy and reduced toxicity in comparison with other anthracyclines. Nevertheless, ~ 30% patients undergoing PIRA treatment still experience relapse and metastasis. Clinical advancements unveiled that cancer stem cells (CSCs) residing in the tumor constitutes a major factor for such limitations and subsequently are the reason for treatment failure. Consequently, eradicating CSCs alongside bulk tumor is a crucial undertaking to attain utmost therapeutic efficacy of the treatment. Nevertheless, majority of the CSCs inhibitors currently under examination lack specificity, show unsynchronized bioavailability with other primary treatments and exhibit notable toxicity in their therapeutic applications, which is primarily attributable to their inadequate tumor-targeting capabilities. Therefore, we have developed a biodegradable polylactic acid based blend block copolymeric NPs for concomitant delivery of CSCs inhibitor Salinomycin (SAL) & chemotherapeutic drug Pirarubicin (PIRA) with an aim to improve the efficacy of treatment and prevent cancer relapse. Prepared NPs showed < 100 nm size and excellent loading with sustained release for both the drugs. Also, PIRA:SAL co-loaded NPs exhibits synergistically enhanced cytotoxicity against cancer cell as well as CSCs. Most importantly, NPs mediated co-delivery of the drugs showed complete tumor eradication, without any reoccurrence throughout the surveillance period. Additionally, NPs treatment didn't show any histopathological alteration in vital organs confirming their non-toxic nature. Altogether, present study concludes that the developed PIRA:SAL NPs have excellent efficacy for tumor regression as well as prevention of cancer relapse, hence can be used as a potential combination therapy for cancer treatment.


Sujet(s)
Doxorubicine , Pyrannes , Pyrannes/administration et posologie , Pyrannes/pharmacologie , Doxorubicine/administration et posologie , Doxorubicine/analogues et dérivés , Doxorubicine/pharmacologie , Humains , Animaux , Lignée cellulaire tumorale , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Nanoparticules/composition chimique , Synergie des médicaments , Cellules souches tumorales/effets des médicaments et des substances chimiques , Souris , Polyesters/composition chimique , Systèmes de délivrance de médicaments/méthodes , Vecteurs de médicaments/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Récidive tumorale locale/traitement médicamenteux , Femelle , Libération de médicament , Polyether Polyketides
11.
Cells ; 13(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273014

RÉSUMÉ

Overcoming temozolomide (TMZ)-resistance is a major challenge in glioblastoma therapy. Therefore, identifying the key molecular player in chemo-resistance becomes urgent. We previously reported the downregulation of PDCD10 in primary glioblastoma patients and its tumor suppressor-like function in glioblastoma cells. Here, we demonstrate that the loss of PDCD10 causes a significant TMZ-resistance during treatment and promotes a rapid regrowth of tumor cells after treatment. PDCD10 knockdown upregulated MGMT, a key enzyme mediating chemo-resistance in glioblastoma, accompanied by increased expression of DNA mismatch repair genes, and enabled tumor cells to evade TMZ-induced cell-cycle arrest. These findings were confirmed in independent models of PDCD10 overexpressing cells. Furthermore, PDCD10 downregulation led to the dedifferentiation of glioblastoma cells, as evidenced by increased clonogenic growth, the upregulation of glioblastoma stem cell (GSC) markers, and enhanced neurosphere formation capacity. GSCs derived from PDCD10 knockdown cells displayed stronger TMZ-resistance and regrowth potency, compared to their parental counterparts, indicating that PDCD10-induced stemness may independently contribute to tumor malignancy. These data provide evidence for a dual role of PDCD10 in tumor suppression by controlling both chemo-resistance and dedifferentiation, and highlight PDCD10 as a potential prognostic marker and target for combination therapy with TMZ in glioblastoma.


Sujet(s)
Protéines régulatrices de l'apoptose , Résistance aux médicaments antinéoplasiques , Glioblastome , Témozolomide , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux , Témozolomide/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , DNA modification methylases/métabolisme , DNA modification methylases/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Enzymes de réparation de l'ADN/métabolisme , Enzymes de réparation de l'ADN/génétique
12.
Oncol Rep ; 52(5)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39301645

RÉSUMÉ

Phenformin, a biguanide compound, has attracted increased attention due to its prominent antitumor activity. As a multi­target agent, the antitumor effects of phenformin involve a wide range of factors, including inhibition of mitochondrial complex I, activation of AMP­activated protein kinase, impact on the tumor microenvironment, suppression of cancer stem cells and others. In addition, phenformin has been shown to markedly augment the effectiveness of various clinical treatment methods, including radiotherapy, chemotherapy, targeted therapy and immunotherapy. It is noteworthy that breakthrough progress has been made in the treatment of cancer with phenformin with application in clinical trials for the treatment of melanoma. Phenformin not only reduces the lesion area of patients, but also enhances the efficacy of dalafinib/trimetinib. In the present review, the novel breakthroughs in the antitumor effects and mechanisms of phenformin were discussed. In addition, the current review focuses on the clinical development value of phenformin, striving to provide new insights into the future research direction of phenformin in the field of tumor treatment.


Sujet(s)
Tumeurs , Phenformine , Microenvironnement tumoral , Humains , Phenformine/pharmacologie , Phenformine/usage thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Animaux , Mélanome/traitement médicamenteux
13.
Sci Rep ; 14(1): 22487, 2024 09 28.
Article de Anglais | MEDLINE | ID: mdl-39341857

RÉSUMÉ

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.


Sujet(s)
Diterpènes , Composés époxy , Cellules souches tumorales , Phénanthrènes , Tumeurs du sein triple-négatives , Microenvironnement tumoral , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Humains , Animaux , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/immunologie , Femelle , Souris , Lignée cellulaire tumorale , Composés époxy/pharmacologie , Composés époxy/usage thérapeutique , Phénanthrènes/pharmacologie , Phénanthrènes/usage thérapeutique , Diterpènes/pharmacologie , Diterpènes/usage thérapeutique , Cyclophosphamide/pharmacologie , Cyclophosphamide/usage thérapeutique , Souris de lignée BALB C , Organophosphates
14.
Sci Rep ; 14(1): 20421, 2024 09 03.
Article de Anglais | MEDLINE | ID: mdl-39227705

RÉSUMÉ

Glioblastoma (GBM) is the most common malignant primary brain cancer that, despite recent advances in the understanding of its pathogenesis, remains incurable. GBM contains a subpopulation of cells with stem cell-like properties called cancer stem cells (CSCs). Several studies have demonstrated that CSCs are resistant to conventional chemotherapy and radiation thus representing important targets for novel anti-cancer therapies. Proton sensing receptors expressed by CSCs could represent important factors involved in the adaptation of tumours to the extracellular environment. Accordingly, the expression of acid-sensing ion channels (ASICs), proton-gated sodium channels mainly expressed in the neurons of peripheral (PNS) and central nervous system (CNS), has been demonstrated in several tumours and linked to an increase in cell migration and proliferation. In this paper we report that the ASIC3 isoform, usually absent in the CNS and present in the PNS, is enriched in human GBM CSCs while poorly expressed in the healthy human brain. We propose here a novel therapeutic strategy based on the pharmacological activation of ASIC3, which induces a significant GBM CSCs damage while being non-toxic for neurons. This approach might offer a promising and appealing new translational pathway for the treatment of glioblastoma.


Sujet(s)
Canaux ioniques sensibles à l'acidité , Tumeurs du cerveau , Prolifération cellulaire , Glioblastome , Cellules souches tumorales , Humains , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Canaux ioniques sensibles à l'acidité/métabolisme , Canaux ioniques sensibles à l'acidité/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Mouvement cellulaire/effets des médicaments et des substances chimiques
15.
Cancer Res Commun ; 4(10): 2638-2652, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39287565

RÉSUMÉ

High-grade serous ovarian cancer remains a poorly understood disease with a high mortality rate. Although most patients respond to cytotoxic therapies, a majority will experience recurrence. This may be due to a minority of drug-resistant cancer stem-like cells (CSC) that survive chemotherapy and are capable of repopulating heterogeneous tumors. It remains unclear how CSCs are supported in the tumor microenvironment (TME) particularly during chemotherapy exposure. Tumor-associated macrophages (TAM) make up half of the immune population of the ovarian TME and are known to support CSCs and contribute to cancer progression. TAMs are plastic cells that alter their phenotype in response to environmental stimuli and thus may influence CSC maintenance during chemotherapy. Given the plasticity of TAMs, we studied the effects of carboplatin on macrophage phenotypes using both THP1- and peripheral blood mononuclear cell (PBMC)-derived macrophages and whether this supports CSCs and ovarian cancer progression following treatment. We found that carboplatin exposure induces an M1-like proinflammatory phenotype that promotes SOX2 expression, spheroid formation, and CD117+ ovarian CSCs, and that macrophage-secreted CCL2/MCP-1 is at least partially responsible for this effect. Depletion of TAMs during carboplatin exposure results in fewer CSCs and prolonged survival in a xenograft model of ovarian cancer. This study supports a role for platinum-based chemotherapies in promoting a transient proinflammatory M1-like TAM that enriches for CSCs during treatment. Improving our understanding of TME responses to cytotoxic drugs and identifying novel mechanisms of CSC maintenance will enable the development of better therapeutic strategies for high-grade serous ovarian cancer. Significance: We show that chemotherapy enhances proinflammatory macrophage phenotypes that correlate with ovarian cancer progression. Given that macrophages are the most prominent immune cell within these tumors, this work provides the foundation for future translational studies targeting specific macrophage populations during chemotherapy, a promising approach to prevent relapse in ovarian cancer.


Sujet(s)
Carboplatine , Évolution de la maladie , Cellules souches tumorales , Tumeurs de l'ovaire , Phénotype , Microenvironnement tumoral , Femelle , Humains , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/immunologie , Animaux , Souris , Carboplatine/pharmacologie , Carboplatine/usage thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
16.
Sci Rep ; 14(1): 22619, 2024 09 30.
Article de Anglais | MEDLINE | ID: mdl-39349745

RÉSUMÉ

Cancer stem cells (CSC) play an important role in carcinogenesis and are acknowledged to be responsible for chemoresistance in cholangiocarcinoma (CCA). Studying CCA CSC has been challenging, due to lack of consensus CSC markers, and to their plastic nature. Since dual expression of the core pluripotent factors SOX2/OCT4 has been shown to correlate with poor outcome in CCA patients, we selected the SOX2/OCT4 activating short half-life GFP-based live reporter (SORE6-dsCopGFP) to study CSC dynamics at the single-cell level. Transduction of five human CCA cell lines resulted in the expression of 1.8-13.1% GFP-positive (SORE6POS) cells. By live imaging, we found that SORE6POS CCA cells possess self-renewal capacity and that they can be induced to differentiate. Significantly, the SORE6POS cells were highly tumorigenic, both in vitro and in vivo, thus implicating the characteristics of primary CSCs. When we then analyzed for selected CSC-related markers, we found that the majority of both CD133+/CD44+, and CD133+/LGR5+ CCA cells were SORE6POS cells. Exposing transduced cells to standard CCA chemotherapy revealed higher growth rate inhibition at 50% (GR50s) for SORE6POS cells compared to GFP-negative (SORE6NEG) ones indicating that these CSC-like cells were more resistant to the treatment. Moreover, the chemotherapy induced SORE6POS from SORE6NEG cells, while retaining the existing SORE6POS population. Finally, treatment of transduced cells with CDK4/6 inhibitors in vitro for 3 days resulted in a lowered CSC number in the culture. Thus, applying a live reporter system allowed us to elucidate the stem cell diversity and drug-induced plasticity of CCA CSCs. These findings have clear implications for future management of such patients.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Cellules souches tumorales , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Humains , Cholangiocarcinome/anatomopathologie , Cholangiocarcinome/métabolisme , Cholangiocarcinome/traitement médicamenteux , Lignée cellulaire tumorale , Tumeurs des canaux biliaires/anatomopathologie , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/métabolisme , Gènes rapporteurs , Facteurs de transcription SOX-B1/métabolisme , Facteurs de transcription SOX-B1/génétique , Analyse sur cellule unique/méthodes , Animaux , Souris , Facteur de transcription Oct-3/métabolisme , Facteur de transcription Oct-3/génétique , Antinéoplasiques/pharmacologie , Plasticité cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques
17.
Molecules ; 29(18)2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39339380

RÉSUMÉ

Cancer stem cells (CSCs) are most likely the main cause of lung cancer formation, metastasis, drug resistance, and genetic heterogeneity. Three-dimensional (3D) ex vivo cell culture models can facilitate stemness improvement and CSC enrichment. Considering the critical role of extracellular matrix (ECM) on CSC properties, the present study developed a thermo-responsive hydrogel using the porcine decellularized lung for 3D cell culture, and the cell-laden hydrogel culturing model was used to explore the CSC characteristics and potential utilization in CSC-specific drug evaluation. Results showed that the lung dECM hydrogel (LEH) was composed of the main ECM components and displayed excellent cellular compatibility. In addition, lung cancer cells 3D cultured in LEH displayed the overexpression of metastasis-related genes and enhanced migration properties, as compared with those in two-dimensional (2D) conditions. Notably, the CSC features, including the expression level of stemness-associated genes, colony formation capability, drug resistance, and the proportion of cancer stem-like cells (CD133+), were also enhanced in 3D cells. Furthermore, the attenuation effect of epigallocatechin gallate (EGCG) on CSC properties in the 3D model was observed, confirming the potential practicability of the 3D culture on CSC-targeted drug screening. Overall, our results suggest that the fabricated LEH is an effective and facile platform for 3D cell culture and CSC-specific drug evaluation.


Sujet(s)
Techniques de cultures cellulaires tridimensionnelles , Hydrogels , Tumeurs du poumon , Cellules souches tumorales , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Humains , Hydrogels/composition chimique , Hydrogels/pharmacologie , Techniques de cultures cellulaires tridimensionnelles/méthodes , Animaux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Matrice extracellulaire décellularisée/pharmacologie , Matrice extracellulaire décellularisée/composition chimique , Poumon/métabolisme , Poumon/anatomopathologie , Poumon/cytologie , Suidae , Catéchine/analogues et dérivés , Catéchine/pharmacologie , Catéchine/composition chimique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Techniques de culture cellulaire/méthodes
18.
Cell Commun Signal ; 22(1): 425, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39223652

RÉSUMÉ

BACKGROUND: Increased cancer stem cell (CSC) content and SOX2 overexpression are common features in the development of resistance to therapy in hormone-dependent breast cancer, which remains an important clinical challenge. SOX2 has potential as biomarker of resistance to treatment and as therapeutic target, but targeting transcription factors is also challenging. Here, we examine the potential inhibitory effect of different polyoxometalate (POM) derivatives on SOX2 transcription factor in tamoxifen-resistant breast cancer cells. METHODS: Various POM derivatives were synthesised and characterised by infrared spectra, powder X-ray diffraction pattern and nuclear magnetic resonance spectroscopy. Estrogen receptor (ER) positive breast cancer cells, and their counterparts, which have developed resistance to the hormone therapy tamoxifen, were treated with POMs and their consequences assessed by gel retardation and chromatin immunoprecipitation to determine SOX2 binding to DNA. Effects on proliferation, migration, invasion and tumorigenicity were monitored and quantified using microscopy, clone formation, transwell, wound healing assays, flow cytometry and in vivo chick chorioallantoic membrane (CAM) models. Generation of lentiviral stable gene silencing and gene knock-out using CRISPR-Cas9 genome editing were applied to validate the inhibitory effects of the selected POM. Cancer stem cell subpopulations were quantified by mammosphere formation assays, ALDEFLUOR activity and CD44/CD24 stainings. Flow cytometry and western blotting were used to measure reactive oxygen species (ROS) and apoptosis. RESULTS: POMs blocked in vitro binding activity of endogenous SOX2. [P2W18O62]6- (PW) Wells-Dawson-type anion was the most effective at inhibiting proliferation in various cell line models of tamoxifen resistance. 10 µM PW also reduced cancer cell migration and invasion, as well as SNAI2 expression levels. Treatment of tamoxifen-resistant cells with PW impaired tumour formation by reducing CSC content, in a SOX2-dependent manner, which led to stem cell depletion in vivo. Mechanistically, PW induced formation of reactive oxygen species (ROS) and inhibited Bcl-2, leading to the death of tamoxifen-resistant cells. PW-treated tamoxifen-resistant cells showed restored sensitivity to tamoxifen. CONCLUSIONS: Together, these observations highlight the potential use of PW as a SOX2 inhibitor and the therapeutic relevance of targeting SOX2 to treat tamoxifen-resistant breast cancer.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Facteurs de transcription SOX-B1 , Tamoxifène , Composés du tungstène , Facteurs de transcription SOX-B1/métabolisme , Facteurs de transcription SOX-B1/génétique , Tamoxifène/pharmacologie , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Composés du tungstène/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Lignée cellulaire tumorale , Animaux
19.
Cell Death Dis ; 15(9): 696, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39349424

RÉSUMÉ

Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3­kinase/protein kinase B (PI3K/AKT) pathway and mitogen­activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.


Sujet(s)
Cellules souches tumorales , Sérine-thréonine kinases TOR , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Humains , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Inhibiteurs de mTOR/pharmacologie , Inhibiteurs de mTOR/usage thérapeutique , Thérapie moléculaire ciblée
20.
Int J Mol Sci ; 25(18)2024 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-39337661

RÉSUMÉ

One of the main causes of poor prognoses in patient with glioblastoma (GBM) is drug resistance to current standard treatment, which includes chemoradiation and adjuvant temozolomide (TMZ). In addition, the concept of cancer stem cells provides new insights into therapy resistance and management also in GBM and glioblastoma stem cell-like cells (GSCs), which might contribute to therapy resistance. Bone morphogenetic protein-4 (BMP4) stimulates astroglial differentiation of GSCs and thereby reduces their self-renewal capacity. Exposure of GSCs to BMP4 may also sensitize these cells to TMZ. A recent phase I trial has shown that local delivery of BMP4 is safe, but a large variation in survival is seen in these treated patients and in features of their cultured tumors. We wanted to combine TMZ and BMP4 (TMZ + BMP4) therapy and assess the inter-tumoral variability in response to TMZ + BMP4 in patient-derived GBM cultures. A phase II trial could then benefit a larger group of patients than those treated with BMP4 only. We first show that simultaneous treatment with TMZ + BMP4 is more effective than sequential treatment. Second, when applying our optimized treatment protocol, 70% of a total of 20 GBM cultures displayed TMZ + BMP4 synergy. This combination induces cellular apoptosis and does not inhibit cell proliferation. Comparative bulk RNA-sequencing indicates that treatment with TMZ + BMP4 eventually results in decreased MAPK signaling, in line with previous evidence that increased MAPK signaling is associated with resistance to TMZ. Based on these results, we advocate further clinical trial research to test patient benefit and validate pathophysiological hypothesis.


Sujet(s)
Protéine morphogénétique osseuse de type 4 , Tumeurs du cerveau , Glioblastome , Cellules souches tumorales , Témozolomide , Humains , Protéine morphogénétique osseuse de type 4/métabolisme , Témozolomide/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Synergie des médicaments , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Apoptose/effets des médicaments et des substances chimiques , Femelle , Mâle , Adulte d'âge moyen , Antinéoplasiques alcoylants/pharmacologie , Sujet âgé , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE