Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.807
Filtrer
1.
N Engl J Med ; 391(9): 789-799, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39231341

RÉSUMÉ

BACKGROUND: The addition of trabectedin to doxorubicin, followed by trabectedin maintenance, may have superior efficacy to doxorubicin alone as first-line treatment in patients with advanced leiomyosarcoma. METHODS: We conducted a phase 3 trial involving patients with metastatic or unresectable leiomyosarcoma who had not received chemotherapy previously. Patients were randomly assigned to receive either single-agent doxorubicin (six cycles) or doxorubicin plus trabectedin (six cycles), with continued trabectedin as maintenance therapy in patients in the doxorubicin-trabectedin group who did not have disease progression. Surgery to resect residual disease was allowed in each group after six cycles of therapy. Analyses of progression-free survival (primary end point) and overall survival (secondary end point) were adjusted for two stratification factors: tumor origin site (uterine vs. soft tissue) and disease stage (locally advanced vs. metastatic). The primary end-point results were reported previously. RESULTS: A total of 150 patients underwent randomization. At a median follow-up of 55 months (interquartile range, 49 to 63), a total of 107 patients had died (47 in the doxorubicin-trabectedin group and 60 in the doxorubicin group). The median overall survival was longer in the doxorubicin-trabectedin group (33 months; 95% confidence interval [CI], 26 to 48) than in the doxorubicin group (24 months; 95% CI, 19 to 31); the adjusted hazard ratio for death was 0.65 (95% CI, 0.44 to 0.95). In a finding consistent with earlier reports, progression-free survival was longer in the doxorubicin-trabectedin group (12 months; 95% CI, 10 to 16) than in the doxorubicin group (6 months; 95% CI, 4 to 7); the adjusted hazard ratio for progression or death was 0.37 (95% CI, 0.26 to 0.53). The incidence of adverse events and the percentage of patients with dose reductions were higher with doxorubicin plus trabectedin than with doxorubicin alone. CONCLUSIONS: Combination therapy with doxorubicin and trabectedin induction, followed by trabectedin maintenance, was associated with improved overall survival and progression-free survival, as compared with doxorubicin alone, among patients with metastatic or surgically unresectable uterine or soft-tissue leiomyosarcoma. (Funded by PharmaMar and others; LMS04 ClinicalTrials.gov number, NCT02997358.).


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Doxorubicine , Léiomyosarcome , Tumeurs des tissus mous , Trabectédine , Tumeurs de l'utérus , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Doxorubicine/administration et posologie , Doxorubicine/effets indésirables , Estimation de Kaplan-Meier , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/mortalité , Léiomyosarcome/anatomopathologie , Chimiothérapie de maintenance , Survie sans progression , Tumeurs des tissus mous/traitement médicamenteux , Tumeurs des tissus mous/mortalité , Tumeurs des tissus mous/anatomopathologie , Analyse de survie , Trabectédine/administration et posologie , Trabectédine/effets indésirables , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/mortalité , Tumeurs de l'utérus/anatomopathologie , Stadification tumorale
3.
J Biochem Mol Toxicol ; 38(10): e23855, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39328005

RÉSUMÉ

Adriamycin is an anticancer anthracycline drug that inhibits the progression of topoisomerase II activity and causes apoptosis. The effective clinical application of the drug is very much limited by its adverse drug reactions on various tissues. Most importantly, Adriamycin causes cardiomyopathy, one of the life-threatening complications of the drug. Altered expression of PPARγ in adipocytes inhibited the glucose and fatty acids uptake by down regulating GLUT4 and CD36 expression and causes cardiotoxicity. Therefore, the influence of Adriamycin in cardiac ailments was investigated in vivo and in vitro. Adriamycin treated rats showed altered ECG profile, arrhythmic heartbeat with the elevated levels of CRP and LDH. Dysregulated lipid profiles with elevated levels of cholesterol and triglycerides were also observed. Possibilities of cardiac problems due to cardiomyopathy were analyzed through histopathology. Adriamycin treated rats showed no signs for atheromatous plaque formation in aorta but disorganized cardiomyocytes with myofibrillar loss and inflammation in heart tissue, indicative of cardiomyopathy. Reduced levels of antioxidant enzymes confirmed the incidence of oxidative stress. Adriamycin treatment significantly reduced glucose and insulin levels, creating energy demand due to decreased glucose and insulin levels with increased fatty acid accumulation, ultimately resulting in oxidative stress mediated cardiomyopathy. Since PPARs play a vital role in regulating oxidative stress, the effect of Adriamycin on PPARγ was analyzed by western blot. Adriamycin downregulated PPARγ in a dose-dependent manner in H9C2 cells in vitro. Overall, our study suggests that Adriamycin alters glucose and lipid metabolism via PPARγ inhibition that leads to oxidative stress and cardiomyopathy that necessitates a different therapeutic approach.


Sujet(s)
Cardiomyopathies , Doxorubicine , Récepteur PPAR gamma , Animaux , Doxorubicine/effets indésirables , Cardiomyopathies/induit chimiquement , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Rats , Récepteur PPAR gamma/métabolisme , Mâle , Lignée cellulaire , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Wistar , Métabolisme énergétique/effets des médicaments et des substances chimiques , Antibiotiques antinéoplasiques/effets indésirables , Antibiotiques antinéoplasiques/toxicité
4.
J Zhejiang Univ Sci B ; 25(9): 756-772, 2024 Sep 12.
Article de Anglais, Chinois | MEDLINE | ID: mdl-39308066

RÉSUMÉ

Doxorubicin (DOX) is a commonly administered chemotherapy drug for treating hematological malignancies and solid tumors; however, its clinical application is limited by significant cardiotoxicity. Cynaroside (Cyn) is a flavonoid glycoside distributed in honeysuckle, with confirmed potential biological functions in regulating inflammation, pyroptosis, and oxidative stress. Herein, the effects of Cyn were evaluated in a DOX-induced cardiotoxicity (DIC) mouse model, which was established by intraperitoneal injections of DOX (5 mg/kg) once a week for three weeks. The mice in the treatment group received dexrazoxane, MCC950, and Cyn every two days. Blood biochemistry, histopathology, immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and western blotting were conducted to investigate the cardioprotective effects and potential mechanisms of Cyn treatment. The results demonstrated the significant benefits of Cyn treatment in mitigating DIC; it could effectively alleviate oxidative stress to a certain extent, maintain the equilibrium of cell apoptosis, and enhance the cardiac function of mice. These effects were realized via regulating the transcription levels of pyroptosis-related genes, such as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), caspase-1, and gasdermin D (GSDMD). Mechanistically, for DOX-induced myocardial injury, Cyn could significantly modulate the expression of pivotal genes, including adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), sirtuin 3 (SIRT3), and nuclear factor erythroid 2-related factor 2 (Nrf2). We attribute it to the mediation of AMPK/SIRT3/Nrf2 pathway, which plays a central role in preventing DOX-induced cardiomyocyte injury. In conclusion, the present study confirms the therapeutic potential of Cyn in DIC by regulating the AMPK/SIRT3/Nrf2 pathway.


Sujet(s)
AMP-Activated Protein Kinases , Cardiotoxicité , Doxorubicine , Myocytes cardiaques , Facteur-2 apparenté à NF-E2 , Pyroptose , Transduction du signal , Sirtuine-3 , Animaux , Doxorubicine/effets indésirables , Pyroptose/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Souris , Facteur-2 apparenté à NF-E2/métabolisme , AMP-Activated Protein Kinases/métabolisme , Sirtuine-3/métabolisme , Cardiotoxicité/prévention et contrôle , Cardiotoxicité/traitement médicamenteux , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Souris de lignée C57BL
5.
J Biochem Mol Toxicol ; 38(10): e23861, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39305037

RÉSUMÉ

Doxorubicin (DOX) is an anthracycline antibiotic widely employed to treat carcinoma. Nevertheless, severe cardiotoxic side effects restrict its clinical use. Esculetin, a natural flavonoid, is found abundantly in plants. This study evaluated the protective effects of esculetin against DOX-induced hepatotoxicity in rat livers. Forty-eight rats were randomly divided into six groups with eight rats in each group: control (I), DOX (II), esculetin (III, 50 mg/kg), esculetin (IV, 100 mg/kg), DOX+esculetin 50 (V, DOX+esculetin 50 mg/kg), and DOX+esculetin 100 (VI, DOX+esculetin 100 mg/kg). The administration of esculetin effectively mitigated alterations in the measured biochemical parameters induced by DOX. Gene expression analyses demonstrated that esculetin treatment significantly reduced the DOX-induced expression of Foxo1, Hspa1a, Hsp4a, Hsp5a, Casp3, and Casp9 while increasing the DOX-induced expression of Foxo3. These findings suggest that esculetin, with its antioxidant and anti-inflammatory effects, might be a therapeutic option for protecting against DOX-induced hepatotoxicity.


Sujet(s)
Lésions hépatiques dues aux substances , Doxorubicine , Ombelliférones , Animaux , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Ombelliférones/pharmacologie , Rats , Mâle , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/prévention et contrôle , Lésions hépatiques dues aux substances/traitement médicamenteux , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Protéines du choc thermique/métabolisme , Protéines du choc thermique/génétique , Facteurs de transcription Forkhead/métabolisme , Caspases/métabolisme , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables , Transduction du signal/effets des médicaments et des substances chimiques
6.
Front Immunol ; 15: 1430648, 2024.
Article de Anglais | MEDLINE | ID: mdl-39229263

RÉSUMÉ

Background: Angioimmunoblastic T-cell lymphoma (AITL) is known for its unfavorable survival prognosis. Chidamide has shown efficacy in relapsed/refractory AITL, but its efficacy in newly diagnosed AITL is uncertain. Objective: This retrospective research aimed to evaluate the effectiveness and safety of chidamide when used with doxorubicin, cyclophosphamide, prednisone, and vincristine (CHOP) in comparison to CHOP by itself for individuals newly diagnosed with AITL, and to examine the impact of transplantation. Method: This was an analysis that compared outcomes among patients who received chidamide + CHOP on a clinical trial vs. historical controls who received CHOP alone, enrolling a total of sixty-six treatment-naive AITL patients between April 2014 and November 2022. Among them, thirty-three received chidamide in addition to CHOP (chidamide group), while thirty-three received CHOP alone (control group). The clinical characteristics were balanced between the two groups. All patients were scheduled to undergo up to six courses of treatment before transplantation. Results: The chidamide group had a significantly longer median overall survival (OS) compared to the control group, with a median OS that was not reached, as opposed to 20 months in the control group (p = 0.002). In the control group, the median progression-free survival (PFS) was 11 months, while in the chidamide group, it was 22 months (p = 0.080). In the high-risk group (IPI ≥ 3), the chidamide group demonstrated notably superior complete response (CR) and overall response rate (ORR) compared to the control cohort (p = 0.002, p = 0.034). The PFS and OS in the chidamide group were not reached, and there were significant differences compared to the control group (p = 0.007, p = 0.003). The median OS of the transplanted group was longer than the non-transplanted group (p = 0.004). On multivariate analysis, chidamide group reduced the hazards of death in the total cohort. Conclusion: As the study was non-random and retrospective, Chidamide combined with chemotherapy should be tested in randomized trials given its potential to improve prognosis in treatment-naive AITL patients. Furthermore, autologous hematopoietic stem cell transplantation (auto-HSCT) has demonstrated enhanced overall survival in individuals with AITL. Clinical trial registration: https://clinicaltrials.gov/, NCT03268889.


Sujet(s)
Aminopyridines , Protocoles de polychimiothérapie antinéoplasique , Benzamides , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Aminopyridines/usage thérapeutique , Aminopyridines/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzamides/usage thérapeutique , Benzamides/administration et posologie , Benzamides/effets indésirables , Cyclophosphamide/usage thérapeutique , Cyclophosphamide/administration et posologie , Doxorubicine/usage thérapeutique , Doxorubicine/administration et posologie , Doxorubicine/effets indésirables , Lymphome T/mortalité , Lymphome T/thérapie , Lymphome T/traitement médicamenteux , Lymphome T/diagnostic , Prednisone/usage thérapeutique , Prednisone/administration et posologie , Études rétrospectives , Résultat thérapeutique , Vincristine/usage thérapeutique , Vincristine/administration et posologie
7.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39273564

RÉSUMÉ

Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction.


Sujet(s)
Facteur de croissance du tissu conjonctif , Doxorubicine , Fibrose , Souris knockout , Stress oxydatif , Animaux , Doxorubicine/effets indésirables , Facteur de croissance du tissu conjonctif/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Délétion de gène , Mâle , Myocarde/métabolisme , Myocarde/anatomopathologie , Souris de lignée C57BL , Cardiotoxicité/génétique , Cardiotoxicité/métabolisme
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1441-1449, 2024 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-39276039

RÉSUMÉ

OBJECTIVE: To investigate the effect of Kaixinsan (KXS, a traditional Chinese medicine formula) for alleviating adriamycin-induced depression-like behaviors in mice bearing breast cancer xenografts and explore the pharmacological mechanism. METHODS: Forty female BALB/c mice were randomized equally into control group, model group, and low- and high-dose KXS treatment groups, and in the latter 3 groups, mouse models bearing orthotopic breast cancer 4T1 cell xenografts were established and treated with adriamycin along with saline or KXS via gavage. Depression-like behaviors of the mice were assessed using open field test and elevated plus-maze test, and the changes in serum levels of depression-related factors were examined. RNA-seq analysis and transmission electron microscopy were used and ferroptosis-related factors were detected to explore the mechanisms of adriamycin-induced depression and the therapeutic mechanism of KXS. The results were verified in SH-SY5Y cells using ferroptosis inhibitor Fer-1 as the positive control. RESULTS: KXS significantly alleviated depression-like behaviors and depression-related serological changes induced by adriamycin in the mouse models. RNA-seq results suggested that KXS alleviated chemotherapy-induced depression by regulating oxidative stress, lipid metabolism and iron ion binding in the prefrontal cortex. Pathological analysis and detection of ferroptosis-related factors showed that KXS significantly reduced ferroptosis in the prefrontal cortex of adriamycin-treated mice. In SH-SY5Y cells, both KXS-medicated serum and the ferroptosis inhibitor were capable of attenuating adriamycin-induced cell ferroptosis. CONCLUSION: KXS alleviates adriamycininduced depression-like behaviors in mice by reducing ferroptosis in the prefrontal cortex of breast cancer-bearing mice.


Sujet(s)
Dépression , Doxorubicine , Ferroptose , Souris de lignée BALB C , Cortex préfrontal , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Souris , Dépression/traitement médicamenteux , Dépression/induit chimiquement , Doxorubicine/effets indésirables , Femelle , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Lignée cellulaire tumorale , Comportement animal/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1571-1581, 2024 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-39276053

RÉSUMÉ

OBJECTIVE: To explore the mechanism underlying the protective effect of Lonicerae japonicae flos (LJF) extract against doxorubicin (DOX) -induced liver injury in mice. METHODS: Network pharmacology methods were used to obtain the intersection genes between LJF targets and disease targets, based on which the protein-protein interaction (PPI) network was constructed using STRING database for screening the core targets using Cytoscape software. DAVID database was used for bioinformatics analysis, and the core components and core targets were verified using molecular docking study. In a mouse model of DOX-induced liver injury, the effect of LJF extract on liver pathologies, serum levels of ALT and AST, and hepatic expressions of HYP, ROS, TNF-α, IL-6, COL-Ⅳ and P53 proteins were evaluated using HE and Masson staining, ELISA, and Western blotting. RESULTS: We identified 12 core targets from 43 intersection genes involving cancer pathway, IL-17 signaling pathway, and TNF signaling pathways. Molecular docking study suggested that 10 core components of LJF could bind to different core targets. The mice with DOX-induced liver injury showed elevated serum AST and ALT levels with obvious liver injury and fibrosis, increased ROS content, and enhanced expressions of TNF-α, IL-6, HYP, COL-Ⅳ and P53 proteins in the liver tissue. All these changes in the mouse models were significantly alleviated by treatment with LJF extract, suggesting obviously lowered levels of oxidative stress, inflammation and fibrosis in the liver tissues. CONCLUSION: LJF extract is capable of alleviating DOX-induced liver injury in mice by downregulating Trp53, TNF and IL-6 to reduce liver oxidative stress, inflammation and fibrosis.


Sujet(s)
Lésions hépatiques dues aux substances , Doxorubicine , Interleukine-6 , Lonicera , Simulation de docking moléculaire , Facteur de nécrose tumorale alpha , Animaux , Doxorubicine/effets indésirables , Souris , Lonicera/composition chimique , Lésions hépatiques dues aux substances/prévention et contrôle , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/traitement médicamenteux , Interleukine-6/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Extraits de plantes/pharmacologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme , Agents protecteurs/pharmacologie , Cartes d'interactions protéiques , Transduction du signal/effets des médicaments et des substances chimiques , Pharmacologie des réseaux
10.
J Biochem Mol Toxicol ; 38(10): e23851, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39267350

RÉSUMÉ

Doxorubicin (Dox) is frequently employed as a chemotherapy agent for breast cancer. As the chemotherapy moves forward, breast cancer cells tend to develop resistance to Dox, besides that, Dox are also easy to cause cardiotoxicity related to cumulative dose. Therefore, how to potentiate the chemosensitivity of breast cancer cells to Dox while attenuating its cardiotoxicity has become a research hotspot. Tanshinone IIA (Tan IIA) is known for its anticancer activity as well as for its cardioprotective effects. In view of the aforementioned facts, we assessed whether Tan IIA possesses synergism and attenuation effect on Dox for breast cancer chemotherapy. Our studies in vitro indicated that, Tan IIA could potentiate the effect of Dox on breast cancer cells proliferation inhibition and apoptosis promotion by inhibiting ERK1/2 pathway, but interestingly, Tan IIA attenuated the cytotoxicity of Dox to myocardial cells by activating ERK1/2 pathway. Additionally, our studies in vivo also suggested that Tan IIA potentiated the chemotherapeutic effect of Dox against breast cancer while attenuating Dox-induced myocardial injury. Given that Tan IIA had a synergism and attenuation effect on Dox, we believed that Tan IIA can be used as an ideal drug in combination with Dox for breast cancer therapy.


Sujet(s)
Abiétanes , Tumeurs du sein , Cardiotoxicité , Doxorubicine , Système de signalisation des MAP kinases , Doxorubicine/effets indésirables , Doxorubicine/pharmacologie , Abiétanes/pharmacologie , Femelle , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Animaux , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/prévention et contrôle , Cardiotoxicité/étiologie , Synergie des médicaments , Cellules MCF-7 , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE