Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.775
Filtrer
1.
Cell Death Dis ; 15(8): 558, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090086

RÉSUMÉ

Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. PCa that relapses after hormonal therapies, referred to as castration resistant PCa (CRPC), often presents with metastases (mCRPC) that are the major cause of mortality. The few available therapies for mCRPC patients include taxanes docetaxel (DTX) and cabazitaxel (CBZ). However, development of resistance limits their clinical use. Mechanistically, resistance arises through upregulation of multidrug resistance (MDR) proteins such as MDR1/ABCB1, making ABCB1 an attractive therapeutic target. Yet, ABCB1 inhibitors failed to be clinically useful due to low specificity and toxicity issues. To study taxanes resistance, we produced CBZ resistant C4-2B cells (RC4-2B) and documented resistance to both CBZ and DTX in cell culture and in 3D prostaspheres settings. RNAseq identified increased expression of ABCB1 in RC4-2B, that was confirmed by immunoblotting and immunofluorescent analysis. ABCB1-specific inhibitor elacridar reversed CBZ and DTX resistance in RC4-2B cells, confirming ABCB1-mediated resistance mechanism. In a cell-based screen using a curated library of cytotoxic drugs, we found that DNA damaging compounds Camptothecin (CPT) and Cytarabine (Ara-C) overcame resistance as seen by similar cytotoxicity in parental C4-2B and resistant RC4-2B. Further, these compounds were cytotoxic to multiple PC cells resistant to taxanes with high ABCB1 expression and, therefore, can be used to conquer the acquired resistance to taxanes in PCa. Finally, inhibition of cyclin-dependent kinases 4/6 (CDK4/6) with small molecule inhibitors (CDK4/6i) potentiated cytotoxic effect of CPT or Ara-C in both parental and resistant cells. Overall, our findings indicate that DNA damaging agents CPT and Ara-C alone or in combination with CDK4/6i can be suggested as a new treatment regimen in CRPC patients, including those that are resistant to taxanes.


Sujet(s)
Sous-famille B de transporteurs à cassette liant l'ATP , Docetaxel , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Tumeurs prostatiques résistantes à la castration , Taxoïdes , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Lignée cellulaire tumorale , Docetaxel/pharmacologie , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Taxoïdes/pharmacologie , Taxoïdes/usage thérapeutique , Antinéoplasiques/pharmacologie
3.
Front Public Health ; 12: 1425734, 2024.
Article de Anglais | MEDLINE | ID: mdl-39091529

RÉSUMÉ

Background: Tislelizumab is the first PD-1 inhibitor in China to demonstrate superior efficacy in second-line or third-line treatment of patients with advanced or metastatic non-small-cell lung cancer (NSCLC). This study aimed to evaluate the cost-effectiveness of tislelizumab compared to docetaxel from a Chinese healthcare system perspective. Methods: A dynamic Markov model was developed to evaluate the cost-effectiveness of tislelizumab in comparison to docetaxel in second or third-line treatment. The efficacy data utilized in the model were derived from the RATIONALE-303 clinical trial, while cost and utility values were obtained from the drug data service platform and published studies. The primary outcomes of the model encompassed quality-adjusted life years (QALYs), costs, and incremental cost-effectiveness ratios (ICERs). One-way sensitivity analysis and probabilistic sensitivity analysis were conducted to validate the robustness of the base case analysis results. Results: The tislelizumab group demonstrated a cost increase of CNY 117,473 and a gain of 0.58 QALYs compared to the docetaxel group, resulting in an ICER value of CNY 202,927 per QALY gained. Conclusion: The administration of tislelizumab in patients with advanced or metastatic NSCLC not only extends the progression-free survival (PFS) and overall survival (OS). Moreover, this treatment demonstrates a favorable cost-effectiveness profile across the Chinese population.


Sujet(s)
Anticorps monoclonaux humanisés , Carcinome pulmonaire non à petites cellules , Évaluation du Coût-Efficacité , Docetaxel , Tumeurs du poumon , Années de vie ajustées sur la qualité , Femelle , Humains , Mâle , Adulte d'âge moyen , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/économie , Antinéoplasiques/économie , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Chine , Docetaxel/usage thérapeutique , Docetaxel/économie , Tumeurs du poumon/traitement médicamenteux , Chaines de Markov
4.
BMC Urol ; 24(1): 135, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951868

RÉSUMÉ

The ARASENS trial recruited 1306 men with metastatic hormone sensitive prostate cancer. It investigated the effect of androgen deprivation therapy (ADT) and systemic therapy docetaxel in combination with a third novel drug - daralutamide, compared with placebo on overall survival. Triple therapy with ADT, docetaxel and darolutamide resulted in improved overall survival rates as compared with ADT, docetaxel and placebo (HR 0.68; 95% CI, 0.57-0.80; p < 0.001). The side effect profile for both treatments was similar. This randomised, double blinded, placebo controlled study, was assessed to have a low risk of bias using the Cochrane Risk of Bias 2 tool.


Sujet(s)
Tumeurs de la prostate , Mâle , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/mortalité , Benzamides/usage thérapeutique , Essais contrôlés randomisés comme sujet , Taux de survie , Antagonistes des androgènes/usage thérapeutique , Docetaxel/usage thérapeutique , Pyrazoles
5.
Support Care Cancer ; 32(8): 554, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39066890

RÉSUMÉ

PURPOSE: Common side effects of taxane chemotherapy are nail toxicity and peripheral neuropathy (CIPN) causing severe impact on the quality of life. Different methods of cryotherapy to prevent these side effects have been tested. We investigated the use of machine-controlled cooling of hands and feet to reduce nail toxicity and CIPN in patients receiving taxane chemotherapy. METHODS: Patients receiving Docetaxel (planned dose ≥ 300 mg/m2) or Paclitaxel (planned dose ≥ 720 mg/m2 - ) in the adjuvant or palliative setting of different cancers were included. The dominant hand and foot were cooled to approximately 10 °C using the Hilotherapy machine. The contralateral hand and foot were used as intrapatient comparison. The primary endpoint was the occurrence of any CIPN due to paclitaxel or nail toxicity due to Docetaxel. Both the intention to treat population (ITT) and the per protocol population (PPP) were analyzed. RESULTS: A total of 69 patients, 21 treated with Docetaxel and 48 with Paclitaxel, were included at our centre between 08/2020 and 08/2022. Nail toxicity due to Docetaxel was overall not significantly improved by cooling in the ITT or PPP but a significant benefit across visits was found for the ITT. CIPN due to Paclitaxel was numerically better in the ITT and significantly better in the PPP. A significant benefit of cooling on CIPN occurrence across visits was found for the ITT and the PPP. Cooling was very well tolerated. CONCLUSION: Cooling of hands and feet has a clinically meaningful impact on reducing occurrence of CIPN and nail toxicity on treatment with taxanes. Effects are more significant over time and are dose dependent. TRIAL REGISTRATION NUMBER: 2020-00381. Date of registration. 24th February 2020.


Sujet(s)
Docetaxel , Onychopathies , Paclitaxel , Neuropathies périphériques , Humains , Femelle , Études prospectives , Adulte d'âge moyen , Mâle , Neuropathies périphériques/induit chimiquement , Neuropathies périphériques/prévention et contrôle , Sujet âgé , Docetaxel/administration et posologie , Docetaxel/effets indésirables , Paclitaxel/administration et posologie , Paclitaxel/effets indésirables , Onychopathies/thérapie , Onychopathies/induit chimiquement , Tumeurs/traitement médicamenteux , Adulte , Taxoïdes/effets indésirables , Taxoïdes/administration et posologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/administration et posologie , Cryothérapie/méthodes , Qualité de vie
6.
Anticancer Res ; 44(8): 3277-3285, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060082

RÉSUMÉ

BACKGROUND/AIM: Lipids are essential for energy production, signaling, and membrane formation, hence increased lipid metabolism may lead to cancer growth. 4-cholesten-3-one (4Cone), a sterol metabolite, has various biological activities, including the inhibition of cancer growth. This study examined whether 4Cone could change the lipid profile of triple-negative breast cancer cells (MDA-MB-231) and whether in combination with the anti-cancer chemotherapy docetaxel (TXT) could further reduce cancer aggressiveness. MATERIALS AND METHODS: The effect of 4Cone, TXT, or their combination (4Cone/TXT) on migration and proliferation was examined utilizing the wound healing and MTT assays. The expression of the lipogenesis-related enzymes was assessed using RT-qPCR and lipid profile was examined using mass spectrometry. RESULTS: 4Cone and TXT individually reduced cell viability and migration of MDA-MB-231 cancer cells; however, their combination (4Cone/TXT) had a greater impact on both attributes. All treated cells showed markedly decreased levels of the multidrug resistance enzyme PGP as well as the lipogenic enzymes FASN, ACC1, SCD1, HMGCR, and DGAT. Furthermore, lipid fingerprints were markedly different in treated cells compared with the untreated group. 4Cone increased the percentage of sphingomyelin (SM) while it decreased the percentage of ceramide (Cer); 4Cone in conjunction with TXT had the reverse effect. Triglyceride levels were reduced in 4Cone- and 4Cone/TXT-treated cells, but interestingly, they increased in TXT-treated cells. Additionally, treated cancer cells exhibited changes in glycerophospholipid subclasses. CONCLUSION: 4Cone alone or in combination with TXT alters the lipid profile by reducing a key lipogenic enzyme, resulting in the inhibition of cell proliferation and migration.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Docetaxel , Lipidomique , Humains , Docetaxel/pharmacologie , Lipidomique/méthodes , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Antinéoplasiques/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Cellules MDA-MB-231
7.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39063165

RÉSUMÉ

Human inflammatory breast cancer (IBC) and canine inflammatory mammary cancer (IMC) are highly aggressive neoplastic diseases that share numerous characteristics. In IBC and IMC, chemotherapy produces a limited pathological response and anti-androgen therapies have been of interest for breast cancer treatment. Therefore, the aim was to evaluate the effect of a therapy based on bicalutamide, a non-steroidal anti-androgen, with doxorubicin and docetaxel chemotherapy on cell proliferation, migration, tumor growth, and steroid-hormone secretion. An IMC-TN cell line, IPC-366, and an IBC-TN cell line, SUM149, were used. In vitro assays revealed that SUM149 exhibited greater sensitivity, reducing cell viability and migration with all tested drugs. In contrast, IPC-366 exhibited only significant in vitro reductions with docetaxel as a single agent or in different combinations. Decreased estrogen levels reduced in vitro tumor growth in both IMC and IBC. Curiously, doxorubicin resulted in low efficacy, especially in IMC. In addition, all drugs reduced the tumor volume in IBC and IMC by increasing intratumoral testosterone (T) levels, which have been related with reduced tumor progression. In conclusion, the addition of bicalutamide to doxorubicin and docetaxel combinations may represent a potential treatment for IMC and IBC.


Sujet(s)
Anilides , Prolifération cellulaire , Docetaxel , Cancers du sein inflammatoires , Tumeurs mammaires de l'animal , Nitriles , Composés tosyliques , Composés tosyliques/pharmacologie , Humains , Animaux , Femelle , Nitriles/pharmacologie , Nitriles/usage thérapeutique , Lignée cellulaire tumorale , Anilides/pharmacologie , Chiens , Cancers du sein inflammatoires/traitement médicamenteux , Cancers du sein inflammatoires/anatomopathologie , Cancers du sein inflammatoires/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Docetaxel/pharmacologie , Tumeurs mammaires de l'animal/traitement médicamenteux , Tumeurs mammaires de l'animal/anatomopathologie , Tumeurs mammaires de l'animal/métabolisme , Doxorubicine/pharmacologie , Souris , Survie cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Testostérone
8.
Biomed Pharmacother ; 177: 117125, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39002444

RÉSUMÉ

Active targeting to cancer involves exploiting specific interactions between receptors on the surface of cancer cells and targeting moieties conjugated to the surface of vectors such that site-specific delivery is achieved. Prostate specific membrane antigen (PSMA) has proved to be an excellent target for active targeting to prostate cancer. We report the synthesis and use of a PSMA-specific ligand (Glu-NH-CO-NH-Lys) for the site-specific delivery of brusatol- and docetaxel-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles to prostate cancer. The PSMA targeting ligand covalently linked to PLGA-PEG3400 was blended with methoxyPEG-PLGA to prepare brusatol- and docetaxel-loaded nanoparticles with different surface densities of the targeting ligand. Flow cytometry was used to evaluate the impact of different surface densities of the PSMA targeting ligand in LNCaP prostate cancer cells at 15 min and 2 h. Cytotoxicity evaluations of the targeted nanoparticles reveal differences based on PSMA expression in PC-3 and LNCaP cells. In addition, levels of reactive oxygen species (ROS) were measured using the fluorescent indicator, H2DCFDA, by flow cytometry. PSMA-targeted nanoparticles loaded with docetaxel and brusatol showed increased ROS generation in LNCaP cells compared to PC-3 at different time points. Furthermore, the targeted nanoparticles were evaluated in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors. Evaluation of the percent relative tumor volume show that brusatol-containing nanoparticles show great promise in inhibiting tumor growth. Our data also suggest that the dual drug-loaded targeted nanoparticle platform improves the efficacy of docetaxel in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors.


Sujet(s)
Antigènes de surface , Docetaxel , Glutamate carboxypeptidase II , Nanoparticules , Tumeurs de la prostate , Mâle , Docetaxel/pharmacologie , Docetaxel/administration et posologie , Animaux , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Glutamate carboxypeptidase II/métabolisme , Antigènes de surface/métabolisme , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Espèces réactives de l'oxygène/métabolisme , Cellules PC-3 , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Danio zébré , Souris nude , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Souris de lignée BALB C , Système d'administration de médicaments à base de nanoparticules/composition chimique
9.
PLoS One ; 19(7): e0306969, 2024.
Article de Anglais | MEDLINE | ID: mdl-38990953

RÉSUMÉ

Docetaxel (Doc) plays a crucial role in clinical antineoplastic practice. However, it is continuously documented that tumors frequently develop chemoresistance and relapse, which may be related to polyploid giant cancer cells (PGCCs). The aim of this study was investigate the formation mechanism and biological behavior of PGCCs induced by Doc. Ovarian cancer cells were treated with Doc, and then the effect of Doc on cellular viability was evaluated by MTT assay and microscopic imaging analysis. The biological properties of PGCCs were further evaluated by Hoechst 33342 staining, cell cycle and DNA content assay, DNA damage response (DDR) signaling detection, ß-galactosidase staining, mitochondrial membrane potential detection, and reverse transcription-quantitative polymerase chain reaction. The results indicated that Doc reduced cellular viability; however, many cells were still alive, and were giant and polyploid. Doc increased the proportion of cells stayed in the G2/M phase and reduced the number of cells. In addition, the expression of γ-H2A.X was constantly increased after Doc treatment. PGCCs showed senescence-associated ß-galactosidase activity and an increase in the monomeric form of JC-1. The mRNA level of octamer-binding transcription factor 4 (OCT4) and krüppel-like factor 4 (KLF4) was significantly increased in PGCCs. Taken together, our results suggest that Doc induces G2/M cell cycle arrest, inhibits the proliferation and activates persistent DDR signaling to promote the formation of PGCCs. Importantly, PGCCs exhibit a senescence phenotype and express stem cell markers.


Sujet(s)
Vieillissement de la cellule , Docetaxel , Facteur-4 de type Kruppel , Cellules souches tumorales , Tumeurs de l'ovaire , Polyploïdie , Humains , Docetaxel/pharmacologie , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/génétique , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Facteur de transcription Oct-3/métabolisme , Facteur de transcription Oct-3/génétique , Cellules géantes/effets des médicaments et des substances chimiques , Cellules géantes/métabolisme , Antinéoplasiques/pharmacologie , Phénotype , Survie cellulaire/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Taxoïdes/pharmacologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique
10.
Support Care Cancer ; 32(8): 513, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39001936

RÉSUMÉ

PURPOSE: Anthracycline-cyclophosphamide followed by docetaxel-containing chemotherapy is effective for perioperative breast cancer treatment. However, these treatments frequently induce oral mucositis (OM), with an incidence ranging from 20 to 50%. The association of OM development between different chemotherapeutic treatments remains unclear. Consequently, this study aimed to compare OM development during docetaxel-containing chemotherapy between patients with and without OM experience during previous anthracycline-cyclophosphamide treatments to assess the association between OM development and treatment regimens. METHODS: Seventy-two patients with breast cancer receiving anthracycline-cyclophosphamide followed by docetaxel-containing chemotherapy as a perioperative treatment were categorized into the control (no prior OM experience with anthracycline-cyclophosphamide) and OM-experience (OM development during previous treatment) groups and retrospectively evaluated. The primary endpoint was the incidence of all-grade OM in the first docetaxel-containing chemotherapy cycle. Additionally, the incidences of OM and dysgeusia during all treatment cycles and factors associated with the incidence of OM were evaluated. RESULTS: The incidence of all-grade OM in the first cycle was significantly higher in the OM-experience group (54.2%) than in the control group (10.4%; P < 0.0001). Furthermore, its incidence in all treatment cycles was higher in the OM-experience group (66.7%) than in the control group (12.5%, P < 0.0001). However, the incidence of dysgeusia did not differ between the groups. Multivariate logistic regression analysis revealed OM experience during previous anthracycline-cyclophosphamide treatment and concomitant pertuzumab use as independent risk factors for OM development in subsequent docetaxel-containing chemotherapy. CONCLUSION: Our study suggests that patients experiencing OM with anthracycline-cyclophosphamide during perioperative breast cancer treatment exhibit symptoms following subsequent docetaxel-containing chemotherapy.


Sujet(s)
Anthracyclines , Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Cyclophosphamide , Docetaxel , Stomatite , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/chirurgie , Femelle , Docetaxel/administration et posologie , Docetaxel/effets indésirables , Cyclophosphamide/administration et posologie , Cyclophosphamide/effets indésirables , Adulte d'âge moyen , Stomatite/induit chimiquement , Stomatite/épidémiologie , Études rétrospectives , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Anthracyclines/effets indésirables , Anthracyclines/administration et posologie , Adulte , Sujet âgé , Incidence , Taxoïdes/effets indésirables , Taxoïdes/administration et posologie , Facteurs de risque
11.
Eur J Pharm Biopharm ; 201: 114386, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38950717

RÉSUMÉ

The goal of the study was to fabricate folic acid functionalized docetaxel (DOC)/erlotinib (ERL)-loaded solid lipid nanoparticles (SLNs) to synergistically increase the anticancer activity against triple-negative breast cancer. DOC/ERL-SLNs were prepared by the high shear homogenization - ultrasound dispersion method (0.1 % w/v for DOC, and 0.3 %w/v for ERL) and optimized using Plackett Burman Design (PBD) followed by Box Behnken Design (BBD). The optimized SLNs demonstrated particle size < 200 nm, PDI < 0.35, and negative zeta potential with entrapment and loading efficiency of ∼80 and ∼4 %, respectively. The SLNs and folic acid functionalized SLNs (FA-SLNs) showed sustained release for both drugs, followed by Higuchi and Korsemeyer-Peppas drug release models, respectively. Further, the in vitro pH-stat lipolysis model demonstrated an approximately 3-fold increase in the bioaccessibility of drugs from SLNs compared to suspension. The TEM images revealed the spherical morphology of the SLNs. DOC/ERL loaded SLNs showed dose- and time-dependent cytotoxicity and exhibited a synergism at a molar ratio of 1:3 in TNBC with a combination index of 0.35 and 0.37, respectively. FA-DOC/ERL-SLNs showed enhanced anticancer activity as evidenced by MMP and ROS assay and further inhibited the colony-forming ability and the migration capacity of TNBC cells. Conclusively, the study has shown that SLNs are encouraging systems to improve the pharmaceutical attributes of poorly bioavailable drugs.


Sujet(s)
Docetaxel , Libération de médicament , Synergie des médicaments , Chlorhydrate d'erlotinib , Lipides , Nanoparticules , Taille de particule , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/traitement médicamenteux , Docetaxel/administration et posologie , Docetaxel/pharmacologie , Docetaxel/pharmacocinétique , Humains , Nanoparticules/composition chimique , Chlorhydrate d'erlotinib/administration et posologie , Chlorhydrate d'erlotinib/pharmacologie , Chlorhydrate d'erlotinib/pharmacocinétique , Lignée cellulaire tumorale , Femelle , Lipides/composition chimique , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Vecteurs de médicaments/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Acide folique/composition chimique , Liposomes
12.
J Cancer Res Ther ; 20(3): 913-917, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-39023597

RÉSUMÉ

INTRODUCTION: Adjuvant chemoradiotherapy (CRT) is the optimal management strategy in resectable gastric cancer. There is a debate about the efficacy of more aggressive CRT plus chemotherapy regimens in adjuvant setting. This study aimed to compare the efficacy of adjuvant CRT plus docetaxel-cisplatin-fluorouracil (DCF) versus CRT plus fluorouracil-folinic acid (FUFA) in stage III gastric cancer. METHODS: Patients with a diagnosis of stage III gastric cancer treated with adjuvant therapy after curative resection were analyzed. Patients' disease characteristics and impacts of the regimens on median disease-free survival (DFS) and median overall survival (OS) were analyzed retrospectively. RESULTS: One hundred sixty-one patients (102 in FUFA arm and 59 in DCF arm) with a median age of 56.0 (29-79) were evaluated. In the DCF arm, there were more renal toxicities (31.6% vs 6.4% P < 0.001), emergency department admissions (64.9% vs 23.7%, P < 0.001), and dose reductions/treatment modifications in the DCF arm (51.6% vs 37.2, P < 0.001). The median follow-up was 23 months (1-124) in the FUFA arm and 26.0 months (1-77) in the DCF arm. The median DFS was 25.0 months (%95 CI, 12.7-37.2) in the DCF arm and 17.0 months (%95 CI, 2.6-31.3) in the FUFA arm, P = 0.66. The median OS was 28.0 months (%95 CI, 17.0-38.9) in the DCF arm and 25.0 months (%95 CI, 11.9-36.0) in the FUFA arm, P = 0.70. CONCLUSION: In conclusion, when compared with FUFA regimen, more aggressive therapy with DCF was more toxic and did not improve OS in adjuvant setting of stage III gastric cancer.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Chimioradiothérapie adjuvante , Cisplatine , Docetaxel , Fluorouracil , Leucovorine , Stadification tumorale , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/thérapie , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/mortalité , Mâle , Adulte d'âge moyen , Femelle , Fluorouracil/administration et posologie , Fluorouracil/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Docetaxel/administration et posologie , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Sujet âgé , Adulte , Études rétrospectives , Chimioradiothérapie adjuvante/méthodes , Leucovorine/administration et posologie , Leucovorine/usage thérapeutique , Résultat thérapeutique
13.
Front Immunol ; 15: 1414529, 2024.
Article de Anglais | MEDLINE | ID: mdl-39076983

RÉSUMÉ

Sinonasal squamous cell carcinoma (SNSCC) is the most common, high-aggressive sinonasal malignancies that have remained relatively stable poor outcomes over the past decade. As a first-line treatment for SNSCC, surgery plus adjuvant radiotherapy is recommended. However, complete surgical resection may not be appropriate due to the proximity of the nasal cavity and sinuses to key structures such as orbit or intracranial. Currently, immune checkpoint inhibitors (ICIs) have been established as one of the first-line therapies for many solid tumors with unresectable stage. However, evidence on the efficacy of ICIs in sinonasal malignancy is scarce and no ICIs are approved for use in SNSCC up to day. In this report, we report a case of a 64-year-old man with SNSCC treated by multi-protocol exploration. The patient achieved pathological complete response (pCR) after receiving two cycles of Docetaxel and cisplatin combined with tislelizumab. To the best of our knowledge, this is the first case of SNSCC treated with tislelizumab that achieved pCR. This case offers real-world evidence that chemotherapy plus immunotherapy is a promising treatment for SNSCC.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Traitement néoadjuvant , Humains , Mâle , Adulte d'âge moyen , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Induction de rémission , Tumeurs des sinus de la face/traitement médicamenteux , Carcinome épidermoïde/traitement médicamenteux , Résultat thérapeutique , Cisplatine/usage thérapeutique , Cisplatine/administration et posologie , Docetaxel/usage thérapeutique , Docetaxel/administration et posologie
14.
Niger J Clin Pract ; 27(7): 827-836, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39082907

RÉSUMÉ

BACKGROUND: Docetaxel (DOC) is the main chemotherapeutic agent for the treatment of advanced metastatic prostate cancer. Docetaxel shows anticancer effects by preventing the depolymerization of microtubules in the cell, therefore preventing cell division. However, the low survival effect of docetaxel has prompted researchers to search for novel therapeutic agents. Fucoidan (FUC) is a sulfated polysaccharide derived from brown algae. It has many bioactivities which makes fucoidan a promising anticancer agent. In this study, the potential anti-tumorigenic and preventive effects of fucoidan with or without docetaxel in prostate cancer were investigated by analyzing different cell death modalities. METHODS: The in-vivo six groups (n = 8) were conducted; preventive (Pt), docetaxel treated after preventive (Pt-D), control, fucoidan (FUC), docetaxel (DOC), and FUC and DOC (FUC+DOC) combination. Apoptotic, necroptotic, and autophagic cell death-related protein expressions were assessed in tumor tissues by using immunohistochemical staining. Oxidative stress-related lipid peroxidation, glutathione peroxidase, and glutathione levels were also determined in tumor tissues. RESULTS: Although apoptotic, necroptotic, and autophagic cell deaths were significantly induced in agent-treated groups compared to the control. Apoptotic cell death was more significantly induced in FUC and FUC+DOC-treated groups. Necroptotic cell death was increased considerably by inducing MLKL protein expression in all treatment groups. In the FUC, Pt, and DOC groups, LC3A/B expressions were significantly increased. DOC, FUC+DOC, and Pt-D treatments caused a significant increase in Beclin-1 expression. Oxidative stress-related MDA, GPX, and GSH levels significantly decreased with FUC treatment. The anti-tumorigenic effects of FUC and DOC were also demonstrated through tumor size reduction. CONCLUSION: According to the findings of this study, FUC inhibited tumor growth temporally and dimensionally, especially in preventive applications. FUC and FUC+DOC combinations in both treatment groups showed anti-tumorigenic effects. The results of this study suggest that fucoidan is a promising anticancer agent against prostate cancer. FUC can be considered as a preventive or treatment agent in prostate cancer therapy with DOC. Further studies are needed to fully elucidate the mechanism of action of fucoidan in metastatic prostate cancer.


Sujet(s)
Apoptose , Docetaxel , Polyosides , Tumeurs de la prostate , Mâle , Polyosides/pharmacologie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Docetaxel/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Humains , Antinéoplasiques/pharmacologie , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Mort cellulaire/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Souris
15.
Cell Rep ; 43(7): 114431, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38968071

RÉSUMÉ

Bromodomain-containing protein 4 (BRD4) has emerged as a promising therapeutic target in prostate cancer (PCa). Understanding the mechanisms of BRD4 stability could enhance the clinical response to BRD4-targeted therapy. In this study, we report that BRD4 protein levels are significantly decreased during mitosis in a PLK1-dependent manner. Mechanistically, we show that BRD4 is primarily phosphorylated at T1186 by the CDK1/cyclin B complex, recruiting PLK1 to phosphorylate BRD4 at S24/S1100, which are recognized by the APC/CCdh1 complex for proteasome pathway degradation. We find that PLK1 overexpression lowers SPOP mutation-stabilized BRD4, consequently rendering PCa cells re-sensitized to BRD4 inhibitors. Intriguingly, we report that sequential treatment of docetaxel and JQ1 resulted in significant inhibition of PCa. Collectively, the results support that PLK1-phosphorylated BRD4 triggers its degradation at M phase. Sequential treatment of docetaxel and JQ1 overcomes BRD4 accumulation-associated bromodomain and extra-terminal inhibitor (BETi) resistance, which may shed light on the development of strategies to treat PCa.


Sujet(s)
Azépines , Protéines du cycle cellulaire , Docetaxel , Résistance aux médicaments antinéoplasiques , Mitose , , Tumeurs de la prostate , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Facteurs de transcription , Triazoles , Humains , Protéines du cycle cellulaire/métabolisme , Mâle , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Phosphorylation , Protéines proto-oncogènes/métabolisme , Mitose/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Facteurs de transcription/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Azépines/pharmacologie , Triazoles/pharmacologie , Docetaxel/pharmacologie , Protéolyse/effets des médicaments et des substances chimiques , Protéines nucléaires/métabolisme , Animaux , Protéine-kinase CDC2/métabolisme , Souris nude , Souris , Proteasome endopeptidase complex/métabolisme , Protéines contenant un bromodomaine , Protéines de répression
16.
Sci Rep ; 14(1): 16164, 2024 07 13.
Article de Anglais | MEDLINE | ID: mdl-39003369

RÉSUMÉ

The present study investigated the relationship between MSH3 and MSH6 genes in lung cancer patients. Genotyping of lung cancer patients and healthy controls was performed. Odds ratio values were calculated and survival analysis performed. Patients with mutant genotype (TT) for MSH6 polymorphism have 1.5-fold risk for the development of lung cancer (p = 0.03). For non-smokers, the mutant-type genotype had a threefold increased risk of lung cancer (p = 0.01). Patients administered with docetaxel and carbo/cisplatin and carrying GT genotype for MSH6 polymorphism, patients reported a decrease in median survival time (4.9 vs 9.13 months). MSH3 and MSH6 polymorphisms are involved in modulating the risk towards lung cancer. MSH6 polymorphism is associated with high mortality rate for patients undergoing cisplatin and docetaxel chemotherapy.


Sujet(s)
Cisplatine , Protéines de liaison à l'ADN , Prédisposition génétique à une maladie , Tumeurs du poumon , Protéine-3 homologue de MutS , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/mortalité , Mâle , Adulte d'âge moyen , Femelle , Cisplatine/usage thérapeutique , Protéine-3 homologue de MutS/génétique , Protéines de liaison à l'ADN/génétique , Polymorphisme de nucléotide simple , Docetaxel/usage thérapeutique , Inde/épidémiologie , Sujet âgé , Études cas-témoins , Génotype , Adulte , Carboplatine/usage thérapeutique
17.
Hinyokika Kiyo ; 70(6): 141-147, 2024 Jun.
Article de Japonais | MEDLINE | ID: mdl-38967025

RÉSUMÉ

The administration of cabazitaxel for patients with castration-resistant prostate cancer (CRPC) requires prior docetaxel therapy. Sequential chemotherapy may have to be discontinued due to docetaxelassociated side effects. This study investigated the relationship between treatment outcome of docetaxel and cabazitaxel and their associated side effects. We retrospectively analyzed 69 patients with CRPC who had been administered docetaxel withand without subsequent cabazitaxel at Toyonaka Municipal Hospital from October 2014 to June 2022. Twenty-eight patients (41%) discontinued docetaxel because of side effects, and the median number of docetaxel cycles at discontinuation was 2 (range : 1-11). Fourteen of these patients received no treatment following docetaxel. A comparison of the 28 patients who had discontinued docetaxel due to side effects with 41 patients who had not revealed a significant difference in the total numbers of chemotherapy cycles (2.5 vs 9 ; P<0.001) and time to treatment failure (56 days vs 301 days ; P= 0.001), with a trend toward shorter overall survival from the start of docetaxel treatment (259 days vs 512 days ; P=0.06). Multivariate analysis identified discontinuation of docetaxel due to side effects (OR=0.07 ; P<0.001) and lower hemoglobin (OR=0.01 ; P=0.001) as significant factors inhibiting the introduction of cabazitaxel. Reducing the side effects of docetaxel, including early drug switching, may allow more CRPC patients to be reached with cabazitaxel. Consequently, the resulting taxane-based chemotherapy may contribute to an additional survival advantage.


Sujet(s)
Docetaxel , Tumeurs prostatiques résistantes à la castration , Taxoïdes , Humains , Mâle , Taxoïdes/effets indésirables , Taxoïdes/administration et posologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Docetaxel/administration et posologie , Docetaxel/effets indésirables , Sujet âgé , Adulte d'âge moyen , Études rétrospectives , Sujet âgé de 80 ans ou plus , Antinéoplasiques/effets indésirables , Antinéoplasiques/administration et posologie , Résultat thérapeutique
18.
Zhonghua Yi Xue Za Zhi ; 104(26): 2401-2408, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38978363

RÉSUMÉ

Objective: To compare the efficacy and safety of carrelizumab combined with the modified TPF regimen (docetaxel, cisplatinand capecitabine) and TPF regimen alone in larynx preservation strategy for locally advanced resectable hypopharyngeal squamous cell carcinoma. Methods: A cohort study was conducted. Patients with locally advanced resectable hypopharyngeal carcinoma (cT3-4aN0-3bM0) who were treated at the Eye & ENT Hospital of Fudan University from January 2017 to April 2023 were enrolled in the study. One group was treated with a modified TPF regimen (TPF group) for 2-3 cycles (retrospective data), and the other group was a prospective phase Ⅱ trial with a modified TPF regimen combined with carrelizumab (TPFC group) for three cycles. The patients with complete or partial remission of the primary focus were treated with sequential radical radiotherapy and/or drug therapy. The patients in the TPFC group were treated with carrelizumab at the end of radiotherapy with a maximum of up to 18 doses. The patients with stable or progressive disease were given radical surgery, and those who refused the surgery were given radical chemoradiotherapy. Objective response rate (ORR), overall survival rate, progression-free survival (PFS) rate, larynx preservation rate (LPR), and adverse reactions were compared between the two groups. Results: There were 51 male patients in the TPFC group, with an median age of 57 (35, 69) years. Meanwhile, 44 patients were in the TPF group, among which 43 were male and one was female, with an median age of 62 (46, 70) years. The ORR of the TPFC group was higher than that of the TPF group [82.4% (42/51) vs 63.6% (28/44), P=0.039]. During a median follow-up of 24.4 (18.5, 31.4) months, the TPFC group showed a higher 2-year survival rate (84.8% vs 64.6%, P=0.013) and 2-year LPR (66.6% vs 48.6%, P=0.045) than those in the TPF group. In patients with poor effect of induction therapy for hypopharyngeal carcinoma, surgical combination therapy significantly prolonged the 2-year PFS rate (77.9% vs 18.2%, P<0.001) and 2-year survival rate (76.9% vs 45.5%, P=0.005)than those of non-surgical combination therapy. The incidences of nausea and/or vomiting, reactive cutaneous capillary endothelial proliferation, thyroid dysfunction, and rash were increased in the TPFC group (all P<0.05). There was no treatment-related death. Conclusion: Carrelizumab combined with a modified TPF regimen has good efficacy and safety and can improve the LPR of locally advanced hypopharyngeal carcinoma.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs de l'hypopharynx , Humains , Tumeurs de l'hypopharynx/traitement médicamenteux , Tumeurs de l'hypopharynx/thérapie , Tumeurs de l'hypopharynx/anatomopathologie , Mâle , Femelle , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte d'âge moyen , Cisplatine/administration et posologie , Études prospectives , Chimiothérapie d'induction , Études de cohortes , Études rétrospectives , Anticorps monoclonaux humanisés/usage thérapeutique , Docetaxel/usage thérapeutique , Docetaxel/administration et posologie , Résultat thérapeutique , Adulte
19.
Biol Pharm Bull ; 47(7): 1326-1330, 2024.
Article de Anglais | MEDLINE | ID: mdl-39048353

RÉSUMÉ

In perioperative chemotherapy for breast cancer, dexamethasone (DEX) is administered at high dose to prevent adverse effects. Abrupt cessation of high-dose DEX treatment induces fatigue, but the incidence of the fatigue is uncertain. In this study, we retrospectively evaluated the incidence of fatigue following DEX administration for supportive therapy and the improvement of fatigue with DEX tapering, a gradual reduction of the daily dose, in breast cancer patients. The subjects were 124 patients with breast cancer receiving epirubicin- or docetaxel-based regimens as perioperative chemotherapy. Of all patients, 16.1% of patients experienced fatigue after cessation of DEX administration. The severity of fatigue was grade 1 in 6.5% of patients, grade 2 in 8.1% of patients, and grade 3 in 1.6% of patients. There were no significant differences in dose and duration of DEX administration between the group with fatigue and the group without fatigue. In almost all patients with fatigue, DEX tapering was performed from the next cycle. The efficacy of DEX tapering was evaluated by comparing the grade and subjective symptoms. Following DEX tapering, the severity of fatigue was significantly reduced (p < 0.05), and the subjective symptom was improved in 94.7% of patients. Therefore, fatigue is occasionally induced after the cessation of DEX administration for supportive therapy in breast cancer patients. The tapering of DEX may be effective for fatigue.


Sujet(s)
Tumeurs du sein , Dexaméthasone , Fatigue , Humains , Dexaméthasone/administration et posologie , Dexaméthasone/effets indésirables , Dexaméthasone/usage thérapeutique , Tumeurs du sein/chirurgie , Tumeurs du sein/traitement médicamenteux , Femelle , Études rétrospectives , Fatigue/traitement médicamenteux , Fatigue/étiologie , Adulte d'âge moyen , Adulte , Sujet âgé , Incidence , Épirubicine/administration et posologie , Épirubicine/effets indésirables , Épirubicine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Soins périopératoires/méthodes , Docetaxel/administration et posologie , Docetaxel/effets indésirables , Docetaxel/usage thérapeutique , Diminution progressive de la dose du médicament , Antinéoplasiques/effets indésirables , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique
20.
Oncol Rep ; 52(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38963044

RÉSUMÉ

Lysine methyltransferase 5A (KMT5A) is the sole mammalian enzyme known to catalyse the mono­methylation of histone H4 lysine 20 and non­histone proteins such as p53, which are involved in the occurrence and progression of numerous cancers. The present study aimed to determine the function of KMT5A in inducing docetaxel (DTX) resistance in patients with breast carcinoma by evaluating glucose metabolism and the underlying mechanism involved. The upregulation or downregulation of KMT5A­related proteins was examined after KMT5A knockdown in breast cancer (BRCA) cells by Tandem Mass Tag proteomics. Through differential protein expression and pathway enrichment analysis, the upregulated key gluconeogenic enzyme fructose­1,6­bisphosphatase 1 (FBP1) was discovered. Loss of FBP1 expression is closely related to the development and prognosis of cancers. A dual­luciferase reporter gene assay confirmed that KMT5A inhibited the expression of FBP1 and that overexpression of FBP1 could enhance the chemotherapeutic sensitivity to DTX through the suppression of KMT5A expression. The KMT5A inhibitor UNC0379 was used to verify that DTX resistance induced by KMT5A through the inhibition of FBP1 depended on the methylase activity of KMT5A. According to previous literature and interaction network structure, it was revealed that KMT5A acts on the transcription factor twist family BHLH transcription factor 1 (TWIST1). Then, it was verified that TWSIT1 promoted the expression of FBP1 by using a dual­luciferase reporter gene experiment. KMT5A induces chemotherapy resistance in BRCA cells by promoting cell proliferation and glycolysis. After the knockdown of the KMT5A gene, the FBP1 related to glucose metabolism in BRCA was upregulated. KMT5A knockdown expression and FBP1 overexpression synergistically inhibit cell proliferation and block cells in the G2/M phase. KMT5A inhibits the expression of FBP1 by methylating TWIST1 and weakening its promotion of FBP1 transcription. In conclusion, KMT5A was shown to affect chemotherapy resistance by regulating the cell cycle and positively regulate glycolysis­mediated chemotherapy resistance by inhibiting the transcription of FBP1 in collaboration with TWIST1. KMT5A may be a potential therapeutic target for chemotherapy resistance in BRCA.


Sujet(s)
Tumeurs du sein , Docetaxel , Résistance aux médicaments antinéoplasiques , Fructose-1,6-diphosphatase , Régulation de l'expression des gènes tumoraux , Protéines nucléaires , Protéine-1 apparentée à Twist , Humains , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Protéine-1 apparentée à Twist/génétique , Protéine-1 apparentée à Twist/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Fructose-1,6-diphosphatase/génétique , Fructose-1,6-diphosphatase/métabolisme , Docetaxel/pharmacologie , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Méthylation de l'ADN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE