Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 62.024
Filtrer
1.
Front Immunol ; 15: 1410250, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979408

RÉSUMÉ

Nasopharyngeal carcinoma with bone marrow metastasis presents a rare and challenging clinical scenario associated with exceedingly poor prognosis. While standard treatment regimens offer limited efficacy and tolerability in such cases, individualized approaches are increasingly necessary. We present the case of a 64-year-old male diagnosed with recurrent nonkeratinizing undifferentiated nasopharyngeal carcinoma with extensive bone marrow metastasis (rTxN0M1). Treatment was initiated with immunotherapy-based combination therapy, consisting of pembrolizumab and low-dose cisplatin, which resulted in an initial response. Subsequently, there was a transition to standard-dose nab-paclitaxel-cisplatin chemotherapy in combination with pembrolizumab, followed by maintenance therapy with pembrolizumab plus fruquintinib. The patient achieved a sustained response with renormalization of tumor markers, imaging findings, and bone biopsies, resulting in complete remission. This case highlights the successful management of nasopharyngeal carcinoma with extensive bone marrow metastasis through an individualized treatment approach incorporating immunotherapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs de la moelle osseuse , Cancer du nasopharynx , Tumeurs du rhinopharynx , Humains , Mâle , Adulte d'âge moyen , Cancer du nasopharynx/thérapie , Cancer du nasopharynx/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du rhinopharynx/thérapie , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs de la moelle osseuse/secondaire , Tumeurs de la moelle osseuse/thérapie , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Immunothérapie/méthodes , Induction de rémission , Paclitaxel/administration et posologie , Paclitaxel/usage thérapeutique , Résultat thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Albumines
2.
Int J Nanomedicine ; 19: 6603-6618, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979533

RÉSUMÉ

Objective: Ovarian cancer cells are prone to acquire tolerance to chemotherapeutic agents, which seriously affects clinical outcomes. The development of novel strategies to enhance the targeting of chemotherapeutic agents to overcome drug resistance and minimize side effects is significant for improving the clinical outcomes of ovarian cancer patients. Methods: We employed folic acid (FA)-modified ZIF-90 nanomaterials (FA-ZIF-90) to deliver the chemotherapeutic drug, cisplatin (DDP), via dual targeting to improve its targeting to circumvent cisplatin resistance in ovarian cancer cells, especially by targeting mitochondria. FA-ZIF-90/DDP could rapidly release DDP in response to dual stimulation of acidity and ATP in tumor cells. Results: FA-ZIF-90/DDP showed good blood compatibility. It was efficiently taken up by human ovarian cancer cisplatin-resistant cells A2780/DDP and aggregated in the mitochondrial region. FA-ZIF-90/DDP significantly inhibited the mitochondrial activity and metastatic ability of A2780/DDP cells. In addition, it effectively induced apoptosis in A2780/DDP cells and overcame cisplatin resistance. In vivo experiments showed that FA-ZIF-90/DDP increased the accumulation of DDP in tumor tissues and significantly inhibited tumor growth. Conclusion: FA-modified ZIF-90 nanocarriers can improve the tumor targeting and anti-tumor effects of chemotherapeutic drugs, reduce toxic side effects, and are expected to be a novel therapeutic strategy to reverse drug resistance in ovarian cancer.


Sujet(s)
Antinéoplasiques , Apoptose , Cisplatine , Résistance aux médicaments antinéoplasiques , Acide folique , Imidazoles , Tumeurs de l'ovaire , Zéolites , Femelle , Cisplatine/pharmacologie , Cisplatine/composition chimique , Cisplatine/pharmacocinétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Zéolites/composition chimique , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Acide folique/composition chimique , Acide folique/pharmacologie , Imidazoles/composition chimique , Imidazoles/pharmacologie , Imidazoles/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments/méthodes , Mitochondries/effets des médicaments et des substances chimiques , Souris , Souris de lignée BALB C , Souris nude , Vecteurs de médicaments/composition chimique , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
3.
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
4.
J Gastric Cancer ; 24(3): 300-315, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38960889

RÉSUMÉ

PURPOSE: Gastric cancer (GC) is among the deadliest malignancies and the third leading cause of cancer-related deaths worldwide. Galectin-1 (Gal-1) is a primary protein secreted by cancer-associated fibroblasts (CAFs); however, its role and mechanisms of action of Gal-1 in GC remain unclear. In this study, we stimulated GC cells with exogenous human recombinant galectin-1 protein (rhGal-1) to investigate its effects on the proliferation, migration, and resistance to cisplatin. MATERIALS AND METHODS: We used simulated rhGal-1 protein as a paracrine factor produced by CAFs to induce GC cells and investigated its promotional effects and mechanisms in GC progression and cisplatin resistance. Immunohistochemical (IHC) assay confirmed that Gal-1 expression was associated with clinicopathological parameters and correlated with the expression of neuropilin-1 (NRP-1), c-JUN, and Wee1. RESULTS: Our study reveals Gal-1 expression was significantly associated with poor outcomes. Gal-1 boosts the proliferation and metastasis of GC cells by activating the NRP-1/C-JUN/Wee1 pathway. Gal-1 notably increases GC cell resistance to cisplatin The NRP-1 inhibitor, EG00229, effectively counteracts these effects. CONCLUSIONS: These findings revealed a potential mechanism by which Gal-1 promotes GC growth and contributes to chemoresistance, offering new therapeutic targets for the treatment of GC.


Sujet(s)
Prolifération cellulaire , Cisplatine , Résistance aux médicaments antinéoplasiques , Galectine 1 , Neuropiline 1 , Tumeurs de l'estomac , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Humains , Galectine 1/génétique , Galectine 1/métabolisme , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Neuropiline 1/métabolisme , Neuropiline 1/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Femelle , Évolution de la maladie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Souris , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie
5.
Cell Mol Life Sci ; 81(1): 292, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976080

RÉSUMÉ

Cisplatin resistance is a major challenge for systemic therapy against advanced bladder cancer (BC). Little information is available on the regulation of cisplatin resistance and the underlying mechanisms require elucidation. Here, we detected that downregulation of the tumor suppressor, PPP2R2B (a serine/threonine protein phosphatase 2 A regulatory subunit), in BC promoted cell proliferation and migration. What's more, low PPP2R2B expression was correlated with cisplatin resistance. In vitro and in vivo experiments verified that PPP2R2B could promote BC sensitivity to cisplatin. In terms of mechanism, we identified a novel function of PPP2R2B as a nucleocytoplasmic transport molecule. PPP2R2B promoted ISG15 entry into the nucleus by mediating binding of IPO5 with ISG15. Nuclear translocation of ISG15 inhibited DNA repair, further increasing ISG15 expression through activation of the STING pathway. Besides, PPP2R2B was down-regulated by SUV39H1-mediated histone 3 lysine 9 trimethylation, which could be restored by the SUV39H1-specific inhibitor, chaetocin. Our data suggest that PPP2R2B expression level is a potential biomarker for chemotherapy response and that chemotherapy in combination with chaetocin may be a feasible treatment strategy for patients with BC.


Sujet(s)
Cisplatine , Cytokines , Résistance aux médicaments antinéoplasiques , Protein Phosphatase 2 , Ubiquitines , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Protein Phosphatase 2/métabolisme , Protein Phosphatase 2/génétique , Humains , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Ubiquitines/métabolisme , Ubiquitines/génétique , Cytokines/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Noyau de la cellule/métabolisme , Antinéoplasiques/pharmacologie , Souris de lignée BALB C , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Femelle , Protéines de tissu nerveux
6.
Int J Mol Med ; 54(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38963035

RÉSUMÉ

Globally, non­small cell lung cancer (NSCLC) is a significant threat to human health, and constitutes >80% of lung cancer cases. Cisplatin (CDDP), a commonly used drug in clinical treatment, has been the focus of research aiming to mitigate its potent toxicity through encapsulation within liposomes. However, challenges, such as a reduced drug loading efficiency and nonspecific release, have emerged as obstacles. The present study aimed to improve the encapsulation efficiency of CDDP within liposomes by pre­preparation of CDDP and modifying the liposome surface through the incorporation of peanut agglutinin (PNA) as a ligand [CDDP­loaded PNA­modified liposomes (CDDP­PNA­Lip)]. This strategy was designed to enhance the delivery of CDDP to tumour tissues, thereby reducing associated side effects. The effect of CDDP­PNA­Lip on the proliferation and migration of NSCLC cell lines with high MUC1 expression was elucidated through in vitro studies. Additionally, the capacity of PNA modification to augment the targeted anti­tumour efficacy of liposomes was assessed through xenograft tumour experiments. The results indicated that in an in vitro uptake assay Rhodamine B (RhB)­loaded PNA­modified liposomes were taken up by cells with ~50% higher efficiency compared with free RhB. In addition, CDDP­PNA­Lip resulted in a 2.65­fold enhancement of tumour suppression in vivo compared with free CDDP. These findings suggested that the encapsulation of CDDP within ligand­modified liposomes may significantly improve its tumour­targeting capabilities, providing valuable insights for clinical drug development.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Cisplatine , Liposomes , Tumeurs du poumon , Agglutinine cacahuète , Cisplatine/pharmacologie , Cisplatine/administration et posologie , Liposomes/composition chimique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Animaux , Agglutinine cacahuète/composition chimique , Lignée cellulaire tumorale , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques , Femelle , Systèmes de délivrance de médicaments/méthodes
7.
BMC Cancer ; 24(1): 833, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997665

RÉSUMÉ

BACKGROUND: This study aimed to investigate the efficacy and safety of pegylated recombinant human granulocyte colony-stimulating factor (PEG-rhG-CSF) for primary prophylaxis of neutropenia in patients with cervical cancer receiving concurrent chemoradiotherapy. METHODS: In this prospective, single-center, single-arm study, we enrolled patients (18-70 years) with 2018 International Federation of Gynecology and Obstetrics (FIGO) stage IIIC1r-IVA and IVB (distant metastasis only with inguinal lymph node metastasis) cervical cancer. Eligible patients should have normal function of the bone marrow (absolute neutrophil count (ANC) ≥ 2.0 × 109/L) and adequate hepatic and renal functions. Key exclusion criteria included: previous chemotherapy and/or radiotherapy; a history of bone marrow dysplasia or other hematopoietic abnormalities. All patients underwent radical radiotherapy (pelvic radiotherapy or extended-field irradiation) plus brachytherapy. The chemotherapy regimen included four cycles of 3-weekly paclitaxel and cisplatin. PEG-rhG-CSF was administered 48-72 h after each treatment cycle. Salvage granulocyte colony-stimulating factor (G-CSF) was only permitted in certain circumstances. The primary endpoint was the incidence of grade 3-4 neutropenia. The secondary endpoints included frequency of febrile neutropenia (FN), chemotherapy completion rate in cycles 2-4, time to complete radiotherapy, and safety. RESULTS: Overall, 52 patients were enrolled in this study from July 2019 to October 2020. The incidence of grade 3-4 neutropenia was 28.8%, with an average duration of grade 3-4 neutropenia persistence of 3.85 days (1-7 days). The incidence rate of FN was 3.8%. The chemotherapy completion rate was 94.2%, 82.7%, and 75.0% for cycles 2-4, respectively. The incidences of grade 3-4 neutropenia for cycles 1-4 were 9.6% (5/52), 8.2% (4/49), 14.0% (6/43), and 2.6% (1/39), respectively. All patients completed radiotherapy within 8 weeks (median, 48 days; range: 41-56 days), except one patient who withdrew consent and did not receive radiotherapy. Severe non-hematologic toxicity was not observed in any patient. CONCLUSION: PEG-rhG-CSF is an effective and safe prophylactic treatment for neutropenia in patients with cervical cancer undergoing concurrent chemoradiotherapy. TRIAL REGISTRATION: Chinese Clinical Trial Registry, ChiCTR1900024494. Date of Registration:13/July/2019.


Sujet(s)
Chimioradiothérapie , Facteur de stimulation des colonies de granulocytes , Neutropénie , Polyéthylène glycols , Protéines recombinantes , Tumeurs du col de l'utérus , Humains , Tumeurs du col de l'utérus/thérapie , Femelle , Adulte d'âge moyen , Adulte , Études prospectives , Chimioradiothérapie/effets indésirables , Chimioradiothérapie/méthodes , Facteur de stimulation des colonies de granulocytes/usage thérapeutique , Facteur de stimulation des colonies de granulocytes/administration et posologie , Polyéthylène glycols/administration et posologie , Polyéthylène glycols/usage thérapeutique , Polyéthylène glycols/effets indésirables , Protéines recombinantes/administration et posologie , Protéines recombinantes/usage thérapeutique , Sujet âgé , Neutropénie/prévention et contrôle , Neutropénie/étiologie , Cisplatine/effets indésirables , Cisplatine/usage thérapeutique , Cisplatine/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Jeune adulte , Adolescent , Paclitaxel/effets indésirables , Paclitaxel/administration et posologie , Paclitaxel/usage thérapeutique
8.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39000064

RÉSUMÉ

Chondrosarcoma (CS) is a rare malignant bone sarcoma that primarily affects cartilage cells in the femur and pelvis. While most subtypes exhibit slow growth with a very good prognosis, some aggressive subtypes have a poorer overall survival. CS is known for its resistance to chemotherapy and radiotherapy, leaving surgery as the sole effective therapeutic option. Cold physical plasma (CPP) has been explored in vitro as a potential therapy, demonstrating positive anti-tumor effects on CS cells. This study investigated the synergistic effects of combining CPP with cytostatics on CS cells. The chemotherapeutic agents cisplatin, doxorubicin, and vincristine were applied to two CS cell lines (CAL-78 and SW1353). After determining their IC20 and IC50, they were combined with CPP in both cell lines to assess their impact on the cell proliferation, viability, metabolism, and apoptosis. This combined approach significantly reduced the cell proliferation and viability while increasing the apoptosis signals compared to cytostatic therapy alone. The combination of CPP and chemotherapeutic drugs shows promise in targeting chemoresistant CS cells, potentially improving the prognosis for patients in clinical settings.


Sujet(s)
Apoptose , Tumeurs osseuses , Prolifération cellulaire , Survie cellulaire , Chondrosarcome , Doxorubicine , Gaz plasmas , Chondrosarcome/traitement médicamenteux , Chondrosarcome/anatomopathologie , Humains , Gaz plasmas/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/thérapie , Antinéoplasiques/pharmacologie , Cisplatine/pharmacologie , Vincristine/pharmacologie , Association thérapeutique
9.
Cell Death Dis ; 15(7): 501, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003251

RÉSUMÉ

Eukaryotic elongation factor 2 (eEF2) kinase (eEF2K) is a stress-responsive hub that inhibits the translation elongation factor eEF2, and consequently mRNA translation elongation, in response to hypoxia and nutrient deprivation. EEF2K is also involved in the response to DNA damage but its role in response to DNA crosslinks, as induced by cisplatin, is not known. Here we found that eEF2K is critical to mediate the cellular response to cisplatin. We uncovered that eEF2K deficient cells are more resistant to cisplatin treatment. Mechanistically, eEF2K deficiency blunts the activation of the DNA damage response associated ATM and ATR pathways, in turn preventing p53 activation and therefore compromising induction of cisplatin-induced apoptosis. We also report that loss of eEF2K delays the resolution of DNA damage triggered by cisplatin, suggesting that eEF2K contributes to DNA damage repair in response to cisplatin. In support of this, our data shows that eEF2K promotes the expression of the DNA repair protein ERCC1, critical for the repair of cisplatin-caused DNA damage. Finally, using Caenorhabditis elegans as an in vivo model, we find that deletion of efk-1, the worm eEF2K ortholog, mitigates the induction of germ cell death in response to cisplatin. Together, our data highlight that eEF2K represents an evolutionary conserved mediator of the DNA damage response to cisplatin which promotes p53 activation to induce cell death, or alternatively facilitates DNA repair, depending on the extent of DNA damage.


Sujet(s)
Caenorhabditis elegans , Cisplatine , Altération de l'ADN , Elongation Factor 2 Kinase , Protéine p53 suppresseur de tumeur , Cisplatine/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Elongation Factor 2 Kinase/métabolisme , Elongation Factor 2 Kinase/génétique , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/génétique , Caenorhabditis elegans/métabolisme , Humains , Réparation de l'ADN/effets des médicaments et des substances chimiques , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Apoptose/effets des médicaments et des substances chimiques
10.
Sci Rep ; 14(1): 16164, 2024 Jul 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
11.
BMC Cancer ; 24(1): 838, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003442

RÉSUMÉ

BACKGROUND: The National Comprehensive Cancer Network (NCCN) guideline recommends consideration of weekly cisplatin as an alternative option for patients with head and neck cancer undergoing definitive chemoradiation. However, in a recent phase III trial (ConCERT), 20% of patients treated with weekly cisplatin could not receive a total of 200 mg/m2, and the association of low adherence to weekly cisplatin and cancer control outcomes remains unclear. To fill this knowledge gap, we performed an observational cohort study of patients with head and neck cancer undergoing definitive chemoradiation with weekly cisplatin. METHODS: Our institutional database was queried for patients with non-metastatic head and neck cancer who underwent definitive chemoradiation with weekly cisplatin (40 mg/m2) between November 2007 and April 2023. Adherence to weekly cisplatin was defined as receiving at least 5 cycles with a total cumulative dose of 200 mg/m2. Survival outcomes were evaluated using Kaplan-Meier method, log-rank tests, Cox proportional hazard multivariable (MVA) analyses. Logistic MVA was performed to identify variables associated with low adherence to weekly cisplatin. Fine-Gray MVA was performed to analyze failure outcomes with death as a competing event. RESULTS: Among 119 patients who met our criteria, 51 patients (42.9%) had low adherence to weekly cisplatin. Median follow up was 19.8 months (interquartile range 8.8-65.6). Low adherence to weekly cisplatin was associated with worse overall survival (adjusted hazards ratio [aHR] 2.94, 95% confidence interval [CI] 1.58-5.47, p < 0.001) and progression-free survival (aHR 2.32, 95% CI 1.29-4.17, p = 0.005). It was also associated with worse distant failure (aHR 4.55, 95% CI 1.19-17.3, p = 0.03), but not locoregional failure (aHR 1.61, 95% CI 0.46-5.58, p = 0.46). KPS < 90 was the only variable associated with low adherence to weekly cisplatin (adjusted odds ratio [aOR] 2.67, 95% CI 1.10-6.65, p = 0.03). CONCLUSION: Our study suggested that over 40% of patients underwent fewer than 5 weekly cisplatin cycles and that low adherence to weekly cisplatin was an independent, adverse prognostic factor for worse survival and distant failure outcomes. Those with reduced adherence to weekly cisplatin were more likely to have poor performance status. Further studies are warranted to improve the adherence to chemotherapy and outcomes.


Sujet(s)
Chimioradiothérapie , Cisplatine , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Humains , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Études rétrospectives , Sujet âgé , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/mortalité , Tumeurs de la tête et du cou/anatomopathologie , Chimioradiothérapie/méthodes , Adhésion au traitement médicamenteux/statistiques et données numériques , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/administration et posologie , Résultat thérapeutique , Calendrier d'administration des médicaments , Adulte , Estimation de Kaplan-Meier
12.
Zhonghua Wei Chang Wai Ke Za Zhi ; 27(7): 740-748, 2024 Jul 25.
Article de Chinois | MEDLINE | ID: mdl-39004991

RÉSUMÉ

Peritoneal metastasis in gastric cancer is associated with rapid disease progression. Hyperthermic intraoperative peritoneal chemotherapy (HIPEC) done immediately after cytoreductive surgery (CRS) has become an important treatment for peritoneal metastasis in gastric cancer patients. However, different treatment options for HIPEC exist with potential influence on survival rates and prognosis in patients, exist. These treatment options include open or closed abdomen technique, perfusion solution, number of catheters, temperature, duration, and drug regimens. This paper aims to provide more evidence on standardization of HIPEC treatment options and technologies by systematically reviewing different drug regimens and technical approaches. The study included 2 randomized controlled trials, 3 phase I/II clinical trials, 2 prospective cohort studies, and 34 retrospective cohort studies, involving 1511 patients. The most common HIPEC option is to dissolve 50-75 mg/m2 of Cisplatin and 30-40 mg/m2 of Mitomycin C in 3-4 L saline solution at 42-43℃. After gastrointestinal anastomosis, 2-3 catheters are used in the HIPEC system with a perfusion flow rate of 500 ml/min. The duration is 60-90 minutes. Anastomotic leakage was low in studies where HIPEC was performed after gastrointestinal anastomosis. The utilization of open HIPEC and a two-drug regimen resulted in improved overall survival rates. The future development of HIPEC aims to enhance tumor-specific therapy by optimizing various aspects, such as identifying the safest and most effective chemotherapy regimens, refining patient selection criteria, and improving perioperative care.


Sujet(s)
Interventions chirurgicales de cytoréduction , Chimiothérapie hyperthermique intrapéritonéale , Tumeurs du péritoine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/thérapie , Interventions chirurgicales de cytoréduction/méthodes , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/traitement médicamenteux , Chimiothérapie hyperthermique intrapéritonéale/méthodes , Association thérapeutique , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Mitomycine/administration et posologie , Mitomycine/usage thérapeutique , Hyperthermie provoquée/méthodes
13.
Int J Biol Sci ; 20(9): 3570-3589, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993556

RÉSUMÉ

Background: Cisplatin (DDP) based combination chemotherapy is a vital method for the treatment of bladder cancer (BLca). Chemoresistance easily occurs in the course of cisplatin chemotherapy, which is one of the important reasons for the unfavorable prognosis of BLca patients. Circular RNAs (circRNAs) are widely recognized for their role in the development and advancement of BLca. Nevertheless, the precise role of circRNAs in DDP resistance for BLca remains unclear. Methods: To study the properties of circATIC, sanger sequencing, agarose gel electrophoresis and treatment with RNase R/Actinomycin D were utilized. RT-qPCR assay was utilized to assess the expression levels of circRNA, miRNA and mRNA in BLca tissues and cells. Functional experiments were conducted to assess the function of circATIC in BLca progression and chemosensitivity in vitro. Various techniques such as FISH, Dual-luciferase reporter assay, TRAP, RNA digestion assay, RIP and ChIRP assay were used to investigate the relationships between PTBP1, circATIC, miR-1247-5p and RCC2. Orthotopic bladder cancer model, xenograft subcutaneous tumor model and xenograft lung metastasis tumor model were performed to indicate the function and mechanism of circATIC in BLca progression and chemosensitivity in vivo. Results: In our study, we observed that circATIC expression was significantly enhanced in BLca tissues and cells and DDP resistant cells. Patients with higher circATIC expression have larger tumor diameter, higher incidence of postoperative metastasis and lower overall survival rate. Further experiments showed that circATIC accelerated BLca cell growth and metastasis and induced DDP resistance. Mechanistically, alternative splicing enzyme PTBP1 mediated the synthesis of circATIC. circATIC could enhance RCC2 mRNA stability via sponging miR-1247-5p or constructing a circATIC/LIN28A/RCC2 RNA-protein ternary complex. Finally, circATIC promotes RCC2 expression to enhance Epithelial-Mesenchymal Transition (EMT) progression and activate JNK signal pathway, thus strengthening DDP resistance in BLca cells. Conclusion: Our study demonstrated that circATIC promoted BLca progression and DDP resistance, and could serve as a potential target for BLca treatment.


Sujet(s)
Cisplatine , Résistance aux médicaments antinéoplasiques , Ribonucléoprotéines nucléaires hétérogènes , Protéine PTB , ARN circulaire , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Cisplatine/usage thérapeutique , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Protéine PTB/métabolisme , Protéine PTB/génétique , Animaux , Lignée cellulaire tumorale , Souris , Souris nude , microARN/métabolisme , microARN/génétique , Mâle , Femelle , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Souris de lignée BALB C , Prolifération cellulaire/effets des médicaments et des substances chimiques
14.
J Ovarian Res ; 17(1): 142, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987777

RÉSUMÉ

BACKGROUND: To identify key genes associated with cisplatin resistance in ovarian cancer, a comprehensive analysis was conducted on three datasets from the GEO database and through experimental validation. METHODS: Gene expression profiles were retrieved from the GEO database. DEGs were identified by comparing gene expression profiles between cisplatin-sensitive and resistant ovarian cancer cell lines. The identified genes were further subjected to GO, KEGG, and PPI network analysis. Potential inhibitors of key genes were identified through methods such as LibDock nuclear molecular docking. In vitro assays and RT-qPCR were performed to assess the expression levels of key genes in ovarian cancer cell lines. The sensitivity of cells to chemotherapy and proliferation of key gene knockout cells were evaluated through CCK8 and Clonogenic assays. RESULTS: Results showed that 12 genes influenced the chemosensitivity of the ovarian cancer cell line SKOV3, and 9 genes were associated with the prognosis and survival outcomes of ovarian cancer patients. RT-qPCR results revealed NDRG1, CYBRD1, MT2A, CNIH3, DPYSL3, and CARMIL1 were upregulated, whereas ERBB4, ANK3, B2M, LRRTM4, EYA4, and SLIT2 were downregulated in cisplatin-resistant cell lines. NDRG1, CYBRD1, and DPYSL3 knock-down significantly inhibited the proliferation of cisplatin-resistant cell line SKOV3. Finally, photofrin, a small-molecule compound targeting CYBRD1, was identified. CONCLUSION: This study reveals changes in the expression level of some genes associated with cisplatin-resistant ovarian cancer. In addition, a new small molecule compound was identified for the treatment of cisplatin-resistant ovarian cancer.


Sujet(s)
Antinéoplasiques , Cisplatine , Biologie informatique , Résistance aux médicaments antinéoplasiques , Tumeurs de l'ovaire , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Femelle , Humains , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Biologie informatique/méthodes , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes/méthodes , Cartes d'interactions protéiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
15.
Cancer Med ; 13(14): e7446, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39015047

RÉSUMÉ

AIM: The Japanese Interventional oncology group (JIVROSG) showed the efficacy and safety of nonselective transarterial chemoembolization (TACE) with fine cisplatin powder (diamminedichloroplatinum; DDP-H) (65 mg/m2) and porous gelatin particles (DDP-H TACE) without lipiodol for extensive multifocal hepatocellular carcinoma (HCC). However, there are no studies on this method following the JIVROSG study. Therefore, we aimed to evaluate the efficacy of this new DDP-H TACE and its effect on liver function. METHODS: We retrospectively reviewed the medical records of TACE-naïve patients with multifocal HCC (Child-Pugh class A, up-to-seven out, no prior history of systemic therapy) who underwent whole-liver DDP-H TACE between January 2006 and December 2019. RESULTS: Sixty patients were included in this study. The median age of the patients was 71 (range, 35-88) years. The median maximum size of tumors was 26 (range, 8-184) mm; 86.7% of patients met the up-to-11 criteria out. The overall survival duration was 30.3 months. At the time of initial evaluation (median, 45 days), the overall response rate was 65.0%; the disease control rate was 86.7% based on the modified response evaluation criteria in solid tumors guideline. Although nine patients' liver function had deteriorated to Child-Pugh class B at initial evaluation, six of them recovered to Child-Pugh class A. Only three patients (5%) showed permanently impaired liver function. CONCLUSIONS: Whole-liver DDP-H TACE without lipiodol or beads effectively reduced tumors and preserved liver function.


Sujet(s)
Carcinome hépatocellulaire , Chimioembolisation thérapeutique , Cisplatine , Gélatine , Tumeurs du foie , Humains , Chimioembolisation thérapeutique/méthodes , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/mortalité , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/mortalité , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Mâle , Sujet âgé , Adulte d'âge moyen , Femelle , Gélatine/administration et posologie , Études rétrospectives , Adulte , Sujet âgé de 80 ans ou plus , Poudres , Résultat thérapeutique , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Huile éthiodée/administration et posologie
16.
J Biochem Mol Toxicol ; 38(8): e23768, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39015062

RÉSUMÉ

Nephrotoxicity remains a major adverse reaction of the anticancer drug cisplatin (CDDP) chemotherapy, which is an important risk factor for chronic renal disease. Ginsenoside Rh2 from Panax ginseng has been shown to protect against CDDP-induced nephrotoxicity in vivo, but its pharmacological effect on renal tubular epithelial cells is not clearly understood. This study examined the molecular mechanisms underlying the nephroprotective effects of Rh2 on CDDP-induced HK-2 cells and acute kidney injury (AKI) mice. As a result of Rh2 treatment, CDDP-induced HK-2 cells showed increased cell viability and reduced lactate dehydrogenase release. Moreover, Rh2 ameliorated CDDP-induced mitochondrial membrane potential, increased antioxidant enzyme activities, and reduced pro-inflammatory cytokine expression to reduce damage. Rh2 inhibited apoptosis and enhanced the antioxidant capacity of HK-2 cells by reducing proteins associated with endoplasmic reticulum (ER) stress, as well as by attenuating tunicamycin-induced ER stress. In addition, treatment of CDDP-induced AKI mice with Rh2 substantially reduced blood urea nitrogen and serum creatinine levels, attenuated histological damage of kidney. Further, Rh2 also improved kidney function by inhibiting ER stress to support in vitro findings. These results consistently demonstrated that Rh2 protects renal tubular epithelial cells from CDDP-induced nephrotoxicity and apoptosis by restoring ER homeostasis, which might suggest a therapeutic potential and providing new insights into AKI alternative therapies.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Stress du réticulum endoplasmique , Cellules épithéliales , Ginsénosides , Tubules rénaux , Ginsénosides/pharmacologie , Cisplatine/effets indésirables , Cisplatine/toxicité , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Animaux , Souris , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Humains , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/prévention et contrôle , Atteinte rénale aigüe/traitement médicamenteux , Mâle , Lignée cellulaire , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée C57BL
17.
Ren Fail ; 46(2): 2378212, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39011587

RÉSUMÉ

PURPOSE: The present study investigated the nephron-testicular protective effects of sesamin against cisplatin (CP)-induced acute renal and testicular injuries. METHODS: Thirty-two male Wistar rats were allocated to receive carboxymethylcellulose (0.5%, as sesamin vehicle), CP (a single i.p. 5 mg/kg dose), CP plus sesamin at 10 or 20 mg/kg orally for 10 days. RESULTS: Data analysis showed significant increases in serum urea, creatinine, interleukin (IL)-1, IL-6, and tumor necrosis factor-α (TNF-α), as well as renal and testicular tissue malondialdehyde and nitric-oxide concentrations in CP-intoxicated rats in comparison to control animals. On the contrary, rats treated with CP only exhibited significantly lower (p < .05) serum testosterone, tissue glutathione, and activities of endogenous antioxidant enzymes compared to control rats. Histopathologically examining CP-intoxicated rats' tissues using H&E and PAS stains showed atrophied glomeruli, interstitial inflammatory cells, atypic tubular epithelium with focal apoptosis, and reduced mucopolysaccharide content. Further, immunohistochemical staining of the same group revealed an increase in p53 and cyclooxygenase-II (Cox-II) expression in renal and testicular tissues. Treatment with sesamin alleviated almost all the changes mentioned above in a dose-dependent manner, with the 20 mg/kg dose restoring several parameters' concentrations to normal ranges. CONCLUSIONS: In brief, sesamin could protect the kidneys and testes against CP toxicity through its antioxidant, anti-inflammatory, and anti-apoptotic effects.


Sujet(s)
Anti-inflammatoires , Antioxydants , Apoptose , Cisplatine , Dioxoles , Rein , Lignanes , Rat Wistar , Testicule , Animaux , Mâle , Lignanes/pharmacologie , Lignanes/usage thérapeutique , Cisplatine/toxicité , Cisplatine/effets indésirables , Rats , Dioxoles/pharmacologie , Antioxydants/pharmacologie , Testicule/effets des médicaments et des substances chimiques , Testicule/anatomopathologie , Testicule/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Anti-inflammatoires/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/prévention et contrôle , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/métabolisme , Antinéoplasiques/toxicité
18.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000202

RÉSUMÉ

The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) protein plays an essential role in the cisplatin (CDDP)-induced generation of reactive oxygen species (ROS). In this study, we evaluated the suitability of ultrasound-mediated lysozyme microbubble (USMB) cavitation to enhance NOX4 siRNA transfection in vitro and ex vivo. Lysozyme-shelled microbubbles (LyzMBs) were constructed and designed for siNOX4 loading as siNOX4/LyzMBs. We investigated different siNOX4-based cell transfection approaches, including naked siNOX4, LyzMB-mixed siNOX4, and siNOX4-loaded LyzMBs, and compared their silencing effects in CDDP-treated HEI-OC1 cells and mouse organ of Corti explants. Transfection efficiencies were evaluated by quantifying the cellular uptake of cyanine 3 (Cy3) fluorescein-labeled siRNA. In vitro experiments showed that the high transfection efficacy (48.18%) of siNOX4 to HEI-OC1 cells mediated by US and siNOX4-loaded LyzMBs significantly inhibited CDDP-induced ROS generation to almost the basal level. The ex vivo CDDP-treated organ of Corti explants of mice showed an even more robust silencing effect of the NOX4 gene in the siNOX4/LyzMB groups treated with US sonication than without US sonication, with a marked abolition of CDDP-induced ROS generation and cytotoxicity. Loading of siNOX4 on LyzMBs can stabilize siNOX4 and prevent its degradation, thereby enhancing the transfection and silencing effects when combined with US sonication. This USMB-derived therapy modality for alleviating CDDP-induced ototoxicity may be suitable for future clinical applications.


Sujet(s)
Cisplatine , Cellules ciliées auditives , Microbulles , Lysozyme , NADPH Oxidase 4 , Ototoxicité , Espèces réactives de l'oxygène , Cisplatine/pharmacologie , Animaux , NADPH Oxidase 4/génétique , NADPH Oxidase 4/métabolisme , Souris , Cellules ciliées auditives/effets des médicaments et des substances chimiques , Cellules ciliées auditives/métabolisme , Espèces réactives de l'oxygène/métabolisme , Ototoxicité/génétique , Lysozyme/génétique , Petit ARN interférent/génétique , Ondes ultrasonores , Techniques de knock-down de gènes , Lignée cellulaire
19.
PLoS One ; 19(7): e0307204, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008481

RÉSUMÉ

Malignant pleural mesothelioma (MPM) is an aggressive cancer with a very poor prognosis. Recently, immune checkpoint inhibition (ICI) has taken center stage in the currently ongoing revolution that is changing standard-of-care treatment for several malignancies, including MPM. As multiple arguments and accumulating lines of evidence are in support of the existence of a therapeutic synergism between chemotherapy and immunotherapy, as well as between different classes of immunotherapeutics, we designed a multicenter, single-arm, phase I/II trial in which both programmed-death-ligand 1 (PD-L1) inhibition and dendritic cell (DC) vaccination are integrated in the first-line conventional platinum/pemetrexed-based treatment scheme for epithelioid MPM patients (Immuno-MESODEC, ClinicalTrials.gov identifier NCT05765084). Fifteen treatment-naïve patients with unresectable epithelioid subtype MPM will be treated with four 3-weekly (±3 days) chemo-immunotherapy cycles. Standard-of-care chemotherapy consisting of cisplatinum (75mg/m2) and pemetrexed (500mg/m2) will be supplemented with the anti-PD-L1 antibody atezolizumab (1200 mg) and autologous Wilms' tumor 1 mRNA-electroporated dendritic cell (WT1/DC) vaccination (8-10 x 106 cells/vaccination). Additional atezolizumab (1680 mg) doses and/or WT1/DC vaccinations (8-10 x 106 cells/vaccination) can be administered optionally following completion of the chemo-immunotherapy scheme. Follow-up of patients will last for up to 90 days after final atezolizumab administration and/or WT1/DC vaccination or 24 months after diagnosis, whichever occurs later. The trial's primary endpoints are safety and feasibility, secondary endpoints are clinical efficacy and immunogenicity. This phase I/II trial will evaluate whether addition of atezolizumab and WT1/DC vaccination to frontline standard-of-care chemotherapy for the treatment of epithelioid MPM is feasible and safe. If so, this novel combination strategy should be further investigated as a promising advanced treatment option for this hard-to-treat cancer.


Sujet(s)
Anticorps monoclonaux humanisés , Antigène CD274 , Vaccins anticancéreux , Cellules dendritiques , Mésothéliome malin , Humains , Mésothéliome malin/traitement médicamenteux , Mésothéliome malin/immunologie , Cellules dendritiques/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Mâle , Femelle , Protéines WT1/immunologie , Tumeurs de la plèvre/immunologie , Tumeurs de la plèvre/traitement médicamenteux , Tumeurs de la plèvre/thérapie , Immunothérapie/méthodes , Adulte d'âge moyen , Adulte , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Sujet âgé , Vaccination , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pémétrexed/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Mésothéliome/traitement médicamenteux , Mésothéliome/immunologie , Mésothéliome/thérapie , Cisplatine/usage thérapeutique , Cisplatine/pharmacologie
20.
Molecules ; 29(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38930946

RÉSUMÉ

Cisplatin, a platinum-based chemotherapeutic, is effective against various solid tumors, but its use is often limited by its nephrotoxic effects. This study evaluated the protective effects of trametinib, an FDA-approved selective inhibitor of mitogen-activated protein kinase kinase 1/2 (MEK1/2), against cisplatin-induced acute kidney injury (AKI) in mice. The experimental design included four groups, control, trametinib, cisplatin, and a combination of cisplatin and trametinib, each consisting of eight mice. Cisplatin was administered intraperitoneally at a dose of 20 mg/kg to induce kidney injury, while trametinib was administered via oral gavage at 3 mg/kg daily for three days. Assessments were conducted 72 h after cisplatin administration. Our results demonstrate that trametinib significantly reduces the phosphorylation of MEK1/2 and extracellular signal-regulated kinase 1/2 (ERK1/2), mitigated renal dysfunction, and ameliorated histopathological abnormalities. Additionally, trametinib significantly decreased macrophage infiltration and the expression of pro-inflammatory cytokines in the kidneys. It also lowered lipid peroxidation by-products, restored the reduced glutathione/oxidized glutathione ratio, and downregulated NADPH oxidase 4. Furthermore, trametinib significantly inhibited both apoptosis and necroptosis in the kidneys. In conclusion, our data underscore the potential of trametinib as a therapeutic agent for cisplatin-induced AKI, highlighting its role in reducing inflammation, oxidative stress, and tubular cell death.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Modèles animaux de maladie humaine , Inflammation , Stress oxydatif , Pyridones , Pyrimidinones , Animaux , Cisplatine/effets indésirables , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Pyridones/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Pyrimidinones/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement , Inflammation/métabolisme , Mâle , Mort cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tubules rénaux/anatomopathologie , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...