Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.816
Filtrer
2.
Eur J Endocrinol ; 191(2): 251-261, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39158090

RÉSUMÉ

OBJECTIVE: Targeted therapy (TT) with BRAF/MEK inhibitors has emerged as a potential treatment in papillary craniopharyngiomas (PCPs). However, standardized data on large cohorts are lacking. Our study aimed to assess real-life efficacy and safety of BRAF/MEK inhibition in patients with PCPs. DESIGN: Retrospective French multicenter study involving BRAF V600E-mutated PCP patients, treated with BRAF/MEK inhibitor combination dabrafenib and trametinib, from April 2019 to July 2023. METHODS: Objective response and clinical and safety outcomes were assessed after 3 months and at the last available follow-up during TT. RESULTS: Sixteen patients (8 females, mean age 50.5 ± 15.75 years), receiving either neoadjuvant therapy (NEO) for non-resectable tumors (n = 6), post-surgical adjuvant therapy (ADJ; n = 8), or palliative therapy (PAL) following failure of multimodal treatment (n = 2), were included.At the last follow-up (mean 7.6 ± 5.3 months), 12 patients showed subtotal response, 3 exhibited partial response, and 1 maintained stable disease. Mean volume reduction was 88.9 ± 4.4%, 73.3 ± 23.4%, and 91.8 ± 4.3% in the NEO, ADJ, and PAL groups, respectively.Targeted therapy resolved headaches in 5/5 patients and visual impairment in 6/9; 2/3 patients had improved neurological symptoms, 1/4 presented weight loss, and 2/14 recovered endocrine function.Targeted therapy was well-tolerated in 62.5% of cases; adverse events led to treatment discontinuation in 5 patients and definitive discontinuation in 3 cases. CONCLUSIONS: In this study, 94% of patients showed partial response or better to TT. Adverse events were acceptable. Further research is needed to establish standardized protocols; however, these results advocate for a NEO approach in invasive PCPs.


Sujet(s)
Craniopharyngiome , Oximes , Tumeurs de l'hypophyse , Protéines proto-oncogènes B-raf , Pyridones , Pyrimidinones , Humains , Femelle , Mâle , Adulte d'âge moyen , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/génétique , Adulte , Études rétrospectives , Craniopharyngiome/traitement médicamenteux , Pyridones/usage thérapeutique , Pyridones/administration et posologie , Pyridones/effets indésirables , Sujet âgé , Tumeurs de l'hypophyse/traitement médicamenteux , Pyrimidinones/usage thérapeutique , Pyrimidinones/administration et posologie , Pyrimidinones/effets indésirables , Oximes/usage thérapeutique , Oximes/administration et posologie , Oximes/effets indésirables , Études de cohortes , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/effets indésirables , Thérapie moléculaire ciblée/méthodes , Imidazoles/usage thérapeutique , Imidazoles/administration et posologie , Imidazoles/effets indésirables , Résultat thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
3.
Cell Rep Med ; 5(8): 101663, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39094577

RÉSUMÉ

The current targeted therapy for BRAFV600E-mutant lung cancer consists of a dual blockade of RAF/MEK kinases often combining dabrafenib/trametinib (D/T). This regimen extends survival when compared to single-agent treatments, but disease progression is unavoidable. By using whole-genome CRISPR screening and RNA sequencing, we characterize the vulnerabilities of both persister and D/T-resistant cellular models. Oxidative stress together with concomitant induction of antioxidant responses is boosted by D/T treatment. However, the nature of the oxidative damage, the choice of redox detoxification systems, and the resulting therapeutic vulnerabilities display stage-specific differences. Persister cells suffer from lipid peroxidation and are sensitive to ferroptosis upon GPX4 inhibition in vivo. Biomarkers of lipid peroxidation are detected in clinical samples following D/T treatment. Acquired alterations leading to mitogen-activated protein kinase (MAPK) reactivation enhance cystine transport to boost GPX4-independent antioxidant responses. Similarly to BRAFV600E-mutant melanoma, histone deacetylase (HDAC) inhibitors decrease D/T-resistant cell viability and extend therapeutic response in vivo.


Sujet(s)
Adénocarcinome pulmonaire , Résistance aux médicaments antinéoplasiques , Inhibiteurs de désacétylase d'histone , Tumeurs du poumon , Phospholipid hydroperoxide glutathione peroxidase , Protéines proto-oncogènes B-raf , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Lignée cellulaire tumorale , Animaux , Inhibiteurs de désacétylase d'histone/pharmacologie , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Oximes/pharmacologie , Imidazoles/pharmacologie , Pyridones/pharmacologie , Pyrimidinones/pharmacologie , Peroxydation lipidique/effets des médicaments et des substances chimiques , Mutation/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Anal Methods ; 16(33): 5710-5722, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39136083

RÉSUMÉ

In the current study, nickel oxide nanoparticles (NiO NPs) modified with dimethylglyoxime (DMG) were deposited onto the cellulose surface (Ni(DMG)2-NiO-Cell) and used as an efficient adsorbent for thin film microextraction (TFME) of tramadol (TRA). The extracted TRA was determined using a high-performance liquid chromatography-ultraviolet detector (HPLC-UV). NiO NPs were synthesized by co-precipitation method on the surface of the cellulose substrate; afterward, its surface was modified by DMG to increase the extraction capability of the thin film toward TRA. The synthesized NiO-Cell and Ni(DMG)2-NiO-Cell thin films were characterized using various techniques. The effect of modification of the NiO thin film with DMG reagent on the extraction efficiency was investigated. The crucial parameters influencing the extraction efficiency, including extraction time, desorption time, desorption solvent, pH and salt content, were investigated via a one-at-a-time approach. The figures of merit for the developed method were evaluated in urine, plasma, and deionized water under the optimized extraction and desorption condition. The limits of detection and limits of quantification were in the range of 0.1 to 1 ng mL-1 and 0.3 to 3 ng mL-1, respectively, for the studied samples. The linear dynamic ranges of the developed TFME-HPLC-UV method were 0.3-1000, 1-2500, and 3-5000 ng mL-1 for the deionized water, urine, and plasma samples, respectively. The reproducibility and repeatability of the developed method was assayed in terms of intra-day, inter-day, and inter-thin film precisions by conducting six-replicate experiments at the concentration level of 0.1 and 1 µg mL-1, which were in the range of 5.9% to 8.3%. The sufficiency and applicability of the developed TFME-HPLC-UV method was investigated by determining TRA in urine and plasma samples, and the resulting relative recoveries (RR%) were 85.9% and 91.7%, respectively.


Sujet(s)
Cellulose , Nickel , Oximes , Tramadol , Chromatographie en phase liquide à haute performance/méthodes , Tramadol/urine , Tramadol/sang , Tramadol/analyse , Tramadol/composition chimique , Humains , Cellulose/composition chimique , Nickel/composition chimique , Nickel/sang , Nickel/urine , Oximes/composition chimique , Adsorption , Nanoparticules métalliques/composition chimique , Limite de détection , Microextraction en phase solide/méthodes , Reproductibilité des résultats
5.
Bull Exp Biol Med ; 177(3): 344-348, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39126546

RÉSUMÉ

The neuroprotective activity of tryptanthrin and its oxime was compared in male Wistar rats with a model of intraluminal occlusion of the middle cerebral artery. Neurobehavioral tests were performed 4, 24, and 48 h after focal cerebral infarction (FCI) using a modified neurological severity score (mNSS); additionally, the horizontal stability test, the plantar sensitivity test of the fore and hind limbs, holding on the tilted cage top test, and negative geotaxis test were performed. The size of FCI and the severity of brain tissue swelling were examined on day 2 after occlusion. Tryptanthrin and its oxime were administered at a dose of 10 mg/kg intraperitoneally during FCI, then daily for 2 days. In the control group, the mean score of neurological deficit remained at a high level for 2 days. FCI size was 43.8±3.4% of hemisphere area, and the hemisphere volume increased by 18.5±2.0% due to brain tissue swelling and edema. Administration of tryptanthrin and its oxime significantly decreased neurological deficits at all control points and reduced FCI size (by 24.2 and 30.4%, respectively) and brain tissue swelling of the affected hemisphere (by 64.9 and 62.7%, respectively). Therefore, the neuroprotective effect of tryptanthrine and its oxime in the acute period of FCI is largely determined by their anti-inflammatory activity.


Sujet(s)
Infarctus du territoire de l'artère cérébrale moyenne , Neuroprotecteurs , Oximes , Quinazolines , Rat Wistar , Animaux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Mâle , Rats , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Oximes/pharmacologie , Oximes/usage thérapeutique , Oedème cérébral/traitement médicamenteux , Oedème cérébral/anatomopathologie , Modèles animaux de maladie humaine , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie
6.
Neurosurg Rev ; 47(1): 458, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39172230

RÉSUMÉ

This study aimed to evaluate the effects of dabrafenib and/or trametinib therapy in BRAF v600-mutant glioma treatment. PubMed, the Cochrane Library, EMBASE and Web of Science were searched from inception to Sep 2023. Inclusion criteria were designed based on the PICO principle to select relevant articles. Search keywords included 'dabrafenib', 'trametinib', 'glioma' and other related keywords. Outcomes included overall survival (OS), progression-free survival (PFS), adverse events (AEs), and death events. Methodological index for non-randomized studies (MINORS) was used to assess the methodological quality. Stata 14.0 was selected to perform the Cochrane Q and I2 statistics to test the heterogeneity among all studies. As for publication bias assessment and sensitivity analysis, the funnel plot, Egger regression test, Begg test, and trim and fill method were selected. Including 8 studies for meta-analysis. The pooled results of the single-arm trials showed that the median PFS and median OS after treatment were 6.10 months and 22.73 months, respectively. Notably, this study found a high incidence of AEs and death events of 50% and 43% after treatment. All the above findings were statistically significant. Also, this study statistically supported the advantage of disease response improvement after the combination therapy in BRAF v600-mutant glioma patients, which were shown as a pooled rate of PR (30%), a pooled rate of CR (18%), and a pooled rate of ORR (39%). And the AE rate was lower in the monotherapy group (AE: 25%) than in the combination treatment group (AE: 60%). Sensitivity analysis indicated that all the results were robust. Based on current literature outcomes, dabrafenib and/or trametinib may lead to the median PFS of 6.10 months and median OS as 22.73 months for BRAF v600-mutant glioma patients, and the safety of monotherapy is better than that of combination therapy. This conclusion needs to be treated with caution and further verified.


Sujet(s)
Tumeurs du cerveau , Gliome , Imidazoles , Mutation , Oximes , Protéines proto-oncogènes B-raf , Pyridones , Pyrimidinones , Humains , Oximes/usage thérapeutique , Pyrimidinones/usage thérapeutique , Pyridones/usage thérapeutique , Imidazoles/usage thérapeutique , Gliome/traitement médicamenteux , Gliome/génétique , Protéines proto-oncogènes B-raf/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
7.
Anticancer Drugs ; 35(8): 761-763, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39115059

RÉSUMÉ

The combination of BRAF and MEK inhibitors demonstrated significant clinical benefit in patients with BRAF-mutant non-small cell lung cancer (NSCLC). However, the molecular mechanisms of acquired resistance to BRAF and MEK inhibition in NSCLC are still unknown. Herein, we report a case of a 76-year-old man with a history of smoking who was diagnosed with BRAF V600E-mutant lung adenocarcinoma (PD-L1 > 50%) and subsequently candidate to first-line therapy with pembrolizumab. After 18 months since the start of immunotherapy, computed tomography scan showed disease progression and a second-line therapy with dabrafenib and trametinib was initiated. Seven months later, due to a suspect disease progression, a left supraclavicular lymphadenectomy was performed and next-generation sequencing analysis revealed the appearance of MET exon 14 skipping mutation, while fluorescence in situ hybridization analysis showed MET amplification. The patient is still on BRAF and MEK inhibitor treatment. Our case highlights the relevance of performing tumor tissue rebiopsy at the time of progression during treatment with BRAF/MEK inhibition with the aim of identifying putative mechanisms of resistance.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Imidazoles , Tumeurs du poumon , Mutation , Oximes , Protéines proto-oncogènes B-raf , Protéines proto-oncogènes c-met , Pyridones , Pyrimidinones , Humains , Pyridones/administration et posologie , Pyrimidinones/administration et posologie , Mâle , Oximes/administration et posologie , Sujet âgé , Protéines proto-oncogènes B-raf/génétique , Imidazoles/administration et posologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines proto-oncogènes c-met/génétique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
8.
Chem Pharm Bull (Tokyo) ; 72(8): 731-746, 2024.
Article de Anglais | MEDLINE | ID: mdl-39085089

RÉSUMÉ

Nitrones are widely used as 1,3-dipoles in organic synthesis, but control of their reactions is not always easy. This review outlines our efforts to make the reactions of nitrones more predictable and easier to use. These efforts can be categorized into (1) 1,3-nucleophilic addition reaction of ketene silyl acetals to nitrones, (2) geometry-controlled cycloaddition of C-alkoxycarbonyl nitrones, (3) stereo-controlled cycloaddition using double asymmetric induction, and (4) generation of nitrones by N-selective modification of oximes.


Sujet(s)
Oxydes d'azote , Oxydes d'azote/composition chimique , Oxydes d'azote/synthèse chimique , Réaction de cycloaddition , Structure moléculaire , Acétals/composition chimique , Acétals/synthèse chimique , Cétones/composition chimique , Cétones/synthèse chimique , Oximes/composition chimique , Oximes/synthèse chimique , Éthylènes/composition chimique , Stéréoisomérie
9.
Int J Biol Macromol ; 275(Pt 2): 133741, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38986985

RÉSUMÉ

BACKGROUND: Sarcopenia, characterized by progressive muscle dysfunction, is a common complication of chronic obstructive pulmonary disease (COPD). Our previous study revealed serum Lipoprotein-associated phospholipaseA2 (Lp-PLA2) level significantly increased in COPD and associated with exercise tolerance. This study further investigated the functions and target potential of Lp-PLA2 for sarcopenia in COPD. METHODS: The circulating Lp-PLA2 level/enzyme activity in COPD patients and age-matched healthy volunteers were measured. Clinical parameters on skeletal muscle were measured and their correlations with Lp-PLA2 were analyzed. We explored the involvement of Lp-PLA2 in vivo and treatment effectiveness of darapladib (a specific Lp-PLA2 inhibitor) in CS-induced muscle dysfunction models. RESULTS: Circulating Lp-PLA2 level/enzyme activity was elevated in COPD patients compared with healthy controls, negatively associated with skeletal muscle mass and function. In CS-induced muscle dysfunction murine models, up-regulated serum Lp-PLA2 level/enzyme activity was verified again. In CS-exposed mouse models, darapladib treatment reversed muscle mass loss and muscle dysfunction, meanwhile rescued upregulation of MuRF1 and atrogin-1, and activation of inflammatory factors, oxidant enzymes and NF-κB signaling. CONCLUSIONS: Lp-PLA2 could be a potential indicator for sarcopenia in COPD. Darapladib, a Lp-PLA2 inhibitor, can alleviate CS-induced skeletal muscle dysfunction and represents a potential therapeutic for sarcopenia in COPD.


Sujet(s)
1-Alkyl-2-acetylglycerophosphocholine esterase , Broncho-pneumopathie chronique obstructive , Sarcopénie , Broncho-pneumopathie chronique obstructive/traitement médicamenteux , Broncho-pneumopathie chronique obstructive/complications , Humains , Sarcopénie/traitement médicamenteux , Sarcopénie/étiologie , 1-Alkyl-2-acetylglycerophosphocholine esterase/métabolisme , 1-Alkyl-2-acetylglycerophosphocholine esterase/sang , Mâle , Animaux , Souris , Femelle , Sujet âgé , Adulte d'âge moyen , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Benzaldéhydes/pharmacologie , Benzaldéhydes/usage thérapeutique , Oximes/usage thérapeutique , Oximes/pharmacologie , Marqueurs biologiques/sang , Modèles animaux de maladie humaine , Dérivés du biphényle , Diéthylamines , Pyrimidinones , Sulfures
10.
J Biochem Mol Toxicol ; 38(7): e23750, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38952032

RÉSUMÉ

The treatment of organophosphate (OP) anticholinesterases currently lacks an effective oxime reactivator of OP-inhibited acetylcholinesterase (AChE) which can penetrate the blood-brain barrier (BBB). Our laboratories have synthesized novel substituted phenoxyalkyl pyridinium oximes and tested them for their ability to promote survival of rats challenged with lethal doses of nerve agent surrogates. These previous studies demonstrated the ability of some of these oximes to promote 24-h survival to rats challenged with a lethal level of highly relevant surrogates for sarin and VX. The reactivation of OP-inhibited AChE in peripheral tissues was likely to be a major contributor to their efficacy in survival of lethal OP challenges. In the present study, twenty of these novel oximes were screened in vitro for reactivation ability for AChE in rat skeletal muscle and serum using two nerve agent surrogates: phthalimidyl isopropyl methylphosphonate (PIMP, a sarin surrogate) and 4-nitrophenyl ethyl methylphosphonate (NEMP, a VX surrogate). The oximes demonstrated a range of 23%-102% reactivation of AChE in vitro across both tissue types. Some of the novel oximes tested in the present study demonstrated the ability to more effectively reactivate AChE in serum than the currently approved oxime, 2-PAM. Therefore, some of these novel oximes have the potential to reverse AChE inhibition in peripheral target tissues and contribute to survival efficacy.


Sujet(s)
Acetylcholinesterase , Anticholinestérasiques , Réactivateurs de la cholinestérase , Muscles squelettiques , Organophosphates , Oximes , Animaux , Oximes/pharmacologie , Oximes/composition chimique , Rats , Acetylcholinesterase/métabolisme , Acetylcholinesterase/sang , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/enzymologie , Anticholinestérasiques/pharmacologie , Anticholinestérasiques/toxicité , Organophosphates/toxicité , Mâle , Réactivateurs de la cholinestérase/pharmacologie , Réactivateurs de la cholinestérase/composition chimique , Composés de pyridinium/pharmacologie , Rat Sprague-Dawley
11.
Chem Biol Interact ; 399: 111131, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-38964639

RÉSUMÉ

Dabrafenib is a BRAF inhibitor that has been demonstrated to be efficacious in the treatment of melanoma and non-small-cell lung cancer patients with BRAF V600E mutations. The objective of this study was to investigate the effects of 51 traditional Chinese medicines on the metabolism of dabrafenib and to further investigate the inhibitory effect of imperatorin. The quantification of dabrafenib and its metabolite hydroxy-dabrafenib was carried out using a sensitive, rapid, and accurate assay method based on ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). The results of in vitro experiments showed that 20 drugs inhibited the metabolism of dabrafenib by more than 80 %. In a further study of imperatorin on dabrafenib, the half-maximal inhibitory concentration (IC50) values of imperatorin on dabrafenib were 0.22 µM and 3.68 µM in rat liver microsomes (RLM) and human liver microsomes (HLM), respectively, while the inhibition mechanisms were non-competitive and mixed type inhibition, respectively. The results of in vivo experiments demonstrated that in the presence of imperatorin, the AUC(0-t), AUC(0-∞), Cmax, and Tmax of dabrafenib were increased by 2.38-, 2.26-, 1.05-, and 6.10-fold, respectively, while CLz/F was decreased by 67.9 %. In addition, Tmax of hydroxy-dabrafenib was increased by 1.4-fold. The results of the research showed that imperatorin had a consistent inhibitory effect on dabrafenib in vitro and in vivo. When the concurrent use of dabrafenib and imperatorin is unavoidable, clinicians should closely monitor for potential adverse events and make timely adjustments to the administered dosage.


Sujet(s)
Furocoumarines , Imidazoles , Microsomes du foie , Oximes , Rat Sprague-Dawley , Oximes/pharmacologie , Imidazoles/pharmacologie , Imidazoles/métabolisme , Animaux , Furocoumarines/pharmacologie , Furocoumarines/métabolisme , Microsomes du foie/métabolisme , Humains , Rats , Mâle , Spectrométrie de masse en tandem , Chromatographie en phase liquide à haute performance
12.
BMC Cancer ; 23(Suppl 1): 1250, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054462

RÉSUMÉ

BACKGROUND: The combination of the checkpoint inhibitor (CPI) pembrolizumab and platinum-based chemotherapy is effective frontline therapy for advanced non-small cell lung cancer (NSCLC) lacking targetable mutations. Indoleamine 2,3- dioxygenase 1 (IDO1), an enzyme involved in kynurenine production, inhibits immune responses. Inhibition of IDO1 may restore antitumor immunity and augment CPI activity. This trial evaluated addition of epacadostat, a potent and highly selective IDO1 inhibitor, to pembrolizumab and chemotherapy for metastatic NSCLC. METHODS: ECHO-306/KEYNOTE-715 was a partial double-blind, randomized phase II study of adults with treatment-naïve stage IV NSCLC not indicated for EGFR-, ALK-, or ROS1-directed therapy. Patients were randomized to one of three treatment arms: epacadostat-pembrolizumab-chemotherapy (E + P + C; blinded), epacadostat-pembrolizumab (E + P; open-label) or placebo-pembrolizumab-chemotherapy (PBO + P + C; blinded). Stratification was by PD-L1 tumor proportion score (< 50% vs. ≥ 50%) and tumor histology (non-squamous vs. squamous). A protocol amendment closed enrollment in the open-label E + P group, excluding it from efficacy analyses. Intravenous pembrolizumab (200 mg) was administered every 21 days and epacadostat 100 mg or matching placebo (oral) twice daily (BID) for ≤ 35 3-week cycles. The primary objective was objective response rate (ORR) for E + P + C vs. PBO + P + C. RESULTS: 178 patients were randomized to E + P + C (n = 91) or PBO + P + C (n = 87); 55 were enrolled in the E + P group. The E + P + C group had a lower confirmed ORR (26.4%; 95% CI 17.7-36.7) than the PBO + P + C group (44.8%; 95% CI 34.1-55.9), with a difference of - 18.5% (95% CI - 32.0 - (- 4.3); one-sided P = 0.9948). The E + P + C group had a numerically higher percentage of confirmed responders with extended response ≥ 6 months (29.2% vs. 15.4%). Circulating kynurenine levels at C1D1 were similar to those at C2D1 in all treatment groups and were not reduced to normal levels with epacadostat 100 mg BID plus P + C. The safety profile of E + P + C was consistent with that for PBO + P + C. CONCLUSIONS: Addition of epacadostat 100 mg BID to pembrolizumab and platinum-based chemotherapy was generally well tolerated but did not improve ORR in patients with treatment-naïve metastatic NSCLC. Evaluating epacadostat doses that normalize circulating kynurenine in combination with CPIs may help determine the clinical potential of this combination. TRIAL REGISTRATION: NCT03322566. Registered October 26, 2017.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Méthode en double aveugle , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Sujet âgé , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Adulte , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Oximes
13.
BMC Cancer ; 23(Suppl 1): 1254, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054467

RÉSUMÉ

BACKGROUND: Advanced head and neck squamous cell carcinoma (HNSCC) has a poor prognosis, and new treatment options are needed. Combining immunotherapies with differing mechanisms of action may enhance clinical benefits compared with single-agent immunotherapy. Epacadostat, an indoleamine 2,3 dioxygenase 1 inhibitor, plus pembrolizumab, a PD-1 inhibitor, showed promising activity in advanced HNSCC in the phase 1/2 KEYNOTE-037/ECHO-202 trial. METHODS: KEYNOTE-669/ECHO-304 is a randomized, open-label, phase 3 study evaluating the efficacy and safety of pembrolizumab plus epacadostat, pembrolizumab monotherapy, and the EXTREME regimen (cetuximab with a platinum [carboplatin or cisplatin] and 5-fluorouracil) in recurrent/metastatic (R/M) HNSCC. Participants had no prior systemic therapy for R/M HNSCC and were randomly assigned (2:1:2) to pembrolizumab 200 mg intravenously every 3 weeks plus epacadostat 100 mg orally twice daily, pembrolizumab monotherapy, or EXTREME. The primary endpoint was objective response rate (ORR; investigator assessment). Secondary endpoints were safety and tolerability. Change in serum kynurenine was an exploratory endpoint. Study enrollment was discontinued early as a strategic decision on May 2, 2018, and response assessment was discontinued after first on-study imaging assessment at week 9. Data cut-off was January 17, 2019. RESULTS: Between December 1, 2017, and May 2, 2018, 89 patients were randomly allocated to pembrolizumab plus epacadostat (n = 35), pembrolizumab monotherapy (n = 19), or EXTREME (n = 35). ORR (95% CI) was 31% (17%-49%) for pembrolizumab plus epacadostat, 21% (6%-46%) for pembrolizumab monotherapy, and 34% (19%-52%) for EXTREME. Treatment-related adverse events (TRAEs) occurred in 82% (n = 28) of patients receiving pembrolizumab plus epacadostat, 63% (n = 12) receiving pembrolizumab monotherapy, and 100% (n = 34) receiving EXTREME. Grade 3-4 TRAEs occurred in 24% (n = 8) of patients receiving pembrolizumab plus epacadostat, 16% (n = 3) receiving pembrolizumab monotherapy, and 82% (n = 28) receiving EXTREME. No deaths occurred due to AEs. Pembrolizumab plus epacadostat treatment reduced kynurenine levels but not to that of healthy subjects. CONCLUSIONS: Pembrolizumab plus epacadostat and pembrolizumab monotherapy provided a similar response rate to EXTREME and demonstrated a manageable safety profile in patients with R/M HNSCC. TRIAL REGISTRATION: NCT03358472. Date of trial registration: November 30, 2017.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Tumeurs de la tête et du cou , Récidive tumorale locale , Carcinome épidermoïde de la tête et du cou , Humains , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Mâle , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Femelle , Adulte d'âge moyen , Récidive tumorale locale/traitement médicamenteux , Sujet âgé , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Adulte , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Sulfonamides/effets indésirables , Oximes
14.
BMC Cancer ; 23(Suppl 1): 1256, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054485

RÉSUMÉ

BACKGROUND: Indoleamine 2,3-dioxygenase 1 (IDO1) levels correlate with poor outcomes in urothelial carcinoma (UC). IDO1 and programmed death-ligand 1 (PD-L1) are often co-expressed. Epacadostat is a potent and highly selective inhibitor of IDO1. In a subgroup analysis of patients with advanced UC participating in a phase I/II study, epacadostat-pembrolizumab treatment produced an objective response rate (ORR) of 35%. METHODS: ECHO-303/KEYNOTE-698 was a double-blinded, randomized phase III study of adults with metastatic or unresectable locally advanced UC with recurrence or progression following first-line platinum-based chemotherapy. Participants were randomized to epacadostat 100 mg twice daily (BID) plus pembrolizumab or placebo plus pembrolizumab until completion of 35 pembrolizumab infusions, disease progression, or unacceptable toxicity. The primary endpoint was investigator-assessed ORR per Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS: Target enrollment was 648 patients; enrollment was halted early based on efficacy results from the phase III ECHO-301/KEYNOTE-252 study in metastatic melanoma. Forty-two patients were randomized to each treatment arm. Median duration of follow-up was 62 days in each arm. The investigator-assessed ORR (unconfirmed) was 26.2% (95% CI 16.35-48.11) for epacadostat plus pembrolizumab and 11.9% (95% CI 4.67-29.50) for placebo plus pembrolizumab. Two complete responses were reported, both in the placebo-plus-pembrolizumab arm. Circulating kynurenine levels increased from C1D1 to C2D1 in the placebo-plus-pembrolizumab arm and numerically decreased in the epacadostat-plus-pembrolizumab arm. The safety profile of epacadostat plus pembrolizumab was similar to that of pembrolizumab monotherapy, although a numerically greater proportion of patients in the combination vs. control arm experienced treatment-related grade ≥ 3 adverse events (16.7% vs. 7.3%). One patient in each arm died due to cardiovascular events, which were not deemed drug-related. No new safety concerns were identified for either agent. CONCLUSIONS: Epacadostat plus pembrolizumab demonstrated anti-tumor activity and was generally tolerable as second-line treatment of patients with unresectable locally advanced or recurrent/progressive metastatic UC. Epacadostat 100 mg BID, when administered with pembrolizumab, did not normalize circulating kynurenine in most patients. Further study of combined IDO1/PD-L1 inhibition in this patient population, particularly with epacadostat doses that result in durable normalization of circulating kynurenine, may be warranted. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03374488. Registered 12/15/2017.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Sulfonamides , Humains , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/usage thérapeutique , Mâle , Femelle , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Sujet âgé , Adulte d'âge moyen , Méthode en double aveugle , Sulfonamides/administration et posologie , Sulfonamides/usage thérapeutique , Oximes/administration et posologie , Oximes/usage thérapeutique , Carcinome transitionnel/traitement médicamenteux , Carcinome transitionnel/anatomopathologie , Sujet âgé de 80 ans ou plus , Adulte , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Tumeurs urologiques/traitement médicamenteux , Tumeurs urologiques/anatomopathologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie
16.
BMC Cancer ; 23(Suppl 1): 1251, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054476

RÉSUMÉ

BACKGROUND: Pembrolizumab is a first-line therapy for certain patients with advanced/metastatic non-small cell lung cancer (NSCLC). Combining pembrolizumab with other immunotherapies may enhance tumor cell killing and clinical outcomes. Epacadostat is a selective inhibitor of indoleamine 2,3-dioxygenase 1, an immuno-regulatory enzyme involved in tryptophan to kynurenine metabolism that inhibits T cell-mediated immune responses. METHODS: In this randomized phase II study, patients with metastatic NSCLC expressing high (≥ 50%) programmed death-ligand 1 (PD-L1) levels received pembrolizumab 200 mg every 21 days plus oral epacadostat 100 mg twice daily (combination) or matching placebo (control). The primary objective was objective response rate (ORR); secondary objectives were progression-free survival (PFS), overall survival (OS), duration of response (DOR) and safety/tolerability. RESULTS: 154 patients were randomized (77 per group). Median (range) follow-up was 6.8 months (0.1-11.4) and 7.0 months (0.2-11.9) in the combination and control groups, respectively Confirmed ORR was similar between groups (combination: 32.5%, 95% CI 22.2-44.1; control: 39.0%, 95% CI 28.0-50.8; difference: - 6.5, 95% CI - 21.5 to 8.7; 1-sided P = 0.8000). Median (range) DOR was 6.2 months (1.9 + to 6.5 +) and not reached (1.9 + to 8.6 +) in the combination and control groups, respectively. Although not formally tested, median PFS was 6.7 and 6.2 months for the combination and control groups, respectively, and median OS was not reached in either group. Circulating kynurenine levels increased from C1D1 to C2D1 (P < 0.01) in the control group and decreased from C1D1 to C2D1 (P < 0.01) in the combination group but were not normalized in most patients. The most frequent serious adverse events (AEs) (≥ 2%) were pneumonia (4.0%), anemia (2.7%), atelectasis (2.7%) and pneumonitis (2.7%) in the combination group and pneumonia (3.9%), pneumonitis (2.6%) and hypotension (2.6%) in the control group. Two deaths due to drug-related AEs were reported, both in the control group. CONCLUSIONS: Addition of epacadostat to pembrolizumab therapy for PD-L1-high metastatic NSCLC was generally well tolerated but did not demonstrate an improved therapeutic effect. Evaluating higher doses of epacadostat that normalize kynurenine levels when given in combination with checkpoint inhibitors may be warranted. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03322540. Registered 10/26/2017.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Antigène CD274 , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Sulfonamides , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/mortalité , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Méthode en double aveugle , Antigène CD274/antagonistes et inhibiteurs , Sulfonamides/administration et posologie , Sulfonamides/usage thérapeutique , Sulfonamides/effets indésirables , Adulte , Oximes/administration et posologie , Oximes/usage thérapeutique , Oximes/effets indésirables , Sujet âgé de 80 ans ou plus , Survie sans progression
17.
BMC Cancer ; 23(Suppl 1): 1252, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054491

RÉSUMÉ

BACKGROUND: Indoleamine 2,3- dioxygenase 1 (IDO1) is an immunosuppressive enzyme that has been correlated with shorter disease-specific survival in patients with urothelial carcinoma (UC). IDO1 may counteract the antitumor effects of immune checkpoint inhibitors. Epacadostat is a potent and highly selective inhibitor of IDO1. In the phase I/II ECHO-202/KEYNOTE-037 study, epacadostat plus pembrolizumab resulted in a preliminary objective response rate (ORR) of 35% in a cohort of patients with advanced UC. METHODS: ECHO-307/KEYNOTE-672 was a double-blinded, randomized, phase III study. Eligible adults had confirmed locally advanced/unresectable or metastatic UC of the urinary tract and were ineligible to receive cisplatin-based chemotherapy. Participants were randomly assigned (1:1) to receive epacadostat (100 mg twice daily) plus pembrolizumab (200 mg every 3 weeks) or placebo plus pembrolizumab for up to 35 pembrolizumab infusions. The primary endpoint was investigator-assessed ORR per Response Evaluation Criteria in Solid Tumors (version 1.1). RESULTS: A total of 93 patients were randomized (epacadostat plus pembrolizumab, n = 44; placebo plus pembrolizumab, n = 49). Enrollment was stopped early due to emerging data from the phase III ECHO-301/KEYNOTE-252 study. The median duration of follow-up was 64 days in both arms. Based on all available data at cutoff, ORR (unconfirmed) was 31.8% (95% CI, 22.46-55.24%) for epacadostat plus pembrolizumab and 24.5% (95% CI, 15.33-43.67%) for placebo plus pembrolizumab. Circulating kynurenine levels numerically increased from C1D1 to C2D1 in the placebo-plus-pembrolizumab arm and decreased in the epacadostat-plus-pembrolizumab arm. Epacadostat-plus-pembrolizumab combination treatment was well tolerated with a safety profile similar to the placebo arm. Treatment discontinuations due to treatment-related adverse events were more frequent with epacadostat (11.6% vs. 4.1%). CONCLUSIONS: Treatment with epacadostat plus pembrolizumab resulted in a similar ORR and safety profile as placebo plus pembrolizumab in cisplatin-ineligible patients with previously untreated locally advanced/unresectable or metastatic UC. At a dose of 100 mg twice daily, epacadostat did not appear to completely normalize circulating kynurenine levels when administered with pembrolizumab. Larger studies with longer follow-up and possibly testing higher doses of epacadostat, potentially in different therapy settings, may be warranted. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03361865, retrospectively registered December 5, 2017.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Cisplatine , Sulfonamides , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/effets indésirables , Mâle , Femelle , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Sujet âgé , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Sulfonamides/effets indésirables , Cisplatine/usage thérapeutique , Cisplatine/administration et posologie , Cisplatine/effets indésirables , Méthode en double aveugle , Adulte d'âge moyen , Tumeurs urologiques/traitement médicamenteux , Tumeurs urologiques/anatomopathologie , Sujet âgé de 80 ans ou plus , Carcinome transitionnel/traitement médicamenteux , Carcinome transitionnel/anatomopathologie , Adulte , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Oximes
18.
Medicina (Kaunas) ; 60(7)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39064466

RÉSUMÉ

A 76-year-old female patient presented with an iodine-refractory papillary thyroid carcinoma (PTC), diagnosed eight years earlier, with several lymph node recurrences requiring successive surgeries. Fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) imaging revealed a new unresectable loco-regional recurrence. The patient was diagnosed with a somatic BRAF V600E mutation. Therefore, dabrafenib and trametinib combination therapy was introduced and closely monitored by a dedicated multidisciplinary team, involving pharmaceutical consultations. As early as six weeks after treatment initiation, the patient reported multiple adverse events (AEs) to the clinical pharmacy team, who provided advice on resolving AEs or improving tolerance. Close interprofessional collaboration among healthcare workers involved in the care pathway allowed for the identification of the most opportune times for temporary suspension of treatment (four suspensions over seven months) or dose reduction (two reductions over 3.5 months). This resulted in a total treatment duration (one year) longer than the average times reported in the literature. The patient showed a rapid and excellent response to treatment immediately after initiation, culminating in a complete metabolic response assessed by [18F]FDG PET/CT imaging at nine months. Twenty-five months after treatment discontinuation, the disease remained controlled. Overall, dabrafenib and trametinib combination could offer excellent outcomes in selected patients with refractory BRAF-mutated PTC, with additional clinical pharmacy initiatives allowing for the optimized management of AEs and prolonged treatment periods.


Sujet(s)
Imidazoles , Oximes , Pyridones , Pyrimidinones , Tumeurs de la thyroïde , Humains , Femelle , Oximes/usage thérapeutique , Oximes/administration et posologie , Imidazoles/usage thérapeutique , Imidazoles/administration et posologie , Pyridones/usage thérapeutique , Pyridones/administration et posologie , Sujet âgé , Pyrimidinones/usage thérapeutique , Pyrimidinones/administration et posologie , Tumeurs de la thyroïde/traitement médicamenteux , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Radio-isotopes de l'iode/usage thérapeutique , Radio-isotopes de l'iode/administration et posologie , Cancer papillaire de la thyroïde/traitement médicamenteux , Résultat thérapeutique , Protéines proto-oncogènes B-raf/génétique
19.
Int Immunopharmacol ; 138: 112653, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38996664

RÉSUMÉ

As one of the main pathmechanisms of Alzheimer's disease (AD), amyloid-ß (Aß) is widely considered to be the prime target for the development of AD therapy. Recently, imidazolylacetophenone oxime ethers or esters (IOEs) have shown neuroprotective effects against neuronal cells damage, suggesting their potential use in the prevention and treatment of AD. Thirty IOEs compounds from our lab in-house library were constructed and screened for the inhibitory effects on Aß42-induced cytotoxicity. Among them, TJ1, as a new IOEs hit, preliminarily showed the effect on inhibiting Aß42-induced cytotoxicity. Furthermore, the inhibitory effects of TJ1 on Aß42 aggregation were tested by ThT assays and TEM. The neuroprotective effects of TJ1 were evaluated in Aß42-stimulated SH-SY5Y cells, LPS-stimulated BV-2 cells, and H2O2- and RSL3-stimulated PC12 cells. The cognitive improvement of TJ1 was assessed in 5xFAD (C57BL/6J) transgenic mouse. These results showed that TJ1 had strong neuroprotective effects and high blood-brain barrier (BBB) permeability without obvious cytotoxicity. TJ1 impeded the self-accumulation process of Aß42 by acting on Aß oligomerization and fibrilization. Besides, TJ1 reversed Aß-, H2O2- and RSL3-induced neuronal cell damage and decreased neuroinflammation. In 5xFAD mice, TJ1 improved cognitive impairment, increased GSH level, reduced the level of Aß42 and Aß plaques, and attenuated the glia reactivation and inflammatory response in the brain,. Taken together, our results demonstrate that TJ1 improves cognitive impairments as a new neuroprotective candidate via targeting amyloidogenesis, which suggests the potential of TJ1 as a treatment for AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Barrière hémato-encéphalique , Modèles animaux de maladie humaine , Souris de lignée C57BL , Souris transgéniques , Neuroprotecteurs , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Humains , Souris , Rats , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Fragments peptidiques/métabolisme , Cellules PC12 , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Oximes/pharmacologie , Oximes/usage thérapeutique , Lignée cellulaire tumorale , Mâle
20.
FEBS Lett ; 598(16): 2011-2027, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38977937

RÉSUMÉ

Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn't impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Imidazoles , Mélanome , Mitogen-Activated Protein Kinase 7 , Oximes , Protéines proto-oncogènes B-raf , Vémurafénib , Oximes/pharmacologie , Humains , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/métabolisme , Imidazoles/pharmacologie , Vémurafénib/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mitogen-Activated Protein Kinase 7/métabolisme , Mitogen-Activated Protein Kinase 7/génétique , Indoles/pharmacologie , Sulfonamides/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE