Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Clin Oncol ; 42(26): 3105-3114, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39028931

RÉSUMÉ

PURPOSE: To assess the safety and efficacy of the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor osimertinib as neoadjuvant therapy in patients with surgically resectable stage I-IIIA EGFR-mutated non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: This was a multi-institutional phase II trial of neoadjuvant osimertinib for patients with surgically resectable stage I-IIIA (American Joint Committee on Cancer [AJCC] V7) EGFR-mutated (L858R or exon 19 deletion) NSCLC (ClinicalTrials.gov identifier: NCT03433469). Patients received osimertinib 80 mg orally once daily for up to two 28-day cycles before surgical resection. The primary end point was major pathological response (MPR) rate. Secondary safety and efficacy end points were also assessed. Exploratory end points included pretreatment and post-treatment tumor mutation profiling. RESULTS: A total of 27 patients were enrolled and treated with neoadjuvant osimertinib for a median 56 days before surgical resection. Twenty-four (89%) patients underwent subsequent surgery; three (11%) patients were converted to definitive chemoradiotherapy. The MPR rate was 14.8% (95% CI, 4.2 to 33.7). No pathological complete responses were observed. The ORR was 52%, and the median DFS was 40.9 months. One treatment-related serious adverse event (AE) occurred (3.7%). No patients were unable to undergo surgical resection or had surgery delayed because of an AE. The most common co-occurring tumor genomic alterations were in TP53 (42%) and RBM10 (21%). CONCLUSION: Treatment with neoadjuvant osimertinib in surgically resectable (stage IA-IIIA, AJCC V7) EGFR-mutated NSCLC did not meet its primary end point for MPR rate. However, neoadjuvant osimertinib did not lead to unanticipated AEs, surgical delays, nor result in a significant unresectability rate.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Traitement néoadjuvant , Humains , Acrylamides/usage thérapeutique , Femelle , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Dérivés de l'aniline/usage thérapeutique , Dérivés de l'aniline/effets indésirables , Mâle , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Récepteurs ErbB/génétique , Sujet âgé , Stadification tumorale , Adulte , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/administration et posologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Antinéoplasiques/administration et posologie , Indoles , Pyrimidines
2.
Nat Commun ; 15(1): 4871, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871738

RÉSUMÉ

The phenomenon of mixed/heterogenous treatment responses to cancer therapies within an individual patient presents a challenging clinical scenario. Furthermore, the molecular basis of mixed intra-patient tumor responses remains unclear. Here, we show that patients with metastatic lung adenocarcinoma harbouring co-mutations of EGFR and TP53, are more likely to have mixed intra-patient tumor responses to EGFR tyrosine kinase inhibition (TKI), compared to those with an EGFR mutation alone. The combined presence of whole genome doubling (WGD) and TP53 co-mutations leads to increased genome instability and genomic copy number aberrations in genes implicated in EGFR TKI resistance. Using mouse models and an in vitro isogenic p53-mutant model system, we provide evidence that WGD provides diverse routes to drug resistance by increasing the probability of acquiring copy-number gains or losses relative to non-WGD cells. These data provide a molecular basis for mixed tumor responses to targeted therapy, within an individual patient, with implications for therapeutic strategies.


Sujet(s)
Instabilité des chromosomes , Récepteurs ErbB , Tumeurs du poumon , Mutation , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Souris , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/anatomopathologie , Thérapie moléculaire ciblée/méthodes , Femelle , Variations de nombre de copies de segment d'ADN , Mâle
3.
Nat Commun ; 15(1): 3741, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702301

RÉSUMÉ

Targeted therapy is effective in many tumor types including lung cancer, the leading cause of cancer mortality. Paradigm defining examples are targeted therapies directed against non-small cell lung cancer (NSCLC) subtypes with oncogenic alterations in EGFR, ALK and KRAS. The success of targeted therapy is limited by drug-tolerant persister cells (DTPs) which withstand and adapt to treatment and comprise the residual disease state that is typical during treatment with clinical targeted therapies. Here, we integrate studies in patient-derived and immunocompetent lung cancer models and clinical specimens obtained from patients on targeted therapy to uncover a focal adhesion kinase (FAK)-YAP signaling axis that promotes residual disease during oncogenic EGFR-, ALK-, and KRAS-targeted therapies. FAK-YAP signaling inhibition combined with the primary targeted therapy suppressed residual drug-tolerant cells and enhanced tumor responses. This study unveils a FAK-YAP signaling module that promotes residual disease in lung cancer and mechanism-based therapeutic strategies to improve tumor response.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Transduction du signal , Facteurs de transcription , Protéines de signalisation YAP , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines de signalisation YAP/métabolisme , Lignée cellulaire tumorale , Animaux , Résistance aux médicaments antinéoplasiques/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Maladie résiduelle , Souris , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Kinase du lymphome anaplasique/métabolisme , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Focal adhesion protein-tyrosine kinases/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Nat Cancer ; 5(6): 938-952, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38637658

RÉSUMÉ

Tailoring optimal treatment for individual cancer patients remains a significant challenge. To address this issue, we developed PERCEPTION (PERsonalized Single-Cell Expression-Based Planning for Treatments In ONcology), a precision oncology computational pipeline. Our approach uses publicly available matched bulk and single-cell (sc) expression profiles from large-scale cell-line drug screens. These profiles help build treatment response models based on patients' sc-tumor transcriptomics. PERCEPTION demonstrates success in predicting responses to targeted therapies in cultured and patient-tumor-derived primary cells, as well as in two clinical trials for multiple myeloma and breast cancer. It also captures the resistance development in patients with lung cancer treated with tyrosine kinase inhibitors. PERCEPTION outperforms published state-of-the-art sc-based and bulk-based predictors in all clinical cohorts. PERCEPTION is accessible at https://github.com/ruppinlab/PERCEPTION . Our work, showcasing patient stratification using sc-expression profiles of their tumors, will encourage the adoption of sc-omics profiling in clinical settings, enhancing precision oncology tools based on sc-omics.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Médecine de précision , Analyse sur cellule unique , Transcriptome , Humains , Analyse sur cellule unique/méthodes , Médecine de précision/méthodes , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs/génétique , Tumeurs/traitement médicamenteux , Analyse de profil d'expression de gènes/méthodes , Femelle , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Biologie informatique/méthodes
5.
Nat Genet ; 56(1): 60-73, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38049664

RÉSUMÉ

In this study, the impact of the apolipoprotein B mRNA-editing catalytic subunit-like (APOBEC) enzyme APOBEC3B (A3B) on epidermal growth factor receptor (EGFR)-driven lung cancer was assessed. A3B expression in EGFR mutant (EGFRmut) non-small-cell lung cancer (NSCLC) mouse models constrained tumorigenesis, while A3B expression in tumors treated with EGFR-targeted cancer therapy was associated with treatment resistance. Analyses of human NSCLC models treated with EGFR-targeted therapy showed upregulation of A3B and revealed therapy-induced activation of nuclear factor kappa B (NF-κB) as an inducer of A3B expression. Significantly reduced viability was observed with A3B deficiency, and A3B was required for the enrichment of APOBEC mutation signatures, in targeted therapy-treated human NSCLC preclinical models. Upregulation of A3B was confirmed in patients with NSCLC treated with EGFR-targeted therapy. This study uncovers the multifaceted roles of A3B in NSCLC and identifies A3B as a potential target for more durable responses to targeted cancer therapy.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Animaux , Souris , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Mutation , Régulation positive/génétique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Cytidine deaminase/génétique , Antigènes mineurs d'histocompatibilité/génétique , Antigènes mineurs d'histocompatibilité/métabolisme
6.
Cancer Discov ; 12(11): 2666-2683, 2022 11 02.
Article de Anglais | MEDLINE | ID: mdl-35895872

RÉSUMÉ

Anticancer therapies have been limited by the emergence of mutations and other adaptations. In bacteria, antibiotics activate the SOS response, which mobilizes error-prone factors that allow for continuous replication at the cost of mutagenesis. We investigated whether the treatment of lung cancer with EGFR inhibitors (EGFRi) similarly engages hypermutators. In cycling drug-tolerant persister (DTP) cells and in EGFRi-treated patients presenting residual disease, we observed upregulation of GAS6, whereas ablation of GAS6's receptor, AXL, eradicated resistance. Reciprocally, AXL overexpression enhanced DTP survival and accelerated the emergence of T790M, an EGFR mutation typical to resistant cells. Mechanistically, AXL induces low-fidelity DNA polymerases and activates their organizer, RAD18, by promoting neddylation. Metabolomics uncovered another hypermutator, AXL-driven activation of MYC, and increased purine synthesis that is unbalanced by pyrimidines. Aligning anti-AXL combination treatments with the transition from DTPs to resistant cells cured patient-derived xenografts. Hence, similar to bacteria, tumors tolerate therapy by engaging pharmacologically targetable endogenous mutators. SIGNIFICANCE: EGFR-mutant lung cancers treated with kinase inhibitors often evolve resistance due to secondary mutations. We report that in similarity to the bacterial SOS response stimulated by antibiotics, endogenous mutators are activated in drug-treated cells, and this heralds tolerance. Blocking the process prevented resistance in xenograft models, which offers new treatment strategies. This article is highlighted in the In This Issue feature, p. 2483.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Humains , Lignée cellulaire tumorale , Réplication de l'ADN , Protéines de liaison à l'ADN/génétique , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/génétique , Tumeurs du poumon/génétique , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/génétique , Ubiquitin-protein ligases/génétique , Animaux , Axl Receptor Tyrosine Kinase
7.
J Clin Invest ; 132(13)2022 07 01.
Article de Anglais | MEDLINE | ID: mdl-35579943

RÉSUMÉ

Molecularly targeted cancer therapy has improved outcomes for patients with cancer with targetable oncoproteins, such as mutant EGFR in lung cancer. Yet, the long-term survival of these patients remains limited, because treatment responses are typically incomplete. One potential explanation for the lack of complete and durable responses is that oncogene-driven cancers with activating mutations of EGFR often harbor additional co-occurring genetic alterations. This hypothesis remains untested for most genetic alterations that co-occur with mutant EGFR. Here, we report the functional impact of inactivating genetic alterations of the mRNA splicing factor RNA-binding motif 10 (RBM10) that co-occur with mutant EGFR. RBM10 deficiency decreased EGFR inhibitor efficacy in patient-derived EGFR-mutant tumor models. RBM10 modulated mRNA alternative splicing of the mitochondrial apoptotic regulator Bcl-x to regulate tumor cell apoptosis during treatment. Genetic inactivation of RBM10 diminished EGFR inhibitor-mediated apoptosis by decreasing the ratio of (proapoptotic) Bcl-xS to (antiapoptotic) Bcl-xL isoforms of Bcl-x. RBM10 deficiency was a biomarker of poor response to EGFR inhibitor treatment in clinical samples. Coinhibition of Bcl-xL and mutant EGFR overcame the resistance induced by RBM10 deficiency. This study sheds light on the role of co-occurring genetic alterations and on the effect of splicing factor deficiency on the modulation of sensitivity to targeted kinase inhibitor cancer therapy.


Sujet(s)
Facteur X , Tumeurs du poumon , Apoptose/génétique , Lignée cellulaire tumorale , Récepteurs ErbB/génétique , Facteur X/usage thérapeutique , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Facteurs d'épissage des ARN , ARN messager/génétique , Motifs de liaison à l'ARN , Protéines de liaison à l'ARN/métabolisme
8.
Sci Transl Med ; 14(638): eabc7480, 2022 03 30.
Article de Anglais | MEDLINE | ID: mdl-35353542

RÉSUMÉ

Residual cancer cells that survive drug treatments with targeted therapies act as a reservoir from which eventual resistant disease emerges. Although there is great interest in therapeutically targeting residual cells, efforts are hampered by our limited knowledge of the vulnerabilities existing in this cell state. Here, we report that diverse oncogene-targeted therapies, including inhibitors of epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), KRAS, and BRAF, induce DNA double-strand breaks and, consequently, ataxia-telangiectasia mutated (ATM)-dependent DNA repair in oncogene-matched residual tumor cells. This DNA damage response, observed in cell lines, mouse xenograft models, and human patients, is driven by a pathway involving the activation of caspases 3 and 7 and the downstream caspase-activated deoxyribonuclease (CAD). CAD is, in turn, activated through caspase-mediated degradation of its endogenous inhibitor, ICAD. In models of EGFR mutant non-small cell lung cancer (NSCLC), tumor cells that survive treatment with small-molecule EGFR-targeted therapies are thus synthetically dependent on ATM, and combined treatment with an ATM kinase inhibitor eradicates these cells in vivo. This led to more penetrant and durable responses in EGFR mutant NSCLC mouse xenograft models, including those derived from both established cell lines and patient tumors. Last, we found that rare patients with EGFR mutant NSCLC harboring co-occurring, loss-of-function mutations in ATM exhibit extended progression-free survival on first generation EGFR inhibitor therapy relative to patients with EGFR mutant NSCLC lacking deleterious ATM mutations. Together, these findings establish a rationale for the mechanism-based integration of ATM inhibitors alongside existing targeted therapies.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , ADN , Réparation de l'ADN , Humains , Tumeurs du poumon/traitement médicamenteux , Souris , Maladie résiduelle
9.
Chem Commun (Camb) ; 58(19): 3166-3169, 2022 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-35170593

RÉSUMÉ

This article reports the synthesis and characterization of a novel self-immolative linker, based on thiocarbonates, which releases a free thiol upon activation via enzymes. We demonstrate that thiocarbonate self-immolative linkers can be used to detect the enzymes penicillin G amidase (PGA) and nitroreductase (NTR) with high sensitivity using absorption spectroscopy. Paired with modern thiol amplification technology, the detection of PGA and NTR were achieved at concentrations of 160 nM and 52 nM respectively. In addition, the PGA probe was shown to be compatible with both biological thiols and enzymes present in cell lysates.


Sujet(s)
Nitroréductases/analyse , Penicillin amidase/analyse , Thiols/composition chimique , Structure moléculaire , Nitroréductases/métabolisme , Penicillin amidase/métabolisme , Spectrométrie de fluorescence
10.
Mol Pharm ; 19(1): 67-79, 2022 01 03.
Article de Anglais | MEDLINE | ID: mdl-34931518

RÉSUMÉ

The development of endosomal disruptive agents is a major challenge in the field of drug delivery and pharmaceutical chemistry. Current endosomal disruptive agents are composed of polymers, peptides, and nanoparticles and have had limited clinical impact. Alternatives to traditional endosomal disruptive agents are therefore greatly needed. In this report, we introduce a new class of low molecular weight endosomal disruptive agents, termed caged surfactants, that selectively disrupt endosomes via reversible PEGylation under acidic endosomal conditions. The caged surfactants have the potential to address several of the limitations hindering the development of current endosomal disruptive agents, such as high toxicity and low excretion, and are amenable to traditional medicinal chemistry approaches for optimization. In this report, we synthesized three generations of caged surfactants and demonstrated that they can enhance the ability of cationic lipids to deliver mRNA into primary cells. We also show that caged surfactants can deliver siRNA into cells when modified with the RNA-binding dye thiazole orange. We anticipate that the caged surfactants will have numerous applications in pharmaceutical chemistry and drug delivery given their versatility.


Sujet(s)
Systèmes de délivrance de médicaments , Acides nucléiques/administration et posologie , Tensioactifs/usage thérapeutique , Systèmes de délivrance de médicaments/méthodes , Endosomes/effets des médicaments et des substances chimiques , Hémolyse/effets des médicaments et des substances chimiques , Humains , Concentration en ions d'hydrogène , ARN messager/administration et posologie , Petit ARN interférent/administration et posologie , Relation structure-activité , Tensioactifs/administration et posologie , Tensioactifs/composition chimique
11.
Curr Opin Chem Biol ; 62: 1-12, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33418513

RÉSUMÉ

The nonreceptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) integrates growth and differentiation signals from receptor tyrosine kinases (RTKs) into the RAS/mitogen-activated protein kinase (MAPK) cascade. Considered 'undruggable' over three decades, SHP2 is now a potentially druggable target with the advent of allosteric SHP2 inhibitors. These agents hold promise for improving patient outcomes, showing efficacy in preclinical cancer models, where SHP2 is critical for either oncogenic signaling or resistance to current targeted agents. SHP2 inhibition may also produce immunomodulatory effects in certain tumor microenvironment cells to help cultivate antitumor immune responses. The first generation of allosteric SHP2 inhibitors is under clinical evaluation to determine safety, appropriate tolerability management, and antitumor efficacy, investigations that will dictate future clinical applications.


Sujet(s)
Antinéoplasiques/composition chimique , Antienzymes/composition chimique , Mitogen-Activated Protein Kinases/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Protéines G ras/métabolisme , Régulation allostérique/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/immunologie , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Antienzymes/immunologie , Antienzymes/pharmacologie , Humains , Immunothérapie , Simulation de docking moléculaire , Liaison aux protéines , Conformation des protéines , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Cell ; 182(5): 1232-1251.e22, 2020 09 03.
Article de Anglais | MEDLINE | ID: mdl-32822576

RÉSUMÉ

Lung cancer, the leading cause of cancer mortality, exhibits heterogeneity that enables adaptability, limits therapeutic success, and remains incompletely understood. Single-cell RNA sequencing (scRNA-seq) of metastatic lung cancer was performed using 49 clinical biopsies obtained from 30 patients before and during targeted therapy. Over 20,000 cancer and tumor microenvironment (TME) single-cell profiles exposed a rich and dynamic tumor ecosystem. scRNA-seq of cancer cells illuminated targetable oncogenes beyond those detected clinically. Cancer cells surviving therapy as residual disease (RD) expressed an alveolar-regenerative cell signature suggesting a therapy-induced primitive cell-state transition, whereas those present at on-therapy progressive disease (PD) upregulated kynurenine, plasminogen, and gap-junction pathways. Active T-lymphocytes and decreased macrophages were present at RD and immunosuppressive cell states characterized PD. Biological features revealed by scRNA-seq were biomarkers of clinical outcomes in independent cohorts. This study highlights how therapy-induced adaptation of the multi-cellular ecosystem of metastatic cancer shapes clinical outcomes.


Sujet(s)
Tumeurs du poumon/génétique , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire , Écosystème , Humains , Tumeurs du poumon/anatomopathologie , Macrophages/anatomopathologie , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Lymphocytes T/anatomopathologie , Microenvironnement tumoral/génétique
13.
Chem Commun (Camb) ; 55(31): 4562-4565, 2019 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-30931453

RÉSUMÉ

In this report, we designed and synthesized a novel fluorescent single tailed surfactant (termed FEDS), which can disrupt endosomes, complex lipofectamine, and can also identify cells that have been transfected. FEDS was able to increase the gene editing efficiency of lipofectamine/Cas9 ribonucleoprotein by 300% via a combination of fluorescent based enrichment and endosomal disruption.


Sujet(s)
Protéine-9 associée à CRISPR/génétique , Édition de gène/méthodes , Lipides/composition chimique , Animaux , Lignée cellulaire , Endosomes/métabolisme , Érythrocytes/cytologie , Érythrocytes/métabolisme , Cytométrie en flux , Colorants fluorescents/composition chimique , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Humains , Lapins
14.
Mol Syst Des Eng ; 3(4): 599-603, 2018 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-30740245

RÉSUMÉ

The development of antibiotics against Gram-negative bacteria is a central problem in drug discovery. In this report, we demonstrate that aromatic sulfonyl fluorides with a nitro group in their ortho position have remarkable antibacterial activity and are active against drug-resistant pathogens, such as methicillin-resistant Staphylococcus aureus (MRSA), multidrug resistant Acinetobacter baumannii, and Pseudomonas aeruginosa.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE