Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 50.307
Filtrer
1.
JMIR Res Protoc ; 13: e51381, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954434

RÉSUMÉ

BACKGROUND: Starting in 2010, the epidermal growth factor receptor (EGFR) kinase inhibitors erlotinib and gefitinib were introduced into routine use in Aotearoa New Zealand (NZ) for treating advanced lung cancer, but their impact in this setting is unknown. OBJECTIVE: The study described in this protocol aims to understand the effectiveness and safety of these new personalized lung cancer treatments and the contributions made by concomitant medicines and other factors to adverse outcomes in the general NZ patient population. A substudy aimed to validate national electronic health databases as the data source and the methods for determining patient eligibility and identifying outcomes and variables. METHODS: This study will include all NZ patients with advanced EGFR mutation-positive lung cancer who were first dispensed erlotinib or gefitinib before October 1, 2020, and followed until death or for at least 1 year. Routinely collected health administrative and clinical data will be collated from national electronic cancer registration, hospital discharge, mortality registration, and pharmaceutical dispensing databases by deterministic data linkage using National Health Index numbers. The primary effectiveness and safety outcomes will be time to treatment discontinuation and serious adverse events, respectively. The primary variable will be high-risk concomitant medicines use with erlotinib or gefitinib. For the validation substudy (n=100), data from clinical records were compared to those from national electronic health databases and analyzed by agreement analysis for categorical data and by paired 2-tailed t tests for numerical data. RESULTS: In the validation substudy, national electronic health databases and clinical records agreed in determining patient eligibility and for identifying serious adverse events, high-risk concomitant medicines use, and other categorical data with overall agreement and κ statistic of >90% and >0.8000, respectively; for example, for the determination of patient eligibility, the comparison of proxy and standard eligibility criteria applied to national electronic health databases and clinical records, respectively, showed overall agreement and κ statistic of 96% and 0.8936, respectively. Dates for estimating time to treatment discontinuation and other numerical variables and outcomes showed small differences, mostly with nonsignificant P values and 95% CIs overlapping with zero difference; for example, for the dates of the first dispensing of erlotinib or gefitinib, national electronic health databases and clinical records differed on average by approximately 4 days with a nonsignificant P value of .33 and 95% CIs overlapping with zero difference. As of May 2024, the main study is ongoing. CONCLUSIONS: A protocol is presented for a national whole-of-patient-population retrospective cohort study designed to describe the safety and effectiveness of erlotinib and gefitinib during their first decade of routine use in NZ for treating EGFR mutation-positive lung cancer. The validation substudy demonstrated the feasibility and validity of using national electronic health databases and the methods for determining patient eligibility and identifying the study outcomes and variables proposed in the study protocol. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry ACTRN12615000998549; https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=368928. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/51381.


Sujet(s)
Récepteurs ErbB , Chlorhydrate d'erlotinib , Géfitinib , Tumeurs du poumon , Mutation , Humains , Chlorhydrate d'erlotinib/usage thérapeutique , Chlorhydrate d'erlotinib/effets indésirables , Géfitinib/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Récepteurs ErbB/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Études rétrospectives , Nouvelle-Zélande , Femelle , Mâle , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/effets indésirables , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Études de cohortes , Adulte d'âge moyen , Sujet âgé
2.
Drug Dev Res ; 85(5): e22231, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38956926

RÉSUMÉ

The close association between inflammation and cancer inspired the synthesis of a series of 1,3,4-oxadiazole derivatives (compounds H4-A-F) of 6-methoxynaphtalene. The chemical structures of the new compounds were validated utilizing Fourier-transform infrared, proton nuclear magnetic resonance, and carbon-13 nuclear magnetic resonance spectroscopic techniques and CHN analysis. Computer-aided drug design methods were used to predict the compounds biological target, ADMET properties, toxicity, and to evaluate the molecular similarities between the design compounds and erlotinib, a standard epidermal growth factor receptor (EGFR) inhibitor. The antiproliferative effects of the new compounds were evaluated by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay, cell cycle analysis, apoptosis detection by microscopy, quantitative reverse transcription-polymerase chain reaction, and immunoblotting, and EGFR enzyme inhibition assay. In silico analysis of the new oxadiazole derivatives indicated that these compounds target EGFR, and that compounds H4-A, H4-B, H4-C, and H4-E show similar molecular properties to erlotinib. Additionally, the results indicated that none of the synthesized compounds are carcinogenic, and that compounds H4-A, H4-C, and H4-F are nontoxic. Compound H4-A showed the best-fit score against EGFR pharmacophore model, however, the in vitro studies indicated that compound H4-C was the most cytotoxic. Compound H4-C caused cytotoxicity in HCT-116 colorectal cancer cells by inducing both apoptosis and necrosis. Furthermore, compounds H4-D, H4-C, and H4-B had potent inhibitory effect on EGFR tyrosine kinase that was comparable to erlotinib. The findings of this inquiry offer a basis for further investigation into the differences between the synthesized compounds and erlotinib. However, additional testing will be needed to assess all of these differences and to identify the most promising compound for further research.


Sujet(s)
Antinéoplasiques , Récepteurs ErbB , Simulation de docking moléculaire , Naproxène , Oxadiazoles , Récepteurs ErbB/antagonistes et inhibiteurs , Humains , Oxadiazoles/pharmacologie , Oxadiazoles/composition chimique , Oxadiazoles/synthèse chimique , Naproxène/pharmacologie , Naproxène/analogues et dérivés , Naproxène/composition chimique , Naproxène/synthèse chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Chlorhydrate d'erlotinib/pharmacologie , Chlorhydrate d'erlotinib/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques
3.
Sci Rep ; 14(1): 15160, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956132

RÉSUMÉ

In order to survive and replicate, Salmonella has evolved mechanisms to gain access to intestinal epithelial cells of the crypt. However, the impact of Salmonella Typhimurium on stem cells and progenitors, which are responsible for the ability of the intestinal epithelium to renew and protect itself, remains unclear. Given that intestinal organoids growth is sustained by stem cells and progenitors activity, we have used this model to document the effects of Salmonella Typhimurium infection on epithelial proliferation and differentiation, and compared it to an in vivo model of Salmonella infection in mice. Among gut segments, the caecum was preferentially targeted by Salmonella. Analysis of infected crypts and organoids demonstrated increased length and size, respectively. mRNA transcription profiles of infected crypts and organoids pointed to upregulated EGFR-dependent signals, associated with a decrease in secretory cell lineage differentiation. To conclude, we show that organoids are suited to mimic the impact of Salmonella on stem cells and progenitors cells, carrying a great potential to drastically reduce the use of animals for scientific studies on that topic. In both models, the EGFR pathway, crucial to stem cells and progenitors proliferation and differentiation, is dysregulated by Salmonella, suggesting that repeated infections might have consequences on crypt integrity and further oncogenesis.


Sujet(s)
Différenciation cellulaire , Récepteurs ErbB , Organoïdes , Salmonelloses , Salmonella typhimurium , Cellules souches , Animaux , Organoïdes/microbiologie , Cellules souches/métabolisme , Souris , Salmonella typhimurium/pathogénicité , Salmonella typhimurium/physiologie , Salmonelloses/microbiologie , Salmonelloses/anatomopathologie , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Prolifération cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL
4.
BMC Pulm Med ; 24(1): 309, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956553

RÉSUMÉ

BACKGROUND: Treatment of non-small lung cancer (NSCLC) has evolved in recent years, benefiting from advances in immunotherapy and targeted therapy. However, limited biomarkers exist to assist clinicians and patients in selecting the most effective, personalized treatment strategies. Targeted next-generation sequencing-based genomic profiling has become routine in cancer treatment and generated crucial clinicogenomic data over the last decade. This has made the development of mutational biomarkers for drug response possible. METHODS: To investigate the association between a patient's responses to a specific somatic mutation treatment, we analyzed the NSCLC GENIE BPC cohort, which includes 2,004 tumor samples from 1,846 patients. RESULTS: We identified somatic mutation signatures associated with response to immunotherapy and chemotherapy, including carboplatin-, cisplatin-, pemetrexed- or docetaxel-based chemotherapy. The prediction power of the chemotherapy-associated signature was significantly affected by epidermal growth factor receptor (EGFR) mutation status. Therefore, we developed an EGFR wild-type-specific mutation signature for chemotherapy selection. CONCLUSION: Our treatment-specific gene signatures will assist clinicians and patients in selecting from multiple treatment options.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Récepteurs ErbB/génétique , Sujet âgé , Pronostic , Études de cohortes , Marqueurs biologiques tumoraux/génétique , Immunothérapie , Carboplatine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pémétrexed/usage thérapeutique , Médecine de précision , Séquençage nucléotidique à haut débit , Antinéoplasiques/usage thérapeutique
6.
PLoS One ; 19(7): e0306429, 2024.
Article de Anglais | MEDLINE | ID: mdl-38980867

RÉSUMÉ

Brucella abortus (Ba) is a pathogen that survives inside macrophages. Despite being its preferential niche, Ba infects other cells, as shown by the multiple signs and symptoms humans present. This pathogen can evade our immune system. Ba displays a mechanism of down-modulating MHC-I on monocytes/macrophages in the presence of IFN-γ (when Th1 response is triggered) without altering the total expression of MHC-I. The retained MHC-I proteins are located within the Golgi Apparatus (GA). The RNA of Ba is one of the PAMPs that trigger this phenomenon. However, we acknowledged whether this event could be triggered in other cells relevant during Ba infection. Here, we demonstrate that Ba RNA reduced the surface expression of MHC-I induced by IFN-γ in the human bronchial epithelium (Calu-6), the human alveolar epithelium (A-549) and the endothelial microvasculature (HMEC) cell lines. In Calu-6 and HMEC cells, Ba RNA induces the retention of MHC-I in the GA. This phenomenon was not observed in A-549 cells. We then evaluated the effect of Ba RNA on the secretion of IL-8, IL-6 and MCP-1, key cytokines in Ba infection. Contrary to our expectations, HMEC, Calu-6 and A-549 cells treated with Ba RNA had higher IL-8 and IL-6 levels compared to untreated cells. In addition, we showed that Ba RNA down-modulates the MHC-I surface expression induced by IFN-γ on human monocytes/macrophages via the pathway of the Epidermal Growth Factor Receptor (EGFR). So, cells were stimulated with an EGFR ligand-blocking antibody (Cetuximab) and Ba RNA. Neutralization of the EGFR to some extent reversed the down-modulation of MHC-I mediated by Ba RNA in HMEC and A-549 cells. In conclusion, this is the first study exploring a central immune evasion strategy, such as the downregulation of MHC-I surface expression, beyond monocytes and could shed light on how it persists effectively within the host, enduring unseen and escaping CD8+ T cell surveillance.


Sujet(s)
Brucella abortus , Cellules endothéliales , Cellules épithéliales , Antigènes d'histocompatibilité de classe I , Interféron gamma , Humains , Interféron gamma/métabolisme , Interféron gamma/pharmacologie , Cellules endothéliales/métabolisme , Cellules endothéliales/microbiologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Cellules épithéliales/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , ARN bactérien/génétique , Lignée cellulaire , Régulation négative/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Brucellose/immunologie , Brucellose/métabolisme , Brucellose/microbiologie , Brucellose/génétique , Appareil de Golgi/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Monocytes/métabolisme , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques
7.
JCO Precis Oncol ; 8: e2300725, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38986051

RÉSUMÉ

PURPOSE: National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) was a multicohort phase 2 trial that assigned patients with advanced pretreated cancers to molecularly targeted therapies on the basis of tumor genomic testing. NCI-MATCH Arm A evaluated afatinib, an EGFR tyrosine kinase inhibitor (TKI) approved for advanced non-small cell lung cancer, in patients with tumors other than lung cancer harboring EGFR mutations. METHODS: Patients with advanced pretreated cancers other than lung cancer found to have selected actionable EGFR mutations were offered participation in Arm A. Previous therapy with an EGFR TKI was not allowed. Patients received afatinib 40 mg once daily continuously until disease progression or unacceptable toxicity. The primary end point was objective response rate (ORR). Secondary end points included progression-free survival (PFS), 6-month PFS, and overall survival (OS). RESULTS: Seventeen patients received protocol therapy. Tumor types included glioblastoma multiforme (GBM) (13), gliosarcoma (1), adenocarcinoma not otherwise specified (NOS) (2), and adenosquamous carcinoma of the breast (1). Fifty-nine percent of patients received ≥2 lines of previous therapy. The ORR was 11.8% (90% CI, 2.1 to 32.6), with one complete response lasting 16.4 months (GBM harboring a rare exon 18 EGFR-SEPT14 fusion) and one partial response lasting 12.8 months (adenocarcinoma NOS with the classic EGFR mutation, p.Glu746_Ala750del). Three patients had stable disease. The 6-month PFS was 15% (90% CI, 0 to 30.7); the median OS was 9 months (90% CI, 4.6 to 14.0). Rash and diarrhea were the most common toxicities. CONCLUSION: Afatinib had modest activity in a cohort of patients with heavily pretreated cancer with advanced nonlung, EGFR-mutated tumors, but the trial's primary end point was not met. Further evaluation of afatinib in GBM with EGFR exon 18 fusions may be of interest.


Sujet(s)
Afatinib , Récepteurs ErbB , Mutation , Humains , Afatinib/usage thérapeutique , Femelle , Mâle , Adulte d'âge moyen , Récepteurs ErbB/génétique , Sujet âgé , Adulte , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Sujet âgé de 80 ans ou plus
8.
Science ; 385(6705): 194-200, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38991070

RÉSUMÉ

Millions of hibernating bats across North America have died from white-nose syndrome (WNS), an emerging disease caused by a psychrophilic (cold-loving) fungus, Pseudogymnoascus destructans, that invades their skin. Mechanisms of P. destructans invasion of bat epidermis remain obscure. Guided by our in vivo observations, we modeled hibernation with a newly generated little brown bat (Myotis lucifugus) keratinocyte cell line. We uncovered the stealth intracellular lifestyle of P. destructans, which inhibits apoptosis of keratinocytes and spreads through the cells by two epidermal growth factor receptor (EGFR)-dependent mechanisms: active penetration during torpor and induced endocytosis during arousal. Melanin of endocytosed P. destructans blocks endolysosomal maturation, facilitating P. destructans survival and germination after return to torpor. Blockade of EGFR aborts P. destructans entry into keratinocytes.


Sujet(s)
Éveil , Ascomycota , Chiroptera , Récepteurs ErbB , Hibernation , Kératinocytes , Animaux , Apoptose , Ascomycota/physiologie , Ascomycota/pathogénicité , Lignée cellulaire , Chiroptera/microbiologie , Chiroptera/physiologie , Endocytose , Récepteurs ErbB/métabolisme , Kératinocytes/microbiologie , Mélanines/métabolisme
9.
FASEB J ; 38(13): e23759, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38949635

RÉSUMÉ

The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.


Sujet(s)
ADP ribose transferases , Récepteurs ErbB , Exotoxines , Tumeurs de la tête et du cou , Immunoglobuline G , Immunotoxines , , Facteurs de virulence , Humains , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/immunologie , Animaux , Immunotoxines/pharmacologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/métabolisme , Souris , Immunoglobuline G/pharmacologie , Lignée cellulaire tumorale , Exotoxines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Cétuximab/pharmacologie , Souris nude , Toxines bactériennes , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Femelle , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie
10.
Drug Dev Res ; 85(5): e22228, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38952003

RÉSUMÉ

Chromone-based compounds have established cytotoxic, antiproliferative, antimetastatic, and antiangiogenic effects on various cancer cell types via modulating different molecular targets. Herein, 17 novel chromone-2-carboxamide derivatives were synthesized and evaluated for their in vitro anticancer activity against 15 human cancer cell lines. Among the tested cell lines, MDA-MB-231, the triple-negative breast cancer cell line, was found to be the most sensitive, where the N-(2-furylmethylene) (15) and the α-methylated N-benzyl (17) derivatives demonstrated the highest growth inhibition with GI50 values of 14.8 and 17.1 µM, respectively. In vitro mechanistic studies confirmed the significant roles of compounds 15 and 17 in the induction of apoptosis and suppression of EGFR, FGFR3, and VEGF protein levels in MDA-MB-231 cancer cells. Moreover, compound 15 exerted cell cycle arrest at both the G0-G1 and G2-M phases. The in vivo efficacy of compound 15 as an antitumor agent was further investigated in female mice bearing Solid Ehrlich Carcinoma. Notably, administration of compound 15 resulted in a marked decrease in both tumor weight and volume, accompanied by improvements in biochemical, hematological, histological, and immunohistochemical parameters that verified the repression of both angiogenesis and inflammation as additional Anticancer mechanisms. Moreover, the binding interactions of compounds 15 and 17 within the binding sites of all three target receptors (EGFR, FGFR3, and VEGF) were clearly illustrated using molecular docking.


Sujet(s)
Antinéoplasiques , 4H-1-Benzopyran-4-ones , Récepteurs ErbB , Simulation de docking moléculaire , Récepteur de type 3 des facteurs de croissance fibroblastique , Tumeurs du sein triple-négatives , Facteur de croissance endothéliale vasculaire de type A , Humains , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Femelle , Animaux , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Récepteur de type 3 des facteurs de croissance fibroblastique/antagonistes et inhibiteurs , Récepteur de type 3 des facteurs de croissance fibroblastique/métabolisme , Souris , 4H-1-Benzopyran-4-ones/pharmacologie , 4H-1-Benzopyran-4-ones/synthèse chimique , 4H-1-Benzopyran-4-ones/composition chimique , 4H-1-Benzopyran-4-ones/usage thérapeutique , Conception de médicament , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
11.
Drug Des Devel Ther ; 18: 2571-2591, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947223

RÉSUMÉ

Purpose: Activating mutations in epidermal growth factor receptor (EGFR) have been identified as key predictive biomarkers for the customized treatment with EGFR tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC), aiding in improving patient response rates and survival. However, resistance challenges the efficacy of these treatments, with limited understanding of post-resistance therapeutic strategies. A deep understanding of the biology and resistance mechanisms of EGFR-mutant NSCLC is crucial for developing new treatment approaches. This study, through bibliometric analysis, summarizes the trends in research on resistance to EGFR-TKIs. Methods: Research papers on NSCLC with EGFR inhibitor resistance were collected from the Web of Science Core Collection (WoSCC). The analysis utilized bibliometric tools like CiteSpace, VOSviewer, and other platforms for comprehensive analysis and visualization of the outcomes. Results: The WoSCC database contains a total of 5866 documents on resistance to EGFR-TKIs treatment, including 4727 articles (93.48%) and 1139 reviews (6.52%), spanning 81 countries and regions, 4792 institutions, with the involvement of 23,594 authors. Since 2016, there has been a significant increase in publications in this field. China has the highest publication output, while the United States has the highest citation count for papers. Harvard University leads in terms of the number of publications. Among the top ten journals with the highest output, Clinical Cancer Research has the highest impact factor at 11.5, with 90% of the journals classified in Q1 or Q2. Rafael Rosell is one of the most influential authors in this field, ranking second in publication volume and fourth in citation count. Research on EGFR-TKIs resistance mainly focuses on genetic testing, resistance mechanisms, and post-resistance treatment strategies. Conclusion: This study provides researchers with a reliable basis and guidance for finding authoritative references, understanding research trends, and exploring potential directions.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Bibliométrie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique
12.
BMJ Open ; 14(7): e078335, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969367

RÉSUMÉ

BACKGROUND: Patients with advanced non-small-cell lung cancer (NSCLC) with activating mutations in the epidermal growth factor receptor (EGFR) gene are a heterogeneous population who often develop brain metastases (BM). The optimal management of patients with asymptomatic brain metastases is unclear given the activity of newer-generation targeted therapies in the central nervous system. We present a protocol for an individual patient data (IPD) prospective meta-analysis to evaluate whether the addition of stereotactic radiosurgery (SRS) before osimertinib treatment will lead to better control of intracranial metastatic disease. This is a clinically relevant question that will inform practice. METHODS: Randomised controlled trials will be eligible if they include participants with BM arising from EGFR-mutant NSCLC and suitable to receive osimertinib both in the first-line and second-line settings (P); comparisons of SRS followed by osimertinib versus osimertinib alone (I, C) and intracranial disease control included as an endpoint (O). Systematic searches of Medline (Ovid), Embase (Ovid), Cochrane Central Register of Controlled Trials (CENTRAL), CINAHL (EBSCO), PsychInfo, ClinicalTrials.gov and the WHO's International Clinical Trials Registry Platform's Search Portal will be undertaken. An IPD meta-analysis will be performed using methodologies recommended by the Cochrane Collaboration. The primary outcome is intracranial progression-free survival, as determined by response assessment in neuro-oncology-BM criteria. Secondary outcomes include overall survival, time to whole brain radiotherapy, quality of life, and adverse events of special interest. Effect differences will be explored among prespecified subgroups. ETHICS AND DISSEMINATION: Approved by each trial's ethics committee. Results will be relevant to clinicians, researchers, policymakers and patients, and will be disseminated via publications, presentations and media releases. PROSPERO REGISTRATION: CRD42022330532.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Radiochirurgie , Revues systématiques comme sujet , Humains , Acrylamides/usage thérapeutique , Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/secondaire , Association thérapeutique , Récepteurs ErbB/génétique , Indoles , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Méta-analyse comme sujet , Mutation , Études prospectives , Pyrimidines , Radiochirurgie/méthodes , Essais contrôlés randomisés comme sujet , Plan de recherche
13.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38977310

RÉSUMÉ

Hippocampal seizures mimicking mesial temporal lobe epilepsy cause a profound disruption of the adult neurogenic niche in mice. Seizures provoke neural stem cells to switch to a reactive phenotype (reactive neural stem cells, React-NSCs) characterized by multibranched hypertrophic morphology, massive activation to enter mitosis, symmetric division, and final differentiation into reactive astrocytes. As a result, neurogenesis is chronically impaired. Here, using a mouse model of mesial temporal lobe epilepsy, we show that the epidermal growth factor receptor (EGFR) signaling pathway is key for the induction of React-NSCs and that its inhibition exerts a beneficial effect on the neurogenic niche. We show that during the initial days after the induction of seizures by a single intrahippocampal injection of kainic acid, a strong release of zinc and heparin-binding epidermal growth factor, both activators of the EGFR signaling pathway in neural stem cells, is produced. Administration of the EGFR inhibitor gefitinib, a chemotherapeutic in clinical phase IV, prevents the induction of React-NSCs and preserves neurogenesis.


Sujet(s)
Récepteurs ErbB , Facteur de croissance de type EGF liant l'héparine , Hippocampe , Cellules souches neurales , Neurogenèse , Crises épileptiques , Transduction du signal , Animaux , Récepteurs ErbB/métabolisme , Cellules souches neurales/métabolisme , Cellules souches neurales/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Souris , Facteur de croissance de type EGF liant l'héparine/métabolisme , Crises épileptiques/métabolisme , Neurogenèse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Modèles animaux de maladie humaine , Géfitinib/pharmacologie , Épilepsie temporale/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Acide kaïnique/pharmacologie , Souris de lignée C57BL
14.
Biomark Med ; 18(9): 431-439, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007837

RÉSUMÉ

Leptomeningeal metastasis (LM) is a devastating complication of malignancy. Diagnosis relies on both contrast enhancement on imaging and malignant cells in cerebral spinal fluid cytology. Though early detection and prompt intervention improves survival, the detection of LM is limited by false negatives. A rare brainstem imaging finding uncovered specifically in EGFR mutation-positive lung cancer patients may represent an early sign of LM. This sign demonstrates high signal on T2 fluid-attenuated inversion recovery and diffusion-weighted imaging sequences, but paradoxically lacks correlative contrast enhancement. Here we report a case of a 72-year-old female EGFR-positive lung cancer patient who developed this lesion following treatment with two first-generation EGFR tyrosine kinase inhibitors then showed subsequent response to osimertinib, an irreversible third-generation EGFR tyrosine kinase inhibitor.


A non-enhancing, T2 FLAIR hyperintense, diffusion-restricting brainstem lesion in an EGFR-positive lung cancer patient may represent an early indicator of leptomeningeal metastases.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Femelle , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/imagerie diagnostique , Carcinome pulmonaire non à petites cellules/métabolisme , Sujet âgé , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/imagerie diagnostique , Tronc cérébral/anatomopathologie , Tronc cérébral/imagerie diagnostique , Tronc cérébral/métabolisme , Dérivés de l'aniline/usage thérapeutique , Acrylamides/usage thérapeutique , Imagerie par résonance magnétique de diffusion , Indoles , Pyrimidines
15.
Medicine (Baltimore) ; 103(28): e38789, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996163

RÉSUMÉ

RATIONALE: Acquired resistance still inevitably occurs in patients treated with third-generation TKI osimertinib. Although the EGFR L718Q mutation has been reported as a scarce mechanism of osimertinib resistance, advanced therapeutic strategies are still in development. In this report, we included 2 cases of patients who acquired EGFR L858R/L718Q mutation after osimertinib and were overcome by dacomitinib. PATIENT CONCERNS: Case 1: A 77-year-old woman was diagnosed with stage IV lung adenocarcinoma. Case 2: A 64-year-old woman was diagnosed with stage IV lung adenocarcinoma. DIAGNOSES: Case 1: The patient was diagnosed with adenocarcinoma with EGFR L858R mutation. Since then, treatment with gefitinib was administrated, leading to a progression-free survival of 18 months. The treatment was switched to osimertinib based on the detection of EGFR T790M mutation, resulting in a progression-free survival of 24 months. Subsequently, EGFR L718Q mutation was detected. Case 2: The patient was diagnosed with adenocarcinoma with EGFR L858R mutation. Icotinib was used as the first-line treatment for 7 months. Osimertinib was applied as the second-line treatment for 13 months based on the EGFR T790M mutation. Subsequently, EGFR L718Q mutation was detected. INTERVENTIONS: Case 1: Dacomitinib was administered. Case 2: Dacomitinib was administered. OUTCOMES: Case 1:The progression-free survival was 8 months. Case 2: The progression-free survival was 3 months. LESSONS: Dacomitinib is a potential treatment option for NSCLC patients with EGFR L718Q mutation after resistance to Osimertinib. Further research is needed to validate the efficacy of Dacomitinib in this context.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Mutation , Quinazolinones , Humains , Femelle , Acrylamides/usage thérapeutique , Dérivés de l'aniline/usage thérapeutique , Récepteurs ErbB/génétique , Sujet âgé , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Résistance aux médicaments antinéoplasiques/génétique , Quinazolinones/usage thérapeutique , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Antinéoplasiques/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Indoles , Pyrimidines
16.
Nat Commun ; 15(1): 5874, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997284

RÉSUMÉ

Mucus injury associated with goblet cell (GC) depletion constitutes an early event in inflammatory bowel disease (IBD). Using single-cell sequencing to detect critical events in mucus dysfunction, we discover that the Kazal-type serine protease inhibitor SPINK4 is dynamically regulated in colitic intestine in parallel with disease activities. Under chemically induced colitic conditions, the grim status in Spink4-conditional knockout mice is successfully rescued by recombinant murine SPINK4. Notably, its therapeutic potential is synergistic with existing TNF-α inhibitor infliximab in colitis treatment. Mechanistically, SPINK4 promotes GC differentiation using a Kazal-like motif to modulate EGFR-Wnt/ß-catenin and -Hippo pathways. Microbiota-derived diacylated lipoprotein Pam2CSK4 triggers SPINK4 production. We also show that monitoring SPINK4 in circulation is a reliable noninvasive technique to distinguish IBD patients from healthy controls and assess disease activity. Thus, SPINK4 serves as a serologic biomarker of IBD and has therapeutic potential for colitis via intrinsic EGFR activation in intestinal homeostasis.


Sujet(s)
Colite , Souris knockout , Animaux , Colite/génétique , Colite/induit chimiquement , Colite/anatomopathologie , Colite/traitement médicamenteux , Colite/métabolisme , Humains , Souris , Cellules caliciformes/métabolisme , Cellules caliciformes/anatomopathologie , Cellules caliciformes/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Souris de lignée C57BL , Inhibiteurs de serine peptidase de type Kazal/génétique , Inhibiteurs de serine peptidase de type Kazal/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Mâle , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/métabolisme , Femelle , Modèles animaux de maladie humaine , Marqueurs biologiques/sang , Marqueurs biologiques/métabolisme , Différenciation cellulaire
17.
BMC Cancer ; 24(1): 835, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997622

RÉSUMÉ

PURPOSE: Extracellular heat shock protein 90 AA1(eHSP90α) is intricately linked to tumor progression and prognosis. This study aimed to investigate the difference in the value of eHSP90α in post-treatment response assessment and prognosis prediction between exon 19 deletion(19DEL) and exon 21 Leu858Arg(L858R) mutation types in lung adenocarcinoma(LUAD). METHODS: We analyzed the relationship between the expression of eHSP90α and clinicopathological features in 89 patients with L858R mutation and 196 patients with 19DEL mutation in LUAD. The Kaplan-Meier survival curve was used to determine their respective cut-off values and analyze the relationship between eHSP90α expression and the survival time of the two mutation types. The area under the curve (AUC) was used to evaluate the diagnostic performance of biomarkers. Then, the prognostic model was developed using the univariate-Cox multivariate-Cox and LASSO-multivariate logistic methods. RESULTS: In LUAD patients, eHSP90α was positively correlated with carcinoembryonic antigen(CEA), carbohydrate antigen 125(CA125), and carbohydrate antigen 153(CA153). The truncated values of eHSP90α in L858R and 19DEL patients were 44.5 ng/mL and 40.8 ng/mL, respectively. Among L858R patients, eHSP90α had the best diagnostic performance (AUC = 0.765), and higher eHSP90α and T helper cells(Th cells) expression were significantly related to shorter overall survival(OS) and worse treatment response. Also, high eHSP90a expression and short progression-free survival(PFS) were significantly correlated. Among 19DEL patients, CEA had the best diagnostic efficacy (AUC = 0.734), and CEA and Th cells were independent prognostic factors that predicted shorter OS. Furthermore, high CA125 was significantly associated with short PFS and poor curative effect. CONCLUSIONS: eHSP90α has a better prognostic value in LUAD L858R patients than 19DEL, which provides a new idea for clinical diagnosis and treatment.


Sujet(s)
Adénocarcinome pulmonaire , Marqueurs biologiques tumoraux , Récepteurs ErbB , Exons , Protéines du choc thermique HSP90 , Tumeurs du poumon , Mutation , Humains , Protéines du choc thermique HSP90/génétique , Femelle , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Mâle , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Pronostic , Récepteurs ErbB/génétique , Exons/génétique , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Délétion de séquence , Adulte
18.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999930

RÉSUMÉ

Although SARS-CoV-2 induces mucin hypersecretion in the respiratory tract, hyposalivation/xerostomia has been reported by COVID-19 patients. We evaluate the submandibular gland (SMGs) pathogenesis in SARS-CoV-2-infected K18-hACE2 mice, focusing on the impact of infection on the mucin production and structural integrity of acini, ductal system, myoepithelial cells (MECs) and telocytes. The spike protein, the nucleocapsid protein, hACE2, actin, EGF, TNF-α and IL-1ß were detected by immunofluorescence, and the Egfr and Muc5b expression was evaluated. In the infected animals, significant acinar hypertrophy was observed in contrast to ductal atrophy. Nucleocapsid proteins and/or viral particles were detected in the SMG cells, mainly in the nuclear membrane-derived vesicles, confirming the nuclear role in the viral formation. The acinar cells showed intense TNF-α and IL-1ß immunoexpression, and the EGF-EGFR signaling increased, together with Muc5b upregulation. This finding explains mucin hypersecretion and acinar hypertrophy, which compress the ducts. Dying MECs and actin reduction were also observed, indicating failure of contraction and acinar support, favoring acinar hypertrophy. Viral assembly was found in the dying telocytes, pointing to these intercommunicating cells as viral transmitters in SMGs. Therefore, EGF-EGFR-induced mucin hypersecretion was triggered by SARS-CoV-2 in acinar cells, likely mediated by cytokines. The damage to telocytes and MECs may have favored the acinar hypertrophy, leading to ductal obstruction, explaining xerostomia in COVID-19 patients. Thus, acinar cells, telocytes and MECs may be viral targets, which favor replication and cell-to-cell viral transmission in the SMG, corroborating the high viral load in saliva of infected individuals.


Sujet(s)
COVID-19 , Récepteurs ErbB , SARS-CoV-2 , Glande submandibulaire , Xérostomie , COVID-19/anatomopathologie , COVID-19/virologie , COVID-19/métabolisme , Animaux , Glande submandibulaire/virologie , Glande submandibulaire/anatomopathologie , Glande submandibulaire/métabolisme , SARS-CoV-2/physiologie , Souris , Xérostomie/étiologie , Xérostomie/anatomopathologie , Xérostomie/virologie , Xérostomie/métabolisme , Récepteurs ErbB/métabolisme , Humains , Angiotensin-converting enzyme 2/métabolisme , Mucine 5B/métabolisme , Cellules acineuses/anatomopathologie , Cellules acineuses/métabolisme , Cellules acineuses/virologie , Interleukine-1 bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Modèles animaux de maladie humaine
19.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000238

RÉSUMÉ

Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. Therefore, the need for new therapeutic strategies is still a challenge. Surgery and chemotherapy represent the first-line interventions; nevertheless, the prognosis for metastatic CRC (mCRC) patients remains unacceptable. An important step towards targeted therapy came from the inhibition of the epidermal growth factor receptor (EGFR) pathway, by the anti-EGFR antibody, Cetuximab, or by specific tyrosine kinase inhibitors (TKI). Cetuximab, a mouse-human chimeric monoclonal antibody (mAb), binds to the extracellular domain of EGFR thus impairing EGFR-mediated signaling and reducing cell proliferation. TKI can affect the EGFR biochemical pathway at different steps along the signaling cascade. Apart from Cetuximab, other anti-EGFR mAbs have been developed, such as Panitumumab. Both antibodies have been approved for the treatment of KRAS-NRAS wild type mCRC, alone or in combination with chemotherapy. These antibodies display strong differences in activating the host immune system against CRC, due to their different immunoglobulin isotypes. Although anti-EGFR antibodies are efficient, drug resistance occurs with high frequency. Resistant tumor cell populations can either already be present before therapy or develop later by biochemical adaptations or new genomic mutations in the EGFR pathway. Numerous efforts have been made to improve the efficacy of the anti-EGFR mAbs or to find new agents that are able to block downstream EGFR signaling cascade molecules. Indeed, we examined the importance of analyzing the anti-EGFR antibody-drug conjugates (ADC) developed to overcome resistance and/or stimulate the tumor host's immunity against CRC growth. Also, patient-derived CRC organoid cultures represent a useful and feasible in vitro model to study tumor behavior and therapy response. Organoids can reflect tumor genetic heterogeneity found in the tissue of origin, representing a unique tool for personalized medicine. Thus, CRC-derived organoid cultures are a smart model for studying the tumor microenvironment and for the preclinical assay of anti-EGFR drugs.


Sujet(s)
Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Organoïdes , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Organoïdes/métabolisme , Organoïdes/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Cétuximab/pharmacologie , Cétuximab/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Thérapie moléculaire ciblée/méthodes , Panitumumab/pharmacologie , Panitumumab/usage thérapeutique , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques
20.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000275

RÉSUMÉ

In tumor cells, interleukin-6 (IL-6) signaling can lead to activation of the epidermal growth factor receptor (EGFR), which prolongs Stat3 activation. In the present experiments, we tested the hypothesis that IL-6 signaling activates EGFR signaling in peripheral and spinal nociception and examined whether EGFR localization and activation coincide with pain-related behaviors in arthritis. In vivo in anesthetized rats, spinal application of the EGFR receptor blocker gefitinib reduced the responses of spinal cord neurons to noxious joint stimulation, but only after spinal pretreatment with IL-6 and soluble IL-6 receptor. Using Western blots, we found that IL-6-induced Stat3 activation was reduced by gefitinib in microglial cells of the BV2 cell line, but not in cultured DRG neurons. Immunohistochemistry showed EGFR localization in most DRG neurons from normal rats, but significant downregulation in the acute and most painful arthritis phase. In the spinal cord of mice, EGFR was highly activated mainly in the chronic phase of inflammation, with localization in neurons. These data suggest that spinal IL-6 signaling may activate spinal EGFR signaling. Downregulation of EGFR in DRG neurons in acute arthritis may limit nociception, but pronounced delayed activation of EGFR in the spinal cord may be involved in chronic inflammatory pain.


Sujet(s)
Récepteurs ErbB , Interleukine-6 , Cellules réceptrices sensorielles , Moelle spinale , Animaux , Récepteurs ErbB/métabolisme , Interleukine-6/métabolisme , Moelle spinale/métabolisme , Rats , Souris , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Mâle , Transduction du signal , Facteur de transcription STAT-3/métabolisme , Arthrite/métabolisme , Géfitinib/pharmacologie , Récepteurs à l'interleukine-6/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Arthrite expérimentale/métabolisme , Lignée cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...