Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.423
Filtrer
1.
Drug Des Devel Ther ; 18: 2653-2679, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974119

RÉSUMÉ

Purpose: Over the last few years, covalent fragment-based drug discovery has gained significant importance. Thus, striving for more warhead diversity, we conceived a library consisting of 20 covalently reacting compounds. Our covalent fragment library (CovLib) contains four different warhead classes, including five α-cyanoacacrylamides/acrylates (CA), three epoxides (EO), four vinyl sulfones (VS), and eight electron-deficient heteroarenes with a leaving group (SNAr/SN). Methods: After predicting the theoretical solubility of the fragments by LogP and LogS during the selection process, we determined their experimental solubility using a turbidimetric solubility assay. The reactivities of the different compounds were measured in a high-throughput 5,5'-dithiobis-(2-nitrobenzoic acid) DTNB assay, followed by a (glutathione) GSH stability assay. We employed the CovLib in a (differential scanning fluorimetry) DSF-based screening against different targets: c-Jun N-terminal kinase 3 (JNK3), ubiquitin-specific protease 7 (USP7), and the tumor suppressor p53. Finally, the covalent binding was confirmed by intact protein mass spectrometry (MS). Results: In general, the purchased fragments turned out to be sufficiently soluble. Additionally, they covered a broad spectrum of reactivity. All investigated α-cyanoacrylamides/acrylates and all structurally confirmed epoxides turned out to be less reactive compounds, possibly due to steric hindrance and reversibility (for α-cyanoacrylamides/acrylates). The SNAr and vinyl sulfone fragments are either highly reactive or stable. DSF measurements with the different targets JNK3, USP7, and p53 identified reactive fragment hits causing a shift in the melting temperatures of the proteins. MS confirmed the covalent binding mode of all these fragments to USP7 and p53, while additionally identifying the SNAr-type electrophile SN002 as a mildly reactive covalent hit for p53. Conclusion: The screening and target evaluation of the CovLib revealed first interesting hits. The highly cysteine-reactive fragments VS004, SN001, SN006, and SN007 covalently modify several target proteins and showed distinct shifts in the melting temperatures up to +5.1 °C and -9.1 °C.


Sujet(s)
Mitogen-Activated Protein Kinase 10 , Protéine p53 suppresseur de tumeur , Ubiquitin-specific peptidase 7 , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/composition chimique , Ubiquitin-specific peptidase 7/antagonistes et inhibiteurs , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/composition chimique , Humains , Mitogen-Activated Protein Kinase 10/métabolisme , Mitogen-Activated Protein Kinase 10/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase 10/composition chimique , Sulfones/composition chimique , Sulfones/pharmacologie , Structure moléculaire , Solubilité , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Relation structure-activité , Acrylamides/composition chimique , Acrylamides/pharmacologie , Acrylates/composition chimique , Acrylates/pharmacologie , Liaison aux protéines
2.
Bioorg Med Chem ; 109: 117796, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38879996

RÉSUMÉ

The USFDA granted regular approval to Osimertinib (AZD9291) on March 2017, for treating individuals with metastatic Non-Small Cell Lung Cancer having EGFR T790M mutation. Clinically, Osimertinib stands at the forefront for the treatment of patients with Non-Small Cell Lung Cancer. Osimertinib forms a covalent bond with the Cys797 residue and predominantly spares binding to WT-EGFR, thereby reducing toxicity and enabling the administration of doses that effectively inhibit T790M. However, a high percentage of patients treated with Osimertinib (AZD9291) developed a tertiary cysteine797 to serine797 (C797S) mutation in the EGFR kinase domain, rendering resistance to it. This comprehensive review sheds light on the chemistry, computational aspects, structural features, and expansive spectrum of biological activities of Osimertinib and its analogues. The in-depth exploration of these facets serves as a valuable resource for medicinal chemists, empowering them to design better Osimertinib analogues. This exhaustive study not only provides insights into improving potency but also emphasizes considerations for mutant selectivity and optimizing pharmacokinetic properties. This review acts as a guiding beacon for the strategic design and development of next-generation Osimertinib analogues.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Inhibiteurs de protéines kinases , Acrylamides/composition chimique , Acrylamides/pharmacologie , Dérivés de l'aniline/composition chimique , Dérivés de l'aniline/pharmacologie , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Relation structure-activité , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Structure moléculaire , Pipérazines/composition chimique , Pipérazines/pharmacologie , Indoles , Pyrimidines
3.
Mar Drugs ; 22(6)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38921583

RÉSUMÉ

The marine environment provides a rich source of distinct creatures containing potentially revolutionary bioactive chemicals. One of these organisms is Caulerpa racemosa, a type of green algae known as green seaweed, seagrapes, or green caviar. This organism stands out because it has great promise for use in medicine, especially in the study of cancer. Through the utilization of computational modeling (in silico) and cellular laboratory experiments (in vitro), the chemical components included in the green seaweed C. racemosa were effectively analyzed, uncovering its capability to treat non-small cell lung cancer (NSCLC). The study specifically emphasized blocking SRC, STAT3, PIK3CA, MAPK1, EGFR, and JAK1 using molecular docking and in vitro. These proteins play a crucial role in the EGFR Tyrosine Kinase Inhibitor Resistance pathway in NSCLC. The chemical Caulersin (C2) included in C. racemosa extract (CRE) has been identified as a potent and effective agent in fighting against non-small cell lung cancer (NSCLC), both in silico and in vitro. CRE and C2 showed a level of inhibition similar to that of osimertinib (positive control/NSCLC drug).


Sujet(s)
Carcinome pulmonaire non à petites cellules , Caulerpa , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Simulation de docking moléculaire , Pharmacologie des réseaux , Inhibiteurs de protéines kinases , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Caulerpa/composition chimique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Algue marine/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Récepteurs ErbB/antagonistes et inhibiteurs , Acrylamides/pharmacologie , Acrylamides/composition chimique
4.
PLoS One ; 19(6): e0304914, 2024.
Article de Anglais | MEDLINE | ID: mdl-38935790

RÉSUMÉ

Lung cancer is one of the most common and deadliest cancers. Preclinical models are essential to study new therapies and combinations taking tumor genetics into account. We have established cell lines expressing the luciferase gene from lines with varied genetic backgrounds, commonly encountered in patients with pulmonary adenocarcinoma. We have characterized these lines by testing their response to multiple drugs. Thus, we have developed orthotopic preclinical mouse models of NSCLC with very high engraftment efficiency. These models allow the easy monitoring of tumor growth, particularly in response to treatment, and of tumor cells dissemination in the body. We show that concomitant treatment with osimertinib (3rd generation tyrosine kinase inhibitor targeting mutated EGFR) and bevacizumab (anti-angiogenic targeting VEGF) can have a beneficial therapeutic effect on EGFR-mutated tumors. We also show that the addition of afatinib to osimertinib-treated tumors in escape leads to tumor growth inhibition. No such effect is observed with selumetinib or simvastatin. These preclinical mouse models therefore make it possible to test innovative therapeutic combinations and are also a tool of choice for studying resistance mechanisms.


Sujet(s)
Acrylamides , Afatinib , Dérivés de l'aniline , Bévacizumab , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Animaux , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Acrylamides/pharmacologie , Afatinib/pharmacologie , Afatinib/usage thérapeutique , Bévacizumab/pharmacologie , Bévacizumab/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Souris , Humains , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Modèles animaux de maladie humaine , Tests d'activité antitumorale sur modèle de xénogreffe , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Quinazolines/administration et posologie , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Pipérazines/administration et posologie , Femelle , Indoles , Pyrimidines
5.
ESMO Open ; 9(6): 103592, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38878323

RÉSUMÉ

BACKGROUND: Osimertinib represents the standard of care for the treatment of advanced non-small-cell lung cancer (NSCLC) harboring classical epidermal growth factor receptor (EGFR) mutations, constituting 80%-90% of all EGFR alterations. In the remaining cases, an assorted group of uncommon alterations of EGFR (uEGFR) can be detected, which confer variable sensitivity to previous generations of EGFR inhibitors, overall with lower therapeutic activity. Data on osimertinib in this setting are limited and strongly warranted. PATIENTS AND METHODS: The ARTICUNO study retrospectively evaluated data on osimertinib activity from patients with advanced NSCLC harboring uEGFR treated in 21 clinical centers between August 2017 and March 2023. Data analysis was carried out with a descriptive aim. Investigators collected response data according to RECIST version 1.1 criteria. The median duration of response, progression-free survival (mPFS), and overall survival were estimated by the Kaplan-Meier method. RESULTS: Eighty-six patients harboring uEGFR and treated with osimertinib were identified. Patients with 'major' uEGFR, that is, G719X, L861X, and S768I mutations (n = 51), had an overall response rate (ORR) and mPFS of 50% and 9 months, respectively. Variable outcomes were registered in cases with rarer 'minor' mutations (n = 27), with ORR and mPFS of 31% and 4 months, respectively. Among seven patients with exon 20 insertions, ORR was 14%, while the best outcome was registered among patients with compound mutations including at least one classical EGFR mutation (n = 13). Thirty patients presented brain metastases (BMs) and intracranial ORR and mPFS were 58% and 9 months, respectively. Amplification of EGFR or MET, TP53 mutations, and EGFR E709K emerged after osimertinib failure in a dataset of 18 patients with available rebiopsy. CONCLUSION: The ARTICUNO study confirms the activity of osimertinib in patients with uEGFR, especially in those with compound uncommon-common mutations, or major uEGFR, even in the presence of BMs. Alterations at the E709 residue of EGFR are associated with resistance to osimertinib.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Études rétrospectives , Acrylamides/usage thérapeutique , Acrylamides/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Récepteurs ErbB/génétique , Dérivés de l'aniline/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Sujet âgé de 80 ans ou plus , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Indoles , Pyrimidines
6.
Biomacromolecules ; 25(7): 4192-4202, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38917475

RÉSUMÉ

The permeability and responsiveness of polymer membranes are absolutely relevant in the design of polymersomes for cargo delivery. Accordingly, we herein correlate the structural features, permeability, and responsiveness of doxorubicin-loaded (DOX-loaded) nonresponsive and stimuli-responsive polymersomes with their in vitro and in vivo antitumor performance. Polymer vesicles were produced using amphiphilic block copolymers containing a hydrophilic poly[N-(2-hydroxypropyl)methacrylamide] (PHPMA) segment linked to poly[N-(4-isopropylphenylacetamide)ethyl methacrylate] (PPPhA, nonresponsive block), poly[4-(4,4,5,5-tetra-methyl-1,3,2-dioxaborolan-2-yl)benzyl methacrylate] [PbAPE, reactive oxygen species (ROS)-responsive block], or poly[2-(diisopropylamino)ethyl methacrylate] (PDPA, pH-responsive block). The PDPA-based polymersomes demonstrated outstanding biological performance with antitumor activity notably enhanced compared to their counterparts. We attribute this behavior to a fast-triggered DOX release in acidic tumor environments as induced by pH-responsive polymersome disassembly at pH < 6.8. Possibly, an insufficient ROS concentration in the selected tumor model attenuates the rate of ROS-responsive vesicle degradation, whereas the nonresponsive nature of the PPPhA block remarkably impacts the performance of such potential nanomedicines.


Sujet(s)
Doxorubicine , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Humains , Animaux , Souris , Perméabilité des membranes cellulaires/effets des médicaments et des substances chimiques , Polymères/composition chimique , Polymères/pharmacologie , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Vecteurs de médicaments/composition chimique , Espèces réactives de l'oxygène/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Acrylamides/composition chimique , Acrylamides/pharmacologie , Concentration en ions d'hydrogène
7.
Bioorg Med Chem Lett ; 107: 129779, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38729317

RÉSUMÉ

Targeted protein degradation is mediated by small molecules that induce or stabilize protein-protein interactions between targets and the ubiquitin-proteasome machinery. Currently, there remains a need to expand the repertoire of viable E3 ligases available for hijacking. Notably, covalent chemistry has been employed to engage a handful of E3 ligases, including DCAF11. Here, we disclose a covalent PROTAC that enables DCAF11-dependent degradation, featuring a cyanoacrylamide warhead. Our findings underscore DCAF11 as an interesting candidate with a capacity to accommodate diverse electrophilic chemistries compatible with targeted protein degradation.


Sujet(s)
Acrylamides , Humains , Acrylamides/composition chimique , Acrylamides/pharmacologie , Acrylamides/synthèse chimique , Structure moléculaire , Protéolyse/effets des médicaments et des substances chimiques , Découverte de médicament , Ubiquitin-activating enzymes/métabolisme , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Relation structure-activité
8.
Mol Cancer ; 23(1): 91, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38715012

RÉSUMÉ

BACKGROUND: Recent evidence has demonstrated that abnormal expression and regulation of circular RNA (circRNAs) are involved in the occurrence and development of a variety of tumors. The aim of this study was to investigate the effects of circ_PPAPDC1A in Osimertinib resistance in NSCLC. METHODS: Human circRNAs microarray analysis was conducted to identify differentially expressed (DE) circRNAs in Osimertinib-acquired resistance tissues of NSCLC. The effect of circ_PPAPDC1A on cell proliferation, invasion, migration, and apoptosis was assessed in both in vitro and in vivo. Dual-luciferase reporter assay, RT-qPCR, Western-blot, and rescue assay were employed to confirm the interaction between circ_PPAPDC1A/miR-30a-3p/IGF1R axis. RESULTS: The results revealed that circ_PPAPDC1A was significantly upregulated in Osimertinib acquired resistance tissues of NSCLC. circ_PPAPDC1A reduced the sensitivity of PC9 and HCC827 cells to Osimertinib and promoted cell proliferation, invasion, migration, while inhibiting apoptosis in Osimertinib-resistant PC9/OR and HCC829/OR cells, both in vitro and in vivo. Silencing circ_PPAPDC1A partially reversed Osimertinib resistance. Additionally, circ_PPAPDC1A acted as a competing endogenous RNA (ceRNA) by targeting miR-30a-3p, and Insulin-like Growth Factor 1 Receptor (IGF1R) was identified as a functional gene for miR-30a-3p in NSCLC. Furthermore, the results confirmed that circ_PPAPDC1A/miR-30a-3p/IGF1R axis plays a role in activating the PI3K/AKT/mTOR signaling pathway in NSCLC with Osimertinib resistance. CONCLUSIONS: Therefore, for the first time we identified that circ_PPAPDC1A was significantly upregulated and exerts an oncogenic role in NSCLC with Osimertinib resistance by sponging miR-30a-3p to active IGF1R/PI3K/AKT/mTOR pathway. circ_PPAPDC1A may serve as a novel diagnostic biomarker and therapeutic target for NSCLC patients with Osimertinib resistance.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , microARN , ARN circulaire , Récepteur IGF de type 1 , Transduction du signal , Humains , microARN/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Récepteur IGF de type 1/génétique , Récepteur IGF de type 1/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Acrylamides/pharmacologie , ARN circulaire/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Dérivés de l'aniline/pharmacologie , Lignée cellulaire tumorale , Animaux , Souris , Apoptose , Mouvement cellulaire/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Femelle , Indoles , Pyrimidines
9.
Bioorg Chem ; 147: 107394, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38691906

RÉSUMÉ

Epidermal growth factor receptor (EGFR) is one of the most studied drug targets for treating non-small-cell lung cancer (NSCLC). However, there are no approved inhibitors for the C797S resistance mutation caused by the third-generation EGFR inhibitor (Osimertinib). Therefore, the development of fourth-generation EGFR inhibitors is urgent. In this study, we clarified the structure-activity relationship of several synthesized compounds as fourth-generation inhibitors against human triple (Del19/T790M/C797S) mutation. Representative compound 52 showed potent inhibitory activity against EGFRL858R/T790M/C797S with an IC50 of 0.55 nM and significantly inhibited the proliferation of the Ba/F3 cell line harboring EGFRL858R/T790M/C797S with an IC50 of 43.28 nM. Moreover, 52 demonstrated good pharmacokinetic properties and excellent in vivo efficacy. Overall, the compound 52 can be considered a promising candidate for overcoming EGFR C797S-mediated mutations.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Résistance aux médicaments antinéoplasiques , Tests de criblage d'agents antitumoraux , Récepteurs ErbB , Tumeurs du poumon , Inhibiteurs de protéines kinases , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Acrylamides/pharmacologie , Acrylamides/composition chimique , Acrylamides/synthèse chimique , Relation structure-activité , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/composition chimique , Dérivés de l'aniline/synthèse chimique , Dérivés de l'aniline/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Structure moléculaire , Animaux , Souris , Lignée cellulaire tumorale , Mutation , Indoles , Pyrimidines
10.
J Med Chem ; 67(10): 8099-8121, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38722799

RÉSUMÉ

Nicotinamide phosphoribosyltransferase (NAMPT) is an attractive therapeutic target for treating select cancers. There are two forms of NAMPT: intracellular NAMPT (iNAMPT, the rate-limiting enzyme in the mammalian NAD+ main synthetic pathway) and extracellular NAMPT (eNAMPT, a cytokine with protumorigenic function). Reported NAMPT inhibitors only inhibit iNAMPT and show potent activities in preclinical studies. Unfortunately, they failed to show efficacy due to futility and toxicity. We developed a series of FK866-based NAMPT-targeting PROTACs and identified LYP-8 as a potent and effective NAMPT degrader that simultaneously diminished iNAMPT and eNAMPT. Importantly, LYP-8 demonstrated superior efficacy and safety in mice when compared to the clinical candidate, FK866. This study highlights the importance and feasibility of applying PROTACs as a superior strategy for interfering with both the enzymatic function of NAMPT (iNAMPT) and nonenzymatic function of NAMPT (eNAMPT), which is difficult to achieve with conventional NAMPT inhibitors.


Sujet(s)
Acrylamides , Conception de médicament , Nicotinamide phosphoribosyltransferase , Pipéridines , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Nicotinamide phosphoribosyltransferase/métabolisme , Acrylamides/pharmacologie , Acrylamides/composition chimique , Acrylamides/synthèse chimique , Animaux , Humains , Pipéridines/pharmacologie , Pipéridines/composition chimique , Souris , Antienzymes/pharmacologie , Antienzymes/composition chimique , Cytokines/métabolisme , Lignée cellulaire tumorale , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
11.
Nat Commun ; 15(1): 4195, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760351

RÉSUMÉ

Osimertinib (Osi) is a widely used epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). However, the emergence of resistance is inevitable, partly due to the gradual evolution of adaptive resistant cells during initial treatment. Here, we find that Osi treatment rapidly triggers adaptive resistance in tumor cells. Metabolomics analysis reveals a significant enhancement of oxidative phosphorylation (OXPHOS) in Osi adaptive-resistant cells. Mechanically, Osi treatment induces an elevation of NCOA4, a key protein of ferritinophagy, which maintains the synthesis of iron-sulfur cluster (ISC) proteins of electron transport chain and OXPHOS. Additionally, active ISC protein synthesis in adaptive-resistant cells significantly increases the sensitivity to copper ions. Combining Osi with elesclomol, a copper ion ionophore, significantly increases the efficacy of Osi, with no additional toxicity. Altogether, this study reveals the mechanisms of NCOA4-mediated ferritinophagy in Osi adaptive resistance and introduces a promising new therapy of combining copper ionophores to improve its initial efficacy.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Ferritines , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Lignée cellulaire tumorale , Ferritines/métabolisme , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Animaux , Souris , Cuivre/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Souris nude , Indoles , Pyrimidines
12.
Eur J Pharm Sci ; 199: 106805, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38763450

RÉSUMÉ

Drug resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is a primary factor affecting their therapeutic efficacy in human non-small cell lung cancer (NSCLC). NSCLC cells can undergo epithelial-mesenchymal transition (EMT) induced by many factors in the tumour microenvironment (TME), which plays a crucial role in tumour drug resistance. In this study, a multicellular lung-on-a-chip that can realise the cell co-culture of the human non-small cell lung cancer cell line HCC827, human foetal lung fibroblasts (HFL-1), and human umbilical vein endothelial cells (HUVECs) is prepared. The TME was simulated on the chip combined with perfusion and other factors, and the drug evaluation of osimertinib was performed to explore the drug resistance mechanism of EGFR-TKIs. In the early stages, a two-dimensional static cell co-culture was achieved by microchip, and the results showed that HFL-1 cells could be transformed into cancer-associated fibroblasts (CAFs), and HCC827 cells could undergo EMT, both of which were mediated by Interleukin-6 (IL-6). Vimentin (VIM) and Alpha Skeletal Muscle Actin (a-SMA) expression of HFL-1 was upregulated, whereas E-cadherin (E-cad) expression of HCC827 was down-regulated. Further, N-cadherin (N-cad) expression of HCC827 was upregulated. In both the static cell co-culture and multicellular lung-on-a-chip, HCC827 cells with CAFs co-culture or IL-6 treatment developed resistance to osimertinib. Further use of the IL-6 antibody inhibitor tocilizumab could reverse EGFR-TKI resistance to a certain extent. Combination therapy with tocilizumab and EGFR-TKIs may provide a novel therapeutic strategy for overcoming EGFR-TKI resistance caused by EMT in NSCLC. Furthermore, the lung-on-a-chip can simulate complex TME and can be used for evaluating tumour resistance and exploring mechanisms, with the potential to become an important tool for personalised diagnosis, treatment, and biomedical research.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , Récepteurs ErbB , Cellules endothéliales de la veine ombilicale humaine , Laboratoires sur puces , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Antinéoplasiques/pharmacologie , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Lignée cellulaire tumorale , Techniques de coculture , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Indoles , Interleukine-6/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines , Microenvironnement tumoral/effets des médicaments et des substances chimiques
13.
Cancer Invest ; 42(5): 425-434, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38818695

RÉSUMÉ

Addressing recurrent cervical cancer poses a substantial challenge. Osimertinib, an FDA-approved EGFR inhibitor, has emerged as a promising option. Our study examined its potential to enhance paclitaxel's efficacy against cervical cancer. Osimertinib effectively hindered cancer cell growth and induced apoptosis across multiple cell lines. Combined with paclitaxel, it exhibited synergy in suppressing cervical cancer cells. Importantly, osimertinib's inhibitory effect was EGFR-independent; it targeted Mnk phosphorylation, reducing eIF4E activity. In mice, the combined osimertinib-paclitaxel treatment surpassed individual drugs in inhibiting cancer growth. These preclinical findings suggest osimertinib's repurposing as a means to improve paclitaxel's effectiveness in cervical cancer treatment.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Paclitaxel , Tumeurs du col de l'utérus , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Humains , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Femelle , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Animaux , Souris , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Lignée cellulaire tumorale , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Indoles , Pyrimidines
14.
Respir Res ; 25(1): 215, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38764025

RÉSUMÉ

BACKGROUND: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of lung cancer patients with mutated EGFR. However, the efficacy of EGFR-TKIs in wild-type EGFR tumors has been shown to be marginal. Methods that can sensitize EGFR-TKIs to EGFR wild-type NSCLC remain rare. Hence, we determined whether combination treatment can maximize the therapeutic efficacy of EGFR-TKIs. METHODS: We established a focused drug screening system to investigate candidates for overcoming the intrinsic resistance of wild-type EGFR NSCLC to EGFR-TKIs. Molecular docking assays and western blotting were used to identify the binding mode and blocking effect of the candidate compounds. Proliferation assays, analyses of drug interactions, colony formation assays, flow cytometry and nude mice xenograft models were used to determine the effects and investigate the molecular mechanism of the combination treatment. RESULTS: Betulinic acid (BA) is effective at targeting EGFR and synergizes with EGFR-TKIs (gefitinib and osimertinib) preferentially against wild-type EGFR. BA showed inhibitory activity due to its interaction with the ATP-binding pocket of EGFR and dramatically enhanced the suppressive effects of EGFR-TKIs by blocking EGFR and modulating the EGFR-ATK-mTOR axis. Mechanistic studies revealed that the combination strategy activated EGFR-induced autophagic cell death and that the EGFR-AKT-mTOR signaling pathway was essential for completing autophagy and cell cycle arrest. Activation of the mTOR pathway or blockade of autophagy by specific chemical agents markedly attenuated the effect of cell cycle arrest. In vivo administration of the combination treatment caused marked tumor regression in the A549 xenografts. CONCLUSIONS: BA is a potential wild-type EGFR inhibitor that plays a critical role in sensitizing EGFR-TKI activity. BA combined with an EGFR-TKI effectively suppressed the proliferation and survival of intrinsically resistant lung cancer cells via the inhibition of EGFR as well as the induction of autophagy-related cell death, indicating that BA combined with an EGFR-TKI may be a potential therapeutic strategy for overcoming the primary resistance of wild-type EGFR-positive lung cancers.


Sujet(s)
Autophagie , Acide bétulinique , Carcinome pulmonaire non à petites cellules , Synergie des médicaments , Récepteurs ErbB , Tumeurs du poumon , Triterpènes pentacycliques , Inhibiteurs de protéines kinases , Animaux , Humains , Souris , Cellules A549 , Acrylamides/pharmacologie , Dérivés de l'aniline/pharmacologie , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Lignée cellulaire tumorale , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Géfitinib/pharmacologie , Indoles , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Souris de lignée BALB C , Souris nude , Simulation de docking moléculaire , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines , Transduction du signal/effets des médicaments et des substances chimiques , Triterpènes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
15.
Rev Mal Respir ; 41(6): 451-454, 2024 Jun.
Article de Français | MEDLINE | ID: mdl-38796386

RÉSUMÉ

Targeted therapies are the standard first-line treatment for metastatic lung adenocarcinoma with certain molecular abnormalities. These abnormalities are particularly common in Southeast Asia and French Polynesia. A 51-year-old Tahitian female non-smoker was diagnosed in 2018 with stage IV lung adenocarcinoma harboring a p.L858R EGFR mutation. She received gefitinib as first-line treatment. Due to locoregional progression and the presence of a resistance mutation (p.T790M of EFGR), she received osimertinib as second-line treatment, after which chemotherapy was proposed as 3rd-line treatment. An additional biopsy detected not only the previously known EGFR mutation, but also a BRAF p.V600E mutation. Following disease progression during chemotherapy, the patient received targeted therapies combining dabrafenib, trametinib and osimertinib. Due to a dissociated response after four months of treatment, a 5th line of paclitaxel bevacizumab was initiated. Subsequent to additional progression and given the ALK rearrangement shown on the re-biopsy, 6th-line treatment with alectinib was proposed. As the response was once again dissociated, a final line was proposed before stopping active treatments due to their toxicity and overall deterioration in the patient's state of health. This exceptional case is characterized by resistance to anti-EGFR through the successive and cumulative acquisition of two new oncogene addictions. The authors underline the importance of re-biopsy at each progression, leading (if at all feasible) to yet around round of targeted therapy.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Dépendance oncogène , Femelle , Humains , Adulte d'âge moyen , Acrylamides/usage thérapeutique , Acrylamides/pharmacologie , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Dérivés de l'aniline/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Récepteurs ErbB/génétique , Géfitinib/usage thérapeutique , Géfitinib/pharmacologie , Indoles , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Mutation , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/génétique , Pyrimidines
16.
BMC Med ; 22(1): 174, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38658988

RÉSUMÉ

BACKGROUND: Osimertinib has become standard care for epidermal growth factor receptor (EGFR)-positive non-small cell lung cancer (NSCLC) patients whereas drug resistance remains inevitable. Now we recognize that the interactions between the tumor and the tumor microenvironment (TME) also account for drug resistance. Therefore, we provide a new sight into post-osimertinib management, focusing on the alteration of TME. METHODS: We conducted a retrospective study on the prognosis of different treatments after osimertinib resistance. Next, we carried out in vivo experiment to validate our findings using a humanized mouse model. Furthermore, we performed single-cell transcriptome sequencing (scRNA-seq) of tumor tissue from the above treatment groups to explore the mechanisms of TME changes. RESULTS: Totally 111 advanced NSCLC patients have been enrolled in the retrospective study. The median PFS was 9.84 months (95% CI 7.0-12.6 months) in the osimertinib plus anti-angiogenesis group, significantly longer than chemotherapy (P = 0.012) and osimertinib (P = 0.003). The median OS was 16.79 months (95% CI 14.97-18.61 months) in the osimertinib plus anti-angiogenesis group, significantly better than chemotherapy (P = 0.026), the chemotherapy plus osimertinib (P = 0.021), and the chemotherapy plus immunotherapy (P = 0.006). The efficacy of osimertinib plus anlotinib in the osimertinib-resistant engraft tumors (R-O+A) group was significantly more potent than the osimertinib (R-O) group (P<0.05) in vitro. The combinational therapy could significantly increase the infiltration of CD4+ T cells (P<0.05), CD25+CD4+ T cells (P<0.001), and PD-1+CD8+ T cells (P<0.05) compared to osimertinib. ScRNA-seq demonstrated that the number of CD8+ T and proliferation T cells increased, and TAM.mo was downregulated in the R-O+A group compared to the R-O group. Subtype study of T cells explained that the changes caused by combination treatment were mainly related to cytotoxic T cells. Subtype study of macrophages showed that proportion and functional changes in IL-1ß.mo and CCL18.mo might be responsible for rescue osimertinib resistance by combination therapy. CONCLUSIONS: In conclusion, osimertinib plus anlotinib could improve the prognosis of patients with a progressed disease on second-line osimertinib treatment, which may ascribe to increased T cell infiltration and TAM remodeling via VEGF-VEGFR blockage.


Sujet(s)
Acrylamides , Inhibiteurs de l'angiogenèse , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Pyrimidines , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Dérivés de l'aniline/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Acrylamides/usage thérapeutique , Acrylamides/pharmacologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Études rétrospectives , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Mâle , Animaux , Souris , Adulte d'âge moyen , Inhibiteurs de l'angiogenèse/usage thérapeutique , Inhibiteurs de l'angiogenèse/administration et posologie , Sujet âgé , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Adulte , Indoles/usage thérapeutique , Indoles/administration et posologie
17.
Commun Biol ; 7(1): 497, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38658677

RÉSUMÉ

Most lung cancer patients with metastatic cancer eventually relapse with drug-resistant disease following treatment and EGFR mutant lung cancer is no exception. Genome-wide CRISPR screens, to either knock out or overexpress all protein-coding genes in cancer cell lines, revealed the landscape of pathways that cause resistance to the EGFR inhibitors osimertinib or gefitinib in EGFR mutant lung cancer. Among the most recurrent resistance genes were those that regulate the Hippo pathway. Following osimertinib treatment a subpopulation of cancer cells are able to survive and over time develop stable resistance. These 'persister' cells can exploit non-genetic (transcriptional) programs that enable cancer cells to survive drug treatment. Using genetic and pharmacologic tools we identified Hippo signalling as an important non-genetic mechanism of cell survival following osimertinib treatment. Further, we show that combinatorial targeting of the Hippo pathway and EGFR is highly effective in EGFR mutant lung cancer cells and patient-derived organoids, suggesting a new therapeutic strategy for EGFR mutant lung cancer patients.


Sujet(s)
Acrylamides , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Indoles , Tumeurs du poumon , Mutation , Pyrimidines , Facteurs de transcription , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lignée cellulaire tumorale , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Protéines de signalisation YAP/métabolisme , Protéines de signalisation YAP/génétique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Géfitinib/pharmacologie , Voie de signalisation Hippo , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Transduction du signal , Facteurs de transcription à domaine TEA , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/pharmacologie , Clustered regularly interspaced short palindromic repeats , Systèmes CRISPR-Cas
18.
Proc Natl Acad Sci U S A ; 121(19): e2315597121, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38687786

RÉSUMÉ

Snakebite envenoming is a neglected tropical disease that causes substantial mortality and morbidity globally. The venom of African spitting cobras often causes permanent injury via tissue-destructive dermonecrosis at the bite site, which is ineffectively treated by current antivenoms. To address this therapeutic gap, we identified the etiological venom toxins in Naja nigricollis venom responsible for causing local dermonecrosis. While cytotoxic three-finger toxins were primarily responsible for causing spitting cobra cytotoxicity in cultured keratinocytes, their potentiation by phospholipases A2 toxins was essential to cause dermonecrosis in vivo. This evidence of probable toxin synergism suggests that a single toxin-family inhibiting drug could prevent local envenoming. We show that local injection with the repurposed phospholipase A2-inhibiting drug varespladib significantly prevents local tissue damage caused by several spitting cobra venoms in murine models of envenoming. Our findings therefore provide a therapeutic strategy that may effectively prevent life-changing morbidity caused by snakebite in rural Africa.


Sujet(s)
Acétates , Venins des élapidés , Indoles , Cétoacides , Nécrose , Morsures de serpent , Animaux , Morsures de serpent/traitement médicamenteux , Souris , Humains , Acrylamides/pharmacologie , Phospholipases A2/métabolisme , Naja , Elapidae , Kératinocytes/effets des médicaments et des substances chimiques , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Repositionnement des médicaments
19.
Biochem Pharmacol ; 224: 116207, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621425

RÉSUMÉ

Osimertinib is a novel epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), acting as the first-line medicine for advanced EGFR-mutated NSCLC. Recently, the acquired resistance to osimertinib brings great challenges to the advanced treatment. Therefore, it is in urgent need to find effective strategy to overcome osimertinib acquired resistance. Here, we demonstrated that SREBP pathway-driven lipogenesis was a key mediator to promote osimertinib acquired resistance, and firstly found Tanshinone IIA (Tan IIA), a natural pharmacologically active constituent isolated from Salvia miltiorrhiza, could overcome osimertinib-acquired resistance in vitro and in vivo via inhibiting SREBP pathway-mediated lipid lipogenesis by using LC-MS based cellular lipidomics analysis, quantitative real-time PCR (qRT-PCR) analysis, western blotting analysis, flow cytometry, small interfering RNAs transfection, and membrane fluidity assay et al. The results showed that SREBP1/2-driven lipogenesis was highly activated in osimertinib acquired resistant NSCLC cells, while knockdown or inhibition of SREBP1/2 could restore the sensitivity of NSCLC to osimertinib via altered the proportion of saturated phospholipids and unsaturated phospholipids in osimertinib acquired-resistant cells. Furthermore, Tanshinone IIA (Tan IIA) could reverse the acquired resistance to osimertinib in lung cancer. Mechanically, Tan IIA inhibited SREBP signaling mediated lipogenesis, changed the profiles of saturated phospholipids and unsaturated phospholipids, and thus promoted osimertinib acquired resistant cancer cells to be attacked by oxidative stress-induced damage and reduce the cell membrane fluidity. The reversal effect of Tan IIA on osimertinib acquired resistant NSCLC cells was also confirmed in vivo, which is helpful for the development of strategies to reverse osimertinib acquired resistance.


Sujet(s)
Abiétanes , Acrylamides , Résistance aux médicaments antinéoplasiques , Lipogenèse , Tumeurs du poumon , Souris nude , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Abiétanes/pharmacologie , Animaux , Acrylamides/pharmacologie , Lipogenèse/effets des médicaments et des substances chimiques , Souris , Dérivés de l'aniline/pharmacologie , Antinéoplasiques/pharmacologie , Souris de lignée BALB C , Lignée cellulaire tumorale , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Protéine-2 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-2 de liaison à l'élément de régulation des stérols/génétique , Mâle , Femelle , Indoles , Pyrimidines
20.
J Med Chem ; 67(9): 7283-7300, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38676656

RÉSUMÉ

The epidermal growth factor receptor (EGFR) tertiary C797S mutation is an important cause of resistance to Osimertinib, which seriously hinders the clinical application of Osimertinib. Developing proteolysis-targeting chimeras (PROTACs) targeting EGFR mutants can offer a promising strategy to overcome drug resistance. In this study, some novel PROTACs targeting C797S mutation were designed and synthesized based on a new EGFR inhibitor and displayed a potent degradation effect in H1975-TM cells harboring EGFRL858R/T790M/C797S. The representative compound C6 exhibited a DC50 of 10.2 nM against EGFRL858R/T790M/C797S and an IC50 of 10.3 nM against H1975-TM. Furthermore, C6 also showed potent degradation activity against various main EGFR mutants, including EGFRDel19/T790M/C797S. Mechanistic studies revealed that the protein degradation was achieved through the ubiquitin-proteasome system. Finally, C6 inhibited tumor growth in the H1975-TM xenograft tumor model effectively and safely. This study identifies a novel and potent EGFR PROTAC to overcome Osimertinib resistance mediated by C797S mutation.


Sujet(s)
Antinéoplasiques , Conception de médicament , Récepteurs ErbB , Mutation , Inhibiteurs de protéines kinases , Protéolyse , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Humains , Animaux , Protéolyse/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Souris , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Souris nude , Acrylamides/pharmacologie , Acrylamides/synthèse chimique , Acrylamides/composition chimique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/synthèse chimique , Dérivés de l'aniline/composition chimique , Souris de lignée BALB C , Relation structure-activité , Chimère ciblant la protéolyse , Indoles , Pyrimidines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...