Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 250
Filtrer
1.
PeerJ ; 12: e17446, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827306

RÉSUMÉ

Objectives: To investigate the interaction between tramadol and representative tyrosine kinase inhibitors, and to study the inhibition mode of drug-interaction. Methods: Liver microsomal catalyzing assay was developed. Sprague-Dawley rats were administrated tramadol with or without selected tyrosine kinase inhibitors. Samples were prepared and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used for analysis. Besides, liver, kidney, and small intestine were collected and morphology was examined by hematoxyline-eosin (H&E) staining. Meanwhile, liver microsomes were prepared and carbon monoxide differential ultraviolet radiation (UV) spectrophotometric quantification was performed. Results: Among the screened inhibitors, crizotinib takes the highest potency in suppressing the metabolism of tramadol in rat/human liver microsome, following non-competitive inhibitory mechanism. In vivo, when crizotinib was co-administered, the AUC value of tramadol increased compared with the control group. Besides, no obvious pathological changes were observed, including cell morphology, size, arrangement, nuclear morphology with the levels of alanine transaminase (ALT) and aspartate transaminase (AST) increased after multiple administration of crizotinib. Meanwhile, the activities of CYP2D1 and CYP3A2 as well as the total cytochrome P450 abundance were found to be decreased in rat liver of combinational group. Conclusions: Crizotinib can inhibit the metabolism of tramadol. Therefore, this recipe should be vigilant to prevent adverse reactions.


Sujet(s)
Crizotinib , Cytochrome P-450 CYP3A , Microsomes du foie , Rat Sprague-Dawley , Tramadol , Animaux , Tramadol/pharmacologie , Crizotinib/pharmacologie , Rats , Microsomes du foie/effets des médicaments et des substances chimiques , Microsomes du foie/métabolisme , Cytochrome P-450 CYP3A/métabolisme , Mâle , Interactions médicamenteuses , Humains , Spectrométrie de masse en tandem , Famille-2 de cytochromes P450/métabolisme , Famille-2 de cytochromes P450/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/pharmacocinétique , Analgésiques morphiniques/pharmacologie
2.
J Transl Med ; 22(1): 585, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902768

RÉSUMÉ

BACKGROUND: In the era of second-generation ALK tyrosine kinase inhibitors (ALK-TKIs), there was a paucity of data regarding the progression patterns, resistant mechanisms, and subsequent therapeutic approaches for ALK-positive (ALK+) non-small cell lung cancer (NSCLC). METHODS: Patients with advanced ALK+ NSCLC were retrospectively selected from our center. Cohort 1 consisted of patients who experienced disease progression after receiving first-line alectinib treatment (n = 20), while Cohort 2 included patients who progressed following sequential treatment with crizotinib and second-generation ALK-TKIs (n = 53). Oligo-progression was defined as the occurrence of disease progression in no more than three lesions. Symptomatic progression was determined when patients developed new symptoms or experienced worsening of pre-existing symptoms during radiological progression. RESULTS: The incidence of central nervous system (CNS) progression and symptomatic CNS progression was significantly lower in Cohort 1 compared to patients treated with crizotinib, with rates of 15.0% vs. 56.6% (p = 0.002) and 5.0% vs. 32.1% (p = 0.016), respectively. A total of 60.3% (44/73) patients underwent repeated biopsy and next-generation sequencing subsequent to the second-generation ALK-TKI resistance, with secondary mutation in ALK kinase domain emerging as the predominant mechanism of resistance (56.8%). Local therapy was applied to 50% of oligo-progression cases. Subsequent ALK-TKIs demonstrated significantly prolonged progression-free survival (PFS) (8.6 m vs. 2.7 m, p = 0.021, HR = 0.43, 95%CI: 0.15-0.85) and long-term overall survival (OS) (NA vs. 11.9 m, p = 0.132, HR = 0.50, 95%CI: 0.18-1.25) in patients harboring ALK resistance mutations, compared to those without such mutations. For patients without ALK-resistant mutations following progression on second-generation ALK-TKIs, there was no statistically significant difference in survival outcomes between subsequent chemotherapy or alternative ALK-TKI treatments. CONCLUSIONS: First-line alectinib demonstrated superior efficacy in protecting the CNS compared to crizotinib. For patients with ALK-resistant mutations following the resistance to second-generation ALK-TKIs, appropriate sensitive ALK-TKI should be administered; for those without such mutations, the selection of chemotherapy or third-generation ALK-TKI should be based on the patient's overall physical health and personal preferences.


Sujet(s)
Kinase du lymphome anaplasique , Carcinome pulmonaire non à petites cellules , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Kinase du lymphome anaplasique/métabolisme , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sujet âgé , Adulte , Crizotinib/usage thérapeutique , Crizotinib/pharmacologie , Études rétrospectives , Mutation/génétique
3.
Mol Cancer ; 23(1): 123, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849845

RÉSUMÉ

BACKGROUND: Pediatric-type diffuse high-grade glioma (pHGG) is the most frequent malignant brain tumor in children and can be subclassified into multiple entities. Fusion genes activating the MET receptor tyrosine kinase often occur in infant-type hemispheric glioma (IHG) but also in other pHGG and are associated with devastating morbidity and mortality. METHODS: To identify new treatment options, we established and characterized two novel orthotopic mouse models harboring distinct MET fusions. These included an immunocompetent, murine allograft model and patient-derived orthotopic xenografts (PDOX) from a MET-fusion IHG patient who failed conventional therapy and targeted therapy with cabozantinib. With these models, we analyzed the efficacy and pharmacokinetic properties of three MET inhibitors, capmatinib, crizotinib and cabozantinib, alone or combined with radiotherapy. RESULTS: Capmatinib showed superior brain pharmacokinetic properties and greater in vitro and in vivo efficacy than cabozantinib or crizotinib in both models. The PDOX models recapitulated the poor efficacy of cabozantinib experienced by the patient. In contrast, capmatinib extended survival and induced long-term progression-free survival when combined with radiotherapy in two complementary mouse models. Capmatinib treatment increased radiation-induced DNA double-strand breaks and delayed their repair. CONCLUSIONS: We comprehensively investigated the combination of MET inhibition and radiotherapy as a novel treatment option for MET-driven pHGG. Our seminal preclinical data package includes pharmacokinetic characterization, recapitulation of clinical outcomes, coinciding results from multiple complementing in vivo studies, and insights into molecular mechanism underlying increased efficacy. Taken together, we demonstrate the groundbreaking efficacy of capmatinib and radiation as a highly promising concept for future clinical trials.


Sujet(s)
Tumeurs du cerveau , Gliome , Protéines proto-oncogènes c-met , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Humains , Gliome/anatomopathologie , Gliome/traitement médicamenteux , Gliome/génétique , Gliome/thérapie , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Souris , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/radiothérapie , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Lignée cellulaire tumorale , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Femelle , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Modèles animaux de maladie humaine , Enfant , Grading des tumeurs , Anilides/pharmacologie , Imidazoles , Triazines
4.
Hum Cell ; 37(4): 1132-1140, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38829559

RÉSUMÉ

Approximately 3-5% of non-small cell lung cancers (NSCLC) harbor ALK fusion genes and may be responsive to anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors. There are only a few reports on cell lines with EML4-ALK variant 3 (v3) and tumoroids that can be subject to long-term culture (> 3 months). In this study, we established tumoroids (PDT-LUAD#119) from a patient with lung cancer harboring EML4-ALK that could be cultured for 12 months. Whole-exome sequencing and RNA sequencing analyses revealed TP53 mutations and an EML4-ALK v3 mutation. PDT-LUAD#119 lung tumoroids were sensitive to the ALK tyrosine kinase inhibitors (ALK TKIs) crizotinib, alectinib, entrectinib, and lorlatinib, similar to NCI-H3122 cells harboring EML4-ALK variant 1 (v1). Unexpectedly, clear squamous cell carcinoma and solid adenocarcinoma were observed in xenografts from PDT-LUAD#119 lung tumoroids, indicating adenosquamous carcinoma. Immunostaining revealed that the squamous cell carcinoma was ALK positive, suggesting a squamous transformation of the adenocarcinoma. Besides providing a novel cancer model to support basic research on ALK-positive lung cancer, PDT-LUAD#119 lung tumoroids will help elucidate the pathogenesis of adenosquamous carcinoma.


Sujet(s)
Kinase du lymphome anaplasique , Carcinome épidermoïde , Tumeurs du poumon , Protéines de fusion oncogènes , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Animaux , Transformation cellulaire néoplasique/génétique , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Crizotinib/pharmacologie , Lignée cellulaire tumorale , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie
5.
PLoS One ; 19(5): e0300644, 2024.
Article de Anglais | MEDLINE | ID: mdl-38758826

RÉSUMÉ

Mechanisms underlying primary and acquired resistance to MET tyrosine kinase inhibitors (TKIs) in managing non-small cell lung cancer remain unclear. In this study, we investigated the possible mechanisms acquired for crizotinib in MET-amplified lung carcinoma cell lines. Two MET-amplified lung cancer cell lines, EBC-1 and H1993, were established for acquired resistance to MET-TKI crizotinib and were functionally elucidated. Genomic and transcriptomic data were used to assess the factors contributing to the resistance mechanism, and the alterations hypothesized to confer resistance were validated. Multiple mechanisms underlie acquired resistance to crizotinib in MET-amplified lung cancer cell lines. In EBC-1-derived resistant cells, the overexpression of SERPINE1, the gene encoding plasminogen activator inhibitor-1 (PAI-1), mediated the drug resistance mechanism. Crizotinib resistance was addressed by combination therapy with a PAI-1 inhibitor and PAI-1 knockdown. Another mechanism of resistance in different subline cells of EBC-1 was evaluated as epithelial-to-mesenchymal transition with the upregulation of antiapoptotic proteins. In H1993-derived resistant cells, MEK inhibitors could be a potential therapeutic strategy for overcoming resistance with downstream mitogen-activated protein kinase pathway activation. In this study, we revealed the different mechanisms of acquired resistance to the MET inhibitor crizotinib with potential therapeutic application in patients with MET-amplified lung carcinoma.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Crizotinib , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Inhibiteur-1 d'activateur du plasminogène , Inhibiteurs de protéines kinases , Protéines proto-oncogènes c-met , Humains , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Inhibiteur-1 d'activateur du plasminogène/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-met/métabolisme , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
6.
ACS Nano ; 18(17): 11025-11041, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38626916

RÉSUMÉ

ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.


Sujet(s)
Crizotinib , Tumeurs du poumon , Nanoparticules magnétiques d'oxyde de fer , Microenvironnement tumoral , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Humains , Souris , Crizotinib/pharmacologie , Crizotinib/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Lignée cellulaire tumorale , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle
8.
J Med Chem ; 67(6): 4916-4935, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38477575

RÉSUMÉ

The emergence of resistance to PARP1 inhibitors poses a current therapeutic challenge, necessitating the development of novel strategies to overcome this obstacle. The present study describes the design and synthesis of a series of small molecules that target both PARP1 and c-Met. Among them, compound 16 is identified as a highly potent dual inhibitor, exhibiting excellent inhibitory activities against PARP1 (IC50 = 3.3 nM) and c-Met (IC50 = 32.2 nM), as well as demonstrating good antiproliferative effects on HR-proficient cancer cell lines and those resistant to PARP1 inhibitors. Importantly, compound 16 demonstrates superior antitumor potency compared to the PARP1 inhibitor Olaparib and the c-Met inhibitor Crizotinib, either alone or in combination, in MDA-MB-231 and HCT116OR xenograft models. These findings highlight the potential of PARP1/c-Met dual inhibitors for expanding the indications of PARP1 inhibitors and overcoming tumor cells' resistance to them.


Sujet(s)
Antinéoplasiques , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Lignée cellulaire tumorale , Poly (ADP-Ribose) polymerase-1 , Crizotinib/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Prolifération cellulaire , Antinéoplasiques/pharmacologie
9.
Clin Chem ; 70(4): 629-641, 2024 04 03.
Article de Anglais | MEDLINE | ID: mdl-38416709

RÉSUMÉ

BACKGROUND: ROS1 fusion-positive (ROS1+) nonsmall cell lung cancer (NSCLC) patients are highly sensitive to tyrosine kinase inhibitor (TKI) treatments. However, acquired TKI resistance remains the major hurdle preventing patients from experiencing prolonged benefits. METHODS: 107 advanced or metastatic ROS1+ NSCLC patients who progressed on crizotinib and lorlatinib were recruited. Tissue and plasma samples were collected at baseline (N = 50), postcrizotinib (N = 91), and postlorlatinib (N = 21), which were all subject to the 139-gene targeted next-generation DNA sequencing. Molecular dynamics modeling was performed to investigate the effects of ROS1 mutations on binding to different TKIs. RESULTS: In patients with postcrizotinib and postlorlatinib samples, an accumulation of on- and off-target resistance alterations after multiple TKI treatments was observed. ROS1 G2032R and MET amplification were the most common on-target and off-target alterations, respectively. Patients with CD74-ROS1 and SLC34A2-ROS1 had longer progression-free survival (PFS) (P < 0.001) and higher rates of resistance mutations (on-target, P = 0.001; off-target, P = 0.077) than other ROS1 fusion variants following crizotinib treatment. Ten distinct on-target resistance mutations were detected after TKI therapies, of which 4 were previously unreported (ROS1 L2010M, G1957A, D1988N, L1982V). Molecular dynamics simulations showed that all 4 mutations were refractory to crizotinib, while G1957A, D1988N, and L1982V were potentially sensitive to lorlatinib and entrectinib. CONCLUSIONS: This study provided a comprehensive portrait of TKI-resistance mechanisms in ROS1+ NSCLC patients. Using in silico simulations of TKI activity, novel secondary mutations that may confer TKI resistance were identified and may support clinical therapeutic decision-making.


Sujet(s)
Aminopyridines , Carcinome pulmonaire non à petites cellules , Lactames , Tumeurs du poumon , Pyrazoles , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Crizotinib/usage thérapeutique , Crizotinib/pharmacologie , Protein-tyrosine kinases/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Lactames macrocycliques/pharmacologie , Lactames macrocycliques/usage thérapeutique , Résistance aux médicaments antinéoplasiques/génétique
10.
ESMO Open ; 9(2): 102237, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38350336

RÉSUMÉ

BACKGROUND: In 2019, we reported the first efficacy and safety analysis of EUCROSS, a phase II trial investigating crizotinib in ROS1 fusion-positive lung cancer. At that time, overall survival (OS) was immature and the effect of crizotinib on intracranial disease control remained unclear. Here, we present the final analysis of OS, systemic and intracranial activity, and the impact of co-occurring aberrations. MATERIALS AND METHODS: EUCROSS was a prospective, single-arm, phase II trial. The primary endpoint was best overall response rate (ORR) using RECIST 1.1. Secondary and exploratory endpoints were progression-free survival (PFS), OS, and efficacy in pre-defined subgroups. RESULTS: Median OS of the intention-to-treat population (N = 34) was 54.8 months [95% confidence interval (CI) 20.3 months-not reached (NR); median follow-up 81.4 months] and median all-cause PFS of the response-evaluable population (N = 30) was 19.4 months (95% CI 10.1-32.2 months). Time on treatment was significantly correlated with OS (R = 0.82; P < 0.0001). Patients with co-occurring TP53 aberrations (28%) had a significantly shorter OS [hazard ratio (HR) 11; 95% CI 2.0-56.0; P = 0.006] and all-cause PFS (HR 4.2; 95% CI 1.2-15; P = 0.025). Patients with central nervous system (CNS) involvement at baseline (N = 6; 20%) had a numerically shorter median OS and all-cause PFS. Median intracranial PFS was 32.2 months (95% CI 23.7 months-NR) and the rate of isolated CNS progression was 24%. CONCLUSIONS: Our final analysis proves the efficacy of crizotinib in ROS1-positive lung cancer, but also highlights the devastating impact of TP53 mutations on survival and treatment efficacy. Additionally, our data show that CNS disease control is durable and the risk of CNS progression while on crizotinib treatment is low.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Protein-tyrosine kinases/génétique , Études prospectives , Protéines proto-oncogènes/génétique , Système nerveux central
11.
Acta Pharmacol Sin ; 45(6): 1252-1263, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38360931

RÉSUMÉ

Although ALK tyrosine kinase inhibitors (ALK-TKIs) have shown remarkable benefits in EML4-ALK positive NSCLC patients compared to conventional chemotherapy, the optimal sequence of ALK-TKIs treatment remains unclear due to the emergence of primary and acquired resistance and the lack of potential prognostic biomarkers. In this study, we systematically explored the validity of sequential ALK inhibitors (alectinib, lorlatinib, crizotinib, ceritinib and brigatinib) for a heavy-treated patient with EML4-ALK fusion via developing an in vitro and in vivo drug testing system based on patient-derived models. Based on the patient-derived models and clinical responses of the patient, we found that crizotinib might inhibit proliferation of EML4-ALK positive tumors resistant to alectinib and lorlatinib. In addition, NSCLC patients harboring the G1269A mutation, which was identified in alectinib, lorlatinib and crizotinib-resistant NSCLC, showed responsiveness to brigatinib and ceritinib. Transcriptomic analysis revealed that brigatinib suppressed the activation of multiple inflammatory signaling pathways, potentially contributing to its anti-tumor activity. Moreover, we constructed a prognostic model based on the expression of IL6, CXCL1, and CXCL5, providing novel perspectives for predicting prognosis in EML4-ALK positive NSCLC patients. In summary, our results delineate clinical responses of sequential ALK-TKIs treatments and provide insights into the mechanisms underlying the superior effects of brigatinib in patients harboring ALKG1269A mutation and resistant towards alectinib, lorlatinib and crizotinib. The molecular signatures model based on the combination of IL6, CXCL1 and CXCL5 has the potential to predict prognosis of EML4-ALK positive NSCLC patients.


Sujet(s)
Adénocarcinome pulmonaire , Antinéoplasiques , Tumeurs du poumon , Protéines de fusion oncogènes , Composés organiques du phosphore , Inhibiteurs de protéines kinases , Pyrimidines , Humains , Composés organiques du phosphore/usage thérapeutique , Composés organiques du phosphore/pharmacologie , Pyrimidines/usage thérapeutique , Pyrimidines/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Animaux , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Pronostic , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques , Lactames/usage thérapeutique , Carbazoles/usage thérapeutique , Carbazoles/pharmacologie , Sulfones/usage thérapeutique , Sulfones/pharmacologie , Crizotinib/usage thérapeutique , Crizotinib/pharmacologie , Lignée cellulaire tumorale , Pipéridines/usage thérapeutique , Pipéridines/pharmacologie , Femelle , Souris , Inflammation/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Pyrazoles/usage thérapeutique , Pyrazoles/pharmacologie , Mâle , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Kinase du lymphome anaplasique/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mutation , Aminopyridines/usage thérapeutique , Aminopyridines/pharmacologie
12.
J Thorac Oncol ; 19(6): 912-927, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38280448

RÉSUMÉ

INTRODUCTION: Iruplinalkib (WX-0593) is a new-generation, potent ALK tyrosine kinase inhibitor (TKI) that has been found to have systemic and central nervous system (CNS) efficacy in ALK-positive NSCLC. We compared the efficacy and safety of iruplinalkib with crizotinib in patients with ALK TKI-naive, locally advanced or metastatic ALK-positive NSCLC. METHODS: In this open-label, randomized, multicenter, phase 3 study, patients with ALK-positive NSCLC were randomly assigned to receive iruplinalkib 180 mg once daily (7-d run-in at 60 mg once daily) or crizotinib 250 mg twice daily. The primary end point was progression-free survival (PFS) assessed by Independent Review Committee (IRC) per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary end points included PFS by investigator, objective response rate (ORR), time to response, duration of response, intracranial ORR and time to CNS progression by IRC and investigator, overall survival, and safety. An interim analysis was planned after approximately 70% (134 events) of all 192 expected PFS events assessed by IRC were observed. Efficacy was analyzed in the intention-to-treat population. Safety was assessed in the safety population, which included all randomized patients who received at least one dose of the study drugs. This study is registered with Center for Drug Evaluation of China National Medical Products Administration (CTR20191231) and Clinicaltrials.gov (NCT04632758). RESULTS: From September 4, 2019, to December 2, 2020, a total of 292 patients were randomized and treated; 143 with iruplinalkib and 149 with crizotinib. At this interim analysis (145 events), the median follow-up time was 26.7 months (range: 3.7-37.7) in the iruplinalkib group and 25.9 months (range: 0.5-35.9) in the crizotinib group. The PFS assessed by IRC was significantly longer among patients in the iruplinalkib group (median PFS, 27.7 mo [95% confidence interval (CI): 26.3-not estimable] versus 14.6 mo [95% CI: 11.1-16.5] in the crizotinib group; hazard ratio, 0.34 [98.02% CI: 0.23-0.52], p < 0.0001). The ORR assessed by IRC was 93.0% (95% CI: 87.5-96.6) in the iruplinalkib group and 89.3% (95% CI: 83.1-93.7) in the crizotinib group. The intracranial ORR was 90.9% (10 of 11, 95% CI: 58.7-99.8) in the iruplinalkib group and 60.0% (nine of 15, 95% CI: 32.3-83.7) in the crizotinib group for patients with measurable baseline CNS metastases. Incidence of grade 3 or 4 treatment-related adverse events was 51.7% in the iruplinalkib group and 49.7% in the crizotinib group. CONCLUSIONS: Iruplinalkib was found to have significantly improved PFS and improved intracranial antitumor activity versus crizotinib. Iruplinalkib may be a new treatment option for patients with advanced ALK-positive and ALK TKI-naive NSCLC. FUNDING: This study was funded by Qilu Pharmaceutical Co., Ltd., Jinan, People's Republic of China, and partly supported by the National Science and Technology Major Project for Key New Drug Development (2017ZX09304015).


Sujet(s)
Carcinome pulmonaire non à petites cellules , Crizotinib , Tumeurs du poumon , Humains , Mâle , Femelle , 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 , Adulte d'âge moyen , Crizotinib/usage thérapeutique , Crizotinib/pharmacologie , Adulte , Sujet âgé , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs
13.
Thorac Cancer ; 15(1): 89-93, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38093515

RÉSUMÉ

The possibility of stratifying patients according to differences in ROS proto-oncogene 1 (ROS1) fusion partners has been discussed. This study aimed to clarify the clinicopathological differences between two SDC4::ROS1 positive NSCLC cases who had different responses to crizotinib. Cytology and pathology samples from two NSCLC cases with SDC4::ROS1 who were diagnosed and treated with crizotinib at Nihon University Itabashi Hospital were obtained. Case 1 has been well-controlled with crizotinib for over 5 years, but case 2 was worse and overall survival was 19 months. Sequencing analysis of ROS1 fusion genes was performed by reverse-transcription-PCR and Sanger's sequencing methods. In addition, thyroid transcription factor (TTF)-1, ROS-1, Ki67, and phosphorylated extracellular signal-regulated kinase (pERK)1/2 expression were investigated using immunohistochemistry. Sequencing analysis showed SDC4 exon2::ROS1 exon 32 (exon33 deleted) in case 1, and coexistence of SDC4 exon2::ROS1 exon 34 and SDC4 exon2::ROS1 exon35 in case 2. The Ki67 index was not different, but ROS1 and pERK1/2 expression levels tended to be higher in the tumor cells of case 2 than in case 1. Therapeutic response to crizotinib and patients' prognosis in ROS1 rearranged NSCLC may be related to the activation of ROS1 signaling, depending on ROS1 and pERK1/2 overexpression status, even if the ROS1 fusion partner is the same.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Crizotinib , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Antigène KI-67 , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Espèces réactives de l'oxygène , Syndécane-4/génétique
14.
J Thorac Oncol ; 19(4): 613-625, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38070598

RÉSUMÉ

INTRODUCTION: Variable partners and breakpoints have been reported in patients with ROS1-rearranged NSCLC. Here, we investigated the association of fusion partners and breakpoints with crizotinib efficacy in NSCLCs with common ROS1 fusions. METHODS: DNA and RNA next-generation sequencing (NGS) and immunohistochemistry were performed to characterize ROS1 fusions. RESULTS: Using DNA NGS, we identified ROS1 fusions in 210 cases, comprising 171 common (CD74/EZR/TPM3/SDC4/SLC34A2-ROS1) and 39 uncommon (variants identified in <5%) ROS1 fusion cases. DNA NGS detected variable ROS1 genomic breakpoints in common ROS1 fusions, whereas RNA NGS found ROS1 breakpoints mainly occurring in exons 32, 34 and 35, resulting in long (exon 32) and short (exon 34 or 35) ROS1 fusions. ROS1 immunohistochemistry revealed that membranous and cytoplasmic staining was predominant in long ROS1 fusions, whereas cytoplasmic staining was predominant in short ROS1 fusions (p = 0.006). For patients who received first-line crizotinib, median progression-free survival (mPFS) was lower in patients with long ROS1 fusions than those with short ROS1 fusions (8.0 versus 24.0 mo, p = 0.006). Moreover, mPFS for patients with and without TP53 mutations was 8.0 and 19.0 months, respectively (p = 0.159); mPFS for patients with and without BIM deletion polymorphism was 5.0 and 22.0 months, respectively (p = 0.003). When analyzing together with fusion partners, patients with long CD74/SLC34A2-ROS1 fusions were found to have shorter PFS than those with other ROS1, regardless of the presence or absence of TP53 mutations (p < 0.001 and p = 0.002, respectively). CONCLUSIONS: Long CD74/SLC34A2-ROS1 fusions, which retain transmembrane regions in ROS1 and fusion partners, are associated with poor response to crizotinib independent of TP53 mutations.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Antigènes d'histocompatibilité de classe II , Tumeurs du poumon , Protéines de fusion oncogènes , Protein-tyrosine kinases , Protéines proto-oncogènes , Cotransporteurs sodium-phosphate de type IIb , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , ADN , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Mutation , Protéines de fusion oncogènes/génétique , Protein-tyrosine kinases/génétique , Protein-tyrosine kinases/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , ARN , Cotransporteurs sodium-phosphate de type IIb/génétique , Protéine p53 suppresseur de tumeur/génétique , Antigènes d'histocompatibilité de classe II/génétique , Antigènes de différenciation des lymphocytes B/génétique
15.
Target Oncol ; 18(6): 905-914, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37966566

RÉSUMÉ

BACKGROUND: Although ALK-translocated (ALK+) advanced non-small cell lung cancers (aNSCLCs) are currently treated with second- or third-generation ALK inhibitors (ALK-TKIs), some patients respond durably to the first-generation ALK-TKI crizotinib. OBJECTIVE: This study aimed to describe the clinical characteristics of these long-term responders. PATIENTS AND METHODS: This national, multicenter, retrospective, non-interventional study included patients with ALK+ aNSCLCs and long-term responses to first (L1)- or subsequent (≥ L2)-line crizotinib, defined, respectively, as treatments lasting > 18 and > 10 months. Median treatment duration (mDOT) was the primary endpoint. RESULTS: A total of 85 patients (32 L1 and 53 ≥ L2 responders) from 23 centers were included (receiving crizotinib between 10/24/2011-10/02/2018): median age of 59 years, 83.6% non-smokers or ex-smokers, 85.9% performance status (PS) 0/1, 94.1% with adenocarcinomas, median of one metastatic site, and 22.4% with brain metastases (BMs). After median follow-up of 73.4 [95% confidence interval, 67.5-79.9] months, respective L1 and ≥ L2 mDOTs were 43.3 [26.7-56.8] and 29.6 [22.6-35.8] months, with overall survival (OS) not reached (NR) and 116.2 [83.4-NR] months. BM presence or absence did not affect mDOT (31.4 versus 32.9 months) but significantly impacted median OS (70.6 versus 158.6 months; p = 0.0008). Progression on crizotinib was paucisymptomatic (74.1%) and oligometastatic (34.8%), especially BMs (42.4%). After crizotinib discontinuation, 65 (76.5%) patients received subsequent systemic therapy: 57 (67.1%) with second-generation ALK-TKIs. Respective mDOTs of first- and second-line post-crizotinib ALK-TKIs lasted 19.4 [14.9-25.6] and 11.1 [4.8-17.9] months, respectively. CONCLUSIONS: Most ALK+ aNSCLC patients with prolonged crizotinib efficacy had paucisymptomatic and oligometastatic disease without BMs. They subsequently benefited from a sequential strategy with other ALK-TKIs.


Sujet(s)
Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/anatomopathologie , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Tumeurs du poumon/anatomopathologie , Études rétrospectives , Kinase du lymphome anaplasique/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/secondaire
16.
Mol Pharm ; 20(12): 6140-6150, 2023 Dec 04.
Article de Anglais | MEDLINE | ID: mdl-37939020

RÉSUMÉ

Glioblastoma (GBM) is a highly aggressive form of brain cancer with a poor prognosis and limited treatment options. The ALK and c-MET inhibitor Crizotinib has demonstrated preclinical therapeutic potential for newly diagnosed GBM, although its efficacy is limited by poor penetration of the blood brain barrier. Here, we identify Crizotinib as a novel inhibitor of nuclear factor-κB (NF-κB)-inducing kinase, which is a key regulator of GBM growth and proliferation. We further show that the conjugation of Crizotinib to a heptamethine cyanine dye, or a near-infrared dye (IR-Crizotinib), attenuated glioma cell proliferation and survival in vitro to a greater extent than unconjugated Crizotinib. Moreover, we observed increased IR-Crizotinib localization to orthotopic mouse xenograft GBM tumors, which resulted in impaired tumor growth in vivo. Overall, IR-Crizotinib exhibited improved intracranial chemotherapeutic delivery and tumor localization with concurrent inhibition of NIK and noncanonical NF-κB signaling, thereby reducing glioma growth in vitro, as well as in vivo, and increasing survival in a preclinical rodent model.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Gliome , Souris , Animaux , Humains , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Facteur de transcription NF-kappa B , Lignée cellulaire tumorale , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Glioblastome/traitement médicamenteux ,
17.
Cancer Biol Ther ; 24(1): 2271212, 2023 12 31.
Article de Anglais | MEDLINE | ID: mdl-37906510

RÉSUMÉ

Anaplastic lymphoma kinase-positive large B-cell lymphoma (ALK+ LBCL) is a rare subtype of non-Hodgkin lymphoma. ALK inhibitors are being tried to treat recurrent/refractory ALK+ LBCL. A majority of patients with ALK+ tumors respond to crizotinib, but partial cases ultimately develop resistance about a year later. Here, we report a case of ALK+ LBCL carrying a new fusion gene involving CDK14 and ALK, CLTC-ALK gene rearrangements and MTOR gene mutation. The patient had progressive disease after combination of crizotinib and chemotherapy treatment about 5.5 months later, accompanied by reduced abundance of CDK14-ALK, increased abundance of CLTC-ALK and a novel MFHAS1 gene mutation. However, MTOR mutation turned negative. The patient received alectinib combined with hyper-CVAD, then followed by alectinib as monotherapy for 21 months. The patient achieved partial response and remained in a stable condition. This case suggests that CDK14-ALK fusion gene may be more sensitive to crizotinib than CLTC-ALK fusion gene. MTOR is associated with the anti-tumor mechanism of ALK inhibitors. MFHAS1 gene mutation and/or CLTC-ALK gene copy number amplification may involve resistance to crizotinib. Furthermore, alectinib may inhibit the carcinogenicity of these gene changes and improve the prognosis of ALK+ LBCL.


The novel CDK14-ALK fusion gene in ALK+ LBCL was sensitive to crizotinib.MFHAS1 gene mutation and/or CLTC-ALK gene copy number amplification may involve resistance to crizotinib.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Lymphome B , Humains , Kinase du lymphome anaplasique/génétique , Carbazoles/pharmacologie , Carbazoles/usage thérapeutique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines du cycle cellulaire/génétique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Protéines de liaison à l'ADN/génétique , Résistance aux médicaments antinéoplasiques/génétique , Séquençage nucléotidique à haut débit , Tumeurs du poumon/anatomopathologie , Lymphome B/traitement médicamenteux , Mutation , Protéines oncogènes/génétique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Sérine-thréonine kinases TOR/génétique
18.
J Thorac Oncol ; 18(12): 1743-1755, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37574132

RÉSUMÉ

INTRODUCTION: This open-label, phase 3 trial (ALTA-3; NCT03596866) compared efficacy and safety of brigatinib versus alectinib for ALK+ NSCLC after disease progression on crizotinib. METHODS: Patients with advanced ALK+ NSCLC that progressed on crizotinib were randomized 1:1 to brigatinib 180 mg once daily (7-d lead-in, 90 mg) or alectinib 600 mg twice daily, aiming to test superiority. The primary end point was blinded independent review committee-assessed progression-free survival (PFS). Interim analysis for efficacy and futility was planned at approximately 70% of 164 expected PFS events. RESULTS: The population (N = 248; brigatinib, n = 125; alectinib, n = 123) was notable for long median duration of prior crizotinib (16.0-16.8 mo) and low rate of ALK fusion in baseline circulating tumor DNA (ctDNA; 78 of 232 [34%]). Median blinded independent review committee-assessed PFS was 19.3 months with brigatinib and 19.2 months with alectinib (hazard ratio = 0.97 [95% confidence interval: 0.66-1.42], p = 0.8672]). The study met futility criterion. Overall survival was immature (41 events [17%]). Exploratory analyses pooled across the treatment groups revealed median PFS of 11.1 versus 22.5 months in patients with versus without ctDNA-detectable ALK fusion at baseline (hazard ratio: 0.48 [95% confidence interval: 0.32-0.71]). Treatment-related adverse events in more than 30% of patients (brigatinib, alectinib) were elevated levels of blood creatine phosphokinase (70%, 29%), aspartate aminotransferase (53%, 38%), and alanine aminotransferase (40%, 36%). CONCLUSIONS: Brigatinib was not superior to alectinib for PFS in crizotinib-pretreated ALK+ NSCLC. Safety was consistent with the well-established and unique profiles of each drug. The low proportion of patients with ctDNA-detectable ALK fusion may account for prolonged PFS with both drugs in ALTA-3.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Kinase du lymphome anaplasique/génétique , Carbazoles/pharmacologie , Carbazoles/usage thérapeutique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Évolution de la maladie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique
19.
Cell Mol Biol (Noisy-le-grand) ; 69(5): 174-178, 2023 May 31.
Article de Anglais | MEDLINE | ID: mdl-37571883

RÉSUMÉ

C-Met receptor and its ligand hepatocyte growth factor (HGF) are overexpressed in a variety of osteosarcoma cell lines and osteosarcoma pathological samples. It is suggested that c-Met/HGF plays an important role in the development of osteosarcoma. This study aimed to explore the anticancer effect of the c-Met-targeted drug crizotinib on osteosarcoma (OS) cells. The effects of crizotinib on the proliferation of osteosarcoma cells (SaOS2, MG-63 and MNNG) at different concentrations were detected by CCK8. Human osteosarcoma cell line MG-63 was used as an in vitro model to evaluate the effects of 2.5 µM crizotinib, 5.0 µM crizotinib and DMSO on cell apoptosis, cell cycle, migration and invasion. The expression of the c-Met signaling pathway in osteosarcoma cells was detected by western blot. The results showed that crizotinib inhibited the proliferation of cell lines in a concentration-dependent manner. Crizotinib significantly increased the number of apoptotic cells compared with the control group. Compared with the control group, crizotinib increased G0/G1 phase cells and decreased S phase cells. Compared with the control group, crizotinib inhibited the migration and invasion of osteosarcoma cells and decreased the expression of c-Met/Gab1/STAT5. This study will provide a promising therapeutic target and theoretical basis for the clinical application of crizotinib in osteosarcoma.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Humains , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Prolifération cellulaire , Lignée cellulaire tumorale , Transduction du signal , Ostéosarcome/métabolisme , Apoptose , Tumeurs osseuses/traitement médicamenteux
20.
Signal Transduct Target Ther ; 8(1): 301, 2023 08 14.
Article de Anglais | MEDLINE | ID: mdl-37574511

RÉSUMÉ

Anaplastic lymphoma kinase (ALK) rearrangements are present in about 5-6% of non-small cell lung cancer (NSCLC) cases and associated with increased risks of central nervous system (CNS) involvement. Envonalkib, a novel ALK inhibitor, demonstrated promising anti-tumor activity and safety in advanced ALK-positive NSCLC in the first-in-human phase I study. This phase III trial (ClinicalTrials.gov NCT04009317) investigated the efficacy and safety of first-line envonalkib in advanced ALK-positive NSCLC cases. Totally 264 participants were randomized 1:1 to receive envonalkib (n = 131) or crizotinib (n = 133). Median independent review committee (IRC)-assessed progression-free survival (PFS) times were 24.87 (95% confidence interval [CI]: 15.64-30.36) and 11.60 (95% CI: 8.28-13.73) months in the envonalkib and crizotinib groups, respectively (hazard ratio [HR] = 0.47, 95% CI: 0.34-0.64, p < 0.0001). IRC-assessed confirmed objective response rate (ORR) was higher (81.68% vs. 70.68%, p = 0.056) and duration of response was longer (median, 25.79 [95% CI, 16.53-29.47] vs. 11.14 [95% CI, 9.23-16.59] months, p = 0.0003) in the envonalkib group compared with the crizotinib group. In participants with baseline brain target lesions, IRC-assessed CNS-ORR was improved with envonalkib compared with crizotinib (78.95% vs. 23.81%). Overall survival (OS) data were immature, and median OS was not reached in either group (HR = 0.84, 95% CI: 0.48-1.47, p = 0.5741). The 12-month OS rates were 90.6% (95% CI, 84.0%-94.5%) and 89.4% (95% CI, 82.8%-93.6%) in the envonalkib and crizotinib groups, respectively. Grade ≥3 treatment-related adverse events were observed in 55.73% and 42.86% of participants in the envonalkib and crizotinib groups, respectively. Envonalkib significantly improved PFS and delayed brain metastasis progression in advanced ALK-positive NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Inhibiteurs de protéines kinases/usage thérapeutique , Kinase du lymphome anaplasique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...