Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20.713
Filtrer
1.
J Gene Med ; 26(7): e3710, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967229

RÉSUMÉ

BACKGROUND: Patients with non-small cell lung cancer (NSCLC) are susceptible to coronavirus disease-2019 (COVID-19), but current treatments are limited. Icariside II (IS), a flavonoid compound derived from the plant epimedin, showed anti-cancer,anti-inflammation and immunoregulation effects. The present study aimed to evaluate the possible effect and underlying mechanisms of IS on NSCLC patients with COVID-19 (NSCLC/COVID-19). METHODS: NSCLC/COVID-19 targets were defined as the common targets of NSCLC (collected from The Cancer Genome Atlas database) and COVID-19 targets (collected from disease database of Genecards, OMIM, and NCBI). The correlations of NSCLC/COVID-19 targets and survival rates in patients with NSCLC were analyzed using the survival R package. Prognostic analyses were performed using univariate and multivariate Cox proportional hazards regression models. Furthermore, the targets in IS treatment of NSCLC/COVID-19 were defined as the overlapping targets of IS (predicted from drug database of TMSCP, HERBs, SwissTarget Prediction) and NSCLC/COVID-19 targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis of these treatment targets were performed aiming to understand the biological process, cellular component, molecular function and signaling pathway. The hub targets were analyzed by a protein-protein interaction network and the binding capacity with IS was characterized by molecular docking. RESULTS: The hub targets for IS in the treatment of NSCLC/COVID-19 includes F2, SELE, MMP1, MMP2, AGTR1 and AGTR2, and the molecular docking results showed that the above target proteins had a good binding degree to IS. Network pharmacology showed that IS might affect the leucocytes migration, inflammation response and active oxygen species metabolic process, as well as regulate the interleukin-17, tumor necrosus factor and hypoxia-inducible factor-1 signaling pathway in NSCLC/COVID-19. CONCLUSIONS: IS may enhance the therapeutic efficacy of current clinical anti-inflammatory and anti-cancer therapy to benefit patients with NSCLC combined with COVID-19.


Sujet(s)
COVID-19 , Carcinome pulmonaire non à petites cellules , Flavonoïdes , Tumeurs du poumon , Simulation de docking moléculaire , Pharmacologie des réseaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , COVID-19/virologie , COVID-19/métabolisme , Flavonoïdes/usage thérapeutique , Flavonoïdes/composition chimique , Flavonoïdes/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/métabolisme , Traitements médicamenteux de la COVID-19 , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Pronostic
2.
Pathol Oncol Res ; 30: 1611733, 2024.
Article de Anglais | MEDLINE | ID: mdl-38953007

RÉSUMÉ

Lung cancer is a leading cause of cancer-related death worldwide in both men and women, however mortality in the US and EU are recently declining in parallel with the gradual cut of smoking prevalence. Consequently, the relative frequency of adenocarcinoma increased while that of squamous and small cell carcinomas declined. During the last two decades a plethora of targeted drug therapies have appeared for the treatment of metastasizing non-small cell lung carcinomas (NSCLC). Personalized oncology aims to precisely match patients to treatments with the highest potential of success. Extensive research is done to introduce biomarkers which can predict the effectiveness of a specific targeted therapeutic approach. The EGFR signaling pathway includes several sufficient targets for the treatment of human cancers including NSCLC. Lung adenocarcinoma may harbor both activating and resistance mutations of the EGFR gene, and further, mutations of KRAS and BRAF oncogenes. Less frequent but targetable genetic alterations include ALK, ROS1, RET gene rearrangements, and various alterations of MET proto-oncogene. In addition, the importance of anti-tumor immunity and of tumor microenvironment has become evident recently. Accumulation of mutations generally trigger tumor specific immune defense, but immune protection may be upregulated as an aggressive feature. The blockade of immune checkpoints results in potential reactivation of tumor cell killing and induces significant tumor regression in various tumor types, such as lung carcinoma. Therapeutic responses to anti PD1-PD-L1 treatment may correlate with the expression of PD-L1 by tumor cells. Due to the wide range of diagnostic and predictive features in lung cancer a plenty of tests are required from a single small biopsy or cytology specimen, which is challenged by major issues of sample quantity and quality. Thus, the efficacy of biomarker testing should be warranted by standardized policy and optimal material usage. In this review we aim to discuss major targeted therapy-related biomarkers in NSCLC and testing possibilities comprehensively.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Proto-oncogène Mas , Humains , 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 , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme
3.
Gen Physiol Biophys ; 43(4): 301-312, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953570

RÉSUMÉ

Vascular endothelial growth factor A (VEGFA) is an important regulator for non-small cell lung cancer (NSCLC). Our study aimed to reveal its upstream pathway to provide new ideas for developing the therapeutic targets of NSCLC. The mRNA and protein levels of VEGFA, ubiquitin-specific peptidase 35 (USP35), and FUS were determined by quantitative real-time PCR and Western blot. Cell proliferation, apoptosis, invasion and angiogenesis were detected using CCK8 assay, EdU assay, flow cytometry, transwell assay and tube formation assay. The interaction between USP35 and VEGFA was assessed by Co-IP assay and ubiquitination assay. Animal experiments were performed to assess USP35 and VEGFA roles in vivo. VEGFA had elevated expression in NSCLC tissues and cells. Interferences of VEGFA inhibited NSCLC cell proliferation, invasion, angiogenesis, and increased apoptosis. USP35 could stabilize VEGFA protein level by deubiquitination, and USP35 knockdown suppressed NSCLC cell growth, invasion and angiogenesis via reducing VEGFA expression. FUS interacted with USP35 to promote its mRNA stability, thereby positively regulating VEGFA expression. Also, USP35 silencing could reduce NSCLC tumorigenesis by downregulating VEGFA. FUS-stabilized USP35 facilitated NSCLC cell growth, invasion and angiogenesis through deubiquitinating VEGFA, providing a novel idea for NSCLC treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Néovascularisation pathologique , Protéine FUS de liaison à l'ARN , Ubiquitination , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Invasion tumorale/génétique , Lignée cellulaire tumorale , Souris , Animaux , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris nude ,
4.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38955468

RÉSUMÉ

In addition to mitochondrial DNA, mitochondrial double-stranded RNA (mtdsRNA) is exported from mitochondria. However, specific channels for RNA transport have not been demonstrated. Here, we begin to characterize channel candidates for mtdsRNA export from the mitochondrial matrix to the cytosol. Down-regulation of SUV3 resulted in the accumulation of mtdsRNAs in the matrix, whereas down-regulation of PNPase resulted in the export of mtdsRNAs to the cytosol. Targeting experiments show that PNPase functions in both the intermembrane space and matrix. Strand-specific sequencing of the double-stranded RNA confirms the mitochondrial origin. Inhibiting or down-regulating outer membrane proteins VDAC1/2 and BAK/BAX or inner membrane proteins PHB1/2 strongly attenuated the export of mtdsRNAs to the cytosol. The cytosolic mtdsRNAs subsequently localized to large granules containing the stress protein TIA-1 and activated the type 1 interferon stress response pathway. Abundant mtdsRNAs were detected in a subset of non-small-cell lung cancer cell lines that were glycolytic, indicating relevance in cancer biology. Thus, we propose that mtdsRNA is a new damage-associated molecular pattern that is exported from mitochondria in a regulated manner.


Sujet(s)
Cytosol , Mitochondries , Prohibitines , ARN double brin , ARN mitochondrial , Humains , Cytosol/métabolisme , Mitochondries/métabolisme , ARN double brin/métabolisme , ARN mitochondrial/métabolisme , ARN mitochondrial/génétique , Lignée cellulaire tumorale , Protéines de répression/métabolisme , Protéines de répression/génétique , Transport des ARN , Exoribonucleases/métabolisme , Exoribonucleases/génétique , Canal anionique-1 voltage-dépendant/métabolisme , Canal anionique-1 voltage-dépendant/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines mitochondriales
5.
BMC Pulm Med ; 24(1): 309, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956553

RÉSUMÉ

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


Sujet(s)
Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Récepteurs ErbB/génétique , Sujet âgé , Pronostic , Études de cohortes , Marqueurs biologiques tumoraux/génétique , Immunothérapie , Carboplatine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pémétrexed/usage thérapeutique , Médecine de précision , Séquençage nucléotidique à haut débit , Antinéoplasiques/usage thérapeutique
7.
Acta Oncol ; 63: 542-551, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967220

RÉSUMÉ

BACKGROUND: Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are oncogenic drivers. Using the Auria Biobank in Finland, we aimed to identify and characterize patients with these gene fusions, and describe their clinical and tumor characteristics, treatments received, and outcomes. MATERIAL AND METHODS: We evaluated pediatrics with any solid tumor type and adults with colorectal cancer (CRC), non-small cell lung cancer (NSCLC), sarcoma, or salivary gland cancer. We determined tropomyosin receptor kinase (TRK) protein expression by pan-TRK immunohistochemistry (IHC) staining of tumor samples from the Auria Biobank, scored by a certified pathologist. NTRK gene fusion was confirmed by next generation sequencing (NGS). All 2,059 patients were followed-up starting 1 year before their cancer diagnosis. RESULTS: Frequency of NTRK gene fusion tumors was 3.1% (4/127) in pediatrics, 0.7% (8/1,151) for CRC, 0.3% (1/288) for NSCLC, 0.9% (1/114) for salivary gland cancer, and 0% (0/379) for sarcoma. Among pediatrics there was one case each of fibrosarcoma (TPM3::NTRK1), Ewing's sarcoma (LPPR1::NTRK2), primitive neuroectodermal tumor (DAB2IP::NTRK2), and papillary thyroid carcinoma (RAD51B::NTRK3). Among CRC patients, six harbored tumors with NTRK1 fusions (three fused with TPM3), one harbored a NTRK3::GABRG1 fusion, and the other a NTRK2::FXN/LPPR1 fusion. Microsatellite instability was higher in CRC patients with NTRK gene fusion tumors versus wild-type tumors (50.0% vs. 4.4%). Other detected fusions were SGCZ::NTRK3 (NSCLC) and ETV6::NTRK3 (salivary gland cancer). Four patients (three CRC, one NSCLC) received chemotherapy; one patient (with CRC) received radiotherapy. CONCLUSION: NTRK gene fusions are rare in adult CRC, NSCLC, salivary tumors, sarcoma, and pediatric solid tumors.


Sujet(s)
Récepteur trkA , Récepteur trkC , Humains , Finlande/épidémiologie , Mâle , Enfant , Femelle , Adulte , Adulte d'âge moyen , Adolescent , Récepteur trkA/génétique , Enfant d'âge préscolaire , Jeune adulte , Récepteur trkC/génétique , Sujet âgé , Biobanques , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Fusion de gènes , Sarcomes/génétique , Sarcomes/anatomopathologie , Tumeurs des glandes salivaires/génétique , Tumeurs des glandes salivaires/anatomopathologie , Récepteur trkB/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Nourrisson , Protéines de fusion oncogènes/génétique , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Séquençage nucléotidique à haut débit , Glycoprotéines membranaires
8.
Front Immunol ; 15: 1413956, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975340

RÉSUMÉ

Introduction: Younger patients with non-small cell lung cancer (NSCLC) (<50 years) represent a significant patient population with distinct clinicopathological features and enriched targetable genomic alterations compared to older patients. However, previous studies of younger NSCLC suffer from inconsistent findings, few studies have incorporated sex into their analyses, and studies targeting age-related differences in the tumor immune microenvironment are lacking. Methods: We performed a retrospective analysis of 8,230 patients with NSCLC, comparing genomic alterations and immunogenic markers of younger and older patients while also considering differences between male and female patients. We defined older patients as those ≥65 years and used a 5-year sliding threshold from <45 to <65 years to define various groups of younger patients. Additionally, in an independent cohort of patients with NSCLC, we use our observations to inform testing of the combinatorial effect of age and sex on survival of patients given immunotherapy with or without chemotherapy. Results: We observed distinct genomic and immune microenvironment profiles for tumors of younger patients compared to tumors of older patients. Younger patient tumors were enriched in clinically relevant genomic alterations and had gene expression patterns indicative of reduced immune system activation, which was most evident when analyzing male patients. Further, we found younger male patients treated with immunotherapy alone had significantly worse survival compared to male patients ≥65 years, while the addition of chemotherapy reduced this disparity. Contrarily, we found younger female patients had significantly better survival compared to female patients ≥65 years when treated with immunotherapy plus chemotherapy, while treatment with immunotherapy alone resulted in similar outcomes. Discussion: These results show the value of comprehensive genomic and immune profiling (CGIP) for informing clinical treatment of younger patients with NSCLC and provides support for broader coverage of CGIP for younger patients with advanced NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Microenvironnement tumoral , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/thérapie , Mâle , Femelle , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/thérapie , Adulte d'âge moyen , Sujet âgé , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Facteurs âges , Études rétrospectives , Facteurs sexuels , Adulte , Génomique/méthodes , Marqueurs biologiques tumoraux/génétique , Analyse de profil d'expression de gènes , Immunothérapie
9.
JCO Precis Oncol ; 8: e2300718, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38976829

RÉSUMÉ

PURPOSE: To use modern machine learning approaches to enhance and automate the feature extraction from the longitudinal circulating tumor DNA (ctDNA) data and to improve the prediction of survival and disease progression, risk stratification, and treatment strategies for patients with 1L non-small cell lung cancer (NSCLC). MATERIALS AND METHODS: Using IMpower150 trial data on patients with untreated metastatic NSCLC treated with atezolizumab and chemotherapies, we developed a machine learning algorithm to extract predictive features from ctDNA kinetics, improving survival and progression prediction. We analyzed kinetic data from 17 ctDNA summary markers, including cell-free DNA concentration, allele frequency, tumor molecules in plasma, and mutation counts. RESULTS: Three hundred and ninety-eight patients with ctDNA data (206 in training and 192 in validation) were analyzed. Our models outperformed existing workflow using conventional temporal ctDNA features, raising overall survival (OS) concordance index to 0.72 and 0.71 from 0.67 and 0.63 for C3D1 and C4D1, respectively, and substantially improving progression-free survival (PFS) to approximately 0.65 from the previous 0.54-0.58, a 12%-20% increase. Additionally, they enhanced risk stratification for patients with NSCLC, achieving clear OS and PFS separation. Distinct patterns of ctDNA kinetic characteristics (eg, baseline ctDNA markers, depth of ctDNA responses, and timing of ctDNA clearance, etc) were revealed across the risk groups. Rapid and complete ctDNA clearance appears essential for long-term clinical benefit. CONCLUSION: Our machine learning approach offers a novel tool for analyzing ctDNA kinetics, extracting critical features from longitudinal data, improving our understanding of the link between ctDNA kinetics and progression/mortality risks, and optimizing personalized immunotherapies for 1L NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , ADN tumoral circulant , Évolution de la maladie , Immunothérapie , Tumeurs du poumon , Apprentissage machine , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/sang , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , ADN tumoral circulant/sang , Tumeurs du poumon/génétique , Tumeurs du poumon/sang , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Immunothérapie/méthodes , Mâle , Femelle , Adulte d'âge moyen , Anticorps monoclonaux humanisés/usage thérapeutique , Sujet âgé , Survie sans progression
10.
J Manag Care Spec Pharm ; 30(7): 660-671, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38950156

RÉSUMÉ

BACKGROUND: Oncology clinical trial enrollment is strongly recommended for patients with cancer who are not eligible for established and approved therapies. Many trials are specific to biomarker-targeted therapies, which are typically managed as specialty pharmacy services. Comprehensive genomic profiling (CGP) of advanced cancers has been shown to detect biomarkers, guide targeted treatment, improve outcomes, and result in the clinical trial enrollment of patients, which is modeled to offset pharmacy costs experienced by US payers, yet payer policy coverage remains inconsistent. A common concern limiting coverage of CGP by payers is the potential of identifying biomarkers beyond guideline-recommended treatments, which creates a perception that insurance companies are being positioned to "pay for research." However, these biomarkers can increase clinical trial eligibility, and specialty pharmacy management may have an interest in maximizing the clinical trial enrollment of members. OBJECTIVE: To investigate if clinical trial enrollment following liquid biopsy CGP for non-small cell lung cancer (NSCLC) is clinically and/or economically impactful from a payer claims perspective. METHODS: Clinical and economic outcomes were studied using a real-world clinical genomic database (including payer claims data) from patients with NSCLC who enrolled in clinical trials immediately following liquid biopsy CGP (using Guardant360) and matched NSCLC patient controls also tested with liquid biopsy CGP. RESULTS: Real-world overall survival was significantly (log-rank P < 0.0001) better for patients enrolled in clinical trials with similar costs of care, albeit with more outpatient encounters among those enrolled compared with matched controls. CONCLUSIONS: The results, together with previous analyses, suggest that, in addition to the clinical benefits associated with targeted therapies directed by CGP and other testing approaches, payers and specialty pharmacy managers may consider clinical trial direction and enrollment as a clinical and economic benefit of liquid biopsy CGP and adopt this into coverage decision frameworks and formularies.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/économie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/économie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Biopsie liquide/économie , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Essais cliniques comme sujet/économie , Marqueurs biologiques tumoraux/génétique , Génomique/économie , États-Unis
11.
J Manag Care Spec Pharm ; 30(7): 649-659, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38950160

RÉSUMÉ

BACKGROUND: Clinical practice guidelines recommend broad-panel genomic profiling to identify actionable genomic alterations for patients with advanced non-small cell lung cancer (aNSCLC). OBJECTIVE: To assess the cost-effectiveness of large-panel next-generation sequencing (LP-NGS) compared with current empirical single-gene test (SGT) patterns to inform first-line treatment decisions for patients with aNSCLC from a US commercial payer perspective, accounting for the effect of testing turnaround time and time to treatment initiation. METHODS: We developed a discrete-event simulation model to estimate the impact of LP-NGS vs SGT for patients with nonsquamous aNSCLC. Discrete events and timing included testing patterns, receipt of the initial test result, treatment initiation (targeted vs nontargeted therapies), switching, retesting, rebiopsies, clinical trial participation, progression on therapy, and death. LP-NGS and SGT cohorts each comprised 100,000 adults with aNSCLC simulated over a 5-year postdiagnosis period, assumed to have the same distribution of genomic alterations. The model predicted the proportion of patients receiving appropriate first-line therapy according to clinical practice guidelines. Economic outcomes included expected life-years gained, quality-adjusted life-years, and the total costs of care over 5 years. Sensitivity and scenario analyses explored the robustness of the base-case model results. RESULTS: In the base-case model, LP-NGS was likely to identify more alterations than SGT. Total 5-year costs per patient were $539,658 for LP-NGS and $544,550 for SGT (net difference, $4,892 lower costs per patient for LP-NGS), which is likely to be cost-effective 95.1% of the time. The most influential model parameters on the 5-year total costs of care were preprogression nondrug medical costs on nontargeted therapy, NGS turnaround time, and clinical trial participation. CONCLUSIONS: This study suggests that LP-NGS to guide first-line treatment decisions is clinically more appropriate (more likely to identify alterations and subsequently allocate patients to clinically appropriate therapy) and provides a dominant cost-effectiveness treatment strategy over 5 years for patients with newly diagnosed aNSCLC in the United States.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Analyse coût-bénéfice , Séquençage nucléotidique à haut débit , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/économie , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/économie , Séquençage nucléotidique à haut débit/économie , Années de vie ajustées sur la qualité , Mâle , Femelle , Adulte d'âge moyen
12.
BMJ Open ; 14(7): e078335, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969367

RÉSUMÉ

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


Sujet(s)
Acrylamides , Dérivés de l'aniline , Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Radiochirurgie , Revues systématiques comme sujet , Humains , Acrylamides/usage thérapeutique , Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/secondaire , Association thérapeutique , Récepteurs ErbB/génétique , Indoles , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Méta-analyse comme sujet , Mutation , Études prospectives , Pyrimidines , Radiochirurgie/méthodes , Essais contrôlés randomisés comme sujet , Plan de recherche
13.
J Cancer Res Clin Oncol ; 150(7): 335, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38969831

RÉSUMÉ

BACKGROUND: Ubiquilin-4 (UBQLN4), a member of the ubiquilin family, has received limited attention in cancer research to date. Here, we investigated for the first time the functional role and mechanism of UBQLN4 in non-small cell lung cancer (NSCLC). METHODS: The Cancer Genome Atlas (TCGA) database was employed to validate UBQLN4 as a differentially expressed gene. Expression differences of UBQLN4 in NSCLC cells and tissues were assessed using immunohistochemistry (IHC) experiment and western blotting (WB) experiment. Kaplan-Meier analysis was conducted to examine the association between UBQLN4 expression and NSCLC prognosis. Functional analyses of UBQLN4 were performed through cell counting kit-8 (CCK-8), colony formation, and transwell invasion assays. The impact of UBQLN4 on tumor-associated signaling pathways was assessed using the path scan intracellular signaling array. In vivo tumorigenesis experiments were conducted to further investigate the influence of UBQLN4 on tumor formation. RESULTS: UBQLN4 exhibited up-regulation in both NSCLC tissues and cells. Additionally, over-expression of UBQLN4 was associated with an unfavorable prognosis in NSCLC patients. Functional loss analyses demonstrated that inhibiting UBQLN4 could suppress the proliferation and invasion of NSCLC cells in both in vitro and in vivo settings. Conversely, functional gain experiments yielded opposite results. Path scan intracellular signaling array results suggested that the role of UBQLN4 is associated with the PI3K/AKT pathway, a correlation substantiated by in vitro and in vivo tumorigenesis experiments. CONCLUSION: We validated that UBQLN4 promotes proliferation and invasion of NSCLC cells by activating the PI3K/AKT pathway, thereby facilitating the progression of NSCLC. These findings underscore the potential of targeting UBQLN4 as a therapeutic strategy for NSCLC.


Sujet(s)
Protéines associées à l'autophagie , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Animaux , Souris , Femelle , Mâle , Pronostic , Lignée cellulaire tumorale , Souris nude , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Adulte d'âge moyen , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de transport , Protéines nucléaires
14.
Per Med ; 21(4): 205-209, 2024.
Article de Anglais | MEDLINE | ID: mdl-38958204

RÉSUMÉ

We report the case of an 87-year-old female patient who was diagnosed with metastatic non-small-cell lung cancer harboring MET exon 14 skipping mutation (MET ex14) and PD-L1 expression of 60%. A first-line treatment with atezolizumab was started with primary resistance. Then, a second-line treatment with capmatinib, a selective type Ib MET tyrosine kinase inhibitor, was started, achieving a partial response. The patient is still alive and on treatment with capmatinib 300 mg twice daily after 20 months, with a good tolerability and no evidence of disease progression.In summary, our patient experienced a long-lasting response (>18 months) with capmatinib as second-line treatment. Further analyses evaluating the efficacy and tolerability of MET tyrosine kinase inhibitors are warranted, especially in the elderly, a non-small-cell lung cancer population whose tumors could more frequently harbor MET ex14 mutation.


[Box: see text].


Sujet(s)
Carcinome pulmonaire non à petites cellules , Exons , Tumeurs du poumon , Mutation , Protéines proto-oncogènes c-met , Humains , Femelle , Protéines proto-oncogènes c-met/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Sujet âgé de 80 ans ou plus , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mutation/génétique , Exons/génétique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/administration et posologie , Benzamides/usage thérapeutique , Benzamides/effets indésirables , Résultat thérapeutique , Acrylamides/usage thérapeutique , Acrylamides/administration et posologie , Acrylamides/effets indésirables , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Imidazoles , Triazines
15.
Medicine (Baltimore) ; 103(28): e38789, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996163

RÉSUMÉ

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


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Mutation , Quinazolinones , Humains , Femelle , Acrylamides/usage thérapeutique , Dérivés de l'aniline/usage thérapeutique , Récepteurs ErbB/génétique , Sujet âgé , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Résistance aux médicaments antinéoplasiques/génétique , Quinazolinones/usage thérapeutique , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Antinéoplasiques/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Indoles , Pyrimidines
16.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000069

RÉSUMÉ

Lung cancer is a leading cause of cancer-related morbidity and mortality worldwide. Metastases in the brain are a common hallmark of advanced stages of the disease, contributing to a dismal prognosis. Lung cancer can be broadly classified as either small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). NSCLC represents the most predominant histology subtype of lung cancer, accounting for the majority of lung cancer cases. Recent advances in molecular genetics, coupled with innovations in small molecule drug discovery strategies, have facilitated both the molecular classification and precision targeting of NSCLC based on oncogenic driver mutations. Furthermore, these precision-based strategies have demonstrable efficacy across the blood-brain barrier, leading to positive outcomes in patients with brain metastases. This review provides an overview of the clinical features of lung cancer brain metastases, as well as the molecular mechanisms that drive NSCLC oncogenesis. We also explore how precision medicine-based strategies can be leveraged to improve NSCLC brain metastases.


Sujet(s)
Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Médecine de précision/méthodes , Mutation , Barrière hémato-encéphalique/métabolisme , Antinéoplasiques/usage thérapeutique
17.
BMC Cancer ; 24(1): 842, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009968

RÉSUMÉ

BACKGROUND: Targeted therapy is now the standard of care in driver-oncogene-positive non-small cell lung cancer (NSCLC). Its initial clinical effects are remarkable. However, almost all patients experience treatment resistance to targeted therapy. Hence, chemotherapy is considered a subsequent treatment option. In patients with driver-oncogene-negative NSCLC, combined immune checkpoint inhibitors (ICIs) and chemotherapy as the first-line therapy has been found to be beneficial. However, the efficacy of ICI plus chemotherapy against driver-oncogene-positive NSCLC other than epidermal growth factor receptor mutation and anaplastic lymphoma kinase fusion is unclear. METHODS: Using the hospital medical records, we retrospectively reviewed advanced or recurrent NSCLC patients who were treated with chemotherapy with or without ICIs at Aichi Cancer Center Hospital between January 2014 and January 2023. Patients with druggable rare mutations such as KRAS-G12C, MET exon 14 skipping, HER2 20 insertion, BRAF-V600E mutations, and ROS1 and RET rearrangements were analyzed. RESULTS: In total, 61 patients were included in this analysis. ICI plus chemotherapy was administered in 36 patients (the ICI-chemo group) and chemotherapy in 25 patients (the chemo group). The median progression-free survival (PFS) rates were 14.0 months in the ICI-chemo group and 4.8 months in the chemo group (hazard ratio [HR] = 0.54, 95% confidence interval [CI] = 0.28-1.01). The median overall survival rates were 31.3 and 21.7 months in the ICI-chemo and chemo groups, respectively (HR = 0.70, 95% CI = 0.33-1.50). Multivariate Cox regression analysis of PFS revealed that HER2 exon 20 insertion mutation was significantly associated with a poorer PFS (HR: 2.39, 95% CI: 1.19-4.77, P = 0.014). Further, ICI-chemo treatment was significantly associated with a better PFS (HR: 0.48, 95% CI: 0.25-0.91, P = 0.025). CONCLUSION: ICI plus chemotherapy improves treatment efficacy in rare driver-oncogene-positive NSCLC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , 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/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Femelle , Études rétrospectives , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes B-raf/génétique , Récepteur ErbB-2/génétique , Protéines proto-oncogènes c-ret/génétique , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes/génétique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , Survie sans progression , Résultat thérapeutique
18.
Int J Biol Sci ; 20(9): 3285-3301, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993559

RÉSUMÉ

Metabolic reprogramming is one of the essential features of tumors that may dramatically contribute to cancer metastasis. Employing liquid chromatography-tandem mass spectrometry-based metabolomics, we analyzed the metabolic profile from 12 pairwise serum samples of NSCLC brain metastasis patients before and after CyberKnife Stereotactic Radiotherapy. We evaluated the histopathological architecture of 144 surgically resected NSCLC brain metastases. Differential metabolites were screened and conducted for functional clustering and annotation. Metabolomic profiling identified a pathway that was enriched in the metabolism of branched-chain amino acids (BCAAs). Pathologically, adenocarcinoma with a solid growth pattern has a higher propensity for brain metastasis. Patients with high BCAT1 protein levels in lung adenocarcinoma tissues were associated with a poor prognosis. We found that brain NSCLC cells had elevated catabolism of BCAAs, which led to a depletion of α-KG. This depletion, in turn, reduced the expression and activity of the m6A demethylase ALKBH5. Thus, ALKBH5 inhibition participated in maintaining the m6A methylation of mesenchymal genes and promoted the occurrence of epithelial-mesenchymal transition (EMT) in NSCLC cells and the proliferation of NSCLC cells in the brain. BCAA catabolism plays an essential role in the metastasis of NSCLC cells.


Sujet(s)
AlkB Homolog 5, RNA demethylase , Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Transition épithélio-mésenchymateuse/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , AlkB Homolog 5, RNA demethylase/métabolisme , AlkB Homolog 5, RNA demethylase/génétique , Mâle , Femelle , Acides aminés à chaine ramifiée/métabolisme , Adulte d'âge moyen , Lignée cellulaire tumorale , Transaminases
19.
Cells ; 13(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38994972

RÉSUMÉ

Understanding tumor-host immune interactions and the mechanisms of lung cancer response to immunotherapy is crucial. Current preclinical models used to study this often fall short of capturing the complexities of human lung cancer and lead to inconclusive results. To bridge the gap, we introduce two new murine monoclonal lung cancer cell lines for use in immunocompetent orthotopic models. We demonstrate how our cell lines exhibit immunohistochemical protein expression (TTF-1, NapA, PD-L1) and common driver mutations (KRAS, p53, and p110α) seen in human lung adenocarcinoma patients, and how our orthotopic models respond to combination immunotherapy in vivo in a way that closely mirrors current clinical outcomes. These new lung adenocarcinoma cell lines provide an invaluable, clinically relevant platform for investigating the intricate dynamics between tumor and the immune system, and thus potentially contributes to a deeper understanding of immunotherapeutic approaches to lung cancer treatment.


Sujet(s)
Adénocarcinome pulmonaire , Carcinome pulmonaire non à petites cellules , Immunothérapie , Tumeurs du poumon , Animaux , Immunothérapie/méthodes , Humains , Lignée cellulaire tumorale , Souris , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Carcinome pulmonaire non à petites cellules/thérapie , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/thérapie , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Modèles animaux de maladie humaine , Femelle
20.
Sci Rep ; 14(1): 15877, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38982267

RÉSUMÉ

Develop a radiomics nomogram that integrates deep learning, radiomics, and clinical variables to predict epidermal growth factor receptor (EGFR) mutation status in patients with stage I non-small cell lung cancer (NSCLC). We retrospectively included 438 patients who underwent curative surgery and completed driver-gene mutation tests for stage I NSCLC from four academic medical centers. Predictive models were established by extracting and analyzing radiomic features in intratumoral, peritumoral, and habitat regions of CT images to identify EGFR mutation status in stage I NSCLC. Additionally, three deep learning models based on the intratumoral region were constructed. A nomogram was developed by integrating representative radiomic signatures, deep learning, and clinical features. Model performance was assessed by calculating the area under the receiver operating characteristic (ROC) curve. The established habitat radiomics features demonstrated encouraging performance in discriminating between EGFR mutant and wild-type, with predictive ability superior to other single models (AUC 0.886, 0.812, and 0.790 for the training, validation, and external test sets, respectively). The radiomics-based nomogram exhibited excellent performance, achieving the highest AUC values of 0.917, 0.837, and 0.809 in the training, validation, and external test sets, respectively. Decision curve analysis (DCA) indicated that the nomogram provided a higher net benefit than other radiomics models, offering valuable information for treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Apprentissage profond , Récepteurs ErbB , Tumeurs du poumon , Mutation , Nomogrammes , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/imagerie diagnostique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs ErbB/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/imagerie diagnostique , Tumeurs du poumon/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Études rétrospectives , Tomodensitométrie/méthodes , Stadification tumorale , Adulte , Courbe ROC , Sujet âgé de 80 ans ou plus ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...