Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 78.514
Filtrer
1.
Cancer Med ; 13(17): e70197, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39233657

RÉSUMÉ

BACKGROUD: Supernatants from various cytological samples, including body cavity effusion, sputum, bronchoalveolar lavage fluid (BALF), and needle aspiration, have been validated for detecting genetic alterations using cell-free DNA (cfDNA) in patients with non-small cell lung cancer (NSCLC). However, the sensitivity of fusion variations detection remains challenging. The protection of cell-free RNA (cfRNA) is critical for resolving the issue. METHODS: A protective solution (PS) was applied for preserving cfRNA in cytological supernatant (CS), and the quality of protected cfRNA was assessed by cycle threshold (CT) values from reverse transcription quantitative polymerase chain reaction (RT-qPCR). Furthermore, we collected an additional set of malignant cytological and matched tumor samples from 84 NSCLC patients, cfDNA & cfRNA extraction and double detection for driver gene mutations was validated using the multi-gene mutations detection by RT-qPCR. RESULTS: Under the optimal protection system, 91.0% (101/111) of cfRNA were protected effectively. Among the 84 NSCLC patient samples, seven cytological samples failed the tests. In comparison with tumor samples, the overall sensitivity and specificity of detecting driver genes of supernatant cfDNA and cfRNA were 93.8% (74/77) and 100% (77/77), respectively. Notably, when focusing exclusively on patients with fusion gene changes, both sensitivity and specificity reached 100% (11/11) for EML4-ALK, ROS1, RET fusions, and MET ex14 skipping. CONCLUSION: These findings suggest that cfDNA & cfRNA extraction and double detection strategy recommended in this study improve the accuracy of driver genes mutations test, especially for RNA-based assay.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Acides nucléiques acellulaires , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/diagnostic , Acides nucléiques acellulaires/génétique , Mutation , Mâle , Femelle , Marqueurs biologiques tumoraux/génétique , Sensibilité et spécificité , Adulte d'âge moyen , Sujet âgé , Protéines de fusion oncogènes/génétique , Protein-tyrosine kinases , Protéines proto-oncogènes
4.
Sci Rep ; 14(1): 20557, 2024 09 04.
Article de Anglais | MEDLINE | ID: mdl-39231973

RÉSUMÉ

The aim of this study was to establish and validate the precision of a novel radiomics approach that integrates 18Fluorine-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)-computed tomography (CT) scan data with clinical information to improve the prognostication of survival rates in patients diagnosed with stage III Non-Small Cell Lung Cancer (NSCLC) who are not candidates for surgery. We evaluated pretreatment 18F-FDG PET-CT scans from 156 individuals diagnosed with stage III inoperable NSCLC at Shandong Cancer Hospital. These individuals were divided into two groups: a training set comprising 110 patients and an internal validation set consisting of 46 patients. By employing random forest classifier and cox proportional hazards model , we identified and utilized relevant features to create predictive models and a nomogram. The effectiveness of these models was assessed through the use of the receiver operating characteristics(ROC) curves, Kaplan-Meier (KM) curves, and the application of the nomogram. Our findings showed that the combined model, which integrates both clinical and radiomic data, outperformed those based solely on clinical or radiomic features in predicting 3-year overall survival(OS). Furthermore, calibration plots revealed a high level of agreement between predicted and actual survival times. The research successfully established a predictive radiomics model that integrates 18F-FDG PET/CT imaging with clinical indicators to enhance survival predictions for patients with stage III inoperable NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Fluorodésoxyglucose F18 , Tumeurs du poumon , Stadification tumorale , Nomogrammes , Tomographie par émission de positons couplée à la tomodensitométrie , Humains , Carcinome pulmonaire non à petites cellules/imagerie diagnostique , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du poumon/imagerie diagnostique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Sujet âgé , Pronostic , Courbe ROC , Adulte , Estimation de Kaplan-Meier , Modèles des risques proportionnels ,
5.
J Laparoendosc Adv Surg Tech A ; 34(9): 798-807, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39288366

RÉSUMÉ

The treatment of non-small cell lung cancer (NSCLC) has evolved tremendously in recent decades as innovations in medical therapies advanced concomitantly with minimally invasive surgical techniques. Despite early skepticism regarding its benefits, video-assisted thoracoscopic surgery (VATS) techniques for the surgical resection of early-stage NSCLC have now become the standard of care. After being the subject of many studies since its inception, VATS has been shown to cause less postoperative pain, have shorter recovery time, and have fewer overall complications when compared to conventional open approaches. Furthermore, some studies have shown it to have comparable oncological outcomes, though more higher evidence studies are needed. Newer technologies and improved surgical instruments, advancements in nodule localization techniques, and improved preoperative staging procedures have allowed for the development of newer, less invasive techniques such as uniportal VATS and parenchymal-sparing sublobar resections, which might further improve postoperative rates of complications in specific cases. These minimally invasive approaches have allowed surgeons to offer surgery to high-risk patients and those who would otherwise not tolerate conventional thoracotomy, though some relative contraindications still exist. This review aims to describe the evolution of VATS lobectomy, current techniques, its indications, contraindications, preoperative testing, benefits, and outcomes in patients with stage I and II NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Stadification tumorale , Pneumonectomie , Chirurgie thoracique vidéoassistée , Humains , Chirurgie thoracique vidéoassistée/méthodes , Tumeurs du poumon/chirurgie , Tumeurs du poumon/anatomopathologie , Pneumonectomie/méthodes , Carcinome pulmonaire non à petites cellules/chirurgie , Carcinome pulmonaire non à petites cellules/anatomopathologie
6.
JCO Glob Oncol ; 10: e2400138, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39303192

RÉSUMÉ

PURPOSE: Anti-PD-1/PD(L)1-based combination therapy is the standard of care in first line (1L) for metastatic nonsquamous non-small cell lung cancer (mnsqNSCLC) without driver alterations. This study aimed to evaluate real-world clinical outcomes in this population. METHODS: Eligible physicians in the United States, Europe, and Japan abstracted information from medical charts of eligible adult patients with mnsqNSCLC (without EGFR/ALK, no known ROS1 alterations) who initiated 1L anti-PD(L)1-based combination therapy for mnsqNSCLC between 2017 and 2021. Kaplan-Meier analyses were used to assess overall survival (OS), time-to-treatment discontinuation (TTD), and real-world progression-free survival (rwPFS) after 1L initiation. RESULTS: Overall, 142 physicians contributed deidentified data from 430 patients' medical charts. The distribution of PD-L1 expression levels was 31.2% with tumor proportion score (TPS) <1%, 42.3% with TPS 1%-49%, and 26.5% with TPS ≥50%. In 1L, patients received anti-PD(L)1 + chemotherapy (84.6%), anti-PD(L)1 + anti-CTLA4 with or without chemotherapy (11.9%), and anti-PD(L)1 + chemotherapy + anti-vascular endothelial growth factor receptor (3.5%). The median OS was 21.7 months (TPS <1%: 18.3 months; TPS 1%-49%: 21.6 months; TPS ≥50%: 24.0 months). The median TTD was 11.0 months (TPS <1%: 9.1 months; TPS 1%-49%: 10.9 months; TPS ≥50%: 12.2 months). The median rwPFS was 11.2 months (TPS <1%: 9.3 months; TPS 1%-49%: 11.1 months; TPS ≥50%: 13.2 months). CONCLUSION: This study assessed the real-world clinical effectiveness of 1L anti-PD(L)1-based combination therapy for mnsqNSCLC. Results from this study were generally consistent with previous clinical trials and published real-world evidence in 1L mnsqNSCLC.


Sujet(s)
Antigène CD274 , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Mâle , Femelle , États-Unis , Sujet âgé , Europe , Japon , Adulte d'âge moyen , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Sujet âgé de 80 ans ou plus , Adulte , Études rétrospectives , Résultat thérapeutique
7.
PLoS One ; 19(9): e0310808, 2024.
Article de Anglais | MEDLINE | ID: mdl-39312569

RÉSUMÉ

BACKGROUND: PD-1/PD-L1 inhibitors (PI) have shown promising results in both neoadjuvant and adjuvant therapies for resectable non-small cell lung cancer (NSCLC). However, substantial evidence from large-scale studies is still lacking for their use in the perioperative setting (neoadjuvant plus adjuvant). This meta-analysis aims to evaluate the integration of perioperative PI (PPI) with neoadjuvant chemotherapy for resectable NSCLC. METHODS: To identify appropriate randomized controlled trials (RCTs), we thoroughly explored six different databases. The primary endpoint was survival, while the secondary measures included pathological responses and adverse events (AEs). RESULTS: Six RCTs involving 2941 patients were included. The PPI group significantly improved overall survival (OS) (hazard ratio [HR]: 0.62 [0.51, 0.77]), event-free survival (EFS) (HR: 0.57 [0.51, 0.64]), pathological complete response (risk ratio [RR]: 5.81 [4.47, 7.57]), and major pathological response (RR: 2.60 [1.77, 3.82]). Benefits in EFS were seen across all subgroups. OS rates at 12-48 months and EFS rates at 6-48 months were higher in the PPI cohort. Furthermore, the advantages in OS and EFS increased with prolonged survival times. The PPI group also exhibited higher rates of surgery and R0 resections. However, the PPI group experienced more grade 3-5 AEs, serious AEs, and treatment discontinuations due to AEs. CONCLUSIONS: The integration of perioperative PI with neoadjuvant chemotherapy can significantly improve survival and pathological responses for resectable NSCLC. However, the increased incidence of grade 3-5 AEs must be carefully evaluated.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Essais contrôlés randomisés comme sujet , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/chirurgie , Tumeurs du poumon/mortalité , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Traitement néoadjuvant/méthodes , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Antigène CD274/antagonistes et inhibiteurs
8.
Front Immunol ; 15: 1379812, 2024.
Article de Anglais | MEDLINE | ID: mdl-39315096

RÉSUMÉ

Introductions: Identifying patients with non-small cell lung cancer (NSCLC) who are optimal candidates for immunotherapy is a cornerstone in clinical decision-making. The tumor immune microenvironment (TIME) is intricately linked with both the prognosis of the malignancy and the efficacy of immunotherapeutic interventions. CD8+ T cells, and more specifically, tissue-resident memory CD8+ T cells [CD8+ tissue-resident memory T (TRM) cells] are postulated to be pivotal in orchestrating the immune system's assault on tumor cells. Nevertheless, the accurate quantification of immune cell infiltration-and by extension, the prediction of immunotherapeutic efficacy-remains a significant scientific frontier. Methods: In this study, we introduce a cutting-edge non-invasive radiomic model, grounded in TIME markers (CD3+ T, CD8+ T, and CD8+ TRM cells), to infer the levels of immune cell infiltration in NSCLC patients receiving immune checkpoint inhibitors and ultimately predict their response to immunotherapy. Data from patients who had surgical resections (cohort 1) were employed to construct a radiomic model capable of predicting the TIME. This model was then applied to forecast the TIME for patients under immunotherapy (cohort 2). Conclusively, the study delved into the association between the predicted TIME from the radiomic model and the immunotherapeutic outcomes of the patients. Result: For the immune cell infiltration radiomic prediction models in cohort 1, the AUC values achieved 0.765, 0.763, and 0.675 in the test set of CD3+ T, CD8+ T, and CD8+ TRM, respectively. While the AUC values for the TIME-immunotherapy predictive value were 0.651, 0.763, and 0.829 in the CD3-immunotherapy response model, CD8-immunotherapy response model, and CD8+ TRM-immunotherapy response model in cohort 2, respectively. The CD8+ TRM-immunotherapy model exhibited the highest predictive value and was significantly better than the CD3-immunotherapy model in predicting the immunotherapy response. The progression-free survival (PFS) analysis based on the predicted levels of CD3+ T, CD8+ T, and CD8+ TRM immune cell infiltration showed that the CD8+ T cell infiltration level was an independent factor (P=0.014, HR=0.218) with an AUC value of 0.938. Discussion: Our empirical evidence reveals that patients with substantial CD8+ T cell infiltration experience a markedly improved PFS compared with those with minimal infiltration, asserting the status of the CD8+ T cell as an independent prognosticator of PFS in the context of immunotherapy. Although CD8+ TRM cells demonstrated the greatest predictive accuracy for immunotherapy response, their predictive strength for PFS was marginally surpassed by that of CD8+ T cells. These insights advocate for the application of the proposed non-invasive radiomic model, which utilizes TIME analysis, as a reliable predictor for immunotherapy outcomes and PFS in NSCLC patients.


Sujet(s)
Lymphocytes T CD8+ , Carcinome pulmonaire non à petites cellules , Immunothérapie , Tumeurs du poumon , Lymphocytes TIL , Microenvironnement tumoral , Humains , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Femelle , Immunothérapie/méthodes , Lymphocytes T CD8+/immunologie , Mâle , Microenvironnement tumoral/immunologie , Adulte d'âge moyen , Sujet âgé , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Résultat thérapeutique , Pronostic ,
9.
Ann Med ; 56(1): 2405075, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39297299

RÉSUMÉ

INTRODUCTION: Artificial intelligence (AI) shows promise for evaluating solitary pulmonary nodules (SPNs) on computed tomography (CT). Accurately determining cancer invasiveness can guide treatment. We aimed to investigate quantitative CT parameters for invasiveness prediction. METHODS: Patients with stage 0-IB NSCLC after surgical resection were retrospectively analysed. Preoperative CTs were evaluated with specialized software for nodule segmentation and CT quantification. Pathology was the reference for invasiveness. Univariate and multivariate logistic regression assessed predictors of high-risk SPN. RESULTS: Three hundred and fifty-five SPN were included. On multivariate analysis, CT value mean and nodule type (ground glass opacity vs. solid) were independent predictors of high-risk SPN. The area under the curve (AUC) was 0.811 for identifying high-risk nodules. CONCLUSIONS: Quantitative CT measures and nodule type correlated with invasiveness. Software-based CT assessment shows potential for noninvasive prediction to guide extent of resection. Further prospective validation is needed, including comparison with benign nodules.


Sujet(s)
Intelligence artificielle , Tumeurs du poumon , Nodule pulmonaire solitaire , Tomodensitométrie , Humains , Nodule pulmonaire solitaire/imagerie diagnostique , Nodule pulmonaire solitaire/chirurgie , Nodule pulmonaire solitaire/anatomopathologie , Femelle , Mâle , Tomodensitométrie/méthodes , Adulte d'âge moyen , Études rétrospectives , Tumeurs du poumon/imagerie diagnostique , Tumeurs du poumon/chirurgie , Tumeurs du poumon/anatomopathologie , Sujet âgé , Carcinome pulmonaire non à petites cellules/imagerie diagnostique , Carcinome pulmonaire non à petites cellules/chirurgie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Adulte , Invasion tumorale , Sujet âgé de 80 ans ou plus
10.
Oncol Rep ; 52(5)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39301655

RÉSUMÉ

Lung cancer is increasingly recognized as a leading cause of cancer­related mortality. Immunotherapy has emerged as a promising therapeutic approach for lung cancer, particularly non­small cell lung cancer (NSCLC). Nonetheless, the response rate to programmed cell death 1 (PD­1) inhibitors remains less than optimal. It has been suggested that protein tyrosine phosphatase 1B (PTP1B) plays a crucial role in the development and progression of cancer by facilitating T cell expansion and cytotoxicity. Our previous research demonstrated that the combination of tumor necrosis factor receptor 2 (TNFR2) with immune activity treatments synergistically suppresses tumor growth. This insight led to exploring the efficacy of a combined treatment strategy involving PD­1 inhibitors, PTP1B inhibitors and TNFR2 antibodies (triple therapy) in NSCLC. In this context, the therapeutic effectiveness of these combination immunotherapies was validated in mouse models with NSCLC by analyzing the expansion and function of immune cells, thereby assessing their impact on tumor growth. The results indicated that inhibiting PTP1B decreases the expression of PD­L1 and TNFR2 on LLC cells, along with an increase in the proportion of CD4+T and CD8+T cells. Compared with other treatment groups, the triple therapy significantly reduced tumor volume in mice with NSCLC and extended their survival. Moreover, this combination therapy altered the distribution of myeloid­derived suppressor cells, dendritic cells, B cells and M1 macrophages, while increasing the proportion of CD8+T cells and reducing the proportion of Treg cells in the spleens, lymph nodes, and tumors of NSCLC models. The triple therapy also resulted in a decrease in PD­L1, PTP1B and TNFR2 expression within NSCLC tumor tissues in mice. Overall, the triple therapy effectively suppressed tumor growth and improved outcomes in mice with NSCLC by modulating immune cell distribution and reducing levels of target immune proteins.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Récepteur au facteur de nécrose tumorale de type II , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Souris , Récepteur au facteur de nécrose tumorale de type II/antagonistes et inhibiteurs , Récepteur au facteur de nécrose tumorale de type II/immunologie , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lignée cellulaire tumorale , Évolution de la maladie , Femelle , Immunothérapie/méthodes , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie
11.
Sci Adv ; 10(38): eado9746, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39303028

RÉSUMÉ

While immune checkpoint inhibitors have revolutionized cancer therapy, many patients exhibit poor outcomes. Here, we show immunotherapy responses in bladder and non-small cell lung cancers are effectively predicted by factoring tumor mutation burden (TMB) into burdens on specific protein assemblies. This approach identifies 13 protein assemblies for which the assembly-level mutation burden (AMB) predicts treatment outcomes, which can be combined to powerfully separate responders from nonresponders in multiple cohorts (e.g., 76% versus 37% bladder cancer 1-year survival). These results are corroborated by (i) engineered disruptions in the predictive assemblies, which modulate immunotherapy response in mice, and (ii) histochemistry showing that predicted responders have elevated inflammation. The 13 assemblies have diverse roles in DNA damage checkpoints, oxidative stress, or Janus kinase/signal transducers and activators of transcription signaling and include unexpected genes (e.g., PIK3CG and FOXP1) for which mutation affects treatment response. This study provides a roadmap for using tumor cell biology to factor mutational effects on immune response.


Sujet(s)
Immunothérapie , Mutation , Humains , Immunothérapie/méthodes , Animaux , Souris , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Résultat thérapeutique , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/thérapie , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Protéines tumorales/génétique , Protéines tumorales/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie
12.
Bull Exp Biol Med ; 177(4): 493-496, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39264561

RÉSUMÉ

The expression of long non-coding RNA (lncRNA) LINC01508 was studied in tumor samples from patients with non-small cell lung cancer (NSCLC), and its clinical significance was evaluated. The expression of LINC01508 lncRNA was measured in 16 pairs of NSCLC samples and its association with clinical and morphological features of the disease and prognosis analyzed. A comparative analysis showed a significant decrease in the expression of LINC01508 in tumor lung tissue in comparison with the surrounding normal lung tissue. The informativity of the diagnostic method was tested using ROC curve analysis and calculation of the area under the curve (AUC) for LINC01508 in NSCLC patients, which showed an AUC of 0.875 (p=0.001). No significant associations of LINC01508 expression level with clinical and morphological characteristics of the disease were found. The analysis of prognostic significance showed that high expression of LINC01508 in NSCLC samples was a favorable prognostic factor. Despite the fact that the results did not reach statistical significance (p=0.107), the median survival rate for patients with low LINC01508 expression was 16 months, while for patients with high expression, it was not reached during the follow-up period. We believe that LINC01508 can be a promising independent prognostic marker for NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/mortalité , Mâle , Femelle , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/métabolisme , Adulte d'âge moyen , Pronostic , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Courbe ROC , Adulte , Pertinence clinique
13.
JAMA Netw Open ; 7(9): e2434180, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39302678

RÉSUMÉ

Importance: Emerging evidence suggests that wearable devices are feasible for monitoring physical activity among patients with lung cancer. However, the association between wearable devices and improvement in patient recovery after surgery remains underexplored. Objective: To evaluate the effects of a wearable device intervention on the recovery of physical activity, cardiopulmonary function, and health-related quality of life (HRQOL) after lung cancer surgery. Design, Setting, and Participants: This nonrandomized clinical trial with a historical control was conducted at a single tertiary cancer center (Samsung Comprehensive Cancer Center) in Seoul, South Korea, between October 18, 2018, and May 24, 2019. Patients were included if they had suspected or confirmed non-small cell lung cancer scheduled for curative surgery more extensive than lobectomy and had an Eastern Cooperative Oncology Group status of 0 or 1. Patients were compared with historical control participants from data collected between September 20, 2017, and September 10, 2018, as part of the Coordinated Approach to Cancer Patients' Health for Lung Cancer (CATCH-LUNG) prospective cohort study. Data analysis was performed between June 21 and July 16, 2020. Intervention: A personalized exercise regimen monitored via a wearable device was administered to intervention patients at home in 3 stages: preoperative (from diagnosis to surgery), immediate (from discharge to 2 months after surgery), and later postoperative (from 2 to 6 months after surgery). Control patients received usual care. Main Outcomes and Measures: The primary outcome was cardiopulmonary function, and the co-primary outcome was physical activity at 6 months after surgery, measured with 6-minute walking distance (6MWD) and number of daily steps, using a linear regression model. Secondary outcomes were changes in cardiopulmonary function, physical activity, and HRQOL, including function and symptoms from baseline to 2 weeks and 6 months after surgery. Additionally, cardiopulmonary function and physical activity (number of daily steps and time spent on moderate-to-vigorous physical activity [MVPA]) at 2 weeks after surgery, physical activity (time spent on MVPA) at 6 months after surgery, and HRQOL, including function and symptoms at 2 weeks and 6 months after surgery, were assessed as secondary outcomes. Results: This trial included 74 patients in the intervention group (mean [SD] age, 60.4 [8.7] years; 31 [41.9%] men and 43 [58.1%] women) and 120 in the control group (mean [SD] age, 60.2 [8.7] years; 65 [54.2%] men and 55 [45.8%] women). Daily steps, MVPA, and 6MWD decreased initially at 2 weeks after surgery but increased thereafter. The control group had a larger decrease in the number of daily steps from baseline compared with the intervention group (-4877 [95% CI, -5861 to -3893] steps vs -1753 [95% CI, -2968 to -539] steps) at 2 weeks after surgery. By 6 months after surgery, the intervention group increased their daily steps by 2220 (95% CI, 1006 to 3435) from baseline, whereas the control group did not return to their baseline number of steps. The intervention group had significantly more daily steps (12 321 [95% CI, 8749-15 761] vs 10 118 [95% CI, 7341-13 420]; P = .007) and had greater vigorous physical activity (33.6 [95% CI, 13.5 to 59.8] vs 18.5 [5.7 to 40.8] minutes; P = .003) at 6 months after surgery compared with the control group. No difference in 6MWD was found. However, the intervention group had better patient-reported physical function (mean [SD] score, 82.2 [17.3] vs 76.9 [17.5]; P = .04), less dyspnea (mean [SD] score, 24.8 [27.1] vs 34.5 [31.6]; P = .03), and less pain (mean [SD] score, 21.4 [20.2] vs 30.1 [26.8]; P = .01) at 2 weeks after surgery and less dyspnea (mean [SD] score, 5.4[12.4] vs 12[23.3]; P = .01) at 6 months after surgery compared with the control group. Conclusions and Relevance: In this nonrandomized clinical trial, integration of perioperative exercise interventions using wearable devices improved physical activity (especially MVPA) and dyspnea at 6 months after lung cancer surgery compared with usual care. This finding suggests a promising role for wearable devices in personalizing perioperative rehabilitation strategies. Trial Registration: ClinicalTrials.gov Identifier: NCT03215537.


Sujet(s)
Exercice physique , Tumeurs du poumon , Qualité de vie , Dispositifs électroniques portables , Humains , Mâle , Femelle , Tumeurs du poumon/chirurgie , Adulte d'âge moyen , Sujet âgé , Études prospectives , République de Corée , Carcinome pulmonaire non à petites cellules/chirurgie
14.
J Cell Mol Med ; 28(18): e70077, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39304978

RÉSUMÉ

In addressing the challenges of lung cancer, attention has increasingly turned to molecular diagnostics and targeted therapies, with nucleolin (NCL) assuming a pivotal role, especially in non-small cell lung cancer. The aberrant activity and cellular distribution of NCL act as crucial biomarkers for early detection and treatment monitoring, showing a strong correlation with disease progression and patient prognosis. Elevated NCL levels signal advanced disease and poorer outcomes, underscoring its significance in evaluating disease severity and therapeutic response. Strategies targeting the molecular interactions of NCL have spurred innovative approaches to inhibit tumour growth, overcome drug resistance and improve treatment efficacy. These advancements are paving the way for personalized therapies tailored to the unique molecular profiles of patients' tumours. Consequently, NCL stands at the forefront of lung cancer management, symbolizing the move towards more precise and individualized oncology care, and marking substantial progress in therapeutic development.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du poumon , , Phosphoprotéines , Protéines de liaison à l'ARN , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/diagnostic , Tumeurs du poumon/génétique , Tumeurs du poumon/thérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Phosphoprotéines/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines 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/diagnostic , Carcinome pulmonaire non à petites cellules/thérapie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Pronostic , Animaux , Thérapie moléculaire ciblée
15.
Sci Rep ; 14(1): 21625, 2024 09 16.
Article de Anglais | MEDLINE | ID: mdl-39285232

RÉSUMÉ

Lung cancer remains the leading cause of cancer death. Treatment with immune checkpoint inhibitor (ICI) alone or combination with chemotherapy served as first-line therapy in non-small cell lung cancer (NSCLC). However, only 20-50% of NSCLC patients respond to ICI. Necroptosis, an inflammatory form of cell death plays an important role in the regulation of tumor immune microenvironment which may affect prognosis and ICI response but its clinical significance in NSCLC patients has remained largely unknown. Therefore, we aimed to analyze the correlation between key necroptotic proteins and necroptosis and clinical outcomes, the status of tumor-infiltrating immune cells, and response to ICI in NSCLC patients. The expression of receptor-interacting protein kinase 3 (RIPK3), mixed lineage kinase domain-like protein (MLKL) and phosphorylated MLKL (pMLKL) were immunolocalized in 125 surgically resected NSCLC patients and 23 NSCLC patients administered with ICI therapy. CD8 + and FOXp3 + T cells and CD163 + M2 macrophages were also immunolocalized. High RIPK3 status was positively correlated with survival of the patients and RIPK3 turned out an independent favorable prognostic factor of the patients. RIPK3 was negatively correlated with CD8 + T cells, while MLKL positively correlated with CD163 + M2 macrophages, suggesting the possible involvement of RIPK3 and MLKL in formulating immunosuppressive microenvironment. In addition, high RIPK3 status tended to be associated with clinical resistance to ICI therapy (P-value = 0.057). Furthermore, NSCLC cells-expressing RIPK3 suppressed T cells response to ICI therapy in vitro. Therefore, RIPK3 and MLKL could induce an immunosuppressive microenvironment, resulting in low response to ICI therapy in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Nécroptose , Protein kinases , Receptor-Interacting Protein Serine-Threonine Kinases , Microenvironnement tumoral , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/immunologie , Nécroptose/effets des médicaments et des substances chimiques , Pronostic , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme
16.
J Exp Med ; 221(11)2024 Nov 04.
Article de Anglais | MEDLINE | ID: mdl-39297884

RÉSUMÉ

The inevitable acquired resistance to osimertinib (AZD9291), an FDA-approved third-generation EGFR tyrosine kinase inhibitor (EGFR-TKI) for the treatment of patients with advanced non-small cell lung cancer (NSCLC) harboring EGFR activating or T790M resistant mutations, limits its long-term clinical benefit. Telomere maintenance via telomerase reactivation is linked to uncontrolled cell growth and is a cancer hallmark and an attractive cancer therapeutic target. Our effort toward understanding the action mechanisms, including resistance mechanisms, of osimertinib has led to the identification of a novel and critical role in maintaining c-Myc-dependent downregulation of hTERT, a catalytic subunit of telomerase, and subsequent inhibition of telomerase/telomere and induction of telomere dysfunction in mediating therapeutic efficacy of osimertinib. Consequently, osimertinib combined with the telomere inhibitor, 6-Thio-dG, which is currently tested in a phase II trial, effectively inhibited the growth of osimertinib-resistant tumors, regressed EGFRm NSCLC patient-derived xenografts, and delayed the emergence of acquired resistance to osimertinib, warranting clinical validation of this strategy to manage osimertinib acquired resistance.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Mutation , Telomerase , Télomère , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Telomerase/génétique , Telomerase/métabolisme , Telomerase/antagonistes et inhibiteurs , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Humains , Récepteurs ErbB/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Animaux , Télomère/métabolisme , Télomère/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Indoles , Pyrimidines
17.
J Immunother Cancer ; 12(9)2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39299754

RÉSUMÉ

BACKGROUND: Major histocompatibility complex class I (MHC-I) loss is frequent in non-small cell lung cancer (NSCLC) rendering tumor cells resistant to T cell lysis. NK cells kill MHC-I-deficient tumor cells, and although previous work indicated their presence at NSCLC margins, they were functionally impaired. Within, we evaluated whether NK cell and CD8 T cell infiltration and activation vary with MHC-I expression. METHODS: We used single-stain immunohistochemistry (IHC) and Kaplan-Meier analysis to test the effect of NK cell and CD8 T cell infiltration on overall and disease-free survival. To delineate immune covariates of MHC-I-disparate lung cancers, we used multiplexed immunofluorescence (mIF) imaging followed by multivariate statistical modeling. To identify differences in infiltration and intercellular communication between IFNγ-activated and non-activated lymphocytes, we developed a computational pipeline to enumerate single-cell neighborhoods from mIF images followed by multivariate discriminant analysis. RESULTS: Spatial quantitation of tumor cell MHC-I expression revealed intratumoral and intertumoral heterogeneity, which was associated with the local lymphocyte landscape. IHC analysis revealed that high CD56+ cell numbers in patient tumors were positively associated with disease-free survival (HR=0.58, p=0.064) and overall survival (OS) (HR=0.496, p=0.041). The OS association strengthened with high counts of both CD56+ and CD8+ cells (HR=0.199, p<1×10-3). mIF imaging and multivariate discriminant analysis revealed enrichment of both CD3+CD8+ T cells and CD3-CD56+ NK cells in MHC-I-bearing tumors (p<0.05). To infer associations of functional cell states and local cell-cell communication, we analyzed spatial single-cell neighborhood profiles to delineate the cellular environments of IFNγ+/- NK cells and T cells. We discovered that both IFNγ+ NK and CD8 T cells were more frequently associated with other IFNγ+ lymphocytes in comparison to IFNγ- NK cells and CD8 T cells (p<1×10-30). Moreover, IFNγ+ lymphocytes were most often found clustered near MHC-I+ tumor cells. CONCLUSIONS: Tumor-infiltrating NK cells and CD8 T cells jointly affected control of NSCLC tumor progression. Coassociation of NK and CD8 T cells was most evident in MHC-I-bearing tumors, especially in the presence of IFNγ. Frequent colocalization of IFNγ+ NK cells with other IFNγ+ lymphocytes in near-neighbor analysis suggests NSCLC lymphocyte activation is coordinately regulated.


Sujet(s)
Lymphocytes T CD8+ , Carcinome pulmonaire non à petites cellules , Antigènes d'histocompatibilité de classe I , Cellules tueuses naturelles , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/immunologie , Mâle , Femelle , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme
18.
Front Immunol ; 15: 1376704, 2024.
Article de Anglais | MEDLINE | ID: mdl-39308861

RÉSUMÉ

Despite treatment advances through immunotherapies, including anti-PD-1/PD-L1 therapies, the overall prognosis of non-small cell lung cancer (NSCLC) patients remains poor, underscoring the need for novel approaches that offer long-term clinical benefit. This review examined the literature on the subject over the past 20 years to provide an update on the evolving landscape of dendritic cell-based immunotherapy to treat NSCLC, highlighting the crucial role of dendritic cells (DCs) in immune response initiation and regulation. These cells encompass heterogeneous subsets like cDC1s, cDC2s, and pDCs, capable of shaping antigen presentation and influencing T cell activation through the balance between the Th1, Th2, and Th17 profiles and the activation of regulatory T lymphocytes (Treg). The intricate interaction between DC subsets and the high density of intratumoral mature DCs shapes tumor-specific immune responses and impacts therapeutic outcomes. DC-based immunotherapy shows promise in overcoming immune resistance in NSCLC treatment. This article review provides an update on key clinical trial results, forming the basis for future studies to characterize the role of different types of DCs in situ and in combination with different therapies, including DC vaccines.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Cellules dendritiques , Tumeurs du poumon , Humains , Cellules dendritiques/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Carcinome pulmonaire non à petites cellules/immunologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/immunologie , Animaux , Immunothérapie/méthodes , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique
19.
Front Immunol ; 15: 1420463, 2024.
Article de Anglais | MEDLINE | ID: mdl-39308869

RÉSUMÉ

With the outbreak of the coronavirus disease 2019 (COVID-19), reductions in T-cell function and exhaustion have been observed in patients post-infection of COVID-19. T cells are key mediators of anti-infection and antitumor, and their exhaustion increases the risk of compromised immune function and elevated susceptibility to cancer. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer with high incidence and mortality. Although the survival rate after standard treatment such as surgical treatment and chemotherapy has improved, the therapeutic effect is still limited due to drug resistance, side effects, and recurrence. Recent advances in molecular biology and immunology enable the development of highly targeted therapy and immunotherapy for cancer, which has driven cancer therapies into individualized treatments and gradually entered clinicians' views for treating NSCLC. Currently, with the development of photosensitizer materials, phototherapy has been gradually applied to the treatment of NSCLC. This review provides an overview of recent advancements and limitations in different treatment strategies for NSCLC under the background of COVID-19. We discuss the latest advances in phototherapy as a promising treatment method for NSCLC. After critically examining the successes, challenges, and prospects associated with these treatment modalities, their profound prospects were portrayed.


Sujet(s)
COVID-19 , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , SARS-CoV-2 , Humains , Carcinome pulmonaire non à petites cellules/thérapie , COVID-19/thérapie , COVID-19/immunologie , Tumeurs du poumon/thérapie , SARS-CoV-2/physiologie , Photothérapie/méthodes , Association thérapeutique , Immunothérapie/méthodes
20.
Int J Biol Sci ; 20(12): 4750-4766, 2024.
Article de Anglais | MEDLINE | ID: mdl-39309428

RÉSUMÉ

Patients with non-small cell lung cancer (NSCLC) are easily resistant to first-line chemotherapy with paclitaxel (PTX) or carboplatin (CBP). N6-methyladenosine (m6A) methyltransferase-like 3 (METTL3) has crucial functions in m6A modification and tumorigenesis. However, its role in chemoresistance of NSCLC is still elusive. Here, we demonstrated that METTL3 inhibitor STM2457 significantly reduced the IC50 values of PTX or CBP in NSCLC cells, and they showed a synergistic effect. Comparing with monotherapy, a combination of STM2457 and PTX or CBP exhibited more potent in vitro and in vivo anti-tumor efficacy. In addition, we found that ATP binding cassette subfamily C member 2 (ABCC2) was responsively elevated in cytomembrane after PTX or CBP treatment, and targeting METTL3 could reverse this effect. Mechanistically, targeting METTL3 decreased the m6A modification of ABCC2 mRNA and accelerated its mRNA degradation. Further studies revealed that YTHDF1 could bind and stabilize the m6A-modified mRNA of ABCC2, while YTHDF1 knockdown promoted it mRNA degradation. These results, taken together, demonstrate that targeting METTL3 enhances the sensitivity of NSCLC cells to PTX or CBP by decreasing the cytomembrane-localized ABCC2 in an m6A-YTHDF1-dependent manner, and suggest that METTL3 may be a potential therapeutic target for acquired resistance to PTX or CBP in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Methyltransferases , Protéine-2 associée à la multirésistance aux médicaments , Protéines associées à la multirésistance aux médicaments , Protéines de liaison à l'ARN , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Humains , Protéines associées à la multirésistance aux médicaments/métabolisme , Protéines associées à la multirésistance aux médicaments/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéine-2 associée à la multirésistance aux médicaments/métabolisme , Lignée cellulaire tumorale , Animaux , Résistance aux médicaments antinéoplasiques/génétique , Souris , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Souris nude , Carboplatine/pharmacologie , Carboplatine/usage thérapeutique , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE