1
|
Zhang Y, Chen Y, Guo C, Li S, Huang C. Systemic immune-inflammation index as a predictor of survival in non-small cell lung cancer patients undergoing immune checkpoint inhibition: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 210:104669. [PMID: 39978427 DOI: 10.1016/j.critrevonc.2025.104669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the association between pretreatment systemic immune-inflammation index (SII) levels and progression-free survival (PFS) and overall survival (OS) in NSCLC patients receiving immune checkpoint inhibitors (ICIs). METHODS A systematic search was conducted across PubMed, Embase, and Web of Science. Hazard ratios (HRs) with 95 % confidence intervals (CIs) for PFS and OS were extracted or calculated. Random-effects models were employed to pool the results and subgroup analyses were performed based on study characteristics, treatment regimens, and analytical methods. RESULTS Two prospective and 11 retrospective studies involving 2342 NSCLC patients treated with ICIs were included. A high pretreatment SII was significantly associated with poor PFS (HR: 2.05, 95 % CI: 1.59-2.64, p < 0.001; I2 = 42 %) and poor OS (HR: 1.54, 95 % CI: 1.29-1.82, p < 0.001; I2 = 22 %). Subgroup analyses according to the country of the study, lines of treatment, cancer stage, methods for determining the cutoffs of SII, and the analytic models showed consistent results (p for subgroup difference all > 0.05). Interestingly, the subgroup analyses indicated a stronger association in patients receiving ICIs alone versus those receiving concurrent chemotherapy (p for subgroup difference = 0.04). CONCLUSIONS High pretreatment SII is associated with worse PFS and OS in NSCLC patients treated with ICIs, particularly for the patients receiving ICIs alone without concurrent chemotherapy.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yeye Chen
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
2
|
Deng Y, Zhang X, Hu F, Lan X. Quantitative 18F-FDG PET/CT Model for predicting pathological complete response to neoadjuvant immunochemotherapy in NSCLC: comparison with RECIST 1.1 and PERCIST. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07342-8. [PMID: 40418330 DOI: 10.1007/s00259-025-07342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE This study aimed to evaluate the predictive value of 18F-FDG PET/CT for pathological complete response (pCR) after neoadjuvant immunochemotherapy in resectable non-small cell lung cancer (NSCLC) and develop a quantitative pCR prediction model. We compared the model's performance with RECIST 1.1 and PERCIST. METHOD A retrospective review was conducted on patients with resectable NSCLC who received neoadjuvant immunochemotherapy from January 2020 to December 2023. Patients with both pre-treatment (18F-FDG PET/CT scan-1) and preoperative scans (18F-FDG PET/CT scan-2) were included. 18F-FDG PET/CT parameters, clinical characteristics, and follow-up data were collected. Logistic regression was used to identify independent predictors and construct the prediction model. The model's predictive performance was compared with RECIST 1.1 and PERCIST criteria. The model was validated with an external cohort from January to September 2024. Postoperative pathological results serve as the gold standard for pCR. RESULTS 36 patients were included for model development, with 19 (52.8%) achieving pCR. ΔTLR% (percentage change between two scans in tumor-to-liver ratio) and SULpeak from scan-2 were significant predictors. The developed prediction model demonstrated outstanding performance with an area under the curve (AUC) of 0.975, 100% sensitivity, and 94.1% specificity. In comparison, RECIST 1.1 showed poor sensitivity (10.5%) but high specificity (100%), while PERCIST had moderate sensitivity (73.7%) and specificity (94.1%). Validation with 8 patients confirmed the model's accuracy. CONCLUSIONS This study suggests that 18F-FDG PET/CT, specifically the ΔTLR% and SULpeak from scan-2, is a reliable predictor of pCR in resectable NSCLC undergoing neoadjuvant immunochemotherapy. The quantitative prediction model outperforms both RECIST 1.1 and PERCIST. These findings highlight the potential clinical utility of this model, although further validation with larger cohorts is required to confirm its robustness and generalizability.
Collapse
Affiliation(s)
- Yueling Deng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
3
|
Zheng Y, Zhang Y, Li J, Su Y. ZC3H12D gene expression exhibits dual effects on the development and progression of lung adenocarcinoma. Sci Rep 2025; 15:17234. [PMID: 40383849 PMCID: PMC12086231 DOI: 10.1038/s41598-025-02163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
While precision oncology requires robust biomarkers, current predictors for lung adenocarcinoma (LUAD) often show limited clinical utility. This study investigates the multifaceted roles of ZC3H12D, a novel immunomodulatory molecule, in LUAD progression and tumor microenvironment regulation. Multi-omics analyses integrated ZC3H12D transcriptomic (511 tumors vs 59 normals), proteomic (74 tumors vs 69 normals), and single-cell RNA-seq data (15 tumors vs 11 normals). Immunohistochemistry validated ZC3H12D expression in 51 matched pairs. Computational biology approaches assessed immune infiltration, genomic instability (TMB/MSI/HRD), and pathway enrichment. Functional validation employed ZC3H12D knockdown in PC9 cells with colony formation and transwell assays. Multi-omics verification confirmed ZC3H12D upregulation in LUAD at both mRNA and protein levels (p < 0.001), with single-cell resolution revealing predominant localization in tumor-infiltrating immune cells. Moreover, ZC3H12D expression positively correlated with immune regulatory genes while inversely associating with genes involved in cellular respiration. Its expression was also linked to clinical markers such as TMB, MSI, HRD, tumor purity, and ploidy. Notably, high ZC3H12D expression revealed Immune-infiltrated microenvironment and favorable prognosis, despite silencing ZC3H12D resulted in significant inhibition of tumor cell proliferation and invasion in vitro (p < 0.001). Our findings demonstrate that high ZC3H12D expression in immune cells appears to enhance antitumor immune activity, whereas lower expression in malignant cells contributes to reduced cellular proliferation and migration. This spatial duality challenges conventional biomarker paradigms and provides mechanistic insights for developing cell type-targeted therapies.
Collapse
Affiliation(s)
- Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, NO.1 East Banshan Road, Gongshu, Hangzhou, 310022, China
| | - Yuhuan Zhang
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd, No.153 Huixin Road, Nanhu, Jiaxing, 314033, China
- Department of R&D, Shanghai Yunying Biopharmaceutical Technology Co., Ltd, No.518 Xinzhuan Road, Songjiang, Shanghai, 201612, China
| | - Jieyi Li
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd, No.153 Huixin Road, Nanhu, Jiaxing, 314033, China.
- Department of R&D, Shanghai Yunying Biopharmaceutical Technology Co., Ltd, No.518 Xinzhuan Road, Songjiang, Shanghai, 201612, China.
| | - Ying Su
- Department of Pathology, Zhejiang Cancer Hospital, NO.1 East Banshan Road, Gongshu, Hangzhou, 310022, China.
| |
Collapse
|
4
|
Chen T, Liu F. Neoadjuvant immunotherapy in early-stage NSCLC: navigating biomarker dilemmas and special population challenges. Lung Cancer 2025; 204:108588. [PMID: 40409027 DOI: 10.1016/j.lungcan.2025.108588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
Neoadjuvant immunotherapy has shown impressive outcomes in treating non-small cell lung cancer (NSCLC) recently due to advancements in immune checkpoint inhibitors (ICIs) research. Neoadjuvant immunotherapy can lower the tumor load, raise the complete surgical (R0) resection rate, and improve clinical outcomes by alleviating the immune system repression caused by tumor cells. This review provides a comprehensive evaluation of neoadjuvant immunotherapy in NSCLC, focusing on: (1) its safety and efficacy profiles, (2) the most recent clinical trial evidence, and (3) critical unresolved challenges including predictive biomarker development, management of driver mutation-positive patients, chronic obstructive pulmonary disease (COPD) comorbidity considerations, and its application in stage III-IVA (oligometastatic) disease. Furthermore, we explore future research directions to optimize neoadjuvant immunotherapy approaches for resectable NSCLC, aiming to guide clinical practice and investigation.
Collapse
Affiliation(s)
- Tong Chen
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
5
|
Dennehy C, Conroy MR, Forde PM. Immunotherapy for resectable lung cancer. Cancer 2025; 131:e35849. [PMID: 40334018 PMCID: PMC12057804 DOI: 10.1002/cncr.35849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 05/09/2025]
Abstract
Lung cancer remains a significant global health challenge, demanding innovative treatment strategies. Immune checkpoint blockade has revolutionized cancer care, leading to improved survival across advanced malignancies and has now become a standard therapy for earlier stage, resectable lung cancer. This review article consolidates the current landscape and future prospects of neoadjuvant and perioperative immunotherapy in lung cancer. The authors outline key findings from clinical trials in resectable lung cancer, including early efficacy, safety profiles, and emerging impact on disease recurrence, and overall survival. Additionally, this review elucidates the challenges encountered, including patient selection criteria, optimal treatment schedules, immune-related adverse events, and impact on surgery. This comprehensive analysis amalgamates current evidence with future directions, providing a roadmap for clinicians, researchers, and stakeholders to navigate the dynamic realm of immunotherapy for surgically resectable lung cancer.
Collapse
Affiliation(s)
- Colum Dennehy
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Michael R. Conroy
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Patrick M. Forde
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
6
|
Zhao F, Bai H, Liu Y, Gao S, Yang C, Wu J, Cheng H, Ma J, Li Y, Ren H, Fu J, Gu S, Zhao X, Qin S. FLT3LG modulates the infiltration of immune cells and enhances the efficacy of anti-PD-1 therapy in lung adenocarcinoma. BMC Cancer 2025; 25:831. [PMID: 40329265 PMCID: PMC12057023 DOI: 10.1186/s12885-025-14220-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Immunotherapy, particularly anti-PD-1 therapy, has assumed a progressively significant position in the management of non-small cell lung cancer (NSCLC), especially in lung adenocarcinoma (LUAD). Nevertheless, a subset of patients exhibit resistance to anti-PD-1 therapy, and the exploration of biomarkers for evaluating the responsiveness to anti-PD-1 therapy necessitates further investigation. FLT3LG is regarded as being associated with tumor diagnosis and immunotherapy in a variety of tumor types, but its function in LUAD is uncertain. METHODS Bioinformatics analysis was conducted to evaluate the clinical value, functional enrichment, genetic correlation, and immune infiltration of FLT3LG in LUAD. We then used a mouse model to detect immune cell infiltration and relevant protein expression by flow cytometry and immunohistochemistry under anti-PD-1 treatment after overexpression of FLT3LG. The serum FLT3LG expression in LUAD patients was detected via ELISA, and PD-L1 expression in tumor samples was detected by immunohistochemistry. RESULTS In LUAD patients, a better prognosis is associated with elevated FLT3LG expression. Among the genes strongly associated with FLT3LG, the majority were involved in immune-related processes and were enriched predominantly in immune-related pathways. Moreover, high expression of FLT3LG was significantly positively correlated with increased infiltration of multiple immune cells, including T cells and natural killer (NK) cells, in lung adenocarcinomas, as well as the expression of several immune cell markers, such as CD4 and CD8a. In a mouse model, overexpression of FLT3LG in mice subjected to subcutaneous graft tumor elicited a pronounced immune response and could enhance the efficacy of anti-PD-1 therapy. CONCLUSION FLT3LG could be considered as a diagnostic and prognostic marker for LUAD and might play a role in enhancing the therapeutic response to immunotherapy in patients with LUAD.
Collapse
Affiliation(s)
- Fengyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Xi'an, China
| | - Yiwei Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China
| | - Shuoze Gao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China
| | - Chengcheng Yang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| | - Jie Wu
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, China
| | - Hao Cheng
- Department of Rehabilitation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Jiao Ma
- Department of Rehabilitation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Yuanyuan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China
| | - Hong Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China
| | - Junke Fu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China
| | - Shanzhi Gu
- Department of Forensic Medicine, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Xinhan Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi Province, 710061, China.
| |
Collapse
|
7
|
Thomas QD, Vendrell JA, Khellaf L, Cavaillon S, Quantin X, Solassol J, Cabello‐Aguilar S. Artificial intelligence-driven microsatellite instability profiling reveals distinctive genetic features in patients with lung cancer. Cancer 2025; 131:e35882. [PMID: 40297960 PMCID: PMC12038786 DOI: 10.1002/cncr.35882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Microsatellite instability (MSI) has emerged as a predictive biomarker for immunotherapy response in various cancers, but its role in non-small cell lung cancer (NSCLC) is not fully understood. METHODS The authors used the bioinformatics tool MIAmS to assess microsatellite status from next-generation sequencing (NGS) data using a tailored microsatellite score. Immunohistochemistry (IHC) assays were also performed to evaluate the correspondence between MSI and deficient mismatch repair (dMMR) status. A retrospective analysis of 1547 lung cancer patients was conducted, focusing on those with an MSI phenotype. Clinical characteristics, co-occurring molecular alterations, tumor mutation burden (TMB), and homologous recombination deficiency (HRD) status were evaluated in this subset. RESULTS Of the 1547 patients analyzed, eight (0.52%) were identified as having MSI through MIAmS, with six (0.39%) of these cases also being dMMR on IHC. All patients with dMMR had an MS score ≥2 and a history of smoking. Most patients showed loss of MLH1 and PMS2 staining on IHC. No correlation was found between MSI status and programmed death-ligand 1 expression, although all MSI patients exhibited high TMB, averaging 21.4 ± 5.6 mutations per megabase. DISCUSSION MSI/dMMR in lung cancer is exceedingly rare, affecting less than 1% of cases. NGS-based analysis combined with bioinformatics tools provides a robust method to identify MSI/dMMR patients, potentially guiding immunotherapy decisions. This comprehensive approach integrates molecular genotyping and MSI detection, offering personalized treatment options for lung cancer patients. NGS-based MSI testing is emerging as the preferred method for detecting microsatellite instability in various tumor types, including rare cancers.
Collapse
Affiliation(s)
- Quentin Dominique Thomas
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Julie Adèle Vendrell
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Lakhdar Khellaf
- Department of PathologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Sarah Cavaillon
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Xavier Quantin
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Jérôme Solassol
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Simon Cabello‐Aguilar
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
- Montpellier BioInformatics for Clinical DiagnosisMolecular Medicine and Genomics PlatformMontpellier University Hospital MontpellierMontpellierFrance
| |
Collapse
|
8
|
Nishimori M, Iwasa H, Miyatake K, Nitta N, Nakaji K, Izumi T, Matsumoto T, Yoshimatsu R, Yamanishi T, Imai R, Kato M, Okada H, Yamagami T. Correlation between PD-L1 expression and FDG-PET/CT visual assessments in non-small cell lung cancer resected specimens. Nucl Med Commun 2025:00006231-990000000-00420. [PMID: 40296446 DOI: 10.1097/mnm.0000000000001984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
PURPOSE This retrospective study aimed to investigate the validity of fluorodeoxyglucose PET (FDG-PET) visual assessments to predict programmed death-ligand 1 (PD-L1) expression levels in patients with non-small cell lung cancer (NSCLC). MATERIALS AND METHODS One hundred and seven NSCLC patients who underwent FDG-PET/computed tomography (CT) scans and PD-L1 expression tests were retrospectively identified. Patients were divided into two groups according to PD-L1 expression: PD-L1 high group (PD-L1 tumor proportion score ≥50%) and PD-L1 low group (<50%). We compared clinicopathological characteristics and PET assessments [maximum standardized uptake value (SUVmax) and Deauville score] between the two groups based on PD-L1 expression. RESULTS High expression of PD-L1 was detected in 25 of 107 cases. In both univariable and multivariable analysis, there were significant differences in PET visual assessments in NSCLC (P < 0.05). Receiver operating characteristics for the PET visual assessments [area under the curve (AUC) = 0.712, 95% confidence interval (CI) 0.628-0.793] and SUVmax (AUC = 0.753, 95% CI 0.647-0.861) showed equivalent accuracy (P = 0.227). Based on histopathology, in adenocarcinoma patients, there were significant differences between PET visual assessments and PD-L1 expression (P < 0.05), while no significant differences were observed in squamous cell carcinoma patients. Based on epidermal growth factor receptor (EGFR) mutation analysis, in patients with EGFR wild type, there were significant differences between PET visual assessments and PD-L1 expression (P = 0.006), while in patients with EGFR mutations, there were no significant differences between PET visual assessments and PD-L1 expression. CONCLUSION Results of PET visual assessments correlated with PD-L1 expression in NSCLC.
Collapse
Affiliation(s)
| | - Hitomi Iwasa
- Department of Diagnostic and Interventional Radiology
| | - Kana Miyatake
- Department of Diagnostic and Interventional Radiology
| | - Noriko Nitta
- Department of Diagnostic and Interventional Radiology
| | - Kosuke Nakaji
- Department of Diagnostic and Interventional Radiology
| | | | | | | | | | - Rikako Imai
- Center for Innovative and Translational Medicine
| | - Mahiru Kato
- Center for Innovative and Translational Medicine
| | - Hironobu Okada
- Department of Thoracic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi 783-8505, Japan
| | | |
Collapse
|
9
|
Babayev P, Ismayilov R, Kuscu O, Akyildiz A, Kurtulan O, Pamuk E, Guler Tezel YG, Gullu IH. Does high PD-L1 expression reduce the risk of relapse after definitive surgery in patients with oral cavity cancer? Acta Otolaryngol 2025:1-6. [PMID: 40266268 DOI: 10.1080/00016489.2025.2489645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Uncovering the molecular mechanisms of cancer pathogenesis is crucial for improving survival outcomes. AIMS This study investigates the impact of programmed death ligand 1 (PD-L1) expression on the risk of progression in patients with oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS The retrospective study included adult patients diagnosed with OSCC between January 2012 and December 2023. PD-L1 expression in tissue removed by definitive surgery was evaluated using immunohistochemistry, and tumor proportion score (TPS) and combined positive score (CPS) were calculated. Survival analyses were performed to assess its prognostic implications. RESULTS Among the 96 patients, TPS and CPS were 20% or above in 52 (54.2%) and 65 (67.7%) cases, respectively. TPS ≥20% was associated with mucosal lip location, earlier disease stage, and decreased risk of progression. The multivariate Cox regression model revealed that stage III/IV disease (HR: 2.176, 95%CI: 1.085-4.376, p = .029) and TPS ≥20% (HR: 0.241, 95%CI: 0.066-0.879, p = .031) were independent risk factors for progression-free survival. CONCLUSIONS The study revealed that increased tumoral PD-L1 expression may reduce the risk of progression in patients with OSCC. SIGNIFICANCE These findings underscore the potential prognostic significance of PD-L1 expression in OSCC following surgical resection.
Collapse
Affiliation(s)
- Parvin Babayev
- Department of Otorhinolaryngology, Hacettepe University Medical School, Ankara, Turkey
| | - Rashad Ismayilov
- Department of Internal Medicine, Hacettepe University Medical School, Ankara, Turkey
| | - Oguz Kuscu
- Department of Otorhinolaryngology, Hacettepe University Medical School, Ankara, Turkey
| | - Arif Akyildiz
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Olcay Kurtulan
- Department of Pathology, Hacettepe University Medical School, Ankara, Turkey
| | - Erim Pamuk
- Department of Otorhinolaryngology, Hacettepe University Medical School, Ankara, Turkey
| | | | - Ibrahim Halil Gullu
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
10
|
Xie Y, Shi Z, Chen T, Li H, Fan M, Xiang X, Liu F. The Lung Cancer Immune Prognostic Score predicts pathologic complete response and survival in NSCLC patients receiving neoadjuvant immunochemotherapy. Front Immunol 2025; 16:1567565. [PMID: 40308604 PMCID: PMC12040963 DOI: 10.3389/fimmu.2025.1567565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Neoadjuvant immunochemotherapy (nICT) has significantly improved event-free survival (EFS) and pathologic complete response (pCR) in patients with resectable non-small cell lung cancer (NSCLC). However, the lack of validated biomarkers limits their ability to predict therapeutic efficacy and survival outcomes. This study aimed to develop a novel inflammatory and nutritional index, the Lung Cancer Immune Prognostic Score (LCIPS), to predict pCR and survival prognosis in patients with NSCLC. Methods This retrospective study included 131 patients with clinical stage IB-IIIB NSCLC who underwent neoadjuvant immunochemotherapy between May 2020 and May 2024. Baseline clinical data and hematological parameters were collected. Lasso regression analysis was employed to identify hematological indices associated with pCR, and the LCIPS was constructed based on these factors. Kaplan-Meier survival analysis and log-rank tests were used to assess survival differences. Logistic regression was performed to identify the predictors of pCR, while Cox regression analysis determined independent prognostic factors for disease-free survival (DFS) and overall survival (OS). The predictive performance of the LCIPS was validated using a nomogram. Results Lasso regression identified three core hematological indices: the albumin-to-globulin ratio (A/G), absolute monocyte count (MONO), and absolute lymphocyte count (LYM). The LCIPS formula was as follows: LCIPS=0.900×A/G+0.761×MONO (109/L) -0.065×LYM (109/L). Receiver operating characteristic (ROC) curve analysis showed that the LCIPS had superior predictive efficacy (area under the curve (AUC) = 0.68) compared to other classical markers. Univariate and multivariate logistic regression analyses identified intraoperative lymph node dissection status and A/G and LCIPS as independent predictors of pCR (p < 0.05). Multivariate Cox regression analysis demonstrated that smoking status and LCIPS were independent prognostic factors for DFS and OS. Nomogram validation indicated robust predictive accuracy for LCIPS. Notably, among immune-related adverse events (irAEs), endocrine- and cardiac-related irAEs significantly affected DFS (p < 0.05). Discussion LCIPS is an independent predictor of pCR in patients with NSCLC receiving neoadjuvant immunochemotherapy and is associated with improved DFS and survival outcomes. This novel index offers valuable guidance for personalized treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
11
|
Cui X, Liu S, Song H, Xu J, Sun Y. Single-cell and spatial transcriptomic analyses revealing tumor microenvironment remodeling after neoadjuvant chemoimmunotherapy in non-small cell lung cancer. Mol Cancer 2025; 24:111. [PMID: 40205583 PMCID: PMC11980172 DOI: 10.1186/s12943-025-02287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the most common pathological type of lung cancer, and the combination of neoadjuvant immunotherapy with chemotherapy has emerged as the first-line treatment for NSCLC. Nevertheless, the efficacy of this therapeutic approach remains variable. The present study aims to examine the impact of chemoimmunotherapy in NSCLC patients, with a view to identifying key molecules, critical cell subpopulations, communication patterns and spatial distributions that potentially correlate with therapeutic sensitivity. A total of 16 lung cancer tissue samples were collected from a cohort of 12 NSCLC patients and subjected to single-cell RNA and spatial transcriptome sequencing. Our data demonstrated that the distribution of CD4 + Treg T cells and mCAFs indicated an immunosuppressive tumor microenvironment, while the accumulation of CD4 + Th17 T cells and iCAFs could act as a positive marker for the sensitivity to chemoimmunotherapy. Furthermore, a significant high level of SELENOP-macrophages was observed in tissues from positive responders, and a strong co-localization between SELENOP-macrophages and antigen-presenting cancer associated fibroblasts (CAFs) in the tumor boundaries was identified, indicating the cooperative roles of these two cell types in response to combined therapy. Moreover, SELENOP-macrophages were observed to be accumulated in tertiary lymphoid structures, which further suggested its critical role in recruiting lymphocytes. Furthermore, analysis of cell-cell communication, based on spatial transcriptomics, suggests that the interactions between SELENOP-macrophages, apCAFs, CD4 + and CD8 + T cells were significantly enhanced in responders. In addition, SELENOP-macrophages recruited CD4 + Naïve, Helper and CD8 + Naïve T cells through pathways such as the cholesterol, interleukin, chemokine and HLA when responding to combined therapy. The present study further unveils the dynamic spatial and transcriptional changes in the tumor microenvironment of non-small cell lung cancer in response to combination therapy.
Collapse
Affiliation(s)
- Xiaolu Cui
- Department of Urology, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Siyuan Liu
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - He Song
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China.
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province , 110004, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China.
| |
Collapse
|
12
|
Peng M, Wang M, Yang X, Wang Y, Xie L, An W, Ge F, Yang C, Wang K. Prediction of PD-L1 expression in NSCLC patients using PET/CT radiomics and prognostic modelling for immunotherapy in PD-L1-positive NSCLC patients. Clin Radiol 2025; 86:106915. [PMID: 40375402 DOI: 10.1016/j.crad.2025.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 05/18/2025]
Abstract
AIM To develop a positron emission tomography/computed tomography (PET/CT)-based radiomics model for predicting programmed cell death ligand 1 (PD-L1) expression in non-small cell lung cancer (NSCLC) patients and estimating progression-free survival (PFS) and overall survival (OS) in PD-L1-positive patients undergoing first-line immunotherapy. MATERIALS AND METHODS We retrospectively analysed 143 NSCLC patients who underwent pretreatment 18F-fluorodeoxyglucose (18F-FDG) PET/CT scans, of whom 86 were PD-L1-positive. Clinical data collected included gender, age, smoking history, Tumor-Node-Metastases (TNM) staging system, pathologic types, laboratory parameters, and PET metabolic parameters. Four machine learning algorithms-Bayes, logistic, random forest, and Supportsupport vector machine (SVM)-were used to build models. The predictive performance was validated using receiver operating characteristic (ROC) curves. Univariate and multivariate Cox analyses identified independent predictors of OS and PFS in PD-L1-positive expression patients undergoing immunotherapy, and a nomogram was created to predict OS. RESULTS A total of 20 models were built for predicting PD-L1 expression. The clinical combined PET/CT radiomics model based on the SVM algorithm performed best (area under curve for training and test sets: 0.914 and 0.877, respectively). The Cox analyses showed that smoking history independently predicted PFS. SUVmean, monocyte percentage and white blood cell count were independent predictors of OS, and the nomogram was created to predict 1-year, 2-year, and 3-year OS based on these three factors. CONCLUSION We developed PET/CT-based machine learning models to help predict PD-L1 expression in NSCLC patients and identified independent predictors of PFS and OS in PD-L1-positive patients receiving immunotherapy, thereby aiding precision treatment.
Collapse
Affiliation(s)
- M Peng
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - M Wang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - X Yang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - Y Wang
- Scientific Research Center Department, Beijing General Electric Company, No.2 Yongchang North Road, Yizhuang Economic and Technological Development Zone, Daxing District, Beijing, 102200, China.
| | - L Xie
- MR Research China, GE Healthcare, Beijing, 100176, China.
| | - W An
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - F Ge
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - C Yang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - K Wang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| |
Collapse
|
13
|
Lu X, Jin P, Tang Q, Zhou M, Xu H, Su C, Wang L, Xu F, Zhao M, Yin Y, Zhang J, Jia Z, Peng X, Zhou J, Wang L, Chen Y, Wang M, Yang M, Chen D, Chen Y. NAD + Metabolism Reprogramming Drives SIRT1-Dependent Deacetylation Inducing PD-L1 Nuclear Localization in Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412109. [PMID: 39988985 PMCID: PMC12005810 DOI: 10.1002/advs.202412109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Cervical cancer (CC) is a major health threat to women, with immunotherapies targeting the programmed death receptor 1/programmed death ligand 1(PD-1/PD-L1) axis showing promise but encountering resistance in a significant patient population. This resistance has driven a critical quest to uncover the underlying mechanisms. This study uncovers a novel metabolic axis involving the nicotinamide adenine dinucleotide (NAD+) salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) and the deacetylase Sirtuin 1 (SIRT1), which regulates PD-L1 expression and nuclear localization in CC. This axis may be a key factor contributing to the resistance observed in immunotherapy. This study reveals that PD-L1 overexpression in cancers is regulated by both transcriptional and post-transcriptional processes. Acetyl-proteomic analysis pinpoints SIRT1 as a central regulator in the deacetylation of histone H3 at lysines 27, which may influence PD-L1 subcellular distribution. This finding reveals the epigenetic control of immune checkpoint proteins by metabolic pathways, offering a new perspective on the regulation of PD-L1. The identification of the NAMPT/SIRT1 metabolic axis as a critical factor suggests that targeting this axis may enhance therapeutic responses.
Collapse
Affiliation(s)
- Xinyi Lu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| | - Pingping Jin
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Qianyun Tang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Zhou
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Hanjie Xu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Chen Su
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Lei Wang
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
- Department of Hepatopancreatobiliary SurgeryJiangnan University Medical CenterJiangsu214002China
| | - Feng Xu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Zhao
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Yongxiang Yin
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Jinqiu Zhang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Zhen Jia
- Department of LaboratoryHaidong Second People's HospitalHaidong810699China
| | - Xinrui Peng
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Jie Zhou
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Lu Wang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Yan Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Wang
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| | - Min Yang
- Molecular Imaging CentreJiangsu Institute of Nuclear MedicineJiangsu214063China
| | - Daozhen Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
- Department of LaboratoryHaidong Second People's HospitalHaidong810699China
| | - Yu Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| |
Collapse
|
14
|
Liu X, Ji Z, Zhang L, Li L, Xu W, Su Q. Prediction of pathological complete response to neoadjuvant chemoimmunotherapy in non-small cell lung cancer using 18F-FDG PET radiomics features of primary tumour and lymph nodes. BMC Cancer 2025; 25:520. [PMID: 40119358 PMCID: PMC11929329 DOI: 10.1186/s12885-025-13905-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Predicting the response to neoadjuvant chemoimmunotherapy in patients with resectable non-small cell lung cancer (NSCLC) facilitates clinical treatment decisions. Our study aimed to establish a machine learning model that accurately predicts the pathological complete response (pCR) using 18F-FDG PET radiomics features. METHODS We retrospectively included 210 patients with NSCLC who completed neoadjuvant chemoimmunotherapy and subsequently underwent surgery with pathological results, categorising them into a training set of 147 patients and a test set of 63 patients. Radiomic features were extracted from the primary tumour and lymph nodes. Using 10-fold cross-validation with the least absolute shrinkage and selection operator method, we identified the most impactful radiomic features. The clinical features were screened using univariate and multivariate analyses. Machine learning models were developed using the random forest method, leading to the establishment of one clinical feature model, one primary tumour radiomics model, and two fusion radiomics models. The performance of these models was evaluated based on the area under the curve (AUC). RESULTS In the training set, the three radiomic models showed comparable AUC values, ranging from 0.901 to 0.925. The clinical model underperformed, with an AUC of 0.677. In the test set, the Fusion_LN1LN2 model achieved the highest AUC (0.823), closely followed by the Fusion_Lnall model with an AUC of 0.729. The primary tumour model achieved a moderate AUC of 0.666, whereas the clinical model had the lowest AUC at 0.631. Additionally, the Fusion_LN1LN2 model demonstrated positive net reclassification improvement and integrated discrimination improvement values compared with the other models, and we employed the SHapley Additive exPlanations methodology to interpret the results of our optimal model. CONCLUSIONS Our fusion radiomics model, based on 18F-FDG-PET, will assist clinicians in predicting pCR before neoadjuvant chemoimmunotherapy for patients with resectable NSCLC.
Collapse
Affiliation(s)
- Xingbiao Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhilin Ji
- Department of Radiology, Tianjin Hospital, Jiefangnan Road, Hexi District, Tianjin, 300211, China
| | - Libo Zhang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Linlin Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Qian Su
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
15
|
Feng Z, Liu H, Huang J, Tan X, Su C, Mo S, Tang J, Wang H, Lin Y, Zhang L. Expression of extracellular HSP90α in pre- and post-treatment samples of driver-gene-negative non-small cell lung cancer. Int Immunopharmacol 2025; 146:113792. [PMID: 39689601 DOI: 10.1016/j.intimp.2024.113792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE This study aimed to explore the significance of the expression of pre- and post-treatment eHSP90α in the treatment response evaluation and prognosis of driver-gene-negative non-small cell lung cancer (NSCLC), as well as the significance of eHSP90α expression in the prognosis of immunotherapy. METHODS We collected pre-treatment eHSP90α in 330 driver-gene-negative NSCLC patients and analyzed its relationship with efficacy evaluation and prognosis. Survival curves were used to determine their respective critical values and the relationship between eHSP90α expression and OS and PFS was analyzed. Then, univariable and multivariable Cox regression analyses and LASSO multivariable logistic regression analyses were used to establish the prognostic and efficacy evaluation models. RESULTS High expression of pre-treatment eHSP90α in driver-gene-negative NSCLC patients is associated with shorter OS. Pre-treatment eHSP90α, immunotherapy, TTF-1, CA125, and age influence OS in NSCLC patients. In the stratified analysis of immunotherapy, pre-treatment eHSP90α in immunotherapy is an independent influencing factor of OS in NSCLC patients, and pre-treatment eHSP90α, age, CA125, and B lymphocytes in non-immunotherapy are independent influencing factors of OS in NSCLC patients. High expression of pre-treatment eHSP90α in NSCLC patients is associated with shorter PFS. Pre-treatment eHSP90α, M1, Ki-67, and immunotherapy are independent influencing factors of PFS in NSCLC patients. In the stratified analysis of immunotherapy, pre-treatment eHSP90α and CEA in immunotherapy are independent influencing factors of PFS in NSCLC patients, and pre-treatment eHSP90α, M1, Ki-67, CA125, Th/Ts, and age in non-immunotherapy are independent factors affecting PFS in NSCLC patients. Pre-treatment eHSP90α was one of the influencing factors in the efficacy evaluation model. CONCLUSIONS The expression of pre-treatment eHSP90α has significant significance for the prognosis and efficacy evaluation of driver-gene-negative NSCLC.
Collapse
Affiliation(s)
- Zhaorong Feng
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haizhou Liu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China; Guangxi Cancer Molecular Medicine Engineering Research Center, Nanning, China; Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning, China
| | - Jinglei Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoyu Tan
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chenglin Su
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shanli Mo
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiaying Tang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haiguang Wang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yanyan Lin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Litu Zhang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China; Guangxi Cancer Molecular Medicine Engineering Research Center, Nanning, China; Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning, China.
| |
Collapse
|
16
|
Xu L, Si H, Zhuang F, Li C, Zhang L, Zhao Y, Chen T, Dong Y, Wang T, Hou L, Hu T, Sun T, She Y, Hu X, Xie D, Wu J, Wu C, Zhao D, Chen C. Predicting therapeutic response to neoadjuvant immunotherapy based on an integration model in resectable stage IIIA (N2) non-small cell lung cancer. J Thorac Cardiovasc Surg 2025; 169:242-253.e4. [PMID: 38763304 DOI: 10.1016/j.jtcvs.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE Accurately predicting response during neoadjuvant chemoimmunotherapy for resectable non-small cell lung cancer remains clinically challenging. In this study, we investigated the effectiveness of blood-based tumor mutational burden (bTMB) and a deep learning (DL) model in predicting major pathologic response (MPR) and survival from a phase 2 trial. METHODS Blood samples were prospectively collected from 45 patients with stage IIIA (N2) non-small cell lung cancer undergoing neoadjuvant chemoimmunotherapy. An integrated model, combining the computed tomography-based DL score, bTMB, and clinical factors, was developed to predict tumor response to neoadjuvant chemoimmunotherapy. RESULTS At baseline, bTMB were detected in 77.8% (35 of 45) of patients. Baseline bTMB ≥11 mutations/megabase was associated with significantly greater MPR rates (77.8% vs 38.5%, P = .042), and longer disease-free survival (P = .043), but not overall survival (P = .131), compared with bTMB <11 mutations/megabase in 35 patients with bTMB available. The developed DL model achieved an area under the curve of 0.703 in all patients. Importantly, the predictive performance of the integrated model improved to an area under the curve of 0.820 when combining the DL score with bTMB and clinical factors. Baseline circulating tumor DNA (ctDNA) status was not associated with pathologic response and survival. Compared with ctDNA residual, ctDNA clearance before surgery was associated with significantly greater MPR rates (88.2% vs 11.1%, P < .001) and improved disease-free survival (P = .010). CONCLUSIONS The integrated model shows promise as a predictor of tumor response to neoadjuvant chemoimmunotherapy. Serial ctDNA dynamics provide a reliable tool for monitoring tumor response.
Collapse
Affiliation(s)
- Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haojie Si
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chongwu Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yue Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yichen Dong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingting Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Hu
- Department of Medicine, Amoy Diagnostics Co, Ltd, Xiamen, China
| | - Tianlin Sun
- Department of Medicine, Amoy Diagnostics Co, Ltd, Xiamen, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
17
|
Hu J, Zhang J, Wan S, Zhang P. Neoadjuvant immunotherapy for non-small cell lung cancer: Opportunities and challenges. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:224-239. [PMID: 39834585 PMCID: PMC11742355 DOI: 10.1016/j.pccm.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Indexed: 01/22/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for resectable non-small cell lung cancer. Numerous trials have explored the use of ICIs, either as monotherapy or in combination with other therapies, in the neoadjuvant setting for stage I-III non-small cell lung cancer. Most trials have demonstrated neoadjuvant immunotherapy to be safe and to have remarkable efficacy, with a high pathological response rate and significantly improved event-free survival. This review summarizes the findings of Phase I-III clinical trials investigating various neoadjuvant regimens, including ICI monotherapy, ICI therapy combined with chemotherapy, ICI plus anti-angiogenic therapy, dual ICI therapy, and ICI therapy in combination with radiotherapy or chemoradiotherapy. We discuss the benefits and outcomes associated with each approach. Despite the results being promising, several unresolved issues remain, including identification of reliable biomarkers, the appropriate duration of therapy, the optimal treatment regimen for tumors with high programmed cell death ligand 1 (PD-L1) expression, the false-negative pathological complete response rate, and the role of digital pathology in assessing the response to treatment. Resistance to immunotherapy, in particular, remains a significant barrier to effective use of ICIs. Given the critical influence of the tumor microenvironment (TME) on the response to treatment, we examine the characteristics of the TME in both responsive and resistant tumors as well as the dynamic changes that occur in the TME in response to neoadjuvant immunotherapy. We also summarize the mechanisms underlying T cell responses following neoadjuvant immunotherapy and provide a perspective on strategies to enhance the understanding of tumor heterogeneity, therapy-driven TME remodeling, and overcoming resistance to therapy. Finally, we propose future directions for advancements in personalized neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Junjie Hu
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Shiyue Wan
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- The 1st School of Medicine, the 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Shihezi University Medical College, Shihezi, Xinjiang 832000, China
| |
Collapse
|
18
|
Mo DC, Huang JF, Lin P, Huang SX, Wang HL, Luo PH, Liang XJ. The role of PD-L1 in patients with non-small cell lung cancer receiving neoadjuvant immune checkpoint inhibitor plus chemotherapy: a meta-analysis. Sci Rep 2024; 14:26200. [PMID: 39482343 PMCID: PMC11527982 DOI: 10.1038/s41598-024-78159-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND The use of immune checkpoint inhibitors (ICIs) as neoadjuvant therapy is a promising novel approach in resectable non-small-cell lung cancer (NSCLC). This study aimed to investigate the prognostic value of PD-L1 in patients with NSCLC receiving neoadjuvant immune checkpoint inhibitor plus chemotherapy (CT). MATERIALS AND METHODS Several databases (PubMed, Embase, and cochrane central register of controlled trials [CENTRAL]) were systematically searched. Randomized controlled trials (RCTs) investigating pathological and survival outcomes with neoadjuvant ICI + CT versus CT alone in NSCLC were analyzed. RESULTS Overall, eight RCTs (n = 3,404) were included. The analyses showed neoadjuvant ICI + CT significantly improved complete pathological response (pCR) and event-free survival (EFS) in either tumor PD-L1 < 1%, ≥ 1%, 1-49%, or ≥ 50% population (both p < 0.0001) compared with neoadjuvant CT alone. The overall survival (OS) data are not yet mature among all included RCTs, and only three RCTs presented OS data by PD-L1 status of patients. The pooled OS favored neoadjuvant ICI + CT in the PD-L1 ≥ 1% population (hazard ratio [HR], 0.45; 95% CI, 0.31-0.65; p < 0.0001), but not in the PD-L1 < 1% population (HR, 0.89; 95% CI, 0.66-1.19; p = 0.43). CONCLUSIONS Compared with neoadjuvant CT alone, neoadjuvant ICI + CT significantly enhanced pCR and EFS for patients with resectable NSCLC regardless of the expression of PD-L1. It seems that only patients with PD-L1 positive tumors may achieve a better OS, but it's currently inconclusive due to immature data, so future research with long-term follow-up is still needed.
Collapse
Affiliation(s)
- Dun-Chang Mo
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China.
| | - Jian-Feng Huang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Peng Lin
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Shang-Xiao Huang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Han-Lei Wang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Peng-Hui Luo
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Xiu-Juan Liang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| |
Collapse
|
19
|
Papaporfyriou A, Bartziokas K, Apessos I, Mueller J, Leivaditis V, Koletsis E, Grapatsas K. Comparative Efficacy and Safety of Neoadjuvant Immunotherapy with Nivolumab vs. Pembrolizumab in Resectable Non-Small Cell Lung Cancer: A Systematic Review. Curr Oncol 2024; 31:6289-6299. [PMID: 39451773 PMCID: PMC11506529 DOI: 10.3390/curroncol31100469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/05/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Immunotherapy has emerged as a promising treatment option due to its favorable toxicity profile. However, selecting the most appropriate immunotherapeutic agent for neoadjuvant use-aimed at curative intent in early-stage NSCLC-based on efficacy and safety remains a critical question. This review aims to compare the efficacy and safety profiles of nivolumab and pembrolizumab when used as neoadjuvant treatments in NSCLC. A systematic review was conducted across PubMed, Scopus, Wiley Online Library, ProQuest Dissertations and Theses Global, and Google Scholar, utilizing the search terms "Nivolumab OR Pembrolizumab AND Neoadjuvant Immunotherapy AND non-small cell lung cancer." Out of 1444 retrieved studies, 4 retrospective studies met the inclusion criteria by providing comparative data on nivolumab and pembrolizumab within the same study cohorts. Despite the critical risk of bias and the evidence quality ranging from moderate to very low across these studies, both nivolumab and pembrolizumab demonstrated efficacy rates exceeding 30% and maintained favorable safety profiles. There is no observed superiority between nivolumab and pembrolizumab in terms of efficacy and safety for the neoadjuvant treatment of early-stage NSCLC.
Collapse
Affiliation(s)
- Anastasia Papaporfyriou
- Department of Pulmonology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Ioulianos Apessos
- Department of Dentoalveolar Surgery, Implantology and Oral Radiology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Jan Mueller
- Department of Pulmonology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Vasileios Leivaditis
- Department of Cardiothoracic and Vascular Surgery, WestpfalzKlinikum, 67655 Kaiserlautern, Germany;
| | - Efstratios Koletsis
- Department of Cardiothoracic Surgery, Patras University Hospital, 26504 Patras, Greece;
| | - Konstantinos Grapatsas
- Department of Thoracic Surgery, West German Cancer Center, University Medical Center Essen-Ruhrlandklinik, University Duisburg-Essen, 45239 Essen, Germany;
| |
Collapse
|
20
|
Hung SK, Lee MS, Chiou WY, Liu DW, Yu CC, Chen LC, Lin RI, Chew CH, Hsu FC, Yang HJ, Chan MWY, Lin HY. Epigenetic modification in radiotherapy and immunotherapy for cancers. Tzu Chi Med J 2024; 36:396-406. [PMID: 39421493 PMCID: PMC11483092 DOI: 10.4103/tcmj.tcmj_3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/20/2024] [Accepted: 06/18/2024] [Indexed: 10/19/2024] Open
Abstract
Radiotherapy (RT) is one of the primary treatment modalities in managing cancer patients. Recently, combined RT and immunotherapy (IT) (i.e., radio-IT [RIT]) have been aggressively investigated in managing cancer patients. However, several issues in conducting RIT are challenging, such as incorporating advanced irradiation techniques, predictive/prognostic biomarkers, and other treatment modalities. Several clinical efforts and novel biomarkers have been introduced and developed to solve these challenges. For example, stereotactic radiosurgery/stereotactic radiotherapy, stereotactic body radiotherapy/stereotactic ablative body radiotherapy, and FLASH-RT have been applied for delivering precise irradiation to lung and liver tumors in conjunction with IT. Besides, several novel IT agents and incorporations of other therapies, such as targeted and thermal therapies, have been further investigated. The present study reviewed the emerging challenges of RIT in modern oncology. We also evaluated clinical practice, bench research, and multimodality treatments. In addition to several clinically applicable biomarkers, we emphasize the roles of advanced irradiation techniques and epigenetic modification as predictive/prognostic biomarkers and potential therapeutic targets. For example, 6(m) A-based epigenetic agents demonstrate the potential to enhance the treatment effects of RIT. However, further prospective randomized trials should be conducted to confirm their roles.
Collapse
Affiliation(s)
- Shih-Kai Hung
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Moon-Sing Lee
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Yen Chiou
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Dai-Wei Liu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chih-Chia Yu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Liang-Cheng Chen
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ru-Inn Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chia-Hui Chew
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Feng-Chun Hsu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Hsuan-Ju Yang
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Hon-Yi Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
21
|
Wang X, Jin H, Feng X, Liang Z, Jin R, Li X. Depiction of the Genetic Alterations and Molecular Landscapes of Thymic Epithelial Tumors: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:2966. [PMID: 39272824 PMCID: PMC11394263 DOI: 10.3390/cancers16172966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Thymic epithelial tumors (TETs), consisting of thymomas, thymic carcinomas (TCs), and thymic neuroendocrine tumors, are rare diseases. Surgery remains the prime option in resectable and early-stage TETs, while chemotherapy, targeted therapy, and immunotherapy are also potential treatment modalities. However, the inadequate comprehension of the molecular landscape of TETs impedes the exploitation of such therapies. Hence, we conducted a meta-analysis which includes 21 studies reporting on genomic alterations in TETs and 14 studies reporting on PD-L1 expression levels, respectively. The pooled estimated rates of the most frequently mutated genes and PD-L1 expression levels were analyzed using the R software. We uncovered that the pooled estimated overall mutation rate is 0.65 ([0.49; 0.81]), and the top three genes with highest mutation frequency in thymomas and TCs are GTF2I (0.4263 [0.3590; 0.4936]), TP53 (0.1101 [0.0000; 0.2586]), and RAS (0.0341 [0.0104; 0.0710]), and TP53 (0.1797 [0.0732; 0.3203]), CDKN2A (0.0608 [0.0139; 0.1378]), and TET2 (0.0318 [0.0087; 0.0639]), respectively. A uniform GTF2I mutational rate in thymomas and TP53 mutational rate in thymic squamous cell carcinomas (TSCCs) are also observed. The pooled estimated expression level of PD-L1 is 0.71 ([0.59-0.81]). This systematic review provides an overview of the gene alteration landscape and PD-L1 expression levels in TETs, discovers several potential confounding factors that may contribute to the high heterogeneity, and facilitates deeper investigations into the elucidation of the molecular landscape of TETs.
Collapse
Affiliation(s)
- Xin Wang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Hongming Jin
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Xiaotong Feng
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Zhijian Liang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Ruoyi Jin
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Xiao Li
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
22
|
Guo W, Qiao T, Li H, Zhao Y, Qin J, Zhang C, Shi C. Peripheral CD8 +PD-1 + T cells as novel biomarker for neoadjuvant chemoimmunotherapy in humanized mice of non-small cell lung cancer. Cancer Lett 2024; 597:217073. [PMID: 38906523 DOI: 10.1016/j.canlet.2024.217073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Neoadjuvant immunotherapy has shown promising clinical activity in the treatment of early non-small cell lung cancer (NSCLC); however, further clarification of the specific mechanism and identification of biomarkers are imperative prior to implementing it as a daily practice. The study investigated the reprogramming of T cells in both tumor and peripheral blood following neoadjuvant chemoimmunotherapy in a preclinical NSCLC mouse model engrafted with a human immune system. Samples were also collected from 21 NSCLC patients (Stage IA-IIIB) who received neoadjuvant chemoimmunotherapy, and the dynamics of potential biomarkers within these samples were measured and further subjected to correlation analysis with prognosis. Further, we initially investigated the sources of the potential biomarkers. We observed in the humanized mouse model, neoadjuvant chemoimmunotherapy could prevent postoperative recurrence and metastasis by increasing the frequency and cytotoxicity of CD8+ T cells in both peripheral blood (p < 0.001) and tumor immune microenvironment (TIME) (p < 0.001). The kinetics of peripheral CD8+PD-1+ T cells reflected the changes in the TIME and pathological responses, ultimately predicting survival outcome of mice. In the clinical cohort, patients exhibiting an increase in these T cells post-treatment had a higher rate of complete or major pathological response (p < 0.05) and increased immune infiltration (p = 0.0012, r = 0.792). We identified these T cells originating from tumor draining lymph nodes and subsequently entering the TIME. In conclusion, the kinetics of peripheral CD8+PD-1+ T cells can serve as a predictor for changes in TIME and optimal timing for surgery, ultimately reflecting the outcomes of neoadjuvant chemoimmunotherapy in both preclinical and clinical setting.
Collapse
Affiliation(s)
- Wenwen Guo
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Clinical Research Center, Xianyang Central Hospital, Xianyang, Shaanxi, 712099, China
| | - Tianyun Qiao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
23
|
Jiang Z, Li Q, Ruan J, Li Y, Zhang D, Xu Y, Liao Y, Zhang X, Gao D, Li Z. Machine Learning-Based Prediction of Pathological Responses and Prognosis After Neoadjuvant Chemotherapy for Non-Small-Cell Lung Cancer: A Retrospective Study. Clin Lung Cancer 2024; 25:468-478.e3. [PMID: 38719649 DOI: 10.1016/j.cllc.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Neoadjuvant chemotherapy has variable efficacy in patients with non-small-cell lung cancer (NSCLC), yet reliable noninvasive predictive markers are lacking. This study aimed to develop a radiomics model predicting pathological complete response and postneoadjuvant chemotherapy survival in NSCLC. MATERIALS AND METHODS Retrospective data collection involved 130 patients with NSCLC who underwent neoadjuvant chemotherapy and surgery. Patients were randomly divided into training and independent testing sets. Nine radiomics features from prechemotherapy computed tomography (CT) images were extracted from intratumoral and peritumoral regions. An auto-encoder model was constructed, and its performance was evaluated. X-tile software classified patients into high and low-risk groups based on their predicted probabilities. survival of patients in different risk groups and the role of postoperative adjuvant chemotherapy were examined. RESULTS The model demonstrated area under the receiver operating characteristic (ROC) curve of 0.874 (training set) and 0.876 (testing set). The larger the area under curve (AUC), the better the model performance. Calibration curve and decision curve analysis indicated excellent model calibration (Hosmer-Lemeshow test, P = .763, the higher the P-value, the better the model fit) and potential clinical applicability. Survival analysis revealed significant differences in overall survival (P = .011) and disease-free survival (P = .017) between different risk groups. Adjuvant chemotherapy significantly improved survival in the low-risk group (P = .041) but not high-risk group (P = 0.56). CONCLUSION This study represents the first successful prediction of pathological complete response achievement after neoadjuvant chemotherapy for NSCLC, as well as the patients' survival, utilizing intratumoral and peritumoral radiomics features.
Collapse
Affiliation(s)
- Zhaojuan Jiang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Qingwan Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Jinqiu Ruan
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Yanli Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Dafu Zhang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Yongzhou Xu
- Department of Clinical & Technique Support, Philips Healthcare, Guangzhou, 510220, China
| | - Yuting Liao
- Department of Clinical & Technique Support, Philips Healthcare, Guangzhou, 510220, China
| | - Xin Zhang
- Department of Clinical & Technique Support, GE Healthcare, Shanghai, 210000, China
| | - Depei Gao
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China.
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China.
| |
Collapse
|
24
|
Wang MJ, Xu SQ, Wu LL, Li ZX, Xie D. Surgical resection due to poor outcome of the immunotherapy of a relapsed mediastinal liposarcoma: a case report. Future Sci OA 2024; 10:FSO906. [PMID: 38827794 PMCID: PMC11140642 DOI: 10.2144/fsoa-2023-0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/18/2023] [Indexed: 06/05/2024] Open
Abstract
The feasibility of surgery after immunotherapy for mediastinal liposarcoma remains uncertain. Besides, the case of immunotherapy for liposarcoma is still lacking. We report a case of recurrence after resection of a left mediastinal liposarcoma. After recurrence, one course of pembrolizumab plus anlotinib hydrochloride showed no tumor shrinkage, and genetic testing showed CDK4 amplification and PD-L1 TPS <1%; therefore, the plan was changed to one course of pembrolizumab plus palbociclib, but the tumor still did not shrink. Thus, second tumor resection was performed. In addition, the postoperative pathology was still well-differentiated liposarcoma. The significance of immunotherapy in liposarcoma still needs to be further explored. In the absence of surgical contraindications, secondary surgery might be feasible.
Collapse
Affiliation(s)
- Ming-Ji Wang
- Department of Thoracic Surgery, Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, PR China
| | - Shu-Quan Xu
- School of Medicine, Tongji University, Shanghai, PR China
| | - Lei-Lei Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Zhi-Xin Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| |
Collapse
|
25
|
Gu C, Teng X, Sun X, Liu J, Zhu Z, Zhang L, Wu Z, Zou R, Pang J, Lyu X. Impact of treatment interval between neoadjuvant immunochemotherapy and surgery in lung squamous cell carcinoma. BMC Cancer 2024; 24:585. [PMID: 38741038 DOI: 10.1186/s12885-024-12333-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVE The optimal timing for surgery following neoadjuvant immunochemotherapy for lung squamous cell carcinoma appears to be a topic of limited data. Many clinical studies lack stringent guidelines regarding this timing. The objective of this study is to explore the effect of the interval between neoadjuvant immunochemotherapy and surgery on survival outcomes in patients with lung squamous cell carcinoma. METHODS This study conducted a retrospective analysis of patients with lung squamous cell carcinoma who underwent neoadjuvant immunochemotherapy between January 2019 and October 2022 at The First Affiliated Hospital, Zhejiang University School of Medicine. Patients were divided into two groups based on the treatment interval: ≤33 days and > 33 days. The primary observational endpoints of the study were Disease-Free Survival (DFS) and Overall Survival (OS). Secondary observational endpoints included Objective response rate (ORR), Major Pathological Response (MPR), and Pathological Complete Remission (pCR). RESULTS Using the Kaplan-Meier methods, the ≤ 33d group demonstrated a superior DFS curve compared to the > 33d group (p = 0.0015). The median DFS for the two groups was 952 days and 590 days, respectively. There was no statistical difference in the OS curves between the groups (p = 0.66), and the median OS was not reached for either group. The treatment interval did not influence the pathologic response of the tumor or lymph nodes. CONCLUSIONS The study observed that shorter treatment intervals were associated with improved DFS, without influencing OS, pathologic response, or surgical safety. Patients should avoid having a prolonged treatment interval between neoadjuvant immunochemotherapy and surgery.
Collapse
Affiliation(s)
- Chen Gu
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
- Zhejiang University School of Medicine, Huangzhou, China
| | - Xiao Teng
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Xuqi Sun
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Jiacong Liu
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Ziyue Zhu
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Lichen Zhang
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Zhigang Wu
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Rui Zou
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China
| | - Jinghua Pang
- Thoracic Surgery, Fenghua People's Hospital, Ningbo, China
| | - Xiayi Lyu
- Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou, 310000, China.
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
26
|
Dawood S, Natarajan V, Danchaivijitr P. Comprehensive molecular profiling identifies actionable biomarkers for patients from Thailand and the United Arab Emirates with advanced malignancies. Front Oncol 2024; 14:1374087. [PMID: 38800398 PMCID: PMC11116666 DOI: 10.3389/fonc.2024.1374087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024] Open
Abstract
Background Comprehensive molecular profiling of tissue samples that can help guide therapy management is not widely available across the globe. Methods Comprehensive molecular profiling through Caris Molecular Intelligence involves the analysis of DNA through next-generation sequencing, chromogenic or fluorescent in situ hybridization, pyrosequencing, and copy number alterations; RNA through whole-transcriptome sequencing and multiplex PCR of RNA; and protein through immunohistochemistry. Results Here we describe the experience of molecular profiling of tumor tissue samples from patients diagnosed with advanced solid tumors and treated in two countries, the United Arab Emirates and Thailand. Tumor cancer cases submitted to Caris Life Sciences (Phoenix, Arizona, USA) for molecular profiling from the UAE and Thailand were retrospectively analyzed (data accessed between 2019 and 2020) for their molecular alterations and clinical biomarkers, without regard to ethnicity. A total of 451 samples from 35 distinct types of advanced cancers were examined for mutations, amplifications, overexpression, exon copy number alterations, microsatellite instability, deficient mismatch repair, tumor mutational burden, and fusions. Interrogating each step of the biological pathway, from DNA to RNA to distinct protein, identified an alteration with an associated therapy for 75% of these tumor samples. The most common alterations identified included elevated PDL-1 that can be targeted with an immune checkpoint inhibitors and amplification of HER2 for which a variety of anti HER2 therapies are available. Conclusion Comprehensive molecular profiling in patients with advanced malignancies can help optimize therapeutic management allowing for improved prognostic outcome.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Oncology Department, Mediclinic City Hospital, Dubai, United Arab Emirates
| | | | - Pongwut Danchaivijitr
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
27
|
Qu J, Wu B, Chen L, Wen Z, Fang L, Zheng J, Shen Q, Heng J, Zhou J, Zhou J. CXCR6-positive circulating mucosal-associated invariant T cells can identify patients with non-small cell lung cancer responding to anti-PD-1 immunotherapy. J Exp Clin Cancer Res 2024; 43:134. [PMID: 38698468 PMCID: PMC11067263 DOI: 10.1186/s13046-024-03046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/13/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Mucosal-associated invariant T (MAIT) cells have been reported to regulate tumor immunity. However, the immune characteristics of MAIT cells in non-small cell lung cancer (NSCLC) and their correlation with the treatment efficacy of immune checkpoint inhibitors (ICIs) remain unclear. PATIENTS AND METHODS In this study, we performed single-cell RNA sequencing (scRNA-seq), flow cytometry, and multiplex immunofluorescence assays to determine the proportion and characteristics of CD8+MAIT cells in patients with metastatic NSCLC who did and did not respond to anti-PD-1 therapy. Survival analyses were employed to determine the effects of MAIT proportion and C-X-C chemokine receptor 6 (CXCR6) expression on the prognosis of patients with advanced NSCLC. RESULTS The proportion of activated and proliferating CD8+MAIT cells were significantly higher in responders-derived peripheral blood mononuclear cells (PBMCs) and lung tissues before anti-PD-1 therapy, with enhanced expression of cytotoxicity-related genes including CCL4, KLRG1, PRF1, NCR3, NKG7, GZMB, and KLRK1. The responders' peripheral and tumor-infiltrating CD8+MAIT cells showed an upregulated CXCR6 expression. Similarly, CXCR6+CD8+MAIT cells from responders showed higher expression of cytotoxicity-related genes, such as CST7, GNLY, KLRG1, NKG7, and PRF1. Patients with ≥15.1% CD8+MAIT cells to CD8+T cells ratio and ≥35.9% CXCR6+CD8+MAIT cells to CD8+MAIT cells ratio in peripheral blood showed better progression-free survival (PFS) after immunotherapy. The role of CD8+MAIT cells in lung cancer immunotherapy was potentially mediated by classical/non-classical monocytes through the CXCL16-CXCR6 axis. CONCLUSION CD8+MAIT cells are a potential predictive biomarker for patients with NSCLC responding to anti-PD-1 therapy. The correlation between CD8+MAIT cells and immunotherapy sensitivity may be ascribed to high CXCR6 expression.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Binggen Wu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Zuoshi Wen
- Department of Cardiology, The First Affiliated Hospital, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
| | - Liangjie Fang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Qian Shen
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jianfu Heng
- Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, P. R. China.
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
28
|
Zhang SL, Tian Y, Yu J, Zhang JH, Sun L, Huang LT, Ma JT, Han CB. Is neoadjuvant immunotherapy necessary in patients with programmed death ligand 1 expression-negative resectable non-small cell lung cancer? A systematic review and meta-analysis. Lung Cancer 2024; 191:107799. [PMID: 38669725 DOI: 10.1016/j.lungcan.2024.107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/30/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVES The aim of this study was to investigate the clinical benefit and necessity of neoadjuvant programmed cell death (or ligand) (PD-(L)1) blockades in resectable non-small cell lung cancer (NSCLC) patients with negative PD-L1 expression. MATERIALS AND METHODS Randomized control trials (RCTs) that compared event-free survival (EFS), overall survival (OS), major pathological response (MPR), and/or pathological complete response (pCR) between neoadjuvant chemo-immunotherapy (nCIT) and neoadjuvant chemotherapy (nCT) for patients with resectable NSCLC stratified by PD-L1 expression were eligible for inclusion in the study. Data regarding the pathological response and EFS were evaluated by the odds ratio (OR) and hazard ratio (HR) with 95% confidence interval (CI) using random and fixed models. RESULTS A total of six RCTs involving 3,194 patients with resectable NSCLC with or without neoadjuvant immunotherapy were included. Compared with nCT alone, nCIT significantly improved pCR (18.3 % vs. 3.0 %; OR, 5.64; 95 % CI, 3.22-9.89; P < 0.001), MPR (38.9 % vs. 15.5 %; OR, 3.57; 95 % CI, 2.10-6.05; P < 0.001), and EFS (HR, 0.75; 95 % CI, 0.62-0.90; P = 0.002) in PD-L1 <1 % NSCLC patients. In addition, PD-L1 ≥1 % was associated with higher rates of pCR (32.8 % vs. 18.3 %; OR, 2.28; 95 % CI, 1.40-3.73; P = 0.001) and MPR (53.9 % vs. 38.9 %; OR, 1.84; 95 % CI, 1.22-2.79; P = 004) and longer EFS (HR, 0.44 vs. 0.75) in the setting of nCIT compared with PD-L1 <1 %. nCIT improved only OS in NSCLC patients with PD-L1 ≥1 % but not in patients with PD-L1 <1 %. CONCLUSIONS The use of nCIT should be recommended for resectable NSCLC patients with negative PD-L1 expression, as nCIT significantly improved the pathological response and EFS in these patients. The benefit to PD-L1-negative patients treated with nCIT on OS remains to be validated.
Collapse
Affiliation(s)
- Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yuan Tian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jing Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jie-Hui Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
29
|
Wang Y, Song Y, Wang R, Wu Y, Li M, Xu K, He R, Wang Z, Li Q, Kong FM(S, Wang T. Clinical factors and major pathological response after neoadjuvant chemoimmunotherapy in potentially resectable lung squamous cell carcinoma. Front Oncol 2024; 14:1265228. [PMID: 38680859 PMCID: PMC11045983 DOI: 10.3389/fonc.2024.1265228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 03/18/2024] [Indexed: 05/01/2024] Open
Abstract
Objective Major pathological response (MPR) helps evaluate the prognosis of patients with lung squamous cell carcinoma (LUSC). However, the clinical factors that affect the achievement of MPR after neoadjuvant chemoimmunotherapy (NCIO) in patients with LUSC remain unclear. This study aimed to explore the clinical factors affecting the MPR after NCIO in patients with potentially resectable LUSC. Methods This retrospective study included patients with stage IIB-IIIC LUSC who underwent surgical resection after receiving NCIO at a center between March 2020 and November 2022. In addition to the postoperative pathological remission rate, sex, age, body mass index (BMI), smoking history, TNM stage, hematological and imaging test results, and other indicators were examined before NCIO. According to the pathological response rate of the surgically removed tumor tissue, the patients were split into MPR and non-MPR groups. Results In total, 91 LUSC patients who met the study's eligibility criteria were enrolled: 32 (35%) patients in the non-MPR group and 59 (65%) in the MPR group, which included 43 cases of pathological complete remission (pCR). Pre-treatment lymphocyte level (LY) (odds ratio [OR] =5.997), tumor burden (OR=0.958), N classification (OR=15.915), radiographic response (OR=11.590), pulmonary atelectasis (OR=5.413), and PD-L1 expression (OR=1.028) were independently associated with MPR (all P < 0.05). Based on these six independent predictors, we developed a nomogram model of prediction having an area under the curve (AUC) of 0.914 that is simple to apply clinically to predict the MPR. The MPR group showed greater disease-free survival (DFS) than the non-MPR group, according to the survival analysis (P < 0.001). Conclusion The MPR rate of NCIO for potentially resectable LUSC was 65%. LY, tumor burden, N classification, radiographic response, pulmonary atelectasis, and PD-L1 expression in patients with LUSC before NCIO were the independent and ideal predictors of MPR. The developed nomogram demonstrated a good degree of accuracy and resilience in predicting the MPR following NCIO, indicating that it is a useful tool for assuring customized therapy for patients with possibly resectable LUSC.
Collapse
Affiliation(s)
- Ye Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- School of Graduate, Dalian Medical University, Dalian, China
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Runze Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Yu Wu
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- School of Graduate, Dalian Medical University, Dalian, China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Ke Xu
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Rong He
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Zheng Wang
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Qingqing Li
- Department of Endoscopy, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- Faculty of Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
30
|
Vaidya P, Cohen EE. Facts and Hopes in Neoadjuvant Immunotherapy: Current Approvals and Emerging Evidence. Clin Cancer Res 2024; 30:1232-1239. [PMID: 37955563 PMCID: PMC10984792 DOI: 10.1158/1078-0432.ccr-23-0583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/27/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
In 2021 and 2022, two immune checkpoint inhibitors received FDA approval in the neoadjuvant setting for the treatment of early-stage triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC). Several more studies have since indicated the benefits, and challenges, of administering neoadjuvant immunotherapy prior to definitive surgery in the gastrointestinal, head and neck, and cutaneous realms. In addition, numerous ongoing phase II and phase III trials are investigating outcomes of neoadjuvant immune treatment in early-stage disease. As such, it is anticipated that more immune checkpoint inhibitors will receive approval for various neoadjuvant indications in the next several years. Medical oncologists, surgeons, and other providers in a multidisciplinary cancer care team will be presented with alternate treatment paradigms and clinical decisions regarding upfront surgery versus neoadjuvant treatment. Here, we describe the current evidence supporting use of immune checkpoint inhibitors for neoadjuvant treatment, ongoing studies, and clinical considerations of this treatment approach.
Collapse
Affiliation(s)
- Poorva Vaidya
- Dept of Internal Medicine, Division of Hematology-Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Ezra E.W. Cohen
- Dept of Internal Medicine, Division of Hematology-Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
31
|
Mei T, Zhou Q, Gong Y. Comparison of the Efficacy and Safety of Perioperative Immunochemotherapeutic Strategies for Resectable Non-small Cell Lung Cancer: a Systematic Review and Network Meta-analysis. Clin Oncol (R Coll Radiol) 2024; 36:107-118. [PMID: 38151439 DOI: 10.1016/j.clon.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023]
Abstract
AIMS The aim of this network meta-analysis was to elucidate the efficacy and safety of various immune checkpoint inhibitors (ICIs) used in combination with chemotherapy for the treatment of non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Data from randomised controlled trials comparing perioperative ICI-chemotherapy and chemotherapy alone were acquired from the EMBASE, Web of Science, Cochrane Library databases, PubMed, and meeting abstracts from inception until August 2023. The endpoints for this analysis were pathological complete response, event-free survival and treatment-related adverse events of any grade or adverse events of grade 3 or higher. RESULTS In total, six randomised controlled trials with 2538 NSCLC patients were selected for this network meta-analysis. Compared with other ICIs, toripalimab + chemotherapy demonstrated increased pathological complete response rates and prolonged event-free survival in NSCLC. In patients with negative/low PD-L1 expression or squamous cell pathology, toripalimab + chemotherapy was the most effective regimen. In contrast, nivolumab + chemotherapy was preferable for patients with high PD-L1 expression or non-squamous cell pathology. Among the analysed regimens, toripalimab + chemotherapy presented the highest risk of adverse events of any grade, whereas nivolumab + chemotherapy showed the highest risk of grade 3-4 adverse events. Conversely, durvalumab + chemotherapy exhibited the lowest risk of grade 3-4 adverse events. CONCLUSIONS Among the evaluated perioperative immunochemotherapy regimens, toripalimab + chemotherapy indicated a significantly increased survival benefit for most resectable NSCLC patients. However, for high PD-L1 expression and non-squamous NSCLC patients, nivolumab + chemotherapy provided the most potent outcomes. Perioperative durvalumab + chemotherapy is a relatively safe treatment. The findings of this investigation are expected to assist clinicians in making informed decisions among promising treatment options.
Collapse
Affiliation(s)
- T Mei
- Division of Thoracic Tumor Multidisciplinary Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China; Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Q Zhou
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| | - Y Gong
- Division of Thoracic Tumor Multidisciplinary Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
32
|
Tan J, Xue Q, Hu X, Yang J. Inhibitor of PD-1/PD-L1: a new approach may be beneficial for the treatment of idiopathic pulmonary fibrosis. J Transl Med 2024; 22:95. [PMID: 38263193 PMCID: PMC10804569 DOI: 10.1186/s12967-024-04884-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a globally prevalent, progressive disease with limited treatment options and poor prognosis. Because of its irreversible disease progression, IPF affects the quality and length of life of patients and imposes a significant burden on their families and social healthcare services. The use of the antifibrotic drugs pirfenidone and nintedanib can slow the progression of the disease to some extent, but it does not have a reverse effect on the prognosis. The option of lung transplantion is also limited owing to contraindications to transplantation, possible complications after transplantation, and the risk of death. Therefore, the discovery of new, effective treatment methods is an urgent need. Over recent years, various studies have been undertaken to investigate the relationship between interstitial pneumonia and lung cancer, suggesting that some immune checkpoints in IPF are similar to those in tumors. Immune checkpoints are a class of immunosuppressive molecules that are essential for maintaining autoimmune tolerance and regulating the duration and magnitude of immune responses in peripheral tissues. They can prevent normal tissues from being damaged and destroyed by the immune response. While current studies have focused on PD-1/PD-L1 and CTLA-4, PD-1/PD-L1 may be the only effective immune checkpoint IPF treatment. This review discusses the application of PD-1/PD-L1 checkpoint in IPF, with the aim of finding a new direction for IPF treatment.
Collapse
Affiliation(s)
- Jie Tan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Qianfei Xue
- Hospital of Jilin University, Changchun, China
| | - Xiao Hu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
33
|
Wu Y, Hu L, Zhang S, Zhang H. The Value of Perioperative Immunotherapy for Non-Small Cell Lung Cancer: A Pool- and Meta-Analysis. Technol Cancer Res Treat 2024; 23:15330338241258164. [PMID: 38872482 PMCID: PMC11179512 DOI: 10.1177/15330338241258164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 06/15/2024] Open
Abstract
Purpose: This study aimed to analyze the efficacy and safety of neoadjuvant and adjuvant immunotherapies for non-small cell lung cancer (NSCLC). Methods: Electronic literature searches were conducted in PubMed, OVID, Web of SCI, Embase, Cochrane Library, and the Chinese National Knowledge Infrastructure databases. The deadline for literature update and retrieval is February 16, 2024. Studies presented at meetings were also screened. Randomized controlled trials (RCTs) and single-arm trials were included, and the data were extracted according to the inclusion and exclusion criteria. Data analysis was performed using Stata (16.0) software. Results: A total of 5850 patients in 11 RCTs and 6 single-arm trial studies involving neoadjuvant and/or adjuvant immune checkpoint inhibitor (ICI)-based therapies were included. Regarding neoadjuvant therapy, the overall complication rate after surgery reached 35% (95% CI, 0.21-0.49). Higher rates of pathological complete response (OR = 7.83; 95% CI, 5.95-10.31; P < .001) and major pathological response (OR = 5.13; 95% CI, 3.56-7.40; P < .001) were found in the resectable NSCLC patients who received neoadjuvant therapy with ICIs combined with chemotherapy compared with patients treated with chemotherapy alone. Of note, compared with chemotherapy, neoadjuvant ICIs combined with chemotherapy significantly improved the overall survival (OS) (HR = 0.65; 95% CI, 0.52-0.82; P < .001) and event-free survival (EFS) (HR = 0.59; 95% CI, 0.52-0.67; P < .001) in patients with resectable NSCLC. Regarding adjuvant therapy, a lower risk of disease progression or death (HR = 0.78; 95% CI, 0.69-0.90; P < .001) was found in the adjuvant ICI group compared with the adjuvant chemotherapy-alone group. In terms of safety, perioperative immunotherapy combined with chemotherapy did not increase toxicity compared with chemotherapy alone. Conclusion: In patients with resectable NSCLC, perioperative immunotherapy was safe and efficacious. Perioperative immunotherapy combined with chemotherapy improved the pathologic response and EFS/DFS/OS over chemotherapy alone without increasing toxicity.
Collapse
Affiliation(s)
- Yanmeng Wu
- China Medical University, Shenyang, China
| | - Lin Hu
- China Medical University, Shenyang, China
| | - Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui Zhang
- Traditional Chinese Medicine Department, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
34
|
Yu S, Zhai S, Gong Q, Xiang C, Gong J, Wu L, Pu X. Neoadjuvant Immunotherapy and Non-Small Cell Lung Cancer: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Am J Clin Oncol 2023; 46:517-528. [PMID: 37749786 PMCID: PMC10589427 DOI: 10.1097/coc.0000000000001046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
OBJECTIVES To systematically evaluate the effectiveness and safety of neoadjuvant immunotherapy for patients with non-small cell lung cancer (NSCLC). METHODS Randomized controlled trials of neoadjuvant immunotherapy in treating patients with NSCLC were comprehensively retrieved from electronic databases, eligible studies, previous systematic reviews and meta-analyses, guidelines, and conference abstracts. The meta-analysis was performed by the Stata/SE 12.0 software. RESULTS Eleven randomized controlled trials were eventually included. The results of the meta-analysis showed that neoadjuvant immunochemotherapy significantly improved the objective response rate compared with neoadjuvant chemotherapy (CT; 62.46% vs 41.88%, P = 0.003), but the objective response rate of neoadjuvant double-immunotherapy was roughly comparable to that of neoadjuvant single-immunotherapy (15.74% vs 10.45%, P = 0.387). Major pathologic response (MPR) rate and pathologic complete response (pCR) rate of neoadjuvant immunochemotherapy and neoadjuvant double-immunotherapy were significantly superior to neoadjuvant CT alone and neoadjuvant single-immunotherapy, respectively. Compared with neoadjuvant CT alone, neoadjuvant immunochemotherapy increased the down-staging rate (40.16% vs 26.70%, P = 0.060), the surgical resection rate (83.69% vs 73.07%, P = 0.231), and R0 resection rate (86.19% vs 77.98%, P = 0.502), but there were no statistically significant differences. Neoadjuvant immunochemotherapy did not increase the postoperative complications rate than neoadjuvant CT alone (40.20% vs 41.30%, P = 0.920). In terms of safety, neoadjuvant immunochemotherapy and neoadjuvant double-immunotherapy did not increase the incidence of treatment-related adverse events (TRAEs) and the grade 3 or higher TRAEs. CONCLUSIONS In summary, neoadjuvant immunochemotherapy had better clinical efficacy than neoadjuvant CT for patients with NSCLC. MPR rate and pCR rate of neoadjuvant immunochemotherapy and neoadjuvant double-immunotherapy were significantly superior to neoadjuvant CT and neoadjuvant single-immunotherapy, respectively, for patients with NSCLC, showing that MPR rate and pCR rate were probably considered as alternative endpoints for survival benefit. TRAEs were comparable between the corresponding groups. The long-term survival outcome of neoadjuvant immunotherapy for patients with NSCLC needs to be further confirmed to better guide clinical practice.
Collapse
Affiliation(s)
- Shaofu Yu
- Department of Clinical Pharmacy, the Second People’s Hospital of Huaihua
- The Second Department of Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha
| | - Shasha Zhai
- Department of Trauma Surgery, the First Affiliated Hospital of Hunan University of Medicine, Huaihua
| | - Qian Gong
- Department of Clinical Pharmacy, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Chunhong Xiang
- Department of Clinical Pharmacy, the Second People’s Hospital of Huaihua
| | - Jianping Gong
- Department of Clinical Pharmacy, the Second People’s Hospital of Huaihua
| | - Lin Wu
- The Second Department of Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha
| | - Xingxiang Pu
- The Second Department of Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha
| |
Collapse
|
35
|
Attieh F, Chartouni A, Boutros M, Mouawad A, Kourie HR. Tackling the immunotherapy conundrum: advances and challenges for operable non-small-cell lung cancer treatment. Immunotherapy 2023; 15:1415-1428. [PMID: 37671552 DOI: 10.2217/imt-2023-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide. Non-small-cell lung cancer (NSCLC) represents the majority of lung cancer cases, and its standard treatment is primarily surgery. Nonetheless, this type of cancer exhibits an important rate of tumor recurrence. Immune checkpoint inhibitors (ICIs) have demonstrated significant survival benefits in many cancers, especially in early-stage NSCLC. This review considers the latest CheckMate816, IMpower010 and KEYNOTE-091 trials that led to US FDA approvals. The new wave of resectable NSCLC trial results are also summarized. Finally, the latest challenges for these treatment modalities, such as the choice between neoadjuvant and adjuvant use, the accurate identification of biomarkers and the presence of driver mutations such as EGFR, are discussed.
Collapse
Affiliation(s)
- Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Antoine Mouawad
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| |
Collapse
|
36
|
Kou J, Huang J, Li J, Wu Z, Ni L. Systemic immune-inflammation index predicts prognosis and responsiveness to immunotherapy in cancer patients: a systematic review and meta‑analysis. Clin Exp Med 2023; 23:3895-3905. [PMID: 36966477 DOI: 10.1007/s10238-023-01035-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/01/2023] [Indexed: 03/27/2023]
Abstract
The systemic immune-inflammation index (SII) is a significant prognostic factor in some cancer types. However, the prognostic role of SII in cancer patients with immunotherapy remains uncertain. We aimed to evaluate the relationship between pretreatment SII and clinical survival outcomes for advanced-stage cancer patients treated with immune checkpoint inhibitors (ICIs). A comprehensive literature search was performed to identify eligible studies concerning the association between pretreatment SII and survival outcomes in advanced cancer patients treated with ICIs. The data were extracted from publications and used to calculate the pooled odds ratio (pOR) for objective response rate (ORR), disease control rate (DCR), and pooled hazard ratio (pHR) for overall survival (OS), progressive-free survival (PFS), along with 95% confidence intervals (95% CIs). Fifteen articles with 2438 participants were included. A higher level of SII indicated a lower ORR (pOR = 0.73, 95% CI 0.56-0.94) and worse DCR (pOR = 0.56, 95% CI 0.35-0.88). High SII was associated with a shorter OS (pHR = 2.33, 95% CI 2.02-2.69) and unfavorable PFS (pHR = 1.85, 95% CI 1.61-2.14). Therefore, high SII level might be a non-invasive and efficacious biomarker of poor tumor response and adverse prognosis of advanced cancer patients with immunotherapy.
Collapse
Affiliation(s)
- Junyan Kou
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jing Huang
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jun Li
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Zhen Wu
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Liwei Ni
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China.
| |
Collapse
|
37
|
Boutros M, Attieh F, Chartouni A, Jalbout J, Kourie HR. Beyond the Horizon: A Cutting-Edge Review of the Latest Checkpoint Inhibitors in Cancer Treatment. Cancer Invest 2023; 41:757-773. [PMID: 37795860 DOI: 10.1080/07357907.2023.2267675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a revolutionary paradigm in oncology, offering a potent arsenal against various malignancies by harnessing the body's own immunological prowess. In a whirlwind of advancement, an abundance of new ICIs have come to light, rendering it a Herculean task for physicians to remain au courant with the rapidly evolving landscape. This comprehensive review meticulously explores the crescendo of clinical investigations and FDA approvals that have come to light during 2022 and 2023, showcasing the metamorphic impact of ICIs in cancer therapeutics. Delving into the pith of pivotal Phase 3 trials across diverse cancer types - including lung, renal, melanoma, and more - the review illuminates the significant strides made in enhancing patient outcomes, alongside the unveiling of novel ICIs that have garnered attention in the oncological community. The analysis extends to the notable presentations at the esteemed ESMO and ASCO conventions, providing a panoramic view of the contemporary advancements in ICI technology. Furthermore, the review underscores the imperative of continuous exploration in overcoming the extant challenges, such as the quest for reliable predictive biomarkers and the optimization of combinatorial strategies to surmount resistance and augment therapeutic efficacy. Through a holistic lens, this article elucidates the monumental impact of ICIs, marking a significant epoch in the odyssey towards rendering cancer a conquerable adversary.
Collapse
Affiliation(s)
- Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Johnny Jalbout
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Faculty of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
38
|
Wankhede D, Grover S, Hofman P. The prognostic value of TMB in early-stage non-small cell lung cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231195199. [PMID: 37667779 PMCID: PMC10475237 DOI: 10.1177/17588359231195199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
Background Tumor mutation burden (TMB) has been validated as a predictive biomarker for immunotherapy response and survival in numerous cancer types. Limited data is available on the inherent prognostic role of TMB in early-stage tumors. Objective To evaluate the prognostic role of TMB in early-stage, resected non-small cell lung cancer (NSCLC). Design Systematic review and meta-analysis of pertinent prospective and retrospective studies. Data sources and methods Publication search was performed in PubMed, Embase, Cochrane Library, and Web of Science databases. Based on the level of heterogeneity, a random- or fixed-effects model was used to calculate pooled effects of hazard ratio (HR) for overall survival (OS) and disease-free survival (DFS). The source of heterogeneity was investigated using sensitivity analysis, subgroup analysis, and publication bias assessment. Results Ten studies comprising 2520 patients were included in this analysis. There was no statistically significant difference in OS (HR, 1.18, 95% CI, 0.70, 1.33; p 0.53, I2 = 80%; phet < 0.0001) and DFS (HR, 1.18, 95% CI, 0.91, 1.52; p = 0.53, I2 = 75%; phet = 0.0001) between the high-TMB and low-TMB group. Subgroup analyses indicated that East Asian ethnicity, and TMB detected using whole exome sequencing, and studies with <100 patients had poor DFS in the high-TMB group. Conclusion The inherent prognostic role of TMB is limited in early-stage NSCLC. Ethnic differences in mutation burden must be considered while designing future trials on neoadjuvant immunotherapy. Further research in the harmonization and standardization of panel-based TMB is essential for its widespread clinical utility.Registration: CRD42023392846.
Collapse
Affiliation(s)
- Durgesh Wankhede
- German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg – Standort Marburg, Marburg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Côte d’Azur, Nice, France
- Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081 and UMR CNRS 7284, Team 4, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Pasteur Hospital, Nice, France
- University Hospital Institute RespirERA, Nice, France
- University Hospital Federation OncoAge, CHU de Nice, University Côte d’Azur, Nice, France
| |
Collapse
|
39
|
Shi Y, Ma X, He D, Dong B, Qiao T. Neoadjuvant SBRT combined with immunotherapy in NSCLC: from mechanisms to therapy. Front Immunol 2023; 14:1213222. [PMID: 37600799 PMCID: PMC10435737 DOI: 10.3389/fimmu.2023.1213222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
The utilisation of neoadjuvant immunotherapy has demonstrated promising preliminary clinical outcomes for early-stage resectable non-small-cell lung cancer (NSCLC). Nevertheless, it is imperative to develop novel neoadjuvant combination therapy regimens incorporating immunotherapy to further enhance the proportion of patients who derive benefit. Recent studies have revealed that stereotactic body radiotherapy (SBRT) not only induces direct tumour cell death but also stimulates local and systemic antitumour immune responses. Numerous clinical trials have incorporated SBRT into immunotherapy for advanced NSCLC, revealing that this combination therapy effectively inhibits local tumour growth while simultaneously activating systemic antitumour immune responses. Consequently, the integration of SBRT with neoadjuvant immunotherapy has emerged as a promising strategy for treating resectable NSCLC, as it can enhance the systemic immune response to eradicate micrometastases and recurrent foci post-resection. This review aims to elucidate the potential mechanism of combination of SBRT and immunotherapy followed by surgery and identify optimal clinical treatment strategies. Initially, we delineate the interplay between SBRT and the local tumour immune microenvironment, as well as the systemic antitumour immune response. We subsequently introduce the preclinical foundation and preliminary clinical trials of neoadjuvant SBRT combined with immunotherapy for treating resectable NSCLC. Finally, we discussed the optimal dosage, schedule, and biomarkers for neoadjuvant combination therapy in its clinical application. In conclusion, the elucidation of potential mechanism of neoadjuvant SBRT combined immunotherapy not only offers a theoretical basis for ongoing clinical trials but also contributes to determining the most efficacious therapy scheme for future clinical application.
Collapse
Affiliation(s)
- Yanhong Shi
- Department of Pathology, Xianyang Central Hospital, Xianyang, China
| | - Xiaoyan Ma
- Department of Pathology, Division of Experimental Diagnostic, KingMed Medical Laboratory (Xi’an) Co., Ltd., Xi’an, China
| | - Dan He
- Department of Pathology, Xi’an Central Hospital, Xi’an, China
| | - Bingwei Dong
- Department of Pathology, Xianyang Central Hospital, Xianyang, China
| | - Tianyun Qiao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
40
|
Gao J, Zhang C, Wei Z, Ye X. Immunotherapy for early-stage non-small cell lung cancer: A system review. J Cancer Res Ther 2023; 19:849-865. [PMID: 37675709 DOI: 10.4103/jcrt.jcrt_723_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/06/2023] [Indexed: 09/08/2023]
Abstract
With the addition of immunotherapy, lung cancer, one of the most common cancers with high mortality rates, has broadened the treatment landscape. Immune checkpoint inhibitors have demonstrated significant efficacy in the treatment of non-small cell lung cancer (NSCLC) and are now used as the first-line therapy for metastatic disease, consolidation therapy after radiotherapy for unresectable locally advanced disease, and adjuvant therapy after surgical resection and chemotherapy for resectable disease. The use of adjuvant and neoadjuvant immunotherapy in patients with early-stage NSCLC, however, is still debatable. We will address several aspects, namely the initial efficacy of monotherapy, the efficacy of combination chemotherapy, immunotherapy-related biomarkers, adverse effects, ongoing randomized controlled trials, and current issues and future directions for immunotherapy in early-stage NSCLC will be discussed here.
Collapse
Affiliation(s)
- Jingyi Gao
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong; Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| | - Chao Zhang
- Department of Oncology, Affiliated Qujing Hospital of Kunming Medical University, QuJing, Yunnan Province, China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| |
Collapse
|
41
|
Gui Z, Du J, Wu N, Shen N, Yang Z, Yang H, Wang X, Zhao N, Zeng Z, Wei R, Ma W, Wang C. Immune regulation and prognosis indicating ability of a newly constructed multi-genes containing signature in clear cell renal cell carcinoma. BMC Cancer 2023; 23:649. [PMID: 37438709 DOI: 10.1186/s12885-023-11150-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/04/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common renal malignancy, although newly developing targeted therapy and immunotherapy have been showing promising effects in clinical treatment, the effective biomarkers for immune response prediction are still lacking. The study is to construct a gene signature according to ccRCC immune cells infiltration landscape, thus aiding clinical prediction of patients response to immunotherapy. METHODS Firstly, ccRCC transcriptome expression profiles from Gene Expression Omnibus (GEO) database as well as immune related genes information from IMMPORT database were combine applied to identify the differently expressed meanwhile immune related candidate genes in ccRCC comparing to normal control samples. Then, based on protein-protein interaction network (PPI) and following module analysis of the candidate genes, a hub gene cluster was further identified for survival analysis. Further, LASSO analysis was applied to construct a signature which was in succession assessed with Kaplan-Meier survival, Cox regression and ROC curve analysis. Moreover, ccRCC patients were divided as high and low-risk groups based on the gene signature followed by the difference estimation of immune treatment response and exploration of related immune cells infiltration by TIDE and Cibersort analysis respectively among the two groups of patients. RESULTS Based on GEO and IMMPORT databases, a total of 269 differently expressed meanwhile immune related genes in ccRCC were identified, further PPI network and module analysis of the 269 genes highlighted a 46 genes cluster. Next step, Kaplan-Meier and Cox regression analysis of the 46 genes identified 4 genes that were supported to be independent prognosis indicators, and a gene signature was constructed based on the 4 genes. Furthermore, after assessing its prognosis indicating ability by both Kaplan-Meier and Cox regression analysis, immune relation of the signature was evaluated including its association with environment immune score, Immune checkpoint inhibitors expression as well as immune cells infiltration. Together, immune predicting ability of the signature was preliminary explored. CONCLUSIONS Based on ccRCC genes expression profiles and multiple bioinformatic analysis, a 4 genes containing signature was constructed and the immune regulation of the signature was preliminary explored. Although more detailed experiments and clinical trials are needed before potential clinical use of the signature, the results shall provide meaningful insight into further ccRCC immune researches.
Collapse
Affiliation(s)
- Ziwei Gui
- Department of Pathology, Second Clinical Medical College of ShanXi Medical University, Tai Yuan City, ShanXi Province, China
| | - Juan Du
- Department of Pathology, Second Clinical Medical College of ShanXi Medical University, Tai Yuan City, ShanXi Province, China
| | - Nan Wu
- Department of Anesthesiology, Second Hospital of ShanXi Medical University, Tai Yuan, ShanXi Province, China
| | - Ningning Shen
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China
| | - Zhiqing Yang
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China
| | - Huijun Yang
- Department of Pathology, Second Clinical Medical College of ShanXi Medical University, Tai Yuan City, ShanXi Province, China
| | - Xuzhi Wang
- Department of Pathology, Second Clinical Medical College of ShanXi Medical University, Tai Yuan City, ShanXi Province, China
| | - Na Zhao
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China
| | - Zixin Zeng
- Department of Pathology, Second Clinical Medical College of ShanXi Medical University, Tai Yuan City, ShanXi Province, China
| | - Rong Wei
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China
| | - Wenxia Ma
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China.
| | - Chen Wang
- Department of Pathology, Second Hospital of ShanXi Medical University, No.382 Wuyi Road, Tai Yuan, ShanXi Province, 030000, China.
| |
Collapse
|
42
|
Wei M, Shen X, Fan X, Li J, Bai J. PD-L1 aptamer-functionalized degradable hafnium oxide nanoparticles for near infrared-II diagnostic imaging and radiosensitization. Front Bioeng Biotechnol 2023; 11:1224339. [PMID: 37351473 PMCID: PMC10282151 DOI: 10.3389/fbioe.2023.1224339] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
Immune checkpoint blockade is now recognized as a paradigm-shifting cancer therapeutic strategy, whereas there remains difficulty in accurately predicting immunotherapy efficacy by PD-L1 expression. In addition, radiotherapy for cancer patients faces the problem of insufficient dose of radiotherapy at the tumor site while which have been not tolerated by normal tissues. In this study, we created PD-L1 aptamer-anchored spherical nucleic acids (SNAs) with a shell made of PD-L1 aptamer and indocyanine green (ICG) embedded in a mesoporous hafnium oxide nanoparticle core (Hf@ICG-Apt). Upon low pH irradiation in the tumor sites, the nano-system enabled the release of ICG in the high PD-L1 expression tumor to develop a high tumor-to-background ratio of 7.97 ± 0.76 and enhanced the ICG tumor retention to more than 48 h. Moreover, Hf@ICG-Apt improved radiation therapy (RT) when combined with radiation. Notably, Hf@ICG-Apt showed scarcely any systemic toxicity in vivo. Overall, this research offered a novel approach for applying reliable monitoring of PD-L1 expression and localization and robust RT sensitization against cancer with good biosafety.
Collapse
Affiliation(s)
- Min Wei
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Shen
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqi Fan
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiwei Li
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingwen Bai
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
43
|
Yang Y, Xin D, Wang H, Guan L, Meng X, Lu T, Bai X, Wang F. A Novel Predictor of Pathologic Complete Response for Neoadjuvant Immunochemotherapy in Resectable Locally Advanced Esophageal Squamous Cell Carcinoma. J Inflamm Res 2023; 16:1443-1455. [PMID: 37042015 PMCID: PMC10083012 DOI: 10.2147/jir.s395231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Purpose Neoadjuvant immunochemotherapy (nICT) for resectable locally advanced esophageal squamous cell carcinoma (LA-ESCC) has attracted widespread attention recently, whose safety and clinical benefit was observed in clinical researches. This study aimed to develop and validate a novel predictor systemic inflammation-tumor markers index (SITI) to predict the pathological complete response (pCR) for resectable LA-ESCC patients receiving nICT. Patients and Methods A total of 147 LA-ESCC patients who underwent nICT followed by surgery from February 2020 to April 2022 were included in the study. The dynamic change of inflammatory indexes was compared at baseline, after two cycles of nICT and postoperative one month. Least absolute shrinkage and selection operator (LASSO) regression was performed to avoid collinearity and identify key indexes, with SITI constructed. After univariate and multivariate stepwise forward logistic analyses, a nomogram for pCR prediction was developed. Results 41(27.9%) patients achieved pCR among 147 resectable LA-ESCC patients received nICT. Compared with baseline, most inflammatory indexes were significantly decreased at postoperative one month. 5 key indexes were identified and then a predictive index named SITI was constructed. The result showed that lower SITI and earlier clinical tumor node metastasis (cTNM) stage were more likely to achieve pCR. The nomogram for pCR prediction had excellent discrimination performance (C-index = 0.791). Conclusion The SITI is an independent predictor for pCR in resectable LA-ESCC patients received nICT. To our knowledge, our nomogram is the first model using systemic inflammation-tumor markers for pCR prediction and may be a promising predictor to effectively differentiate pCR for nICT in LA-ESCC patients.
Collapse
Affiliation(s)
- Yalan Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Dao Xin
- Department of Medical Oncology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Huike Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Lulu Guan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiangrui Meng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Taiying Lu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiwen Bai
- Department of Translational Medicine, Nanchang University Queen Mary School, Nanchang, People’s Republic of China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Correspondence: Feng Wang, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China, Email
| |
Collapse
|
44
|
Yuan W, Xiao JH, Zhang JS, Mao BL, Wang PZ, Wang BL. Identification of a cuproptosis and copper metabolism gene-related lncRNAs prognostic signature associated with clinical and immunological characteristics of hepatocellular carcinoma. Front Oncol 2023; 13:1153353. [PMID: 37056336 PMCID: PMC10086263 DOI: 10.3389/fonc.2023.1153353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Background The relationship between cuproptosis and HCC is still in the exploratory stage. Long noncoding RNAs (lncRNAs) have recently been linked to the progression of hepatocellular carcinoma (HCC). However, the clinical significance of lncRNAs associated with cuproptosis remains unclear. Methods Based on The Cancer Genome Atlas (TCGA) liver hepatocellular carcinoma (LIHC) dataset, we identified characteristic prognostic lncRNAs by univariate, LASSO, and multifactorial regression analysis, and constructed a prognostic signature of cuproptosis-related lncRNAs in HCC. The role of lncRNAs were identified through CCK-8, clone formation in Huh-7 cells with high expression of FDX1. Prognostic potential of the characteristic lncRNAs was evaluated in each of the two cohorts created by randomly dividing the TCGA cohort into a training cohort and a test cohort in a 1:1 ratio. Immune profiles in defined subgroups of cuproptosis-related lncRNA features as well as drug sensitivity were analyzed. Results We constructed a multigene signature based on four characteristic prognostic lncRNAs (AL590705.3, LINC02870, KDM4A-AS1, MKLN1-AS). These four lncRNAs participated in the development of cuproptosis. HCC patients were classified into high-risk and low-risk groups based on the median value of the risk score. The receiver operating characteristic curve area under the curve values for 1-, 3-, and 5-year survival were 0.773, 0.728, and 0.647, respectively, for the training cohort, and 0.764, 0.671, and 0.662, respectively, for the test cohort. Univariate and multifactorial regression analyses indicated that this prognostic feature was an independent prognostic factor for HCC. Principal component analysis plots clearly distinguished between low- and high-risk patients in terms of their probability of survival. Furthermore, gene set enrichment analysis showed that a variety of processes associated with tumor proliferation and progression were enriched in the high-risk group compared with the low-risk group. Moreover, there were significant differences in the expression of immune cell subpopulations, immune checkpoint genes, and potential drug screening, which provided distinct therapeutic recommendations for individuals with various risks. Conclusions We constructed a novel cuproptosis-associated lncRNA signature with a significant predictive value for the prognosis of patients with HCC. Cuproptosis-associated lncRNAs are associated with the tumor immune microenvironment of HCC and even the efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Hepatobiliary Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jun-hao Xiao
- Department of Clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jian-song Zhang
- Department of Clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ben-liang Mao
- Department of Clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng-zhen Wang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Bai-lin Wang
- Department of Hepatobiliary Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Roulleaux Dugage M, Albarrán-Artahona V, Laguna JC, Chaput N, Vignot S, Besse B, Mezquita L, Auclin E. Biomarkers of response to immunotherapy in early stage non-small cell lung cancer. Eur J Cancer 2023; 184:179-196. [PMID: 36963241 DOI: 10.1016/j.ejca.2023.01.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023]
Abstract
Immunotherapy with immune-checkpoint inhibitors (ICIs) targeting programmed cell death 1 or programmed death-ligand 1 has revolutionised the treatment of advanced non-small cell lung cancer (NSCLC) and has been investigated in early NSCLC, alone or in combination with chemotherapy, anti-CTLA-4 antibodies and radiotherapy. Although more mature data are needed before setting a change of paradigm in early stages, reports of pathological response rates and disease-free survival are promising, especially with neoadjuvant multimodality approaches. Nevertheless, major pathological response rates for neoadjuvant anti-PD-(L)1 monotherapy rarely exceed 40%, and biomarkers for characterising patients who may benefit the most from ICIs are lacking. These biomarkers have a distinct value from the metastatic setting, with highly different tumour biologies. Among the most investigated so far in this context, programmed death-ligand 1 expression and, to a lesser extent, tumour mutational burden seem to correlate better with higher pathological response rates and survival. Epidermal growth factor receptor, Serine/Threonine Kinase 11and Kelch-like ECH-associated protein 1 mutations rise as essential determinations for the treatment selection in early-stage NSCLC. Emerging and promising approaches comprise evaluation of blood-based ratios, microbiota, and baseline intratumoural TCR clonality. Circulating tumour DNA will be of great help in the near future when selecting best candidates for adjuvant ICIs, monitoring the tumour response to the neoadjuvant treatment in order to improve the rates of complete resections in the early stage.
Collapse
Affiliation(s)
- Matthieu Roulleaux Dugage
- Department of Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France; Laboratoire D'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, Île-de-France, France
| | - Víctor Albarrán-Artahona
- Medical Oncology Department, Hospital Clinic de Barcelona, Spain; Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | | | - Nathalie Chaput
- Laboratoire D'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, Île-de-France, France
| | | | - Benjamin Besse
- Department of Oncology, Gustave Roussy, Villejuif, Île-de-France, France
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic de Barcelona, Spain; Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Edouard Auclin
- Department of Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France.
| |
Collapse
|
46
|
Peng Y, Li Z, Fu Y, Pan Y, Zeng Y, Liu J, Xiao C, Zhang Y, Su Y, Li G, Wu F. Progress and perspectives of perioperative immunotherapy in non-small cell lung cancer. Front Oncol 2023; 13:1011810. [PMID: 36761954 PMCID: PMC9905802 DOI: 10.3389/fonc.2023.1011810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death. Lung cancer mortality has decreased over the past decade, which is partly attributed to improved treatments. Curative surgery for patients with early-stage lung cancer is the standard of care, but not all surgical treatments have a good prognosis. Adjuvant and neoadjuvant chemotherapy are used to improve the prognosis of patients with resectable lung cancer. Immunotherapy, an epoch-defining treatment, has improved curative effects, prognosis, and tolerability compared with traditional and ordinary cytotoxic chemotherapy, providing new hope for patients with non-small cell lung cancer (NSCLC). Immunotherapy-related clinical trials have reported encouraging clinical outcomes in their exploration of different types of perioperative immunotherapy, from neoadjuvant immune checkpoint inhibitor (ICI) monotherapy, neoadjuvant immune-combination therapy (chemoimmunotherapy, immunotherapy plus antiangiogenic therapy, immunotherapy plus radiotherapy, or concurrent chemoradiotherapy), adjuvant immunotherapy, and neoadjuvant combined adjuvant immunotherapy. Phase 3 studies such as IMpower 010 and CheckMate 816 reported survival benefits of perioperative immunotherapy for operable patients. This review summarizes up-to-date clinical studies and analyzes the efficiency and feasibility of different neoadjuvant therapies and biomarkers to identify optimal types of perioperative immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Yurong Peng
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhuo Li
- The Ophthalmologic Center of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yucheng Fu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Pan
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Zeng
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junqi Liu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chaoyue Xiao
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yingzhe Zhang
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yahui Su
- XiangYa School of Public Health, Central South University, Changsha, Hunan, China
| | - Guoqing Li
- XiangYa School of Public Health, Central South University, Changsha, Hunan, China
| | - Fang Wu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
47
|
Wu X, Chau YF, Bai H, Zhuang X, Wang J, Duan J. Progress on neoadjuvant immunotherapy in resectable non-small cell lung cancer and potential biomarkers. Front Oncol 2023; 12:1099304. [PMID: 36761426 PMCID: PMC9902866 DOI: 10.3389/fonc.2022.1099304] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/31/2022] [Indexed: 01/25/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are highly concerned in the treatment of non-small cell lung cancer (NSCLC), represented by inhibitors of programmed death protein 1 (PD-1) and its ligand (PD-L1), and inhibitors of cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). The introduction of immunotherapy in the treatment of perioperative NSCLC has improved the prognosis to a great extent, as demonstrated by several phase II and III clinical trials. The target population for immunotherapy in early-stage NSCLC is still under discussion, and the biomarkers for neoadjuvant immunotherapy population selection are the next pending problem. The predictive efficacy of many potential makers is still being explored, including PD-L1 expression levels, tumor mutation burden, circulating tumor DNA, components of the tumor microenvironment, and several clinical factors. We summarize key findings on the utility of ICIs in clinical trials of preoperative NSCLC patients and conclude analyses of relevant biomarkers to provide a better understanding of potentially predictive biomarkers in neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Xinyu Wu
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Yi Fung Chau
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Hua Bai
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Xiaofei Zhuang
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China,Department of Medical Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China,*Correspondence: Jianchun Duan,
| |
Collapse
|
48
|
Godoy LA, Chen J, Ma W, Lally J, Toomey KA, Rajappa P, Sheridan R, Mahajan S, Stollenwerk N, Phan CT, Cheng D, Knebel RJ, Li T. Emerging precision neoadjuvant systemic therapy for patients with resectable non-small cell lung cancer: current status and perspectives. Biomark Res 2023; 11:7. [PMID: 36650586 PMCID: PMC9847175 DOI: 10.1186/s40364-022-00444-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
Over the past decade, targeted therapy for oncogene-driven NSCLC and immune checkpoint inhibitors for non-oncogene-driven NSCLC, respectively, have greatly improved the survival and quality of life for patients with unresectable NSCLC. Increasingly, these biomarker-guided systemic therapies given before or after surgery have been used in patients with early-stage NSCLC. In March 2022, the US FDA granted the approval of neoadjuvant nivolumab and chemotherapy for patients with stage IB-IIIA NSCLC. Several phase II/III trials are evaluating the clinical efficacy of various neoadjuvant immune checkpoint inhibitor combinations for non-oncogene-driven NSCLC and neoadjuvant molecular targeted therapies for oncogene-driven NSCLC, respectively. However, clinical application of precision neoadjuvant treatment requires a paradigm shift in the biomarker testing and multidisciplinary collaboration at the diagnosis of early-stage NSCLC. In this comprehensive review, we summarize the current diagnosis and treatment landscape, recent advances, new challenges in biomarker testing and endpoint selections, practical considerations for a timely multidisciplinary collaboration at diagnosis, and perspectives in emerging neoadjuvant precision systemic therapy for patients with resectable, early-stage NSCLC. These biomarker-guided neoadjuvant therapies hold the promise to improve surgical and pathological outcomes, reduce systemic recurrences, guide postoperative therapy, and improve cure rates in patients with resectable NSCLC.
Collapse
Affiliation(s)
- Luis A Godoy
- Division of Thoracic Surgery, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Joy Chen
- Medical Student, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jag Lally
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Kyra A Toomey
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Prabhu Rajappa
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Roya Sheridan
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Shirish Mahajan
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Nicholas Stollenwerk
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Service, Pulmonology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Chinh T Phan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Service, Pulmonology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Danny Cheng
- Department of Radiology, Interventional Radiology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Robert J Knebel
- Department of Radiology, Interventional Radiology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA.
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA.
| |
Collapse
|
49
|
Hu M, Chong R, Liu W, Liu S, Liu X. Characteristic of molecular subtype based on lysosome-associated genes reveals clinical prognosis and immune infiltration of gastric cancer. Front Oncol 2023; 13:1155418. [PMID: 37197421 PMCID: PMC10183605 DOI: 10.3389/fonc.2023.1155418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
Background Lysosome are involved in nutrient sensing, cell signaling, cell death, immune responses and cell metabolism, which play an important role in the initiation and development of multiple tumors. However, the biological function of lysosome in gastric cancer (GC) has not been revealed. Here, we aim to screen lysosome-associated genes and established a corresponding prognostic risk signature for GC, then explore the role and underlying mechanisms. Methods The lysosome-associated genes (LYAGs) were obtained from MSigDB database. Differentially expressed lysosome-associated genes (DE-LYAGs) of GC were acquired based on the TCGA database and GEO database. According to expression profiles of DE-LYAGs, we divided the GC patients into different subgroups and then explored tumor microenvironment (TME) landscape and immunotherapy response in LYAG subtypes using GSVA, ESTIMATE and ssGSEA algorithms. Univariate Cox regression analysis, LASSO algorithm and multivariate Cox regression analysis were adopted to identify the prognostic LYAGs and then establish a risk model for patients with GC. The Kaplan-Meier analysis, Cox regression analysis and ROC analysis were utilized to evaluate the performance of the prognostic risk model. Clinical GC specimens were also used to verify the bioinformatics results by qRT-PCR assay. Results Thirteen DE-LYAGs were obtained and utilized to distinguish three subtypes in GC samples. Expression profiles of the 13 DE-LYAGs predicted prognosis, tumor-related immunological abnormalities and pathway dysregulation in these three subtypes. Furthermore, we constructed a prognostic risk model for GC based on DEG in the three subtypes. The Kaplan-Meier analysis suggested that higher risk score related to short OS rate. The Cox regression analysis and ROC analysis indicated that risk model had independent and excellent ability in predicting prognosis of GC patients. Mechanistically, a remarkable difference was observed in immune cell infiltration, immunotherapy response, somatic mutation landscape and drug sensitivity. qRT-PCR results showed that compared with corresponding adjacent normal tissues, most screened genes showed significant abnormal expressions and the expression change trends were consistent with the bioinformatics results. Conclusions We established a novel signature based on LYAGs which could be served as a prognostic biomarker for GC. Our study might provide new insights into individualized prognostication and precision treatment for GC.
Collapse
Affiliation(s)
- Maodong Hu
- Department of Gastroenterology, Huangdao District People’s Hospital, Qingdao, China
| | - Ruifeng Chong
- Department of General Surgery, Chengyang District People’s Hospital, Qingdao, China
| | - Weilin Liu
- General Surgery Department, Qingdao Hongdao People's Hospital, Chengyang District Center for Disease Control and Prevention, Qingdao, China
| | - Shuangyong Liu
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaolei Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Xiaolei Liu,
| |
Collapse
|
50
|
Wang H, Liu T, Chen J, Dang J. Neoadjuvant immunotherapy and neoadjuvant chemotherapy in resectable non-small cell lung cancer: A systematic review and single-arm meta-analysis. Front Oncol 2022; 12:901494. [PMID: 36212419 PMCID: PMC9533019 DOI: 10.3389/fonc.2022.901494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/02/2022] [Indexed: 12/25/2022] Open
Abstract
Background It remains uncertain whether neoadjuvant immune checkpoint inhibitor (nICI) is superior to neoadjuvant chemotherapy (nCT) in resectable non-small cell lung cancer. In addition, there are outstanding questions for nICI such as the ideal treatment mode and predictors. Methods PubMed, Embase, Cochrane Library, Web of Science, and scientific meetings were searched for eligible single-arm or multi-arm trials until 31 December 2021. The primary outcomes of interest were major pathological response (MPR) and pathological complete response (pCR). The random-effect model was used for statistical analysis. Results Twenty-four trials of nICI (n = 1,043) and 29 trials of nCT (n = 2,337) were identified. nICI combination therapy was associated with higher MPR (63.2%, 95% CI: 54.2%-72.1%) and pCR (35.3%, 95% CI: 27.4%-43.3%) rates compared to nCT (16.2%, 95% CI: 7.5%-25.0%, P < 0.001 and 5.5%, 95% CI: 3.5%-7.5%, P < 0.001) and nICI monotherapy (23.3%, 95% CI: 12.7%-33.8%, P < 0.001, and 6.5%, 95% CI: 1.7%-11.2%, P < 0.001). As for safety, nICI monotherapy had the best tolerability; nICI combination showed a similar surgical resection rate and higher R0 resection rate compared to nCT. PD-1 inhibitor and high PD-L1 expression (≥1% or ≥50%) were correlated with higher MPR and pCR rates compared to PD-L1 inhibitor and PD-L1 expression <1%. Conclusions nICI combination therapy is associated with higher MPR and pCR rates compared to nCT and nICI monotherapy. PD-1 inhibitor seems to be superior to PD-L1 inhibitor. PD-L1 status appears to be predictive of MPR and pCR for patients receiving nICI. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=278661, CRD42021278661.
Collapse
Affiliation(s)
- He Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Tingting Liu
- Department of Radiation Oncology, Anshan Cancer Hospital, Anshan, China
| | - Jun Chen
- Department of Radiation Oncology, Shenyang Tenth People’s Hospital, Shenyang, China
| | - Jun Dang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|