51
|
Wang X, Xu Y, Xu J, Chen Y, Song C, Jiang G, Chen R, Mao W, Zheng M, Wan Y. Establishment and validation of nomograms for predicting survival of lung invasive adenocarcinoma based on the level of pathological differentiation: a SEER cohort-based analysis. Transl Cancer Res 2023; 12:804-827. [PMID: 37180650 PMCID: PMC10174764 DOI: 10.21037/tcr-22-2308] [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: 09/28/2022] [Accepted: 03/10/2023] [Indexed: 05/16/2023] [Imported: 02/28/2025]
Abstract
BACKGROUND The pathological differentiation of invasive adenocarcinoma (IAC) has been linked closely with epidemiological characteristics and clinical prognosis. However, the current models cannot accurately predict IAC outcomes and the role of pathological differentiation is confused. This study aimed to establish differentiation-specific nomograms to explore the effect of IAC pathological differentiation on overall survival (OS) and cancer-specific survival (CSS). METHODS The data of eligible IAC patients between 1975 and 2019 were collected from the Surveillance, Epidemiology, and End Results (SEER) database, and randomly divided in a ratio of 7:3 into a training cohort and a validation cohort. The associations between pathological differentiation and other clinical characteristics were evaluated using chi-squared test. The OS and CSS analyses were performed using the Kaplan-Meier estimator, and the log-rank test was used for nonparametric group comparisons. Multivariate survival analysis was performed using a Cox proportional hazards regression model. The discrimination, calibration, and clinical performance of nomograms were assessed by area under receiver operating characteristic curve (AUC), calibration plots, and decision curve analysis (DCA). RESULTS A total of 4,418 IAC patients (1,001 high-differentiation, 1,866 moderate-differentiation, and 1,551 low-differentiation) were identified. Seven risk factors [age, sex, race, tumor-node-metastasis (TNM) stage, tumor size, marital status, and surgery] were screened to construct differentiation-specific nomograms. Subgroup analyses showed that disparate pathological differentiation played distinct roles in prognosis, especially in patients with older age, white race, and higher TNM stage. The AUC of nomograms for OS and CSS in the training cohort were 0.817 and 0.835, while in the validation cohort were 0.784 and 0.813. The calibration curves showed good conformity between the prediction of the nomograms and the actual observations. DCA results indicated that these nomogram models could be used as a supplement to the prediction of the TNM stage. CONCLUSIONS Pathological differentiation should be considered as an independent risk factor for OS and CSS of IAC. Differentiation-specific nomogram models with good discrimination and calibration capacity were developed in the study to predict the OS and CSS in 1-, 3- and 5-year, which could be used predict prognosis and select appropriate treatment options.
Collapse
|
research-article |
2 |
|
52
|
Pi Z, Liu W, Song C, Zhu C, Liu J, Wang L, He Z, Yang C, Wu L, Liu T, Geng Z, Tebbutt SJ, Liu N, Wan Y, Zhang F, Mao W. Multi-level insights into the immuno-oncology-microbiome axis: From biotechnology to novel therapies. IMETA 2024; 3:e240. [PMID: 39429874 PMCID: PMC11487608 DOI: 10.1002/imt2.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 10/22/2024] [Imported: 02/28/2025]
Abstract
The multifaceted interactions among the immune system, cancer cells and microbial components have established a novel concept of the immuno-oncology-microbiome (IOM) axis. Microbiome sequencing technologies have played a pivotal role in not only analyzing how gut microbiota affect local and distant tumors, but also providing unprecedented insights into the intratumor host-microbe interactions. Herein, we discuss the emerging trends of transiting from bulk-level to single cell- and spatial-level analyses. Moving forward with advances in biotechnology, microbial therapies, including microbiota-based therapies and bioengineering-inspired microbes, will add diversity to the current oncotherapy paradigm.
Collapse
|
letter |
1 |
|
53
|
ZHENG H, YAN D, WANG P, YIN Y, ZHANG Q, ZHANG L, LIANG H, SHI D, HU L, MAO W, ZHANG J, LI Y. Survival with lung transplantation and extracorporeal membrane oxygenation: a systematic review and meta-analysis. MINERVA RESPIRATORY MEDICINE 2022; 61. [DOI: 10.23736/s2784-8477.21.01980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025] [Imported: 02/28/2025]
|
|
3 |
|
54
|
Mao WJ, Xia W, Chen JY. [Bilateral lung transplantation for pulmonary destruction after concentrated sulfuric acid inhalation: report of one case]. ZHONGHUA LAO DONG WEI SHENG ZHI YE BING ZA ZHI = ZHONGHUA LAODONG WEISHENG ZHIYEBING ZAZHI = CHINESE JOURNAL OF INDUSTRIAL HYGIENE AND OCCUPATIONAL DISEASES 2012; 30:312-313. [PMID: 22804948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] [Imported: 02/28/2025]
|
Case Reports |
13 |
|
55
|
Jiang G, Liu W, Wang X, Wang Z, Song C, Chen R, He Z, Li H, Zheng M, Mao W. The causality between systemic inflammatory regulators and chronic respiratory diseases: A bidirectional Mendelian-randomization study. Cytokine 2024; 174:156470. [PMID: 38071841 DOI: 10.1016/j.cyto.2023.156470] [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: 09/09/2023] [Revised: 10/29/2023] [Accepted: 12/04/2023] [Indexed: 01/01/2024] [Imported: 02/28/2025]
Abstract
INTRODUCTION Accumulative evidence suggests the associations between systemic inflammatory regulators and chronic respiratory diseases (CRDs). However, the intrinsic causation remains implicit. Therefore, this study aimed to examine causative associations by mendelian randomization (MR) and to identify valuable active factors. METHODS Based on data from the GWAS database, we performed MR analyses of 41 serum cytokines from 8,293 Finnish and European descent cohorts from GBMI and UKBB for five major CRDs. We mainly applied inverse variance weighted regression, supplemented by MR-Egger regression, weighted median, maximum likelihood, weighted mode, and simple mode algorithms. Moreover, sensitivity analyses were conducted using Cochrane's Q test, MR-Egger intercept, MR-PRESSO Global test and MR-Steiger filtering. Eventually, the consistency of MR results was assessed by leave-one-out. RESULTS Our results suggest that 12 genetically predicted systemic inflammatory regulators probably participate in the progression of CRDs, including four risk factors (IL-1RA, IL-4, MIP-1A, PDGF-BB) and one protective factor (IL-6) in IPF, two protective factors (SCF, SDF-1A) in COPD, and two protective factors (SCF, SDF-1A) in asthma, two protective factors (GROA, IL-2RA) were also included in asthma, whereas only one factor (HGF) was protective against bronchiectasis. Additionally, two protective factors (FGF-BASIC, G-CSF) were identified in sarcoidosis. Sensitivity analyses showed no horizontal pleiotropy and significant heterogeneity. Finally, based on the findings of inverse MR analysis, no inverse causal association was uncovered, confirming the robustness of results. CONCLUSION Our study unearths potential associations between systemic inflammatory modulators and common CRDs, providing new insights for inflammation-mediated CRD prevention and therapeutic approaches.
Collapse
|
|
1 |
|
56
|
Mao WJ, Chen JY, Zheng MF, Chen R, He YJ, Liu F, Ye SG, Lu RG. [Lung transplantation for phase Ⅲ silicosis: a series of 32 cases]. ZHONGHUA WAI KE ZA ZHI [CHINESE JOURNAL OF SURGERY] 2016; 54:902-907. [PMID: 27916032 DOI: 10.3760/cma.j.issn.0529-5815.2016.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] [Imported: 02/28/2025]
Abstract
Objective: To evaluate the effect of lung transplantation for phase Ⅲ silicosis. Methods: From September 2002 to September 2015, 32 patients with end-stage silicosis underwent lung transplantation at Department of Thoracic Surgery, Affiliated Wuxi People's Hospital, Nanjing Medical University. There were 29 male and 3 female patients aged from 24 to 63 years. Thirty-two patients were diagnosed as phase Ⅲ silicosis by the local occupational disease prevention and control center. Fifteen patients were type Ⅰ respiratory failure and 17 patients were type Ⅱ. There were 14 cases accepted bilateral sequential lung transplantation and 18 cases accepted single lung transplantation, including 13 cases with right single lung transplantation and 5 cases with left single lung transplantation. Extracorporeal membrane oxygenation was used in 13 patients. Pulmonary function monitoring was performed at 3 months, 6 months, 1 year, and 2 years after lung transplantation. Clinical characteristics were compared using t-test, χ2 test and Fisher exact test between groups, Kaplan-Meier survival curve and Log-rank test were used to find out the factors affecting survival. Results: All the patients received lung transplantation successfully. One patient died of multiple organ failure, 1 died of sepsis, and 1 succumbed to sudden cardiac death. Twenty-nine patients were discharged from hospital. During follow-up, there were 5 deaths, two patients died of sepsis 7 months postoperatively, 1 died of renal failure 5 months post-transplant, 1 died of sudden cardiac death, and the remaining 1 patient died of bronchiolitis obliterans. Twenty-four patients lived a good quality of life, with survival rates of 90.6% at 3 months, 80.8% at 1 year, 76.7% at 3 years, and 76.7% at 5 years. Significant difference was not observed between single and bilateral lung transplantation about long-term survival rate. During follow-up pulmonary function post-transplant (3 months, 6 months, 1 year, and 2 years) were improved dramatically compared with preoperative level, and patients lived a good quality of life. Conclusion: Lung transplantation is beneficial for patients with phase Ⅲ silicosis, long-term survival is probable.
Collapse
|
|
9 |
|
57
|
Song C, Liu W, Jiang G, He Z, Wang R, Wang X, Chen R, Mao W, Zhu S. Identification and validation of a novel NK cells-related signature to predict prognosis and immune microenvironment in LUAD. Immunobiology 2023; 228:152751. [PMID: 37774597 DOI: 10.1016/j.imbio.2023.152751] [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: 08/13/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] [Imported: 02/28/2025]
Abstract
BACKGROUND The prevalence and fatality rates of lung cancer are experiencing a rapid escalation. Natural Killer (NK) cells have been established to have a crucial role in both tumor initiation and progression. Nevertheless, uncertainties persist regarding their precise implications in the prognosis of LUAD. METHODS The data were obtained from reputable sources, such as the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) database, and our internally generated sequencing data. Utilizing the TCGA data as a background, we selected intersecting genes, validated by cluster analysis, to establish a Cox model and validated it using the GEO datasets. Furthermore, we conducted extensive analyses to investigate the significance of potential biomarkers in relation to immune cell infiltration, single-cell data, differential gene expression, and drug sensitivity. RESULTS 67 immune-related genes associated with NK cells (NK-IRGs) were identified in the TCGA datasets, whose research potential was demonstrated by cluster analysis. A prognostic signature was identified utilizing the univariate and multivariate Cox model, resulting in the identification of five genes, which was validated using GEO datasets. Additionally, the nomogram's calibration curve demonstrated exceptional concordance between the projected and actual survival rates. Subsequent investigations uncovered that this prognostic signature demonstrated its independence as a risk factor. Notably, in the low-risk group, NK cells exhibited elevated levels of immune checkpoint molecules, indicating heightened sensitivity to immune therapy. These findings highlight the potential of utilizing this signature as a valuable tool in the selection of patients who could benefit from targeted immune interventions.
Collapse
|
|
2 |
|
58
|
Chen Y, Xu Y, Wang J, Prisinzano P, Yuan Y, Lu F, Zheng M, Mao W, Wan Y. Statins Lower Lipid Synthesis But Promote Secretion of Cholesterol-Enriched Extracellular Vesicles and Particles. Front Oncol 2022; 12:853063. [PMID: 35646709 PMCID: PMC9133486 DOI: 10.3389/fonc.2022.853063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] [Imported: 02/28/2025] Open
Abstract
Lipid droplets are lipid-rich cytosolic organelles that play roles in cell signaling, membrane trafficking, and many other cellular activities. Recent studies revealed that lipid droplets in cancer cells have various biological functions, such as energy production, membrane synthesis, and chemoresistance, thereby fostering cancer progression. Accordingly, the administration of antilipemic agents could improve anti-cancer treatment efficacy given hydrophobic chemotherapeutic drugs could be encapsulated into lipid droplets and then expelled to extracellular space. In this study, we investigated whether statins could promote treatment efficacy of lipid droplet-rich ovarian SKOV-3 cells and the potential influences on generation and composition of cell-derived extracellular vesicles and particles (EVP). Our studies indicate that statins can significantly lower lipid biosynthesis. Moreover, statins can inhibit proliferation, migration, and invasion of SKOV-3 cells and enhance chemosensitivity in vitro and in vivo. Furthermore, statins can lower EVP secretion but enforce the release of cholesterol-enriched EVPs, which can further lower lipid contents in parental cells. It is the first time that the influence of statins on EVP generation and EVP-lipid composition is observed. Overall, we demonstrated that statins could inhibit lipid production, expel cholesterol to extracellular space via EVPs, and improve chemosensitivity.
Collapse
|
research-article |
3 |
|
59
|
Mao WJ, Nie XW, Xia W, Zheng MF, Chen R, He YJ, Liu F, Ye SG, Lu RG, Chen JY. [Influencing factors for postoperative survival of patients with stage III silicosis treated by lung transplantation]. ZHONGHUA LAO DONG WEI SHENG ZHI YE BING ZA ZHI = ZHONGHUA LAODONG WEISHENG ZHIYEBING ZAZHI = CHINESE JOURNAL OF INDUSTRIAL HYGIENE AND OCCUPATIONAL DISEASES 2016; 34:659-664. [PMID: 27866541 DOI: 10.3760/cma.j.issn.1001-9391.2016.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] [Imported: 02/28/2025]
Abstract
Objective: To investigate the clinical effect, postoperative complications, and causes of death in the treatment of stage III silicosis with lung transplantation and the influencing factors for survival. Methods: A retrospective analysis was performed for the clinical data of 32 patients with stage III silicosis who underwent lung transplantation in our hospital from September 2002 to September 2015. The survival, causes of death, and postoperative complications were analyzed. The Kaplan-Meier method was used to plot survival curves, the log-rank test was used to compare the influence of each factor on survival rates, and the multivariate Cox proportional hazards regression model was used to evaluate the influence of each factor on survival. Results: All the patients underwent successful lung transplantation. The 3-month and 1-, 3-, and 5-year postoperative cumulative survival rates were 90.6%, 80.8%, 76.7%, and 76.7%, respectively. Eight patients died during the postoperative follow-up, among whom 1 died of multiple organ failure, 3 died of severe infection, 2 died of sudden cardiac death, 1 died of renal failure, and 1 died of bronchiolitis obliterans. The major postoperative complications included primary graft dysfunction (PGD) in 10 patients, severe infection in 7 patients, acute rejection reaction in 3 patients, bronchiolitis obliterans in 5 patients, bleeding in 5 patients, anastomotic complication in 2 patients, and renal dysfunction in 3 patients. The Kaplan-Meier survival analysis showed that sex, postoperative PGD, postoperative infection, massive intraoperative blood loss, preoperative pulmonary arterial hypertension were influencing factors for postoperative survival rates (P<0.05). The multivariate Cox regression model showed that male sex was the protective factor (P<0.05) and postoperative PGD and massive intraoperative blood loss were independent risk factors for death after transplantation (P<0.05). Conclusion: Lung transplantation is a method for the treatment of silicosis. Postoperative PGD and massive intraoperative blood loss are independent risk factors for death after transplantation. Survival rates are affected by postoperative PGD, infection, massive intraoperative blood loss, and preoperative pulmonary arterial hypertension.
Collapse
|
|
9 |
|
60
|
Jiang G, Wang X, Xu Y, He Z, Lu R, Song C, Jin Y, Li H, Wang S, Zheng M, Mao W. The diagnostic potential role of thioredoxin reductase and TXNRD1 in early lung adenocarcinoma: A cohort study. Heliyon 2024; 10:e31864. [PMID: 38882339 PMCID: PMC11177154 DOI: 10.1016/j.heliyon.2024.e31864] [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: 09/21/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] [Imported: 02/28/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the primary form of lung cancer, yet the reliable biomarkers for early diagnosis remain insufficient. Thioredoxin reductase (TrxR) is strongly linked to the occurrence, development, and drug resistance of lung cancer, making it a potential biomarker. However, further research is required to assess its diagnostic value in LUAD. METHODS A retrospective analysis was performed on patients who underwent pulmonary nodule resection at our center from 2018 to 2022. Clinical data, including preoperative TrxR levels, imaging, and laboratory characteristics, were identified as study variables. Two prediction models were constructed using multiple logistic regression, and their prediction performance was evaluated comprehensively. Besides, bioinformatics analyses of TrxR coding genes including differential expression, functional enrichment, immune infiltration, drug sensitivity, and single-cell landscape were performed based on TCGA database, which were subsequently validated by Human Protein Atlas. RESULTS A total of 506 eligible patients (72 benign lesions, 77 AISs, 185 MIAs and 172 IACs) were identified in the clinical cohort. Two TrxR-based models were developed, which were able to distinguish between benign and malignant pulmonary nodules, as well as pathological subtypes of LUAD, respectively. The models exhibited good predictive ability with all AUC values ranging from 0.7 to 0.9. Based on calibration curves and clinical decision analysis, the nomogram models showed high reliability. Functional analysis indicated that TXNRD1 primarily participated in cell cycle and lipid metabolism. Immune infiltration analysis showed that TXNRD1 has a strong association with immune cells and could impact immunotherapy. Then, we identified small molecular compounds that inhibit TXNRD1 and confirmed TXNRD1 expression by single-cell landscape and immunohistochemistry. CONCLUSION This study validated the diagnostic value of TrxR and TXNRD1 in clinical cohorts and transcriptional data, respectively. TrxR and TXNRD1 could be used in the risk diagnosis of early LUAD and facilitate personalized treatment strategies.
Collapse
|
research-article |
1 |
|
61
|
Xia W, Xu H, Mao W, Chen J. [Extracorporeal membrane oxygenation as a bridge to lung transplantation]. ZHONGHUA WEI ZHONG BING JI JIU YI XUE 2018; 30:1167-1172. [PMID: 30592952 DOI: 10.3760/cma.j.issn.2095-4352.2018.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/28/2025] [Imported: 02/28/2025]
Abstract
OBJECTIVE To evaluate the effect of extracorporeal membrane oxygenation (ECMO) as a bridge to lung transplantation (LTx). METHODS The clinical data of 18 patients with end-stage lung diseases was retrospectively reviewed, using ECMO as a bridge to LTx in intensive care unit of Affiliated Wuxi People's Hospital from January 2015 to December 2017. Clinical parameters were obtained from these patients, including gender, age, primary disease, preoperative lactate level, preoperative leukocyte, operation modality (unilateral or bilateral), type of ECMO, ECMO support time as a bridge to LTx, ECMO support time after operation, total usage time of ECMO, ECMO associated complications, primary graft dysfunction (PGD), successful ECMO weaning, and survival. Patients were divided according to type of ECMO, whether successfully weaned from ECMO or not, and primary disease. Clinical data was compared, and the Kaplan-Meier survival of 180-day was studied. RESULTS (1) The overall situation showed: A total of 18 patients were enrolled, with 14 males and 4 females, age ranged from 23 to 78 years old. Primary disease included 6 cases of idiopathic pulmonary fibrosis (IPF), 3 cases of idiopathic pulmonary hypertension (IPAH), 8 cases of interstitial pneumonia and 1 case of silicosis. Nine patients received venous-venous (V-V) ECMO and 9 venous-artery (V-A) ECMO as a bridge to LTx; 15 patients received LTx successfully, and failed in 3 cases. The average bridge time was 57.5 (14.5, 116.5) hours. ECMO associated complications included 6 cases with bleeding, 12 cases with renal failure, 2 cases with thrombosis, 2 cases with oxygenator leak, and 1 case with leg ischemia. There were 7 unilateral (5 right lungs and 2 left lungs) and 8 bilateral LTx. Three patients died before LTx due to septic shock. Nine patients died after LTx, 4 for septic shock, 4 for multiple organ failure, and 1 for sudden cardiac death. Six patients survived after LTx. (2) Group comparison showed: There was no significant difference in gender, age, ECMO support time as a bridge to LTx, ECMO support time after operation, total ECMO usage time, incidence of PGD, successful weaning from ECMO, and 180-day survival rate between V-V ECMO group (n = 7) and V-A ECMO group (n = 8). There was no significant difference in gender, age, primary disease, type of ECMO, operation modality, preoperative leukocyte count between groups of successfully weaned from ECMO (n = 11) and the failed (n = 7). Lower level of preoperative lactate acid (mmol/L: 3.01±1.51 vs. 8.27±3.49, t = -3.770, P = 0.006), shorter total ECMO usage time (hours: 72.82±40.53 vs. 210.71±107.10, t = -3.907, P = 0.001), and higher 180-day survival rate (54.5% vs. 0, P = 0.038) were found in the group of successfully weaned from ECMO, when compared with the failed group. (3) Kaplan-Meier survival analysis showed that postoperative survival rates of 7, 30, 60, and 180 days of 18 patients was 72.2%, 38.9%, 33.3%, and 33.3%, respectively. Among them, the postoperative survival rates of 7, 30, 60, and 180 days in the group of successfully weaned from ECMO (n = 11) were higher than those in group of failed (n = 7; 81.8% vs. 57.1%, 63.6 % vs. 0, 54.5% vs. 0, 54.5% vs. 0, respectively; log-rank test: χ2 = 8.009, P = 0.005). The postoperative survival rates of 7, 30, 60, and 180 days in IPF group (n = 6) were lower than those in non-IPF group (n = 12; 33.3% vs. 83.3%, 16.7% vs. 50.0%, 16.7% vs. 41.7%, 16.7% vs. 41.7%; log-rank test: χ2 = 4.161, P = 0.041). CONCLUSIONS The use of ECMO as a bridge to LTx may provide survival benefit for LTx recipients. V-V ECMO provides effective life support for patients without severe heart failure, and V-A ECMO for patients with unstable hemodynamics. Preoperative lactate level and total ECMO duration time were closely related to ECMO weaning rate. Primary diagnosis may affect prognosis.
Collapse
|
|
7 |
|
62
|
Wang Y, Hou H, Luo H, Xun J, Ma C, Yang H, Bai D, Yousuf S, Lyu H, Zhang T, Wan X, Yao X, Ma T, Zhou Y, Zhu Z, Zeng M, An S, Bai Q, Bai Y, Cao G, Cao T, Cao Y, Chang C, Chang L, Chen B, Chen D, Chen D, Chen H, Chen J, Chen J, Chen W, Chen X, Chen Y, Chen Z, Cheng C, Cheng Q, Dai X, Deng C, Deng F, Deng J, Dong C, Dong L, Duan L, Duan Y, Fan Q, Fang C, Fang T, Fang W, Fang Z, Fu M, Fu M, Gao C, Gao H, Gao W, Gao X, Gao Y, Geng Y, Gong W, Gu S, Gu X, Gu Z, Guo J, Guo J, Guo Q, Guo X, Guo X, Han D, Han Z, Hao Y, He J, He J, He J, He R, Hou G, Hu B, Hu H, Hu Y, Hu Y, Hu Y, Huang G, Huang H, Huang J, Huang S, Jia B, Jian X, Jiang C, Jiang K, Jiang L, Jiang S, Jiao J, Jin H, Jin J, Kong S, Lai X, Leng Y, Li B, Li B, Li F, Li H, Li H, et alWang Y, Hou H, Luo H, Xun J, Ma C, Yang H, Bai D, Yousuf S, Lyu H, Zhang T, Wan X, Yao X, Ma T, Zhou Y, Zhu Z, Zeng M, An S, Bai Q, Bai Y, Cao G, Cao T, Cao Y, Chang C, Chang L, Chen B, Chen D, Chen D, Chen H, Chen J, Chen J, Chen W, Chen X, Chen Y, Chen Z, Cheng C, Cheng Q, Dai X, Deng C, Deng F, Deng J, Dong C, Dong L, Duan L, Duan Y, Fan Q, Fang C, Fang T, Fang W, Fang Z, Fu M, Fu M, Gao C, Gao H, Gao W, Gao X, Gao Y, Geng Y, Gong W, Gu S, Gu X, Gu Z, Guo J, Guo J, Guo Q, Guo X, Guo X, Han D, Han Z, Hao Y, He J, He J, He J, He R, Hou G, Hu B, Hu H, Hu Y, Hu Y, Hu Y, Huang G, Huang H, Huang J, Huang S, Jia B, Jian X, Jiang C, Jiang K, Jiang L, Jiang S, Jiao J, Jin H, Jin J, Kong S, Lai X, Leng Y, Li B, Li B, Li F, Li H, Li H, Li J, Li K, Li L, Li L, Li M, Li P, Li W, Li W, Li X, Xuemeng L, Li Y, Li Y, Li Z, Liang L, Liang R, Liang Z, Liu Q, Liu D, Liu H, Liu J, Liu L, Liu L, Liu M, Liu R, Liu S, Liu T, Liu W, Liu W, Liu X, Liu Y, Liu Y, Liu Y, Liu Y, Liu Z, Liu Z, Liu Z, Long C, Long Y, Lu C, Lu C, Lu C, Lu Q, Luan Y, Luo P, Luo S, Ma N, Ma X, Ma Y, Mao W, Meng Y, Ni Y, Ni Y, Ning K, Niu D, Peng K, Peng Z, Qian X, Qiu Z, Qu H, Qu Z, Ren Y, Ren Z, Shen Y, Shi L, Shi L, Shi W, Shi Y, Song T, Song X, Song X, Song X, Su Q, Su Y, Sun L, Sun Q, Sun T, Sun Y, Tang H, Tang W, Yu T, Tian S, Wang S, Wang B, Wang C, Jin W, Wang L, Wang L, Wang L, Wang M, Wang M, Wang P, Wang S, Wang S, Wang X, Wang X, Wei M, Wei Y, Wei Y, Wei Y, Wen C, Wen X, Wu L, Wu S, Wu Y, Xia S, Xia X, Xia Y, Xiang X, Xiao C, Xiao W, Xiao Y, Xie R, Xing R, Xu H, Xu W, Xu Z, Xue H, Yan C, Yan Q, Yan S, Yan X, Yang M, Yang Y, Yang Z, Yang Z, Yao G, Yao Y, Yi X, Yin C, Yin M, Yu S, Yu Y, Yu Y, Yuan F, Zhai S, Zhang B, Zhang C, Zhang F, Zhang F, Zhang H, Zhang J, Zhang J, Zhang K, Zhang L, Zhang L, Zhang L, Zhang M, Zhang Q, Zhang R, Zhang T, Zhang T, Zhang W, Zhang Y, Zhang Y, Zhang Y, Zhang Z, Zhang Z, Zhang Z, Zhao B, Zhao Y, Zhao Y, Zhao Z, Zheng D, Zheng Y, Zhi W, Zhong J, Zhong X, Zhou W, Zhou X, Zhou Z, Zhou Z, Zhu C, Zhu F, Zhu X, Zou Y, Zhou H, Lei L, Bi Y, Shi H, Sun H, Jin S, Ren W, Dai L, Wang X, Lan C, Liu H, Liu S, Yin Y, Shi C, Gan R, Zhao F, Yu J, Chen T, Hong X, Yang H, Zhang B, Chen S, Li X, Gao Y, Liu Y. iMeta Conference 2024: Building an innovative scientific research ecosystem for microbiome and One Health. IMETA 2024; 3:e251. [PMID: 39742310 PMCID: PMC11683455 DOI: 10.1002/imt2.251] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] [Imported: 02/28/2025]
Abstract
The iMeta Conference 2024 provides a platform to promote the development of an innovative scientific research ecosystem for microbiome and One Health. The four key components - Technology, Research (Biology), Academic journals, and Social media - form a synergistic ecosystem. Advanced technologies drive biological research, which generates novel insights that are disseminated through academic journals. Social media plays a crucial role in engaging the public and facilitating scientific communication, thus amplifying the impact of research. Together, these elements create a self-sustaining loop that fosters continuous innovation and collaboration in the field of bioinformatics, biotechnology and microbiome research.
Collapse
|
Editorial |
1 |
|
63
|
Xie X, Liu W, Yuan Z, Chen H, Mao W. Bridging epigenomics and tumor immunometabolism: molecular mechanisms and therapeutic implications. Mol Cancer 2025; 24:71. [PMID: 40057791 PMCID: PMC11889836 DOI: 10.1186/s12943-025-02269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] [Imported: 04/02/2025] Open
Abstract
Epigenomic modifications-such as DNA methylation, histone acetylation, and histone methylation-and their implications in tumorigenesis, progression, and treatment have emerged as a pivotal field in cancer research. Tumors undergo metabolic reprogramming to sustain proliferation and metastasis in nutrient-deficient conditions, while suppressing anti-tumor immunity in the tumor microenvironment (TME). Concurrently, immune cells within the immunosuppressive TME undergo metabolic adaptations, leading to alterations in their immune function. The complicated interplay between metabolites and epigenomic modulation has spotlighted the significance of epigenomic regulation in tumor immunometabolism. In this review, characteristics of the epigenomic modification associated with tumors are systematically summarized alongside with their regulatory roles in tumor metabolic reprogramming and immunometabolism. Classical and emerging approaches are delineated to broaden the boundaries of research on the crosstalk research on the crosstalk between tumor immunometabolism and epigenomics. Furthermore, we discuss potential therapeutic strategies that target tumor immunometabolism to modulate epigenomic modifications, highlighting the burgeoning synergy between metabolic therapies and immunotherapy as a promising avenue for cancer treatment.
Collapse
|
Review |
1 |
|
64
|
Liu W, Pi Z, Mao W. The trio of circadian clock, intestinal flora, and cancer. Trends Mol Med 2025; 31:4-6. [PMID: 39304425 DOI: 10.1016/j.molmed.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] [Imported: 02/28/2025]
Abstract
Dysbiotic intestinal flora and a disrupted circadian clock are intimately related to cancer biological behaviors, yet their interwoven regulatory mechanisms remain an explorative field. Studies by Ren et al. and Liu et al. provide deeper insights into the potential roles of intestinal flora and the circadian clock in colorectal tumorigenesis and lung metastasis.
Collapse
|
|
1 |
|
65
|
He Z, Wang R, Song C, Liu J, Chen R, Zheng M, Liu W, Jiang G, Mao W. Exploring the causal relationship between immune cells and idiopathic pulmonary fibrosis: a bi-directional Mendelian randomization study. BMC Pulm Med 2024; 24:145. [PMID: 38509507 PMCID: PMC10956372 DOI: 10.1186/s12890-024-02942-w] [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: 11/16/2023] [Accepted: 03/01/2024] [Indexed: 03/22/2024] [Imported: 02/28/2025] Open
Abstract
BACKGROUND The potential pathogenic mechanism of idiopathic pulmonary fibrosis is widely recognized to involve immune dysregulation. However, the current pool of studies has yet to establish a unanimous agreement regarding the correlation between various types of immune cells and IPF. METHODS By conducting a two-sample Mendelian randomization analysis using publicly available genetic data, the study examined the causal relationship between IPF and 731 immune cells. To ensure the reliability of the results, combined sensitivity analyses and inverse Mendelian analyses were conducted. Moreover, within subgroups, multivariate Mendelian randomization analyses were utilized to investigate the autonomous causal connection between immune cell characteristics and IPF. RESULTS After adjusting for false discovery rate, it was discovered that 20 immunophenotypes exhibited a significant association with IPF. After subgrouping for multivariate Mendelian randomization analysis, there were six immunophenotypes that remained significantly associated with IPF. These included CD33 + HLA DR + CD14dim (OR = 0.96, 95% CI 0.93-0.99, P = 0.033), HLA DR + NK (OR = 0.92, 95% CI 0.85-0.98, P = 0.017), CD39 + CD8 + T cell %T cell (OR = 0.93, 95% CI 0.88-0.99, P = 0.024), CD3 on activated & secreting Treg (OR = 0.91, 95% CI 0.84-0.98, P = 0.026), PDL-1 on CD14- CD16 + monocyte (OR = 0.89, 95% CI 0.84-0.95, P = 8 × 10-4), and CD45 on CD33 + HLA DR + CD14- (OR = 1.08, 95% CI 1.01-1.15, P = 0.011). CONCLUSION Our study reveals a noteworthy association between IPF and various immune cells, providing valuable insights for clinical research and aiding the advancement of immunologically-based therapeutic strategies.
Collapse
|
research-article |
1 |
|
66
|
Liu W, Pi Z, Wang X, Shang C, Song C, Wang R, He Z, Zhang X, Wan Y, Mao W. Microbiome and lung cancer: carcinogenic mechanisms, early cancer diagnosis, and promising microbial therapies. Crit Rev Oncol Hematol 2024; 196:104322. [PMID: 38460928 DOI: 10.1016/j.critrevonc.2024.104322] [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: 09/26/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] [Imported: 02/28/2025] Open
Abstract
Microbiomes in the lung, gut, and oral cavity are correlated with lung cancer initiation and progression. While correlations have been preliminarily established in earlier studies, delving into microbe-mediated carcinogenic mechanisms will extend our understanding from correlation to causation. Building upon the causative relationships between microbiome and lung cancer, a novel concept of microbial biomarkers has emerged, mainly encompassing cancer-specific bacteria and circulating microbiome DNA. They might function as noninvasive liquid biopsy techniques for lung cancer early detection. Furthermore, potential microbial therapies have displayed initial efficacy in lung cancer treatment, providing multiple avenues for therapeutic intervention. Herein, we will discuss the molecular mechanisms and signaling pathways through which microbes influence lung cancer initiation and development. Additionally, we will summarize recent findings on microbial biomarkers as a member of tumor liquid biopsy techniques and provide an overview of the latest advances in various microbe-assisted/mediated therapeutic approaches for lung cancer.
Collapse
|
Review |
1 |
|
67
|
Shi J, Mao W, Song Y, Wang Y, Zhang L, Xu Y, Gu H, Yao S, Yao Y, Liu Z, Raghavan V, Wang J. Butyrate alleviates food allergy by improving intestinal barrier integrity through suppressing oxidative stress‐mediated Notch signaling. IMETA 2025. [DOI: 10.1002/imt2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/14/2025] [Indexed: 05/04/2025] [Imported: 05/04/2025]
Abstract
AbstractFood allergy (FA) has received increased attention in recent years. Multiple studies have highlighted the crucial role of short‐chain fatty acids (SCFAs) in the development of IgE‐mediated FA. Here, a case‐control approach was employed to analyze SCFAs profiles in children with FA, while an ovalbumin (OVA)‐sensitized mouse model was utilized to explore the underlying mechanism by which SCFAs mitigate FA. Children with food‐sensitized tolerance (FST) (n = 20) or FA (n = 20), and healthy controls (HC) (n = 20) were recruited to analyze SCFAs profiles. The HC group exhibited higher SCFAs levels in fecal samples than the FST, FA, and FST + FA groups. Data from an OVA‐sensitized mouse model showed that butyrate exhibited a more significant effect on reducing allergic reactions compared to other SCFAs. Compared to the negative control group, OVA‐induced oxidative stress (OS) triggered excessive Notch signaling activation, which subsequently impaired both tight junctions integrity and mucosal barrier function in murine intestinal epithelial cells (IECs). Gut dysbiosis induced mucus layer erosion, thereby elevating IECs exposure to food antigens and OS, which potentiated Notch signaling activation. However, butyrate counteracted this loop by restoring microbiota structure and suppressing reactive oxygen species (ROS)/Notch cascades. Strikingly, low‐dose butyrate (0.25–1 mM) protected rat small intestine crypt epithelial cells (IEC‐6) by inhibiting ROS, whereas high‐dose (2–5 mM) exacerbated oxidative injury and triggered activation of Notch signaling. Our study revealed the potential molecular mechanisms through which butyrate alleviates food allergy, providing a potential therapeutic strategy for its management.
Collapse
|
|
1 |
|
68
|
Wang K, Li Z, Xuan Y, Zhao Y, Deng C, Wang M, Xie C, Yuan F, Pang Q, Mao W, Cai D, Zhong Z, Mei J. Pan-cancer analysis of NFE2L2 mutations identifies a subset of lung cancers with distinct genomic and improved immunotherapy outcomes. Cancer Cell Int 2023; 23:229. [PMID: 37794491 PMCID: PMC10552358 DOI: 10.1186/s12935-023-03056-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] [Imported: 02/28/2025] Open
Abstract
BACKGROUND Mutations in the KEAP1-NFE2L2 signaling pathway were linked to increased tumorigenesis and aggressiveness. Interestingly, not all hotspot mutations on NFE2L2 were damaging; some even were activating. However, there was conflicting evidence about the association between NFE2L2 mutation and Nrf2-activating mutation and responsiveness to immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) and other multiple cancers. METHODS The study with the largest sample size (n = 49,533) explored the landscape of NFE2L2 mutations and their impact response/resistance to ICIs using public cohorts. In addition, the in-house WXPH cohort was used to validate the efficacy of immunotherapy in the NFE2L2 mutated patients with NSCLC. RESULTS In two pan-cancer cohorts, Nrf2-activating mutation was associated with higher TMB value compared to wild-type. We identified a significant association between Nrf2-activating mutation and shorter overall survival in pan-cancer patients and NSCLC patients but not in those undergoing ICIs treatment. Similar findings were obtained in cancer patients carrying the NFE2L2 mutation. Furthermore, in NSCLC and other cancer cohorts, patients with NFE2L2 mutation demonstrated more objective responses to ICIs than patients with wild type. Our in-house WXPH cohort further confirmed the efficacy of immunotherapy in the NFE2L2 mutated patients with NSCLC. Lastly, decreased inflammatory signaling pathways and immune-depleted immunological microenvironments were enriched in Nrf2-activating mutation patients with NSCLC. CONCLUSIONS Our study found that patients with Nrf2-activating mutation had improved immunotherapy outcomes than patients with wild type in NSCLC and other tumor cohorts, implying that Nrf2-activating mutation defined a distinct subset of pan-cancers and might have implications as a biomarker for guiding ICI treatment, especially NSCLC.
Collapse
|
research-article |
2 |
|
69
|
Pi Z, Liu W, Mao W. One size might fit all: Indole-3-propionic acid potentiates pan-cancer immunotherapy. Cell Host Microbe 2024; 32:627-630. [PMID: 38723599 DOI: 10.1016/j.chom.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 02/27/2025] [Imported: 02/27/2025]
Abstract
Microbial-based therapies have the potential to combat immunotherapy resistance, extending the boundaries of oncological therapeutics. In a recent issue of Cell, Jia et al. demonstrates an example of microbial collaboration to produce a postbiotic that promotes the stemness program of CD8+ T cells to augment immunotherapy at the pan-cancer level.
Collapse
|
|
1 |
|
70
|
Liu W, Pi Z, Liu NN, Mao W. Into the era of mycobiome-driven cancer research. Trends Cancer 2024; 10:389-392. [PMID: 38494372 DOI: 10.1016/j.trecan.2024.02.009] [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: 02/02/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] [Imported: 02/27/2025]
Abstract
The cancer mycobiome has recently become a research hotspot. While the intratumor mycobiota is implicated in cancer initiation and progression, the gut mycobiota functions as biomarkers for cancer diagnosis and treatment. In this forum article we highlight the involvement of the mycobiome in correlation-, causation-, and prediction-oriented cancer research and discuss the potential of this burgeoning field.
Collapse
|
|
1 |
|
71
|
Liu W, Pi Z, Mao W. Beyond probiotics: postbiotics sensitize cancer cells to immune checkpoint inhibitors. Trends Cancer 2024; 10:5-7. [PMID: 38101956 DOI: 10.1016/j.trecan.2023.12.001] [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: 11/30/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] [Imported: 02/28/2025]
Abstract
Accumulated research corroborates the feasibility of microbial antitumor therapies, enriching the diversity of cancer therapeutics. Recently, Ferrari et al. revealed the role of postbiotics in sensitizing cancer cells to immune checkpoint inhibitors (ICIs) and synergizing immunotherapy. Particularly, phytosphingosine induces upstream MYD88/NF-κB and downstream NLRC5 activation, enhances T cell effector responses, and inhibits tumor growth.
Collapse
|
|
1 |
|
72
|
Song C, Liu W, Wan Y, Mao W. Editorial: Immunotherapy and multimodality therapy for lung cancer. Front Immunol 2024; 15:1372513. [PMID: 38343541 PMCID: PMC10853457 DOI: 10.3389/fimmu.2024.1372513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/15/2024] [Imported: 02/28/2025] Open
|
Editorial |
1 |
|
73
|
Lv R, Guo Y, Liu W, Dong G, Liu X, Li C, Ren Y, Zhang Z, Neo SY, Mao W, Wu J. Revolutionizing cancer treatment: the emerging potential and potential challenges of in vivo self-processed CAR cell therapy. Theranostics 2024; 14:7424-7447. [PMID: 39659573 PMCID: PMC11626932 DOI: 10.7150/thno.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/23/2024] [Indexed: 12/12/2024] [Imported: 02/28/2025] Open
Abstract
Chimeric antigen receptor (CAR) cell immunotherapies, including CAR-T, CAR-Macrophages, CAR-Natural Killer, CAR-γδ T, etc., have demonstrated significant advancements in the treatment of both hematologic malignancies and solid tumors. Despite the notable successes of traditional CAR cell manufacturing, its application remains constrained by the complicated production process and expensive costs. Consequently, efforts are focused on streamlining CAR cell production to enhance efficacy and accessibility. Among numerous proposed strategies, direct in vivo generation of CAR cells represents the most substantial technical challenge, yet holding great promise for achieving clinical efficacy. Herein, we outlined the current state-of-the-art in vivo CAR therapy, including CAR technology development, transfection vectors, and influence factors of construction of CAR in vivo. We also reviewed the types and characteristics of different delivery systems and summarized the advantages of in vivo CAR cell therapy, such as rapid preparation and cost-effectiveness. Finally, we discussed the limitations, including technical issues, challenges in target and signal design, and cell-related constraints. Meanwhile, strategies have correspondingly been proposed to advance the development of CAR cell therapy, in order to open the new horizons on cancer treatment.
Collapse
|
Review |
1 |
|
74
|
Mao W, Wang S, Chen R, He Y, Lu R, Zheng M. Erratum to "lncRNA NORAD promotes lung cancer progression by competitively binding to miR-28-3p with E2F2". Open Med (Wars) 2022; 17:2138-2139. [PMID: 36660018 PMCID: PMC9816454 DOI: 10.1515/med-2022-0626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] [Imported: 02/28/2025] Open
Abstract
[This corrects the article DOI: 10.1515/med-2022-0538.].
Collapse
|
Published Erratum |
3 |
|
75
|
Mei J, Fu Z, Cai Y, Song C, Zhou J, Zhu Y, Mao W, Xu J, Yin Y. Erratum: SECTM1 is upregulated in immuno-hot tumors and predicts immunotherapeutic efficacy in multiple cancers. iScience 2023; 26:108302. [PMID: 37953953 PMCID: PMC10632109 DOI: 10.1016/j.isci.2023.108302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023] [Imported: 02/28/2025] Open
Abstract
[This corrects the article DOI: 10.1016/j.isci.2023.106027.].
Collapse
|
Published Erratum |
2 |
|