Buscar en
Clinics
Toda la web
Inicio Clinics Prognostic and Clinicopathological Value of ZWINT Expression Levels in Patients ...
Journal Information
Vol. 76.
(January 2021)
Share
Share
Download PDF
More article options
Visits
687
Vol. 76.
(January 2021)
REVIEW ARTICLE
Open Access
Prognostic and Clinicopathological Value of ZWINT Expression Levels in Patients with Lung Adenocarcinoma: A Systematic Review and Meta-analysis
Visits
687
Ran ZhuI, Huaguo WangI, Ling LinII,
Corresponding author
southwest1993@126.com

Corresponding author.
I Department of Clinical Laboratory, The First People's Hospital of Ziyang, Sichuan, China
II Department of Respiratory and Critical Care Medicine, The First People's Hospital of Ziyang, Sichuan, China
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (5)
Show moreShow less

The current study found that high Zeste White 10 interactor (ZWINT) expression is related to the poor prognosis of patients with a variety of cancers. This study mainly explored the relationship between the expression level of ZWINT and the prognosis of patients with lung adenocarcinoma (LUAD). Briefly, four English databases and two high-throughput sequencing databases were searched and relevant data for meta-analysis were extracted. Pooled mean difference and 95% confidence interval (CI) were used to assess the relationships between clinical features and the expression of ZWINT. Pooled hazard ratio and 95% CI were also used to assess the relationships between clinical features and the expression level of ZWINT. This meta-analysis was registered in PROSPERO (CRD42021249475). A total of 16 high-quality datasets comprising 2,847 LUAD patients were included in this study. Higher ZWINT expression levels were found in patients younger than 65 years, males, and smokers, and were correlated with advanced TNM stages and poor prognosis. Notably, there was no publication bias in this meta-analysis. Overall, our findings indicate that ZWINT is a potential biomarker for poor prognosis and clinicopathological outcomes of patients with LUAD.

KEYWORDS:
Prognosis
ZWINT
Lung Adenocarcinoma
Meta-Analysis
TCGA
Full Text
INTRODUCTIONBackground

Cancer is the leading cause of death and the reduction in life expectancy on a global scale. The burden of cancer as well as cancer incidence and mortality rates are increasing rapidly worldwide (1). In 2020, an estimated 19.3 million new cancer cases were diagnosed globally (18.1 million excluding nonmelanoma skin cancers), with nearly 10 million deaths (9.9 million excluding nonmelanoma skin cancers). Breast cancer has surpassed lung cancer as the most common cancer in women; however, lung cancer remains the leading cause of cancer-related death (1). In China, lung cancer is also the leading cause of cancer death and morbidity for both men and women (2). Of the pathological types of lung cancer, most are lung adenocarcinoma (LUAD) (3).

Zeste White 10 interactor (ZWINT) is an important protein that regulates centromeric division, playing a key role in chromosomal motion and mitosis (4). ZWINT is significantly overexpressed in a variety of cancers and is closely associated with the prognosis of patients with these cancers. Previously, ZWINT knockout was found to inhibit the migration, apoptosis, and colony formation of cancer cells while its downregulation reduced tumor volume. High ZWINT expression was also demonstrated to be closely related to the poor prognosis of patients with LUAD (5). Shorter relapse-free survival, overall survival (OS), and metastatic relapse-free survival may also be associated with higher ZWINT expression in patients with breast cancer (6).

With the advent of precision medicine and the development of sequencing technology, advancements in personalized genomics research have occurred because of the development of individual protocols for cancer and other diseases based on a person’s genetic information (7,8). Moreover, the medical model is gradually changing from empirical-based to an evidence-based model. At present, evidence-based medicine is mainly assessed via systematic evaluation and meta-analysis (9). Meta-analysis is a type of systematic evaluation in which data are statistically processed by quantitative synthesis, termed quantitative systematic evaluation. The greatest advantage of meta-analysis is that it avoids the limitation of a single small-sample clinical trial and can evaluate controversial results and resolve contradictions between studies, thereby providing good evidence for clinical decision making (10). In this study, we conducted a meta-analysis to clarify the prognostic and clinicopathological value of ZWINT in LUAD.

MATERIALS AND METHODSStudy Search

PubMed, EMBASE, Web of Science, and Cochrane Library were searched and the medical subject headings of LUAD and ZWINT were defined according to https://www.ncbi.nlm.nih.gov/mesh/. Data from two high-throughput sequencing databases, namely The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), were also retrieved for subsequent analysis. The search terms for PubMed were ((((Lung Adenocarcinomas) OR (Lung Adenocarcinoma)) OR (Adenocarcinoma, Lung)) OR (Adenocarcinomas, Lung)) AND ((((((ZW10 interacting protein-1) OR (Zwint1 protein)) OR (ZW10 interactor)) OR (Zwint-1 protein)) OR (ZW10 interacting protein 1)) OR (ZW10 interactor protein)). The retrieval method was adjusted according to the characteristics of the database and each database was searched from its inception to May 1, 2021. The languages of publications were limited to English and Chinese. As computer retrieval was limited by the literature and the indexing and retrieval strategy of the database itself, the recall and precision of the results may be affected. Therefore, in addition to computer retrieval, we manually searched all references in the original studies to ensure that all eligible studies were included.

Inclusion Criteria

The criteria for study inclusion were as follows: 1. included the relationship between ZWINT expression and LUAD prognostic indicators, such as OS and progression-free survival (PFS); 2. included corresponding statistical indicators, such as the hazard ratio (HR) and 95% confidence interval (CI), and if the HR and 95% CI were not clearly reported, the corresponding values could be calculated according to the information provided in the study; 3. included the most complete or latest study with the same research results; and 4. included human subjects. Nonoriginal studies, such as reviews, meta-analyses, case reports, and comments, were excluded from the analysis.

Selection and Inclusion Processes

Evidently repeated publications were first removed through a literature search, and publications that were obviously unrelated to this study were excluded via careful reading of the titles and abstracts. The full texts of studies that potentially met the inclusion criteria were examined. Different results for the same study were integrated, and the full text of the selected literature was carefully read to determine whether it met the inclusion criterion of an original article for the systematic review. When the relevant information needed in the literature research was incomplete or unclear, this was either obtained by reasonable deduction from the literature or clarified by the corresponding author. Finally, we decided whether the study should be included. For TCGA and GEO data, we also obtained relevant literature and screened them using a similar approach.

Quality Evaluation

To avoid bias in the quality evaluation, two reviewers assessed the included studies. The two reviewers independently used the Newcastle-Ottawa Scale (NOS) to evaluate quality (11), and communication and negotiation were carried out accordingly to ensure consistent application of the evaluation standards. The quality of the literature was evaluated formally. If the selection and evaluation of the literature differed between reviewers, a discussion was held or relevant professional researchers were asked to evaluate the studies.

Data Extraction

Two authors independently conducted literature quality assessment and data extraction, and then performed an in-depth reading of the text. Studies were then selected according to the above inclusion criteria. During the data extraction process, the data extracted by one reviewer were regularly checked by the other to identify differences in a timely manner. In the event of different decisions, a judgment was made by joint discussion or with the help of other professional researchers.

We downloaded RNA-seq data from TCGA in the fragments per kilobase of exon model per million mapped fragments (FPKM) format from the official website (https://portal.gdc.cancer.gov/) and converted the data to the ENSG ID gene symbol. We also downloaded clinical data for patients with LUAD. For the GEO database (https://www.ncbi.nlm.nih.gov/geo/), we downloaded raw microarray expression matrix data and converted the probe name to the gene symbol according to different sequencing platforms using the limma package and log2 translation (12). If more than one probe was used, the average value was employed. The corresponding clinical information was also downloaded.

Meta-analysis

Upon comparing the expression level of ZWINT with different clinical features, the standard mean difference (SMD) and 95% CI were used as statistics for the combined analysis. When the correlation between expression level of ZWINT and prognosis was investigated, the HR and 95% CI were used as statistics for the combined analysis. The Q and I2 tests were used to assess heterogeneity. Values of p<0.05 and I2>50% indicated high heterogeneity among the studies; the random effect model was used for meta-analysis. Values of p>0.05 and I2<50% indicated no or low heterogeneity among the studies; a fixed effect model was employed for this analysis. The results are presented as a forest map. By eliminating one study at a time, the remaining studies were combined to assess the degree of change in the results of sensitivity analysis. Begg's test was used to evaluate publication bias.

Construction and validation of the nomogram of ZWINT expression

TCGA database has the most detailed information about the clinical characteristics and follow-up of LUAD patients. Therefore, we used TCGA data to construct a nomogram containing ZWINT expression levels to prove the clinical application value of ZWINT. Xtile software (version 3.6.1) was used to derive the best cutoff value of ZWINT and divide patients into high expression and low expression groups. The principle is to group different values as cutoff values for statistical tests. The result with the smallest p value can be considered the best cutoff value. Kaplan-Meier curves and log rank tests were used to detect the difference in prognosis between the high and low ZWINT expression level groups.

We combined the clinical information of patients and the expression level of ZWINT for multivariate Cox regression analysis. For statistically significant factors (p<0.05), a nomogram was generated. In the survival analysis, the disease status and factor values will change with time. Accordingly, the use of a time-dependent ROC curve is undoubtedly a better choice. Therefore, a time-dependent ROC curve was used to determine the predictive ability of the nomogram. In addition, a calibration curve was used to verify the predictive ability of the nomogram.

RESULTSLiterature and Dataset Search

After excluding repetitive studies, 65 relevant studies from four English databases were retained according to the inclusion and exclusion criteria. After careful reading of the 65 manuscripts, we found that no data could be extracted. However, in the two high-throughput sequencing databases, the following datasets were found to contain enough data for subsequent analysis: GSE3141 (13), GSE8894 (14), GSE13213 (15), GSE14814 (16), GSE26939 (17), GSE29013 (18), GSE30219 (19), GSE31210 (20), GSE37745 (21), GSE41271 (22), GSE42127 (23), GSE50081 (24), GSE68465 (25), GSE72094 (26), GSE83227 (27), and TCGA (28). The process of document retrieval and inclusion is shown in Figure 1. The basic information of the 16 datasets is presented in Table 1. The data of 2,847 patients with LUAD were included in this study and the clinical characteristics of these patients are shown in Table S1. According to the literature corresponding to these databases, these studies are of high quality and have high NOS scores (Table 2). This meta-analysis was registered in PROSPERO (CRD42021249475).

Figure 1.

The general screening process for inclusion in the study.

(0.11MB).
Table 1.

Basic characteristics of the included datasets.

Datasets  First author  Year  Platform  No. of included LUAD patients  Country  PMID 
GSE3141  Andrea H Bild  2005  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  58  USA  16273092 
GSE8894  Eung-Sirk Lee  2007  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  63  South Korea  19010856 
GSE13213  Shuta Tomida  2008  GPL6480 Agilent-014850 Whole Human Genome Microarray 4×44K G4112F (Probe Name version)  117  Japan  19414676 
GSE14814  Chang-Qi Zhu  2009  GPL96 [HG-U133A] Affymetrix Human Genome U133A Array  71  Canada  20823422 
GSE26939  Wilkerson MD  2011  GPL9053 Agilent-UNC-custom-4×44K  116  USA  22590557 
GSE29013  Yang Xie  2011  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  30  USA  21742808 
GSE30219  Sophie Rousseaux  2011  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  106  France  23698379 
GSE31210  Hirokazu Okayama  2011  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  226  Japan  23028479 
GSE37745  Miriam Lohr  2012  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  106  Sweden  26608184 
GSE41271  Luc Girard  2012  GPL6884 Illumina HumanWG-6 v3.0 expression beadchip  181  USA  27354471 
GSE42127  Hao Tang  2012  GPL6884 Illumina HumanWG-6 v3.0 expression beadchip  133  USA  23357979 
GSE50081  Sandy D Der  2013  GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array  181  Canada  24305008 
GSE68465  Kerby Shedden  2015  GPL96 [HG-U133A] Affymetrix Human Genome U133A Array  442  USA  18641660 
GSE72094  M B Schabath  2015  GPL15048 Rosetta/Merck Human RSTA Custom Affymetrix 2.0 microarray [HuRSTA_2a520709.CDF]  398  USA  26477306 
GSE83227  A Bhattacharjee  2016  GPL8300 [HG_U95Av2] Affymetrix Human Genome U95 Version 2 Array  137  USA  11707567 
TCGA  Eric A Collisson  2020  Illumina Hiseq  482  USA  25079552 
Table S1.

The clinical characteristics of included datasets.

DatasetsAgeSexSmoking historyT stageN stageM stageAJCC stage
Female  Male  No  Yes  T1  T2  T3  T4  N0  N1  N2  N3  M0  M1  II  III  IV 
GSE3141  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  − 
GSE8894  59.41 ±10.40  28  33  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  − 
GSE13213  60.68± 10.16  57  60  56  61  54  50  87  22  117  79  13  25 
GSE14814  59.03 ±9.43  34  37  −  −  −  −  −  −  −  −  −  −  −  −  42  29 
GSE26939  64.04 ±10.88  63  53  12  101  −  −  −  −  −  −  −  −  100  62  19  19 
GSE29013  63.90 ±9.69  10  20  28  −  −  −  −  −  −  −  −  −  −  16 
GSE30219  61.49 ±9.28  19  66  −  −  71  12  82  85  −  −  −  − 
GSE31210  59.58 ±7.40  121  105  115  111  −  −  −  −  −  −  −  −  −  −  168  58 
GSE37745  62.94 ±9.22  60  46  −  −  −  −  −  −  −  −  −  −  102  70  19  13 
GSE41271  −  90  93  26  156  −  −  −  −  −  −  −  −  98  101  28  49 
GSE42127  65.76 ±10.29  65  68  −  −  −  −  −  −  −  −  −  −  133  89  22  20 
GSE50081  68.72 ±9.71  62  65  23  102  43  82  94  33  127  92  35 
GSE68465  64.42 ±10.10  220  223  49  300  150  251  28  12  299  88  53  −  −  −  −  −  − 
GSE72094  69.30 ±9.33  240  202  33  334  −  −  −  −  −  −  −  −  369  17  264  69  63  17 
GSE83227  63.09 ±10.08  81  56  17  115  42  68  75  22  122  15  85  25  11  15 
TCGA  65.10 ±10.02  258  224  64  418  165  254  45  18  319  92  68  461  21  263  118  80  21 
Table 2.

Study quality and bias in the retrospective cohort studies according to the Newcastle-Ottawa Scale (NOS) checklist.

DatasetsTotal scoreCohort selectionComparabilityOutcome 
Representativeness of the Exposed Cohort  Selection of the Non-Exposed Cohort  Ascertainment of Exposure  Demonstration that the outcome of interest was not present at study initiation  Comparability of cohorts based on the design or analysis  Assessment of outcome  Was follow-up long enough for outcomes to occur  Adequacy of the follow-up of cohorts 
GSE3141   
GSE8894 
GSE13213  ** 
GSE14814 
GSE26939 
GSE29013 
GSE30219 
GSE31210  ** 
GSE37745 
GSE41271 
GSE42127  ** 
GSE50081  ** 
GSE68465 
GSE72094 
GSE83227 
TCGA 
Associations Between ZWINT Expression and Clinical Characteristics

The results of the association analyses of ZWINT expression and clinical characteristics are shown in Table 3 and Figure 2A-J. The expression level of ZWINT was higher in patients younger than 65 years than in patients older than 65 years (SMD=0.109, 95% CI=0.028 and 0.190, p=0.009). Male patients had higher ZWINT expression levels than female patients (SMD=0.198, 95% CI=0.133 and 0.262, p<0.001). The expression levels of ZWINT were higher in patients with a history of smoking than in nonsmoking patients (SMD=0.428, 95% CI=0.310 and 0.545, p<0.001). The expression of ZWINT was also higher in patients with higher TNM stages as depicted by the following statistics: T stage (T2/T1: SMD=0.428, 95% CI=0.310 and 0.545, p<0.001; T3-T4/T1-T2: SMD=0.295, 95% CI=0.124 and 0.466, p=0.001), N stage (N1/N0: SMD=0.199, 95% CI=0.057 and 0.341, p=0.006; T3-T4/T1-T2: SMD=0.183, 95% CI=0.011 and 0.355, p=0.037), M stage (M1/M0: SMD=0.293, 95% CI=0.036 and 0.550, p=0.025), and AJCC stage (II/I: SMD=0.287, 95% CI=0.178 and 0.396, p<0.001; III-IV/I-II: SMD=0.126, 95% CI=0.008 and 0.243, p=0.036). The above meta-analysis was based on a fixed effect model because of zero or low heterogeneity.

Table 3.

Main results and publication bias for the meta-analysis between BUB1B and clinicopathological features, overall survival (OS), and progression-free survival (PFS).

Clinicopathological features/OS/PFS  Number of included datasets  SMD/HR(95%CI)  Z, p value  Heterogeneity test (I2, p value)  Publication bias (Begg's test) (Z, p value)  Pooling model 
Age (<=65/>65)  14  0.109 (0.028, 0.190)  2.63, 0.009  0%, 0.510  0.44, 0.661  Fixed 
Sex (Male/Female)  15  0.198 (0.133, 0.262)  6.01, <0.001  43.3%, 0.058  0.69, 0.488  Fixed 
Smoking status (Yes/No)  10  0.428 (0.310, 0.546)  7.11, <0.001  31.5%, 0.157  0.36, 0.721  Fixed 
T stage (T2/T1)  0.428 (0.310, 0.545)  7.14, <0.001  37.2%, 0.158  1.88, 0.060  Fixed 
T stage (T3-T4/T1-T2)  0.295 (0.124, 0.466)  3.38, 0.001  43.6%, 0.150  0.34, 0.734  Fixed 
N stage (N1/N0)  0.199 (0.057, 0.341)  2.74, 0.006  26.8%, 0.233  1.13, 0.260  Fixed 
N stage (N2-N3/N0-N1)  0.183 (0.011, 0.355)  2.09, 0.037  0%, 0.459  0.34, 0.734  Fixed 
M stage (M1/M0)  0.293 (0.036, 0.550)  2.24, 0.025  7.4%, 0.365  −0.24, 1.000  Fixed 
AJCC stage (II/I)  12  0.287 (0.178, 0.396)  5.15, <0.001  0%, 0.776  0.89, 0.373  Fixed 
AJCC stage (III-IV/I-II)  0.126 (0.008,0.243)  2.10, 0.036  0%, 0.989  1.77, 0.067  Fixed 
OS  15  1.263 (1.187, 1.340)  32.41, <0.001  39.4%, 0.058  0.59, 0.553  Fixed 
PFS  11  1.243 (1.150, 1.336)  26.17, <0.001  32.2%, 0.142  0.31, 0.755  Fixed 
Figure 2.

The results of the meta-analysis for the association of ZWINT expression with patient (A) age (<=65/>65), (B) sex (male/female), (C) smoking status (yes/no), (D) T stage (T2/T1), (E) T stage (T3-T4/T1-T2), (F) N stage (N1/N0), (G) N stage (N2-N3/N0-N1), (H) M stage (M1/M0), (I) AJCC stage (stage II/stage I), (J) AJCC stage (stage III-IV/stage I-II), (K) OS, and (L) PFS.

(0.35MB).
Association of ZWINT Expression with Prognosis

High ZWINT expression levels indicated worse OS and PFS for LUAD patients. Fifteen datasets contained survival data, which could be used to calculate the OS; the pooled results were HR=1.263, 95% CI=1.187-1.340, and p<0.001 with low heterogeneity using a fixed effect model. Eleven datasets contained survival data, which could be used to calculate the PFS; the pooled results were HR=1.243, 95% CI=1.150-1.336, and p<0.001 with low heterogeneity using a fixed effect model. The results are shown in Figure 2K and L, and Table 1.

Sensitivity Analyses and Publication Bias

As shown in Figure 3, no study was found to significantly affect the total pooled results alone, suggesting that this meta-analysis provided reliable results. As shown in Figure 4 and Table 1, no significant publication bias was found among all studies.

Figure 3.

Sensitivity analyses of patient (A) age (<=65/>65), (B) sex (male/female), (C) smoking status (yes/no), (D) T stage (T2/T1), (E) T stage (T3-T4/T1-T2), (F) N stage (N1/N0), (G) N stage (N2-N3/N0-N1), (H) M stage (M1/M0), (I) AJCC stage (stage II/stage I), (J) AJCC stage (stage III-IV/stage I-II), (K) OS, and (L) PFS.

(0.16MB).
Figure 4.

The results of publication bias for patient (A) age (<=65/>65), (B) sex (male/female), (C) smoking status (yes/no), (D) T stage (T2/T1), (E) T stage (T3-T4/T1-T2), (F) N stage (N1/N0), (G) N stage (N2-N3/N0-N1), (H) M stage (M1/M0), (I) AJCC stage (stage II/stage I), (J) AJCC stage (stage III-IV/stage I-II), (K) OS, and (L) PFS.

(0.11MB).
Construction and validation of the nomogram of ZWINT expression

Significant differences in OS (Figure 5C) and PFS (Figure 5F) were found between the high and low ZWINT expression groups. Multivariate Cox regression analysis also revealed that age, T stage, N stage, receiving radiotherapy, and ZWINT expression were independent prognostic factors for OS, while T stage, receiving radiotherapy, receiving chemotherapy, and ZWINT expression were independent prognostic factors for PFS (Table S2). The nomograms for predicting OS and PFS are shown in Figure 5A and 5B. The area under the curve (AUC) of the nomogram for OS at 1 year, 3 years, and 5 years was 0.711, 0.751, and 0.704 (Figure 5D), respectively, while that for PFS at 1 year, 3 years, and 5 years was 0.747, 0.744, and 0.731 (Figure 5G), respectively. The calibration curve (Figure 5E and 5H) also suggested that our nomogram has great prediction ability.

Figure 5.

Nomogram of the association between ZWINT expression level and OS (A) and PFS (B). (C). Kaplan-Meier survival curve depicting the OS of patients with different ZWINT expression levels. (D). Time-dependent ROC curve of OS. (E). Calibration curve of OS. (F). Kaplan-Meier survival curve depicting the PFS of patients with different ZWINT expression levels. (G). Time-dependent ROC curve of PFS. (H). Calibration curve of PFS.

(0.19MB).
Table S2.

The results of multivariate Cox regression analysis.

VariableHR95%HR lowerOSPFS
95% HR higher  P value  HR  95%HR lower  95%HR higher  p-value 
Age  1.020  1.003  1.038  0.022  1.009  0.994  1.025  0.236 
Sex (Female/Male)  0.969  0.695  1.350  0.851  0.928  0.683  1.262  0.634 
Race (White/Others)  1.017  0.682  1.517  0.935  0.907  0.616  1.336  0.621 
T stage  1.334  1.085  1.641  0.006  1.281  1.042  1.574  0.019 
N stage  1.521  1.242  1.862  0.000  0.891  0.722  1.099  0.281 
M stage  1.565  0.792  3.093  0.198  1.102  0.501  2.425  0.809 
Chemotherapy (Yes/No)  0.789  0.541  1.150  0.217  1.833  1.305  2.575  <0.001 
Radiotherapy (Yes/No)  2.018  1.348  3.020  0.001  3.245  2.294  4.590  <0.001 
Smoking history (No/Yes)  0.992  0.616  1.599  0.974  1.149  0.735  1.797  0.542 
ZWINT expression level (Low/High)  1.601  1.147  2.234  0.006  1.485  1.079  2.045  0.015 
DISCUSSION

In this study, we extensively searched four major English databases and two high-throughput sequencing databases; however, no literature reports on ZWINT and the prognosis of patients with LUAD were available. Nonetheless, in two high-throughput sequencing databases, we found datasets related to ZWINT expression and LUAD. Using the data in these datasets, we conducted a meta-analysis of individual patient data (IPD). The results of the IPD meta-analysis indicated significantly high ZWINT expression in patients younger than 65 years old, men, smoking patients, and patients with higher TNM stages. This study confirmed that ZWINT is related to the prognosis of LUAD patients. Further analysis revealed that the positive expression of ZWINT in LUAD was closely related to TNM stage. The higher the TNM stage, the higher the positive expression rate of ZWINT, which suggested that ZWINT might be involved in the occurrence, development, invasion, and metastasis of LUAD. However, the regulatory mechanism and function of ZWINT expression in esophageal cancer are not completely clear. Therefore, ZWINT may be used as a biomarker for predicting poor clinicopathology and the prognosis of patients with LUAD.

The protein encoded by ZWINT is composed of 278 amino acids, and plays a regulatory role in the cell cycle (29). Previously, ZWINT was found to be related to chromosome instability, which promotes the occurrence and development of a variety of malignant tumors (30). ZWINT has also been found to be related to the occurrence and development of a variety of tumors. For example, Wang et al. (31) used bioinformatics to investigate and analyze the differences in gene expression between normal and nasopharyngeal carcinoma tissues and found that significantly high ZWINT expression was related to nasopharyngeal carcinoma tissues. Akabane et al. (32) found that KIFC1 was positive in 67 (52%) of 129 patients with colorectal cancer based on immunohistochemistry; this positivity was also found to be related to the low OS rate. Moreover, the expression of ZWINT was found to be significantly correlated with KIFC1 expression, and KIFC1 and ZWINT knockout cells were observed to reduce the tumor formation ability (32). Kim et al. (33) found that the invasion and migration abilities of ZWINT-deficient pancreatic cancer cells were decreased; the expression levels of MMP2 and MMP9 were decreased; and the cell cycle arrested in the G2/M phase. The apoptosis rate was also gradually increased, and was accompanied by caspase-3 activation and anti-poly (ADP ribose) polymerase cleavage (33). The relative level of ZWINT expression decreased gradually with the progression of the cell cycle and decreased sharply during mitotic withdrawal. Treatment with cycloheximide reduced the level of ZWINT, while treatment with MG132 to inhibit the endogenous ubiquitin proteasome increased the level of ZWIN-1 in HEK293T cells and HeLa cells. These data suggest that ZWINT may be degraded by the endogenous ubiquitin proteasome (34).

ZWINT is also related to the pathological mechanism of lung cancer and may serve as a new biomarker. Using qRT-PCR, Peng et al. found that ZWINT was markedly overexpressed in lung cancer tissue and that knocking out ZWINT could reduce the proliferation of ncih226 and A549 cells; inhibit the migration, invasion, apoptosis, and colony formation of cancer cells; and reduce the tumor volume (5). Further, these researchers combined the clinical and survival follow-up data from TCGA to confirm that high ZWINT expression is associated with poor prognosis in patients with LUAD but not in patients with lung squamous cell carcinoma (LUSC). Some studies have confirmed that ZWINT is not only related to the prognosis of LUAD, but can also be used as a biomarker for the diagnosis of early lung cancer with high sensitivity (35,36).

With the continuous development of high-throughput sequencing technology and precision medicine, an increasing number of studies are focusing on the relationships between genes and diseases, especially cancer (37) and other nontumor chronic diseases (38). The studies included in this meta-analysis were high-throughput sequencing analyses. Although this technology has many advantages, it is associated with a high cost, complex operation, and difficult clinical application. Notably, immunohistochemistry has the advantages of simple operation, low economic cost, localization, and characterization. Further, compared with other protein detection methods, immunohistochemistry provides more direct and accurate localization and has high qualitative sensitivity. Accordingly, it is the preferred method for localization detection and analyses, and is especially useful for the transposition of some factors. The current research supports the proposal that tumors are essentially genetic diseases (39,40). In fact, ZWINT could serve as a new biomarker for LUAD.

To the best of our knowledge, this is the first IPD meta-analysis of the relationship between ZWINT expression and LUAD prognosis. This IPD meta-analysis overcame the shortcomings of limited survival data, insufficient amounts of long-term follow-up data, and insufficient utilization of outcome indicators for each research object, while obtaining more accurate conclusions (41). However, our research has limitations. First, although we included a sufficient number of studies in this analysis, our overall sample size was still slightly small. Second, because of the large time span of the included studies, the TNM stages of some patients may have been determined based on different criteria. Finally, most of the research data were derived from Europe and the United States. Accordingly, data from countries with high cancer incidence rates, especially China and Asia, as well as a global representation, are insufficient and lacking.

CONCLUSIONS

This meta-analysis revealed high ZWINT expression in young, male LUAD patients who smoke and have high TNM stages. Further, high ZWINT expression was found to be significantly associated with poor prognosis. However, such findings need to be further confirmed with a larger sample size and well-designed clinical trials.

AUTHOR CONTRIBUTIONS

Zhu R and Wang H conceived and designed the study, acquired and analyzed the data, and wrote the manuscript. Lin L contributed to data analysis and manuscript preparation. All authors read and approved the final version of the manuscript and agree to be accountable for all aspects of the research with regard to accuracy or integrity.

ACKNOWLEDGMENTS

We are grateful for the English editing services of American Journal Experts.

APPENDIX

Table S1.

The clinical characteristics of included datasets.

DatasetsAgeSexSmoking historyT stageN stageM stageAJCC stage
Female  Male  No  Yes  T1  T2  T3  T4  N0  N1  N2  N3  M0  M1  II  III  IV 
GSE3141  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  − 
GSE8894  59.41 ±10.40  28  33  −  −  −  −  −  −  −  −  −  −  −  −  −  −  −  − 
GSE13213  60.68± 10.16  57  60  56  61  54  50  87  22  117  79  13  25 
GSE14814  59.03 ±9.43  34  37  −  −  −  −  −  −  −  −  −  −  −  −  42  29 
GSE26939  64.04 ±10.88  63  53  12  101  −  −  −  −  −  −  −  −  100  62  19  19 
GSE29013  63.90 ±9.69  10  20  28  −  −  −  −  −  −  −  −  −  −  16 
GSE30219  61.49 ±9.28  19  66  −  −  71  12  82  85  −  −  −  − 
GSE31210  59.58 ±7.40  121  105  115  111  −  −  −  −  −  −  −  −  −  −  168  58 
GSE37745  62.94 ±9.22  60  46  −  −  −  −  −  −  −  −  −  −  102  70  19  13 
GSE41271  −  90  93  26  156  −  −  −  −  −  −  −  −  98  101  28  49 
GSE42127  65.76 ±10.29  65  68  −  −  −  −  −  −  −  −  −  −  133  89  22  20 
GSE50081  68.72 ±9.71  62  65  23  102  43  82  94  33  127  92  35 
GSE68465  64.42 ±10.10  220  223  49  300  150  251  28  12  299  88  53  −  −  −  −  −  − 
GSE72094  69.30 ±9.33  240  202  33  334  −  −  −  −  −  −  −  −  369  17  264  69  63  17 
GSE83227  63.09 ±10.08  81  56  17  115  42  68  75  22  122  15  85  25  11  15 
TCGA  65.10 ±10.02  258  224  64  418  165  254  45  18  319  92  68  461  21  263  118  80  21 
Table S2.

The results of multivariate Cox regression analysis.

VariableHR95%HR lowerOSPFS
95% HR higher  P value  HR  95%HR lower  95%HR higher  p-value 
Age  1.020  1.003  1.038  0.022  1.009  0.994  1.025  0.236 
Sex (Female/Male)  0.969  0.695  1.350  0.851  0.928  0.683  1.262  0.634 
Race (White/Others)  1.017  0.682  1.517  0.935  0.907  0.616  1.336  0.621 
T stage  1.334  1.085  1.641  0.006  1.281  1.042  1.574  0.019 
N stage  1.521  1.242  1.862  0.000  0.891  0.722  1.099  0.281 
M stage  1.565  0.792  3.093  0.198  1.102  0.501  2.425  0.809 
Chemotherapy (Yes/No)  0.789  0.541  1.150  0.217  1.833  1.305  2.575  <0.001 
Radiotherapy (Yes/No)  2.018  1.348  3.020  0.001  3.245  2.294  4.590  <0.001 
Smoking history (No/Yes)  0.992  0.616  1.599  0.974  1.149  0.735  1.797  0.542 
ZWINT expression level (Low/High)  1.601  1.147  2.234  0.006  1.485  1.079  2.045  0.015 

REFERENCES
[1]
H Sung , J Ferlay , RL Siegel , M Laversanne , I Soerjomataram , A Jemal , et al.
Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries.
[2]
X Zou , M Jia , X Wang , X Zhi .
[Changing Epidemic of Lung Cancer & Tobacco and Situation of Tobacco Control in China].
[3]
BD Hutchinson , GS Shroff , MT Truong , JP Ko .
Spectrum of Lung Adenocarcinoma.
Semin Ultrasound CT MRI, 40 (2019), pp. 255-264
[4]
H Wang , X Hu , X Ding , Z Dou , Z Yang , AW Shaw , et al.
Human Zwint-1 specifies localization of Zeste White 10 to kinetochores and is essential for mitotic checkpoint signaling.
[5]
F Peng , Q Li , SQ Niu , GP Shen , Y Luo , M Chen , et al.
ZWINT is the next potential target for lung cancer therapy.
[6]
HN Li , WH Zheng , YY Du , G Wang , ML Dong , ZF Yang , et al.
ZW10 interacting kinetochore protein may serve as a prognostic biomarker for human breast cancer: An integrated bioinformatics analysis.
[7]
IR König , O Fuchs , G Hansen , E von Mutius , MV Kopp .
What is precision medicine?.
[8]
SM Rego , MP Snyder .
High Throughput Sequencing and Assessing Disease Risk.
Cold Spring Harb Perspect Med, 9 (2019),
[9]
RI Horwitz , A Hayes-Conroy , R Caricchio , BH Singer .
From Evidence Based Medicine to Medicine Based Evidence.
[10]
YH Lee .
An overview of meta-analysis for clinicians.
[11]
CK Lo , D Mertz , M Loeb .
Newcastle-Ottawa Scale: comparing reviewers’ to authors’ assessments.
[12]
ME Ritchie , B Phipson , D Wu , Y Hu , CW Law , W Shi , et al.
limma powers differential expression analyses for RNA-sequencing and microarray studies.
[13]
AH Bild , G Yao , JT Chang , Q Wang , A Potti , D Chasse , et al.
Oncogenic pathway signatures in human cancers as a guide to targeted therapies.
[14]
ES Lee , DS Son , SH Kim , J Lee , J Jo , J Han , et al.
Prediction of recurrence-free survival in postoperative non-small cell lung cancer patients by using an integrated model of clinical information and gene expression.
[15]
S Tomida , T Takeuchi , Y Shimada , C Arima , K Matsuo , T Mitsudomi , et al.
Relapse-related molecular signature in lung adenocarcinomas identifies patients with dismal prognosis.
[16]
CQ Zhu , K Ding , D Strumpf , BA Weir , M Meyerson , N Pennell , et al.
Prognostic and predictive gene signature for adjuvant chemotherapy in resected non-small-cell lung cancer.
[17]
MD Wilkerson , X Yin , V Walter , N Zhao , CR Cabanski , MC Hayward , et al.
Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation.
[18]
Y Xie , G Xiao , KR Coombes , C Behrens , LM Solis , G Raso , et al.
Robust gene expression signature from formalin-fixed paraffin-embedded samples predicts prognosis of non-small-cell lung cancer patients.
[19]
S Rousseaux , A Debernardi , B Jacquiau , AL Vitte , A Vesin , H Nagy-Mignotte , et al.
Ectopic activation of germline and placental genes identifies aggressive metastasis-prone lung cancers.
[20]
H Okayama , T Kohno , Y Ishii , Y Shimada , K Shiraishi , R Iwakawa , et al.
Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas.
[21]
M Lohr , B Hellwig , K Edlund , JS Mattsson , J Botling , M Schmidt , et al.
Identification of sample annotation errors in gene expression datasets.
[22]
L Girard , J Rodriguez-Canales , C Behrens , DM Thompson , IW Botros , H Tang , et al.
An Expression Signature as an Aid to the Histologic Classification of Non-Small Cell Lung Cancer.
[23]
H Tang , G Xiao , C Behrens , J Schiller , J Allen , CW Chow , et al.
A 12-gene set predicts survival benefits from adjuvant chemotherapy in non-small cell lung cancer patients.
[24]
SD Der , J Sykes , M Pintilie , CQ Zhu , D Strumpf , N Liu , et al.
Validation of a histology-independent prognostic gene signature for early-stage, non-small-cell lung cancer including stage IA patients.
[25]
K Shedden , JM Taylor , SA Enkemann , MS Tsao , TJ Yeatman , Director’s Challenge Consortium for the Molecular Classification of Lung Adenocarcinoma, et al.
Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study.
[26]
MB Schabath , EA Welsh , WJ Fulp , L Chen , JK Teer , ZJ Thompson , et al.
Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma.
[27]
A Bhattacharjee , WG Richards , J Staunton , C Li , S Monti , P Vasa , et al.
Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses.
[28]
Cancer Genome Atlas Research Network .
Comprehensive molecular profiling of lung adenocarcinoma.
[29]
H Endo , K Ikeda , T Urano , K Horie-Inoue , S Inoue .
Terf/TRIM17 stimulates degradation of kinetochore protein ZWINT and regulates cell proliferation.
[30]
JK Famulski , L Vos , X Sun , G Chan .
Stable hZW10 kinetochore residency, mediated by hZwint-1 interaction, is essential for the mitotic checkpoint.
[31]
B Wang , W Wang , H Wang , W Liu .
Microarray Analysis of Novel Genes Involved in Nasopharyngeal Carcinoma.
[32]
S Akabane , N Oue , Y Sekino , R Asai , PQ Thang , D Taniyama , et al.
KIFC1 regulates ZWINT to promote tumor progression and spheroid formation in colorectal cancer.
[33]
JH Kim , Y Youn , JC Lee , J Kim , JH Hwang .
Involvement of the NF-κB signaling pathway in proliferation and invasion inhibited by Zwint-1 deficiency in Pancreatic Cancer Cells.
[34]
Y He , R Li , L Gu , H Deng , Y Zhao , Y Guo , et al.
Anaphase-promoting complex/cyclosome-Cdc-20 promotes Zwint-1 degradation.
[35]
D Jiang , Y Wang , M Liu , Q Si , T Wang , L Pei , et al.
A panel of autoantibodies against tumor-associated antigens in the early immunodiagnosis of lung cancer.
[36]
T Wang , H Liu , L Pei , K Wang , C Song , P Wang , et al.
Screening of tumor-associated antigens based on Oncomine database and evaluation of diagnostic value of autoantibodies in lung cancer.
[37]
Y Liao , H Xiao , M Cheng , X Fan .
Bioinformatics Analysis Reveals Biomarkers With Cancer Stem Cell Characteristics in Lung Squamous Cell Carcinoma.
[38]
Y Liao , C Huang , J Wang , X Fan .
Association of Surfactant-Associated Protein D Gene Polymorphisms with the Risk of COPD: a Meta-Analysis.
Clinics (Sao Paulo), 74 (2019), pp. e855
[39]
C Philpott , H Tovell , IM Frayling , DN Cooper , M Upadhyaya .
The NF1 somatic mutational landscape in sporadic human cancers.
[40]
CP Liao , RC Booker , JP Brosseau , Z Chen , J Mo , E Tchegnon , et al.
Contributions of inflammation and tumor microenvironment to neurofibroma tumorigenesis.
[41]
JF Tierney , M Clarke , LA Stewart .
Is there bias in the publication of individual patient data meta-analyses?.

No potential conflict of interest was reported.

Copyright © 2021. CLINICS
Article options
Tools
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos