Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2015

Open Access 01.12.2015 | Research article

Do relevant markers of cancer stem cells CD133 and Nestin indicate a poor prognosis in glioma patients? A systematic review and meta-analysis

verfasst von: Bin Wu, Caixing Sun, Fang Feng, Minghua Ge, Liang Xia

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2015

Abstract

Background

CD133 and Nestin, as the markers of cancer stem cells, have recently been reported frequently in the pathogenesis and development of human gliomas. However, the prognostic role of CD133 and Nestin in gliomas still remains controversial. In this study, we aimed to evaluate the association between the expression of CD133 and Nestin and the outcome of glioma patients by conducting a systematic review and meta-analysis.

Methods

We performed systematically electronic and manual searches through the database of Pubmed and embase (until to December 25, 2014) for titles and abstracts which investigated the relationships between CD133 and Nestin expression and outcome of glioma patients. A systematic review and meta-analysis was executed to generate Pooled hazard ratios (HRs) with 95 % confidence intervals (CIs) for overall survival (OS) and progression-free survival (PFS).

Results

A total of 1,490 patients from 32 studies (13 articles) were included in the analysis. 19 studies and 13 studies investigated correlation between CD133 expression or Nestin and survival in gliomas, respectively. Our results showed that high CD133 expression in patients with glioma was associated with poor prognosis in terms of OS (HR 1.69; 95 % CI, 1.16–2.47; P =0.0060) and PFS (HR, 1.64; 95 % CI, 1.12–2.39; P = 0.010). In addition, high Nestin expression were associated with worse OS (HR 1.751; 95 % CI, 1.19–2.58, p = 0.004) but has no significant association with PFS (HR 1.55; 95 % CI, 0.96–2.51, p = 0.074). Even more important, the results of the subgroup meta-analyses show that that high CD133 expression was associated with worse prognosis in terms of OS and PFS in patients with WHO IV glioma but not WHO II-III. On the other hand, Nestin high expression was associated with worse prognosis in terms of OS and PFS in patients with WHO II-III glioma but not WHO IV.

Conclusion

High level of CD133 expression trends to correlate with a worse OS and PFS in glioma patients, especially WHO IV gliomas and Nestin high expression trends to correlate with a worse OS in glioma patients especially WHO II–III, revealing both the markers of cancer stem cells may as the potential pathological prognostic markers for glioma patients.
Hinweise
Bin Wu, Caixing Sun and Fang Feng contributed equally to this work.

Competing interests

All the authors declare that they have no competing interests.

Authors’ contributions

WB, SC and FF carried out the electronic and manual searches. XL and WB independently inspected all candidate articles and conducted study selection. XL and GM conducted data extraction. XL participated in the design of the study and performed the statistical analysis. XL conceived of the study and participated in its design and coordination and the drafted the manuscript. All authors read and approved the final manuscript.

Introduction

Glioma is the most common primary brain tumor with the most grade malignancy, although in recent years the diagnosis and treatment of gliomas have made great progress, the prognosis of patients with glioma remains poor [1]. There is an urgent need to find a reliable marker to predict the prognosis of glioma, thereby providing the basis for the choice of a reasonable individualized treatment plan [2].
Cancer stem cell theory considers that the occurrence of tumors derives from some special cells, these cells are called cancer stem cells with the similar characteristics to embryonic stem cells, such as self-renewing and unlimited proliferation, multi-directional differentiation and anti-chemoradiotherapy and so on [3, 4]. Due to these characteristics of cancer stem cells, the traditional treatments, such as radiation and chemotherapy, can not effectively remove the cancer stem cells, the remaining tumor stem cells continue proliferation and differentiation, leading to tumor recurrence [4, 5]. There are a variety of markers used to isolate glioma stem cells, CD133 and Nestin are the most commonly used two markers that are widely expressed in various tumor cells, such as malignant glioma, liver cancer, ovarian cancer, colon cancer, lung cancer, etc. [611]. In recent years, a number of studies analyze the relationship between the markers of tumor stem cells CD133, Nestin and prognosis of patients with glioma, but due to differences in research method, sample size and the study population, the findings of a single sample are difficult to extend to the entire population and the obtained conclusions are inconsistent. This study used Meta-analysis method to systematically evaluate the literatures on the relationship between the expression of Nestin, CD133 that were multiple markers involving glioma stem cells and the prognosis of patients with glioma.

Materials and methods

Search strategy and study selection

A systematic literature search of the PubMed and Embase databases was conducted on studies evaluating the effect of the markers of cancer stem cells (CD133 and Nestin) on glioma patient survival. Our search strategy included terms (“Glioma” or “Glioblastoma”) and (“CD133 antigen” or “AC133 antigen” or “prominin-1” or “PROML1” or “Nestin”) and (“Survival” or “Mortality” or “Prognosis”). The literature search was conducted in 25 December 2014 and updated in 5 January 2015. Furthermore, a manual search of reference lists from the relevant original articles and review articles was also performed for additional relevant publications.
Two independent reviewers (Xia L and Wu B) independently inspected all candidate articles. Discrepancies were resolved by discussion. Studies that met all the following inclusion criteria were included in the review: (i) The diagnosis of glioma was made based on pathological examination; (ii) The association of the expression CD133 or Nestin with OS or PFS about gliomas was reported; (iii) The study provided the direct estimation of hazard ratios (HRs) and there was 95 % confidence intervals (CIs), or the date could be calculated by p values and other data reported. (iv) We included the studies with the largest sample size if the same glioma patient population were found to overlap among publications.

Definitions and data extraction

The OS (overall survival) was defined as the time interval between the medical treatment and the death of patient or the last follow-up. The PFS (progression free survival) was calculated as the time interval between the date of treatment and the detection of the tumor recurrence or death from any cause. Both two reviewers independently carried out data extraction from including studies and any discrepancies were resolved by discussion between the two. The following data were extracted from all including studies: the first author’s name, year of publication, country, sample size, patient age, WHO grade, detect method of CD133 or Nestin expression, cut‑off level, follow up period, survival analysis and prognostic outcomes (PFS and OS). Any discrepancies were resolved through discussion amongst the authors.

Quality assessment of primary studies

Quality assessment of included primary studies was independently executed by two reviewers (Xia L and Wu B) using the Newcastle–Ottawa Quality Assessment Scale (NOS). NOS scores of ≥6 were defined as high-quality studies. Any disagreement was determined by joint discussion.

Statistical analysis

All analyses were performed by using stata 12.0 statistical software (Stata Corporation, College Station, TX, USA). Hazard ratio (HR) and 95 % confidence intervals (CI) were got directly from each study or from estimation of Kaplan-Meier survival curves according to the methods by Parmer et al. An HR less than one was defined as a better prognosis in glioma patients with IDH mutation, whereas an HR more than one indicated a poor prognosis. We the most powerful one (multivariate analysis was superior to univariate analysis. And the latter one weighted over unadjusted Kaplan-Meier analysis) was chose, if several HR estimates were presented in the same study.
The heterogeneity of the included trials was assessed by the Cochrane’s Q statistic for each meta-analysis. We carried out both fixed-effects (Mantel–Haenszel method) and random effects (DerSimonian–Laird method) models and producted the pooled HRs. Thanks to a priory of assumptions about the likelihood of heterogeneity across primary studies, the random-effects model was chosen. In addition, subgroup analyses were performed to investigate the potential causes of heterogeneity according to study country, sample size, patient age, follow-up period, detect method of CD133 or Nestin expression, cut‑off level and WHO grade.
Publication bias was first investigated by Funnel plots and then performed for each of the pooled study groups using the Begg’s test. All p values were two-sided and the significance level was set at 5 %.

Results

The study inclusion procedure and study characteristics

The selection procedure of the eligible studies was presented in Fig. 1. In brief, a total of 1153 studies were identified from our initial electronic search. Of these, we eliminated 306 studies owing to overlapping data sets. Of which, 847 abstracts were considered relevant and full texts were reviewed in detail. By the end of the review 13 literatures on glioma [1224] (9 CD133 and 6 nestin; 1,490 patients), meeting our inclusion criteria for meta-analysis, were left with sufficient data for extraction.
The baseline characteristics of the literatures enrolled were summarized in Tables 1 and 2. Thirty two studies were included in those studies including 19 studies investigated the association between CD133 expression and outcome of glioma patients and 13 studies for Nestin. All studies were published between 2008 and 2014. Of these, the majority of the studies were executed in Europe (n = 23). Others were conducted in Asia (n = 8) and USA (n = 1). The total sample size from all studies was 1490 and the sample size was 24–379 patients and the range of medium age was 37.5–60.1 years. Of which, 13 studies evaluated grade II–III gliomas and 17 examined grade IV glioma. HRs and 95 % CI for OS or PFS in 27 studies could be directly extracted and was produced by Kaplan-Meier analysis for the 5 remaining studies. The most frequently used cutoff values for the high versus low/ present versus absent expression of CD133 or Nestin were the median (n = 15) and values calculated by using several semiquantitative methods.
Table 1
General characteristics of included studies (about the relationships between CD133 expression and OS or PFS in glioma patients)
First author
Year
Origin country
No. of patients
Median/mean age (year)
WHO grade
Detect method
Cut off level
Survival end points
Follow up period
Survival analysis
Adjusted variables
Ardebili [10]
2011
Slovenia
24
mean 60
IV
QRT-PCR
30 000 (─, 2-ΔΔCt > 30.000)
OS
NR
univariate (KM)
NR
Kase [11]
2013
Estonia
42
30–77
IV
IHC
median
OS
NR
multivariate
Radiotherapy dose, Chemotherapy, Karnofsky performance score
Kim [12]
2011
Korea
88
mean 54.9 (13–80)
IV
IHC
50 %
OS
mean 13.9 months (1–53)
multivariate
age, sex, Karnofsky performance scale (KPS) score, extent of removal, chemoradiotherapeutic modality, temozolomide modality, after temozolomide therapy and stem cell marker expression
        
PFS
   
Melguizo [13]
2012
Italy
75
24–81
IV
IHC
25 %
OS
NR
multivariate (KM)
NR
        
PFS
   
Metellus [14]
2011
France
48
mean 60.1 ± 9.2
IV
QPCR
1.03
OS
Median 18.9 months
multivariable
Age, extent of surgery, MGMT status (MethyLight)
        
PFS
   
Shibahara [15]
2013
Japan
112
Media 57 (7–77)
IV
WB
CD133/b-actin ratio = 1
OS
Median 25.7 months (3–152)
multivariable
Ki67 LI (≥35 %), 9p homozygous deletion and 10q loss
Shin [16]
2013
Korea
67
NR
IV
IHC
50 %
OS
NR
multivariable
status
        
PFS
   
Zeppernik [17]
2008
Germany
48
NR
II–III
IHC
1 %
OS
mean 86 months (±39)
multivariate
age, WHO grade and extent of resection
        
PFS
   
Dahlrot [18]
2014
Denmark
25
NR
II
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status
        
PFS
   
Dahlrot [18]
2014
Denmark
26
NR
III
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status
        
PFS
   
Dahlrot [18]
2014
Denmark
185
NR
IV
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status
        
PFS
   
Table 2
General characteristics of included studies (about the relationships between Nestin expression and OS or PFS in glioma patients)
First author
Year
Origin country
No. of patients
Median/mean age (year)
WHO grade
Detect method
Cut off level
Survival end points
Follow up period
Survival analysis
Adjusted variables
Hatanpa [19]
2014
USA
50
median 37.5 years (20–66)
II–III
IHC
median
OS
4.3 year
multivariate (KM)
nestin, IDH, WHO grade, oligodendroglioma component (astrocytoma or oligoastrocytoma), MIB-1, age, sex, extent of resection and type of adjuvant, therapy
Milde [20]
2012
Russia
379
NR
II–III
IHC
50 %
OS
median 53 months
multivariate
cytogenetic group only, or cytogenetic group and posterior fossa group
        
PFS
   
wan [21]
2011
Germany
283
NR
II–IV
IHC
median
OS
Mean 11.21 ± 4.26 years
multivariate
WHO grade, patient age at diagnosis and extent of resection
Kim [12]
2011
Korea
88
mean 54.9 years (13–80)
IV
IHC
50 %
OS
mean 13.9 (1–53) months
multivariate
age, sex, KPS, extent of removal, chemoradiotherapeutic modality, temozolomide modality, after temozolomide therapy and stem cell marker expression
        
PFS
   
Arai [22]
2012
Japan
64
median 55 years (11–79)
III–IV
IHC
60 %
OS
1–67 months
KM
NR
Dahlrot [18]
2014
Denmark
25
NR
II
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status
        
PFS
   
Dahlrot [18]
2014
Denmark
26
NR
III
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status
        
PFS
   
Dahlrot [18]
2014
Denmark
185
NR
IV
IF
median
OS
Media 12 months
multivariate
age, performance status (PS) and IDH1 status

CD133 expression and OS in gliomas

A total of 11 studies were involved in the association between CD133 expression and OS of glioma patients, among which statistically significant heterogeneity was observed (I2 = 77.8 %). Therefore, a random model was applicable to calculate a pooled HR and 95 % CI, the combined analysis showed that upon comparing patients with a low expression of CD133, patients possessing high CD133 expression had a significantly poorer OS (HR = 1.69, 95 % CI: 1.16 to 2.47; P = .0006)(Fig. 2).
In order to avoid the influence of heterogeneity, further subgroup analyses were conducted and stratified based on the study origin, sample size, follow up period, patient age, test method, cut‑off level and WHO grade. And the results showed that almost the subgroup analysis with origin country did not alter the prognostic role of glioma in OS substantially. And, increased expression of CD133 predicted a significantly worse OS in following subgroups including sample size ≤ 50, follow up period > media 12 months or no referred median/mean age referred, IHC test and cut‑off level not median. However, we did not discover any significant association in other subgroups (Table 3). Even more important, when in subtotal analysis stratified by WHO grade of glioma, 8 studies of WHO IV glioma exhibited a significant association between increased expression of CD133 and poor OS (HR 1.73; 95 % CI, 1.20–2.50, p = 0.004 I2 = 76.3). While, we did not find any significant association in subgroups of WHO II–III giomas (HR 2.38; 95 % CI, 0.30–18.82, p = 0.441 I2 = 87.0).
Table 3
Subgroup analyses of the relationships between CD133 ovexpression and overall survival or progression-free-survival
Comparison variables
Overall survival
Progression-free survival
Number of studies, Heterogeneity (I2 statistics; %)
HR 95 % CI, P value
Interaction, P value
Number of studies, Heterogeneity (I2 statistics; %)
HR 95 % CI, P value
Interaction, P value
Total
11 (77.8)
1.69 (1.16–2.47), 0.006
NA
8 (68.2)
1.64 (1.12–2.39), 0.010
NA
Origin country
      
Europe
8 (81.0)
1.63 (1.03–2.61), 0.039
0.030
6 (76.5)
1.74 (1.06–2.83), 0.027
0.579
Asian
3 (42.5)
1.85 (1.02–3.35), 0.044
 
2 (0.0)
1.37 (0.86–2.18), 0.188
 
Sample size
      
>50
4 (77.4)
1.45 (0.91–2.31), 0.116
0.000
3 (0.0)
1.37 (1.01–1.73), 0.009
0.092
≤50
7 (67.6)
1.94 (1.07–3.54), 0.030
 
5 (77.0)
1.99 (0.94–4.22), 0.073
 
Follow up period
      
1,>media 12months
4 (74.1)
2.48 (1.13–5.45), 0.024
0.000
3 (86.7)
2.64 (0.98–7.11), 0.054
0.060
2,≤media 12months
3 (43.0)
0.90 (0.54–1.49), 0.678
 
3 (0.0)
1.12 (0.79–1.57), 0.530
 
3,No Referreed
4 (0)
2.11 (1.52–2.93), 0.000
 
2 (0.0)
1.64 (1.19–2.27), 0.003
Median/mean age y
      
Referred
6 (0.0)
1.85 (1.45–2.37), 0.000
0.000
3 (0.0)
1.59 (1.24–2.04), 0.000
0.713
No Referred
5 (79.8)
1.57 (0.70–3.55), 0.274
 
5 (80.8)
1.79 (0.81–3.97), 0.153
 
Test method
      
IHC
5 (64.8)
2.16 (1.25–3.73), 0.006
0.000
4 (80.9)
2.25 (1.11–4.58), 0.025
0.122
Others
6 (71.2)
1.37 (0.85–2.22), 0.202
 
4 (20.4)
1.31 (0.93–1.84), 0.122
 
Cut‑off level
      
1:median
4 (61.4)
1.09 (0.66–1.79) 0.748
0.000
3 (0.0)
1.12 (0.79–1.57), 0.530
0.024
2:others
7 (51.3)
2.17 (1.44–3.27), 0.000
 
5 (74.5)
2.07 (1.25–3.43), 0.005
 
WHO grade
      
IV
8 (76.3)
1.73 (1.20–2.50), 0.004
0.915
5 (0.00)
1.46 (1.19–1.80), 0.000
0.143
II–III
3 (87.0)
2.38 (0.30–18.82), 0.411
 
3 (88.1)
2.17 (0.42–11.28), 0.355
 
Regarding the publication bias in the studies, we found no funnel plot asymmetry. Furthermore, Begger’s test was applied to provide statistical evidence for funnel plot symmetry. As expected, the P value of Begger’s test was 0.350 (Fig. 3). Hence, there was no evidence for significant publication bias in the meta-analysis.

CD133 expression and PFS in gliomas

Eight studies provided information concerning the association between CD133 expression and PFS of glioma patients. Similarly, a random model was applicable to calculate a pooled HR and 95 % CI, since significant heterogeneity was observed in the pooled studies (I2 = 80.5 %). The combined analysis exhibited a significant association between increased expression of CD133 and poor PFS (HR 1.73; 95 % CI, 1.86–2.83, p = 0.027) (Fig. 4).
Further subgroup analyses were conducted and stratified based on the study origin, sample size, follow up period, patient age, test method, cut‑off level and WHO grade. And the results show that increased expression of CD133 predicted a significantly worse OS in following subgroups including sample size ≤ 50, follow up period > media 12 months or no referred median/mean age referred, IHC test and cut‑off level not median. However, we did not discover any significant association in other subgroups (Table 3). Similarly, the results in subtotal analysis stratified by WHO grade of glioma showed that five studies of WHO IV glioma exhibited a significant association between increased expression of CD133 and poor OS (HR 1.46; 95 % CI, 1.19–1.80, p = 0.000 I2 = 0.00). However, we did not find any significant association in subgroups of WHO II–III giomas (HR 2.17; 95 % CI, 0.42–11.28, p = 0.355 I2 = 88.1).
At the same time, no funnel plot asymmetry was found in the studies and the Begger’s test did not show any evidence of publication bias (P = 0.902; Fig. 3).

Nestin expression and OS in gliomas

Eight eligible studies provided the estimation of the HR and 95 % CI for the correlation between Nestin expression and OS of glioma patients, among which statistically significant heterogeneity was observed (I2 = 75.8 %). Therefore, a random model was applicable to calculate a pooled HR and 95 % CI and the combined analysis showed that upon comparing patients with a low expression of Nestin, patients with high Nestin expression had a significantly poorer OS (HR = 1.75, 95 % CI: 1.19 to 2.58; P = 0.004) (Fig. 5).
The subgroup analyses were conducted and stratified based on the study origin, sample size, follow up period, patient age, test method, cut‑off level and WHO grade. And the results showed that increased expression of CD133 predicted a significantly worse OS in following subgroups including sample size ≤ 50, follow up period > media 12 months or no referred, median/mean age referred, IHC test and cut‑off level not median. However, we did not discover any significant association in other subgroups (Table 4). Even more important, when in subtotal analysis stratified by WHO grade of glioma, four studies of WHO II–III glioma exhibited a significant association between increased expression of Nestin and poor OS (HR 3.11; 95 % CI, 1.45–6.67, p = 0.004 I2 = 57.2). However, we did not discover any significant association in subgroups of WHO IV giomas (HR 1.09; 95 % CI, 0.83–1.44, p = 0.518 I2 = 0.000).
Table 4
Subgroup analyses of the relationships between Nestin expression and overall survival or progression-free-survival
Comparison variables
Overall survival
Progression-free survival
Number of studies, Heterogeneity (I2 statistics; %)
HR 95 % CI, P value
Interaction, P value
Number of studies, Heterogeneity (I2 statistics; %)
HR 95 % CI, P value
Interaction, P value
Total
8 (75.8)
1.75 (1.19–2.58), 0.004
NA
5 (71.5)
1.55 (0.96–2.51), 0.074
NA
Origin country
      
Europe
5 (73.1)
1.52 (1.04–2.21), 0.029
0.014
4 (74.1)
1.76 (0.98–3.14), 0.057
0.142
Asian
2 (81.8)
1.92 (0.43–8.62), 0.393
 
1 (NA)
1.55 (0.96–2.51), 0.973
 
USA
1 (NA)
13.42 (2.56–70.34), 0.002
    
Sample size
      
>50
5 (71.6)
1.41 (1.00–1.98), 0.051
0.005
3 (82.6)
1.39 (0.77–2.50), 0.277
0.322
≤50
3 (71.2)
3.61 (0.99–13.17), 0.052
 
2 (36.5)
2.14 (0.85–5.35), 0.105
 
Follow up period
      
1,>media 12months
2 (68.7)
4.96 (1.08–22.78), 0.040
0.001
1 (NA)
2.40 (1.60–3.40), 0.000
0.008
2,≤media 12months
3 (68.8)
1.64 (0.74–3.67), 0.225
 
3 (55.4)
1.49 (0.76–2.94), 0.246
 
3,No Referreed
3 (66.8)
1.35 (0.77–2.38), 0.300
 
1 (NA)
1.01 (0.57–1.79), 0.973
 
Median/mean age y
      
Referred
3 (83.4)
3.36 (0.72–15.61), 0.123
0.154
1 (NA)
1.01 (0.57–1.80), 0.973
0.142
No Referred
5 (75.8)
1.52 (1.04–2.21), 0.029
 
4 (74.7)
1.76 (0.98–3.14), 0.057
 
Test method
      
IHC
5 (82.1)
2.11 (1.10–4.04), 0.025
0.671
2 (83.6)
1.60 (0.69–3.73), 0.275
0.063
Others
3 (68.8)
1.64 (0.74–3.67), 0.225
 
3 (55.4)
1.49 (0.76–2.51), 0.246
 
Cut‑off level
      
1:median
5 (74.1)
1.55 (1.00–2.42), 0.050
0.021
3 (55.4)
1.49 (0.76–2.94), 0.246
0.063
2:others
3 (75.6)
2.10 (0.88–5.00), 0.093
 
2 (83.6)
1.60 (0.69–3.73), 0.275
 
WHO grade
      
IV
2 (0.0)
1.09 (0.83–1.44), 0.518
0.000
2 (0.00)
1.04 (0.77–1.41), 0.808
0.000
II–III
4 (57.2)
3.11 (1.45–6.67), 0.004
 
3 (0.00)
2.34 (1.68–3.27), 0.000
 
The publication bias in the studies was conducted, the funnel plot asymmetry was not found. Then, we applied Begger’s test to provide statistical evidence for funnel plot symmetry. As expected, the P value of Begger’s test was 0.266 (Fig. 3). Hence, there was no evidence for significant publication bias in the meta-analysis.

Nestin expression and PFS in gliomas

The combined analysis of the five studies did not exhibit a significant association between increased expression of Nestin and poor PFS (HR 1.55; 95 % CI, 0.96–2.51, p = 0.074) (Fig. 6).
Further subgroup analyses were conducted and stratified based on the study origin, sample size, follow up period, patient age, test method, cut-off level and WHO grade. And the results showed that increased expression of Nestin predicted a significantly worse OS in following subgroups including sample size ≤ 50, follow up period > media 12 months or no referred, median/mean age referred, IHC test and cut‑off level not median. However, we did not discover any significant association in other subgroups (Table 4). Similarly, the results in subtotal analysis stratified by WHO grade of glioma showed that two studies of WHO IV glioma exhibited a significant association between increased expression of Nestin and poor OS (HR 2.34; 95 % CI, 1.68–3.27, p = 0.000 I2 = 0.000). However, we did not find any significant association in subgroups of WHO II–III giomas (HR 1.04; 95 % CI, 0.77-1.41, p = 0.808 I2 = 0.000).
At the same time, no funnel plot asymmetry was found in the studies and the Begger’s test did not show any evidence of publication bias (P = 0.221; Fig. 3).

Discussion

Cancer stem cells (CSC), a small portion of cell population with the characteristics of stem cells, are present in the tumor tissue. It is the root to form the tumor cells with different degree of differentiation because of a capacity of self-renewing and the multi-directional differentiative potential [1, 3, 25]. At the earliest, CSC is found in the blood system tumors [26]. Recently, with the development of flow cytometry and in vitro tumor formation technology, cancer stem cells have been isolated and identified in a variety of solid tumors [27, 28]. The proposed cancer stem cell theory makes people have a new understanding on the biological behavior of tumors: the tumor is not only a genetic disease, but also a disease of stem cells. Stem cells become cancer stem cells after gene mutations, which is the root of tumor recurrence and metastasis [29].
Glioma stem cells can be sorted by finding its markers in order to conducted the targeted chemotherapy for glioma stem cells, which can effectively improve the specificity of chemotherapy and reduce the side effects on the normal engine body and cells, thereby finding a new breakthrough for cancer treatment [30]. Recently, the markers of glioma stem cell that are studied more than others include CD133, nestin, HMGA1, A2B5, etc. [3134]. It has made some breakthroughs from the continuous in-depth study of these markers, but there is a lot of controversy.
At present, a number of studies have shown that glioma stem cell markers CD133 and Nestin are closely related to the prognosis of patients with glioma, but some individual studies show that there is no clear relationship between CD133, Nestin and the prognosis of patients with glioma. We used Meta-analysis to systematically evaluate the literatures on the relationship between the glioma stem cells markers Nestin, CD133 and the prognosis of patients with glioma in order to accurately and objectively evaluate the application value of CDl33 and Nestin in prognosis of glioma.
The current meta-analysis is the first to systematically estimate the association between cancer stem cell markers and glioma survival. In this study, the results showed that CSCs marker CD133 was associated with worse OS and PFS in glioma patients and Nestin was associated with worse OS but not PFS. Especially, subgroup analysis showed that the overexpression of CD133 had a more significant predictive value for glioma patients with WHO grade II–III, but Nestin for WHO grade IV.
Limitations of this study include: (1) the number of selected cases in the research is too few, especially only 1490 cases. (2) Although this study has tried to collect all the relevant data, the potential publication bias is inevitable, some data may be missing. The missing information may reduce the reliability of CDl33 and Nestin expression as a prognostic indicator of glioma. (3) Although immunohistochemistry and RT-PCR are the detection methods for CDl33, Nestin protein and gene, there are still some differences. This article collected all relevant documents in protein and gene level, there may be some heterogeneity in the methods. (4) the criterion of positive decision for the selected CDl33 and Nestin is different (median, mean, 25 %, 50 %, 1 %, or 3 others), leading to the heterogeneity of studies. (5) Follow-up time is different. So the study used a random effects model and subgroup analysis to compensate for these deficiencies.
Above all, we found that high CDl33 expression may be independent risk factor for glioma patients’ prognosis, especially WHO IV gliomas and high Nestin expression may be independent risk factor for glioma patients’ prognosis with grade WHO II–III. Based on the current findings, assessing CDl33 and Nestin expression could provide better prognostic information for patients with glioma and be used as a novel therapeutic target. Further large-scale cohort studies are needed to validate our results.

Acknowledgment

The study was sponsored by the Natural Science Program of Zhejiang Department of Finance (No.: LY12H16032) and Youth Scientific Innovation Foundation of Zhejiang Cancer Hospital (No.: QN201402).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

All the authors declare that they have no competing interests.

Authors’ contributions

WB, SC and FF carried out the electronic and manual searches. XL and WB independently inspected all candidate articles and conducted study selection. XL and GM conducted data extraction. XL participated in the design of the study and performed the statistical analysis. XL conceived of the study and participated in its design and coordination and the drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Stupp R, Tonn JC, Brada M, Pentheroudakis G. High-grade malignant glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21 Suppl 5:v190–3.CrossRefPubMed Stupp R, Tonn JC, Brada M, Pentheroudakis G. High-grade malignant glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21 Suppl 5:v190–3.CrossRefPubMed
2.
Zurück zum Zitat Huse JT, Aldape KD. The evolving role of molecular markers in the diagnosis and management of diffuse glioma. Clin Cancer Res. 2014;20:5601–11.CrossRefPubMed Huse JT, Aldape KD. The evolving role of molecular markers in the diagnosis and management of diffuse glioma. Clin Cancer Res. 2014;20:5601–11.CrossRefPubMed
3.
Zurück zum Zitat Dontu G, Liu S, Wicha MS. Stem cells in mammary development and carcinogenesis: implications for prevention and treatment. Stem Cell Rev. 2005;1:207–13.CrossRefPubMed Dontu G, Liu S, Wicha MS. Stem cells in mammary development and carcinogenesis: implications for prevention and treatment. Stem Cell Rev. 2005;1:207–13.CrossRefPubMed
4.
Zurück zum Zitat Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol. 2005;45:872–7.CrossRefPubMed Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol. 2005;45:872–7.CrossRefPubMed
5.
Zurück zum Zitat Niero EL, Rocha-Sales B, Lauand C, Cortez BA, de Souza MM, Rezende-Teixeira P, et al. The multiple facets of drug resistance: one history, different approaches. J Exp Clin Cancer Res. 2014;33:37.CrossRefPubMedCentralPubMed Niero EL, Rocha-Sales B, Lauand C, Cortez BA, de Souza MM, Rezende-Teixeira P, et al. The multiple facets of drug resistance: one history, different approaches. J Exp Clin Cancer Res. 2014;33:37.CrossRefPubMedCentralPubMed
6.
Zurück zum Zitat Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed
7.
Zurück zum Zitat Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T, Moriwaki H. Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun. 2006;351:820–4.CrossRefPubMed Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T, Moriwaki H. Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun. 2006;351:820–4.CrossRefPubMed
8.
Zurück zum Zitat Ferrandina G, Bonanno G, Pierelli L, Perillo A, Procoli A, Mariotti A, et al. Expression of CD133-1 and CD133-2 in ovarian cancer. Int J Gynecol Cancer. 2008;18:506–14.CrossRefPubMed Ferrandina G, Bonanno G, Pierelli L, Perillo A, Procoli A, Mariotti A, et al. Expression of CD133-1 and CD133-2 in ovarian cancer. Int J Gynecol Cancer. 2008;18:506–14.CrossRefPubMed
9.
Zurück zum Zitat Kojima M, Ishii G, Atsumi N, Fujii S, Saito N, Ochiai A. Immunohistochemical detection of CD133 expression in colorectal cancer: a clinicopathological study. Cancer Sci. 2008;99:1578–83.CrossRefPubMed Kojima M, Ishii G, Atsumi N, Fujii S, Saito N, Ochiai A. Immunohistochemical detection of CD133 expression in colorectal cancer: a clinicopathological study. Cancer Sci. 2008;99:1578–83.CrossRefPubMed
10.
Zurück zum Zitat Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15:504–14.CrossRefPubMed Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15:504–14.CrossRefPubMed
11.
Zurück zum Zitat Zhang M, Song T, Yang L, Chen R, Wu L, Yang Z, et al. Nestin and CD133: valuable stem cell-specific markers for determining clinical outcome of glioma patients. J Exp Clin Cancer Res. 2008;27:85.CrossRefPubMedCentralPubMed Zhang M, Song T, Yang L, Chen R, Wu L, Yang Z, et al. Nestin and CD133: valuable stem cell-specific markers for determining clinical outcome of glioma patients. J Exp Clin Cancer Res. 2008;27:85.CrossRefPubMedCentralPubMed
12.
Zurück zum Zitat Ardebili SY, Zajc I, Gole B, Campos B, Herold-Mende C, Drmota S, et al. CD133/prominin1 is prognostic for GBM patient’s survival, but inversely correlated with cysteine cathepsins’ expression in glioblastoma derived spheroids. Radiol Oncol. 2011;45:102–15.CrossRefPubMedCentralPubMed Ardebili SY, Zajc I, Gole B, Campos B, Herold-Mende C, Drmota S, et al. CD133/prominin1 is prognostic for GBM patient’s survival, but inversely correlated with cysteine cathepsins’ expression in glioblastoma derived spheroids. Radiol Oncol. 2011;45:102–15.CrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Kase M, Minajeva A, Niinepuu K, Kase S, Vardja M, Asser T, et al. Impact of CD133 positive stem cell proportion on survival in patients with glioblastoma multiforme. Radiol Oncol. 2013;47:405–10.CrossRefPubMedCentralPubMed Kase M, Minajeva A, Niinepuu K, Kase S, Vardja M, Asser T, et al. Impact of CD133 positive stem cell proportion on survival in patients with glioblastoma multiforme. Radiol Oncol. 2013;47:405–10.CrossRefPubMedCentralPubMed
14.
Zurück zum Zitat Kim KJ, Lee KH, Kim HS, Moon KS, Jung TY, Jung S, et al. The presence of stem cell marker-expressing cells is not prognostically significant in glioblastomas. Neuropathology. 2011;31:494–502.CrossRefPubMed Kim KJ, Lee KH, Kim HS, Moon KS, Jung TY, Jung S, et al. The presence of stem cell marker-expressing cells is not prognostically significant in glioblastomas. Neuropathology. 2011;31:494–502.CrossRefPubMed
15.
Zurück zum Zitat Melguizo C, Prados J, Gonzalez B, Ortiz R, Concha A, Alvarez PJ, et al. MGMT promoter methylation status and MGMT and CD133 immunohistochemical expression as prognostic markers in glioblastoma patients treated with temozolomide plus radiotherapy. J Transl Med. 2012;10:250.CrossRefPubMedCentralPubMed Melguizo C, Prados J, Gonzalez B, Ortiz R, Concha A, Alvarez PJ, et al. MGMT promoter methylation status and MGMT and CD133 immunohistochemical expression as prognostic markers in glioblastoma patients treated with temozolomide plus radiotherapy. J Transl Med. 2012;10:250.CrossRefPubMedCentralPubMed
16.
Zurück zum Zitat Metellus P, Nanni-Metellus I, Delfino C, Colin C, Tchogandjian A, Coulibaly B, et al. Prognostic impact of CD133 mRNA expression in 48 glioblastoma patients treated with concomitant radiochemotherapy: a prospective patient cohort at a single institution. Ann Surg Oncol. 2011;18:2937–45.CrossRefPubMed Metellus P, Nanni-Metellus I, Delfino C, Colin C, Tchogandjian A, Coulibaly B, et al. Prognostic impact of CD133 mRNA expression in 48 glioblastoma patients treated with concomitant radiochemotherapy: a prospective patient cohort at a single institution. Ann Surg Oncol. 2011;18:2937–45.CrossRefPubMed
17.
Zurück zum Zitat Shibahara I, Sonoda Y, Saito R, Kanamori M, Yamashita Y, Kumabe T, et al. The expression status of CD133 is associated with the pattern and timing of primary glioblastoma recurrence. Neuro Oncol. 2013;15:1151–9.CrossRefPubMedCentralPubMed Shibahara I, Sonoda Y, Saito R, Kanamori M, Yamashita Y, Kumabe T, et al. The expression status of CD133 is associated with the pattern and timing of primary glioblastoma recurrence. Neuro Oncol. 2013;15:1151–9.CrossRefPubMedCentralPubMed
18.
Zurück zum Zitat Shin JH, Lee YS, Hong YK, Kang CS. Correlation between the prognostic value and the expression of the stem cell marker CD133 and isocitrate dehydrogenase1 in glioblastomas. J Neurooncol. 2013;115:333–41.CrossRefPubMed Shin JH, Lee YS, Hong YK, Kang CS. Correlation between the prognostic value and the expression of the stem cell marker CD133 and isocitrate dehydrogenase1 in glioblastomas. J Neurooncol. 2013;115:333–41.CrossRefPubMed
19.
Zurück zum Zitat Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008;14:123–9.CrossRefPubMed Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008;14:123–9.CrossRefPubMed
20.
Zurück zum Zitat Dahlrot RH, Hansen S, Jensen SS, Schroder HD, Hjelmborg J, Kristensen BW. Clinical value of CD133 and nestin in patients with glioma: a population-based study. Int J Clin Exp Pathol. 2014;7:3739–51.PubMedCentralPubMed Dahlrot RH, Hansen S, Jensen SS, Schroder HD, Hjelmborg J, Kristensen BW. Clinical value of CD133 and nestin in patients with glioma: a population-based study. Int J Clin Exp Pathol. 2014;7:3739–51.PubMedCentralPubMed
21.
Zurück zum Zitat Hatanpaa KJ, Hu T, Vemireddy V, Foong C, Raisanen JM, Oliver D, et al. High expression of the stem cell marker nestin is an adverse prognostic factor in WHO grade II–III astrocytomas and oligoastrocytomas. J Neurooncol. 2014;117:183–9.CrossRefPubMedCentralPubMed Hatanpaa KJ, Hu T, Vemireddy V, Foong C, Raisanen JM, Oliver D, et al. High expression of the stem cell marker nestin is an adverse prognostic factor in WHO grade II–III astrocytomas and oligoastrocytomas. J Neurooncol. 2014;117:183–9.CrossRefPubMedCentralPubMed
22.
Zurück zum Zitat Milde T, Hielscher T, Witt H, Kool M, Mack SC, Deubzer HE, et al. Nestin expression identifies ependymoma patients with poor outcome. Brain Pathol. 2012;22:848–60.CrossRefPubMed Milde T, Hielscher T, Witt H, Kool M, Mack SC, Deubzer HE, et al. Nestin expression identifies ependymoma patients with poor outcome. Brain Pathol. 2012;22:848–60.CrossRefPubMed
23.
Zurück zum Zitat Wan F, Herold-Mende C, Campos B, Centner FS, Dictus C, Becker N, et al. Association of stem cell-related markers and survival in astrocytic gliomas. Biomarkers. 2011;16:136–43.CrossRefPubMed Wan F, Herold-Mende C, Campos B, Centner FS, Dictus C, Becker N, et al. Association of stem cell-related markers and survival in astrocytic gliomas. Biomarkers. 2011;16:136–43.CrossRefPubMed
24.
Zurück zum Zitat Arai H, Ikota H, Sugawara K, Nobusawa S, Hirato J, Nakazato Y. Nestin expression in brain tumors: its utility for pathological diagnosis and correlation with the prognosis of high-grade gliomas. Brain Tumor Pathol. 2012;29:160–7.CrossRefPubMed Arai H, Ikota H, Sugawara K, Nobusawa S, Hirato J, Nakazato Y. Nestin expression in brain tumors: its utility for pathological diagnosis and correlation with the prognosis of high-grade gliomas. Brain Tumor Pathol. 2012;29:160–7.CrossRefPubMed
25.
Zurück zum Zitat Tomao F, Papa A, Rossi L, Strudel M, Vici P, Lo Russo G, et al. Emerging role of cancer stem cells in the biology and treatment of ovarian cancer: basic knowledge and therapeutic possibilities for an innovative approach. J Exp Clin Cancer Res. 2013;32:48.CrossRefPubMedCentralPubMed Tomao F, Papa A, Rossi L, Strudel M, Vici P, Lo Russo G, et al. Emerging role of cancer stem cells in the biology and treatment of ovarian cancer: basic knowledge and therapeutic possibilities for an innovative approach. J Exp Clin Cancer Res. 2013;32:48.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90:5002–12.PubMed Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90:5002–12.PubMed
27.
Zurück zum Zitat Nosrati A, Naghshvar F, Khanari S. Cancer stem cell markers CD44, CD133 in primary gastric adenocarcinoma. Int J Mol Cell Med. 2014;3:279–86.PubMedCentralPubMed Nosrati A, Naghshvar F, Khanari S. Cancer stem cell markers CD44, CD133 in primary gastric adenocarcinoma. Int J Mol Cell Med. 2014;3:279–86.PubMedCentralPubMed
28.
Zurück zum Zitat Di C, Zhao Y. Multiple drug resistance due to resistance to stem cells and stem cell treatment progress in cancer (Review). Exp Ther Med. 2015;9:289–93.PubMedCentralPubMed Di C, Zhao Y. Multiple drug resistance due to resistance to stem cells and stem cell treatment progress in cancer (Review). Exp Ther Med. 2015;9:289–93.PubMedCentralPubMed
29.
Zurück zum Zitat Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, et al. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest. 2014;32:470–95.CrossRefPubMed Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, et al. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest. 2014;32:470–95.CrossRefPubMed
30.
Zurück zum Zitat Kong BH, Moon JH, Huh YM, Shim JK, Lee JH, Kim EH, et al. Prognostic value of glioma cancer stem cell isolation in survival of primary glioblastoma patients. Stem Cells Int. 2014;2014:838950.CrossRefPubMedCentralPubMed Kong BH, Moon JH, Huh YM, Shim JK, Lee JH, Kim EH, et al. Prognostic value of glioma cancer stem cell isolation in survival of primary glioblastoma patients. Stem Cells Int. 2014;2014:838950.CrossRefPubMedCentralPubMed
31.
Zurück zum Zitat Campos B, Herold-Mende CC. Insight into the complex regulation of CD133 in glioma. Int J Cancer. 2011;128:501–10.CrossRefPubMed Campos B, Herold-Mende CC. Insight into the complex regulation of CD133 in glioma. Int J Cancer. 2011;128:501–10.CrossRefPubMed
32.
Zurück zum Zitat Jin X, Jung JE, Beck S, Kim H. Cell surface Nestin is a biomarker for glioma stem cells. Biochem Biophys Res Commun. 2013;433:496–501.CrossRefPubMed Jin X, Jung JE, Beck S, Kim H. Cell surface Nestin is a biomarker for glioma stem cells. Biochem Biophys Res Commun. 2013;433:496–501.CrossRefPubMed
33.
Zurück zum Zitat Fan H, Guo H, Zhang IY, Liu B, Luan L, Xu S, et al. The different HMGA1 expression of total population of glioblastoma cell line U251 and glioma stem cells isolated from U251. Brain Res. 2011;1384:9–14.CrossRefPubMed Fan H, Guo H, Zhang IY, Liu B, Luan L, Xu S, et al. The different HMGA1 expression of total population of glioblastoma cell line U251 and glioma stem cells isolated from U251. Brain Res. 2011;1384:9–14.CrossRefPubMed
34.
Zurück zum Zitat Xu M, Yao Y, Hua W, Wu Z, Zhong P, Mao Y, et al. Mouse glioma immunotherapy mediated by A2B5+ GL261 cell lysate-pulsed dendritic cells. J Neurooncol. 2014;116:497–504.CrossRefPubMed Xu M, Yao Y, Hua W, Wu Z, Zhong P, Mao Y, et al. Mouse glioma immunotherapy mediated by A2B5+ GL261 cell lysate-pulsed dendritic cells. J Neurooncol. 2014;116:497–504.CrossRefPubMed
Metadaten
Titel
Do relevant markers of cancer stem cells CD133 and Nestin indicate a poor prognosis in glioma patients? A systematic review and meta-analysis
verfasst von
Bin Wu
Caixing Sun
Fang Feng
Minghua Ge
Liang Xia
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2015
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-015-0163-4

Weitere Artikel der Ausgabe 1/2015

Journal of Experimental & Clinical Cancer Research 1/2015 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.