Skip to main content
Erschienen in: World Journal of Surgical Oncology 1/2015

Open Access 01.12.2015 | Research

Survivin and angiotensin-converting enzyme polymorphisms with risk of colorectal cancer: a systematic review and meta-analysis

verfasst von: Xile Zhou, Caizhao Lin

Erschienen in: World Journal of Surgical Oncology | Ausgabe 1/2015

Abstract

Background

Colorectal cancer (CRC) is the most common cause of cancer death worldwide. Numerous studies have identified the roles of survivin −31 G/C and angiotensin-converting enzyme insertion/deletion (ACE I/D) polymorphisms in CRC risk; however, the results remain inconclusive. This study was to investigate associations between these two polymorphisms and CRC susceptibility.

Methods

A comprehensive literature search was conducted to collect relevant case–control studies published between 2000 and 2014. The extracted data were statistically analyzed, and the odds ratios (ORs) with 95% confidence intervals (CIs) were employed to estimate the strength of association.

Results

A total of 11 studies were included in the meta-analysis. For survivin G/C polymorphism, six articles reported 1,840 cases and 1,804 controls. Overall, we found the frequency of C allele is higher in CRC cases than that in the healthy controls (57.2% vs. 48.0%), and C allele significantly increased the risk of CRC compared to G allele in allele model (OR = 1.46, 95% CI = 1.33–1.60, P < 0.00001). This association was also found in other genetic models (P < 0.00001). Stratified analysis by ethnicity showed significant association in each genetic model among the Asian population. For ACE I/D polymorphism, five studies included 758 cases and 6,755 controls. No significant association was found in any genetic models.

Conclusions

Our results showed that survivin −31 G/C polymorphism might contribute to risk of CRC, especially in the Asian populations. However, the ACE I/D polymorphism is not a genetic factor concerning the risk for CRC. More studies with larger sample sizes are required in the future.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

XLZ and CZL conceived and designed the study, performed the experiments, contributed the material/analysis tools, and revised the statistical analyses and paper writing. Both authors read and approved the final manuscript.

Background

Colorectal cancer (CRC) is one of the three most common cancers in the world and is a major contributor to cancer-related death [1]. Each year, a global incidence exceeding 1.2 million new cases emerge and 600,000 deaths occur [2]. Its incidence rates continue to increase in economically transitioning countries. According to colorectal cancer statistics, an estimated 71,830 men and 65,000 women will be diagnosed with CRC, and 26,270 men and 24,040 women will die of this disease in 2014 [3]. The etiological factors and pathogenetic mechanisms underlying CRC development appear to be complex and heterogeneous. In the last two decades, studies have demonstrated that CRC cells undergo major epigenetic alterations [4]. Among which, genetic variants in oncogenes have been extensively investigated as the essential role in cancer etiology [5].
The survivin gene, located in chromosome 17q25, is the smallest member of the inhibitor of apoptosis (IAP) gene family [6]. It is a multifunctional protein and is required to preserve tissue or organism viability [7]. Survivin is expressed in many human cancers and involved in the regulation of cell division and survival [8]. The survivin −31 G/C polymorphism may modulate susceptibility to cancer by influencing the expression of survivin. A recent study has supported a role of survivin in colorectal carcinogenesis, while the −31 G/C polymorphism may constitute a marker of survival [9].
The angiotensin-converting enzyme (ACE), a major component of the renin-angiotensin system (RAS), plays a crucial role in the regulation of circulatory homeostasis such as blood pressure and serum electrolytes [10]. It is located on human chromosome 17q23 and has been involved in the pathogenesis of human cancers [11,12]. Epidemiologic studies have indicated that inhibition of ACE activity could suppress tumor growth and angiogenesis, decreasing the risk and mortality rate [13]. Recent research showed that long-term/high-dose exposure to ACE inhibitor may decrease the incidence of CRC [14]. ACE variants might affect its activity. Among which, ACE I/D (rs4646994) polymorphism in intron 16 of this gene, based on insertion (I) or deletion (D) of a 287-bp Alu sequence, was the most widely studied and leaded to a change in the plasma ACE level. The carriers of D allele were shown in higher ACE activity.
Numerous studies have shown the association of these genetic polymorphisms with CRC [9,15-25]. However, the results remained inconsistent. In the present study, we performed meta-analyses to evaluate and summarize the contribution of the two polymorphisms to CRC susceptibility in different populations.

Methods

A comprehensive literature search was conducted using the online electronic database of Embase, Medline, PubMed, CNKI (China National Knowledge Infrastructure), and Wanfang. We retrieved the relevant articles published between January 2000 and March 2014 using the following terms: “colorectal cancer or colorectal carcinoma”, “angiotensin-converting enzyme or ACE”, “survivin”, and “polymorphism or variant or mutation” as well as their combinations. The references of retrieved articles were searched manually. When the same authors or laboratory reported the issue on the same group of people, only full-text articles of the most recent studies were included.

Study selection

The inclusion criteria were as follows: 1) case–control or cohort studies, 2) evaluating the contribution of survivin −31 G/C and ACE I/D polymorphisms with CRC risk, 3) the results presented in odds ratio (OR) with its 95% confidence interval (CI), and 4) genotype distributions in the cases and controls were available to extract.

Data extraction

Two investigators independently assessed the data from the included studies. Any disagreement was discussed with a third expert to reach a final consensus. The following information was extracted: the name of first author, publication year, country, ethnicity, sample size, genotyping method, and the genotype frequencies in the cancer cases and controls.

Statistical analysis

The overall effect was measured by ORs with its 95% CI. The Z test was employed to determine the significance of the pooled ORs, and a P value less than 0.05 was considered statistically significant. The per-allele model (C vs. G for survivin, D vs. I for ACE), dominant model (GC + CC vs. GG for survivin, ID + DD vs. II for ACE), recessive model (CC vs. GC + GG survivin, DD vs. ID + II for ACE), and additive model (CC vs. GG for survivin, DD vs. II for ACE) were examined to assess these association. The I 2 test and the Q statistic test were used to assess the between-study heterogeneity. The Mantel-Haenszel (M-H) fixed-effects model is used when the P value is more than 0.10 for the Q test and less than 50% for I 2; otherwise, the random-effects model is used. The publication bias was assessed by visual funnel plot inspection. Review Manager (version 5.2, The Cochrane Collaboration) was used to conduct the statistical analyses. All the tests were two sided.

Results

Study selection and characteristics

The initial search identified 284 references. Of those, 107 records were excluded for duplication and 177 articles were judged potentially relevant. Following the title and abstract screening, 40 full-text articles met inclusion criteria. Overall, a total of 11 studies (eight in English and three in Chinese) were finally included in this review. Figure 1 showed the study flow.
Of the 11 case–control studies, there were five studies of Asians and six of Caucasian. For ACE I/D polymorphism, five studies containing 758 cases and 6,755 controls were included. For survivin G/C polymorphism, six articles reported 1,840 cases and 1,804 controls. Table 1 summarized the main characteristics of included studies. Table 2 showed the information of genotypes and alleles for each case–control study.
Table 1
Main characteristic of the eligible studies
First author
Year
Country
Ethnicity
Cases
Controls
Genotyping method
Survivin (−31 G/C)
Gazouli [16]
2009
Greece
Caucasian
312
362
PCR-RFLP
Huang [17]
2010
China
Asian
702
711
PCR-RFLP
Antonacopoulou [9]
2011
Greece
Caucasian
163
132
Taqman
Duan [18]
2012
China
Asian
206
129
PCR-RFLP
Liu [19]
2012
China
Asian
182
200
PCR-LDR
Li [20]
2013
China
Asian
275
270
PCR-RFLP
ACE (I/D)
Nikiteas [21]
2007
Greece
Caucasian
92
102
PCR
Rocken [22]
2007
Germany
Caucasian
141
189
PCR
Van der [23]
2008
Netherlands
Caucasian
176
6,015
PCR
Toma [24]
2008
Romanian
Caucasian
108
150
PCR
Liu SY [25]
2011
China
Asian
241
299
PCR-PAGE
Table 2
Distribution of genotypes in the individual studies
 
Cases
Control
Survivin
GG
GC
CC
G
C
GG
GC
CC
G
C
Gazouli [16]
68
131
113
267
357
123
163
76
409
315
Huang [17]
144
302
256
590
814
180
345
186
705
717
Antonacopoulou [9]
63
84
16
210
116
66
50
16
182
82
Duan [18]
31
92
83
154
258
32
66
31
130
128
Liu [19]
36
76
70
148
216
54
93
53
201
199
Li [20]
42
123
110
207
343
55
138
77
248
292
ACE
II
ID
DD
I
D
II
ID
DD
I
D
Nikiteas [21]
15
27
50
57
127
6
44
52
56
148
Rocken [22]
37
69
35
143
139
41
95
53
177
201
Van derv [23]
34
97
45
165
187
1,332
3,006
1,677
5,670
6,360
Toma [24]
25
50
33
100
116
30
73
47
133
167
Liu SY [25]
71
138
32
280
202
95
158
46
348
250

Association of survivin G/C polymorphism and CRC risk

Table 3 listed the results of allele and genotypes of survivin polymorphism in this meta-analysis. The between-study heterogeneity was not significant and the fixed-effect model was employed. Overall, the frequency of C allele was higher in CRC cases than that in the healthy controls (57.2% vs. 48.0%). As shown in Figure 2, we found that C allele significantly increased the risk of CRC compared to G allele in the allele model (OR = 1.46, 95% CI = 1.33–1.60, P < 0.00001). This significant association was also found in other genetic models (CC vs. GG: OR = 1.95, 95% CI = 1.62–2.35, P < 0.00001; CC + GC vs. GG: OR = 1.51, 95% CI = 1.29–1.76, P < 0.00001; CC vs. GC + GG: OR = 1.72, 95% CI = 1.49–1.99, P < 0.00001) as shown in Figures 3 and 4. When evaluating the effect of the polymorphism by ethnicity, we found a significant association in all genetic models among the Asian population (C vs. G: OR = 1.42, 95% CI = 1.27–1.58, P < 0.00001; CC vs. GG: OR = 1.88, 95% CI = 1.52–2.33, P < 0.00001; CC + GC vs. GG: OR = 1.41, 95% CI = 1.17–1.70, P = 0.0003; CC vs. GC + GG: OR = 1.70, 95% CI = 1.44–2.01, P < 0.00001) in a fixed-effect model. Among the Caucasian population, Figure 5 showed that C allele carrier was significantly associated with the increased risk of CRC compared to healthy control groups (CC + GC vs. GG: OR = 1.75, 95% CI = 1.33–2.31, P < 0.0001). No relationship was found in other genetic models (P > 0.05).
Table 3
Meta-analysis of survivin −31 G/C polymorphism in CRC by ethnicity analysis
 
Total
Asian
Caucasian
OR (95% CI)
P
Ph
OR (95% CI)
P
Ph
OR (95% CI)
P
Ph
C vs. G
1.46 (1.33, 1.60)
<0.00001
0.37/7%
1.42 (1.27, 1.58)
<0.00001
0.64/0%
1.50 (1.07, 2.10)
0.02
0.09/64%
CC vs. GG
1.95 (1.62, 2.35)
<0.00001
0.25/25%
1.88 (1.52, 2.33)
<0.00001
0.62/0%
1.78 (0.71, 4.45)
0.22
0.04/77%
CC + GC vs. GG
1.51 (1.29, 1.76)
<0.00001
0.67/0%
1.41 (1.17, 1.70)
0.0003
0.72/0%
1.75 (1.33, 2.31)
<0.0001
0.61/0%
CC vs. GC + GG
1.72 (1.49, 1.99)
<0.00001
0.23/28%
1.70 (1.44, 2.01)
<0.00001
0.80/0%
1.37 (0.52, 3.62)
0.52
0.02/83%

Association of ACE I/D polymorphism and CRC risk

Table 4 displayed the summary of all genetic comparisons between ACE I/D polymorphism and CRC risk. The I 2 was less than 50% and the P value was greater than 0.01, suggesting no heterogeneity was present. As shown in Figure 6, the result suggested that the variant D allele did not have a significant increased risk of CRC compared with those individuals without D allele (D vs. I: OR = 0.96, 95% CI = 0.84–1.08, P = 0.48). No significant association was found in other genetic models (DD vs. II: OR = 0.86, 95% CI = 0.66–1.12, P = 0.25; DD + ID vs. II: OR = 0.97, 95% CI = 0.79–1.19, P = 0.77; DD vs. ID + II: OR = 0.91, 95% CI = 0.74–1.12, P = 0.39).
Table 4
Meta-analysis of ACE I/D polymorphism in CRC
Genotype
OR (95% CI)
P
Ph
I 2 (%)
Model
D vs. I
0.96 (0.84, 1.08)
0.48
0.87
0
Fixed
DD vs. II
0.86 (0.66, 1.12)
0.25
0.48
0
Fixed
DD + ID vs. II
0.97 (0.79, 1.19)
0.77
0.12
46
Fixed
DD vs. ID + II
0.91 (0.74, 1.12)
0.39
0.93
0
Fixed

Sensitivity analysis and publication bias

A single study included in the meta-analysis was deleted each time to reflect the influence of the individual data set to the pooled ORs. The corresponding pooled ORs were not materially changed, which confirmed the stability of our overall result. The shape of funnel plots did not reveal any evidence of funnel plot asymmetry (Figure 7).

Discussion

CRC is a major cause of morbidity and mortality worldwide. Although surgical resection remains a curative option for CRC treatment, most patients with CRC are not suited for surgical operations because of the advanced stage of the lesions when diagnosed. Thus, there is an urgent need to identify newer biomarkers to evaluate the severity of this disease, predict the prognosis of patients, and target the pathogenic genes.
Recently, survivin has been reported to be overexpressed in almost all human tumors. It is considered as a tumor-associated antigen for immune-based clinical approaches in CRC [26]. Researchers have suggested that survivin is implicated in the progression of dysplasia to neoplasia in the colon [27]. A significant role of survivin in CRC progression and recurrence of colon cancer liver metastases has been previously demonstrated [28]. The mechanisms of the survivin re-expression in malignancies are still unclear due to the complexity of its regulation. Genetic variations located in the promoter of the survivin gene have impact on the protein expression and consequently contributed to the genetic susceptibility to cancers. The −31 G/C polymorphism is the most extensively studied variant located in the promoter region of survivin, and the C allele has been identified to be associated with a higher transcription activity of survivin [29]. This meta-analysis showed that the survivin −31 G/C polymorphism was associated with increased CRC risk in total populations as well as in Asians, while no association was found in Caucasian except the dominant model. Our result is in agreement with the study conducted by Qin et al., which identified that this polymorphism is associated with elevated cancer risk [30].
Survivin acts as an antagonist of apoptotic cell death and functions as a regulator of mitosis [31,32]. It is a potentially interesting prognostic marker, implicating a shift from adenoma with low dysplasia to high dysplasia during human colorectal tumorigenesis [27]. Survivin overexpression was related with the increase of invasion and the metastasis of CRC [33]. A study conducted by Choi et al. suggested that the nuclear expression of survivin might be associated with the metastasis of CRC to the liver [34]. Survivin can be regulated by or cooperated with other genes. The stimulation of survivin expression by TCF/β catenin might contribute to the molecular pathogenesis of CRC [35]. The positive expression of survivin protein, directly correlated to that of Bcl-2, acted on different stages of apoptosis to promote jointly the development of CRC in a synergistic way [36]. The combined expression levels of Aldh1, survivin, and EpCAM as strong independent prognostic factors for survival and tumor recurrence in colon cancer patients reflect tumor aggressiveness [37]. Furthermore, survivin is also a therapeutic target in cancers [38]. Survivin expression increases during the normal mucosa-adenoma-carcinoma sequence and is maintained throughout the progression of disease, which strengthens its appeal as a therapeutic target [39]. The expression of survivin may play a role in identifying a subgroup of patients who could benefit from a targeted therapy against survivin in CRC [40].
Much evidence indicates that ACE associated with the pathology of carcinomas. It is differentially expressed in several malignancies. ACE is a monomeric glycoprotein that is distributed in many tissues and biological fluids [41]. ACE can influence tumor cell migration, proliferation, metastatic behavior, and angiogenesis [42]. The expression of ACE is upregulated in several cancers with functions of angiogenesis and tumor cell growth [43].
Previous meta-analysis showed that the ACE I/D polymorphism is associated with hepatocellular carcinoma (HCC), indicating that this polymorphism contributes to HCC progression in the Chinese population [44]. Recent analysis suggested that the ACE I/D polymorphism might not be a common risk factor for overall cancer susceptibility [43]. Our results did not show any association between ACE I/D polymorphism and CRC in each genetic models (P > 0.05). This is in accordance with meta-analysis conducted by Liu et al. and Zhang et al. [12,45].
Several limitations were presented in this meta-analysis. Firstly, the number of included studies in the subgroup analysis was small which may have a relatively lower power. Secondly, other covariates such as age, sex, and smoking condition should be considered if they are available in each individual study to obtain a more precise result. Thirdly, the gene-gene interaction which is important in developing complex diseases should also be included.

Conclusions

In conclusion, the results from the present meta-analysis suggest that the survivin −31 G/C polymorphism might be correlated with an increased risk of CRC, indicating it may serve as a biomarker of disease progression. However, ACE I/D polymorphism is not associated with CRC risk. Further large and well-designed studies in various populations are needed to confirm our results. Moreover, the studies of gene-gene and gene-environment interactions between these polymorphisms and CRC risk should also be performed and considered.

Acknowledgements

We thank Jinhai Wang in the Department of Colorectal Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University for the data analysis in this study.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

XLZ and CZL conceived and designed the study, performed the experiments, contributed the material/analysis tools, and revised the statistical analyses and paper writing. Both authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Wasserman M, Baxter N, Rosen B, Burnstein M, Halverson A. Systematic review of internet patient information on colorectal cancer surgery. Dis Colon Rectum. 2014;57(1):64–9.CrossRefPubMed Wasserman M, Baxter N, Rosen B, Burnstein M, Halverson A. Systematic review of internet patient information on colorectal cancer surgery. Dis Colon Rectum. 2014;57(1):64–9.CrossRefPubMed
2.
Zurück zum Zitat Mäkinen MJ. Serrated polyps and colorectal cancer risk. Colorectal Cancer. 2014;3(1):77–91.CrossRef Mäkinen MJ. Serrated polyps and colorectal cancer risk. Colorectal Cancer. 2014;3(1):77–91.CrossRef
3.
Zurück zum Zitat Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64(2):104–17.CrossRefPubMed Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64(2):104–17.CrossRefPubMed
4.
Zurück zum Zitat Derks S, van Engeland M. Epigenetic disturbances in colorectal cancer. Epigenetic Therapy of Cancer. Springer; 2014. p. 283–98. Derks S, van Engeland M. Epigenetic disturbances in colorectal cancer. Epigenetic Therapy of Cancer. Springer; 2014. p. 283–98.
5.
7.
Zurück zum Zitat Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61–70.CrossRefPubMed Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61–70.CrossRefPubMed
8.
Zurück zum Zitat Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med. 1997;3(8):917–21.CrossRefPubMed Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med. 1997;3(8):917–21.CrossRefPubMed
9.
Zurück zum Zitat Antonacopoulou AG, Floratou K, Bravou V, Kottorou A, Dimitrakopoulos F-I, Marousi S, et al. The survivin-31 snp in human colorectal cancer correlates with survivin splice variant expression and improved overall survival. Cell Oncol. 2011;34(4):381–91.CrossRef Antonacopoulou AG, Floratou K, Bravou V, Kottorou A, Dimitrakopoulos F-I, Marousi S, et al. The survivin-31 snp in human colorectal cancer correlates with survivin splice variant expression and improved overall survival. Cell Oncol. 2011;34(4):381–91.CrossRef
10.
Zurück zum Zitat Krege JH, Kim H-S, Moyer JS, Jennette JC, Peng L, Hiller SK, et al. Angiotensin-converting enzyme gene mutations, blood pressures, and cardiovascular homeostasis. Hypertension. 1997;29(1):150–7.CrossRefPubMed Krege JH, Kim H-S, Moyer JS, Jennette JC, Peng L, Hiller SK, et al. Angiotensin-converting enzyme gene mutations, blood pressures, and cardiovascular homeostasis. Hypertension. 1997;29(1):150–7.CrossRefPubMed
11.
Zurück zum Zitat Feng Y, Wan H, Liu J, Zhang R, Ma Q, Han B, et al. The angiotensin-converting enzyme 2 in tumor growth and tumor-associated angiogenesis in non-small cell lung cancer. Oncol Rep. 2010;23(4):941–8.PubMed Feng Y, Wan H, Liu J, Zhang R, Ma Q, Han B, et al. The angiotensin-converting enzyme 2 in tumor growth and tumor-associated angiogenesis in non-small cell lung cancer. Oncol Rep. 2010;23(4):941–8.PubMed
12.
Zurück zum Zitat Zhang Y, He J, Deng Y, Zhang J, Li X, Xiang Z, et al. The insertion/deletion (I/D) polymorphism in the angiotensin-converting enzyme gene and cancer risk: a meta-analysis. BMC Med Genet. 2011;12(1):159.CrossRefPubMedCentralPubMed Zhang Y, He J, Deng Y, Zhang J, Li X, Xiang Z, et al. The insertion/deletion (I/D) polymorphism in the angiotensin-converting enzyme gene and cancer risk: a meta-analysis. BMC Med Genet. 2011;12(1):159.CrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Mc Menamin ÚC, Murray LJ, Cantwell MM, Hughes CM. Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers in cancer progression and survival: a systematic review. Cancer Causes Control. 2012;23(2):221–30.CrossRefPubMed Mc Menamin ÚC, Murray LJ, Cantwell MM, Hughes CM. Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers in cancer progression and survival: a systematic review. Cancer Causes Control. 2012;23(2):221–30.CrossRefPubMed
14.
Zurück zum Zitat Makar GA, Holmes JH, Yang Y-X. Angiotensin-converting enzyme inhibitor therapy and colorectal cancer risk. J Natl Cancer Inst. 2014;106(2):djt374.CrossRefPubMedCentralPubMed Makar GA, Holmes JH, Yang Y-X. Angiotensin-converting enzyme inhibitor therapy and colorectal cancer risk. J Natl Cancer Inst. 2014;106(2):djt374.CrossRefPubMedCentralPubMed
15.
Zurück zum Zitat Sameer A, Nissar S, Bashir S, Bashir A, Siddiqi M. ACE polymorphism in colorectal cancer patients of Kashmiri population—a short report. Open Colorectal Canc J. 2011;4:1–5.CrossRef Sameer A, Nissar S, Bashir S, Bashir A, Siddiqi M. ACE polymorphism in colorectal cancer patients of Kashmiri population—a short report. Open Colorectal Canc J. 2011;4:1–5.CrossRef
16.
Zurück zum Zitat Gazouli M, Tzanakis N, Rallis G, Theodoropoulos G, Papaconstantinou I, Kostakis A, et al. Survivin −31G/C promoter polymorphism and sporadic colorectal cancer. Int J Colorectal Dis. 2009;24(2):145–50.CrossRefPubMed Gazouli M, Tzanakis N, Rallis G, Theodoropoulos G, Papaconstantinou I, Kostakis A, et al. Survivin −31G/C promoter polymorphism and sporadic colorectal cancer. Int J Colorectal Dis. 2009;24(2):145–50.CrossRefPubMed
17.
Zurück zum Zitat Huang J, Wang J, Wang L, Liu H, Wei Y, Huang M, et al. Association between Survivin Promoter -31C/G Polymorphism and Genetic Susceptibility to Sporadic Colorectal Cancer. J SUN Yat-sen Univ (Med Sci). 2010;31(1):59–63 (in chinese). Huang J, Wang J, Wang L, Liu H, Wei Y, Huang M, et al. Association between Survivin Promoter -31C/G Polymorphism and Genetic Susceptibility to Sporadic Colorectal Cancer. J SUN Yat-sen Univ (Med Sci). 2010;31(1):59–63 (in chinese).
18.
Zurück zum Zitat Duan F. The Study on the Relationship between Survivin promoter SNP and the Colorectal Cancer. Master's thesis. Lu Zhou medical college, 2012. (in chinese) Duan F. The Study on the Relationship between Survivin promoter SNP and the Colorectal Cancer. Master's thesis. Lu Zhou medical college, 2012. (in chinese)
19.
Zurück zum Zitat Liu Y. The Association of Survivin Polymorphism with the Risk of Colorectal Cancer. Master's thesis. Hebei medical college, 2012. (in chinese) Liu Y. The Association of Survivin Polymorphism with the Risk of Colorectal Cancer. Master's thesis. Hebei medical college, 2012. (in chinese)
20.
Zurück zum Zitat Li XB, Li SN, Yang ZH, Cao L, Duan FL, Sun XW. Polymorphisms of survivin and its protein expression are associated with colorectal cancer susceptibility in Chinese population. DNA Cell Biol. 2013;32(5):236–42.CrossRefPubMedCentralPubMed Li XB, Li SN, Yang ZH, Cao L, Duan FL, Sun XW. Polymorphisms of survivin and its protein expression are associated with colorectal cancer susceptibility in Chinese population. DNA Cell Biol. 2013;32(5):236–42.CrossRefPubMedCentralPubMed
21.
Zurück zum Zitat Nikiteas N, Tsigris C, Chatzitheofylaktou A, Yannopoulos A. No association with risk for colorectal cancer of the insertion/deletion polymorphism which affects levels of angiotensin-converting enzyme. In Vivo. 2007;21(6):1065–68.PubMed Nikiteas N, Tsigris C, Chatzitheofylaktou A, Yannopoulos A. No association with risk for colorectal cancer of the insertion/deletion polymorphism which affects levels of angiotensin-converting enzyme. In Vivo. 2007;21(6):1065–68.PubMed
22.
Zurück zum Zitat Rocken C, Neumann K, Carl-McGrath S, Lage H, Ebert MP, Dierkes J, et al. The gene polymorphism of the angiotensin I-converting enzyme correlates with tumor size and patient survival in colorectal cancer patients. Neoplasia. 2007;9(9):716–22.CrossRefPubMedCentralPubMed Rocken C, Neumann K, Carl-McGrath S, Lage H, Ebert MP, Dierkes J, et al. The gene polymorphism of the angiotensin I-converting enzyme correlates with tumor size and patient survival in colorectal cancer patients. Neoplasia. 2007;9(9):716–22.CrossRefPubMedCentralPubMed
23.
Zurück zum Zitat Van der Knaap R, Siemes C, Coebergh JW, Van Duijn CM, Hofman A, Stricker BH. Renin-angiotensin system inhibitors, angiotensin I-converting enzyme gene insertion/deletion polymorphism, and cancer: the Rotterdam Study. Cancer. 2008;112(4):748–57.CrossRefPubMed Van der Knaap R, Siemes C, Coebergh JW, Van Duijn CM, Hofman A, Stricker BH. Renin-angiotensin system inhibitors, angiotensin I-converting enzyme gene insertion/deletion polymorphism, and cancer: the Rotterdam Study. Cancer. 2008;112(4):748–57.CrossRefPubMed
24.
Zurück zum Zitat Toma M, Cimponeriu D, Apostol P, Stavarachi M, Cojocaru M, Belusica L, et al. Lack of association between ACE ID polymorphism and colorectal cancer in Romanian patients. Chirurgia (Bucharest, Romania: 1990). 2008;104(5):553–56. Toma M, Cimponeriu D, Apostol P, Stavarachi M, Cojocaru M, Belusica L, et al. Lack of association between ACE ID polymorphism and colorectal cancer in Romanian patients. Chirurgia (Bucharest, Romania: 1990). 2008;104(5):553–56.
25.
Zurück zum Zitat Liu SY, Sima X, Wang CH, Gao M. The association between ACE polymorphism and risk of colorectal cancer in a Chinese population. Clin Biochem. 2011;44(14):1223–26.CrossRefPubMed Liu SY, Sima X, Wang CH, Gao M. The association between ACE polymorphism and risk of colorectal cancer in a Chinese population. Clin Biochem. 2011;44(14):1223–26.CrossRefPubMed
26.
Zurück zum Zitat Casati C, Dalerba P, Rivoltini L, Gallino G, Deho P, Rini F, et al. The apoptosis inhibitor protein survivin induces tumor-specific CD8+ and CD4+ T cells in colorectal cancer patients. Cancer Res. 2003;63(15):4507–15.PubMed Casati C, Dalerba P, Rivoltini L, Gallino G, Deho P, Rini F, et al. The apoptosis inhibitor protein survivin induces tumor-specific CD8+ and CD4+ T cells in colorectal cancer patients. Cancer Res. 2003;63(15):4507–15.PubMed
27.
Zurück zum Zitat Kawasaki H, Toyoda M, Shinohara H, Okuda J, Watanabe I, Yamamoto T, et al. Expression of survivin correlates with apoptosis, proliferation, and angiogenesis during human colorectal tumorigenesis. Cancer. 2001;91(11):2026–32.CrossRefPubMed Kawasaki H, Toyoda M, Shinohara H, Okuda J, Watanabe I, Yamamoto T, et al. Expression of survivin correlates with apoptosis, proliferation, and angiogenesis during human colorectal tumorigenesis. Cancer. 2001;91(11):2026–32.CrossRefPubMed
28.
Zurück zum Zitat Fan L-F, Dong W-G, Jiang C-Q, Qian Q, Yu Q-F. Role of hypoxia-inducible factor-1α and survivin in colorectal carcinoma progression. Int J Color Dis. 2008;23(11):1057–64.CrossRef Fan L-F, Dong W-G, Jiang C-Q, Qian Q, Yu Q-F. Role of hypoxia-inducible factor-1α and survivin in colorectal carcinoma progression. Int J Color Dis. 2008;23(11):1057–64.CrossRef
29.
Zurück zum Zitat Jang JS, Kim KM, Kang KH, Choi JE, Lee WK, Kim CH, et al. Polymorphisms in the < i > survivin </i > gene and the risk of lung cancer. Lung Cancer. 2008;60(1):31–9.CrossRefPubMed Jang JS, Kim KM, Kang KH, Choi JE, Lee WK, Kim CH, et al. Polymorphisms in the < i > survivin </i > gene and the risk of lung cancer. Lung Cancer. 2008;60(1):31–9.CrossRefPubMed
30.
Zurück zum Zitat Qin Q, Zhang C, Zhu H, Yang X, Xu L, Liu J, et al. Association between survivin-31G > C polymorphism and cancer risk: meta-analysis of 29 studies. J Cancer Res Clin Oncol. 2014;140(2):179–88.CrossRefPubMed Qin Q, Zhang C, Zhu H, Yang X, Xu L, Liu J, et al. Association between survivin-31G > C polymorphism and cancer risk: meta-analysis of 29 studies. J Cancer Res Clin Oncol. 2014;140(2):179–88.CrossRefPubMed
31.
Zurück zum Zitat Skoufias DA, Mollinari C, Lacroix FB, Margolis RL. Human survivin is a kinetochore-associated passenger protein. J Cell Biol. 2000;151(7):1575–82.CrossRefPubMedCentralPubMed Skoufias DA, Mollinari C, Lacroix FB, Margolis RL. Human survivin is a kinetochore-associated passenger protein. J Cell Biol. 2000;151(7):1575–82.CrossRefPubMedCentralPubMed
32.
Zurück zum Zitat Dohi T, Okada K, Xia F, Wilford CE, Samuel T, Welsh K, et al. An IAP-IAP complex inhibits apoptosis. J Biol Chem. 2004;279(33):34087–90.CrossRefPubMed Dohi T, Okada K, Xia F, Wilford CE, Samuel T, Welsh K, et al. An IAP-IAP complex inhibits apoptosis. J Biol Chem. 2004;279(33):34087–90.CrossRefPubMed
33.
Zurück zum Zitat Chu XY, Chen LB, Wang JH, Su QS, Yang JR, Lin Y, et al. Overexpression of survivin is correlated with increased invasion and metastasis of colorectal cancer. J Surg Oncol. 2012;105(6):520–8.CrossRefPubMed Chu XY, Chen LB, Wang JH, Su QS, Yang JR, Lin Y, et al. Overexpression of survivin is correlated with increased invasion and metastasis of colorectal cancer. J Surg Oncol. 2012;105(6):520–8.CrossRefPubMed
34.
Zurück zum Zitat Choi J, Chang H. The expression of MAGE and SSX, and correlation of COX2, VEGF, and survivin in colorectal cancer. Anticancer Res. 2012;32(2):559–64.PubMed Choi J, Chang H. The expression of MAGE and SSX, and correlation of COX2, VEGF, and survivin in colorectal cancer. Anticancer Res. 2012;32(2):559–64.PubMed
35.
Zurück zum Zitat Kim PJ, Plescia J, Clevers H, Fearon ER, Altieri DC. Survivin and molecular pathogenesis of colorectal cancer. Lancet. 2003;362(9379):205–9.CrossRefPubMed Kim PJ, Plescia J, Clevers H, Fearon ER, Altieri DC. Survivin and molecular pathogenesis of colorectal cancer. Lancet. 2003;362(9379):205–9.CrossRefPubMed
36.
Zurück zum Zitat Ma Y, Wang HS. Correlations of Bcl-2 and survivin gene protein expressions in colorectal cancer. Appl Mech Mater. 2013;423:362–5.CrossRef Ma Y, Wang HS. Correlations of Bcl-2 and survivin gene protein expressions in colorectal cancer. Appl Mech Mater. 2013;423:362–5.CrossRef
37.
Zurück zum Zitat Goossens-Beumer I, Zeestraten E, Benard A, Christen T, Reimers M, Keijzer R, et al. Clinical prognostic value of combined analysis of Aldh1, survivin, and EpCAM expression in colorectal cancer. Br J Cancer. 2014;110(12):2935–44.CrossRefPubMed Goossens-Beumer I, Zeestraten E, Benard A, Christen T, Reimers M, Keijzer R, et al. Clinical prognostic value of combined analysis of Aldh1, survivin, and EpCAM expression in colorectal cancer. Br J Cancer. 2014;110(12):2935–44.CrossRefPubMed
39.
Zurück zum Zitat Hernandez JM, Farma JM, Coppola D, Hakam A, Fulp WJ, Chen D-T, et al. Expression of the antiapoptotic protein survivin in colon cancer. Clin Colorectal Cancer. 2011;10(3):188–93.CrossRefPubMed Hernandez JM, Farma JM, Coppola D, Hakam A, Fulp WJ, Chen D-T, et al. Expression of the antiapoptotic protein survivin in colon cancer. Clin Colorectal Cancer. 2011;10(3):188–93.CrossRefPubMed
40.
Zurück zum Zitat Krieg A, Werner TA, Verde PE, Stoecklein NH, Knoefel WT. Prognostic and clinicopathological significance of survivin in colorectal cancer: a meta-analysis. PLoS One. 2013;8(6):e65338.CrossRefPubMedCentralPubMed Krieg A, Werner TA, Verde PE, Stoecklein NH, Knoefel WT. Prognostic and clinicopathological significance of survivin in colorectal cancer: a meta-analysis. PLoS One. 2013;8(6):e65338.CrossRefPubMedCentralPubMed
41.
Zurück zum Zitat Guang C, Phillips RD, Jiang B, Milani F. Three key proteases–angiotensin-I-converting enzyme (ACE), ACE2 and renin—within and beyond the renin-angiotensin system. Arch Cardiovasc Dis. 2012;105(6):373–85.CrossRefPubMed Guang C, Phillips RD, Jiang B, Milani F. Three key proteases–angiotensin-I-converting enzyme (ACE), ACE2 and renin—within and beyond the renin-angiotensin system. Arch Cardiovasc Dis. 2012;105(6):373–85.CrossRefPubMed
42.
Zurück zum Zitat Yates CJ, Masuyer G, Schwager SL, Akif M, Sturrock ED, Acharya KR. Molecular and thermodynamic mechanisms of the chloride-dependent human angiotensin-I-converting enzyme (ACE). J Biol Chem. 2014;289(3):1798–814.CrossRefPubMedCentralPubMed Yates CJ, Masuyer G, Schwager SL, Akif M, Sturrock ED, Acharya KR. Molecular and thermodynamic mechanisms of the chloride-dependent human angiotensin-I-converting enzyme (ACE). J Biol Chem. 2014;289(3):1798–814.CrossRefPubMedCentralPubMed
43.
Zurück zum Zitat Xie Y, You C, Chen J. An updated meta-analysis on association between angiotensin I-converting enzyme gene insertion/deletion polymorphism and cancer risk. Tumor Biol. 2014;35(7):6567–79.CrossRef Xie Y, You C, Chen J. An updated meta-analysis on association between angiotensin I-converting enzyme gene insertion/deletion polymorphism and cancer risk. Tumor Biol. 2014;35(7):6567–79.CrossRef
44.
Zurück zum Zitat Yuan F, Zhang L-S, Li H-Y, Liao M, Lv M, Zhang C. Influence of angiotensin I-converting enzyme gene polymorphism on hepatocellular carcinoma risk in China. DNA Cell Biol. 2013;32(5):268–73.CrossRefPubMedCentralPubMed Yuan F, Zhang L-S, Li H-Y, Liao M, Lv M, Zhang C. Influence of angiotensin I-converting enzyme gene polymorphism on hepatocellular carcinoma risk in China. DNA Cell Biol. 2013;32(5):268–73.CrossRefPubMedCentralPubMed
45.
Zurück zum Zitat Liu J-F, Xie H-J, Cheng T-M. Lack of any association between insertion/deletion (I/D) polymorphisms in the angiotensin-converting enzyme gene and digestive system cancer risk: a meta-analysis. Asian Pac J Cancer Prev. 2013;14(12):7271–5.CrossRefPubMed Liu J-F, Xie H-J, Cheng T-M. Lack of any association between insertion/deletion (I/D) polymorphisms in the angiotensin-converting enzyme gene and digestive system cancer risk: a meta-analysis. Asian Pac J Cancer Prev. 2013;14(12):7271–5.CrossRefPubMed
Metadaten
Titel
Survivin and angiotensin-converting enzyme polymorphisms with risk of colorectal cancer: a systematic review and meta-analysis
verfasst von
Xile Zhou
Caizhao Lin
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
World Journal of Surgical Oncology / Ausgabe 1/2015
Elektronische ISSN: 1477-7819
DOI
https://doi.org/10.1186/s12957-015-0461-5

Weitere Artikel der Ausgabe 1/2015

World Journal of Surgical Oncology 1/2015 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.