Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 08 October 2021
Sec. Gastrointestinal Cancers: Colorectal Cancer
This article is part of the Research Topic Detection and Characterization of Gastrointestinal (Early) Cancer View all 30 articles

The Chemokine CXCL7 Is Related to Angiogenesis and Associated With Poor Prognosis in Colorectal Cancer Patients

Longhai Li*&#x;Longhai Li1*†Kai Jiang&#x;Kai Jiang2†Dongpeng Li&#x;Dongpeng Li3†Dongxiao LiDongxiao Li1Zitong FanZitong Fan4Guosheng DaiGuosheng Dai5Sheng TuSheng Tu2Xiangyu Liu*Xiangyu Liu1*Guangyou Wei*Guangyou Wei1*
  • 1Department of Science and Education, Bozhou Hospital of Anhui Medical University, Bozhou, China
  • 2Department of Cardiovascular Medicine, Bozhou Hospital of Anhui Medical University, Bozhou, China
  • 3Department of Pathology, Bozhou Hospital of Anhui Medical University, Bozhou, China
  • 4Department of medicine, Anhui University of Science and Technology, Huainan, China
  • 5Department of Otorhinolaryngology, Bozhou Hospital of Anhui Medical University, Bozhou, China

Objective: The present study was designed to investigate the role of the chemokine CXCL7 in angiogenesis and explore its prognostic value in colorectal cancer (CRC).

Methods: A total of 160 CRC patients who had undergone surgery were included in this study, and staged according to the guidelines of the AJCC, 7th Edition. Expression of CXCL7 and VEGF was detected by immunohistochemical (IHC) staining and divided into high and low expression subgroups. The correlation between CXCL7 and VEGF expression was evaluated by Spearman’s rank-correlation coefficient. Prognosis based on CXCL7 and VEGF was evaluated using the Cox proportional hazards regression model and a nomogram of 5-year overall survival (OS) time.

Results: CXCL7 was highly expressed in tumor tissues (65.63% vs 25.00% in paracancerous tissue, P < 0.001), as was VEGF. CXCL7 and VEGF expression correlated well with N and TNM stage cancers (all P < 0.001). Importantly, CXCL7 was positively correlated with VEGF expression in CRC tissues. CXCL7 was an independent predictor of poor OS of CRC patients (HR = 2.216, 95% CI: 1.069-4.593, P = 0.032), and co-expression of CXCL7 and VEGF of predicted poor OS of 56.96 months.

Conclusion: Expression of CXCL7 correlated with VEGF and was associated with poor clinical outcomes in CRC patients.

Introduction

Colorectal cancer (CRC) is one of the deadliest malignant tumors with over 1.8 million new cases worldwide each year, and contributes to about 10% of tumor-related deaths (1). In fact, 746,000 men and 614,000 women were diagnosed with CRC in developed countries, accounting for 55% of all cancer cases (2). Although the incidence of CRC has been stable and has actually declined in some developed countries owing to the improvements in lifestyle, environmental factors, medical and health conditions (3), new cases of CRC are expected to increase to 2.2 million and deaths to 1.1 million globally by 2030 (4). Therefore, it is vital to control the occurrence and development of CRC to better manage the substantial health burden. The incidence and mortality of CRC are increasing in China due to the aging population and advanced stages of the disease (57). Patients aged 75 years and over constitute a large proportion of new CRC cases (above 40%), and about 45% patients are in advanced stages when first diagnosed (8, 9). The 5-year survival rate is 92% in stage I CRC patients, but drops to 10% when patients are in advanced stage IV (10). Although progress in CRC diagnosis and treatment, including traditional surgical resection, radiotherapy, chemotherapy, and newly targeted immunotherapy, has been achieved, the long-term prognosis, especially in terms of 5-year survival rate, has not improved significantly in recent decades (11).

Angiogenesis is a physiological process whereby new blood vessels are generated from existing vessels, and is regulated by a variety of signaling pathways that dynamically balance pro- and anti-angiogenic responses under normal physiological conditions (12). However, angiogenesis can become abnormally active in response to injuries, the menstrual cycle, and some metabolic diseases (13). Many studies have shown that angiogenesis plays an important role in cancer by providing oxygen, nutrients and other factors in the tumor microenvironment to stimulate tumor growth and accelerate disease progression (14). Numerous factors are involved in angiogenesis, being roughly divided into two types: proangiogenic and antiangiogenic. Vascular endothelial growth factor (VEGF) and its corresponding receptors play an extremely important role in tumor angiogenesis. VEGF is a secreted glycoprotein dimer that promotes angiogenesis by binding to its receptors (15). VEGF exerts its biological functions by activating tyrosine protein kinase. Meanwhile, VEGF can increase capillary permeability, which allows fibrinogen and other proteins to diffuse into the extracellular matrix where they crosslink into a fibrous gel, thus effectively promoting formation of a capillary network (15, 16). VEGF is highly expressed in many tumors where it is the strongest driver of angiogenesis, thus contributing to tumor growth and metastasis (17, 18).

Chemokines are low-molecular-weight signaling proteins (8–12 kDa) that can regulate many biological processes, including cellular immune response, glucose metabolism and angiogenesis (1922). Chemokines play important regulatory roles by binding to their specific receptors (23). Recently, numerous studies have shown that various chemokines play important roles in tumorigenesis, including regulation of proliferation, tumor metastasis, angiogenesis, among others (2427). Chemokines also affect cancer diagnosis and treatment by interacting with tumor cells and the tumor immune environment (28, 29). CXCL7, also called neutrophil activating peptide 2, functions by binding to its receptors CXCR1 and CXCR2, and plays important roles in a variety of tumor processes (3032).

Our previous study showed that CXCL7 is highly expressed in CRC with poor prognosis (30, 33). However, the role of CXCL7 in angiogenesis is unknown. In the current study we probed the function of CXCL7 in angiogenesis and evaluated its prognostic value by analyzing clinical data from a total of 160 CRC patients, including 2 in Stage I, 67 in Stage II, 69 in Stage III and 22 in Stage IV CRC. We assessed the correlation between CXCL7 and VEGF expression and systematically explored the value of CXCL7 for prognosis in CRC.

Materials and Methods

Patient Selection and Tissue Sample Collection

The subjects were enrolled according to the following criteria. (1) Patients were firstly diagnosed in Bozhou Hospital of Anhui Medical University from July 2013 to July 2018. (2) Patients whose condition met the criteria for surgical resection were enrolled in the study. (3) Preoperative clinical diagnosis was consistent with postoperative pathological diagnosis in indicating colorectal cancer. (4) Patients had not received radiotherapy and chemotherapy prior to surgery. (5) The patient’s clinical data was relatively complete. Other terms and selection criteria for healthy participants were described in our previous study (30, 33). These criteria were met by 200 patients. However, 40 people were not eligible during the follow-up. Therefore, 160 patients were included in this study finally (Supplementary Figure 1). Patient data included demographic characteristics and clinical pathology results. All patients were staged according to AJCC (American Joint Committee on Cancer) guidelines, 7th Edition (34). The study was approved by the Institutional Research Ethics Committee of our hospital, and 160 written informed consents were obtained. Tumor and adjacent non-tumor tissues were collected from all 160 patients, then fixed by formalin and embedded in paraffin.

Immunohistochemical Staining Assay

Serial sections of 4 μm thickness were cut from each of the 320 paraffin blocks. IHC staining was performed according to the manufacturer’s instructions using anti-CXCL7 mouse monoclonal antibody (1:200, Abcom Ltd., Cam-bridge, UK) and anti-VEGF rabbit monoclonal antibody (1:300, Abcom Ltd., Cam-bridge, UK). The IHC process made use of an automated immunostainer (Thermo Fisher Scientific, MA, USA). IHC staining was scored by two pathologists, and scores were only accepted when they were in agreement. Light microscopy images were obtained from each slice at 40× magnification. Overall IHC staining was scored as either 0 (none), 1 (weak), 2 (moderate), or 3 (strong). The percentage of positively-stained cells was recorded as either 0 (≤5%), 1(6%–25%), 2(26%–75%), or 3(>75%). The final score was 0–9, calculated as the product of the scores for overall IHC staining and percentage positive staining. The cutoff value for IHC staining was calculated using the receiver operating characteristic (ROC) curve and the Youden Index (sensitivity+specificity-1). The final score was taken to represent high expression when it was greater than the cutoff value, and low expression when less than or equal to the cutoff.

Methods of Postoperative Follow-Up and Collection of Related Information

Two doctors were responsible for follow-up and collection of patient information. Postoperative survival times of CRC patients were collected at 3 months and 1, 2, 3, 5, and 8 years after surgery; follow-up was halted if the patient died. Patient data was categorized according to (1) CXCL7 and VEGF staining in cancer and adjacent normal tissues, (2) age and gender, and (3) pathological parameters (30).

Statistical Analysis

The results of categorical variables were presented as frequencies (n) and percentages (%) in this study. The mean (M) and standard deviation (SD) were used to represent numerical variables. Expression of CXCL7 and VEGF were sequentially tested in different variables by Pearson’s chi-square test. Differences between CXCL7 and VEGF expression were analyzed by the Mann-Whitney U-test. Spearman’s rank-correlation test was used to evaluate the correlation between CXCL7 and VEGF expression. The 5-year overall survival (OS) time was calculated using Kaplan-Meier analysis, and graphs of survival rate are shown in Results. Cox proportional hazard regression models were used for analyzing risk factors used in prognoses for CRC patients. First, statistically significant indicators were selected by univariate analysis. Then, multivariate regression models were employed to seek out independent prognostic factors. R 4.0.0 software was used to draw a nomogram of 5-year OS. In addition, the concordance index (C-index), the area under the ROC curve (AUC), and the calibration curve were used to evaluate the predictive ability of the nomogram model. SPSS 22.0 software (IBM; Armonk, NY, USA) was used to analyze all data in this study. Statistical differences were significant for P value < 0.05.

Results

General Clinical Information of CRC Patients

One hundred and sixty patients who had been definitively diagnosed with CRC were recruited, including 73 women (45.63%, 73/160) and 87 men (54.37%, 87/160). The average age was 59.46 ± 9.13 years for women and 58.17 ± 11.24 years for men (Table 1). Patients were classified into two groups based on age: ≤ 60 (55.00%, 88/160) and > 60 (45.00%, 72/160). Other CRC patient information was shown in Table 1.

TABLE 1
www.frontiersin.org

Table 1 Association of CXCL7 and VEGF with clinical pathological characteristics and the correlation between two markers in CRC patients.

Expression of CXCL7 and VEGF in CRC Tissues

IHC was used to monitor expression of CXCL7 and VEGF in CRC tumors and adjacent normal tissues. Based on the ROC curve, a score of 3.5 was selected as the cutoff value for CXCL7, and 2.8 for VEGF. Similar to our previous research, CXCL7 was highly expressed in tumor tissues (65.63%, 105/160) but not in paracancerous tissue (25.00%, 40/160, P < 0.001; Figures 1A, B). Positive staining was mainly concentrated in the cytoplasm and membranes of CRC cells (Figure 1A). Similarly, we found that VEGF was also highly expressed in CRC tissues (64.38%, 103/160 vs. 26.25%, 42/160 in normal tissues, Figures 1C, D). Number of ICH score was shown in Figure 1E.

FIGURE 1
www.frontiersin.org

Figure 1 Expression of CXCL7 and VEGF in CRC patients. (A) Expression of CXCL7 in tumor tissues (magnification: 40× and 200×). (B) Expression of CXCL7 in normal tissues (40× and 200×). (C) Expression of VEGF in tumor tissues (40× and 200×). (D) Expression of VEGF in normal tissues (40× and 200×). (E) Patients were classified according to CXCL7 and VEGF IHC score from 0 to 9. Scale bar, 50 μm.

Associations Between CXCL7, VEGF and Clinicopathological Characteristics in CRC Patients

In order to probe the clinical significance of CXCL7 and VEGF in CRC, associations between these two markers and clinicopathological information were analyzed by the chi-square (χ2) test, and differences in expression were assessed by the Mann-Whitney U test. CXCL7 was correlated with N- and TNM-stage cancer (both P < 0.001; Figures 2A, B and Table 1), as was VEGF (both P < 0.001; Figures 2C, D and Table 1). However, there were no significant correlations between CXCL7 or VEGF and other clinicopathological characteristics (all P > 0.05; Table 1).

FIGURE 2
www.frontiersin.org

Figure 2 Associations between CXCL7 and VEGF and clinicopathological characteristics. (A) Expression of CXCL7 in different N-stage tumors. (B) Expression of CXCL7 in different TNM stages. (C) VEGF in N0 and N1-N2 stage tumors. (D) VEGF in I-II and III-IV stages. The Mann‐Whitney U-test was used for analysis. ***P < 0.001.

Clinical Correlation Between CXCL7 and VEGF in CRC Patients

To analyze whether expression of CXCL7 was related to the level of VEGF in CRC tissue, Spearman’s test was used to evaluate the correlation. Interestingly, there was a significant correlation between CXCL7 and VEGF in all CRC tissues (correlation coefficient: 0.796, P < 0.001, Figure 3A). We then explored whether there were correlations with other clinical features. Scatter plots were used to visualize results for the two markers. As shown in Figure 3, the correlation coefficients ranged from 0.523 to 0.842 (all P < 0.01), with the highest being for the female subgroup (r= 0.842) and the lowest for the M1 subgroup (r= 0.523, Figures 3A–Y and Table 1). These results illustrated a strong correlation between CXCL7 and VEGF in CRC tissues.

FIGURE 3
www.frontiersin.org

Figure 3 CXCL7 positively correlated with VEGF in CRC patients grouped according to various clinical features. (A) All CRC patients. (B) Male subgroup. (C) Female subgroup. (D) Age ≤ 60 years subgroup. (E) Age > 60 years subgroup. (F) Colon subgroup. (G) Rectum subgroup. (H) Left subgroup. (I) Right subgroup. (J) Tumor size <4 cm subgroup. (K) Tumor size ≥4 cm subgroup. (L) T1-T2 subgroup. (M) T3-T4 subgroup. (N) N0 subgroup. (O) N1-N2 subgroup. (P) M0 subgroup. (Q) M1 subgroup. (R) I-II subgroup. (S) III-IV subgroup. (T) Non-neural invasion subgroup. (U) Neural invasion subgroup. (V) Non-vascular invasion subgroup. (W) Vascular invasion subgroup. (X) Well differentiated subgroup. (Y) Moderately-poorly differentiated subgroup. The non-parametric Spearman’s test was used for these analyses.

High Expression of CXCL7 and VEGF Related to Poor Prognosis in CRC Patients

For the purpose of establishing the prognostic significance of CXCL7 and VEGF in CRC, the 160 patients were observed to have a median OS time of 67.00 (95% confidence interval [CI]: 61.37–72.63) months and a 5-year survival rate of 62.60% (Figure 4A). Interestingly, OS time was affected by extent of tumor invasion. There were significant differences in OS among the tumor stages: T (hazard ratio [HR]: 1.711; 95% CI: 1.048–2.793; P =0.032; Figure 4G), N (HR: 3.717; 95% CI: 2.160-6.397; P <0.001; Figure 4H), M (HR: 3.696; 95% CI: 1.991–6.862; P <0.001; Figure 4I), TNM (HR: 3.765; 95% CI: 1.828–7.756; P <0.001; Figure 4J), and level of CXCL7 (HR: 2.343; 95% CI: 1.431–3.837; P =0.001; Figure 4N) and VEGF (HR: 1.931; 95% CI: 1.189–3.136, P = 0.008; Figure 4O). Details are shown in Table 2. However, no significant differences were found among the other factors (Figures 4B–F, K–M and Table 2; all P >0.05). Multivariate analysis showed that high CXCL7 in tissues was independent of factors associated with poor OS of CRC patients (HR=2.363; 95% CI: 1.359–4.108; P = 0.002; Table 2). Patients with high levels of both CXCL7 and VEGF had poor outcomes (OS: 56.96 months vs 78.00 months in patients with low levels of each, HR: 2.410; 95% CI: 1.405-4.132; P = 0.001; Figure 4P).

FIGURE 4
www.frontiersin.org

Figure 4 Kaplan-Meier curves for CRC patients stratified on the basis of CXCL7, VEGF and clinicopathological features. (A) OS in all 160 patients. (B) OS in male vs. female patients (P = 0.133). (C) OS in age ≤60 vs. age >60 patients (P = 0.117). (D) OS in colon vs. rectum patients (P = 0.899). (E) OS in left side vs. right side patients (P = 0.252). (F) OS in tumor size <4 cm vs. ≥4 cm patients (P = 0.740). (G) OS in T1-T2 vs. T3-T4 patients (P = 0.032). (H) OS in N0 vs. N1-N2 patients (P < 0.001). (I) OS in M0 vs. M1 patients (P < 0.001). (J) OS in I-II vs. III-IV patients (P < 0.001). (K) OS in non-neural invasion vs. neural invasion patients (P = 0.271). (L) OS in non-vascular invasion vs. vascular invasion patients (P = 0.859). (M) OS in well differentiated vs. moderately-poorly differentiated patients (P = 0.271). (N) OS in CXCL7 low expression (CXCL7L) vs. CXCL7 high expression (CXCL7H) patients (P = 0.001). (O) OS in VEGF low expression (VEGFL) vs. VEGF high expression (VEGFH) patients (P = 0.008). (P) OS in CXCL7H + VEGFH vs. CXCL7L +VEGFL patients (P = 0.001).

TABLE 2
www.frontiersin.org

Table 2 Cox proportional hazard regression models for CXCL7, VEGF and clinical pathological characteristics.

A Prognostic Nomogram for Comprehensively Evaluating OS in CRC Patients

As nomograms are widely used in prognostic assessments, especially for cancer, we generated a nomogram using the R software package to analyze the value of CXCL7, VEGF and various pathological features in making a CRC prognosis (Figure 5A). First, a standard point-scale reference line was established on the top of the nomogram. Then, CXCL7, VEGF and related clinical and pathological information were listed in order. The total point score was then calculated for prediction of 5-year OS (only 5-year OS was evaluated in this study). From the nomogram, patients could be provided with a personalized evaluation for guiding clinical treatment following surgery. The length of each variable was associated with patient survival. TNM stage, neural invasion, CXCL7 expression and differentiation had the greatest impact on prognosis.

FIGURE 5
www.frontiersin.org

Figure 5 Nomogram of CRC patients’ 5-year survival time following surgery. (A) Nomogram for 5‐year OS on the basis of CXCL7 and VEGF expression and basic clinical data. (B) ROC curve for developing the nomogram. (C) Calibration curve of the nomogram.

The nomogram was also quantitatively assessed using the concordance index (C-index), area under the ROC curve (AUC), and calibration curve. The C-index was 0.747 (95% CI: 0.701–0.826). The 5‐year survival AUC was 0.734 (95% CI: 0.672–0.797; Figure 5B), and the calibration curve was shown in Figure 5C. Overall, the nomogram for 5-year OS could be used to make a reliable prognosis for CRC patients.

Discussion

CRC is a serious public health challenge associated with high mortality and morbidity worldwide, especially in developing countries such as China (35). Although numerous improvements related to diagnosis and treatment have been applied in clinical practice, the number of tumor-related deaths has continued to rise over the past few years (36). Surgery and postoperative radiotherapy and chemotherapy are standard treatments for CRC. Improvement of OS following surgery for CRC requires systematic evaluation and management (37). TNM stage has been used as a predictor of survival time in recent decades (38), although it is not always accurate. For this reason, recurrence and metastasis after treatment are usually included as risk factors for prognosis. Of course, a great many studies in the past few decades have identified novel prognostic biomarkers (3941). Various studies have found that angiogenesis plays an important role in recurrence and metastasis (42, 43). Even though many proteins and signaling molecules have been associated with angiogenesis in CRC cells (44, 45), the connection between CXCL7 and angiogenesis has not previously been explored. In this study, we performed series of experiments to evaluate the association of CXCL7 with angiogenesis, and assess its value in prognosis of CRC patients.

CXCL7, a neutrophil-activating chemokine, is primarily derived from peripheral platelets (46). Traditionally, CXCL7 has been considered to be involved in regulating glycolysis, mitosis, and prostaglandin synthesis, among other processes (46, 47). Recently, several studies have reported that CXCL7 is associated with many types of tumors, and plays important roles in tumor proliferation and metastasis (48, 49). We used IHC to show that CXCL7 was significantly over-expressed in CRC tumor cells, correlated with N-stage and TNM-stage cancer, and associated with poor patient outcomes. CXCL7 was evaluated as a CRC biomarker in our previous study (30), and together with the present study has been shown be a potentially reliable marker for making prognoses in CRC patients.

Angiogenesis, an important physiological process, also plays a pivotal role in tumor proliferation, invasion and metastasis (12, 13). A variety of proteins are involved in regulation of angiogenesis, including VEGF and other cytokines and chemokines (13, 14). Here, VEGF was highly expressed in CRC tumors and correlated with poor prognosis. As the role of VEGF in angiogenesis is widely known, anti-VEGF treatment drugs such as bevacizumab and many traditional plant polysaccharides (50, 51) are commonly used in anti-cancer treatments.

Previously we have focused on CXCL7 in serum from CRC patients (30), and its potential application to prognosis of obstructive colorectal cancer (33); however, the relationship between CXCL7 and angiogenesis has not previously been evaluated. In recent years, the role of chemokines in regulating angiogenesis has been revealed in several studies (52, 53), including the ability of CCL19 to suppress angiogenesis by downregulating VEGF-A expression in CRC cells (53). So, numerous studies have provided us with data upon which to base an investigation of the role of CXCL7 in angiogenesis in CRC patients. We chose VEGF as a marker of angiogenesis and used IHC to evaluate its expression in CRC cells. Then, the relationship between CXCL7 and VEGF expression was analyzed, and we found that the two were positively correlated. Based on these results, we conclude that CXCL7 prominently correlates with VEGF and clinicopathological features in CRC patients. Furthermore, univariate analysis showed that high expression of CXCL7 with VEGF is a risk factor that can be considered in prognosis of CRC patients. However, VEGF was not an independent risk factor in CRC patients. The nomogram and its corresponding evaluation index also showed that CXCL7 was associated with outcome in CRC patients. Based on the above results, we suggest that CXCL7 may regulate expression of VEGF in CRC tissues, maybe for the reason that angiogenic functions can be induced by chemokine-regulated inflammatory stimuli (48, 5456). Because the treatment methods are also very important for the patient’s prognosis, the related surgery, radiotherapy and chemotherapy methods will be taken into consideration for comprehensively exploring the factors which could affect patient survival time in future study. Furthermore, the relationship between CXCL7 and VEGF should be further studied in vivo and in vitro, including in CRC serum and cell lines.

In summary, this study analyzed the correlation between CXCL7 and VEGF, and illustrated that co-expression of the two markers leads to a poor prognosis in CRC patients. As chemokines have been suggested to be therapeutic targets in many studies (57, 58), CXCL7 may be a new target for regulating angiogenic signaling pathways, especially those controlling VEGF expression.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding authors.

Author Contributions

LL, KJ, DPL, DXL, GD, ST, GW, and XL conceived the study. LL, DPL, and ST performed the experiments and were responsible for statistical analysis. LL, KJ, and DPL were in charge of writing review, revision and other related work in this study. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the Bozhou science and technology major special projects (No. bzzd2020012), the Bozhou Key R&D projects (No.bzzc2021020) and the incubation project of Bozhou People’s Hospital (No.Byf202017).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2021.754221/full#supplementary-material

Supplementary Figure 1 | Flowchart for inclusion and exclusion of CRC patients.

Abbreviations

CRC, colorectal cancer; VEGF, vascular endothelial growth factor; IHC, immunohistochemical; TNM, tumor-node-metastasis; T, depth of tumor invasion; N, nodal involvement; M, distant metastasis; OS, overall survival time; C-index, concordance index; ROC, receiver operating characteristic; Mean (M) and standard deviation (SD); AUC, area under the ROC curve; HR, high risk factor.

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin (2018) 68(6):394–424. doi: 10.3322/caac.21492

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the Global Cancer Incidence and Mortality in 2018: GLOBOCAN Sources and Methods. Int J Cancer (2019) 144(8):1941–53. doi: 10.1002/ijc.31937

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Yang T, Li X, Montazeri Z, Little J, Farrington SM, Ioannidis JPA, et al. Gene-Environment Interactions and Colorectal Cancer Risk: An Umbrella Review of Systematic Reviews and Meta-Analyses of Observational Studies. Int J Cancer (2019) 145(9):2315–29. doi: 10.1002/ijc.32057

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Patterns and Trends in Colorectal Cancer Incidence and Mortality. Gut (2017) 66(4):683–91. doi: 10.1136/gutjnl-2015-310912

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Jacobs D, Zhu R, Luo J, Grisotti G, Heller DR, Kurbatov V, et al. Defining Early-Onset Colon and Rectal Cancer. Front Oncol (2018) 8:504. doi: 10.3389/fonc.2018.00504

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Wan J, Yang J, Qiao CX, Sun X, Di A, Zhang L, et al. MicroRNA-362 Inhibits Cell Proliferation and Invasion by Directly Targeting SIX1 in Colorectal Cancer. Yonsei Med J (2019) 60(5):414–22. doi: 10.3349/ymj.2019.60.5.414

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Chong RC, Ong MW, Tan KY. Managing Elderly With Colorectal Cancer. J Gastrointest Oncol (2019) 10(6):1266–73. doi: 10.21037/jgo.2019.09.04

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Saleh R, Taha RZ, Toor SM, Sasidharan Nair V, Murshed K, Khawar M, et al. Expression of Immune Checkpoints and T Cell Exhaustion Markers in Early and Advanced Stages of Colorectal Cancer. Cancer Immunol Immunother (2020) 69(10):1989–99. doi: 10.1007/s00262-020-02593-w

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Malvezzi M, Arfé A, Bertuccio P, Levi F, La Vecchia C, Negri E. European Cancer Mortality Predictions for the Year 2011. Ann Oncol (2011) 22(4):947–56. doi: 10.1093/annonc/mdq774

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal Cancer Statistics, 2020. CA Cancer J Clin (2020) 70(3):145–64. doi: 10.3322/caac.21601

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Dekker E, Rex DK. Advances in CRC Prevention: Screening and Surveillance. Gastroenterology (2018) 154(7):1970–84. doi: 10.1053/j.gastro.2018.01.069

PubMed Abstract | CrossRef Full Text | Google Scholar

12. De Heer EC, Jalving M, Harris AL. HIFs, Angiogenesis, and Metabolism: Elusive Enemies in Breast Cancer. J Clin Invest (2020) 130(10):5074–87. doi: 10.1172/JCI137552

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Cao J, Liu X, Yang Y, Wei B, Li Q, Mao G, et al. Decylubiquinone Suppresses Breast Cancer Growth and Metastasis by Inhibiting Angiogenesis via the ROS/p53/ BAI1 Signaling Pathway. Angiogenesis (2020) 23(3):325–38. doi: 10.1007/s10456-020-09707-z

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Bausch D, Fritz S, Bolm L, Wellner UF, Fernandez-Del-Castillo C, Warshaw AL, et al. Hedgehog Signaling Promotes Angiogenesis Directly and Indirectly in Pancreatic Cancer. Angiogenesis (2020) 23(3):479–92. doi: 10.1007/s10456-020-09725-x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Xie M, Yu T, Jing X, Ma L, Fan Y, Yang F, et al. Exosomal Circshkbp1 Promotes Gastric Cancer Progression via Regulating the miR-582-3p/HUR/VEGF Axis and Suppressing HSP90 Degradation. Mol Cancer (2020) 19(1):112. doi: 10.1186/s12943-020-01208-3

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Braile M, Marcella S, Cristinziano L, Galdiero MR, Modestino L, Ferrara AL, et al. VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci (2020) 21(15):5294. doi: 10.3390/ijms21155294

CrossRef Full Text | Google Scholar

17. Hosaka K, Yang Y, Seki T, Du Q, Jing X, He X, et al. Therapeutic Paradigm of Dual Targeting VEGF and PDGF for Effectively Treating FGF-2 Off-Target Tumors. Nat Commun (2020) 11(1):3704. doi: 10.1038/s41467-020-17525-6

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Eguchi R, Wakabayashi I. HDGF Enhances VEGF‐dependent Angiogenesis and FGF‐2 Is a VEGF‐independent Angiogenic Factor in Non‐Small Cell Lung Cancer. Oncol Rep (2020) 44(1):14–28. doi: 10.3892/or.2020.7580

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The Cytokine Storm in COVID-19: An Overview of the Involvement of the Chemokine/Chemokine-Receptor System. Cytokine Growth Factor Rev (2020) 53:25–32. doi: 10.1016/j.cytogfr.2020.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Groblewska M, Litman-Zawadzka A, Mroczko B. The Role of Selected Chemokines and Their Receptors in the Development of Gliomas. Int J Mol Sci (2020) 21(10):3704. doi: 10.3390/ijms21103704

CrossRef Full Text | Google Scholar

21. Morein D, Erlichman N, Ben-Baruch A. Beyond Cell Motility: The Expanding Roles of Chemokines and Their Receptors in Malignancy. Front Immunol (2020) 11:952. doi: 10.3389/fimmu.2020.00952

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Wang L, Zhang S, Wang X. The Metabolic Mechanisms of Breast Cancer Metastasis. Front Oncol (2021) 10:602416. doi: 10.3389/fonc.2020.602416

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Jin Y, Lin Q, Fei H, Xue L, Li L, Xi Q, et al. Bioinformatics Analysis of Potential Therapeutic Targets and Prognostic Biomarkers Amid CXC Chemokines in Ovarian Carcinoma Microenvironment. J Oncol (2021) 2021:8859554. doi: 10.1155/2021/8859554

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Korbecki J, Grochans S, Gutowska I, Barczak K, Baranowska-Bosiacka I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int J Mol Sci (2020) 21(20):7619. doi: 10.3390/ijms21207619

CrossRef Full Text | Google Scholar

25. Korbecki J, Kojder K, Simińska D, Bohatyrewicz R, Gutowska I, Chlubek D, et al. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of the Ligands of Receptors CCR1, CCR2, CCR3, and CCR4. Int J Mol Sci (2020) 21(21):8412. doi: 10.3390/ijms21218412

CrossRef Full Text | Google Scholar

26. Adekoya TO, Richardson RM. Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int J Mol Sci (2020) 21(12):4449. doi: 10.3390/ijms21124449

CrossRef Full Text | Google Scholar

27. Liu H, Yang Z, Lu W, Chen Z, Chen L, Han S, et al. Chemokines and Chemokine Receptors: A New Strategy for Breast Cancer Therapy. Cancer Med (2020) 9(11):3786–99. doi: 10.1002/cam4.3014

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Ferrari SM, Fallahi P, Elia G, Ragusa F, Ruffilli I, Paparo SR, et al. Thyroid Autoimmune Disorders and Cancer. Semin Cancer Biol (2020) 64:135–46. doi: 10.1016/j.semcancer.2019.05.019

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Pawluczuk E, Łukaszewicz-Zając M, Mroczko B. The Role of Chemokines in the Development of Gastric Cancer -Diagnostic and Therapeutic Implications. Int J Mol Sci (2020) 21(22):8456. doi: 10.3390/ijms21228456

CrossRef Full Text | Google Scholar

30. Li L, Zhang L, Tian Y, Zhang T, Duan G, Liu Y, et al. Serum Chemokine CXCL7 as a Diagnostic Biomarker for Colorectal Cancer. Front Oncol (2019) 9:921. doi: 10.3389/fonc.2019.00921

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Finsterbusch M, Norman MU, Hall P, Kitching AR, Hickey MJ. Platelet Retention in Inflamed Glomeruli Occurs via Selective Prolongation of Interactions With Immune Cells. Kidney Int (2019) 95(2):363–74. doi: 10.1016/j.kint.2018.08.042

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Cheng Y, Wang X, Wang B, Zhou H, Dang S, Shi Y, et al. Aging-Associated Oxidative Stress Inhibits Liver Progenitor Cell Activation in Mice. Aging (2017) 9(5):1359–74. doi: 10.18632/aging.101232

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Li LH, Zhang LH, Zhang T, Qi XX, Cheng G, Xia LX. Serum Chemokine CXCL7 as a Potential Novel Biomarker for Obstructive Colorectal Cancer. Front Oncol (2021) 10:599363. doi: 10.3389/fonc.2020.599363

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Meng ZW, Pan W, Hong HJ, Chen JZ, Chen YL. Modified Staging Classification for Intrahepatic Cholangiocarcinoma Based on the Sixth and Seventh Editions of the AJCC/UICC TNM Staging Systems. Med (Baltimore) (2017) 96(34):e7891. doi: 10.1097/MD.000000000000789

CrossRef Full Text | Google Scholar

35. Abulimiti A, Zhang X, Shivappa N, Hébert JR, Fang YJ, Huang CY, et al. The Dietary Inflammatory Index Is Positively Associated With Colorectal Cancer Risk in a Chinese Case-Control Study. Nutrients (2020) 12(1):232. doi: 10.3390/nu12010232

CrossRef Full Text | Google Scholar

36. Gu MJ, Huang QC, Bao CZ, Li YJ, Li XQ, Ye D, et al. Attributable Causes of Colorectal Cancer in China. BMC Cancer (2018) 18(1):38. doi: 10.1186/s12885-017-3968-z

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Hashiguchi Y, Muro K, Saito Y, Ito Y, Ajioka Y, Hamaguchi T, et al. Japanese Society for Cancer of the Colon and Rectum (JSCCR) Guidelines 2019 for the Treatment of Colorectal Cancer. Int J Clin Oncol (2020) 25(1):1–42. doi: 10.1007/s10147-019-01485-z

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Lawler M, Alsina D, Adams RA, Anderson AS, Brown G, Fearnhead NS, et al. Critical Research Gaps and Recommendations to Inform Research Prioritisation for More Effective Prevention and Improved Outcomes in Colorectal Cancer. Gut (2018) 67(1):179–93. doi: 10.1136/gutjnl-2017-315333

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Jung G, Hernández-Illán E, Moreira L, Balaguer F, Goel A. Epigenetics of Colorectal Cancer: Biomarker and Therapeutic Potential. Nat Rev Gastroenterol Hepatol (2020) 17(2):111–30. doi: 10.1038/s41575-019-0230-y

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Sveen A, Kopetz S, Lothe RA. Biomarker-Guided Therapy for Colorectal Cancer: Strength in Complexity. Nat Rev Clin Oncol (2020) 17(1):11–32. doi: 10.1038/s41571-019-0241-1

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Sawicki T, Ruszkowska M, Danielewicz A, Niedźwiedzka E, Arłukowicz T, Przybyłowicz KE. A Review of Colorectal Cancer in Terms of Epidemiology, Risk Factors, Development, Symptoms and Diagnosis. Cancers (2021) 13(9):2025. doi: 10.3390/cancers13092025

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Long W, Wang M, Luo X, Huang G, Chen J. Murrangatin Suppresses Angiogenesis Induced by Tumor Cell-Derived Media and Inhibits AKT Activation in Zebrafish and Endothelial Cells. Drug Des Devel Ther (2018) 12:3107–15. doi: 10.2147/DDDT.S145956

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Fahey E, Doyle SL. IL-1 Family Cytokine Regulation of Vascular Permeability and Angiogenesis. Front Immunol (2019) 10:1426. doi: 10.3389/fimmu.2019.01426

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Zheng S, Han MY, Xiao ZX, Peng JP, Dong Q. Clinical Significance of Vascular Endothelial Growth Factor Expression and Neovascularization in Colorectal Carcinoma. World J Gastroenterol (2003) 9(6):1227–30. doi: 10.3748/wjg.v9.i6.1227

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Yin C, Jiang C, Liao F, Rong Y, Cai X, Guo G, et al. Initial LDH Level can Predict the Survival Benefit From Bevacizumab in the First-Line Setting in Chinese Patients With Metastatic Colorectal Cancer. Onco Targets Ther (2014) 7:1415–22. doi: 10.2147/OTT.S64559

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Brown AJ, Joseph PR, Sawant KV, Rajarathnam K. Chemokine CXCL7 Heterodimers: Structural Insights, CXCR2 Receptor Function, and Glycosaminoglycan Interactions. Int J Mol Sci (2017) 18(4):748. doi: 10.3390/ijms18040748

CrossRef Full Text | Google Scholar

47. Brown AJ, Sepuru KM, Sawant KV, Rajarathnam K. Platelet-Derived Chemokine CXCL7 Dimer Preferentially Exists in the Glycosaminoglycan-Bound Form: Implications for Neutrophil-Platelet Crosstalk. Front Immunol (2017) 8:1248. doi: 10.3389/fimmu.2017.01248

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Yu M, Berk R, Kosir MA. CXCL7-Mediated Stimulation of Lymphangiogenic Factors VEGF-C, VEGF-D in Human Breast Cancer Cells. J Oncol (2010) 2010:939407. doi: 10.1155/2010/939407

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Chiang HC, Wang YS, Chou CH, Liao AT, Chu RM, Lin CS. Overexpression of Chemokine Ligand 7 Is Associated With the Progression of Canine Transmissible Venereal Tumor. BMC Vet Res (2012) 8:216. doi: 10.1186/1746-6148-8-216

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Haunschild CE, Tewari KS. Bevacizumab Use in the Frontline, Maintenance and Recurrent Settings for Ovarian Cancer. Future Oncol (2020) 16(7):225–46. doi: 10.2217/fon-2019-0042

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Gan QX, Wang J, Hu J, Lou GH, Xiong HJ, Peng CY, et al. Modulation of Apoptosis by Plant Polysaccharides for Exerting Anti-Cancer Effects: A Review. Front Pharmacol (2020) 11:792. doi: 10.3389/fphar.2020.00792

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Reich N, Beyer C, Gelse K, Akhmetshina A, Dees C, Zwerina J, et al. Microparticles Stimulate Angiogenesis by Inducing ELR(+) CXC-Chemokines in Synovial Fibroblasts. J Cell Mol Med (2018) 15(4):756–62. doi: 10.1111/j.1582-4934.2010.01051.x

CrossRef Full Text | Google Scholar

53. Xu Z, Zhu C, Chen C, Zong Y, Feng H, Liu D, et al. CCL19 Suppresses Angiogenesis Through Promoting miR-206 and Inhibiting Met/ERK/Elk-1/HIF-1α /VEGF-A Pathway in Colorectal Cancer. Cell Death Dis (2018) 9(10):974. doi: 10.1038/s41419-018-1010-2

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and Mortality and Epidemiology of Breast Cancer in the World. Asian Pac J Cancer Prev (2016) 17(S3):43–6. doi: 10.7314/apjcp.2016.17.s3.43

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Fülle L, Steiner N, Funke M, Gondorf F, Pfeiffer F, Siegl J, et al. RNA Aptamers Recognizing Murine CCL17 Inhibit T Cell Chemotaxis and Reduce Contact Hypersensitivity In Vivo. Mol Ther (2018) 26(1):95–104. doi: 10.1016/j.ymthe.2017.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Groblewska M, Mroczko B. Pro- and Antiangiogenic Factors in Gliomas: Implications for Novel Therapeutic Possibilities. Int J Mol Sci (2021) 22(11):6126. doi: 10.3390/ijms22116126

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Korbecki J, Kojder K, Barczak K, Simińska D, Gutowska I, Chlubek D, et al. Hypoxia Alters the Expression of CC Chemokines and CC Chemokine Receptors in a Tumor-A Literature Review. Int J Mol Sci (2020) 21(16):5647. doi: 10.3390/ijms21165647

CrossRef Full Text | Google Scholar

58. Bronger H, Magdolen V, Goettig P, Dreyer T. Proteolytic Chemokine Cleavage as a Regulator of Lymphocytic Infiltration in Solid Tumors. Cancer Metastasis Rev (2019) 38(3):417–30. doi: 10.1007/s10555-019-09807-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: CXCL7, VEGF, angiogenesis, prognosis, colorectal cancer

Citation: Li L, Jiang K, Li D, Li D, Fan Z, Dai G, Tu S, Liu X and Wei G (2021) The Chemokine CXCL7 Is Related to Angiogenesis and Associated With Poor Prognosis in Colorectal Cancer Patients. Front. Oncol. 11:754221. doi: 10.3389/fonc.2021.754221

Received: 06 August 2021; Accepted: 13 September 2021;
Published: 08 October 2021.

Edited by:

Yuming Jiang, Stanford University, United States

Reviewed by:

Louise Catherine Connell, Cornell University, United States
Yuqian Feng, Zhejiang Chinese Medical University, China

Copyright © 2021 Li, Jiang, Li, Li, Fan, Dai, Tu, Liu and Wei. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Longhai Li, pe_lilonghai@126.com; Guangyou Wei, 13515682626@163.com; Xiangyu Liu, 632780327@qq.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.