Back to Journals » Cancer Management and Research » Volume 12

Clinical Significance of Red Cell Distribution Width and Circulating Tumor Cells with an Epithelial–Mesenchymal Transition Phenotype in Lung Adenocarcinoma

Authors Peng H, Tan X, Wang Y, Dai L, Liang G, Guo J, Chen M

Received 25 February 2020

Accepted for publication 20 May 2020

Published 26 June 2020 Volume 2020:12 Pages 5105—5117

DOI https://doi.org/10.2147/CMAR.S251271

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Eileen O'Reilly



Huajian Peng,* Xiang Tan,* Yongyong Wang, Lei Dai, Guanbiao Liang, Jianji Guo, Mingwu Chen

Department of Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Mingwu Chen
Department of Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
Email [email protected]

Objective: To determine the prognostic value of red cell distribution width (RDW) and circulating tumor cells with epithelial–mesenchymal transition phenotype (M-CTC) in lung adenocarcinoma (LUAD).
Patients and Methods: Clinical and laboratory data of 60 patients with LUAD were collected. CTCs were isolated from their peripheral blood using the CanPatrolTM CTC enrichment method. The indicators of RDW and neutrophil lymphocyte ratio (NLR) were calculated based on the laboratory standards.
Results: A total of 60 LUAD patients were enrolled, of which 19 (31.7%) had high RDW (> 0.14) and 32 (53.3%) were positive for M-CTCs. There was no significant correlation between RDW and the clinical characteristics. M-CTC was not significantly associated with tumor size and differentiation, age, gender, tumor stage, and histological type but correlated significantly with lymphatic metastasis (P = 0.044), high NLR (> 2.26, P = 0.023), and high RDW (> 0.14, P = 0.036). Furthermore, the M-CTC+ LUAD patients had a significantly poor recurrence-free survival (RFS; Log rank P =0.001, HR = 2.749, 95% CI = 1.489– 5.078) and overall survival (OS; Log rank P =0.022, HR = 2.283, 95% CI = 1.128– 4.622) compared to the M-CTC patients. Similarly, high RDW also correlated with worse RFS (Log rank P = 0.008, HR = 2.331, 95% CI = 1.248– 4.353) and OS (Log rank P = 0.004, HR = 0.004, 95% CI = 1.398– 5.525).
Conclusion: M-CTC is significantly related to RDW and NLR, and an independent prognostic factor in LUAD.

Keywords: circulating tumor cell, epithelial–mesenchymal, red cell distribution width, lung adenocarcinoma, survival

Introduction

Lung cancer is the leading cause of morbidity and mortality in China and worldwide. Non-small cell lung cancer (NSCLC) accounts for nearly 80% of all lung cancer cases, and includes large cell carcinoma, squamous cell carcinoma and adenocarcinoma, of which lung adenocarcinoma (LUAD) has the highest prevalence of 50%.1,2 Although the development of novel diagnostic and therapeutic approaches has improved the prognosis of NSCLC patients, the 5-year survival rate of patients with LUAD is only 4–17%,3,4 mainly due to the lack of simple and effective prognostic biomarkers. Therefore, novel biomarkers need to be identified in order to improve early diagnosis and treatment of LUAD patients.

Studies have established the prognostic relevance of complete blood counts (CBC) in various malignancies, including lung cancer.57 CBC parameters are reliable indices of local and systemic inflammation,810 and cancer patients frequently show significant changes in neutrophil, lymphocyte and platelet counts, red cell distribution width (RDW), systemic immune inflammation index (SII), platelet to lymphocyte ratio (PLR), neutrophil to lymphocyte ratio (NLR) and monocyte to lymphocyte ratio (MLR).8,11 RDW (%) is a measure of the variability in erythrocyte volume and calculated as the standard deviation of erythrocyte volume/average cell volume × 100. High RDW is associated with the prognosis of liver cancer,12 breast cancer13 and gastric cancer,14 and likely caused by chronic inflammation and poor nutritional status (such as deficiency of iron, folic acid and vitamin B12)15,16 that frequently accompanies cancer.

The existence of circulating tumor cells (CTCs) was first proposed by Ashworth in 1869.17 CTCs are epithelial cells that are shed from the primary tumor into circulation and cause tumor metastasis.18 They are classified into the epithelial (E-CTCs), epithelial–mesenchymal transition (M-CTCs) and mixed (E/M-CTCs) phenotypes.19 Epithelial–mesenchymal transition (EMT) endows cancer cells with greater invasiveness and is crucial to the process of metastasis.2022 Consistent with this, studies show that M-CTCs are closely related to the prognosis and other characteristics of gastric cancer,23 breast cancer,24 liver cancer25 and NSCLC.26 However, despite millions of tumor cells entering the bloodstream every day, the detection rate of CTCs is very low27 due to their clearance by immune cells or other factors.28 However, other blood cells like neutrophils and platelets can enhance the survival and distant metastasis of CTCs.29 For instance, an aberrantly high peripheral blood NLR is significantly correlated to tumor development, since neutrophils secrete vascular endothelial growth factor (VEGF) and proteases that promote CTCs adhesion and seeding in distant organs.30,31 Lymphocytes on the other hand prevent tumor metastasis by inducing cell death and inhibiting tumor cell proliferation and migration,28,32 which determines patients’ immune response to malignant tumors.33 Furthermore, the inflammatory response and oxidative stress-induced damage to red blood cells increases RDW, which alters the blood flow3436 and may further disseminate the CTCs.

Although the relationship between RDW and tumor prognosis has been established before, the specific role of RDW in LUAD remains to be elucidated. The aim of this study was to explore the correlation between NLR, RDW and M-CTC in LUAD, and determine their respective prognostic values. To this end, we collected clinical and pathological data of 60 LUAD patients, and isolated and typed the peripheral blood CTCs using the advanced CanPatrolTM CTC enrichment technology and in situ hybridization respectively.

Patients and Methods

Study Population and Design

Sixty LUAD patients were enrolled between April 2014 and July 2014 at the First Affiliated Hospital of Guangxi Medical University (Nanning, China) (Supplemnatery Table S1). The inclusion criteria were as follows: (i) pathologically confirmed LUAD, (ii) radical lobectomy and systemic lymph node dissection, (iii) no distant metastasis before surgery, (iv) no history of radiotherapy or chemotherapy before surgery, and (v) availability of complete medical records. The platelet (P), neutrophil (N), monocytes (M) and lymphocyte (L) counts, and the RDW were measured by routine tests in the week before surgery. SII was calculated as P × N/L, NLR as N/L, MLR as M/L, and PLR as P/L. Five milliliter peripheral blood was collected from patients within three days after surgery into anticoagulant-coated tubes for CTCs isolation or biochemical assays. The study was conducted in accordance with the Declaration of Helsinki. The study was also approved by the ethical committee of the First Affiliated Hospital of Guangxi Medical College, and all patients provided written informed consent.

Isolation of CTCs

The erythrocytes were removed from the peripheral blood samples using the erythrocyte lysis buffer, and the plasma was filtered through an 8μm pore size filter membrane using the CanPatrolTM37,38 immune capture and nanofiltration-based CTC enrichment system. The isolated CTCs were then typed for CD45 and the EMT markers (EpCAM and vimentin) using RNA in situ hybridization (ISH).37

RNA ISH

The CTCs were digested with protease (Qiagen GmbH, Hilden, Germany) and hybridized with EpCAM, vimentin and CD45 probes (Invitrogen, Thermo Fisher Scientific Inc., Waltham, MA, USA) at 42°C for 2 hours. After washing thrice with 1 mL washing buffer to remove unbound probes, the cells were incubated with preamplifier solution at 42°C for 20 minutes, cooled, washed again, and incubated with the amplifier solution at room temperature for 1h. The cells were then incubated with Alexa Fluor 594-vimentin, Alexa Fluor 488-EpCAM and Alexa Fluor 647-CD45 at 42°C for 20 minutes, washed, and counter-stained with 4ʹ,6-diamidino-2-phenylindole (DAPI) for 5 minutes at room temperature. The stained cells were observed under a fluorescence microscope (Olympus Corporation, Tokyo, Japan), and the EpCAM+ vimentin (E-CTCs), EpCAM+ vimentin+ (biphenotypic E/MCTCs) and EpCAM vimentin+ (M-CTCs) phenotypes were identified.

Follow-Up

All patients were followed up through outpatient review or telephone interviews till July 30, 2019. Recurrence-free survival (RFS) was defined as the date from surgery to disease recurrence or the last follow-up. Overall survival (OS) was defined as the time from surgery to death for any reason or the last recorded follow-up visit.

Statistical Analysis

All statistical analysis was performed using SPSS version 19.0 (SPSS Inc., Chicago, Illinois, USA) and the graphs were drawn using GraphPad Prism version 5.0 (GraphPad software, Inc., La Jolla, CA, USA). Time-dependent receiver operating characteristic (ROC) curves were plotted in order to establish the cutoffs for low and high NLR, PLR, MLR, SII and RDW relative to the respective baseline values, optimal sensitivity, specificity, and area under the curve (AUC) for prediction of death from all causes. Kaplan–Meier survival curves were plotted to determine RFS and OS of patients demarcated on the basis of M-CTC, NLR, PLR, MLR, SII and RDW. The hazard rates (HRs) and 95% confidence intervals (CI) were calculated by univariate and multivariate Cox proportional hazard regression model. P values less than 0.05 were considered statistically significant.

Results

A total of 60 patients with LUAD were enrolled from April 2014 to July 2014, and their characteristics are summarized in Table 1. There were 30 male and female patients each (50%), and their median age was 59 years (33–79 years, 59.68 ± 9.16 years). Forty-four patients were younger than 65 years and 16 were older than 65 years. In addition, 12 patients (25%) had a history of smoking and 45 (75%) were non-smokers. In terms of oncological parameters, 28 patients (46.7%) had no lymphatic metastasis and 32 (53.3%) presented with lymphatic metastasis, while 18 (30%) and 42 (70%) patients had primary tumors > 4 cm and ≤ 4 cm respectively. Furthermore, 23 (38.3%), 14 (23.3%), 17 (28.3%) and 6 (10%) patients were respectively at stage I, II, III and IV, resulting in 37 (61.7%) patients at early stage (I + II) and 23 (38.3%) at the advanced stage (III + IV). Finally, 30 (50%) patients had poorly differentiated tumors, 24 (40%) moderately differentiated tumors and 6 (10%) presented highly differentiated tumors.

Table 1 Patient Characteristics of the Entire Series

The average NLR in patient peripheral blood is 3.329 ± 3.877 (1.11–29.27). According to the ROC curve, the cut-off value, sensitivity, specificity and area under the curve (AUC) for NLR in our cohort were respectively 2.26, 69.7%, 70.4% and 0.7250 (95% CI=0.5943–0.8557) (Figure 1A). The patients were divided into the NLR ≤ 2.26 (31, 51.7%) and NLR > 2.26 (29, 48.3%) groups, and as shown in Table 2, NLR was significantly correlated with the staging (P = 0.0032, OR = 3.352, 95% CI = 1.111–10.115) but not with other clinical characteristics. The average value of PLR is 157.87 ± 79.28 (44.90–398.99), and its cut-off value in the current study was 108.94 (Figure 1B). The ROC curve also indicated that the sensitivity and specificity were 90.9–40.7% respectively, and the AUC was 0.5960 (95% CI = 0.4454–0.7465). There were 46 (76.7%) patients with high PLR and 14 (23.3%) with low PLR. The relationship between PLR and clinical characteristics are summarized in Table 2, which indicate no significant correlation. The cut-off value of MLR was calculated to be 0.245 (Figure 1C) compared to its average value of 0.347 ± 0.03 (0.10–1.49). The sensitivity and specificity were 75.8–59.3% respectively, and the AUC was 0.6526 (95% CI = 0.5093–0.7960). There were 30 (50%) patients with MLR> 0.245 and 30 (50%) with MLR≤0.245. As shown in Table 3, MLR was significantly correlated with lymphatic metastasis (P = 0.048, OR = 2.949, 95% CI = 1.011–8.599) and tumor stage (P = 0.027, OR = 3.800, 95% CI = 1.165–12.392).

Table 2 Association Between Patients/Tumor Characteristics with NLR and PLR

Table 3 Association Between Patients/Tumor Characteristics with MLR and SII

Figure 1 The ROC curves for inflammation index: (A) NLR; (B) PLR; (C) MLR; (D) SII and (E) RDW.

The average value of SII is 837.01 ± 851.80 (113.60–5790.15), and its cut-off value was 491.75 in the current study (Figure 1D). The ROC curve indicated that the sensitivity, specificity and AUC were respectively 72.7%, 63% and 0.7026 (95% CI = 0.5670–0.8382). Accordingly, 34 (56.7%) and 26 (43.3%) patients were divided into the SII> 491.75 and SII≤491.75 groups respectively. As shown in Table 3, SII was significantly correlated to lymphatic metastasis (P = 0.013, OR = 3.949, 95% CI = 1.340–11.644), tumor size (P = 0.037, OR = 3.850, 95% CI = 1.086–13.647) and stage (P = 0.010, OR = 4.725, 95% CI = 1.444–15.457). The average value of RDW is 0.1383 ± 0.0118 (0.11–0.17), and its cut-off was determined to be 0.14 from the ROC curve (Figure 1E). The sensitivity and specificity of RDW were 45.9–85.2% respectively, and the AUC was 0.6229 (95% CI = 0.4780–0.7678). Nineteen patients showed RDW>0.14 and 41 had RDW≤0.14. Although RDW was not significantly associated with the clinical characteristics (Table 4), it increased with stage progression (Figure 2) and showed statistical significance with stage I and II (P = 0.0020).

Table 4 Association Between Patients/Tumor Characteristics with RDW

Figure 2 Distribution of RDW in LUAD patients according to tumor stage.

The distribution of CTC phenotypes among the 60 LUAD patients is shown in Figure 3AC. The positive rate of CTCs was 95% (0 to 68), and the median and average values were 5 and 9.5 ± 12.6 respectively (Table 5). The CTC load increased with disease progression, but did not reach statistical significance (Figure 4A). The positive rates of M-CTC increased steadily to 34.8%, 57.1%, 64.7–83.3% at stages I, II, III and IV respectively (P = 0.0186 between stage I and IV, Figure 4B), while that of CTCs and E-CTCs were unaffected by LUAD progression. Furthermore, the M-CTC+ patients had significantly higher risk of lymphatic metastasis (P = 0.044, OR = 2.950, 95% CI = 1.0-0-8.451), RDW (P = 0.036, OR = 3.578, 95% CI = 1.085–11.795) and NLR (P = 0.023, OR = 3.436, 95% CI = 1.187–9.947) compared to the M-CTC patients (Table 6, Figure 5A and B).

Table 5 Positive Expression Rate of CTCs in Each LUAD Stage n (%)

Table 6 Association Between Patients/Tumor Characteristics with M-CTC

Figure 3 Representative images showing CTCs phenotypes. (A) E-CTC; (B) E/M-CTC and (C) M-CTC. Red CK19 and green – Twist. Magnification – 100×.

Figure 4 Distribution of CTC and M-CTC counts in LUAD patients according to tumor stage. (A) Total CTCs and (B) M-CTC.

Figure 5 NLR and RDW in M-CTC+ and M-CTC patients. (A) NLR and (B) RDW.

All patients were followed for at least 60 months, during which period 33 (55%) died and 45 (75%) experienced recurrence. NLR, PLR, MLR, SII, LDW and M-CTC levels had different impacts on RFS (Figure 6AF) and OS (Figure 7AF). High NLR was associated with significantly worse RFS (P = 0.0086) and OS (P = 0.0014), whereas patients with low PLR had better OS compared to those with high PLR (P=0.0076), although RFS (P=0.104) was not significantly affected. In addition, high MLR showed a significant correlation with worse RFS (P = 0.0481) and OS (P = 0.005). Patients with low SII had markedly better OS compared to patients with high SII (P = 0.0044), while RFS was not significantly affected by this parameter (P = 0.1004). Low RDW also correlated to favorable RFS (P = 0.0009) and OS (P = 0.0015), and consistent with this, the M-CTC patients showed both better RFS (P = 0.0001) and OS (P = 0.0106).

Figure 6 Kaplan–Meier curve of RFS in LUAD patients: (A) NLR; (B) PLR; (C) MLR; (D) SII; (E) RDW and (F) M-CTC.

Figure 7 Kaplan–Meier curve of OS in LUAD patients: (A) NLR; (B) PLR; (C) MLR; (D) SII; (E) RDW and (F) M-CT.

Univariate analysis showed that gender (P = 0.047, HR = 0.547, 95% CI = 0.302–0.992), smoking (P = 028, HR = 2.076, 95% CI = 1.083–3.977), M-CTC (P = 0.001, HR = 2.749, 95% CI = 1.489–5.078), lymphatic metastasis (P = 0.006, HR = 2.316, 95% CI = 1.266–4.283), tumor size (P = 0.003, HR = 2.562, 95% CI = 1.378 −4.763), stage (P = 0.041, HR = 1.873, 95% CI = 1.026–3.420), degree of differentiation (P = 0.004, HR = 0.412, 95% CI = 0.225–0.756) and RDW (P = 0.008, HR = 2.331, 95% CI = 1.248–4.353) were significantly associated with RFS (Table 7), of which M-CTC was an independent factor of recurrence as per multivariate analysis (P = 0.003, HR = 2.818, 95% CI = 1.431–5.531). Likewise, smoking (P = 0.006, HR = 2.711, 95% CI = 1.334–5.511), M-CTC (P = 0.022, HR2.283, 95% CI = 1.128–4.622), tumor size (P = 0.015, HR = 2.349, 95% CI = 1.179–4.681), staging (P = 0.003, HR = 2.746, 95% CI = 1.394–5.409), degree of differentiation (P = 0.006, HR = 0.366, 95% CI = 0.180–0.746), NLR (P = 0.004, HR = 2.879, 95% CI = 1.398–5.930), MLR (P = 0.013, HR = 2.649, 95% CI = 1.233–5.689), PLR (P = 0.025, HR = 3.299, 95% CI = 1.159–9.389), SII (P = 0.011, HR = 2.635, 95% CI = 1.254–5.539) and RDW (P = 0.004, HR = 2.779, 95% CI = 1.398–5.525) were significantly correlated with OS, and M-CTC (P = 0.022, HR = 2.490, 95% CI = 1.141–5.431), stage (P = 0.040, HR = 2.452, 95% CI = 1.040–5.782) and RDW (P = 0.032, HR = 2.508, 95% CI = 1.084–5.804) were the independent factors (Table 8).

Table 7 Univariate and Multivariate Statistical Analyses of Recurrence-Free Survival

Table 8 Univariate and Multivariate Statistical Analyses of Overall Survival

Discussion

This study is the first to explore the relationship between RDW and M-CTC, and determine their prognostic relevance in LUAD. RDW is a widely available by the vast majority of automated analysis. Reflecting the size heterogeneity of the circuiting erythrocytes, higher RDW values are suggestive of increased variation of red cell volumes (anisocytosis). We found that patients with higher RDW and M-CTC load had worse prognosis, and both increased with tumor progression. In addition, RDW was also determined to an independent risk factor, although the underlying mechanism through which RDW affects prognosis is still unclear. Tumor progression frequently triggers an inflammatory response that further exacerbates tumor growth, invasion and angiogenesis, and eventually promotes metastases.29,32,39 Inflammation also lowers red blood cell survival by destroying their membranes, leading to increased RDW and red blood cell atypia, thereby altering blood flow through microcirculation and likely promoting M-CTC dispersion.34 However, further research is needed to elucidate the relationship between RDW, inflammation and tumor metastasis.

CTCs are closely associated with distant metastasis in various malignancies. We found that both CTC and M-CTC counts increased with tumor progression, and patients with lymphatic metastasis had higher M-CTC positive rates. M-CTCs are regarded as the most malignant CTC. Therefore, patients with positive of M-CTC have a greater chance of early recurrence. Current research also confirms this. Metastasis involves EMT of tumor cells that results in the loss of cell-to-cell contact and cellular polarity, along with degradation of the extracellular matrix and basement membrane, which increase tumor cell migration and invasion into adjacent tissues.40,41 In line with this, both RFS and PFS were significantly worse in the M-CTC+ LUAD patients, and M-CTC was also an independent factor of worse prognosis.

NLR and RDW are established risk factors in multiple malignancies, and the M-CTC count was positively correlated with both factors in the LUAD patients in agreement with the findings of Wu et al.42 Studies show that neutrophils secrete vascular endothelial growth factor (VEGF) and proteases into circulation, which promote CTCs adhesion and seeding in distant organs.30,31 Lymphocytes on the other hand inhibit tumor metastasis by inducing cell death and28,32 mediating an immune response against the malignant tumors.33 Furthermore, inflammation and oxidative stress-induced damage to red blood cells increases RDW and alters microcirculation,3436 which further promote CTC metastasis.

There are certain limitations to our study. For instance, the study was retrospective in nature and performed at a single center on a small number of patients. In addition, we did not elucidate the relationship between M-CTC, RDW and NLR. Our findings need to be validated in multicenter prospective studies on larger cohorts. Nevertheless, we showed for the first time that RDW is associated with M-CTC and LUAD prognosis.

Conclusion

RDW and M-CTC are independent predictors of prognosis in patients with LUAD, and RDW is an economical and convenient prognostic biomarker for LUAD.

Acknowledgments

The authors thank Prof. Nuo Yang and Prof. Huafu Zhou, which are work in Department of Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, for their contributions in manuscript revision. This work was supported in part by the National Natural Science Foundation of China (81660387) and Development and Application of Medical and Health Appropriate Technology of Guangxi (S201654).

Disclosure

Huajian Peng and Xiang Tan are co-first authors. The authors declare that they have no competing interests.

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

2. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–132. doi:10.3322/caac.21338

3. Herbst RS, Heymach JV, Lippman SM. Lung cancer. N Engl J Med. 2008;359(13):1367–1380. doi:10.1056/NEJMra0802714

4. Moore W, Talati R, Bhattacharji P, Bilfinger T. Five-year survival after cryoablation of stage I non–small cell lung cancer in medically inoperable patients. J Vasc Interventional Radiol. 2015;26(3):312–319. doi:10.1016/j.jvir.2014.12.006

5. Wang Y, Li Y, Chen P, Xu W, Wu Y, Che G. Prognostic value of the pretreatment systemic immune-inflammation index (SII) in patients with non-small cell lung cancer: a meta-analysis. Ann Transl Med. 2019;7(18):433. doi:10.21037/atm.2019.08.116

6. Toda M, Tsukioka T, Izumi N, et al. Platelet-to-lymphocyte ratio predicts the prognosis of patients with non-small cell lung cancer treated with surgery and postoperative adjuvant chemotherapy. Thoracic Cancer. 2018;9(1):112–119. doi:10.1111/1759-7714.12547

7. Watanabe K, Yasumoto A, Amano Y, et al. Mean platelet volume and lymphocyte-to-monocyte ratio are associated with shorter progression-free survival in EGFR-mutant lung adenocarcinoma treated by EGFR tyrosine kinase inhibitor. PLoS One. 2018;13(9):e0203625. doi:10.1371/journal.pone.0203625

8. Balkwill F, Mantovani A. Cancer and inflammation: implications for pharmacology and therapeutics. Clin Pharmacol Ther. 2010;87(4):401–406. doi:10.1038/clpt.2009.312

9. Zheng L, Zou K, Yang C, Chen F, Guo T, Xiong B. Inflammation-based indexes and clinicopathologic features are strong predictive values of preoperative circulating tumor cell detection in gastric cancer patients. Clin Transl Oncol. 2017;19(9):1125–1132. doi:10.1007/s12094-017-1649-7

10. Bozkaya Y, Kurt B, Gurler F. A prognostic parameter in advanced non-small cell lung cancer: the ratio of hemoglobin-to-red cell distribution width. Int J Clin Oncol. 2019;24(7):798–806. doi:10.1007/s10147-019-01417-x

11. Passardi A, Scarpi E, Cavanna L, et al. Inflammatory indexes as predictors of prognosis and bevacizumab efficacy in patients with metastatic colorectal cancer. Oncotarget. 2016;7(22):33210–33219. doi:10.18632/oncotarget.8901

12. Zhao T, Cui L, Li A. The significance of RDW in patients with hepatocellular carcinoma after radical resection. Cancer Biomarkers. 2016;16(4):507–512. doi:10.3233/CBM-160591

13. Seretis. Is red cell distribution width a novel biomarker of breast cancer activity? Data from a pilot study. J Clin Med Res. 2013. doi:10.4021/jocmr1214w

14. Hirahara N, Tajima Y, Fujii Y, et al. Comprehensive analysis of red blood cell distribution width as a preoperative prognostic predictor in gastric cancer. Anticancer Res. 2019;39(6):3121–3130. doi:10.21873/anticanres.13448

15. Ferrucci L, Guralnik JM, Woodman RC, et al. Proinflammatory state and circulating erythropoietin in persons with and without anemia. Am J Med. 2005;118(11):1288.e1211–1288.e1219. doi:10.1016/j.amjmed.2005.06.039

16. Adamson S. The anemia of chronic disorders: studies of marrow regulation and iron metabolism. Blood. 1975;45:55–65. doi:10.1182/blood.V45.1.55.55

17. Pantel K, Alix-Panabieres C. Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010;16(9):398–406. doi:10.1016/j.molmed.2010.07.001

18. Kang BJ, Ra SW, Lee K, et al. Circulating tumor cell number is associated with primary tumor volume in patients with lung adenocarcinoma. Tuberc Respir Dis. 2020;83(1):61–70. doi:10.4046/trd.2019.0048

19. Tsongalis GJ. Branched DNA technology in molecular diagnostics. Am J Clin Pathol. 2006;126(3):448–453. doi:10.1309/90BU6KDXANFLN4RJ

20. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–454. doi:10.1038/nrc822

21. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112(12):1776–1784. doi:10.1172/JCI200320530

22. Chopin JPTD. Epithelial cell plasticity in development and tumor progression. Cancer Metastasis. 1999;18:31–42. doi:10.1023/A:1006256219004

23. Li TT, Liu H, Li FP, et al. Evaluation of epithelial-mesenchymal transitioned circulating tumor cells in patients with resectable gastric cancer: relevance to therapy response. World j Gastroenterol. 2015;21(47):13259–13267. doi:10.3748/wjg.v21.i47.13259

24. Mooney SM, Talebian V, Jolly MK, et al. The GRHL2/ZEB feedback loop-a key axis in the regulation of EMT in breast cancer. J Cell Biochem. 2017;118(9):2559–2570. doi:10.1002/jcb.25974

25. Hu B, Yang XR, Xu Y, et al. Systemic immune-inflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clin Cancer Res. 2014;20(23):6212–6222. doi:10.1158/1078-0432.CCR-14-0442

26. Li S, Chen Q, Li H, Wu Y, Feng J, Yan Y. Mesenchymal circulating tumor cells (CTCs) and OCT4 mRNA expression in CTCs for prognosis prediction in patients with non-small-cell lung cancer. Clin Transl Oncol. 2017;19(9):1147–1153. doi:10.1007/s12094-017-1652-z

27. Chang YS, Di Tomaso E, McDonald DM, Jones R, Jain RK, Munn LL. Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc Natl Acad Sci U S A. 2000;97(26):14608–14613. doi:10.1073/pnas.97.26.14608

28. Chen F, Wang S, Fang Y, et al. Feasibility of a novel one-stop ISET device to capture CTCs and its clinical application. Oncotarget. 2017;8(2):3029–3041. doi:10.18632/oncotarget.13823

29. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi:10.1016/j.cell.2011.02.013

30. Lazova R, Laberge GS, Duvall E, et al. A melanoma brain metastasis with a donor-patient hybrid genome following bone marrow transplantation: first evidence for fusion in human cancer. PLoS One. 2013;8(6):e66731. doi:10.1371/journal.pone.0066731

31. Cools-Lartigue J, Spicer J, McDonald B, et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123:3446–3458. doi:10.1172/JCI67484

32. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–444. doi:10.1038/nature07205

33. Halazun KJ, Hardy MA, Rana AA, et al. Negative impact of neutrophil-lymphocyte ratio on outcome after liver transplantation for hepatocellular carcinoma. Ann Surg. 2009;250(1):141–151. doi:10.1097/SLA.0b013e3181a77e59

34. Patel KV, Mohanty JG, Kanapuru B, Hesdorffer C, Ershler WB, Rifkind JM. Association of the red cell distribution width with red blood cell deformability. Adv Exp Med Biol. 2013;765:211–216.

35. Ichinose J, Murakawa T, Kawashima M, et al. Prognostic significance of red cell distribution width in elderly patients undergoing resection for non-small cell lung cancer. J Thorac Dis. 2016;8(12):3658–3666. doi:10.21037/jtd.2016.12.44

36. Zhao Z, Liu T, Li J, Yang W, Liu E, Li G. Elevated red cell distribution width level is associated with oxidative stress and inflammation in a canine model of rapid atrial pacing. Int J Cardiol. 2014;174(1):174–176. doi:10.1016/j.ijcard.2014.03.189

37. Wu S, Liu S, Liu Z, et al. Classification of circulating tumor cells by epithelial-mesenchymal transition markers. PLoS One. 2015;10(4):e0123976. doi:10.1371/journal.pone.0123976

38. Qi LN, Xiang BD, Wu FX, et al. Circulating tumor cells undergoing EMT provide a metric for diagnosis and prognosis of patients with hepatocellular carcinoma. Cancer Res. 2018;78(16):4731–4744. doi:10.1158/0008-5472.CAN-17-2459

39. Huang D-P, Ma R-M, Xiang Y-Q. Utility of red cell distribution width as a prognostic factor in young breast cancer patients. Medicine. 2016;95(17):e3430. doi:10.1097/MD.0000000000003430

40. Hollier BG, Evans K, Mani SA. The epithelial-to-mesenchymal transition and cancer stem cells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia. 2009;14(1):29–43. doi:10.1007/s10911-009-9110-3

41. Mego M, Karaba M, Minarik G, et al. Circulating tumor cells with epithelial-to-mesenchymal transition phenotypes associated with inferior outcomes in primary breast cancer. Anticancer Res. 2019;39(4):1829–1837. doi:10.21873/anticanres.13290

42. Wu F, Zhu J, Mao Y, Li X, Hu B, Zhang D. Associations between the epithelial-mesenchymal transition phenotypes of circulating tumor cells and the clinicopathological features of patients with colorectal cancer. Dis Markers. 2017;2017:1–6. doi:10.1155/2017/9474532

Creative Commons License © 2020 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.