Oncotarget

Research Papers:

Elevated pretreatment platelet distribution width and platelet count predict poor prognosis in nasopharyngeal carcinoma

PDF |  HTML  |  How to cite

Oncotarget. 2017; 8:106089-106097. https://doi.org/10.18632/oncotarget.22528

Metrics: PDF 1354 views  |   HTML 2130 views  |   ?  

XueCheng Xie, XiaoChun Zeng, SuJuan Cao, XiaoMao Hu, QiaoJing Shi, Dan Li, ShiYuan Zhou, Ping Gu _ and ZhongShan Zhang

Abstract

XueCheng Xie1,*, XiaoChun Zeng1,*, SuJuan Cao1, XiaoMao Hu1, QiaoJing Shi1, Dan Li1, ShiYuan Zhou2, Ping Gu3 and ZhongShan Zhang1

1Department of Oncology, The Affiliated Hospital of XiangNan University, Chenzhou, China

2Department of Nuclear Medicine, The Affiliated Hospital of XiangNan University, Chenzhou, China

3Department of Child Healthcare, Chenzhou First People’s Hospital, Chenzhou, China

*These authors have contributed equally to this work

Correspondence to:

Ping Gu, email: [email protected]

ZhongShan Zhang, email: [email protected]

Keywords: nasopharyngeal carcinoma; platelet distribution width; prognosis; overall survival

Received: July 31, 2017     Accepted: October 28, 2017     Published: November 20, 2017

ABSTRACT

Background: Previous studies have demonstrated that platelets play a multifaceted role in cancer progression and metastasis. However, the value of platelet indices for predicting survival in nasopharyngeal carcinoma (NPC) patients remains unknown. The aim of this study was to evaluate the predictive significance of platelet indices in NPC cases.

Materials and Methods: A total of 168 patients who were diagnosed with NPC between January 2011 and June 2012 were recruited. The optimal cut-off values for the platelet indices were determined using a receiver operating characteristic curve. The Kaplan-Meier method and Cox regression were used to evaluate the prognostic impact of the potential predictors.

Results: Of the 168 patients, high platelet distribution width (PDW) and platelet count (PLT) levels were observed in 81 (48.21%) and 68 (40.48%) of the patients, respectively. An increased PDW was associated with the depth of invasion (T stage, P = 0.019), lymph node metastasis (N stage, P = 0.026), and clinical stage (P < 0.001). Moreover, the survival analysis showed that the overall survival of the patients with a PDW > 16.3 fL or platelet count > 266 × 109/L was associated with a poorer prognosis (both P < 0.001). In the multivariate Cox regression model, the PDW (P < 0.001), PLT (P = 0.001), T stage (P < 0.001), N stage (P = 0.006), clinical stage (P = 0.005), and Epstein-Barr virus DNA (P = 0.039) were independent prognostic factors for the overall survival.

Conclusions: The PDW and PLT are easily available via a routine blood test, and our study showed that the PDW and PLT could be prognostic predictors in NPC patients. However, further studies are required to confirm this conclusion.


INTRODUCTION

Nasopharyngeal carcinoma (NPC) is the most frequently diagnosed tumor originating in the nasopharynx. It exhibits a distinct endemic distribution, with a particularly high incidence in Southern China and its surrounding regions [1]. Radiotherapy, with or without chemotherapy, is the standard treatment for patients with NPCs, while the prognosis for patients with tumor-node-metastasis (TNM) stages I and II is infusive [2]. Currently, TNM staging remains the gold standard for predicting the outcome of NPC [3]. However, even within the same staging category, there is variability in the patient outcomes because of the inability of the TNM system to reflect biological heterogeneity among the cancer cases [4]. Recent studies have reported a number of additional NPC prognostic markers, such as circulating Epstein-Barr virus (EBV) DNA loads [5], pretreatment serum lactate dehydrogenase [6], and microRNA signatures [7]. Due to the current limitations, such as detection difficulty, cost efficiency, and interlaboratory variability, the identification of appropriate and effective NPC prognostic markers is still of great value.

Multiple studies have suggested that platelets play a significant role in cancer progression and metastasis. For example, activated platelets promote cancer cell growth, aberrant angiogenesis, and invasion [8, 9]. Some platelet indices, such as the platelet count (PLT), mean platelet volume (MPV), platelet distribution width (PDW), and platelet-lymphocyte ratio (PLR), can be easily tested and have been demonstrated to be related to the prognosis of various cancers, including non-small cell lung cancer, breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, and laryngeal cancer [1015].

The MPV shows the average size of the platelets in the bloodstream, but it does not reflect the microscopically observed changes in the platelet size. The PDW is calculated as the coefficient of variation in the MPV, and high PDW values show that the change in the MPV is greater than normal [16]. Recently, some studies have found that an elevated pretreatment PLT or PLR was significantly and independently associated with a poor overall survival (OS) in NPC patients [17, 18].

These abovementioned studies only evaluated one or two biomarkers, without considering others. Moreover, the prognostic values of the PDW and MPV in NPC have not yet been established. Overall, the clinical significance of the platelet indices in the prognosis of NPC still remains unclear. Thus, the aim of this analysis was to investigate the prognostic value of the platelet indices mentioned above for patients with NPC, and to evaluate the correlation between these potential prognostic predictors and the clinical-pathological parameters.

RESULTS

The patient characteristics are outlined in Table 1. Overall, there were 119 (70.83%) male patients and 49 (29.17%) female patients, and the median age at diagnosis was 49 years old (range 29–76). In terms of the staging system, 28 cases (16.67%) were categorized as stage I or stage II, and 140 patients (83.33%) were stage III or stage IV (including stage IVA and IVB). The median follow-up time for the current cohort was 65.2 months (range 11.4–77.2). Finally, there were 112 cancer-related deaths at the time of the last follow-up.

Table 1: Baseline characteristics of the patients according to the PDW and PLT

Variables

Total n =168

PDW (fL)

P

PLT (×109/L)

P

≤16.3 (n=87)

>16.3 (n=81)

≤266 (n=100)

>266(n=68)

Age (years)

0.120

0.176

 < 60

146

79

67

84

62

 ≥ 60

22

8

14

16

6

Gender

0.252

0.773

  Male

119

65

54

70

49

  Female

49

22

27

30

19

Smoking

0.615

0.485

  Yes

59

29

30

33

26

  No

109

58

51

67

42

BMI(kg/m2)

0.091

0.491

 >23.7

82

37

45

51

31

 ≤23.7

86

50

36

49

37

Diabetes

0.290

0.053

  Yes

11

4

7

3

8

  No

157

83

74

97

60

Family history of NPC

0.735

0.245

 Yes

18

10

8

13

5

 No

150

77

73

87

63

 KPS

0.176

0.069

 ≤80

76

35

41

51

25

 > 80

92

52

40

49

43

Undifferentiated carcinoma

0.440

0.209

 Yes

84

46

38

54

30

 No

84

41

43

46

38

T stage

0.019

0.287

  T1-2

80

49

31

51

29

  T3-4

88

38

50

49

39

N stage

0.026

0.574

  N0-1

60

38

22

34

26

  N2-3

108

49

59

66

42

Clinical stage

< 0.001

0.574

 I-II

28

24

4

18

10

 III-IVB

140

63

77

82

58

 CCRT

0.519

0.086

 Yes

116

62

54

64

52

 No

52

25

27

36

16

IMRT

0.533

0.147

 Yes

126

67

59

79

47

 No

42

20

22

21

21

EBV DNA

0.096

0.194

 High

114

54

60

64

50

 low

54

33

21

36

18

Abbreviations: PDW, platelet distribution width; PLT, platelet count; BMI, Body mass index; UC, Undifferentiated carcinoma; KPS, Karnofsky performance score; CCRT, concurrent chemoradiotherapy; IMRT, intensity-modulated radiotherapy; EBV: Epstein–Barr.

According to the ROC curve analysis, the optimum cut-off values of the PLT, MPV, PDW, and PLR for the 5-year OS were 266 × 109/L (sensitivity 49.1%, specificity 76.8%), 10.6 fL(sensitivity 67.0%, specificity 76.8%), 16.3 fL(sensitivity 59.8%, specificity 75.0%), and 130.22 (sensitivity 78.6%, specificity 55.4%), respectively. The areas under the curve (AUCs) for the PLT, MPV, PDW, and PLR were 0.632 (95% CI: 0.554–0.705, P = 0.003), 0.713 (95% CI: 0.638–0.780, P < 0.001), 0.707 (95% CI: 0.631–0.774, P < 0.001), and 0.632 (95% CI: 0.554–0.705, P = 0.004), respectively (Figure 1 and Table 2). Consistent with a previous report [5], we used 1500 copies/mL as the cut-off value, and all of the patients were divided into either low (< 1500 copies/mL) or high (≥ 1500 copies/mL) pretreatment EBV DNA groups.

Table 2: Receiver operating characteristic curve analyses showing the utility of platelet indices for NPC

Platelet indices

Cut-off values

AUC

Sensibility

Specificity

95% CI

P-value

PLT (×109/L)

266

0.632

49.1

76.8

0.554-0.705

0.003

MPV (fL)

10.6

0.713

67.0

76.8

0.638-0.780

<0.001

PDW (fL)

16.3

0.707

59.8

75.0

0.631-0.774

<0.001

PLR

130.22

0.632

78.6

55.4

0.554-0.705

0.004

Abbreviations: see Table 1.

The ROC curves grouped by PLT, MPV, PDW and PLR.

Figure 1: The ROC curves grouped by PLT, MPV, PDW and PLR. ROC, receiver operating characteristic. Notes: The blue line represents PLT, the green line MPV, and the orange line PDW, and the imaginary line PLR.

The univariate Cox proportional hazards analyses revealed that the age at diagnosis (categorical variable), depth of invasion (T stage, T3–4 vs. T1–2), lymph node metastasis (N stage, N2–3 vs. N0–1), clinical stage (III–IV vs. I–II), platelet count (× 109/L) (> 266 vs. ≤ 266), MPV (fL) (> 10.6 vs. ≤ 10.6), PDW (fL) (> 16.3 vs. ≤ 16.3), PLR (> 130.22 vs. ≤ 130.22), and EBV DNA (high vs. low) were significantly associated with the OS (Table 3). However, the other parameters were not found to be correlated with the OS. All of the factors with P values of less than 0.05 in the univariate analyses were included in the subsequent multivariate Cox proportional hazards models.

Table 3: Result of the univariate analysis of overall survival in NPC patients

Characteristic

Hazard ratio

95% CI

P-value

Sex (male vs. female)

1.435

0.925-2.227

0.107

Age (≥60 vs. < 60 years)

1.719

1.035-2.852

0.036

Smoking (yes vs. no)

1.448

0.985-2.129

0.060

BMI (kg/m2) (>23.7 vs. ≤23.7)

1.335

0.917-1.943

0.132

Diabetes (yes vs. no)

1.767

0.856-3.647

0.124

Family history of NPC (yes vs. no)

0.986

0.563-1.728

0.962

KPS (≤80 vs. > 80)

1.168

0.805-1.696

0.414

Undifferentiated cancer (yes vs. no)

0.948

0.652-1.380

0.782

T stage(T3-4 vs. T1-2)

4.786

2.963-7.728

<0.001

N stage (N2–3 vs. N0–1)

4.099

2.536-6.651

<0.001

Clinical stage (III–IVB) vs. (I–II)

43.441

10.284 – 183.507

<0.001

CCRT (no vs. yes)

0.936

0.620-1.413

0.753

IMRT (no vs. yes)

1.292

0.839-1.988

0.245

PLT (>266 vs. ≤266) (×109/L)

1.979

1.361-2.877

<0.001

MPV (>10.6vs.≤10.6) fL

0.385

0.257-0.577

<0.001

PDW (>16.3 vs. ≤16.3) fL

2.986

2.030 – 4.394

<0.001

PLR (>130.22 vs. ≤130.22)

2.890

1.835 – 4.553

<0.001

EBV DNA (high vs. low) (copies/mL)

2.192

1.418 – 3.388

<0.001

Abbreviations: see Table 1.

In the multivariate analyses, we demonstrated that the PDW (HR 2.362, 95% CI: 1.554–3.590, P < 0.001), PLT (HR 2.017, 95% CI: 1.329–3.060, P = 0.001), T stage (HR 2.768, 95% CI: 1.603–4.779, P < 0.001), N stage (HR 2.165, 95% CI: 1.245–3.765, P = 0.006), clinical stage (HR 9.534, 95% CI: 1.998–45.498, P = 0.005), and EBV DNA (HR 1.619, 95% CI: 1.024–2.560, P = 0.039) were independent prognostic factors for NPC patients (Table 4).

Table 4: Multivariate Cox proportional hazards regression analysis

Characteristic

Hazard ratio

95% CI

P-value

Age (≥60 vs. < 60 years)

1.696

0.927-3.104

0.087

T stage(T3-4 vs. T1-2)

2.768

1.603-4.779

< 0.001

N stage (N2–3 vs. N0–1)

2.165

1.245-3.765

0.006

Clinical stage (III–IVB) vs. (I–II)

9.534

1.998-45.498

0.005

PLT (>266 vs. ≤266) (×109/L)

2.017

1.329-3.060

0.001

MPV (>10.6vs.≤10.6) fL

0.645

0.404-1.028

0.065

PDW (>16.3 vs. ≤16.3) fL

2.362

1.554-3.590

< 0.001

PLR (>130.22 vs. ≤130.22)

1.523

0.933-2.487

0.092

EBV DNA (high vs. low) (copies/mL)

1.619

1.024-2.560

0.039

Abbreviations: see Table 1.

The relationship between the PDW, PLT, and clinicopathological characteristics in NPC patients is shown in Table 1. There were no differences in the age distribution (P = 0.120), gender (P = 0.252), smoking history (P = 0.615), body mass index (P = 0.091), diabetes incidence (P = 0.290), family history of NPC (P = 0.735), KPS (P = 0.176), undifferentiated carcinoma incidence (P = 0.440), CCRT (P = 0.519), IMRT (P = 0.533), and EBV DNA (P = 0.096) with regard to the PDW. However, the PDW was associated with the T stage (P = 0.019), N stage (P = 0.026), and clinical stage (P < 0.001). Moreover, none of the clinicopathological features were significantly associated with the PLT.

The 5-year OS rate in this cohort was 69.05%, while the 5-year overall survival rates in those patients with PDW ≤ 16.3 fL and PDW > 16.3 fL were 86.21% and 50.62%, respectively. The results of the Kaplan-Meier and log-rank tests showed that the OS of the NPC patients with PDWs greater than 16.3 fL was shorter than that of the patients with PDWs less than or equal to 16.3 fL (P < 0.001). In addition, the OS of those patients with an increased PLT was also significantly shorter than in those with lower PLT levels (P < 0.001). Additionally, an adjusted Cox proportional hazard survival curve was used to investigate the effects of the PDW and PLT on survival. The outcomes were the same as those obtained while using the Kaplan-Meier method (Figure 2).

Figure 2:

Figure 2: Kaplan-Meier survival curves for NPC patients according to whether they show high or lower (A) PDW and (C) PLT levels. Multivariate Cox survival curves for NPC patients according to whether they show high or lower (B) PDW and (D) PLT levels.

DISCUSSION

To the best of our knowledge, this is the first study to investigate the prognostic value of the pretreatment PDW in NPC patients, and the first to systematically evaluate the relationships between the PLT, MPV, PDW, PLR, and the prognosis of NPC. This study revealed that a PDW with a cut-off 16.3 fL and a PLT with a cut-off 266 × 109/L had independent prognostic significances for the OS in NPC patients.

Platelets, a blood component, have been generally recognized as mediating tumor cell growth, metastasis, and angiogenesis. Activated platelets are capable of interacting with cancer cells through paracrine signaling or direct contact, consequently promoting tumor cell growth and survival [19]. An elevated blood platelet count is a common phenomenon in many kinds of malignancies and is linked to reduced survival in patients with various tumor types, including lung, breast, gastric, colorectal, pancreatic, brain, endometrial, and ovarian cancer. Although these results somewhat overlapped with those of the present study, our present study confirmed the key role of activated platelets in NPC using a simple platelet parameter. These data are also in accordance with the present knowledge that antiplatelet therapy is considered to be one portion of adjuvant cancer therapy [8].

It is now generally recognized that the PDW can not only measure platelet volume heterogeneity, but also reflect platelet activity. Recently, one study reported that a reduced PDW is an unfavorable predictive factor for non-small cell lung cancer patient survival [13]. Contrarily, an elevated PDW has been demonstrated to have a poor prognostic impact in melanoma [20], thyroid cancer [21], colorectal cancer [12], and laryngeal cancer [11]. However, the clinical value of the PDW has not yet been reported in NPC patients. Consistent with most studies, our results showed that an increased PDW was not only associated with the T stage, N stage, and clinical stage, but also significantly associated with a poorer OS in NPC patients. Moreover, an elevated PDW was an independent prognostic index for the OS in NPC patients.

New study has demonstrated that a high pretreatment MPV level in invasive breast cancer patients is a potential predictive factor and significant independent prognostic factor [14]. The study conducted by Omar et al. [22] reported that an increased MPV level may be used as a prognostic biomarker to estimate a poor OS in patients with lung cancer. However, the researchers found that the MPV levels were significantly increased in those patients with gastric ulcers and in the control subjects when compared with the gastric cancer patients [23]. This result seems to suggest that the MPV is a protective factor in gastric cancer. Moreover, this retrospective cohort study is the first study to investigate the prognostic value of pretreatment MPV levels in NPC patients. The results revealed that those patients with high MPV levels had 0.645 times the risk of death when compared to those with a low MPV levels, albeit the difference was not statistically significant after the multivariate analysis.

A previous study showed that elevated PLR values were associated with poor cancer-specific survival, OS, and distant metastasis free survival in patients with NPC [18]. In this study, the results of the multivariate analysis revealed that those patients with high PLR levels had 1.52 times the risk of death when compared to those with low PLR levels; however, the difference was not statistically significant (P > 0.092). The discrepancies above may be attributed to the different tumor tissues, different sample sizes, and variability in the measurement methods. Of course, there is the fact that we have additionally adjusted for some potential prognostic factors, including the PLT and PDW.

At present, the biological mechanisms underlying the correlation between the PDW, PLT, and tumorigenesis or progression in NPC remains poorly understood. The platelet volume is determined during both megakaryopoiesis and thrombopoiesis. In megakaryocytic maturation, the platelet production and platelet size can be modulated by cytokines, such as interleukin-6 (IL-6), the granulocyte colony stimulating factor, and the macrophage colony stimulating factor [24]. In addition, the tumor-associated production of the granulocyte-macrophage colony stimulating factor or thrombopoietin mediated by IL-6 is considered to be responsible for the increase in the PLT observed in cancer patients [25]. There is consistent evidence that IL-6 is overexpressed in EBV-infected nasopharyngeal epithelial cancer cells [26]. By activating the IL-6-associated transcription factors (nuclear factor kappa-light-chain-enhancer of activated B cells and signal transducer and activator of transcription 3), the enhanced circulating IL-6 not only contributes to NPC’s proliferative properties but also mediates platelet generation. Furthermore, elevated platelet levels promote cancer progression and metastasis by protecting the circulating tumor cells from immune surveillance and death [27]. Another reasonable mechanism is that platelets facilitate a hypercoagulable state in cancer. The increased number of platelets produces a procoagulant microenvironment that enables the cancer cells to cover themselves with platelets and escape the host’s immune system [28].

There were some study limitations that should be addressed. First, our study may have been restricted by its retrospective design. Second, the number of subjects included in our study was relatively small, and the power of the statistical analysis was relatively weak; therefore, larger studies are warranted to validate our findings. Finally, all of the included studies were from Chinese populations; consequently, the conclusion might be limited to East Asian individuals.

In summary, the current study results suggest that the PDW and PLT could be new independent prognostic factors in NPC patients. However, additional investigations are warranted to fully understand the potential mechanism. These easily measured and inexpensive biomarkers may be used as practical predictors in daily clinical practice in the future.

MATERIALS AND METHODS

Patient recruitment and data collection

A total of 168 NPC patients were admitted to the Department of Oncology of the Affiliated Hospital of XiangNan University between January 2011 and June 2012. All of the included patients met the following criteria: (1) histological confirmation of NPC and no previous anticancer treatment; (2) no distant metastasis; (3) had received complete blood differential count records within 3 days prior to treatment; (4) available clinical information. The exclusion criteria were as follows: (1) Karnofsky Performance Scale (KPS) score < 70; (2) had other previously treated and/or synchronous malignancies; (3) the presence of a concomitant disease that can affect the platelet count, including severe hypertension, autoimmune disease, inflammation, liver cirrhosis, splenic disease, and a history of blood transfusion.

The pretreatment hematological parameters were measured via a Beckman Coulter LH750 automated analyzer (Beckman Coulter, California, USA). The EBV DNA concentrations were obtained using a quantitative polymerase chain reaction (DA 7600; Da-an, Zhongshan, China) before treatment, as previously reported [29].

This study was approved by the Institutional Review Board of the Affiliated Hospital of XiangNan University.

Staging, treatment, and follow-up

All of the patients were reassigned based on the 7th Edition of the American Joint Committee on Cancer (AJCC) cancer staging system for NPC as assessed via the compilation of the magnetic resonance imaging, chest radiograph, abdominal ultrasonography, whole-body bone scanning, and/or positron emission tomography-computed tomography results.

The treatment strategies for all of the patients were based on the National Comprehensive Cancer Network Guidelines. All of the patients received definitive radiotherapy (RT) using three-dimensional conformal RT (3D-CRT) and intensity-modulated RT (IMRT) techniques. Those patients with stage I–II malignancies were treated by RT alone or with concurrent chemoradiotherapy (CCRT). Those patients with stage III–IVB malignancies were treated with CCRT with or without induction/adjuvant chemotherapy. The CCRT regimen was the use of 30 mg/m2 of cisplatin every week for 6–7 cycles during the RT period. The regimen for the induction/adjuvant chemotherapy included cisplatin (75 mg/m2 on day 1) and 5-fluorouracil (1,000 mg/m2 via a 96-hour continuous infusion from days 2–5) administrated every 21 days for 2-3 cycles.

After completing treatment, the patients were followed up every three months for the first three years, with the intervals gradually increasing to 6–12 months after three years. The last follow-up date was June 30, 2017. The overall survival (OS) was calculated from the date of the diagnosis to the date of death or the last follow-up. The median follow-up time was 65.2 months.

Statistical analysis

The statistical analysis was performed using the Statistical Program for Social Sciences (SPSS) software, version 16.0 (SPSS Inc., Chicago, IL, USA). The optimal cut-off values of the platelet indices were determined by using a receiver operating characteristic (ROC) curve. The univariate and multivariate survival analyses were determined using the Cox proportional hazards model. The correlations between the potential prognostic predictors and the clinicopathological variables were evaluated via the chi-squared test. The Kaplan-Meier method and the corresponding log rank test were used for the survival analyses of the categorical variables. All of the tests were two-tailed, and the level of significance was P < 0.05.

Author contributions

XC Xie and XC Zeng mainly participated in literature search, study design, writing and critical revision. SJ Cao, XM Hu, QJ S, D Li and SY Zhou mainly participated in data collection, data analysis and data interpretation. P Gu and ZS Zhang contributed to the technical and material support. All authors read and approved the final manuscript.

CONFLICTS OF INTEREST

None of the authors have any conflicts of interest to declare.

FUNDING

This study was funded by the project of XiangNan University (2015XB17).

REFERENCES

1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015; 65: 87-108. https://doi.org/10.3322/caac.21262.

2. Maingon P, Blanchard P, Bidault F, Calmels L. [Radiotherapy for nasopharyngeal carcinoma]. [Article in French]. Cancer Radiother. 2016; 20: S104-9. https://doi.org/10.1016/j.canrad.2016.07.031.

3. Lee AW, Ma BB, Ng WT, Chan AT. Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol. 2015; 33: 3356-64. https://doi.org/10.1200/jco.2015.60.9347.

4. Chua ML, Wee JT, Hui EP, Chan AT. Nasopharyngeal carcinoma. Lancet. 2016; 387: 1012-24. https://doi.org/10.1016/s0140-6736(15)00055-0.

5. Lin JC, Wang WY, Chen KY, Wei YH, Liang WM, Jan JS, Jiang RS. Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med. 2004; 350: 2461-70. https://doi.og/10.1056/NEJMoa032260.

6. Wan XB, Wei L, Li H, Dong M, Lin Q, Ma XK, Huang PY, Wen JY, Li X, Chen J, Ruan DY, Lin ZX, Chen ZH, et al. High pretreatment serum lactate dehydrogenase level correlates with disease relapse and predicts an inferior outcome in locally advanced nasopharyngeal carcinoma. Eur J Cancer. 2013; 49: 2356-64. https://doi.org/10.1016/j.ejca.2013.03.008.

7. Liu N, Chen NY, Cui RX, Li WF, Li Y, Wei RR, Zhang MY, Sun Y, Huang BJ, Chen M, He QM, Jiang N, Chen L, et al. Prognostic value of a microRNA signature in nasopharyngeal carcinoma: a microRNA expression analysis. Lancet Oncol. 2012; 13: 633-41. https://doi.org/10.1016/s1470-2045(12)70102-x.

8. Mezouar S, Frere C, Darbousset R, Mege D, Crescence L, Dignat-George F, Panicot-Dubois L, Dubois C. Role of platelets in cancer and cancer-associated thrombosis: experimental and clinical evidences. Thromb Res. 2016; 139: 65-76. https://doi.org/10.1016/j.thromres.2016.01.006.

9. Buergy D, Wenz F, Groden C, Brockmann MA. Tumor-platelet interaction in solid tumors. Int J Cancer. 2012; 130: 2747-60. https://doi.org/10.1002/ijc.27441.

10. Zhang X, Cui MM, Fu S, Li LL, Liu YS, Liu ZP, Liu T, Wang RT, Yu KJ. Platelet distribution width correlates with prognosis of gastric cancer. Oncotarget. 2017; 8: 20213-9. https://doi.org/10.18632/oncotarget.15561.

11. Zhang H, Liu L, Fu S, Liu YS, Wang C, Liu T, Liu ZP, Wang RT, Yu KJ. Higher platelet distribution width predicts poor prognosis in laryngeal cancer. Oncotarget. 2017; 8: 48138-44. https://doi.org/10.18632/oncotarget.18306.

12. Song X, Zhu H, Pei Q, Tan F, Li C, Zhou Z, Zhou Y, Yu N, Li Y, Pei H. Significance of inflammation-based indices in the prognosis of patients with non-metastatic colorectal cancer. Oncotarget. 2017; 8: 45178-89. https://doi.org/10.18632/oncotarget.16774.

13. Cui MM, Li N, Liu X, Yun ZY, Niu Y, Zhang Y, Gao B, Liu T, Wang RT. Platelet distribution width correlates with prognosis of non-small cell lung cancer. Sci Rep. 2017; 7: 3456. https://doi.org/10.1038/s41598-017-03772-z.

14. Gu M, Zhai Z, Huang L, Zheng W, Zhou Y, Zhu R, Shen F, Yuan C. Pre-treatment mean platelet volume associates with worse clinicopathologic features and prognosis of patients with invasive breast cancer. Breast Cancer. 2016; 23: 752-60. https://doi.org/10.1007/s12282-015-0635-6.

15. Liu P, Zhu Y, Liu L. Elevated pretreatment plasma D-dimer levels and platelet counts predict poor prognosis in pancreatic adenocarcinoma. Onco Targets Ther. 2015; 8: 1335-40. https://doi.org/10.2147/ott.s82329.

16. Thompson CB, Jakubowski JA. The pathophysiology and clinical relevance of platelet heterogeneity. Blood. 1988; 72: 1-8.

17. Chen YP, Chen C, Mai ZY, Gao J, Shen LJ, Zhao BC, Chen MK, Chen G, Yan F, Huang TY, Xia YF. Pretreatment platelet count as a predictor for survival and distant metastasis in nasopharyngeal carcinoma patients. Oncol Lett. 2015; 9: 1458-66. https://doi.org/10.3892/ol.2015.2872.

18. Jiang R, Zou X, Hu W, Fan YY, Yan Y, Zhang MX, You R, Sun R, Luo DH, Chen QY, Huang PY, Hua YJ, Guo L, et al. The elevated pretreatment platelet-to-lymphocyte ratio predicts poor outcome in nasopharyngeal carcinoma patients. Tumour Biol. 2015; 36: 7775-87. https://doi.org/10.1007/s13277-015-3505-0.

19. Sharma D, Brummel-Ziedins KE, Bouchard BA, Holmes CE. Platelets in tumor progression: a host factor that offers multiple potential targets in the treatment of cancer. J Cell Physiol. 2014; 229: 1005-15. https://10.1002/jcp.24539.

20. Li N, Diao Z, Huang X, Niu Y, Liu T, Liu ZP, Wang RT, Yu KJ. Increased platelet distribution width predicts poor prognosis in melanoma patients. Sci Rep. 2017; 7: 2970. https://doi.org/10.1038/s41598-017-03212-y.

21. Yu YJ, Li N, Yun ZY, Niu Y, Xu JJ, Liu ZP, Liu T, Wang RT, Yu KJ. Preoperative mean platelet volume and platelet distribution associated with thyroid cancer. Neoplasma. 2017; 64: 594-8. https://doi.org/10.4149/neo_2017_414.

22. Omar M, Tanriverdi O, Cokmert S, Oktay E, Yersal O, Pilanci KN, Menekse S, Kocar M, Sen CA, Ordu C, Goksel G, Meydan N, Barutca S. Role of increased mean platelet volume (MPV) and decreased MPV/platelet count ratio as poor prognostic factors in lung cancer. Clin Respir J. 2016. https://doi.org/10.1111/crj.12605.

23. Yun ZY, Li N, Zhang X, Zhang H, Bu Y, Sun Y, Liu T, Wang RT, Yu KJ. Mean platelet volume, platelet distribution width and carcinoembryonic antigen to discriminate gastric cancer from gastric ulcer. Oncotarget. 2017; 8: 62600-5. https://doi.org/10.18632/oncotarget.15898.

24. Kaushansky K. Growth factors and hematopoietic cell fate. A new feature: controversies in hematology. Blood. 1998; 92: 345-4.

25. Kaser A, Brandacher G, Steurer W, Kaser S, Offner FA, Zoller H, Theurl I, Widder W, Molnar C, Ludwiczek O, Atkins MB, Mier JW, Tilg H. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood. 2001; 98: 2720-5.

26. Zhang G, Tsang CM, Deng W, Yip YL, Lui VW, Wong SC, Cheung AL, Hau PM, Zeng M, Lung ML, Chen H, Lo KW, Takada K, et al. Enhanced IL-6/IL-6R signaling promotes growth and malignant properties in EBV-infected premalignant and cancerous nasopharyngeal epithelial cells. PLoS One. 2013; 8: e62284. https://doi.org/10.1371/journal.pone.0062284.

27. Meikle CK, Kelly CA, Garg P, Wuescher LM, Ali RA, Worth RG. Cancer and thrombosis: the platelet perspective. Front Cell Dev Biol. 2016; 4: 147. https://doi.org/10.3389/fcell.2016.00147.

28. Santilli F, Boccatonda A, Davi G. Aspirin, platelets, and cancer: the point of view of the internist. Eur J Intern Med. 2016; 34: 11-20. https://doi.org/10.1016/j.ejim.2016.06.004.

29. Shao JY, Li YH, Gao HY, Wu QL, Cui NJ, Zhang L, Cheng G, Hu LF, Ernberg I, Zeng YX. Comparison of plasma Epstein-Barr virus (EBV) DNA levels and serum EBV immunoglobulin A/virus capsid antigen antibody titers in patients with nasopharyngeal carcinoma. Cancer. 2004; 100: 1162-70. https://doi.org/10.1002/cncr.20099.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 22528