Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2025 Jun 15;15(6):2451–2468. doi: 10.62347/MQBC5709

Diagnostic and prognostic value of coagulation markers and platelet-derived growth factor-BB in evaluating intensity-modulated radiotherapy efficacy in nasopharyngeal carcinoma

Haizhong Zhang 1, Deli Ye 2
PMCID: PMC12256396  PMID: 40667550

Abstract

Objectives: To evaluate the diagnostic and prognostic value of coagulation markers - including activated partial thromboplastin time (APTT), prothrombin time (PT), fibrinogen (FIB), platelet count (PLT), and D-dimer (DD) - and platelet-derived growth factor-BB (PDGF-BB) in patients with nasopharyngeal carcinoma (NPC) undergoing intensity-modulated radiotherapy (IMRT). Methods: A total of 210 NPC patients receiving IMRT and 160 healthy controls were enrolled. Baseline levels of PDGF-BB and coagulation markers were compared between groups. The association of PDGF-BB with clinical staging was analyzed, and receiver operating characteristic (ROC) curve analysis was used to assess its diagnostic performance. Cox regression analyses were performed to identify independent predictors of five-year survival. A dynamic nomogram was developed to provide individualized survival predictions. Results: NPC patients exhibited significantly higher levels of PDGF-BB, APTT, PT, FIB, PLT, and DD compared to healthy controls (all P < 0.001). PDGF-BB was positively correlated with TNM stage (stage III/IV vs. I/II, P < 0.001), T stage (P = 0.005), and N stage (P = 0.020). Multivariate Cox regression identified low PDGF-BB (< 628.18) (HR = 0.492, P = 0.009), low DD (< 746.1) (HR = 0.456, P = 0.002), age 51-64 years (HR = 2.057, P = 0.032) and ≥ 65 years (HR = 4.138, P < 0.001), EBV DNA negativity (HR = 0.273, P = 0.012), and TNM stage III/IV (HR = 3.042, P = 0.023) as independent prognostic factors. Conclusions: PDGF-BB and DD, alongside age, EBV DNA status, and TNM stage, are promising biomarkers for NPC prognosis. A dynamic nomogram integrating these factors offers accurate survival prediction and supports personalized treatment strategies in NPC management.

Keywords: Nasopharyngeal carcinoma, intensity-modulated radiotherapy, PDGF-BB, coagulation markers, prognosis

Introduction

Nasopharyngeal carcinoma (NPC), a malignancy arising in the nasopharyngeal epithelium, exhibits a distinct geographic distribution, with high prevalence in Southeast Asia-particularly in southern China, where incidence rates reach 20-50 per 100,000 in regions such as Guangdong, Hong Kong, and Macao [1,2]. Its pathogenesis is multifactorial, involving genetic predisposition, environmental exposure, Epstein-Barr virus (EBV) infection, and lifestyle factors including smoking and alcohol consumption [3]. Intensity-modulated radiotherapy (IMRT) remains the cornerstone of NPC treatment due to its ability to deliver precise radiation doses to the tumor while minimizing damage to adjacent healthy tissues [4,5]. However, the insidious onset and non-specific early symptoms often result in late-stage diagnoses, complicating treatment and worsening prognosis [6].

Despite advances in imaging and molecular diagnostics, early detection and accurate prognostic assessment of NPC remain challenging [7]. Current diagnostic modalities, such as computed tomography (CT), magnetic resonance imaging (MRI), and EBV DNA quantification, provide essential information but have limited utility for early-stage identification and real-time disease monitoring [8,9]. Traditional prognostic indicators-such as TNM staging and patient age-demonstrate variable predictive power across populations [10,11]. Recent studies have highlighted the potential involvement of coagulation markers and platelet-derived growth factor-BB (PDGF-BB) in tumor biology, implicating them in cancer progression, metastasis, and treatment responsiveness [12]. This study aims to evaluate the diagnostic and prognostic value of coagulation markers and PDGF-BB in NPC patients undergoing IMRT, with the goal of improving individualized treatment strategies.

The role of the coagulation system extends beyond hemostasis and into cancer biology. Emerging evidence indicates that it contributes to multiple stages of tumorigenesis, including initiation, progression, and metastasis [13]. PDGF-BB, a growth factor secreted by platelets and certain tumor cells, plays a crucial role in cell proliferation, migration, and angiogenesis [14]. In malignancies, PDGF-BB acts as a double-edged sword: it promotes tumor angiogenesis and sustains tumor growth by enhancing vascularization, while also remodeling the tumor microenvironment to facilitate metastasis and therapy resistance [15]. Previous research has suggested associations between PDGF-BB levels and tumor aggressiveness, metastatic potential, and treatment outcomes, positioning it as a promising biomarker for diagnosis and prognosis [16].

Although prior studies have explored the roles of coagulation indices and PDGF-BB in various malignancies, their specific functions in NPC remain unclear. This study investigates the relationships between these biomarkers and NPC clinical characteristics, treatment responses, and 5-year survival outcomes by comparing their levels in NPC patients and healthy controls. The findings are expected to provide new insights into the diagnostic and prognostic utility of coagulation markers and PDGF-BB in NPC, potentially informing more effective and personalized management strategies.

Methods and materials

Sample size calculation

Based on a previously reported 5-year mortality rate of approximately 30% for nasopharyngeal carcinoma (NPC) [17], the required sample size was calculated using the formula: N = Z2 [P (1-P)]/E2, where P = 0.30, E = 0.05, Z = 1.96. The calculated sample size was approximately 323. However, the actual number of participants was determined by the availability of eligible clinical samples.

General information

This study included 210 patients diagnosed with NPC and treated at Longyou County People’s Hospital between January 2017 and December 2019. Additionally, 160 healthy individuals undergoing routine physical examinations during the same period were enrolled as the control group. The study was approved by the Medical Ethics Committee of Longyou County People’s Hospital (Figure 1).

Figure 1.

Figure 1

Sample screening flow chart.

Inclusion and exclusion criteria

Inclusion criteria: (1) Histopathological confirmation of NPC [18]. (2) Age between 18 and 70 years. (3) Scheduled to receive IMRT. (4) Availability of complete clinical, imaging, and pathological data required for the study.

Exclusion Criteria: (1) Presence of severe cardiovascular or cerebrovascular disease, hepatic/renal dysfunction, diabetes, or other conditions affecting coagulation function. (2) History of primary or secondary coagulation disorders. (3) History of malignancies other than NPC. (4) Pregnancy or lactation. (5) Current use of anticoagulant or antiplatelet medications. (6) Stage IVB NPC.

Treatment regimens

Patients received IMRT via linear accelerators (e.g., Varian TrueBeam). Treatment protocols varied by clinical stage:

Stage I (T1N0M0): Radical radiotherapy alone; 66-70 Gy to the nasopharynx and 54-60 Gy to the cervical lymph nodes.

Stage II (T0-2N0-1M0): For T2N1 cases, concurrent chemoradiotherapy (typically cisplatin-based) is recommended; others may receive radiotherapy alone or in combination with chemotherapy. Radiation doses are the same as in Stage I.

Stages III-IVA (locally advanced): Induction chemotherapy (e.g., TP or GP regimen) followed by concurrent chemoradiotherapy, or concurrent chemoradiotherapy plus adjuvant chemotherapy. Radiation doses are 66-70 Gy for the nasopharynx and 54-60 Gy for the cervical lymph nodes, including bilateral retropharyngeal and level II-Vb lymph nodes.

Clinical data collection

Comprehensive baseline data were collected to compare clinical characteristics between NPC patients and healthy controls, providing a foundation for subsequent analysis.

Demographic and behavioral variables included age, sex, education level, smoking history, alcohol consumption history, TNM stage, tumor differentiation, and EBV DNA status.

All data were obtained from the hospital’s electronic medical records to ensure accuracy and completeness.

Laboratory indicator collection and testing

Laboratory indicators - including PDGF-BB, activated partial thromboplastin time (APTT), prothrombin time (PT), fibrinogen (FIB), platelet count (PLT), and D-dimer (DD), were measured using standardized protocols and equipment.

(1) PDGF-BB (pg/mL): Measured by ELISA (ELISA Bio, Shanghai; Batch No. ml105299). (2) PLT was determinedusing an automated hematology analyzer (Sysmex XN-9000, Japan). (3) APTT, PT, FIB, and DD were measured using a coagulation analyzer (Sysmex CS-5100, Japan).

All blood samples were collected upon patient admission, and processed and stored following standardized protocols to ensure reliability.

Clinical prognosis follow-up

Five-year survival outcomes were determined through structured follow-up. Patients were followed every 6 months for the first year, and annually thereafter, up to 5 years post-treatment. Survival status and clinical progress were recorded at each visit.

Outcome measurements

Primary outcome measures

Comparison of the baseline levels of coagulation markers (APTT, PT, FIB, PLT, DD) and PDGF-BB between healthy individuals and NPC patients.

Identification of independent prognostic factors influencing five-year survival in NPC patients using Cox regression analysis.

Secondary outcome measures

Comparison of coagulation markers and PDGF-BB levels among NPC patients with different clinical characteristics (e.g., age, gender, smoking history, etc.).

Comparison of expression levels of coagulation markers and PDGF-BB in patients with varying treatment responses (complete response [CR], partial response [PR], stable disease [SD], and progressive disease [PD]).

Evaluation of the expression and predictive value of coagulation markers and PDGF-BB in patients with improved treatment outcomes.

Statistical analysis

Statistical analyses were conducted using SPSS 26.0 and R 4.3.3. The Kolmogorov-Smirnov (K-S) test was first applied to assess data normality. For normally distributed continuous variables presented as (mean ± SD) deviation, independent samples t-tests were used to compare differences between groups. Continuous variables with non-normal distribution were expressed as median with interquartile range (IQR, 25th-75th percentile), and group comparisons were performed using the Mann-Whitney U test.

Categorical variables expressed as numbers (percentages) (e.g., sex, EBV DNA status) were analyzed using the chi-square test.

Cox proportional hazards regression (using the survival package in R) was employed to identify independent prognostic factors for five-year survival. Hazard ratios (HRs) and corresponding P-values were reported. A dynamic nomogram was constructed using the DynNom package in R to integrate clinical and laboratory variables, enabling real-time, individualized survival prediction. All tests were two-sided, and P < 0.05 was considered statistically significant.

Results

Comparison of baseline characteristics

There were no significant differences in age (P = 0.849), gender (P = 0.541), education level (P = 0.653), smoking history (P = 0.234), or alcohol consumption (P = 0.270) between NPC patients and healthy controls. Tumor-related characteristics such as T stage, N stage, TNM stage, and differentiation degree also showed no significant differences. However, the EBV DNA positivity rate was significantly higher in the NPC group than in controls (all P < 0.001; Table 1).

Table 1.

Comparison and presentation of clinical characteristics

Variable Total Control group (n = 160) NPC group (n = 210) χ2 P
Age
    ≤ 50 135 61 74 0.327 0.849
    51-64 152 64 88
    ≥ 65 83 35 48
Gender
    Male 234 104 130 0.374 0.541
    Female 136 56 80
Educational level
    ≤ Junior high school 149 61 88 0.852 0.653
    Senior high school 152 70 82
    ≥ University 69 29 40
Smoking history (cigarettes/day)
    < 10 202 93 109 1.417 0.234
    ≥ 10 168 67 101
Alcohol consumption history (ml/d)
    < 1000 224 102 122 1.216 0.270
    ≥ 1000 146 58 88
T staging
    T1 80
    T2 66
    T3 55
    T4 9
N staging
    N0 46
    N1 38
    N2 80
    N3 46
TNM staging
    I 18
    II 32
    III 105
    IV 55
Differentiation degree
    Well differentiated 40
    Moderately differentiated 47
    Poorly differentiated 123
EBV DNA positivity
    Yes 184 16 168 177.994 < 0.001
    No 186 144 42

Notes: TNM, Tumor, Node, Metastasis; T, Tumor; N, Node; EBV, Epstein-Barr Virus.

Comparison of the baseline levels of coagulation markers and PDGF-BB

NPC patients showed significantly elevated levels of PDGF-BB compared to healthy controls (P < 0.001). Similarly, APTT, PT, FIB, PLT, and DD levels were all significantly higher in the NPC group (all P < 0.001), suggesting notable coagulation and platelet-related changes in NPC pathophysiology (Table 2).

Table 2.

Comparison of the distribution of baseline levels of coagulation markers and PDGF-BB

Variable Total Control group (n = 160) NPC group (n = 210) Statistic P
PDGF-BB (pg/mL) 284.40 ± 251.34 30.08 ± 10.04 478.17 ± 155.61 36.354 < 0.001
APTT (s) 37.56 ± 4.51 35.33 ± 2.94 39.26 ± 4.76 9.198 < 0.001
PT (s) 12.50 [11.90, 13.10] 12.10 [11.50, 12.50] 12.90 [12.30, 13.50] 9.886 < 0.001
FIB (g/L) 3.57 ± 0.99 3.12 ± 0.53 3.91 ± 1.13 8.121 < 0.001
PLT (×109/L) 230.82 ± 66.69 203.11 ± 42.51 251.93 ± 73.76 7.477 < 0.001
DD (ng/mL) 342.75 [216.95, 626.59] 226.10 [170.05, 285.28] 593.92 [394.48, 827.78] 12.991 < 0.001

Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer.

Comparison of coagulation markers and PDGF-BB in NPC patients with different clinical characteristics

NPC patients were stratified into low and high PDGF-BB expression groups. No significant differences were found in age, sex, education level, smoking, alcohol consumption, EBV DNA status, or tumor differentiation (all P > 0.05). However, significant associations were observed with TNM stage (P < 0.001), T stage (P = 0.005), N stage (P = 0.020), and receipt of chemotherapy (P < 0.001), suggesting a potential link between PDGF-BB expression and disease severity or treatment need (Table 3). Comparisons for other coagulation markers are shown in Tables S1, S2, S3, S4, S5.

Table 3.

Analysis of differences in clinical characteristics stratified by PDGF-BB expression levels

Variable Total PDGF-BB (pg/mL) χ2 P

Low expression (n = 105) High expression (n = 105)
Age
    ≤ 50 74 37 37 4.636 0.098
    51-64 88 50 38
    ≥ 65 48 18 30
Gender
    Male 130 68 62 0.727 0.394
    Female 80 37 43
Educational level
    ≤ Junior high school 88 45 43 0.884 0.643
    Senior high school 82 38 44
    ≥ University 40 22 18
Smoking history (cigarettes/day)
    < 10 109 60 49 2.308 0.129
    ≥ 10 101 45 56
Alcohol consumption history (ml/d)
    < 1000 122 66 56 1.956 0.162
    ≥ 1000 88 39 49
EBV DNA positivity
    Yes 168 81 87 1.071 0.301
    No 42 24 18
Differentiation degree
    Well differentiated 40 17 23 1.320 0.517
    Moderately differentiated 47 23 24
    Poorly differentiated 123 65 58
TNM staging
    I 18 18 0 21.534 < 0.001
    II 32 18 14
    III 105 45 60
    IV 55 24 31
T staging
    T1 80 47 33 12.955 0.005
    T2 66 37 29
    T3 55 20 35
    T4 9 1 8
N staging
    N0 46 32 14 9.791 0.020
    N1 38 16 22
    N2 80 34 46
    N3 46 23 23
Receive chemotherapy
    Yes 165 64 85 10.189 0.001
    No 45 41 20

Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; EBV, Epstein-Barr Virus; TNM, Tumor, Node, Metastasis; T, Tumor; N, Node.

Comparison of coagulation markers and PDGF-BB expression across clinical stages

PDGF-BB levels differed significantly across TNM stages, with stage IV patients showing significantly higher levels than stage I patients (P < 0.001). In contrast, APTT, PT, FIB, PLT, and DD levels showed no significant differences among stages (all P > 0.05; Figure 2).

Figure 2.

Figure 2

Expression of coagulation markers and PDGF-BB across clinical stages. A: Distribution of PDGF-BB levels across different TNM stages. B: Distribution of APTT levels across different TNM stages. C: Distribution of PT levels across different TNM stages. D: Distribution of FIB levels across different TNM stages. E: Distribution of PLT levels across different TNM stages. F: Distribution of DD levels across different TNM stages. Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer. ****P < 0.0001.

Comparison of coagulation markers and PDGF-BB expression between early- and advanced-stage NPC patients

PDGF-BB levels were significantly higher in advanced-stage (III/IV) than in early-stage (I/II) patients (P < 0.001). PT and FIB also showed significant differences between these groups (both P < 0.05). However, no significant differences were observed in APTT, PLT, or DD (all P > 0.05; Figure 3).

Figure 3.

Figure 3

Expression differences of coagulation markers and PDGF-BB in early- and advanced-stage patients. A: Distribution of PDGF-BB in Stage I/II and Stage III/IV patients. B: Distribution of APTT in Stage I/II and Stage III/IV patients. C: Distribution of PT in Stage I/II and Stage III/IV patients. D: Distribution of FIB in Stage I/II and Stage III/IV patients. E: Distribution of PLT in Stage I/II and Stage III/IV patients. F: Distribution of DD in Stage I/II and Stage III/IV patients. Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer. *P < 0.05, ****P < 0.0001.

Comparison of coagulation markers and PDGF-BB expression across different treatment responses

No significant differences were observed in the expression levels of PDGF-BB, APTT, PT, FIB, PLT, or DD among patients with different treatment responses (all P > 0.05; Figure 4).

Figure 4.

Figure 4

Expression of coagulation markers and PDGF-BB in patients with different treatment responses. A: Expression of PDGF-BB in patients with different treatment outcomes. B: Expression of APTT in patients with different treatment outcomes. C: Expression of PT in patients with different treatment outcomes. D: Expression of FIB in patients with different treatment outcomes. E: Expression of PLT in patients with different treatment outcomes. F: Expression of DD in patients with different treatment outcomes. Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer; CR, Complete Response; PR, Partial Response; SD, Stable Disease; PD, Progressive Disease.

Comparison of coagulation markers and PDGF-BB expression in patients with improved therapeutic outcomes

Similarly, no statistically significant differences were found in the levels of PDGF-BB, APTT, PT, FIB, PLT, or DD between patients who showed improved treatment outcomes and those who did not (all P > 0.05), as shown in Figure 5.

Figure 5.

Figure 5

Expression of coagulation markers and PDGF-BB in patients with different treatment responses. A: Expression of PDGF-BB in patients with improved outcomes versus disease progression. B: Expression of APTT in patients with improved outcomes versus disease progression. C: Expression of PT in patients with improved outcomes versus disease progression. D: Expression of FIB in patients with improved outcomes versus disease progression. E: Expression of PLT in patients with improved outcomes versus disease progression. F: Expression of DD in patients with improved outcomes versus disease progression. Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer; CR, Complete Response; PR, Partial Response; SD, Stable Disease; PD, Progressive Disease.

Prognostic value of coagulation markers and PDGF-BB for five-year survival

Overall survival outcomes

During the five-year follow-up period, 72 patients died, yielding a mortality rate of 34.29%. The average time to death was 19.83 months. These findings provided a foundation for assessing the prognostic significance of coagulation markers and PDGF-BB in NPC.

Univariate cox regression analysis

Univariate Cox regression revealed several significant predictors of five-year survival:

Each 1-unit increase of PDGF-BB levels was associated with a higher mortality risk (HR = 1.004, P < 0.001). DD also showed significant prognostic value (HR = 1.001, P < 0.001) Increased risk was observed in patients aged 51-64 years (HR = 1.998, P = 0.039) and ≥ 65 years (HR = 5.801, P < 0.001). EBV DNA positivity was associated with worse survival (HR = 0.201, P = 0.002). TNM stage III/IV: Significantly worse prognosis (HR = 5.486, P < 0.001). As for T stage T3/T4, increased mortality risk (HR = 1.855, P = 0.010).

Other variables - including sex, education level, smoking history, alcohol consumption, tumor differentiation, N stage, and chemotherapy-did not significantly impact survival (all P > 0.05; Table 4).

Table 4.

Univariate Cox regression analysis: prognostic value of coagulation markers and PDGF-BB for five-year survival

Variable Beta Std Err P HR Lower Upper
PDGF-BB 0.004 0.001 < 0.001 1.004 1.003 1.006
APTT 0.001 0.024 0.981 1.001 0.954 1.049
PT 0.069 0.150 0.647 1.071 0.799 1.436
FIB -0.093 0.104 0.369 0.911 0.744 1.116
PLT -0.001 0.002 0.683 0.999 0.996 1.002
DD 0.001 < 0.001 < 0.001 1.001 1.001 1.002
Age
    ≤ 50
    51-64 0.692 0.336 0.039 1.998 1.035 3.858
    ≥ 65 1.758 0.332 < 0.001 5.801 3.024 11.127
Gender
    Male
    Female -0.202 0.248 0.414 0.817 0.503 1.327
Educational level
    ≤ Junior high school
    Senior high school 0.026 0.265 0.921 1.027 0.611 1.726
    ≥ University 0.109 0.318 0.731 1.116 0.598 2.081
Smoking history (cigarettes/day)
    < 10
    ≥ 10 0.317 0.237 0.181 1.373 0.863 2.182
Alcohol consumption history (ml/d)
    < 1000
    ≥ 1000 0.228 0.237 0.336 1.256 0.790 1.996
EBV DNA positivity
    Yes
    No -1.603 0.515 0.002 0.201 0.073 0.552
Differentiation degree
    Well differentiated
    Moderately differentiated -0.202 0.392 0.607 0.817 0.379 1.763
    Poorly differentiated 0.210 0.314 0.504 1.234 0.667 2.284
TNM staging
    I + II
    III + IV 1.702 0.464 < 0.001 5.486 2.210 13.622
T staging
    T1 + T2
    T3 + T4 0.618 0.239 0.010 1.855 1.160 2.965
N staging
    N0
    N1-3 0.598 0.328 0.068 1.819 0.957 3.457
Receive chemotherapy
    Yes
    No -0.357 0.287 0.213 0.700 0.399 1.227

Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; APTT, Activated Partial Thromboplastin Time; PT, Prothrombin Time; FIB, Fibrinogen; PLT, Platelet Count; DD, D-dimer; EBV, Epstein-Barr Virus; TNM, Tumor, Node, Metastasis; T, Tumor; N, Node; HR Hazard Ratio.

Kaplan-Meier survival curve analysis

Kaplan-Meier survival curves confirmed the prognostic significance of several factors. Significant survival differences (all P < 0.001) were observed based on the following items. Patients ≥ 65 years had notably lower survival than younger groups.

Positive EBV DNA status was associated with poorer survival outcomes. Patients with advanced TNM and T stages exhibited worse prognoses. High expression levels of PDGF-BB and DD were significantly associated with reduced survival. Detailed curves are presented in Figure 6.

Figure 6.

Figure 6

KM survival curves: influence of different clinical characteristics on five-year survival. A: Survival curves grouped by age. B: Survival curves grouped by EBV-DNA. C: Survival curves of patients with different TNM stages. D: Survival curves of patients with different T stages. E: Survival curves of patients with high vs. low PDGF-BB expression. F: Survival curves of patients with high vs. low DD expression. Notes: K-M, Kaplan-Meier; EBV, Epstein-Barr Virus; PDGF-BB, Platelet-Derived Growth Factor-BB; DD, D-dimer.

Multivariate cox regression analysis

Multivariate analysis identified the following independent predictors of five-year survival:

PDGF-BB levels < 628.18 were associated with better survival compared to ≥ 628.18 (HR = 0.492, P = 0.009). DD levels < 746.1 showed significant protective effect compared to ≥ 746.1 (HR = 0.456, P = 0.002). Strong independent effect for age 51-64 years (HR = 2.057, P = 0.032) and ≥ 65 years (HR = 4.138, P < 0.001). EBV DNA negativity was associated with better survival compared to positivity (HR = 0.273, P = 0.012). TNM stage III/IV was associated with increased mortality risk (HR = 3.042, P = 0.023). No sig- nificant effect was found in T stage (P = 0.624; Table 5).

Table 5.

Multivariate Cox regression analysis: prognostic value of coagulation markers and clinical characteristics for five-year survival

Variable Beta Std Err P HR Lower Upper
PDGF-BB
    ≥ 628.18
    < 628.18 -0.710 0.273 0.009 0.492 0.288 0.840
DD
    ≥ 746.1
    < 746.1 -0.785 0.249 0.002 0.456 0.280 0.743
Age
    ≤ 50
    51-64 0.721 0.337 0.032 2.057 1.064 3.979
    ≥ 65 1.420 0.350 < 0.001 4.138 2.084 8.215
EBV DNA
    Positive
    Negative -1.300 0.519 0.012 0.273 0.098 0.754
TNM staging
    I + II
    III + IV 1.113 0.491 0.023 3.042 1.163 7.958
T staging
    T1 + T2
    T3 + T4 0.122 0.250 0.624 1.130 0.692 1.845

Notes: PDGF-BB, Platelet-Derived Growth Factor-BB; DD, D-dimer; EBV, Epstein-Barr Virus; TNM, Tumor, Node, Metastasis; T, Tumor; HR, Hazard Ratio.

Application of the dynamic nomogram in predicting five-year survival

Figures 7 and 8 illustrate the dynamic nomogram developed using the DynNom package in R. This tool enables real-time prediction of five-year survival probabilities based on individual patient characteristics, including age, EBV DNA status, TNM stage, T stage, and PDGF-BB level.

Figure 7.

Figure 7

Patient 5-year prognosis nomogram. Notes: DynNom, Dynamic Nomogram; PDGF-BB, Platelet-Derived Growth Factor-BB; EBV, Epstein-Barr Virus; TNM, Tumor, Node, Metastasis; T, Tumor.

Figure 8.

Figure 8

Five-year survival rate prediction based on the dynamic nomogram. Notes: DynNom, Dynamic Nomogram; PDGF-BB, Platelet-Derived Growth Factor-BB; EBV, Epstein-Barr Virus; TNM, Tumor, Node, Metastasis; T, Tumor.

By inputting specific values for these variables, clinicians can dynamically calculate and visualize personalized survival probabilities. For example, a patient aged ≥ 65 years with EBV DNA negativity, TNM stage III/IV, T3/T4 staging, and high PDGF-BB levels would generate a survival curve reflecting a lower five-year survival probability.

This dynamic nomogram provides an intuitive, interactive platform for individualized survival prediction, enhancing clinical decision-making in the management of NPC.

Discussion

NPC is highly prevalent in Southeast Asia, particularly in Guangdong, Hong Kong, and Macao [19]. Coagulation abnormalities within the tumor microenvironment are closely associated with cancer progression [20]. PDGF-BB, a key regulator of cell proliferation, migration, and angiogenesis, has been strongly implicated in the development of various solid tumors [21]. This study compared PDGF-BB levels and coagulation parameters between NPC patients and healthy controls, aiming to investigate their roles in diagnosis, disease staging, treatment efficacy, and prognosis, thereby providing a foundation for precision diagnosis and individualized treatment strategies in NPC.

Our findings revealed significantly elevated PDGF-BB levels in NPC patients compared to healthy individuals, highlighting its important role in tumor initiation and progression [22]. PDGF-BB secreted by tumor cells promotes proliferation and migration, while its interaction with PDGF receptors on vascular endothelial cells activates the MAPK and PI3K/Akt pathways, facilitating neovascularization. This vascular remodeling supplies oxygen and nutrients to the tumor, enhancing its growth [22]. Additionally, NPC patients exhibited significantly higher levels of coagulation markers-APTT, PT, FIB, PLT, and DD, reflecting a dysregulated coagulation system within the tumor microenvironment [23]. These abnormalities may result from tumor-derived procoagulant factors, local inflammatory responses, and extracellular matrix remodeling, all of which promote microthrombus formation and help tumor cells evade immune surveillance [23].

Previous studies have shown that coagulation markers, with the exception of thrombin time, are associated with NPC stage and metastatic risk [24]. Elevated PDGF-BB levels derived from platelets have also been observed in breast cancer, underscoring its role in tumor angiogenesis and metastasis across multiple solid tumors [25,26]. With the development of advanced detection technologies such as surface-enhanced Raman scattering-lateral flow assay biosensors, PDGF-BB can now be detected in prostate cancer plasma at picogram-per-milliliter levels, supporting its potential as an early diagnostic biomarker for NPC [27].

Further analysis revealed a strong positive correlation between PDGF-BB levels and TNM, T, and N stages, with higher expression observed in patients with advanced-stage NPC. Tumor-derived PDGF-BB facilitates angiogenesis and extracellular matrix remodeling by activating downstream signaling in endothelial and stromal cells, thereby promoting tumor invasiveness and metastatic potential [28,29]. Although APTT, PT, FIB, PLT, and DD showed stage-dependent variation trends, their collective alterations reflect underlying coagulation dysfunction and inflammation during tumor progression. Notably, He et al. [30] reported that elevated preoperative plasma FIB levels were positively associated with TNM stage and metastatic risk, highlighting the utility of multi-marker panels for tumor staging and prognostic evaluation.

To assess treatment response, this study systematically compared biomarker profiles across patient groups stratified by clinical efficacy. Despite thorough statistical analysis, no significant differences were found in PDGF-BB or coagulation marker levels (APTT, PT, FIB, PLT, DD) between patients with treatment response and those with disease progression. Nevertheless, ROC analysis indicated that PDGF-BB retained predictive value in identifying treatment-related improvement. The complexity of clinical efficacy assessment may limit the ability of individual coagulation markers to reflect therapeutic outcomes, which are often influenced by diverse treatment protocols, tumor heterogeneity, and patient-specific factors such as therapy tolerance.

Supporting this view, Yang et al. [31] reported that reductions in PDGF-BB levels after treatment were associated with better clinical outcomes, reinforcing its potential as a response biomarker. Therefore, while no significant group differences were observed in this cohort, PDGF-BB may still play a critical role in monitoring therapeutic efficacy. Future studies incorporating broader clinical variables, diverse treatment regimens, and extended follow-up are warranted to further clarify its role in efficacy evaluation.

Using multivariate Cox regression analysis, this study identified PDGF-BB as an independent risk factor for five-year survival in NPC patients, with each unit increase in PDGF-BB levels associated with a proportional rise in mortality risk. In univariate analysis, DD also demonstrated significant prognostic value. Additionally, age, EBV DNA status, and TNM stage were found to significantly influence survival outcomes, with elderly patients, EBV DNA-positive individuals, and those with advanced-stage disease exhibiting poorer prognoses [32].

Among coagulation markers, DD was notably elevated in NPC patients and has been widely validated as a prognostic biomarker. Chen et al. [33] reported that elevated DD reflects a hypercoagulable state and is significantly associated with worse disease-free survival, distant metastasis-free survival, and overall survival, findings that align with our results. He et al. [34] further demonstrated that combined detection of DD and albumin enhances prognostic accuracy in NPC. DD levels are closely related to tumor invasiveness and metastatic potential, reinforcing their utility as a prognostic marker. Moreover, when integrated with traditional clinical factors such as TNM stage and age, survival prediction accuracy is further improved.

PDGF-BB also plays a central role in both coagulation and tumor biology. As highlighted by Liang et al. [35], PDGF-BB facilitates tumor proliferation, migration, and angiogenesis, and is strongly associated with aggressive behavior and metastatic capacity. Our study further supports this role in the NPC microenvironment.

In our analysis, age, EBV DNA status, and TNM stage emerged as strong independent prognostic indicators. Specifically, older patients, EBV DNA-positive cases, and advanced-stage tumors were consistently linked to reduced survival. EBV DNA is an established biomarker in NPC, and the study by Mazurek et al. [36] emphasized its dual value in diagnosis and prognosis. Elevated EBV DNA levels are linked to poor local control and a markedly increased risk of distant metastasis.

TNM staging remains the cornerstone of prognostic evaluation in clinical practice. However, integrating TNM stage with emerging biomarkers such as PDGF-BB and DD can significantly enhance survival prediction accuracy. Kaplan-Meier survival curves in our study demonstrated that high PDGF-BB and DD levels were strongly correlated with poorer five-year survival, validating their prognostic utility.

Interestingly, studies in other cancers also support PDGF-BB’s prognostic value. For example, in esophageal cancer, post-radiotherapy reductions in PDGF-BB levels were significantly associated with longer survival durations [37]. This finding supports the potential application of PDGF-BB in monitoring treatment efficacy and survival outcomes in NPC as well.

Based on multivariate Cox regression results, we developed a dynamic nomogram incorporating PDGF-BB, age, EBV DNA status, TNM stage, and T stage to predict individualized five-year survival. This model offers a user-friendly, interactive platform for real-time survival prediction and risk assessment, enabling clinicians to tailor treatment strategies according to patient-specific clinical profiles. With further validation in multi-center cohorts, this model holds promise for broader application in NPC and other malignancies, contributing to the advancement of precision oncology.

This study has several limitations. First, its single-center, retrospective design and limited sample size may restrict the generalizability of findings. Second, variability in detection technologies and sample processing may have reduced the sensitivity of certain coagulation parameters in predicting treatment response, limiting the utility of single-marker analysis.

Future studies should aim to conduct multi-center, prospective trials with larger cohorts to validate the diagnostic and prognostic value of PDGF-BB and coagulation biomarkers in NPC. Investigating combined biomarker panels, dynamic changes during treatment, and the molecular mechanisms underlying PDGF-BB and coagulation dysregulation will improve predictive models and guide therapeutic interventions. Integration of clinical, molecular, and imaging data will be essential for enhancing early detection, personalized treatment, and overall clinical outcomes in NPC patients.

In conclusion, PDGF-BB and key coagulation markers, particularly DD, represent promising biomarkers for prognostic assessment in nasopharyngeal carcinoma. When combined with clinical factors such as age, EBV DNA status, and TNM staging, these biomarkers can significantly enhance the accuracy of personalized survival predictions, offering valuable guidance for clinical decision-making in NPC patients receiving IMRT.

Disclosure of conflict of interest

None.

Supporting Information

ajcr0015-2451-f9.pdf (234.2KB, pdf)

References

  • 1.Chang ET, Ye W, Zeng YX, Adami HO. The evolving epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 2021;30:1035–1047. doi: 10.1158/1055-9965.EPI-20-1702. [DOI] [PubMed] [Google Scholar]
  • 2.Tang LL, Chen YP, Chen CB, Chen MY, Chen NY, Chen XZ, Du XJ, Fang WF, Feng M, Gao J, Han F, He X, Hu CS, Hu DS, Hu GY, Jiang H, Jiang W, Jin F, Lang JY, Li JG, Lin SJ, Liu X, Liu QF, Ma L, Mai HQ, Qin JY, Shen LF, Sun Y, Wang PG, Wang RS, Wang RZ, Wang XS, Wang Y, Wu H, Xia YF, Xiao SW, Yang KY, Yi JL, Zhu XD, Ma J. The Chinese Society of Clinical Oncology (CSCO) clinical guidelines for the diagnosis and treatment of nasopharyngeal carcinoma. Cancer Commun (Lond) 2021;41:1195–1227. doi: 10.1002/cac2.12218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Qiao H, Tan XR, Li H, Li JY, Chen XZ, Li YQ, Li WF, Tang LL, Zhou GQ, Zhang Y, Liang YL, He QM, Zhao Y, Huang SY, Gong S, Li Q, Ye ML, Chen KL, Sun Y, Ma J, Liu N. Association of intratumoral microbiota with prognosis in patients with nasopharyngeal carcinoma from 2 hospitals in China. JAMA Oncol. 2022;8:1301–1309. doi: 10.1001/jamaoncol.2022.2810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ng WT, Soong YL, Ahn YC, AlHussain H, Choi HCW, Corry J, Grégoire V, Harrington KJ, Hu CS, Jensen K, Kwong DL, Langendijk JA, Le QT, Lee NY, Lin JC, Lu TX, Mendenhall WM, O’Sullivan B, Ozyar E, Pan JJ, Peters LJ, Poh SS, Rosenthal DI, Sanguineti G, Tao Y, Wee JT, Yom SS, Chua MLK, Lee AWM. International recommendations on reirradiation by intensity modulated radiation therapy for locally recurrent nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2021;110:682–695. doi: 10.1016/j.ijrobp.2021.01.041. [DOI] [PubMed] [Google Scholar]
  • 5.Liu X, Zhang Y, Yang KY, Zhang N, Jin F, Zou GR, Zhu XD, Xie FY, Liang XY, Li WF, He ZY, Chen NY, Hu WH, Wu HJ, Shi M, Zhou GQ, Mao YP, Guo R, Sun R, Huang J, Liang SQ, Wu WL, Su Z, Li L, Ai P, He YX, Zang J, Chen L, Lin L, Huang SH, Xu C, Lv JW, Li YQ, Hong SB, Jie YS, Li H, Huang SW, Liang YL, Wang YQ, Peng YL, Zhu JH, Zang SB, Liu SR, Lin QG, Li HJ, Tian L, Liu LZ, Zhao HY, Lin AH, Li JB, Liu N, Tang LL, Chen YP, Sun Y, Ma J. Induction-concurrent chemoradiotherapy with or without sintilimab in patients with locoregionally advanced nasopharyngeal carcinoma in China (CONTINUUM): a multicentre, open-label, parallel-group, randomised, controlled, phase 3 trial. Lancet. 2024;403:2720–2731. doi: 10.1016/S0140-6736(24)00594-4. [DOI] [PubMed] [Google Scholar]
  • 6.Yuan Y, Ye F, Wu JH, Fu XY, Huang ZX, Zhang T. Early screening of nasopharyngeal carcinoma. Head Neck. 2023;45:2700–2709. doi: 10.1002/hed.27466. [DOI] [PubMed] [Google Scholar]
  • 7.Gong R, Yang C, Abbas G, Wang D, Zhang X, Nie G, Ding H. Diagnosis of nasopharyngeal carcinoma using an ultrasensitive immunoassay method based on nanoparticles. Nanoscale. 2023;15:3475–3481. doi: 10.1039/d2nr05848k. [DOI] [PubMed] [Google Scholar]
  • 8.Tang J, Chen H, Chen F. CT and MRI presentation of nasopharyngeal tuberculosis (with a case report) Radiol Case Rep. 2024;19:1702–1707. doi: 10.1016/j.radcr.2024.01.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hsu CL, Chang YS, Li HP. Molecular diagnosis of nasopharyngeal carcinoma: past and future. Biomed J. 2025;48:100748. doi: 10.1016/j.bj.2024.100748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pan JJ, Mai HQ, Ng WT, Hu CS, Li JG, Chen XZ, Chow JCH, Wong E, Lee V, Ma LY, Guo QJ, Liu Q, Liu LZ, Xu TT, Gong XC, Qiang MY, Au KH, Liu TC, Chiang CL, Xiao YP, Lin SJ, Chen YB, Guo SS, Wong CHL, Tang LQ, Xu ZY, Jia YZ, Peng WS, Hu LP, Lu TZ, Jiang F, Cao CN, Xu W, Ma J, Blanchard P, Williams M, Glastonbury CM, King AD, Patel SG, Seethala RR, Colevas AD, Fan DM, Chua MLK, Huang SH, O’Sullivan B, Lydiatt W, Lee AWM. Ninth version of the AJCC and UICC nasopharyngeal cancer TNM staging classification. JAMA Oncol. 2024;10:1627–1635. doi: 10.1001/jamaoncol.2024.4354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ding C, Dai DY, Luo ZK, Wang GY, Dong Z, Qin GJ, Du XJ, Ma J. Evaluation of a novel model incorporating serological indicators into the conventional TNM staging system for nasopharyngeal carcinoma. Oral Oncol. 2024;151:106725. doi: 10.1016/j.oraloncology.2024.106725. [DOI] [PubMed] [Google Scholar]
  • 12.Adelborg K, Larsen JB, Hvas AM. Disseminated intravascular coagulation: epidemiology, biomarkers, and management. Br J Haematol. 2021;192:803–818. doi: 10.1111/bjh.17172. [DOI] [PubMed] [Google Scholar]
  • 13.Jing H, Wu X, Xiang M, Wang C, Novakovic VA, Shi J. Microparticle phosphatidylserine mediates coagulation: involvement in tumor progression and metastasis. Cancers (Basel) 2023;15:1957. doi: 10.3390/cancers15071957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.You D, Wang Y, Xu J, Yang R, Wang W, Wang X, Cao X, Li Y, Yu L, Wang W, Shi Y, Zhang C, Yang H, He Y, Bian L. MiR-3529-3p from PDGF-BB-induced cancer-associated fibroblast-derived exosomes promotes the malignancy of oral squamous cell carcinoma. Discov Oncol. 2023;14:166. doi: 10.1007/s12672-023-00753-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kozłowski M, Borzyszkowska D, Mirko J, Turoń-Skrzypińska A, Piotrowska K, Tołoczko-Grabarek A, Kwiatkowski S, Tarnowski M, Rotter I, Cymbaluk-Płoska A. Preoperative serum levels of PDGF-AB, PDGF-BB, TGF-α, EGF and ANG-2 in the diagnosis of endometrial cancer. Cancers (Basel) 2023;15:4815. doi: 10.3390/cancers15194815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Inanc M, Sirakaya HA, Karaman H, Bozkurt O. The prognostic importance of VEGF-A, PDGF-BB and c-MET in patients with metastatic colorectal cancer. J Oncol Pharm Pract. 2020;26:1878–1885. doi: 10.1177/1078155220904151. [DOI] [PubMed] [Google Scholar]
  • 17.Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229–263. doi: 10.3322/caac.21834. [DOI] [PubMed] [Google Scholar]
  • 18.Bossi P, Chan AT, Licitra L, Trama A, Orlandi E, Hui EP, Halámková J, Mattheis S, Baujat B, Hardillo J, Smeele L, van Herpen C, Castro A, Machiels JP ESMO Guidelines Committee. Nasopharyngeal carcinoma: ESMO-EURACAN clinical practice guidelines for diagnosis, treatment and follow-up(†) Ann Oncol. 2021;32:452–465. doi: 10.1016/j.annonc.2020.12.007. [DOI] [PubMed] [Google Scholar]
  • 19.Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet. 2019;394:64–80. doi: 10.1016/S0140-6736(19)30956-0. [DOI] [PubMed] [Google Scholar]
  • 20.Peng PH, Wu CC, Liu SC, Chang KP, Chen CD, Chang YT, Hsu CW, Chang YS, Yu JS. Quantitative plasma proteome analysis reveals aberrant level of blood coagulation-related proteins in nasopharyngeal carcinoma. J Proteomics. 2011;74:744–757. doi: 10.1016/j.jprot.2011.02.023. [DOI] [PubMed] [Google Scholar]
  • 21.Ren X, Li L, Wu J, Lin K, He Y, Bian L. PDGF-BB regulates the transformation of fibroblasts into cancer-associated fibroblasts via the lncRNA LURAP1L-AS1/LURAP1L/IKK/IκB/NF-κB signaling pathway. Oncol Lett. 2021;22:537. doi: 10.3892/ol.2021.12798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Xiu-Ying H, Yue-Xiang Z, Hui-Si Y, Hong-Zhou Y, Qing-Jie X, Ting-Hua W. PDGFBB facilitates tumorigenesis and malignancy of lung adenocarcinoma associated with PI3K-AKT/MAPK signaling. Sci Rep. 2024;14:4191. doi: 10.1038/s41598-024-54801-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wang D, Cui SP, Chen Q, Ren ZY, Lyu SC, Zhao X, Lang R. The coagulation-related genes for prognosis and tumor microenvironment in pancreatic ductal adenocarcinoma. BMC Cancer. 2023;23:601. doi: 10.1186/s12885-023-11032-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yin J, Zhu SS. Routine coagulation molecules predict nasopharyngeal carcinoma and associated metastases. Br J Biomed Sci. 2019;76:178–183. doi: 10.1080/09674845.2019.1631943. [DOI] [PubMed] [Google Scholar]
  • 25.Han H, Cao FL, Wang BZ, Mu XR, Li GY, Wang XW. Expression of angiogenesis regulatory proteins and epithelial-mesenchymal transition factors in platelets of the breast cancer patients. ScientificWorldJournal. 2014;2014:878209. doi: 10.1155/2014/878209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Barron GA, Goua M, Wahle KWJ, Bermano G. Circulating levels of angiogenesis-related growth factors in breast cancer: a study to profile proteins responsible for tubule formation. Oncol Rep. 2017;38:1886–1894. doi: 10.3892/or.2017.5803. [DOI] [PubMed] [Google Scholar]
  • 27.Cao X, Song Q, Sun Y, Mao Y, Lu W, Li L. A SERS-LFA biosensor combined with aptamer recognition for simultaneous detection of thrombin and PDGF-BB in prostate cancer plasma. Nanotechnology. 2021;32 doi: 10.1088/1361-6528/ac1754. [DOI] [PubMed] [Google Scholar]
  • 28.Fang J, Huang X, Han X, Zheng Z, Hu C, Chen T, Yang X, Ouyang X, Chen Z, Wei H. Endothelial progenitor cells promote viability and nerve regenerative ability of mesenchymal stem cells through PDGF-BB/PDGFR-β signaling. Aging (Albany NY) 2020;12:106–121. doi: 10.18632/aging.102604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yang Y, Andersson P, Hosaka K, Zhang Y, Cao R, Iwamoto H, Yang X, Nakamura M, Wang J, Zhuang R, Morikawa H, Xue Y, Braun H, Beyaert R, Samani N, Nakae S, Hams E, Dissing S, Fallon PG, Langer R, Cao Y. The PDGF-BB-SOX7 axis-modulated IL-33 in pericytes and stromal cells promotes metastasis through tumour-associated macrophages. Nat Commun. 2016;7:11385. doi: 10.1038/ncomms11385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.He SS, Wang Y, Yang L, Chen HY, Liang SB, Lu LX, Chen Y. Plasma fibrinogen correlates with metastasis and is associated with prognosis in human nasopharyngeal carcinoma. J Cancer. 2017;8:403–409. doi: 10.7150/jca.17028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yang Q, Nie YH, Cai MB, Li ZM, Zhu HB, Tan YR. Gemcitabine combined with cisplatin has a better effect in the treatment of recurrent/metastatic advanced nasopharyngeal carcinoma. Drug Des Devel Ther. 2022;16:1191–1198. doi: 10.2147/DDDT.S353898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wu YX, Tian BY, Ou XY, Wu M, Huang Q, Han RK, He X, Chen SL. A novel model for predicting prognosis and response to immunotherapy in nasopharyngeal carcinoma patients. Cancer Immunol Immunother. 2024;73:14. doi: 10.1007/s00262-023-03626-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chen WH, Tang LQ, Wang FW, Li CP, Tian XP, Huang XX, Mai SJ, Liao YJ, Deng HX, Chen QY, Liu H, Zhang L, Guo SS, Liu LT, Yan SM, Li CF, Zhang JP, Liu Q, Liu XW, Liu LZ, Mai HQ, Zeng MS, Xie D. Elevated levels of plasma D-dimer predict a worse outcome in patients with nasopharyngeal carcinoma. BMC Cancer. 2014;14:583. doi: 10.1186/1471-2407-14-583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.He SS, Wang Y, Wang CT, Zhu MY, Yang XL, Chen DM, Chen Y. A combined marker based on plasma D-dimer and serum albumin levels in patients with nasopharyngeal carcinoma is associated with poor survival outcomes in a retrospective cohort study. J Cancer. 2019;10:3691–3697. doi: 10.7150/jca.32387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liang T, Liu W, Xie J, Wang Y, Chen G, Liao W, Song L, Zhang X. Serum EA-IgA and D-dimer, but not VCA-IgA, are associated with prognosis in patients with nasopharyngeal carcinoma: a meta-analysis. Cancer Cell Int. 2021;21:329. doi: 10.1186/s12935-021-02035-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mazurek AM, Wygoda A, Rutkowski T, Olbryt M, Pietrowska M, Celejewska A, Składowski K, Widłak P. Prognostic significance of Epstein-Barr virus viral load in patients with T1-T2 nasopharyngeal cancer. J Med Virol. 2020;92:348–355. doi: 10.1002/jmv.25606. [DOI] [PubMed] [Google Scholar]
  • 37.Er P, Qian D, Zhang W, Zhang B, Wei H, Zhang T, Chen X, Wang Y, Zhao J, Wang Q, Pang Q, Wang P. The expression of PDGF-BB predicts curative effect in locally advanced esophageal squamous cell carcinoma treated by radiotherapy. Aging (Albany NY) 2020;12:6586–6599. doi: 10.18632/aging.102993. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ajcr0015-2451-f9.pdf (234.2KB, pdf)

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES