Skip to main content

Main menu

  • Home
  • About
    • About CBM
    • Editorial Board
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Other Publications
    • cbm

User menu

  • My alerts

Search

  • Advanced search
Cancer Biology & Medicine
  • Other Publications
    • cbm
  • My alerts
Cancer Biology & Medicine

Advanced Search

 

  • Home
  • About
    • About CBM
    • Editorial Board
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Follow cbm on Twitter
  • Visit cbm on Facebook
Research ArticleOriginal Article

Prognostic significance of a combined and controlled nutritional status score and EBV-DNA in patients with advanced nasopharyngeal carcinoma: a long-term follow-up study

Hui Lu, Shanshan Guo, Liting Liu, Qiuyan Chen, Yujing Liang, Sailan Liu, Xuesong Sun, Qingnan Tang, Xiaoyun Li, Ling Guo, Haoyuan Mo, Linquan Tang and Haiqiang Mai
Cancer Biology & Medicine April 2022, 19 (4) 551-564; DOI: https://doi.org/10.20892/j.issn.2095-3941.2020.0627
Hui Lu
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
2Department of Radiation Oncology, Guangzhou Concord Cancer Center, Guangzhou 510045, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shanshan Guo
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Liting Liu
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Qiuyan Chen
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yujing Liang
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sailan Liu
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xuesong Sun
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Qingnan Tang
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xiaoyun Li
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ling Guo
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Haoyuan Mo
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Linquan Tang
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Linquan Tang
  • For correspondence: [email protected] [email protected]
Haiqiang Mai
1State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
3Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Haiqiang Mai
  • For correspondence: [email protected] [email protected]
  • Article
  • Figures & Data
  • References
  • Info & Metrics
  • PDF
Loading

Abstract

Objective: Several studies have reported that the controlling nutritional status (CONUT) score is a prognostic predictor for survival among patients with different types of cancer. We assessed the prognostic value of changes in the CONUT score during treatment and the ΔCONUT-EBV DNA score in patients with advanced nasopharyngeal carcinoma (NPC).

Methods: We retrospectively analyzed 433 patients with advanced NPC having no evidence of metastasis from January 2007 to June 2011; the patients underwent radical concurrent chemoradiotherapy (CCRT) at Sun Yat-sen University Cancer Center and were grouped based on their ΔCONUT and ΔCONUT-EBV DNA scores. Kaplan-Meier curves were used to compare the patient outcomes according to the cut-off ΔCONUT score and the ΔCONUT-EBV DNA scoring system.

Results: Among all patients, overall survival (OS) was independently predicted by a high ΔCONUT score (P = 0.031) and high EBV DNA (P < 0.001). The ΔCONUT-EBV DNA score [OS area under the curve (AUC) = 0.621; progression free survival (PFS)-AUC = 0.612; distant metastasis-free survival (DMFS)-AUC = 0.622] was more predictive of OS, PFS, and DMFS in patients with advanced NPC than the ΔCONUT score (OS-AUC = 0.547; PFS-AUC = 0.533; DMFS-AUC = 0.522) and pretreatment plasma EBV DNA levels alone (OS-AUC = 0.600; PFS-AUC = 0.591, DMFS-AUC = 0.610). The ΔCONUT-EBV DNA score was significantly correlated with OS, PFS, and DMFS in patients with advanced NPC treated with CCRT.

Conclusions: The ΔCONUT-EBV DNA score may be useful in clinical practice as a convenient biomarker for predicting the outcomes in patients with advanced NPC treated with CCRT.

keywords

  • Controlling nutritional status score
  • Epstein-Barr virus deoxyribonucleic acid
  • nasopharyngeal carcinoma
  • prognosis
  • predictive factor

Introduction

Nasopharyngeal carcinoma (NPC) is one of the most prevalent malignancies in Southeast Asia, including southern China1. Epstein-Barr virus deoxyribonucleic acid (EBV DNA) has been used as a plasma marker for population screening2, prognoses3, and disease recurrence surveillance4,5. Owing to the high sensitivity of irradiation, radiotherapy alone and concurrent chemoradiotherapy (CCRT) are the primary treatments for nonmetastatic NPC. Moreover, intensity-modulated radiotherapy (IMRT) is the currently preferred method, because it reduces the risk of short-term side effects and long-term sequelae6. Over the years, immune-nutritional scores, such as the body mass index (BMI)7, albumin-globulin ratio8, and prognostic nutritional index (PNI)9,10, have been reported to be prognostic markers that influence NPC treatment outcomes.

Recently, studies have shown that the controlling nutritional status (CONUT) score, a novel immunological and nutritional score, is a prognostic predictor for survival prognosis in many cancer types11–16. This score is calculated from the total lymphocyte count, total cholesterol, and serum albumin levels17. However, data on the prognostic value of the CONUT score for patients with advanced NPC are still unavailable. Moreover, the point at which the nutritional score should be measured during anti-cancer treatment remains unclear. Thus, this study aimed to clarify the prognostic and predictive values of the CONUT score at different time points to predict survival among patients with advanced NPC.

However, nutritional scores, such as the CONUT score, PNI, and BMI, are not always reliable in predicting the outcomes of cancers, because they lack tumor-related factors. Previous reports have suggested that tumor-related factors, such as the EBV DNA level3 and tumor node metastasis (TNM) stage18,19, are likely the most reliable prognostic predictors for NPC. Plasma marker EBV DNA has been used for screening2, prognoses3, and disease recurrence surveillance4,5. Lin et al.20 reported an inferior overall survival (OS) in patients with higher baseline plasma EBV DNA levels. The addition of pretreatment plasma EBV DNA to the 8th edition of the AJCC/UICC TNM stage classification greatly improved its prognostic performance21–23. Many studies support a positive relationship between the plasma EBV DNA level and tumor burden24,25. Unfortunately, cut-off values of plasma EBV DNA levels varied across these studies, and an optimal cut-off value of this liquid biopsy marker is still being determined26,27. Here, we characterized the complementary role of the ΔCONUT score in combination with pretreatment plasma EBV DNA. The ΔCONUT-EBV DNA score, which is based on the ΔCONUT score and pretreatment plasma EBV DNA, was integrated with nutritional score and tumor-related factors, so it may have more prognostic information compared to only 1 parameter.

Materials and methods

Patients and study design

We retrospectively included and analyzed 433 patients with advanced NPC, but with no evidence of metastasis (M0) from January 2007 to June 2011, who underwent radical CCRT at Sun Yat-sen University Cancer Center. The exclusion and inclusion criteria were as follows: (I) patients with newly-diagnosed and pathologically proven NPC; (II) patients with complete clinical information and follow-up data; (III) patients with complete laboratory data; (IV) patients who underwent radical CCRT during the course of anti-cancer treatment; (V) patients without other malignant cancers, serious illnesses (severe acute or chronic diseases); (VI) patients without distant metastasis; and (VII) patients with confirmed locoregionally advanced (stages II–IVA) NPC, as defined by the 2010 Union for International Cancer Control (UICC) system (Supplementary Figure S1). Laboratory data and clinical and epidemiological characteristics of the patients, such as age, gender, date of diagnosis, UICC T stage, UICC N stage, clinical stage, total lymphocyte count (/mm3), total cholesterol (mg/dL) and serum albumin (g/dL) levels, were assessed from their medical records. All patients were followed-up until April 2020 or until death. Our study protocol was approved by the ethics committee of Sun Yat-sen University Cancer Center (Approval No. GZR2014-069).

Treatment strategies

Institutional guidelines recommend CCRT ± neoadjuvant/adjuvant chemotherapy (CCRT ± NC/AC) for patients with stage II–IVA NPC. In the present study, 397 (91.7%) of the patients received CCRT only, 24 (5.5%) received CCRT + AC, and 12 (2.8%) received CCRT + NC. All patients received IMRT. The prescribed doses were 66–70 Gy to the primary tumor and 54–64 Gy to the involved cervical lymph nodes and low risk clinical target volume.

The CONUT score and cut-off value

The CONUT score was calculated based on the total cholesterol level, total lymphocyte counts, and serum albumin level (Supplementary Table S1)17. Pretreatment laboratory data (pre-CONUT score) were obtained within 14 days before anti-cancer treatment. Post-CONUT scores were calculated based on the results of blood tests within 5 days before the completion of CCRT. Individual difference value (ΔCONUT score) of the post-treatment to pretreatment CONUT score was calculated as follows: (post - CONUT score) - (pre-CONUT score). The area under the curve (AUC) estimation method was used to determine the predictive value of the pretreatment CONUT score, the post-treatment CONUT score, and the ΔCONUT score for OS. The receiver operating characteristic (ROC) curve analysis method was used to determine the optimal ΔCONUT score cut-off value that was significantly correlated with OS.

Definition of the ΔCONUT-EBV DNA score

The cut-off value of the ΔCONUT score was 3, which was used as the criterion to divide the included 433 patients into ΔCONUT-low (score ≤ 3) and ΔCONUT-high (score > 3) groups. The median cut-off value of EBV DNA was considered in all patients. The patients were divided into the following 2 groups according to the cut-off value of pretreatment plasma EBV DNA level (2,110 copies/mL): EBV DNA-low (score ≤ 2,110 copies/mL) and EBV DNA-high (score > 2,110 copies/mL) groups. Based on the cut-off values of the ΔCONUT score and pretreatment plasma EBV DNA, the ΔCONUT-EBV DNA score was defined. Patients with an EBV DNA score of ≤2,110 copies/mL [N = 217; low-risk group (LRG)] were assigned a score of 1; those with both a ΔCONUT score of ≤ 3 and an EBV DNA score of > 2,110 copies/mL [N = 170; middle-risk group (MRG)] were assigned a score of 2; and those with both a ΔCONUT score of > 3 and an EBV DNA score of > 2,110 copies/mL [N = 46; high risk group (HRG)] were assigned a score of 3 (Table 3).

Follow-up

All the patients were routinely followed up every 3–4 months throughout the first 2 years, every 6 months for the next 2 years, and annually thereafter, and were monitored either until April 2020 or until death. OS was defined as the period from the initial diagnosis to the patient’s death or last follow-up, regardless of whether it was or was not related to NPC. The median follow-up period was 9.52 (range, 8.88–10.74) years. The following end points (time to the first defining event) were assessed: OS, progression-free survival (PFS), and DMFS.

Statistical analysis

Pathological and clinical characteristics, and laboratory data were compared between the 2 groups using Fisher’s exact test. Survival curves were estimated using the Kaplan-Meier method with the log-rank test. Univariate and multivariate analyses were performed using the Cox proportional hazards model. In all patients, the median cut-off values were considered for age and EBV DNA. Age (cut-off: 46 years), gender (female/male), UICC T stage (T1–2, T3–4), UICC N stage (N0–1, N2–3), clinical stage (stage II, stage III–IVA), EBV DNA score (cut-off: 2,110 copies/mL), and ΔCONUT score (cut-off, 3) were the parameters considered in the Cox proportional hazards model. To further investigate the characteristics of patients between the different ΔCONUT-EBV DNA score groups, we performed further experiments. Fisher’s exact test with a two-sided significance level was used to compare the pathological and clinical characteristics, and laboratory data between the different ΔCONUT-EBV DNA score groups; all characteristics are shown in Table 1 before the Bonferroni-Holm correction was included. These analyses were also corrected for testing using the Bonferroni-Holm method. All data were analyzed using SPSS statistical software for Windows, version 22.0 (SPSS, Chicago, IL, USA). All P values were two-sided. P < 0.05 and Bonferroni-corrected P < 0.05 were considered statistically significant.

View this table:
  • View inline
  • View popup
Table 1

Baseline characteristics of 433 patients with locoregionally advanced nasopharyngeal carcinoma

Results

Clinical and pathological characteristics, and treatment outcomes

A total of 397 (91.7%) patients received CCRT only, 24 (5.5%) received CCRT + AC, and 12 (2.8%) received CCRT + NC. All patients in this cohort received IMRT. Of the 433 patients, 39 (9.0%) developed locoregional recurrence, 75 (17.3%) developed distant metastases, and 98 (22.6%) died. The 5-year OS, PFS, and DMFS were 86.4%, 77.8%, and 84.2%, respectively.

We used the continuous variable pre-CONUT, post-CONUT, and ΔCONUT scores as the test variables, and OS as the state variable. The ΔCONUT score AUC was 0.547 [95% confidence interval (CI): 0.479–0.615], which was higher than the AUC of the other CONUT scores [pre-CONUT score (AUC = 0.453; 95% CI: 0.387–0.520) and post-CONUT score (AUC = 0.526; 95% CI: 0.458–0.594)] (Figure 1A). According to the ROC curve analysis results, the optimal ΔCONUT score cut-off value that significantly correlated with OS was 3 (AUC = 0.547; Figure 1A).

Figure 1
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1

(A) Receiver operating characteristic (ROC) curve analysis of the pretreatment controlling nutritional status (CONUT), post-treatment CONUT, and ΔCONUT scores of interest for predicting overall survival (OS). (B) OS based on the ΔCONUT score.

Based on the ΔCONUT score cut-off determined using the ROC curve analysis, the 433 patients were subdivided into the ΔCONUT-low (score ≤ 3; N = 345) and ΔCONUT-high (score > 3; N = 88) groups. The clinical and demographic characteristics of the patients in the 2 groups are shown in Table 1. Age, gender, cancer stage, histology, smoking status, EBV DNA levels, viral capsid antigen (VCA)/immunoglobulin a (IgA) titers, and early antigen (EA)/IgA titer distributions did not significantly differ between the 2 groups (Table 1).

Figure 1B shows the Kaplan-Meier curves for OS according to the ΔCONUT score-based groups. The patients in the ΔCONUT-high group were more likely to experience a shorter OS than those in the ΔCONUT-low group (Figure 1B; P = 0.030). The 5-year OS rates for patients with low and high ΔCONUT scores were 88.7% and 77.3%, respectively.

Prognostic value of the ΔCONUT score

Univariate analyses identified an age > 46 years (P = 0.011), male (P = 0.008), advanced N stage (P = 0.003), high ΔCONUT score (P = 0.031), and high EBV DNA level (P < 0.001) as factors significantly associated with a worse OS (Table 2). Multivariable analyses showed that age [hazard ratio (HR): 1.580; 95.0% CI: 1.054–2.369; P = 0.027], gender (HR: 1.937; 95.0% CI: 1.115–3.367; P = 0.019), N stage (HR: 1.597; 95.0% CI: 1.060–2.406; P = 0.025), EBV DNA level (HR: 1.753; 95.0% CI: 1.142–2.693; P = 0.010), and ΔCONUT score (HR: 1.574; 95.0% CI: 1.009–2.454; P = 0.045) were independent prognostic factors for OS (Table 2).

View this table:
  • View inline
  • View popup
Table 2

Univariate and multivariate analysis for OS

Prognostic values of the ΔCONUT-EBV DNA score

In addition, patients were stratified according to their pretreatment plasma EBV DNA levels. The results showed that the ΔCONUT score was not associated with OS in patients with low EBV DNA levels (≤2,110 copies/mL; Figure 2A). Notably, the high ΔCONUT score group had a significantly worse OS than the low ΔCONUT score group, but only for patients with high pretreatment plasma EBV DNA levels (>2,110 copies/mL; P = 0.014; Figure 2B).

Figure 2
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2

Survival curves of patients in different Epstein-Barr virus deoxyribonucleic acid (EBV DNA) groups. (A) Overall survival (OS) based on the ΔCONUT score in 217 patients with advanced nasopharyngeal carcinoma (NPC) with EBV DNA ≤ 2,110 copies/mL. (B) OS based on the ΔCONUT score in 216 patients with advanced NPC with EBV DNA > 2,110 copies/mL.

We then suspected that the pretreatment plasma EBV DNA level could help improve the prognostic value of ΔCONUT in patients with advanced NPC. The ΔCONUT-EBV DNA score, a novel prognostic marker, was calculated based on the ΔCONUT score and the pretreatment plasma EBV DNA level (Table 3). Using this ΔCONUT-EBV DNA scoring system, we divided patients into low-risk (LRG; N = 217), middle-risk (MGR; N = 170), and high-risk (HRG; N = 46) groups (Table 3). The baseline characteristics of the different groups are shown in Table 4. Patients with a low ΔCONUT-EBV DNA score had earlier UICC N stages (Bonferroni-Holm corrected P = 0.003), earlier clinical stages (Bonferroni-Holm corrected P = 0.006), and lower EA/IgA values (Bonferroni-Holm corrected; P = 0.036) than patients with a medium ΔCONUT-EBV DNA score (MRG). Significant differences were present between LRG and HRG in the UICC N stage (Bonferroni-Holm corrected; P < 0.001), but other variables were not significantly different. There was no significant difference in the age, gender, stage, histology, smoking status, VCA/IgA titers, and EA/IgA titers between the HRG and MRG (Table 4).

View this table:
  • View inline
  • View popup
Table 3

ΔCONUT-EBV DNA score based on risk stratification with CONUT scores and EBV DNA for advanced nasopharyngeal carcinoma patients

View this table:
  • View inline
  • View popup
Table 4

Characteristics of NPC patients in 3 different risk (ΔCONUT-EBV DNA score) groups

ROC analysis was used to evaluate the effect of the ΔCONUT score, pretreatment plasma EBV DNA level, and ΔCONUT-EBV DNA score on the prognosis. The results showed that the ΔCONUT-EBV DNA scores (OS: AUC = 0.621, 95% CI: 0.556–0.685; PFS: AUC = 0.612, 95% CI: 0.552–0.672; DMFS: AUC = 0.622, 95% CI: 0.552–0.672; Figure 3A–3C) were more predictive of OS, PFS, and DMFS in patients with advanced NPC than the ΔCONUT scores (OS: AUC = 0.547, 95% CI: 0.480–0.613; PFS: AUC = 0.533, 95% CI: 0.472–0.594; DMFS: AUC = 0.522, 95% CI: 0.449–0.595; Figure 3A–3C) or pretreatment plasma EBV DNA levels alone (OS: AUC = 0.600, 95% CI: 0.537–0.663; PFS: AUC = 0.591, 95% CI: 0.532–0.650; DMFS: AUC = 0.610, 95% CI: 0.541–0.678; Figure 3A–3C).

Figure 3
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3

Receiver operating characteristic (ROC) curve analysis of the ΔCONUT score, the Epstein-Barr virus deoxyribonucleic acid (EBV DNA) level, and the ΔCONUT-EBV DNA score of interest for predicting 5-year overall survival (OS) (A), progression-free survival (PFS) (B), and distant metastasis-free survival (DMFS) (C); Survival analysis for 5-year OS (D), PFS (E), and DMFS (F) based on the ΔCONUT-EBV DNA score.

There were significant differences among these 3 groups in terms of the 5-year OS (92.6%, 84.1%, and 65.2%; P < 0.001) (Figure 3D), PFS (83.9%, 76.4%, and 54.3%; P < 0.001) (Figure 3E), and DMFS (89.8%, 81.1%, and 68.9%; P < 0.001) (Figure 3F). Notably, HRG and MRG had a significantly worse OS (HRG vs. LGR, P < 0.001; MRG vs. LGR, P = 0.009) (Figure 3D), PFS (HRG vs. LGR, P < 0.001; MRG vs. LGR, P = 0.013) (Figure 3E), and DMFS (HRG vs. LGR, P < 0.001; MRG vs. LGR, P = 0.003) (Figure 3F) than the LRG. Kaplan-Meier analysis and the log-rank test also identified a significantly poorer OS (HRG vs. MGR, P = 0.014) (Figure 3D) and PFS (HRG vs. MGR, P = 0.006) (Figure 3E) among patients with a high ΔCONUT-EBV DNA score than among those with a medium ΔCONUT-EBV DNA score.

The results of our multivariate Cox analysis showed that the ΔCONUT-EBV DNA score was an independent prognostic factor for OS (HR: 1.652; 95% CI: 1.242–2.198; P = 0.001), PFS (HR: 1.772; 95% CI: 1.385–2.267; P < 0.001), and DMFS (HR: 1.874, 95% CI: 1.371–2.562; P < 0.001) in patients with advanced NPC who were treated with CCRT (Table 5).

View this table:
  • View inline
  • View popup
Table 5

Univariate and multivariate analysis for 5-year OS, PFS, and DMFS combined with the novel prognosis predictor ΔCONUT score and EBV DNA

Discussion

Studies have recently shown that the CONUT score is a prognostic predictor for survival prognosis in many cancer types11–16. Moreover, the point at which the nutritional score should be measured during anti-cancer treatment remains unclear. In the present study, the prognostic values of the pre-CONUT, post-CONUT, and ΔCONUT scores were evaluated and compared in 433 patients with advanced NPC. The ΔCONUT score was found to be superior to the pre-CONUT and post-CONUT scores in terms of the predictive ability for prognosis, and it was an independent prognostic factor for OS. Moreover, our study suggested that a combination of the ΔCONUT score and the EBV DNA level was a novel tool for the prediction of poor future outcomes in patients with NPC.

Many reports have suggested that the CONUT score is a useful and convenient biomarker for estimating the nutritional status and predicting prognoses among patients with non-metastatic renal cell carcinoma12, esophageal cancer13,14, pancreatic ductal adenocarcinoma15, and stage 2/3 gastric cancer15. The CONUT score, an immunological and nutritional score, is calculated from the total lymphocyte count and total cholesterol and serum albumin levels17. Lymphocytes were found to be associated with cellular immunity against malignant cells28,29. Cholesterol reportedly reflects the nutritional status and cancer malignancy status30,31. Serum albumin level is an indicator of nutritional status32; however, it is more widely recognized as a marker of inflammation33,34. Previous studies have shown that low lymphocyte counts and low cholesterol and serum albumin levels are associated with poor prognoses in different cancers35–39.

This study is the first to assess the influence of changes in the CONUT score during treatment, so the ΔCONUT-EBV DNA score has potential applications in the development of nutritional and individualized treatments for the prognoses of patients with advanced NPC. In the present study, the ΔCONUT score was found to be superior to the pre-CONUT and post-CONUT scores in predicting survival for patients with NPC.

The mechanisms explaining why a high ΔCONUT score is associated with worse OS are not fully known. Side effects of anti-cancer treatment affect the nutritional status among patients with head and neck cancers. Oral mucositis is one of the most common side effects of anti-cancer treatments such as radiation therapy, chemotherapy, and immunotherapy40,41. It occurs in almost all patients receiving radiation therapy for head and neck cancers42–44. Clinically, its symptoms include anorexia, malnutrition (significant weight loss), and systemic infections. It also can influence treatment efficacy by causing interruptions in treatment or by resulting in dose reductions in chemotherapy and radiotherapy45. In turn, dose modification and treatment interruption have been associated with decreased survival46–48. Taken together, the ΔCONUT score might predict the prognoses of patients with NPC based on a combination of host malnutrition and immunity.

As mentioned in the Introduction, nutritional scores only reflect 1 aspect of the nutritional status and ignore tumor-related factors. In combination with nasopharyngeal and neck magnetic resonance imaging, nasopharyngoscopy, plasma EBV DNA levels and other blood indicators are routinely assessed in most hospitals. Across numerous studies, plasma EBV DNA levels have been shown to be associated with NPC stage49–51, suggesting a reliable and direct correlation between tumor burden and plasma EBV DNA levels52. The EBV DNA level is likely a reliable prognostic predictor and tumor-related factor for NPC3,53,54. The association between survival outcomes and the plasma EBV DNA levels have been investigated in many studies20,24,25. The cut-off values of plasma EBV DNA levels varied across these studies, and an optimal cut-off value of this liquid biopsy marker is still being determined27,55. Measurement of plasma EBV DNA levels is commonly used to diagnose NPC and to evaluate anti-cancer treatment and prognosis. Studies support a positive relationship between the EBV DNA level and tumor burden24,25. Finally, we further determined the complementary role of the ΔCONUT score in combination with pretreatment plasma EBV DNA.

In the subgroup analysis, the ΔCONUT score was found to be correlated with the OS of patients with a higher EBV DNA level, but not with the OS of patients with a lower EBV DNA level. Thus, the ΔCONUT-EBV DNA score, a novel combination prognostic marker, was introduced to improve the predictive value of the ΔCONUT score among patients with advanced NPC. Compared to the ΔCONUT score or pretreatment plasma EBV DNA level alone, the ΔCONUT-EBV DNA score was more predictive of the OS, PFS, and DMFS for patients with advanced NPC. Thus, patients with NPC who have a medium or high CONUT-EBV DNA score could benefit from a more intensive follow-up, even after curative CCRT with a nutritional intervention during treatment; this may be clinically beneficial in improving the treatment outcomes of these patients.

Nutritional and inflammation status have significant effects on the prognoses of cancer patients. Many studies have already shown that inflammatory markers, nutritional indices, or inflammation-based prognostic scores including red blood cell56, total lymphocyte count57, albumin58–60, hemoglobin61,62, serum pre-albumin63,64, transferrin65, serum C-reactive protein66, BMI7,67–69 and prognostic nutritional index (PNI)70 are closely associated with treatment outcomes in patients with NPC. Using these blood indicators could only reflect 1 aspect of the nutritional status, so the sensitivity of the scores may differ. These indicators could be easily affected by metabolism, food, and disease status71–74. The utility of BMI and PNI are useful to assess the nutritional status in clinical practice. Individual nutritional factors, such as hemoglobin, albumin, and BMI were not prognostic factors in Wang’s cohort of patients with NPC75. However, BMI and PNI are easily influenced by gender, age, and disease status. Using only BMI, PNI or 1 blood indicator to assess the nutritional status of patients may therefore introduce errors76. These nutritional or immune-nutritional scores are not always reliable in predicting the outcomes of cancers because they lack tumor-related factors. In addition, none of these scores are designed specifically for NPC patients.

However, the tumor-related factors, such as TNM stage and plasma EBV DNA levels only reflect 1 aspect of the tumor-related status. These factors are always reliable in predicting the treatment outcomes of cancers. The EBV DNA level is likely a useful prognostic predictor and tumor-related factor for NPC. The association between survival outcomes and plasma EBV DNA levels have been investigated in many studies20,24,25. The cut-off values of the plasma EBV DNA level varied across these studies, and an optimal cut-off value of this liquid biopsy marker is still being determined27,55.

The relationship between survival outcomes and nutritional or tumor-related factors needs to be further established in NPC patients, although both nutritional scores and plasma EBV DNA levels are used in the clinic. However, there are only a few clinical studies that have characterized the applicability of different nutritional scores when combined with tumor-related factors and the plasma EBV DNA levels in patients with NPC. The ΔCONUT-EBV DNA score includes plasma EBV DNA level and also assesses the change of immune-nutritional status during anti-cancer treatment, resulting in a particularly relevant multidimensional score. The novel combined ΔCONUT-EBV DNA scoring system provides more comprehensive prognostic information than individual nutritional indexes.

Our study had some limitations. First, it was a retrospective study performed at a single center. Second, other nutritional scores, such as PNI, BMI, and platelet:lymphocyte ratio, were not assessed. Thus, we do not know if changes in other nutritional scores during the treatment influenced the survival of patients with advanced NPC. Third, information on the side effects of anti-cancer treatment and food intake during the treatment was insufficient for the further analysis of OS, PFS, and DMFS. Thus, further prospective studies are required to establish the value of the ΔCONUT-EBV DNA score as a biomarker of prognosis and treatment outcomes in NPC.

Conclusions

Taken together, the ΔCONUT-EBV DNA score, which is based on the ΔCONUT score and pretreatment plasma EBV DNA, has been integrated with the nutritional score and tumor-related factors, which may have more predictive information when compared to only 1 parameter. This score could be important and useful in clinical practice as a convenient, inexpensive biomarker for predicting outcomes for patients with advanced NPC treated with CCRT. As a novel and convenient biomarker, the ΔCONUT-EBV DNA score can be used in the development of nutritional and individualized treatments.

Supporting Information

[cbm-19-551-s001.pdf]

Grant support

This work was supported by grants from the National Key R&D Program of China (Grant Nos. 2017YFC1309003 and 2017YFC0908500), the National Natural Science Foundation of China (Grant Nos. 81425018, 81672868, and 81802775), the Sci-Tech Project Foundation of Guangzhou City (Grant No. 201707020039), the Sun Yat-sen University Clinical Research 5010 Program, the Special Support Plan of Guangdong Province (Grant No. 2014TX01R145), the Natural Science Foundation of Guangdong Province (Grant Nos. 2017A030312003 and 2018A0303131004), the Natural Science Foundation of Guangdong Province for Distinguished Young Scholar (Grant No. 2018B030306001), the Sci-Tech Project Foundation of Guangdong Province (Grant No. 2014A020212103), the Health & Medical Collaborative Innovation Project of Guangzhou City (Grant Nos. 201400000001 and 201803040003), the Pearl River S&T Nova Program of Guangzhou (Grant No. 201806010135), the Planned Science and Technology Project of Guangdong Province (Grant No. 2019B020230002), the National Science & Technology Pillar Program during the Twelfth Five-year Plan Period (Grant No. 2014BAI09B10), the Natural Science Foundation of Guangdong Province (Grant No. 2017A030312003, and Fundamental Research Funds for the Central Universities.

Conflict of interest statement

No potential conflicts of interest are disclosed.

Footnotes

  • ↵*These authors contributed equally to this work.

  • Received October 8, 2020.
  • Accepted December 25, 2020.
  • Copyright: © 2022, Cancer Biology & Medicine
https://creativecommons.org/licenses/by/4.0/

This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY) 4.0, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

References

  1. 1.↵
    1. Tan GW,
    2. Sivanesan VM,
    3. Abdul Rahman FI,
    4. Hassan F,
    5. Hasbullah HH,
    6. Ng CC, et al.
    A novel and non-invasive approach utilising nasal washings for the detection of nasopharyngeal carcinoma. Int J Cancer. 2019; 145: 2260–6.
    OpenUrl
  2. 2.↵
    1. Chan KCA,
    2. Woo JKS,
    3. King A,
    4. Zee BCY,
    5. Lam WKJ,
    6. Chan SL, et al.
    Analysis of plasma Epstein-Barr virus DNA to screen for nasopharyngeal cancer. N Engl J Med. 2017; 377: 513–22.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Lo YM,
    2. Chan AT,
    3. Chan LY,
    4. Leung SF,
    5. Lam CW,
    6. Huang DP, et al.
    Molecular prognostication of nasopharyngeal carcinoma by quantitative analysis of circulating Epstein-Barr virus DNA. Cancer Res. 2000; 60: 6878–81.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Lo YM,
    2. Chan LY,
    3. Chan AT,
    4. Leung SF,
    5. Lo KW,
    6. Zhang J, et al.
    Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res. 1999; 59: 5452–5.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Leung SF,
    2. Lo YM,
    3. Chan AT,
    4. To KF,
    5. To E,
    6. Chan LY, et al.
    Disparity of sensitivities in detection of radiation-naive and postirradiation recurrent nasopharyngeal carcinoma of the undifferentiated type by quantitative analysis of circulating Epstein-Barr virus DNA1,2. Clin Cancer Res. 2003; 9: 3431–4.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Chen L,
    2. Zhang Y,
    3. Lai SZ,
    4. Li WF,
    5. Hu WH,
    6. Sun R, et al.
    10-Year results of therapeutic ratio by intensity-modulated radiotherapy versus two-dimensional radiotherapy in patients with nasopharyngeal carcinoma. Oncologist. 2019; 24: e38–45.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Huang JF,
    2. Sun RJ,
    3. Jiang WJ,
    4. Wu P,
    5. Zhang L,
    6. Xu MQ, et al.
    Systematic nutrition management for locally advanced nasopharyngeal carcinoma patients undergoing radiotherapy. Onco Targets Ther. 2019; 12: 8379–86.
    OpenUrl
  8. 8.↵
    1. Gundog M,
    2. Basaran H.
    Pretreatment low prognostic nutritional index and low albumin-globulin ratio are predictive for overall survival in nasopharyngeal cancer. Eur Arch Otorhinolaryngol. 2019; 276: 3221–30.
    OpenUrl
  9. 9.↵
    1. Wei GB,
    2. Lu YY,
    3. Liao RW,
    4. Chen QS,
    5. Zhang KQ.
    Prognostic nutritional index predicts prognosis in patients with metastatic nasopharyngeal carcinoma. Onco Targets Ther. 2016; 9: 5955–61.
    OpenUrl
  10. 10.↵
    1. Zeng X,
    2. Liu G,
    3. Pan Y,
    4. Li Y.
    Prognostic value of clinical biochemistry-based indexes in nasopharyngeal carcinoma. Front Oncol. 2020; 10: 146.
    OpenUrl
  11. 11.↵
    1. Harimoto N,
    2. Yoshizumi T,
    3. Inokuchi S,
    4. Itoh S,
    5. Adachi E,
    6. Ikeda Y, et al.
    Prognostic significance of preoperative controlling nutritional status (CONUT) score in patients undergoing hepatic resection for hepatocellular carcinoma: a multi-institutional study. Ann Surg Oncol. 2018; 25: 3316–23.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Song H,
    2. Xu B,
    3. Luo C,
    4. Zhang Z,
    5. Ma B,
    6. Jin J, et al.
    The prognostic value of preoperative controlling nutritional status score in non-metastatic renal cell carcinoma treated with surgery: a retrospective single-institution study. Cancer Manag Res. 2019; 11: 7567–75.
    OpenUrl
  13. 13.↵
    1. Hikage M,
    2. Taniyama Y,
    3. Sakurai T,
    4. Sato C,
    5. Takaya K,
    6. Okamoto H, et al.
    The influence of the perioperative nutritional status on the survival outcomes for esophageal cancer patients with neoadjuvant chemotherapy. Ann Surg Oncol. 2019; 26: 4744–53.
    OpenUrl
  14. 14.↵
    1. Toyokawa T,
    2. Kubo N,
    3. Tamura T,
    4. Sakurai K,
    5. Amano R,
    6. Tanaka H, et al.
    The pretreatment Controlling Nutritional Status (CONUT) score is an independent prognostic factor in patients with resectable thoracic esophageal squamous cell carcinoma: results from a retrospective study. BMC Cancer. 2016; 16: 722.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Terasaki F,
    2. Sugiura T,
    3. Okamura Y,
    4. Ito T,
    5. Yamamoto Y,
    6. Ashida R, et al.
    The preoperative controlling nutritional status (CONUT) score is an independent prognostic marker for pancreatic ductal adenocarcinoma. Updates Surg. 2020; 73: 251–9.
    OpenUrl
  16. 16.↵
    1. Ryo S,
    2. Kanda M,
    3. Ito S,
    4. Mochizuki Y,
    5. Teramoto H,
    6. Ishigure K, et al.
    The controlling nutritional status score serves as a predictor of short- and long-term outcomes for patients with stage 2 or 3 gastric cancer: analysis of a multi-institutional data set. Ann Surg Oncol. 2019; 26: 456–64.
    OpenUrl
  17. 17.↵
    1. Ignacio de Ulibarri J,
    2. Gonzalez-Madrono A,
    3. de Villar NG,
    4. Gonzalez P,
    5. Gonzalez B,
    6. Mancha A, et al.
    CONUT: a tool for controlling nutritional status. First validation in a hospital population. Nutr Hosp. 2005; 20: 38–45.
    OpenUrlPubMed
  18. 18.↵
    1. Chan AT.
    Nasopharyngeal carcinoma. Ann Oncol. 2010; 21 Suppl 7: vii308–12.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Lee AW,
    2. Ma BB,
    3. Ng WT,
    4. Chan AT.
    Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol. 2015; 33: 3356–64.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Lin JC,
    2. Wang WY,
    3. Chen KY,
    4. Wei YH,
    5. Liang WM,
    6. Jan JS, et al.
    Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med. 2004; 350: 2461–70.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Guo R,
    2. Tang LL,
    3. Mao YP,
    4. Du XJ,
    5. Chen L,
    6. Zhang ZC, et al.
    Proposed modifications and incorporation of plasma Epstein-Barr virus DNA improve the TNM staging system for Epstein-Barr virus-related nasopharyngeal carcinoma. Cancer. 2019; 125: 79–89.
    OpenUrlCrossRefPubMed
  22. 22.
    1. Lee VH,
    2. Kwong DL,
    3. Leung TW,
    4. Choi CW,
    5. O’Sullivan B,
    6. Lam KO, et al.
    The addition of pretreatment plasma Epstein-Barr virus DNA into the eighth edition of nasopharyngeal cancer TNM stage classification. Int J Cancer. 2019; 144: 1713–22.
    OpenUrl
  23. 23.↵
    1. Xu C,
    2. Chen YP,
    3. Liu X,
    4. Li WF,
    5. Chen L,
    6. Mao YP, et al.
    Establishing and applying nomograms based on the 8th edition of the UICC/AJCC staging system to select patients with nasopharyngeal carcinoma who benefit from induction chemotherapy plus concurrent chemoradiotherapy. Oral Oncol. 2017; 69: 99–107.
    OpenUrl
  24. 24.↵
    1. Ma BB,
    2. King A,
    3. Lo YM,
    4. Yau YY,
    5. Zee B,
    6. Hui EP, et al.
    Relationship between pretreatment level of plasma Epstein-Barr virus DNA, tumor burden, and metabolic activity in advanced nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2006; 66: 714–20.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Chan KC,
    2. Chan AT,
    3. Leung SF,
    4. Pang JC,
    5. Wang AY,
    6. Tong JH, et al.
    Investigation into the origin and tumoral mass correlation of plasma Epstein-Barr virus DNA in nasopharyngeal carcinoma. Clin Chem. 2005; 51: 2192–5.
    OpenUrlFREE Full Text
  26. 26.↵
    1. McMillan GH.
    Looking at problems–a new approach to injury records. Occup Health (Lond). 1980; 32: 286–98.
    OpenUrlPubMed
  27. 27.↵
    1. Qu H,
    2. Huang Y,
    3. Zhao S,
    4. Zhou Y,
    5. Lv W.
    Prognostic value of Epstein-Barr virus DNA level for nasopharyngeal carcinoma: a meta-analysis of 8128 cases. Eur Arch Otorhinolaryngol. 2020; 277: 9–18.
    OpenUrl
  28. 28.↵
    1. Romaniuk A,
    2. Lyndin M.
    Immune microenvironment as a factor of breast cancer progression. Diagn Pathol. 2015; 10: 79.
    OpenUrlPubMed
  29. 29.↵
    1. Bates JP,
    2. Derakhshandeh R,
    3. Jones L,
    4. Webb TJ.
    Mechanisms of immune evasion in breast cancer. BMC Cancer. 2018; 18: 556.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Chen P,
    2. Han L,
    3. Wang C,
    4. Jia Y,
    5. Song Q,
    6. Wang J, et al.
    Preoperative serum lipids as prognostic predictors in esophageal squamous cell carcinoma patients with esophagectomy. Oncotarget. 2017; 8: 41605–19.
    OpenUrl
  31. 31.↵
    1. Lee H,
    2. Jeong CW,
    3. Kwak C,
    4. Kim HH,
    5. Seo SI,
    6. Lee HM, et al.
    Preoperative cholesterol level is associated with worse pathological outcomes and postoperative survival in localized renal cell carcinoma patients: a propensity score-matched study. Clin Genitourin Cancer. 2017; 15: e935–41.
    OpenUrl
  32. 32.↵
    1. de Ulibarri Perez JI,
    2. Fernandez G,
    3. Rodriguez Salvanes F,
    4. Diaz Lopez AM.
    Nutritional screening; control of clinical undernutrition with analytical parameters. Nutr Hosp. 2014; 29: 797–811.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Roxburgh CS,
    2. McMillan DC.
    Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol. 2010; 6: 149–63.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. McMillan DC.
    Systemic inflammation, nutritional status and survival in patients with cancer. Curr Opin Clin Nutr Metab Care. 2009; 12: 223–6.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Remark R,
    2. Becker C,
    3. Gomez JE,
    4. Damotte D,
    5. Dieu-Nosjean MC,
    6. Sautes-Fridman C, et al.
    The non-small cell lung cancer immune contexture. A major determinant of tumor characteristics and patient outcome. Am J Respir Crit Care Med. 2015; 191: 377–90.
    OpenUrlCrossRefPubMed
  36. 36.
    1. Campian JL,
    2. Sarai G,
    3. Ye X,
    4. Marur S,
    5. Grossman SA.
    Association between severe treatment-related lymphopenia and progression-free survival in patients with newly diagnosed squamous cell head and neck cancer. Head Neck. 2014; 36: 1747–53.
    OpenUrlCrossRefPubMed
  37. 37.
    1. Deng W,
    2. Xu C,
    3. Liu A,
    4. van Rossum PSN,
    5. Deng W,
    6. Liao Z, et al.
    The relationship of lymphocyte recovery and prognosis of esophageal cancer patients with severe radiation-induced lymphopenia after chemoradiation therapy. Radiother Oncol. 2019; 133: 9–15.
    OpenUrl
  38. 38.
    1. Kanda M,
    2. Tanaka C,
    3. Kobayashi D,
    4. Uda H,
    5. Inaoka K,
    6. Tanaka Y, et al.
    Preoperative albumin-bilirubin grade predicts recurrences after radical gastrectomy in patients with pT2-4 gastric cancer. World J Surg. 2018; 42: 773–81.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. Wu N,
    2. Chen G,
    3. Hu H,
    4. Pang L,
    5. Chen Z.
    Low pretherapeutic serum albumin as a risk factor for poor outcome in esophageal squamous cell carcinomas. Nutr Cancer. 2015; 67: 481–5.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Pico JL,
    2. Avila-Garavito A,
    3. Naccache P.
    Mucositis: its occurrence, consequences, and treatment in the oncology setting. Oncologist. 1998; 3: 446–51.
    OpenUrlFREE Full Text
  41. 41.↵
    1. Bensinger W,
    2. Schubert M,
    3. Ang KK,
    4. Brizel D,
    5. Brown E,
    6. Eilers JG, et al.
    NCCN Task Force Report. prevention and management of mucositis in cancer care. J Natl Compr Canc Netw. 2008; 6 (Suppl 1): S1–21; quiz S22-24.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Sonis ST,
    2. Elting LS,
    3. Keefe D,
    4. Peterson DE,
    5. Schubert M,
    6. Hauer-Jensen M, et al.
    Perspectives on cancer therapy-induced mucosal injury: pathogenesis, measurement, epidemiology, and consequences for patients. Cancer. 2004; 100: 1995–2025.
    OpenUrlCrossRefPubMed
  43. 43.
    1. Mortensen HR,
    2. Overgaard J,
    3. Specht L,
    4. Overgaard M,
    5. Johansen J,
    6. Evensen JF, et al.
    Prevalence and peak incidence of acute and late normal tissue morbidity in the DAHANCA 6&7 randomised trial with accelerated radiotherapy for head and neck cancer. Radiother Oncol. 2012; 103: 69–75.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Blijlevens N,
    2. Schwenkglenks M,
    3. Bacon P,
    4. D’Addio A,
    5. Einsele H,
    6. Maertens J, et al.
    Prospective oral mucositis audit: oral mucositis in patients receiving high-dose melphalan or BEAM conditioning chemotherapy–European Blood and Marrow Transplantation Mucositis Advisory Group. J Clin Oncol. 2008; 26: 1519–25.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    1. Trotti A,
    2. Bellm LA,
    3. Epstein JB,
    4. Frame D,
    5. Fuchs HJ,
    6. Gwede CK, et al.
    Mucositis incidence, severity and associated outcomes in patients with head and neck cancer receiving radiotherapy with or without chemotherapy: a systematic literature review. Radiother Oncol. 2003; 66: 253–62.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Rosenthal DI.
    Consequences of mucositis-induced treatment breaks and dose reductions on head and neck cancer treatment outcomes. J Support Oncol. 2007; 5: 23–31.
    OpenUrlPubMed
  47. 47.
    1. Russo G,
    2. Haddad R,
    3. Posner M,
    4. Machtay M.
    Radiation treatment breaks and ulcerative mucositis in head and neck cancer. Oncologist. 2008; 13: 886–98.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    1. Campos MI,
    2. Campos CN,
    3. Aarestrup FM,
    4. Aarestrup BJ.
    Oral mucositis in cancer treatment: natural history, prevention and treatment. Mol Clin Oncol. 2014; 2: 337–40.
    OpenUrl
  49. 49.↵
    1. Lo Y,
    2. Chan L,
    3. Lo K,
    4. Leung S,
    5. Zhang J,
    6. Chan A, et al.
    Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res. 1999; 59: 1188–91.
    OpenUrlAbstract/FREE Full Text
  50. 50.
    1. Vo J,
    2. Nei W,
    3. Hu M,
    4. Phyo W,
    5. Wang F,
    6. Fong K, et al.
    Comparison of circulating tumour cells and circulating cell-free Epstein-Barr virus DNA in patients with nasopharyngeal carcinoma undergoing radiotherapy. Sci Rep. 2016; 6: 13.
    OpenUrlCrossRef
  51. 51.↵
    1. Prayongrat A,
    2. Chakkabat C,
    3. Kannarunimit D,
    4. Hansasuta P,
    5. Lertbutsayanukul C.
    Prevalence and significance of plasma Epstein-Barr Virus DNA level in nasopharyngeal carcinoma. J Radiat Res. 2017; 58: 509–16.
    OpenUrl
  52. 52.↵
    1. Tan R,
    2. Phua S,
    3. Soong Y,
    4. Oon L,
    5. Chan K,
    6. Lucky S, et al.
    Clinical utility of Epstein-Barr virus DNA and other liquid biopsy markers in nasopharyngeal carcinoma. Cancer Commun (London, England). 2020; 40: 564–85.
    OpenUrl
  53. 53.↵
    1. Chan A,
    2. Lo Y,
    3. Zee B,
    4. Chan L,
    5. Ma B,
    6. Leung S, et al.
    Plasma Epstein-Barr virus DNA and residual disease after radiotherapy for undifferentiated nasopharyngeal carcinoma. J Natl Cancer Inst. 2002; 94: 1614–9.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Wang W,
    2. Twu C,
    3. Chen H,
    4. Jan J,
    5. Jiang R,
    6. Chao J, et al.
    Plasma EBV DNA clearance rate as a novel prognostic marker for metastatic/recurrent nasopharyngeal carcinoma. Clin Cancer Res. 2010; 16: 1016–24.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. Zhang J,
    2. Shu C,
    3. Song Y,
    4. Li Q,
    5. Huang J,
    6. Ma X.
    Epstein-Barr virus DNA level as a novel prognostic factor in nasopharyngeal carcinoma: a meta-analysis. Medicine. 2016; 95: e5130.
  56. 56.↵
    1. Yao ZH,
    2. Tian GY,
    3. Wan YY,
    4. Kang YM,
    5. Guo HS,
    6. Liu QH, et al.
    Prognostic nutritional index predicts outcomes of malignant pleural mesothelioma. J Cancer Res Clin Oncol. 2013; 139: 2117–23.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Chen K,
    2. Liu Y,
    3. Li W,
    4. Chen J,
    5. Gu Y,
    6. Geng Q, et al.
    The prognostic nutritional index predicts survival for patients with extranodal natural killer/T cell lymphoma, nasal type. Ann Hematol. 2015; 94: 1389–400.
    OpenUrl
  58. 58.↵
    1. Li G,
    2. Gao J,
    3. Liu Z,
    4. Tao Y,
    5. Xu B,
    6. Tu Z, et al.
    Influence of pretreatment ideal body weight percentile and albumin on prognosis of nasopharyngeal carcinoma: long-term outcomes of 512 patients from a single institution. Head Neck. 2014; 36: 660–6.
    OpenUrl
  59. 59.
    1. Liu S,
    2. Tsai W,
    3. Wong Y,
    4. Lin J,
    5. Poon C,
    6. Chao S, et al.
    Nutritional factors and survival of patients with oral cancer. Head Neck. 2006; 28: 998–1007.
    OpenUrlCrossRefPubMed
  60. 60.↵
    1. Tartari R,
    2. Ulbrich-Kulczynski J,
    3. Filho A.
    Measurement of mid-arm muscle circumference and prognosis in stage IV non-small cell lung cancer patients. Oncol Lett. 2013; 5: 1063–7.
    OpenUrl
  61. 61.↵
    1. Xie X,
    2. Yao M,
    3. Chen X,
    4. Lu W,
    5. Lv Q,
    6. Wang K, et al.
    Reduced red blood cell count predicts poor survival after surgery in patients with primary liver cancer. Medicine. 2015; 94: e577.
  62. 62.↵
    1. Mayr N,
    2. Wang J,
    3. Zhang D,
    4. Montebello J,
    5. Grecula J,
    6. Lo S, et al.
    Synergistic effects of hemoglobin and tumor perfusion on tumor control and survival in cervical cancer. Int J Radiat Oncol Biol Phys. 2009; 74: 1513–21.
    OpenUrlPubMed
  63. 63.↵
    1. Iwasa M.
    [Nutritional assessment of patients with esophageal cancer. “Nutritional Assessment Index (NAI)” to estimate nutritional conditions in pre-and postoperative period]. Nihon Geka Gakkai Zasshi. 1983; 84: 1031–41.
    OpenUrlPubMed
  64. 64.↵
    1. Onizuka K,
    2. Migita S,
    3. Yamada H,
    4. Matsumoto I.
    [Serum protein fractions in patients with laryngeal cancer undergoing radiation therapy. Possibility as a prognostic factor]. Fukuoka Igaku Zasshi. 1999; 90: 46–58.
    OpenUrlPubMed
  65. 65.↵
    1. Lai S,
    2. Perng R.
    Impact of nutritional status on the survival of lung cancer patients. Zhonghua Yi Xue Za Zhi. 1998; 61: 134–40.
    OpenUrl
  66. 66.↵
    1. Tang L,
    2. Li C,
    3. Chen Q,
    4. Zhang L,
    5. Lai X,
    6. He Y, et al.
    High-sensitivity C-reactive protein complements plasma Epstein-Barr virus deoxyribonucleic acid prognostication in nasopharyngeal carcinoma: a large-scale retrospective and prospective cohort study. Int J Radiat Oncol Biol Phys. 2015; 91: 325–36.
    OpenUrlCrossRefPubMed
  67. 67.↵
    1. OuYang PY,
    2. Zhang LN,
    3. Tang J,
    4. Lan XW,
    5. Xiao Y,
    6. Gao YH, et al.
    Evaluation of body mass index and survival of nasopharyngeal carcinoma by propensity-matched analysis: an observational case-control study. Medicine (Baltimore). 2016; 95: e2380.
  68. 68.
    1. Sinicrope F,
    2. Foster N,
    3. Yothers G,
    4. Benson A,
    5. Seitz J,
    6. Labianca R, et al.
    Body mass index at diagnosis and survival among colon cancer patients enrolled in clinical trials of adjuvant chemotherapy. Cancer. 2013; 119: 1528–36.
    OpenUrlCrossRefPubMed
  69. 69.↵
    1. Zhang S,
    2. Yang H,
    3. Luo K,
    4. Huang Q,
    5. Chen J,
    6. Yang F, et al.
    The impact of body mass index on complication and survival in resected oesophageal cancer: a clinical-based cohort and meta-analysis. Br J Cancer. 2013; 109: 2894–903.
    OpenUrlPubMed
  70. 70.↵
    1. Yang L,
    2. Xia L,
    3. Wang Y,
    4. Hong S,
    5. Chen H,
    6. Liang S, et al.
    Low prognostic nutritional index (PNI) predicts unfavorable distant metastasis-free survival in nasopharyngeal carcinoma: a propensity score-matched analysis. PLoS One. 2016; 11: e0158853.
  71. 71.↵
    1. Fuhrman M,
    2. Charney P,
    3. Mueller C.
    Hepatic proteins and nutrition assessment. J Am Diet Assoc. 2004; 104: 1258–64.
    OpenUrlCrossRefPubMed
  72. 72.
    1. Dequanter D,
    2. Lothaire P.
    Serum albumin concentration and surgical site identify surgical risk for major post-operative complications in advanced head and neck patients. B-ENT. 2011; 7: 181–3.
    OpenUrlPubMed
  73. 73.
    1. Guerra L,
    2. Rosa A,
    3. Romani R,
    4. Gurski R,
    5. Schirmer C,
    6. Kruel C.
    Serum transferrin and serum prealbumin as markers of response to nutritional support in patients with esophageal cancer. Nutr Hosp. 2009; 24: 241–2.
    OpenUrlPubMed
  74. 74.↵
    1. Hong J,
    2. Hua Y,
    3. Su L,
    4. Zhang H,
    5. Lv W,
    6. Chen X, et al.
    Modified-nutrition index is a significant prognostic factor for the overall survival of the nasopharyngeal carcinoma patients who undergo intensity-modulated radiotherapy. Nutr Cancer. 2017; 69: 1011–8.
    OpenUrl
  75. 75.↵
    1. Wang Y,
    2. He S,
    3. Cai X,
    4. Chen H,
    5. Yang X,
    6. Lu L, et al.
    The novel prognostic score combining red blood cell distribution width and body mass index (COR-BMI) has prognostic impact for survival outcomes in nasopharyngeal carcinoma. J Cancer. 2018; 9: 2295–301.
    OpenUrl
  76. 76.↵
    1. Lin Y,
    2. Chang K,
    3. Lin Y,
    4. Chang T.
    Evaluation of effect of body mass index and weight loss on survival of patients with nasopharyngeal carcinoma treated with intensity-modulated radiation therapy. Radiat Oncol. 2015; 10: 136.
    OpenUrl
PreviousNext
Back to top

In this issue

Cancer Biology & Medicine: 19 (4)
Cancer Biology & Medicine
Vol. 19, Issue 4
15 Apr 2022
  • Table of Contents
  • Index by author
Print
Download PDF
Email Article

Thank you for your interest in spreading the word on Cancer Biology & Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Prognostic significance of a combined and controlled nutritional status score and EBV-DNA in patients with advanced nasopharyngeal carcinoma: a long-term follow-up study
(Your Name) has sent you a message from Cancer Biology & Medicine
(Your Name) thought you would like to see the Cancer Biology & Medicine web site.
Citation Tools
Prognostic significance of a combined and controlled nutritional status score and EBV-DNA in patients with advanced nasopharyngeal carcinoma: a long-term follow-up study
Hui Lu, Shanshan Guo, Liting Liu, Qiuyan Chen, Yujing Liang, Sailan Liu, Xuesong Sun, Qingnan Tang, Xiaoyun Li, Ling Guo, Haoyuan Mo, Linquan Tang, Haiqiang Mai
Cancer Biology & Medicine Apr 2022, 19 (4) 551-564; DOI: 10.20892/j.issn.2095-3941.2020.0627

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Prognostic significance of a combined and controlled nutritional status score and EBV-DNA in patients with advanced nasopharyngeal carcinoma: a long-term follow-up study
Hui Lu, Shanshan Guo, Liting Liu, Qiuyan Chen, Yujing Liang, Sailan Liu, Xuesong Sun, Qingnan Tang, Xiaoyun Li, Ling Guo, Haoyuan Mo, Linquan Tang, Haiqiang Mai
Cancer Biology & Medicine Apr 2022, 19 (4) 551-564; DOI: 10.20892/j.issn.2095-3941.2020.0627
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and methods
    • Results
    • Discussion
    • Conclusions
    • Supporting Information
    • Grant support
    • Conflict of interest statement
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • References
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Akt isoforms differentially provide for chemoresistance in prostate cancer
  • Somatic copy number alterations are predictive of progression-free survival in patients with lung adenocarcinoma undergoing radiotherapy
  • Improved diagnosis of thyroid cancer aided with deep learning applied to sonographic text reports: a retrospective, multi-cohort, diagnostic study
Show more Original Article

Similar Articles

Keywords

  • Controlling nutritional status score
  • Epstein-Barr virus deoxyribonucleic acid
  • nasopharyngeal carcinoma
  • prognosis
  • predictive factor

Navigate

  • Home
  • Current Issue

More Information

  • About CBM
  • About CACA
  • About TMUCIH
  • Editorial Board
  • Subscription

For Authors

  • Instructions for authors
  • Journal Policies
  • Submit a Manuscript

Journal Services

  • Email Alerts
  • Facebook
  • RSS Feeds
  • Twitter

 

© 2022 Cancer Biology & Medicine

Powered by HighWire