Skip to main content

Prognostic value of sarcopenia and inflammatory indices synergy in patients with esophageal squamous cell carcinoma undergoing chemoradiotherapy

Abstract

Background and purpose

Sarcopenia has been demonstrated to be adversely correlated with the prognosis of various cancers. Our study aimed to estimate the prognostic value of sarcopenia in conjunction with inflammatory indices [neutrophil-to-lymphocyte ratio (NLR)] for evaluating the prognosis of patients with esophageal squamous cell carcinoma (ESCC) undergoing chemoradiotherapy.

Materials and methods

This study retrospectively analyzed 255 patients with ESCC who received chemoradiotherapy from January 2012 to December 2018. Multivariate Cox regression analysis was employed to identify prognostic values of assessed factors following a novel prognostic scoring system (SMI-NLR), covering sarcopenia and NLR during different treatment courses.

Results

Kaplan–Meier analysis revealed significantly greater overall survival (OS) rates in the nonsarcopenia group than in the sarcopenia group (P = 0.011). The low NLR group (< 4.84) demonstrated significantly higher OS rates than the high NLR group (≥ 4.84) (P < 0.001). The SMI-NLR prognostic model was established through multivariate analysis, revealing that Karnofsky performance status [hazard ratio (HR) = 0.285; 95% confidence interval (CI) = 0.117–0.699; P = 0.006], clinical staging (HR = 5.223; 95% CI = 1.879–14.514; P = 0.002), and preSMI-NLR (HR = 0.544; 95% CI = 0.330–0.898; P = 0.017) were independent factors affecting the prognosis of patients with ESCC. Nomograms were constructed based on these data providing more accurate 1-, 3-, and 5-year survival rates for patients with ESCC.

Conclusion

Our study indicates the effectiveness of the combined sarcopenia and NLR prognostic model for the prognostic evaluation of patients with ESCC having undergone chemoradiotherapy.

Peer Review reports

Introduction

Esophageal malignancies have gained global attention due to their high incidence and mortality rates. Recent data indicate over 570,000 new cases annually [1]. Esophageal cancer is primarily categorized into esophageal squamous cell carcinoma (ESCC) and adenocarcinoma, with a significantly varying geographical distribution, and adenocarcinoma is predominant in Europe and America, whereas ESCC is more common in Asia [2,3,4]. Surgical resection remains the treatment of choice, but the condition is discovered at advanced stages in approximately 70% of patients, and chemoradiotherapy is the recommended standard of care for those unable or unwilling to undergo surgery [5,6,7]. Smoking, alcohol consumption, and poor dietary habits possess the greatest risks for developing esophageal cancer making effective cessation and early screening and diagnosis of paramount importance [4, 8, 9]. The overall prognosis remains poor despite the elaboration of new strategies, including targeted and immunotherapies, offering more options to patients [10, 11].

Currently, tumor staging serves as an essential measure for guiding clinical treatment and evaluating prognoses in patients with esophageal cancer; however, significant disparities remain within the same clinical stages in the latter [12]. Therefore, the development of effective screening tools determined high-risk individuals and conducted tailored treatment demonstrated significant clinical value. Recent studies have revealed that easily measurable blood biomarkers, such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PNI), and systemic immune-inflammation index, can reflect systemic inflammation levels along with the efficacy of performed treatment and survival prognosis in patients with cancer [13,14,15].

Sarcopenia, as outlined by the European Working Group on Sarcopenia in Older People and the Asian Working Group on Sarcopenia, is a syndrome exhibiting progressively reduced and generalized loss of skeletal muscle mass and strength associated with physical disability, deteriorated quality of life, and higher mortality risks [16,17,18]. Recent research indicates that systemic inflammation may be an integral part of the onset of sarcopenia and that the prognostic properties of combined evaluation of systemic inflammation and sarcopenia reveal more stability and effectiveness than single-factor [19,20,21]. The interaction of the factors in pre-treatment sarcopenia and systemic inflammation and their potential effect on the outcomes in patients with esophageal cancer remains unclear.

However, the synergistic effect of sarcopenia and systemic inflammation on the prognosis of patients with esophageal cancer remains unclear. Hence, this study focuses on analyzing the interplay between sarcopenia and the NLR in patients with esophageal cancer subjected to chemoradiotherapy and its effect on patient prognosis. We aim to construct a novel prognostic scoring model based on these biomarkers to forecast more precisely long-term survival outcomes for patients receiving chemoradiotherapy and provide more targeted and effective treatment plans. Additionally, we analyzed the changes in skeletal muscle index (SMI) and NLR levels before and after therapy and their influence on prognoses, further validating the accuracy of prognostic prediction with these factors through subgroup analysis.

Materials and methods

Study population

This study retrospectively evaluated 255 patients with ESCC who received chemoradiotherapy at Sichuan Cancer Hospital from January 2012 to December 2018. The following inclusion criteria were applied: (1) histologically confirmed ESCC; (2) ESCC that was either unresectable or the patient was unwilling to undergo surgery; (3) a Karnofsky performance status (KPS) score of ≥ 70; (4) a radiation dose of ≥ 40 Gy; (5) availability of complete blood count from one week before treatment initiation; (6) no evidence of distant metastases; (7) TNM staging based on the 7th edition of the American Joint Committee on Cancer staging system.

Treatment

All the patients underwent radiotherapy, with spiral computed tomography (CT) scans used for tumor target delineation and visualizing adjacent nonaffected organs. Three-dimensional conformal radiotherapy (3D-CRT) or intensity-modulated radiotherapy (IMRT), with PTV doses of 50–72 Gy over 25–36 fractions, were performed across 5–7 weeks. Constraints were set with V20 of ≤ 25% for both lungs, V30 of ≤ 40% for the heart, V40 of ≤ 30%, and a maximum spinal cord dose of ≤ 45.0 Gy. Some patients received 1–6 cycles of platinum-based monotherapy or combination chemotherapy, while older patients were subjected to oral tegafur treatment, and those intolerant to first-line chemotherapy were given raltitrexed.

Blood biomarker calculation methods: Pre-treatment and post-treatment serum albumin levels, lymphocyte counts, and neutrophil counts were collected. According to previous reports, PNI was defined as 10 × serum albumin level (g/dL) + 0.005 × lymphocyte count (mm3); NLR was defined as the neutrophil count divided by the lymphocyte count [15].

Body composition calculation

Patient CT images were obtained from the PACS system of the Sichuan Cancer Hospital Imaging Department. Two trained radiologists, blinded to the outcomes, delineated the skeletal muscle area (SMA) at the L3 level using semiautomated SliceOmatic software (version 5.0; Tomovision, Montreal, QC, Canada), based on attenuation thresholds of -29 to + 150 Hounsfield Units, that covered psoas, quadratus lumborum, erector spinae, transversus abdominis, internal oblique, external oblique, and rectus abdominis muscles. The SMI was calculated as SMA (cm2)/height2 (m2) [22], with the diagnostic thresholds for sarcopenia set at 40.8 cm2/m2 for men and 34.9 cm2/m2 for women following the research within Asian populations [18].

Outcome measures and follow-up

Follow-up was conducted every 3 months in the first year, every 6 months in the following 2 years, and annually thereafter. Routine evaluations included physical examinations, blood tests, ultrasonography, tumor marker assays, chest CT imaging, and esophagography with barium meal. All patients were followed up through outpatient visits and telephone interviews.

Statistical methods

Statistical Package for the RStudio version 4.3.1 (R Foundation for Statistical Computing) were used for data analysis and visualization. Chi-square or Fisher’s exact test was applied for categorical variables and Student’s t-test for continuous variables. The optimal stratification of continuous covariates for different treatment stages, such as NLR, PNI, and SMI-NLR cut-off values, was performed with log-rank tests [23, 24]. Patients were allocated to low and high groups according to the latter. The Kaplan–Meier method, with differences between curves analyzed using the log-rank test, was used to plot Survival curves. Uni- and multivariate analyses were performed with Cox proportional hazards models and a significance level was established at α of 0.05 and the statistical significance at a p-value of < 0.05.

Results

Patient characteristics

This study enrolled 255 patients with ESCC meeting the inclusion criteria. The median age of the participants was 64 years (range: 34–87 years), while the gender distribution was reflected in 200 (78.5%) men and 55 (21.5%) women. Of them, 147 (57.6%) and 108 (42.4%) patients reported with and without smoking habit. Alcohol consumption was reported in 140 (54.9%) patients, while 115 (45.1%) were non-drinkers. Tumors of < 5 cm in size were observed in 102 cases (40.0%), and those of ≥ 5 cm were observed in 153 (60.0%) patients. Stages I–II of the disease were noted in 16 (6.3%) cases, and stages III–IV were detected in 239 (96.7%) cases. Additionally, 39 (15.3%) patients received no chemotherapy, whereas 216 (84.7%) patients did (Table 1).

Table 1 The distribution of background variables stratified by survival status

The relationship between patient prognosis and sarcopenia

In a study of 255 patients, 1-, 3-, and 5-year overall survival (OS) rates were 68.9%, 41.8%, and 30.9%, respectively. Patients with a pre-treatment NLR (preNLR) < 4.84 showed higher OS rates of 73.1%, 46.3%, and 34.7%, compared to those with NLR ≥ 4.84, who had OS rates of 50.0%, 21.4%, and 15.3%. Kaplan–Meier analysis revealed significantly higher OS in the former group (p < 0.001, Fig. 1A and B). Additionally, non-sarcopenic patients had 1-, 3-, and 5-year OS rates of 72.3%, 45.4%, and 34.1%, respectively, surpassing the 57.6%, 37.3%, and 20.3% rates observed in sarcopenic patients. Kaplan–Meier analysis significantly higher OS rates in the former than in the latter (p = 0.011, Fig. 1C and D).

Fig. 1
figure 1

The Kaplan–Meier survival curves of SMI、NLR in the OS of overall. preSMI, pre-treatment skeletal muscle index; preNLR, pre-treatment neutrophil-to-lymphocyte ratio, postSMI, post-treatment skeletal muscle index; postNLR, post-treatment neutrophil-to-lymphocyte ratio

Skeletal muscle mass index and sarcopenia incidence

Pre-treatment SMI (preSMI) and preNLR levels were analyzed in gender groups, and the associations with age, KPS score, tumor length, and clinical stage were revealed, all demonstrating statistical significance (p < 0.05, Fig. 2A-H). Sarcopenia was present in 59 (23.1%) patients before treatment, including 47 men (23.5%) and 12 women (21.8%). Sarcopenia was found in 66 (25.9%) patients, including 51 men (25.5%) and 15 women (27.3%), after the therapy (Table 2).

Fig. 2
figure 2

The distribution of SMI in different groups. KPS, Karnofsky performance score; BMI, Body Mass Index; SMI, skeletal muscle index; NLR, neutrophil-to-lymphocyte ratio; PreSMI-NLR, pre-treatment skeletal muscle index combined with pre-treatment neutrophil-to-lymphocyte ratio; postSMI-NLR, post-treatment skeletal muscle index combined with post-treatment neutrophil-to-lymphocyte ratio

Table 2 The distribution of background variables stratified by survival status

Correlation between sarcopenia and inflammation

Uni- and multivariate logistic regression analyses were performed to investigate the correlation between systemic inflammation levels and SMI. The former identified clinical stage and preNLR as predictors for preSMI. Multivariate analysis revealed that clinical stage (hazard ratio [HR] = 0.494, 95% confidence interval [CI] = 0.290–0.844; p = 0.010) and preNLR (HR = 1.099; 95% CI = 1.039–1.162; p = 0.001) were the essential predictors for preSMI (Table 3). Considering the significant role of inflammatory components in sarcopenia, combined with the aforementioned analysis, we have hypothesized that SMI conjugated with NLR is a potentially effective tool for evaluating the prognoses in patients undergoing chemoradiotherapy. We combined SMI and NLR levels to create four subgroups to further differentiate patients according to distinct outcomes. Patients with sarcopenia and preNLR of ≥ 4.84 demonstrated the poorest survival rates, while those without the former and preNLR of < 4.84 exhibited the highest survival rates (p = 0.004). However, the survival rates for the patients from the former group were similar to those without sarcopenia but with preNLR of ≥ 4.84 (p > 0.05, Fig. 1E and F).

Table 3 Univariate and multivariate Cox regression analyses of factors associated with Sarcopenia

Establishment of a prognostic model based on sarcopenia and NLR

Optimal stratification was implemented to determine the best cut-off values for continuous covariates resulting in a pre-treatment SMI-NLR (preSMI-NLR) threshold of 11.04 and a post-treatment SMI-NLR (postSMI-NLR) of 3.53. The preSMI-NLR for the whole sample was 18.27 ± 11.20, and the postSMI-NLR composed 8.71 ± 24.6, showing a downward trend (Fig. 2I and J). Spearman’s analysis was performed on various clinical parameters associated with preSMI-NLR. Significant correlations were obtained for body mass index (BMI), SMA, SMI, PNI, NLR, tumor length, and clinical stage (p < 0.05, Fig. 3).

Fig. 3
figure 3

The Person analysis of SMI-NLR and related-factors. KPS, Karnofsky performance score; BMI, Body Mass Index; SMI, skeletal muscle index; NLR, neutrophil-to-lymphocyte ratio; SMI-NLR,skeletal muscle index combined with neutrophil-to-lymphocyte ratio

Kaplan–Meier analysis revealed that the high preSMI-NLR group demonstrated significantly higher OS rates than the low preSMI-NLR (p < 0.001, Fig. 4). The univariate analysis reflected the link between KSP, tumor length, clinical stage, BMI, chemotherapy, SMI, PNI, NLR, and preSMI-NLR and the prognosis of patients with ESCC (p < 0.05). Multivariate analysis revealed that KSP (HR = 0.285; 95% CI = 0.117–0.699; p = 0.006), clinical stage (HR = 5.223; 95% CI = 1.879–14.514; p = 0.002), and preSMI-NLR (HR = 0.544; 95% CI = 0.330–0.898; p = 0.017) were independent factors that affected the prognosis of patients with ESCC (Table 4).

Fig. 4
figure 4

The Kaplan–Meier survival curves of SMI-NLR in the OS of overall. PreSMI-NLR, pre-treatment skeletal muscle index combined with pre-treatment neutrophil-to-lymphocyte ratio; postSMI-NLR, post-treatment skeletal muscle index combined with post-treatment neutrophil-to-lymphocyte ratio

Table 4 Univariate and multivariate Cox regression analyses of factors associated with overall survival

Construction of a risk-scoring prognostic model

This study revealed significant non-conformities in the prognoses within the same TNM stage while TNM staging is essential in the treatment and prognostic assessment in patients with cancer. We based our risk scoring on a multivariate Cox regression hazard model to provide a quantitative method for better outcome predictions. The risk score was designed to reveal the prognostic outcomes of patients. Heatmaps of prognosis, survival curves, and time-dependent survival curves for different risk scores significantly differed (p < 0.05). Results revealed that individuals with higher risk scores demonstrated lower survival rates than those with lower risk. The prognostic receiver operating characteristic curves for 1-, 3-, and 5-year areas under the curve were 70.0, 73.6, and 75.5, respectively (Fig. 5A-F). Furthermore, we designed a nomogram incorporating independent prognostic factors to precisely predict ESCC patient outcomes following radiotherapy and chemotherapy. This nomogram, validated by its superior predictive accuracy and well-performing calibration plots compared to the ideal model, effectively complements the TNM staging system (Fig. 6A-C).

Fig. 5
figure 5

The prognostic risk score model of ECSS based on SMI-NLR score. AUC, area under curve; SMI-NLR,skeletal muscle index combined with neutrophil-to-lymphocyte ratio

Fig. 6
figure 6

The Nomogram constructed of ECSS based on SMI-NLR score. KPS, Karnofsky performance score; BMI, Body Mass Index; preSMI-NLR, pre-treatment skeletal muscle index combined with pre-treatment neutrophil-to-lymphocyte ratio

Discussion

Sarcopenia, a term first introduced by Rosenberg et al. [25] is a pathological condition that progressively develops with age, featuring a gradual decrease in overall muscle mass and strength leading to adverse events, such as falls and disability. Traditionally, sarcopenia has been considered a common phenomenon in the older adult age group. However, an increasing body of research has revealed a high incidence of sarcopenia in patients with cancer affecting not only their quality of life but also provoking adverse reactions to anti-cancer treatments and worsening prognoses [26, 27]. The skeletal muscle area at the third lumbar vertebra outlined in CT scans is the core part of SMI, which has become the “gold standard” for assessing muscle mass. SMI provides more information on body composition and can evaluate more accurately the nutritional status of patients.

Current studies reveal that sarcopenia significantly affects the survival rate of patients with head and neck, esophageal, lung, and gastrointestinal malignant tumors [20, 28,29,30,31]. Consequently, the definition of SMI varies considerably following ethnic and disease-specific characteristics. A study based on a European population defined the condition as that with SMI of < 52.4 cm^2/m^2 for men and SMI < 38.5 cm^2/m^2 for women. This study enrolled 97 patients with locally advanced esophageal cancer and revealed that the sarcopenia prevalence of 44% was associated with the increased overall mortality rate, with sarcopenia being an important prognostic risk factor (HR = 32.1 [24.4–34.0], p = 0.008) [31]. Similarly, Qian et al. used the aforementioned definition of sarcopenia and detected a prevalence rate of 79.8% in Asian patients with ESCC undergoing radiotherapy or CRT, which was associated with poorer PFS and OS rates. Sarcopenia, albumin levels, and tumor staging were determined as essential factors predicting survival [32].

The predictive role of sarcopenia is consistent with our study results; however, considering the differences in population ethnicity and disease types, our study adopted Asian population SMI cut-off values of 40.8 cm^2/m^2 for male patients and 34.9 cm^2/m^2 for female [18]. Our analysis investigated changes in SMI levels during different treatment processes, with sarcopenia occurring pre-treatment in 59 (23.1%) individuals and post-treatment in 66 (25.9%) individuals showing significant differences through paired analysis (p < 0.05). Furthermore, our study revealed that lower KPS levels and advanced clinical stages were significantly associated with decreased OS rates.

Multiple studies have indicated the pivotal role of chronic inflammation in the onset and progression of esophageal cancer [33, 34]. NLR is a biomarker reflecting systemic inflammatory status, and it has attracted attention in recent years for its application in evaluating prognoses for patients with cancer [35,36,37]. Neutrophils can secrete immunosuppressive mediators and angiogenic factors, forming tumor microenvironment and thereby promoting tumor progression [38, 39]. Conversely, circulating lymphocytes may inhibit tumor proliferation, invasion, and metastasis by enhancing tumor immune surveillance [40]. Our study revealed that patients with a low NLR had significantly higher survival rates than those with a high NLR. Nevertheless, postNLR failed to yield analogous outcomes. This discrepancy could stem from the inability of postNLR to fulfill its predictive function accurately, owing to myelosuppression, including neutropenia, and the employment of granulocyte colony-stimulating factors following radiotherapy and chemotherapy, which lead to significant variances in blood markers. Moreover, systemic inflammation levels were somewhat associated with the presence of cancer-related sarcopenia. Patients with esophageal cancer, who are prone to malnutrition due to prolonged inadequate food intake, advanced tumor staging, persistent high-inflammatory states, and intensive anti-tumor treatments, may activate the ubiquitin–proteasome pathway. This process can cause elevated levels of inflammatory cytokines, such as interleukin-1 (IL-1), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), which enhance the binding of ubiquitin to target proteins and accelerate protein degradation contributing to muscle contractile dysfunction by inducing the expression of E3 ligases [41].

Currently, the strong evidence regarding the role of sarcopenia and systemic inflammation in the prognosis of esophageal cancer is limited. A prospective cohort study involving 2470 patients with colorectal cancer revealed the positive association between sarcopenia and high NLR levels, and their combination could effectively identify the poorer prognoses of patients with early colorectal cancer [42]. Our study first discovered sarcopenia as an independent prognostic factor for patients with esophageal cancer receiving chemoradiotherapy. Simultaneously, we outlined NLR as a predictive factor for sarcopenia. The outcomes indicate that the combined sarcopenia–NLR model has better predictive power than considering sarcopenia or NLR alone and has been validated as an independent prognostic factor for esophageal cancer patients undergoing chemoradiotherapy. Interventions that affect tumor growth and treatment resistance for patients at higher risk can help improve their quality of life and survival prognosis.

However, our research has particular limitations. First, it is designed as retrospective, and the assessment of sarcopenia was based on imaging to evaluate SMI in patients, in whom the data on muscle function were not provided. Second, as a single-center study with a relatively small sample size, biases may exist that warrant validation in larger multi-center prospective studies. Furthermore, the complex network between tumor inflammation and body composition and its underlying mechanisms still requires further investigation.

Conclusion

Our study has combined two predictive factors of NLR and sarcopenia across different treatment processes that serve as one independent prognostic for patients with esophageal cancer receiving chemoradiotherapy. This predictive model is characterized by simplicity, reliability, and effective identification of high-risk patients and has certain clinical guiding significance.

Availability of data and materials

The original data supporting the findings of this study are available from the corresponding author upon reasonable request. Corresponding Author: Tao Li, email: litaoxmf@126.com.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Rustgi AK, El-Serag HB. Esophageal carcinoma. N Engl J Med. 2014;371(26):2499–509.

    Article  PubMed  Google Scholar 

  3. Castro C, Bosetti C, Malvezzi M, Bertuccio P, Levi F, Negri E, et al. Patterns and trends in esophageal cancer mortality and incidence in Europe (1980–2011) and predictions to 2015. Ann Oncol. 2014;25(1):283–90.

    Article  CAS  PubMed  Google Scholar 

  4. Kamangar F, Nasrollahzadeh D, Safiri S, Sepanlou SG, Fitzmaurice C, Ikuta KS, et al. The global, regional, and national burden of oesophageal cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol Hepatol. 2020;5(6):582–97.

    Article  Google Scholar 

  5. Zhang AD, Su XH, Shi GH, Han C, Wang L, Liu H, et al. Survival comparision of three-dimensional radiotherapy alone vs. chemoradiotherapy for esophageal squamous cell carcinoma. Arch Med Res. 2020;51(5):419–28.

    Article  CAS  PubMed  Google Scholar 

  6. van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366(22):2074–84.

    Article  PubMed  Google Scholar 

  7. Lordick F, Mariette C, Haustermans K, Obermannová R, Arnold D. Oesophageal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27:v50–7.

    Article  CAS  PubMed  Google Scholar 

  8. Arnold M, Laversanne M, Brown LM, Devesa SS, Bray F. Predicting the future burden of esophageal cancer by histological subtype: international trends in incidence up to 2030. Official J American College Gastroenterol| ACG. 2017;112(8):1247–55.

    Google Scholar 

  9. Uhlenhopp DJ, Then EO, Sunkara T, Gaduputi V. Epidemiology of esophageal cancer: update in global trends, etiology and risk factors. Clin J Gastroenterol. 2020;13(6):1010–21.

    Article  PubMed  Google Scholar 

  10. Yang Y-M, Hong P, Xu WW, He Q-Y, Li B. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Ther. 2020;5(1):229.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Triantafyllou T, Wijnhoven BPL. Current status of esophageal cancer treatment. Chin J Cancer Res. 2020;32(3):271–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rice TW, Ishwaran H, Blackstone EH, Hofstetter WL, Kelsen DP, Apperson-Hansen C. Recommendations for clinical staging (cTNM) of cancer of the esophagus and esophagogastric junction for the 8th edition AJCC/UICC staging manuals. Dis Esophagus. 2016;29(8):913–9.

    Article  PubMed  Google Scholar 

  13. Valero C, Lee M, Hoen D, Weiss K, Kelly DW, Adusumilli PS, et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021;12(1):729.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jomrich G, Paireder M, Kristo I, Baierl A, Ilhan-Mutlu A, Preusser M, et al. High systemic immune-inflammation index is an adverse prognostic factor for patients with gastroesophageal adenocarcinoma. Ann Surg. 2021;273(3):532–41.

    Article  PubMed  Google Scholar 

  15. Casadei Gardini A, Scarpi E, Valgiusti M, Monti M, Ruscelli S, Matteucci L, et al. Prognostic role of a new index (multi inflammatory index) in patients with metastatic colorectal cancer: results from the randomized ITACa trial. Ther Adv Med Oncol. 2020;12:1758835920958363.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Writing Group for the European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the Extended Group for EWGSOP2. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(1):16–31.

    Article  PubMed  Google Scholar 

  17. Chen LK, Woo J, Assantachai P, Auyeung TW, Chou MY, Iijima K, et al. Asian working group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J American Med Direct Assoc. 2020;21(3):300-7. e2.

    Article  Google Scholar 

  18. Chen X-Y, Li B, Ma BW, Zhang XZ, Chen WZ, Lu LS, et al. Sarcopenia is an effective prognostic indicator of postoperative outcomes in laparoscopic-assisted gastrectomy. Eur J Surg Oncol. 2019;45(6):1092–8.

    Article  PubMed  Google Scholar 

  19. Weng CS, Huang WC, Chang CL, Jan YT, Chen TC, Lee J. Association of malignant ascites with systemic inflammation and muscle loss after treatment in advanced-stage ovarian cancer. J Cachexia Sarcopenia Muscle. 2023;14(5):2114–25.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Lin JX, Lin JP, Xie JW, Wang JB, Lu J, Chen QY, et al. Prognostic value and association of sarcopenia and systemic inflammation for patients with gastric cancer following radical gastrectomy. Oncologist. 2019;24(11):e1091–101.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Borges TC, Gomes TL, Pichard C, Laviano A, Pimentel GD. High neutrophil to lymphocytes ratio is associated with sarcopenia risk in hospitalized cancer patients. Clin Nutr. 2021;40(1):202–6.

    Article  CAS  PubMed  Google Scholar 

  22. Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab. 2008;33(5):997–1006.

    Article  PubMed  Google Scholar 

  23. Zhuang CL, Zhang FM, Li W, Wang KH, Xu HX, Song CH, et al. Associations of low handgrip strength with cancer mortality: a multicentre observational study. J Cachexia Sarcopenia Muscle. 2020;11(6):1476–86.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zhang Q, Song MM, Zhang X, Ding JS, Ruan GT, Zhang XW, et al. Association of systemic inflammation with survival in patients with cancer cachexia: results from a multicentre cohort study. J Cachexia Sarcopenia Muscle. 2021;12(6):1466–76.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Rosenberg IH. Sarcopenia: origins and clinical relevance. J Nutr. 1997;127(5):990S-S991.

    Article  CAS  PubMed  Google Scholar 

  26. Nipp RD, Fuchs G, El-Jawahri A, Mario J, Troschel FM, Greer JA, et al. Sarcopenia is associated with quality of life and depression in patients with advanced cancer. Oncologist. 2018;23(1):97–104.

    Article  PubMed  Google Scholar 

  27. Okumura S, Kaido T, Hamaguchi Y, Kobayashi A, Shirai H, Fujimoto Y, et al. Impact of skeletal muscle mass, muscle quality, and visceral adiposity on outcomes following resection of intrahepatic cholangiocarcinoma. Ann Surg Oncol. 2017;24:1037–45.

    Article  PubMed  Google Scholar 

  28. van Rijn-Dekker MI, van den Bosch L, van den Hoek JG, Bijl HP, van Aken ES, van der Hoorn A, et al. Impact of sarcopenia on survival and late toxicity in head and neck cancer patients treated with radiotherapy. Radiother Oncol. 2020;147:103–10.

    Article  PubMed  Google Scholar 

  29. Detopoulou P, Voulgaridou G, Papadopoulou S. Cancer, phase angle and sarcopenia: the role of diet in connection with lung cancer prognosis. Lung. 2022;200(3):347–79.

    Article  CAS  PubMed  Google Scholar 

  30. Choi MH, Oh SN, Lee IK, Oh ST, Won DD. Sarcopenia is negatively associated with long-term outcomes in locally advanced rectal cancer. J Cachexia Sarcopenia Muscle. 2018;9(1):53–9.

    Article  PubMed  Google Scholar 

  31. Mallet R, Modzelewski R, Lequesne J, Mihailescu S, Decazes P, Auvray H, et al. Prognostic value of sarcopenia in patients treated by radiochemotherapy for locally advanced oesophageal cancer. Radiat Oncol. 2020;15(1):1–9.

    Article  Google Scholar 

  32. Qian J, Si Y, Zhou K, Tian Y, Guo Q, Zhao K, et al. Sarcopenia is associated with prognosis in patients with esophageal squamous cell cancer after radiotherapy or chemoradiotherapy. BMC Gastroenterol. 2022;22(1):211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhao H, Wu L, Yan G, Chen Y, Zhou M, Wu Y, et al. Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther. 2021;6(1):263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  PubMed  Google Scholar 

  35. Ng SP, Bahig H, Jethanandani A, Sturgis EM, Johnson FM, Elgohari B, et al. Prognostic significance of pre-treatment neutrophil-to-lymphocyte ratio (NLR) in patients with oropharyngeal cancer treated with radiotherapy. Br J Cancer. 2021;124(3):628–33.

    Article  CAS  PubMed  Google Scholar 

  36. Wolfe AR, Siedow M, Nalin A, DiCostanzo D, Miller ED, Diaz DA, et al. Increasing neutrophil-to-lymphocyte ratio following radiation is a poor prognostic factor and directly correlates with splenic radiation dose in pancreatic cancer. Radiother Oncol. 2021;158:207–14.

    Article  CAS  PubMed  Google Scholar 

  37. Lorente D, Mateo J, Templeton A, Zafeiriou Z, Bianchini D, Ferraldeschi R, et al. Baseline neutrophil–lymphocyte ratio (NLR) is associated with survival and response to treatment with second-line chemotherapy for advanced prostate cancer independent of baseline steroid use. Ann Oncol. 2015;26(4):750–5.

    Article  CAS  PubMed  Google Scholar 

  38. Wang Y, Chen J, Yang L, Li J, Wu W, Huang M, et al. Tumor-contacted neutrophils promote metastasis by a CD90-TIMP-1 juxtacrine–paracrine loop. Clin Cancer Res. 2019;25(6):1957–69.

    Article  CAS  PubMed  Google Scholar 

  39. Germann M, Zangger N, Sauvain MO, Sempoux C, Bowler AD, Wirapati P, et al. Neutrophils suppress tumor-infiltrating T cells in colon cancer via matrix metalloproteinase-mediated activation of TGF β. EMBO Mol Med. 2020;12(1):e10681.

    Article  CAS  PubMed  Google Scholar 

  40. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137–48.

    Article  CAS  PubMed  Google Scholar 

  41. Davis MP, Panikkar R. Sarcopenia associated with chemotherapy and targeted agents for cancer therapy. Ann Palliat Med. 2019;8(1):86–101.

    Article  PubMed  Google Scholar 

  42. Feliciano EMC, Kroenke CH, Meyerhardt JA, Prado CM, Bradshaw PT, Kwan ML, et al. Association of systemic inflammation and sarcopenia with survival in nonmetastatic colorectal cancer: results from the C SCANS study. JAMA Oncol. 2017;3(12):e172319.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Enago (www.enago.cn) for the manuscript review and editing support.

Funding

This work was supported by grants from the Project of Wu Jieping Medical Foundation (NO: 320.6750).

Author information

Authors and Affiliations

Authors

Contributions

LX, YL, JL and TL were responsible for conceptualizing and designing this study, data collection, data interpretation, and manuscript drafting. XN and XZ played a major role in body composition assessment and data analysis. HB and CL participated in acquisition of clinical records, data analysis, and revision of the manuscript. All authors read and approved the final version of manuscript.

Corresponding authors

Correspondence to Jiahua Lyu or Tao Li.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Medical Ethics Committee of Sichuan Cancer Hospital and was conducted in accordance with the principles of the Helsinki Declaration and its amendments. All participants provided written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiao, L., Liu, Y., Zhang, X. et al. Prognostic value of sarcopenia and inflammatory indices synergy in patients with esophageal squamous cell carcinoma undergoing chemoradiotherapy. BMC Cancer 24, 860 (2024). https://doi.org/10.1186/s12885-024-12602-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12602-1

Keywords