Home Combining the Albumin-to-fibrinogen ratio and pathologic factors predicts survival in surgically treated patients with esophageal squamous cell carcinoma
Article Open Access

Combining the Albumin-to-fibrinogen ratio and pathologic factors predicts survival in surgically treated patients with esophageal squamous cell carcinoma

  • Xueshun Zhang , Xinyu Li , Lei Xing and Ping Ren ORCID logo EMAIL logo
Published/Copyright: December 24, 2024

Abstract

Objectives

The primary objectives of this research were to examine the prognostic significance of the albumin-to-fibrinogen ratio (AFR) in patients who have undergone surgery for esophageal squamous cell carcinoma (ESCC) and to develop an easily implementable predictive model with clinical utility.

Methods

The present study retrospectively analyzed the clinical data of 414 patients who underwent R0 resection after being diagnosed with stage I–III ESCC. The prognostic value of AFR was evaluated using Kaplan-Meier survival curves and COX proportional risk regression modeling, and the effectiveness of AFR compared with other inflammatory markers was evaluated. Additionally, a nomogram prediction model was developed, and its accuracy was evaluated using the receiver operating characteristic (ROC) curves, decision curve analysis (DCA), and calibration curves.

Results

AFR was significantly correlated with tumor length, T-stage, N-stage, pathological stage, and vascular infiltration (p<0.05 for all). The multivariate analysis results demonstrated that AFR was an independent prognostic factor that affected patient outcomes, whereas other inflammatory and nutritional biomarkers did not. Furthermore, the overall C-index of the nomogram risk prediction model was 0.737 (95 %-CI: 0.700–0.776). The calibration curves showed that the 3- and 5-year overall survival (OS) probabilities predicted by the nomogram were consistent with actual observations. Moreover, the DCA and ROC curves showed that our model had better clinical utility.

Conclusions

Preoperative AFR, a clinical indicator based on inflammation and nutrition, plays a clear role in the predictions of patient prognosis. The prognostic prediction model incorporating pathological factors and AFR demonstrates simplicity, efficacy, and exceptional accuracy.

Introduction

Esophageal cancer (EC) represents a malignant neoplasm characterized by elevated morbidity and mortality rates. According to GLOBOCAN 2022, EC is the eleventh most commonly diagnosed cancer and the seventh leading cause of cancer death worldwide [1]. Epidemiological data reveal that the histological subtypes of esophageal cancer differ significantly, with ESCC being the most prevalent form in Asia [2]. The prognosis for EC remains unfavorable on a global scale, primarily due to the disease often being diagnosed at an advanced stage, which frequently leads to severe malnutrition resulting from inadequate nutritional intake [3].

In cancer patients, the tumor-lymph node-metastasis (TNM) staging system and histopathological grading are commonly used to assess prognosis and guide antitumor therapy. However, the TNM system primarily emphasizes the pathologic features of the lesion and ignores the patient’s systemic condition, which may lead to inaccuracies in assessing their long-term survival [4]. Some scholars have suggested that considering additional clinical features may improve the accuracy and validity of prognosis in certain cancer types [5], 6]. Therefore, it is necessary to incorporate additional clinical factors when developing a prognostic prediction model for patients undergoing surgery for ESCC.

Inflammation and nutrition are pivotal factors influencing tumor prognosis, significantly impacting tumor progression, angiogenesis, metastasis, and overall prognostic outcomes [7], 8]. Hematological components, such as relevant immune cells and acute temporal reactive proteins, are biomarkers that reflect systemic inflammation and nutrition, and their levels can indicate the degree of chronic inflammation and malnutrition. Additionally, specific nutritional and inflammatory markers have been associated with clinical regression in esophageal cancer, highlighting their potential relevance in patient management. These include the lymphocyte-to-monocyte ratio (LMR), platelet-to-lymphocyte ratio (PLR), neutrophil-to-lymphocyte ratio (NLR), prognostic nutritional index (PNI), and systemic immune-inflammation index (SII) [9], [10], [11].

The peripheral blood AFR is an emerging clinical biomarker that integrates serum albumin and fibrinogen levels, thereby providing a comprehensive reflection of the patient’s nutritional, coagulation, and inflammatory status [12]. Consequently, AFR has been utilized as a prognostic marker for individuals with hepatocellular carcinoma and gastric cancer [13], 14]. Nevertheless, there is a lack of extensive studies focusing on the role of preoperative AFR in predicting long-term survival for patients diagnosed with ESCC, especially when compared to other inflammatory biomarkers.

Therefore, in this study, we retrospectively investigated AFR levels in patients with ESCC before undergoing esophagectomy and evaluated its relationship with clinicopathologic factors and prognosis. In addition, we compared AFR with other nutritional and inflammatory markers and constructed survival prediction models.

Materials and methods

Patients

The research comprised 414 individuals diagnosed with stage I–III ESCC who received surgical treatment at the First Hospital of Jilin University between January 2014 and December 2022. The inclusion criteria were as follows: (1) confirmed diagnosis of ESCC through endoscopy and pathology, (2) not receiving any neoadjuvant oncological treatment before surgery, (3) no prior history of malignancies, no acute or chronic infections such as bacterial, fungal, or tuberculosis, and no hematological system diseases or rheumatoid immunity-related diseases, (4) surgery was performed to achieve R0 resection, and (5) availability of complete perioperative and follow-up data. Patients who died from factors unrelated to esophageal cancer, patients who had a combination of other primary cancers, and patients who had received neoadjuvant antitumor therapy were excluded. All participants underwent radical esophagectomy along with systematic lymph node dissection, and staging was conducted in accordance with the 8th edition of the AJCC TNM staging system [15] for esophageal cancer. The research protocol was approved by the Research Ethics Committee of the First Hospital of Jilin University (No. 2024-1157). As this was a retrospective study, informed consent was exempted.

Data collection

The clinicopathological data included sex, age, body mass index (BMI), tumor site, tumor size, and TNM stage. Blood was collected before breakfast 2 weeks before surgery. Hematological tests included neutrophils, lymphocytes, monocytes, platelet count, globulin, albumin, and fibrinogen. The AFR is defined as the ratio of albumin content to fibrin content, while the LMR is the lymphocyte count divided by the monocyte count. The PLR represents the platelet count over the lymphocyte count, and the NLR is calculated by dividing the neutrophil count by the lymphocyte count. The SII is obtained by multiplying the platelet count by the neutrophil count and then dividing by the lymphocyte count. Additionally, the albumin-to-globulin ratio (AGR) is the albumin level compared to the globulin level, and the PNI is calculated as the albumin concentration in grams per liter plus five times the lymphocyte count expressed in 109 per liter.

Follow-up

Follow-up assessments were performed from the time of surgery until January 2023 or until the patient’s death. Overall survival was defined as the interval from the date of surgery to either death attributable to esophageal cancer or the last follow-up visit. The initial follow-up occurred 1-month post-surgery, with subsequent evaluations scheduled every 3 months during the first year. In the second year, follow-ups were conducted biannually, and after 5 years, annual assessments were implemented.

Statistical analysis

Data analysis for this study was conducted using SPSS 26.0 and R 3.6.1 software RMS package. Medians and quartiles were used to represent the measurement information. The count data was expressed as a percentage. To determine the optimal cut-off points for the AFR and other biomarkers within the preoperative patient group, X-tile 3.6 software was utilized. Utilizing the identified optimal cut-off values, patients were divided into two distinct categories: those with elevated AFR levels and those with decreased AFR levels. Following this classification, the relationship between AFR and various clinical characteristics was analyzed employing either the chi-square test or Fisher’s exact test.

The Kaplan-Meier method was used to create survival curves, and log-rank tests were performed to assess the differences between these curves. Additionally, both univariate and multivariate analyses were carried out on clinical variables using the Cox proportional hazards regression approach. The hazard ratio (HR) and its corresponding 95 % confidence interval (CI) were computed to evaluate the associations between prognostic factors and survival duration. Based on the independent risk factors identified in the multivariate analysis, a nomogram prediction model was developed using the R package RMS to estimate the probability of OS at 3 and 5 years for patients undergoing surgical intervention for ESCC. Furthermore, the model’s discrimination and accuracy were assessed through the generation of the concordance index (C-index), receiver operating characteristic (ROC) curves, decision curve analysis (DCA), and calibration curves, all performed using R 3.6.1. p<0.05 was considered statistically significant.

Results

Patient and tumor characteristics

This study included a total of 414 patients diagnosed with stage I–III ESCC, with their clinical characteristics detailed in Table 1. Among the participants, 184 patients (44.4 %) were under the age of 60, while 230 patients (55.6 %) were 60 years or older. The cohort comprised 22 female patients (5.3 %) and 392 male patients (94.7 %). Additionally, 292 patients (70.5 %) reported a history of smoking, and 311 patients (75.1 %) indicated alcohol consumption. Notably, 65 patients (15.7 %) had a prior diagnosis of hypertension, and 26 patients (6.3 %) had a history of diabetes mellitus. The proportions of patients with tumor length <3 cm and ≥3 cm were 39.6 and 60.4 %, respectively. The majority of tumors were located in the lower esophagus (64.7 %), followed by the middle esophagus (29.2 %). Pathologic differentiation showed that the proportions of poor, moderate, and well-differentiated were 21.3, 74.2, and 4.5 %, respectively. Pathological TNM staging revealed that 17.1 % of patients (71 individuals) were classified as stage I, 26.1 % (108 individuals) as stage II, and the majority, 56.8 % (235 individuals), as stage III. In T staging, 92 patients (22.2 %) were in the T1 stage, 65 patients (15.7 %) were in the T2 stage, and 257 patients (62.1 %) were in the T3 stage. In N staging, 163 patients (39.4 %) were in the stage N0, 151 patients (36.5 %) were in the stage N1, and 100 patients (24.1 %) were in the stage N2. There were 220 (53.1 %) and 166 (40.1 %) patients with lymphangio vascular invasion and perineural invasion, respectively. Follow-up data revealed that 223 patients (53.9 %) underwent adjuvant therapy after surgery.

Table 1:

Clinical characteristics in 414 patients with ESCC.

Categories Patients, n (%) Categories Patients, n (%)
Sex TNM stage
Male 392 (94.7) 71 (17.1)
Female 22 (5.3) 108 (26.1)
Age (years) 235 (56.8)
<60 184 (44.4) Differentiation
≥60 230 (55.6) Poor 88 (21.3)
Smoking Moderate 307 (74.2)
No 122 (29.5) Well 19 (4.5)
Yes 292 (70.5) T stage
Alcohol T1 92 (22.2)
No 103 (24.9) T2 65 (15.7)
Yes 311 (75.1) T3 257 (62.1)
Hypertension N stage
No 349 (84.3) N0 163 (39.4)
Yes 65 (15.7) N1 151 (36.5)
Diabetes N2 100 (24.1)
No 388 (93.7) Perineural invasion
Yes 26 (6.3) No 248 (59.9)
Tumor length, cm Yes 166 (40.1)
<3 164 (39.6) Lymphangio vascular invasion
≥3 250 (60.4) No 194 (46.9)
Tumor location Yes 220 (53.1)
Upper 25 (6.1) Adjuvant treatment
Middle 121 (29.2) No 191 (46.1)
Lower 268 (64.7) Yes 223 (53.9)

X-tile determines cut-off values for clinical indicators

As shown in Figure 1, X-tile software was used to determine the optimal thresholds for preoperative nutritional and inflammatory biomarkers for all patients; the optimal cut-off value for AFR was 9.0. According to Supplementary Figure S1, the optimal cut-off values for NLR, PLR, LMR, SII, PNI, albumin, and AGR were 2.0, 123.3, 4.8, 826.6, 46.3, 39.4 and 1.4, respectively.

Figure 1: 
The cutoff values of preoperative AFR in 414 patients with ESCC using X-tile software. (A) Tile plots graphically represent the data, with different values grouped as cutoffs for statistical tests. The data with the smallest p-value can be considered the best cutoffs, manifested as the brightest pixel dots on the tiled plot. (B) The histogram shows the distribution of the number of patients on either side of the cutoff value.
Figure 1:

The cutoff values of preoperative AFR in 414 patients with ESCC using X-tile software. (A) Tile plots graphically represent the data, with different values grouped as cutoffs for statistical tests. The data with the smallest p-value can be considered the best cutoffs, manifested as the brightest pixel dots on the tiled plot. (B) The histogram shows the distribution of the number of patients on either side of the cutoff value.

Correlation analysis between AFR and clinicopathologic characteristics

In this study, AFR was categorized into two groups based on the optimal cut-off value: the low-level group (AFR≤9.0) and the high-level group (AFR>9.0). This classification facilitated an examination of the relationship between AFR and various clinicopathological characteristics, as illustrated in Table 2. A low AFR demonstrated a significant association with tumor length, T-stage, N-stage, pathologic stage, and choroidal infiltration (all p<0.05). In addition, low AFR was significantly associated with anastomotic leakage resulting from surgical treatment (p<0.001). However, AFR was not correlated with sex, age, smoking, alcohol consumption, tumor location, degree of differentiation, and perineural invasion (all p>0.05).

Table 2:

Associations between AFR and clinicopathological characteristics in ESCC patients.

Categories AFR χ 2 p-Value
≤9.0 >9.0
Sex 0.224a
Male 61 331
Female 1 21
Age (years) 3.302 0.069
<60 21 163
≥60 41 189
Smoking 0.007 0.935
No 18 104
Yes 44 248
Alcohol 0.034 0.855
No 16 87
Yes 46 265
Tumor length, cm 16.812 <0.001
<3 10 154
≥3 52 198
Differentiation 4.836 0.089
Poor 19 69
Moderate 42 265
Well 1 18
Tumor location 0.847 0.655
Upper 4 21
Middle 21 100
Lower 37 231
TNM stage 10.977 0.004
7 64
8 100
47 188
T stage
T1 7 85 12.678 0.002
T2 4 61
T3 51 206
N stage 7.048 0.029
N0 15 148
N1 28 123
N2 19 81
Perineural invasion 2.978 0.084
No 31 217
Yes 31 135
Lymphangio vascular invasion 6.244 0.012
No 20 174
Yes 42 178
Adjuvant treatment 4.414 0.039
No 21 170
Yes 41 182
Anastomotic leakage 18.858 <0.001
No 44 319
Yes 18 33
  1. aFisher’s exact probability method.

Survival analysis

The overall median follow-up duration for the entire cohort was 56 months, with a range spanning from 2 to 118 months. The median survival duration was recorded at 42 months (95 %-CI: 31.85–52.15). By the end of the follow-up timeline, 224 individuals, accounting for 54.1 % of the cohort, had succumbed to factors related to EC. The observed OS rates were 84.1 % at 1 year, 54.6 % at the 3-year mark, and 43.3 % at 5 years.

Our results showed a more significant trend of decreasing survival curves in patients with low-level AFR. Moreover, the 5-year OS rate was markedly higher in patients exhibiting elevated levels of the AFR compared to those with low AFR levels (49.7 % vs. 10.1 %, p<0.001). Meanwhile, high-level SII, high-level PLR, high-level NLR, and low-level LMR showed poorer OS (all p<0.005), but there was no statistical significance in OS for different levels of albumin, AGR, and PNI (all p>0.05). The survival curves are depicted in Figure 2.

Figure 2: 
Cumulative 5-year survival curves for ESCC patients. (A) AFR, (B) LMR, (C) NLR, (D) PLR, (E) SII, (F) PNI, (G) AGR, and (H) albumin. AFR, Albumin-to-fibrinogen ratio; LMR, lymphocyte-to-monocyte ratio; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; SII, systemic immune-inflammation index; PNI, prognostic nutritional index; AGR, albumin-to-globulin ratio.
Figure 2:

Cumulative 5-year survival curves for ESCC patients. (A) AFR, (B) LMR, (C) NLR, (D) PLR, (E) SII, (F) PNI, (G) AGR, and (H) albumin. AFR, Albumin-to-fibrinogen ratio; LMR, lymphocyte-to-monocyte ratio; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; SII, systemic immune-inflammation index; PNI, prognostic nutritional index; AGR, albumin-to-globulin ratio.

As shown in Table 3, univariate analysis revealed that gender, smoking history, hypertension, tumor length, lymphangio vascular invasion, perineural invasion, pathological stage, T stage, N stage, degree of differentiation, NLR, PLR, SII, LMR, and AFR significantly affected the prognosis of patients who had undergone surgery for ESCC (all p<0.05). Conversely, factors such as age, alcohol consumption, BMI, cardiovascular disease, diabetes mellitus, tumor location, albumin levels, AGR, and PNI did not demonstrate any significant association with patient prognosis. The multivariate analysis identified independent risk factors influencing patient prognosis, including the AFR, lymphangio vascular invasion, pathological stage, and tumor differentiation degree (all p<0.05).

Table 3:

Univariate and multivariate analysis for OS in 414 patients with ESCC.

Categories Univariate analysis Multivariate analysis
HR (95 % CI) p-Value HR (95 % CI) p-Value
Sex
Male 1 1
Female 0.400 (0.178–0.901) 0.027 0.517 (0.221–1.210) 0.128
Age (years)
<60 1
≥60 1.005 (0.773–1.308) 0.968
Smoking
No 1
Yes 1.172 (0.851–1.614) 0.330
Alcohol
No 1 1
Yes 1.371 (1.007–1.866) 0.045 0.962 (0.691–1.340) 0.821
Adjuvant treatment
No 1
Yes 1.250 (0.957–1.633) 0.102
Hypertension
No 1 1
Yes 1.533 (1.098–2.139) 0.012 1.387 (0.985–1.952) 0.061
Diabetes
No 1
Yes 1.011 (0.588–1.737) 0.969
Cardiovascular disease
No 1
Yes 0.742 (0.330–1.672) 0.472
BMI
Underweight 1
Normal weight 0.634 (0.391–1.028) 0.064
Overweight 0.738 (0.441–1.234) 0.247
Obesity 0.544 (0.238–1.243) 0.149
Tumor length, cm
<3 1 1
≥3 1.716 (1.295–2.275) <0.001 1.306 (0.962–1.774) 0.087
Tumor location
Upper 1
Middle 1.660 (0.877–3.143) 0.120
Lower 1.368 (0.741–2.527) 0.316
Albumin
≤39.4 1
>39.4 1.225 (0.941–1.594) 0.131
NLR
≤2.0 1 1
>2.0 1.583 (1.216–2.062) <0.001 1.039 (0.752–1.436) 0.816
PLR
≤123.3 1 1
>123.3 1.458 (1.121–1.898) 0.005 1.199 (0.877–1.638) 0.225
AFR
≤9.0 1 1
>9.0 2.748 (2.021–3.737) <0.001 1.734 (1.194–2.518) 0.004
SII
≤826.6 1 1
>826.6 2.132 (1.523–2.985) <0.001 0.918 (0.594–1.417) 0.699
PNI
≤46.3 1
>46.3 1.205 (0.917–1.584) 0.182
AGR
≤1.4 1
>1.4 1.202 (0.923–1.564) 0.172
LMR
≤4.8 1 1
>4.8 1.408 (1.070–1.851) 0.014 0.916 (0.668–1.257) 0.588
TNM stage
1 1
3.350 (1.739–6.451) <0.001 2.419 (1.221–4.791) 0.011
6.022 (3.270–11.089) <0.001 2.787 (1.400–5.546) 0.004
Differentiation
Poor 1 1
Moderate 0.340 (0.257–0.449) <0.001 0.441 (0.328–0.594) <0.001
Well 0.143 (0.058–0.355) <0.001 0.229 (0.091–0.573) 0.002
Lymphangio vascular invasion
No 1 1
Yes 2.551 (1.929–3.373) <0.001 1.564 (1.124–2.175) 0.008
Perineural invasion
No 1 1
Yes 1.860 (1.431–2.418) <0.001 1.142 (0.860–1.516) 0.359
  1. HR, hazard ratio; CI, confidence interval; NLR, neutrophil-to-lymphocyte ratio; SII, systemic immune-inflammatory index; PLR, platelet-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio; PNI, prognostic nutritional index; AGR, albumin-to-globulin ratio.

Subgroup analysis

The patients were categorized into two groups based on tumor stage, as illustrated in Figure 3. For patients in stages I and II, the survival curves revealed that those with a low AFR had a significantly lower 3-year OS rate compared to those in the high AFR group (20.0 % vs. 72.0 %, p<0.001). Similarly, among stage III patients, the survival curves indicated that the low AFR group also exhibited a significantly diminished 3-year OS rate relative to the high AFR group (29.2 % vs. 47.9 %, p<0.001).

Figure 3: 
Survival curves for subgroup analysis of ESCC patients. AFR survival curves in patients with (A) stage I + II and (B) stage III ESCC.
Figure 3:

Survival curves for subgroup analysis of ESCC patients. AFR survival curves in patients with (A) stage I + II and (B) stage III ESCC.

A nomogram predicts survival risk based on AFR

The independent risk factors derived from the multifactorial COX-based analysis were incorporated into the nomogram. This model allows for the assessment of the impact of each independent factor on patient survival at 1, 3, and 5 years after surgery, as depicted in Figure 4. Among them, the C-index of the model was 0.737 (95 %-CI: 0.700–0.776).

Figure 4: 
Nomogram predicts survival risk based on AFR and pathological factors in patients with ESCC.
Figure 4:

Nomogram predicts survival risk based on AFR and pathological factors in patients with ESCC.

The constructed model underwent validation. The internally validated calibration curves indicated that the 3- and 5-year OS probabilities predicted by the nomogram closely aligned with actual observations, as illustrated in Figure 5A and B. The DCA revealed that the model’s net gain in predicting 5-year OS was superior to that of other individual prediction models, including TNM staging and histological differentiation. The time-dependent ROC curve also showed that the nomogram incorporating AFR outperformed the models constructed from pathological factors alone in predicting 5-year OS in patients with ESCC, as shown in Figure 5C and D.

Figure 5: 
Validation of nomogram accuracy and effectiveness. (A) 3-Year OS correction curve, (B) 5-year OS correction curve, (C) the decision curve analysis, (D) the time-dependent receiver operating characteristic curve. LVI, lymphangio vascular invasion.
Figure 5:

Validation of nomogram accuracy and effectiveness. (A) 3-Year OS correction curve, (B) 5-year OS correction curve, (C) the decision curve analysis, (D) the time-dependent receiver operating characteristic curve. LVI, lymphangio vascular invasion.

Discussion

Despite significant advancements in comprehensive treatment strategies that emphasize radical surgical intervention, the prognosis for patients diagnosed with esophageal cancer continues to be unfavorable [16]. Researchers widely accept that pathological staging and histologic grading can be used to assess the prognosis of patients undergoing surgery for esophageal cancer, but their predictive value remains limited. Increasingly more studies have shown that serum corresponding protein levels, inflammatory cell counts, and many binding indicators can reflect the prognosis of patients with malignant tumors [17]; for example, plasma fibrinogen, serum albumin, and lymphocyte-monocyte ratios have been suggested to be associated with patient prognosis [18], [19], [20].

AFR consists of two parameters, serum albumin and fibrinogen. The production of serum albumin occurs in hepatocytes, and the concentration of albumin in the blood serves as an indicator of the body’s nutritional condition [21]. A variety of studies have indicated that patients with malignant tumors who present low serum albumin levels tend to have a worse prognosis [22], 23]. The other computational element of AFR is fibrinogen, which plays an indispensable role in tumor formation and development [24]. A prospective trial investigating non-small cell lung cancer indicated that AFR may serve as a promising biomarker for predicting both the clinical efficacy and outcomes of surgical resection and radiotherapy [25].

The results of this study indicate that low levels of AFR are strongly associated with many aggressive tumor characteristics, including greater tumor length, deeper tumor invasion, positive lymph node metastasis, lymphangio vascular invasion, and advanced TNM staging. A study of 88 patients with esophageal small cell carcinoma by Wang et al. [26] demonstrated that low levels of AFR led to a more pronounced propensity for tumor malignancy. Moreover, low levels of AFR are closely associated with surgical complications, as patients with esophageal cancer exhibiting low AFR have a higher likelihood of developing anastomotic leakage postoperatively. The study by You et al. [27] also demonstrated that preoperative AFR levels are a valid predictor of serious complications in patients undergoing radical laparoscopic gastrectomy. In addition, the survival analysis results indicated that patients with low AFR levels experienced shorter OS and a poorer prognosis, aligning with findings from previously published studies [28].

In clinical practice, numerous hematological indices are used to assess the prognosis of tumor patients, and these indices have different predictive values [29]. In this study, several nutritional and inflammatory factors were included. The univariate analysis identified several factors significantly associated with OS, including SII, PLR, NLR, and LMR. The survival curves illustrated that elevated levels of SII, PLR, and NLR, along with decreased levels of LMR, were correlated with poorer OS. However, only AFR was established as an independent prognostic factor in the multivariate analyses. This suggests that AFR outperforms other nutritional and inflammatory markers in influencing and predicting the prognosis of ESCC patients. Our conclusions are consistent with the findings of Zhang et al. [28], but they only compared the prognostic role of AFR vs. AGR in ESCC patients.

We constructed a nomogram prediction model based on clinical indicators such as AFR and pathological stage, which can intuitively reflect the different effects of pathological factors and inflammatory indicators on patient prognosis. Firstly, in our model, patients with low levels of AFR, low-grade differentiation, lymphangio vascular invasion, and later pathological staging had lower 3- and 5-year survival rates. Secondly, our nomogram exhibited higher accuracy in predicting the survival of patients with ESCC compared to the model based solely on pathological factors. Finally, our predictive model identifies at-risk populations, thus allowing clinicians to focus on their nutritional status earlier and enhance nutritional support. According to our prediction model, if patients have low AFR levels, even though their pathological staging is early, they need to be followed up rigorously and further postoperative intensive therapy may be considered.

Cancer development and progression involve not only the tumor cells themselves but also the tumor microenvironment. Inflammation is a common feature of tumor microenvironment abnormalities, and a sustained inflammatory response may lead to normal cell damage and cancer progression [30]. As an inflammatory marker, there are a few possible mechanisms by which AFR can affect tumor development. Serum albumin is essential for the maintenance of important physiological functions in the body, including binding, transportation, and maintenance of plasma osmolality. At the same time, serum albumin removes reactive oxygen species and has an antioxidant effect [31], while reactive oxygen species play a crucial role in tumorigenesis. In addition, decreased serum albumin levels may impair the body’s immune system and make it less resistant to tumors [32]. When inflammation occurs, the migration and invasion of tumor cells into normal tissues result in microhemorrhages, and the coagulation cascade reaction of fibrinogen helps platelets adhere to tumor cells to form microthrombi. This platelet clot forms at the interface between tumor and host cells and is involved in protecting cancer cells from natural killer cell toxicity and enhancing tumor cells’ ability to bind and traverse the endothelial barrier to establish metastases [33], 34]. Platelet-fibrin thrombi also shield intravascular tumor cells from immune detection by host inflammatory and natural killer (NK) cells, enveloping the tumor cells in a platelet-fibrin matrix and presenting human leukocyte antigen (HLA) class I molecules [35].

This study confirms that AFR affects the prognosis of patients with ESCC. Nonetheless, there are several limitations that require attention. First, this was a single-center, retrospective study that included only 414 eligible patients, a relatively small sample size, and because of resource constraints, we were unable to perform external validation of data from other centers, which may have led to unstable results. Second, attention needs to be paid to how optimal thresholds are determined for the clinical use of AFR and other inflammatory and nutritional markers, as different studies use different methods to determine optimal thresholds. Finally, this study focused on the general condition of patients, preoperative nutritional and inflammatory indicators, and tumor-related pathology; it did not thoroughly investigate the impact of other external factors on the long-term survival of esophageal cancer patients. Therefore, future studies with multicenter and large patient cohorts need to be conducted.

Conclusions

Preoperative AFR is an inflammation- and nutrition-based clinical indicator that can influence the prognosis of patients with ESCC. It is also more sensitive than other nutritional and inflammatory indicators. Our findings suggest that patients exhibiting low preoperative AFR levels experience reduced long-term survival compared to those with elevated AFR levels. A prognostic prediction model that incorporates pathologic factors and AFR has a high level of accuracy and usability.


Corresponding author: Ping Ren, Department of Thoracic Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, China, E-mail:

Funding source: the Natural Science Foundation of Jilin Province

Award Identifier / Grant number: No. 20210101263JC

Acknowledgments

We sincerely thank all the patients for participating in this study.

  1. Research ethics: The local Institutional Review Board deemed the study exempt from review.

  2. Informed consent: Informed consent was obtained from all individuals included in this study.

  3. Author contributions: Study design and technical support: PR. Quality control of data: PR, XZ. Data collection: XZ. Data processing and analysis: XZ, XL. Manuscript writing: XZ, XL, LX. Manuscript review and revision: all authors.

  4. Use of Large Language Models, AI and Machine Learning Tools: During the preparation of this work, we used ChatGPT solely for the purpose of language editing and proofreading of the manuscript. We confirm that the use of ChatGPT did not involve the generation of original research content, hypotheses, or data analysis. All scientific content, findings, and conclusions presented in this manuscript are our own, and ChatGPT was used solely to refine the language. After using this tool, we reviewed and edited the content as needed and took full responsibility for the content of the publication.

  5. Conflict of interests: Authors state no conflict of interest.

  6. Research funding: This study was supported by the Natural Science Foundation of Jilin Province (No. 20210101263JC).

  7. Data availability: The data supporting the conclusions of this study can be obtained upon request from the corresponding author.

References

1. Bray, F, Laversanne, M, Sung, H, Ferlay, J, Siegel, RL, Soerjomataram, I, et al.. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229–63. https://doi.org/10.3322/caac.21834.Search in Google Scholar PubMed

2. Santucci, C, Mignozzi, S, Malvezzi, M, Collatuzzo, G, Levi, F, La Vecchia, C, et al.. Global trends in esophageal cancer mortality with predictions to 2025, and in incidence by histotype. Cancer Epidemiol 2023;87:102486. https://doi.org/10.1016/j.canep.2023.102486.Search in Google Scholar PubMed

3. D’Journo, XB, Thomas, PA. Current management of esophageal cancer. J Thorac Dis 2014;6:S253–64. https://doi.org/10.3978/j.issn.2072-1439.2014.04.16.Search in Google Scholar PubMed PubMed Central

4. Yang, Y, He, C. Construction and validation of a nomogram model to predict the overall survival rate of esophageal cancer patients receiving neoadjuvant chemotherapy: a population-based study. Front Surg 2022;9:1066092. https://doi.org/10.3389/fsurg.2022.1066092.Search in Google Scholar PubMed PubMed Central

5. Zhuang, Z, Li, Y, Hong, Y, Chen, L, Qian, J, Lin, J, et al.. A novel prognostic score based on systemic inflammatory biomarkers for patients with oral squamous cell carcinoma. Oral Dis 2022;28:631–8. https://doi.org/10.1111/odi.13774.Search in Google Scholar PubMed

6. Lai, C, Zhang, C, Lv, H, Huang, H, Ke, X, Zhou, C, et al.. A novel prognostic model predicts overall survival in patients with nasopharyngeal carcinoma based on clinical features and blood biomarkers. Cancer Med 2021;10:3511–23. https://doi.org/10.1002/cam4.3839.Search in Google Scholar PubMed PubMed Central

7. McMillan, DC. The systemic inflammation-based glasgow prognostic score: a decade of experience in patients with cancer. Cancer Treat Rev 2013;39:534–40. https://doi.org/10.1016/j.ctrv.2012.08.003.Search in Google Scholar PubMed

8. Alwarawrah, Y, Kiernan, K, MacIver, NJ. Changes in nutritional status impact immune cell metabolism and function. Front Immunol 2018;9:1055. https://doi.org/10.3389/fimmu.2018.01055.Search in Google Scholar PubMed PubMed Central

9. McLaren, PJ, Bronson, NW, Hart, KD, Vaccaro, GM, Gatter, KM, Thomas, CRJr., et al.. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios can predict treatment response to neoadjuvant therapy in esophageal cancer. J Gastrointest Surg 2017;21:607–13. https://doi.org/10.1007/s11605-016-3351-4.Search in Google Scholar PubMed

10. Zhao, K, Wang, C, Shi, F, Li, M, Yu, J. Lymphocyte-monocyte ratio as a predictive marker for pathological complete response to neoadjuvant therapy in esophageal squamous cell carcinoma. Transl Cancer Res 2020;9:3842–53. https://doi.org/10.21037/tcr-19-2849.Search in Google Scholar PubMed PubMed Central

11. Zhang, H, Shang, X, Ren, P, Gong, L, Ahmed, A, Ma, Z, et al.. The predictive value of a preoperative systemic immune-inflammation index and prognostic nutritional index in patients with esophageal squamous cell carcinoma. J Cell Physiol 2019;234:1794–802. https://doi.org/10.1002/jcp.27052.Search in Google Scholar PubMed

12. Li, S, Zhang, D, Zeng, S, Wu, T, Wang, Y, Zhang, H, et al.. Prognostic value of preoperative albumin-to-fibrinogen ratio in patients with bladder cancer. J Cancer 2021;12:5864–73. https://doi.org/10.7150/jca.61068.Search in Google Scholar PubMed PubMed Central

13. Yan, H, Chen, S, Qiong, Y, Cai, L. Preoperative prealbumin-to-fibrinogen ratio to predict survival outcomes in hepatocellular carcinoma patients after hepatic resection. J Med Biochem 2022;41:290–8. https://doi.org/10.5937/jomb0-32980.Search in Google Scholar PubMed PubMed Central

14. Zhang, J, Li, SQ, Liao, ZH, Jiang, YH, Chen, QG, Huang, B, et al.. Prognostic value of a novel FPR biomarker in patients with surgical stage II and III gastric cancer. Oncotarget 2017;8:75195–205. https://doi.org/10.18632/oncotarget.20661.Search in Google Scholar PubMed PubMed Central

15. Amin, MB, Edge, SR, Greene, FL, Byrd, DR, Brookland, RK, Washington, MK, editors, et al.. AJCC cancer staging manual, 8th ed New York: Springer; 2017.Search in Google Scholar

16. He, M, Qi, Z, Qiu, R, Hu, Y, Li, J, Li, Y, et al.. Correlates of long-term survival of patients with pN+ esophageal squamous cell carcinoma after esophagectomy. J Oncol 2021;2021:6675691–10. https://doi.org/10.1155/2021/6675691.Search in Google Scholar PubMed PubMed Central

17. Rinninella, E, Cintoni, M, Raoul, P, Pozzo, C, Strippoli, A, Bria, E, et al.. Effects of nutritional interventions on nutritional status in patients with gastric cancer: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN 2020;38:28–42. https://doi.org/10.1016/j.clnesp.2020.05.007.Search in Google Scholar PubMed

18. Suzuki, T, Shimada, H, Nanami, T, Oshima, Y, Yajima, S, Washizawa, N, et al.. Prognostic significance of hyperfibrinogenemia in patients with esophageal squamous cell carcinoma. Int J Clin Oncol 2017;22:461–8. https://doi.org/10.1007/s10147-016-1087-5.Search in Google Scholar PubMed

19. Wei, J, Jin, M, Shao, Y, Ning, Z, Huang, J. High preoperative serum prealbumin predicts long-term survival in resected esophageal squamous cell cancer. Cancer Manag Res 2019;11:7997–8003. https://doi.org/10.2147/cmar.s214037.Search in Google Scholar PubMed PubMed Central

20. Xu, X, Jing, J. Inflammation-related parameter serve as prognostic biomarker in esophageal squamous cell carcinoma. Front Oncol 2022;12:900305. https://doi.org/10.3389/fonc.2022.900305.Search in Google Scholar PubMed PubMed Central

21. Soeters, PB, Wolfe, RR, Shenkin, A. Hypoalbuminemia: pathogenesis and clinical significance. J Parenter Enteral Nutr 2019;43:181–93. https://doi.org/10.1002/jpen.1451.Search in Google Scholar PubMed PubMed Central

22. Wu, N, Chen, G, Hu, H, Pang, L, Chen, Z. Low pretherapeutic serum albumin as a risk factor for poor outcome in esophageal squamous cell carcinomas. Nutr Cancer 2015;67:481–5. https://doi.org/10.1080/01635581.2015.1004726.Search in Google Scholar PubMed

23. Goh, SL, De Silva, RP, Dhital, K, Gett, RM. Is low serum albumin associated with postoperative complications in patients undergoing oesophagectomy for oesophageal malignancies? Interact Cardiovasc Thorac Surg 2015;20:107–13. https://doi.org/10.1093/icvts/ivu324.Search in Google Scholar PubMed

24. Mosesson, MW. Fibrinogen and fibrin structure and functions. J Thromb Haemostasis 2005;3:1894–904. https://doi.org/10.1111/j.1538-7836.2005.01365.x.Search in Google Scholar PubMed

25. Li, SQ, Jiang, YH, Lin, J, Zhang, J, Sun, F, Gao, QF, et al.. Albumin-to-fibrinogen ratio as a promising biomarker to predict clinical outcome of non-small cell lung cancer individuals. Cancer Med 2018;7:1221–31. https://doi.org/10.1002/cam4.1428.Search in Google Scholar PubMed PubMed Central

26. Wang, Y, Li, J, Chang, S, Zhou, K, Che, G. Low albumin to fibrinogen ratio predicts poor overall survival in esophageal small cell carcinoma patients: a retrospective study. Cancer Manag Res 2020;12:2675–83. https://doi.org/10.2147/cmar.s250293.Search in Google Scholar PubMed PubMed Central

27. You, X, Zhou, Q, Song, J, Gan, L, Chen, J, Shen, H. Preoperative albumin-to-fibrinogen ratio predicts severe postoperative complications in elderly gastric cancer subjects after radical laparoscopic gastrectomy. BMC Cancer 2019;19:931. https://doi.org/10.1186/s12885-019-6143-x.Search in Google Scholar PubMed PubMed Central

28. Zhang, H, Ren, P, Ma, M, Zhu, X, Zhu, K, Xiao, W, et al.. Prognostic significance of the preoperative albumin/fibrinogen ratio in patients with esophageal squamous cell carcinoma after surgical resection. J Cancer 2021;12:5025–34. https://doi.org/10.7150/jca.58022.Search in Google Scholar PubMed PubMed Central

29. Jiang, Y, Xu, D, Song, H, Qiu, B, Tian, D, Li, Z, et al.. Inflammation and nutrition-based biomarkers in the prognosis of oesophageal cancer: a systematic review and meta-analysis. BMJ Open 2021;11:e048324. https://doi.org/10.1136/bmjopen-2020-048324.Search in Google Scholar PubMed PubMed Central

30. Denk, D, Greten, FR. Inflammation: the incubator of the tumor microenvironment. Trends Cancer 2022;8:901–14. https://doi.org/10.1016/j.trecan.2022.07.002.Search in Google Scholar PubMed

31. De, SG, di Masi, A, Ascenzi, P. Serum albumin: a multifaced enzyme. Int J Mol Sci 2021;22:10086. https://doi.org/10.3390/ijms221810086.Search in Google Scholar PubMed PubMed Central

32. Scrimshaw, NS, SanGiovanni, JP. Synergism of nutrition, infection, and immunity: an overview. Am J Clin Nutr 1997;66:464s–77s. https://doi.org/10.1093/ajcn/66.2.464s.Search in Google Scholar

33. Franco, AT, Corken, A, Ware, J. Platelets at the interface of thrombosis, inflammation, and cancer. Blood 2015;126:582–8. https://doi.org/10.1182/blood-2014-08-531582.Search in Google Scholar PubMed PubMed Central

34. Palumbo, JS, Talmage, KE, Massari, JV, La Jeunesse, CM, Flick, MJ, Kombrinck, KW, et al.. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood 2007;110:133–41. https://doi.org/10.1182/blood-2007-01-065995.Search in Google Scholar PubMed PubMed Central

35. Kwaan, HC, Lindholm, PF. Fibrin and fibrinolysis in cancer. Semin Thromb Hemost 2019;45:413–22. https://doi.org/10.1055/s-0039-1688495.Search in Google Scholar PubMed


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/oncologie-2024-0353).


Received: 2024-07-13
Accepted: 2024-11-25
Published Online: 2024-12-24

© 2024 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Editorial
  3. The essential development of integrative oncology
  4. Review Articles
  5. Novel post-translational modifications in cancer
  6. The assessment of 5-aminolevulinic acid photodynamic therapy in glioblastomas
  7. Histone methylation and acetylation in cancer: mechanism, progression, and targets
  8. Current advancements in PD-L1 modulation by CMTM6 in malignant tumors
  9. Research Articles
  10. Combination of oxabicycloheptene sulfonate (OBHS) and antidiabetic drugs synergistically inhibits proliferation in human breast cancer cells
  11. Cell indices as predictors of progression-free survival in multiple myeloma: a retrospective cohort study
  12. The Y44 residue point mutation in CXCL14 reveals the significance of CXCL14 protein stability in anti-tumor activity
  13. Oxeiptosis gene expression profiling identified TCN1 as a prognostic factor for breast cancer
  14. Integrated analyses reveal prognostic markers associated with cancer-associated fibroblasts in oral squamous cell carcinoma
  15. The roles of G protein-coupled receptor genes and the tumor microenvironment in esophageal squamous cell carcinoma
  16. Combining the Albumin-to-fibrinogen ratio and pathologic factors predicts survival in surgically treated patients with esophageal squamous cell carcinoma
  17. Joint multi-omics screening of neutrophil-related biomarkers for lung adenocarcinoma
  18. High-throughput sequencing reveals twelve cell death pattern prognostic target genes as potential drug-response-associated genes in the treatment of colorectal cancer cells with palmatine hydrochloride
  19. Article Commentary
  20. A commentary: optimizing the prevention of osteoradionecrosis in head and neck cancer treatment
Downloaded on 3.10.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2024-0353/html?licenseType=open-access
Scroll to top button