Retrospective Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. Sep 27, 2024; 16(9): 2910-2924
Published online Sep 27, 2024. doi: 10.4240/wjgs.v16.i9.2910
Preoperative systemic inflammatory response index as a prognostic marker for distal cholangiocarcinoma after pancreatoduodenectomy
Wen-Hui Zhang, Cheng-Run Zhang, Jin-Can Huang, Shao-Cheng Lyu, Ren Lang, Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
Yu Zhao, Department of Urology Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
ORCID number: Wen-Hui Zhang (0000-0002-5769-4942); Shao-Cheng Lyu (0000-0002-2757-7868); Ren Lang (0000-0001-9759-6725).
Co-corresponding authors: Shao-Cheng Lyu and Ren Lang.
Author contributions: Zhang WH and Zhao Y collected the related papers and drafted the manuscript; Zhang WH, Zhao Y, Zhang CR and Huang JC collected data; Zhang WH, Lyu SC and Lang R contributed to conceiving the research, analyzing data and revised the manuscript; Lyu SC and Lang R provided administrative support; All authors read and approved the final manuscript.
Supported by the Beijing Natural Science Foundation, No. 7222303.
Institutional review board statement: This study was reviewed and approved by the Ethics Committee of Beijing Chao-Yang Hospital, No. 2024-D-511.
Informed consent statement: This study has been granted exemption from informed consent. All procedures and protocols were reviewed and approved by the Institutional Review Board (IRB) prior to the commencement of the study. The IRB determined that the study met the criteria for exemption under the federal regulations governing human subject’s research. We have followed all necessary ethical guidelines and regulations throughout the study.
Conflict-of-interest statement: All the Authors have no conflict of interest related to the manuscript.
Data sharing statement: All data and materials are available from the corresponding author.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ren Lang, MD, PhD, Director, Doctor, Professor, Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing 100020, China. dr_langren@126.com
Received: July 3, 2024
Revised: August 5, 2024
Accepted: August 13, 2024
Published online: September 27, 2024
Processing time: 76 Days and 18.7 Hours

Abstract
BACKGROUND

The relationship between preoperative inflammation status and tumorigenesis as well as tumor progression is widely acknowledged.

AIM

To assess the prognostic significance of preoperative inflammatory biomarkers in patients with distal cholangiocarcinoma (dCCA) who underwent pancreatoduodenectomy (PD).

METHODS

This single-center study included 216 patients with dCCA after PD between January 1, 2011, and December 31, 2022. The individuals were categorized into two sets based on their systemic inflammatory response index (SIRI) levels: A low SIRI group (SIRI < 1.5, n = 123) and a high SIRI group (SIRI ≥ 1.5, n = 93). Inflammatory biomarkers were evaluated for predictive accuracy using receiver operating characteristic curves. Both univariate and multivariate Cox proportional hazards analyses were performed to estimate SIRI for overall survival (OS) and recurrence-free survival (RFS).

RESULTS

The study included a total of 216 patients, with 58.3% being male and a mean age of 65.6 ± 9.6 years. 123 patients were in the low SIRI group and 93 were in the high SIRI group after PD for dCCA. SIRI had an area under the curve value of 0.674 for diagnosing dCCA, showing better performance than other inflammatory biomarkers. Multivariate analysis indicated that having a SIRI greater than 1.5 independently increased the risk of dCCA following PD, leading to lower OS [hazard ratios (HR) = 1.868, P = 0.006] and RFS (HR = 0.949, P < 0.001). Additionally, survival analysis indicated a significantly better prognosis for patients in the low SIRI group (P < 0.001).

CONCLUSION

It is determined that a high SIRI before surgery is a significant risk factor for dCCA after PD.

Key Words: Distal cholangiocarcinoma; Pancreatoduodenectomy; Biomarker; Systemic inflammatory response index; Prognosis

Core Tip: The objective of this research was to evaluate the prognostic relevance of the systemic inflammatory response index (SIRI) in patients diagnosed with distal cholangiocarcinoma (dCCA) post-pancreatoduodenectomy (PD). A retrospective collection of all clinicopathological data was undertaken to further investigate the utility of SIRI in prognostication for dCCA patients post-PD. The results indicated that SIRI could potentially serve as a prognostic indicator for both overall survival and recurrence-free survival in dCCA patients following PD. Elevated levels of SIRI could predict recurrence and long-term survival in dCCA patients, thereby providing valuable guidance for treatment strategies.



INTRODUCTION

Distal cholangiocarcinoma (dCCA) is a type of cholangiocarcinoma that is located between the choledochal duct and about 1.5 cm closer to the ampulla of Vater[1,2]. Pancreatoduodenectomy (PD) is the primary curative treatment approach for patients with dCCA. Nevertheless, even with PD, the 5-year survival rates for dCCA are approximately 30%, primarily because of the considerable morbidity linked to this operation[3]. Late-stage diagnosis and lack of effective treatments worsen the limited prognosis for dCCA patients, making it crucial to improve outcomes for those undergoing PD[4].

New studies show that preoperative clinical variables can be used to create predictive models for cancer prognosis[5-9]. Preoperative inflammatory biomarkers obtained from laboratory tests have shown associations with tumor progression, metastasis, and poor survival outcomes. In dCCA, preoperative prognostic markers include neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), and neutrophil-to-monocyte ratio (NMR)[10]. Moreover, the systemic immune-inflammation index (SII), which is determined by multiplying the number of platelets in the blood with the NLR, has proven to be effective in forecasting prolonged survival in patients with dCCA who have had PD surgery[11]. Another significant biomarker, the albumin-to-bilirubin ratio (ABR), which reflects hepatic function, has shown a significant correlation with poor prognosis in gastrointestinal tumors such as hepatocellular carcinoma (HCC), pancreatic cancer, and gastric cancer[12-14]. The inflammation-combined prognostic index (ICPI), created as a new prognostic tool for gastric cancer, has demonstrated potential in forecasting survival results[15]. The aspartate transaminase-to-platelet (APRI), primarily used to evaluate hepatic fibrosis, has been found to play diverse roles in various cancers[16]. The systemic inflammatory response index (SIRI), which is determined by multiplying the number of neutrophils in the blood by the ratio of monocytes to lymphocytes, has shown to be a useful predictor for the return of tumors and premature death in various tumors such as HCC, pancreatic cancer, colorectal cancer, and colon cancer[17-19]. Notably, SII, ICPI, and SIRI have shown greater significance as independent factors compared to PLR, NLR, NMR, and LMR alone[20,21]. However, the interpretation of SIRI in individuals with dCCA post-PD is still debated. Further research is needed to elucidate its role in this specific patient population. This study aims to assess if SIRI can predict long-term survival in patients who have had PD for dCCA.

MATERIALS AND METHODS
Study population

A retrospective analysis at Beijing Chao-Yang Hospital examined using SIRI as a preoperative marker for prolonged survival in dCCA patients undergoing PD. Data from patients who had PD between January 1, 2011, to December 31, 2022 was collected from the hospital’s electronic medical record system. The research protocol followed the ethical principles set forth in the 1975 Declaration of Helsinki. Approval for the retrospective study was granted by the hospital’s ethics committees, with all patients providing written informed consent at June 26th 2024 (No. 2024-D-511).

Exposure definition

In this study, several inflammatory biomarkers were defined and calculated based on blood test results obtained from the enrolled patients. NLR was determined by dividing the number of neutrophils by the number of lymphocytes. PLR was calculated by dividing the platelet count by the lymphocyte count. LMR was calculated by dividing the lymphocyte count by the monocyte count. NMR was calculated by neutrophil counts divided by monocyte counts. ABR was calculated by the ratio of the absolute count of albumin to bilirubin. The ICPI score was calculated by combining the values of LMR, NLR, and PLR, with the formula 2.9 multiplied by LMR, plus 2.8 multiplied by NLR, plus 2.8 multiplied by PLR. Furthermore, the APRI was determined by dividing the aspartate transaminase (AST) by the upper limit of normal, multiplying by 100, and then dividing by the platelet counts. The SII was calculated by multiplying the platelet count by the NLR. The calculation of SIRI involved multiplying the monocyte count by the NLR. Before surgery, venous blood samples were collected from all patients for analysis of complete blood count, biochemical examination, and tumor biomarker tests, which included carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9), within 24 hours. Leukopenia, leukocytosis, and cholangitis, which could be potential alterations resulting from acute infectious or inflammatory conditions, were excluded to focus on chronic effects related to cancer. Patients’ pathological stages were determined according to the 8th edition of the Union for International Cancer Control[20].

Outcome ascertainment and follow-up

Follow-up data was collected for all enrolled patients through various means, including telephone interviews and outpatient or inpatient observations, until April 2023 or death. Surveillance post-surgery typically involves scheduled visits every 3-6 months within the initial 2 years, followed by visits every 6-12 months for up to 5 years or as per clinical necessity[21]. This monitoring regimen commonly includes a blend of clinical assessments, laboratory analyses encompassing liver function tests and tumor markers, as well as thoracic, abdominal, and pelvic computed tomography (CT) scans. Patients experiencing postoperative biliary obstruction necessitate thorough multidisciplinary assessment to pinpoint the obstruction site, assess for recurrence, and devise an optimal drainage strategy[21]. The recurrence of dCCA primarily hinges on a comprehensive evaluation involving clinical manifestations, tumor markers, and imaging assessments. Following surgery, patients may present with abdominal pain, fever, jaundice, cachexia, and other symptoms. Concurrently, postoperative monitoring involves tracking fluctuations in tumor markers. Subsequently, the diagnosis of recurrence is confirmed through contrast-enhanced CT and magnetic resonance imaging to identify biliary duct thickening or new lesion formation. Positron emission tomography-CT plays a crucial role in assessing tumor activity and detecting potential metastases outside the primary surgical site[21]. Recurrent dCCA often manifests as biliary obstruction, accompanied by symptoms such as recurrent fever, signaling the need for heightened vigilance towards the likelihood of recurrence. Once recurrence is detected, a combination of options such as surgery, radiotherapy or chemotherapy are considered and used depending on the patient's condition. Of note, none of the patients diagnosed with dCCA before surgery received chemotherapy or radiotherapy containing gemcitabine or cisplatin. Overall survival (OS) was defined as the time elapsed between the treatment date and the date of death or last follow-up. Recurrence-free survival (RFS) was defined as the duration between the treatment date and the date of recurrence.

Statistical analysis

Continuous variables will be presented as means and standard deviations or medians and interquartile ranges, depending on their distribution. Group comparisons will be done using the Student’s t test or Mann-Whitney U test for continuous variables, and χ2 test or Fisher’s exact test for categorical variables. The normality of continuous variables will be assessed with the Kolmogorov-Smirnov test. Receiver operating characteristic (ROC) curves will determine the best threshold values for predicting dCCA prognosis after PD. Kaplan-Meier curves will be used for survival analysis, with differences in time-to-event endpoints assessed using statistical tests. Univariable regression analysis will identify risk factors for death and recurrence in dCCA patients post-PD. Variables with a P value less than 0.2 from the univariable Cox analysis will be considered in the multivariable Cox regression to identify risk factors that are independent. Missing data will be addressed through multiple imputation to minimize bias and enhance the accuracy of the analysis. Statistical significance will be determined by a two-tailed P value below 0.05. Analysis and presentation of data will be conducted with statistical product and service solutions 26.0 (International Business Machines Corporation, Armonk, NY, United States) and GraphPad Prism 8.0 (GraphPad Software, San Diego, CA, United States), respectively.

RESULTS
Demographic and baseline characteristics of patients

A flowchart depicting the inclusion and exclusion of patients has been provided in Figure 1. A sum of 216 patients who were diagnosed with dCCA after PD and met the inclusion and exclusion criteria were recruited during the entire study period. After a thorough examination, it was determined that the average age of the patients who were registered was 65.6 ± 9.6 years, with males making up 58.3% (126 out of 216). Records indicate a median follow-up time of 36.5 months.

Figure 1
Figure 1 Flowchart of the study cohort. dCCA: Distal cholangiocarcinoma.

In Table 1, there is a summary of the initial traits of the low and high SIRI groups. The low SIRI group consisted of 123 patients, while the high SIRI group comprised 93 patients. Significant variations were noted between the two groups in terms of age (P = 0.004), cardiocerebrovascular disease (P = 0.001), preoperative biliary drainage (P < 0.001), platelet counts (P = 0.026), white blood count (WBC) (P < 0.001), neutrophil counts (P < 0.001), monocyte counts (P < 0.001), lymphocyte counts (P < 0.001), PLR (P < 0.001), NLR (P < 0.001), LMR (P < 0.001), ABR (P = 0.034), SII (P < 0.001), and ICPI (P < 0.001). Compared to the low SIRI group, hemoglobin counts (P = 0.027), albumin counts (P = 0.013), total bilirubin (TB) levels (P = 0.040), CA19-9 levels (P = 0.001) were significantly higher in the high SIRI group. Nonetheless, there were no significant differences (P > 0.05) in sex, smoking, diabetes, epigastrium surgery, alanine aminotransferase, AST, direct bilirubin, g-glutamyl transpeptidase, CEA, NMR, APRI, operative outcomes including operative duration, total blood loss, blood transfusion, postoperative outcomes including ventilator use duration, intensive care unit duration and postoperative time between the two groups. The results from the pathological analysis of both groups were examined and are displayed in Table 2. There were no statistically significant differences in pathological stage, tumor differentiation, microscopic lymphatic invasion, microscopic venous invasion, microscopic arterial invasion, peripheral organ invasion, venous invasion type, perineural invasion, distant metastasis, incisal edge and residual tumor status. It is important to mention that there were notable variations between the two groups regarding tumor size (P = 0.045) and type of venous invasion (P = 0.014).

Table 1 The clinical characteristics of primary cohort.
Variables
Total (n = 216)
Low SIRI (n = 123)
High SIRI (n = 93)
P value
Patient basic factors
Age (year)
≥ 707935440.004a
< 701378849
Gender
Male12667590.186
Female905634
Main symptom
Abdominal pain231670.213
Jaundice1829983
Other1183
Smoking
Yes6431330.101
No1529260
Diabetes
Yes5831270.530
No1589266
Epigastrium surgery
Yes201370.445
No19611086
Cardiocerebrovascular disease
Yes10950590.001a
No1077334
Preoperative biliary drainage
ERCP24186< 0.001a
PTBD813645
No1116942
Laboratory parameters
Platelet (109/L)
≥ 3114318250.026a
< 31117310568
WBC (109/L)
≥ 7.2651649< 0.001a
< 7.215110744
Neutrophil (109/L)
≥ 3.61375186< 0.001a
< 3.679727
Monocyte (109/L)
≥ 0.5781761< 0.001a
< 0.513810632
Lymphocyte (109/L)
≥ 1.849427< 0.001a
< 1.81678186
ALT (U/L)
≥ 190.53523120.252
< 190.518110081
AST (U/L)
≥ 16.5206118880.650
< 16.51055
Hemoglobin (g/L)
≥ 11213283490.027a
< 112844044
Albumin (g/dL)
≥ 35.710066340.013a
< 35.71165759
TB (μmol/L)
≥ 144.48340430.040a
< 144.41338350
DB (μmol/L)
≥ 113.88442420.100
< 113.81328151
-GGT (U/L)
≥ 646.56639270.673
< 646.51508466
CEA (ng/mL)
≥ 0.2209117920.118
< 0.2761
CA19-9 (U/mL)
≥ 235.86124370.001a
< 235.81559956
PLR
≥ 1431456580< 0.001a
< 143715813
NLR
≥ 3.11123280< 0.001a
< 3.11049113
LMR
≥ 3.2100928< 0.001a
< 3.21163185
NMR
≥ 11.26937320.499
< 11.21478661
ABR
≥ 0.312076440.034a
< 0.3964749
SII
≥ 6821233984< 0.001a
< 68293849
ICPI
≥ 445.41335776< 0.001a
< 445.4836617
APRI
≥ 0.3178103750.554
< 0.3382018
Operative outcomes
Operative duration (hours)10.1 ± 2.610.1 ± 2.610.1 ± 2.60.900
Total blood loss (mL)618.8 ± 517.5621.3 ± 535.6616.8 ± 505.20.950
Blood transfusion (mL)308.3 ± 538.8259.6 ± 511.0345.9 ± 558.70.244
Postoperative outcomes
Ventilator use duration (hours)20.9 ± 27.518.4 ± 20.922.9 ± 31.60.234
ICU duration (days)3.5 ± 3.33.4 ± 3.23.7 ± 3.60.347
Postoperative time (days)27.0 ± 18.226.0 ± 15.827.7 ± 19.90.498
Table 2 Pathological characteristics of the patients classified by the systemic inflammatory response index.
Variables
Total (n = 216)
Low SIRI (n = 123)
High SIRI (n = 93)
P value
pT stage
Tis3210.654
T1, T226179
T3, T418410381
Unknown312
pN stage
N011167440.676
N1744133
N2291415
Unknown211
pM stage
M0204118860.271
M11257
Tumor differentiation
Well251960.097
Moderate905436
Poor261313
Other753738
Tumor size (cm)
≥ 1.814074660.045a
< 1.8744925
Unknown202
Microscopic lymphatic invasion
Yes10355480.578
No1116744
Unknown211
Microscopic venous invasion
Yes3617190.214
No17910673
Unknown101
Microscopic arterial invasion
Yes6240.344
No20712087
Unknown312
Peripheral organ invasion
Yes3210.670
No21012090
Unknown312
Venous invasion type
1-224159
3-4122100.014a
No18010674
Perineural invasion
Yes189104850.268
No25187
Unknown211
Distant metastasis
Yes12570.189
No20411876
Incisal edge
Positive2414100.884
Negative19210983
Residual tumor status
R0191109820.490
R119109
R2624
Preoperative inflammatory biomarkers and prognosis of patients with dCCA

ROC curves were utilized to establish the connection between preoperative inflammatory markers and patient outcomes in dCCA following PD (Figure 2). The optimal cutoff values for CA19-9, PLR, NLR, LMR, NMR, APRI, ABR, ICPI, SII, and SIRI to predict OS were identified as 235.8, 143, 3.1, 3.2, 11.2, 0.3, 0.3, 445.4, 682, and 1.5, respectively. The corresponding area under the curves (AUCs) for these biomarkers were calculated as 0.661, 0.637, 0.672, 0.654, 0.555, 0.503, 0.632, 0.636, 0.667, and 0.674, respectively (Supplementary Table 1). The results of the ROC analysis indicated that APRI exhibited the highest sensitivity of 0.880, while CA19-9 and NMR demonstrated the highest specificity of 0.907 and 0.773, respectively, compared to other markers. The postoperative RFS rates for the markers mentioned above ranged from 0.503 to 0.604, with AUC values of 0.516, 0.543, 0.553, 0.556, 0.565, 0.568, and 0.569 (Supplementary Table 2). In the ROC analysis, it was found that LMR had the highest sensitivity at 0.975, whereas ABR showed the highest specificity at 0.958 among all the markers.

Figure 2
Figure 2 The receiver operating characteristic curves explore the value of preoperative inflammatory biomarkers in predicting the long-time prognosis in distal cholangiocarcinoma after pancreatoduodenectomy. A: The value of receiver operating characteristic (ROC) in predicting overall survival in distal cholangiocarcinoma (dCCA) after pancreatoduodenectomy (PD); B: The value of ROC in predicting recurrence-free survival in dCCA after PD. PLR: Platelet-to-lymphocyte ratio; NLR: Neutrophil to lymphocyte ratio; LMR: Lymphocyte to monocyte ratio; NMR: Neutrophil-to-monocyte ratio; APRI: Aspartate transaminase-to-platelet; ABR: Albumin to bilirubin ratio; ICPI: Inflammation-combined prognostic index; SII: Systemic immune-inflammation index; SIRI: Systemic inflammatory response index.

The outcomes of univariate and multivariate Cox proportional hazard regression analyses for forecasting OS and RFS were showed in Table 3 and 4. Several independent factors significantly associated with OS were identified through multivariate Cox regression analysis.

Table 3 Cox regression analysis of the effects of clinicopathological factors on the overall survival of patients.
VariablesUnivariate analysis
Multivariate analysis
HR (95%CI)
P value
HR (95%CI)
P value
Age (year)1.022 (1.003-1.042)0.025a1.008 (0.987-1.029)0.471
Cardiocerebrovascular disease1.424 (1.020-1.986)0.038a1.428 (0.989-2.063)0.058
Preoperative biliary drainage1.166 (0.974-1.396)0.0940.746 (0.401-1.389)0.355
Platelet (109/L)1.002 (1.000-1.004)0.1011.001 (0.996-1.007)0.661
WBC (109/L)1.044 (0.975-1.118)0.219
Neutrophil (109/L)1.089 (1.011-1.174)0.025a1.110 (0.900-1.369)0.330
Monocyte (109/L)2.000 (1.048-3.818)0.036a0.078 (0.003-1.761)0.109
Lymphocyte (109/L)0.624 (0.469-0.830)0.001a0.598 (0.287-1.245)0.169
Hemoglobin (g/L)0.990 (0.981-0.999)0.022a1.001 (0.989-1.013)0.895
Albumin (g/dL)1.560 (1.104-2.204)0.012a0.983 (0.948-1.019)0.345
TB (μmol/L)1.002 (1.001-1.004)0.003a1.002 (1.000-1.004)0.030a
CA19-9 (U/mL) (> 235.8)1.000 (1.000-1.000)0.000a1.000 (0.999-1.000)0.000a
PLR (> 143)1.003 (1.002-1.005)0.000a1.000 (0.995-1.005)0.898
NLR (> 3.1)1.093 (1.042-1.146)0.000a0.702 (0.525-0.939)0.017
LMR (> 3.2)0.807 (0.726-0.898)0.000a0.934 (0.712-1.225)0.621
NMR (> 11.2)0.992 (0.969-1.015)0.490
ABR (> 0.3)0.937 (1.091-2.054)0.403
SII (> 682)1.000 (1.000-1.001)0.000a1.000 (0.999-1.001)0.644
SIRI (> 1.5)1.167 (1.092-1.247)0.000a1.868 (1.194-2.920)0.006a
ICPI (> 445.4)1.001 (1.001-1.002)0.000a3.108 (1.710-5.651)0.305
APRI (> 0.3)1.016 (0.877-1.177)0.831
Tumor size (cm) (> 1.8)1.292 (1.116-1.497)0.001a1.343 (1.130-1.596)0.001a
Venous invasion type1.371 (0.430-4.368)0.594
Table 4 Cox regression analysis of the effects of clinicopathological factors on the recurrence-free survival of patients.
VariablesUnivariate analysis
Multivariate analysis
HR (95%CI)
P value
HR (95%CI)
P value
Age (year)1.001 (0.980-1.023)0.931
Cardiocerebrovascular disease1.225 (0.844-1.777)0.285
Preoperative biliary drainage0.679 (0.343-1.342)0.265
Platelet (109/L)1.002 (0.999-1.005)0.229
WBC (109/L)0.978 (0.906-1.056)0.565
Neutrophil (109/L)0.999 (0.914-1.091)0.982
Monocyte (109/L)0.839 (0.409-1.720)0.632
Lymphocyte (109/L)0.695 (0.516-0.936)0.017a0.834 (0.478-1.456)0.523
Hemoglobin (g/L)1.001 (0.991-1.011)0.872
Albumin (g/dL)1.019 (0.989-1.051)0.221
TB (μmol/L)1.001 (0.999-1.002)0.469
CA19-9 (U/mL) (> 69.3)1.000 (1.000-1.000)0.970
PLR (> 159.8)1.003 (1.001-1.005)0.001a1.000 (0.995-1.004)0.811
NLR (> 2.8)1.129 (1.024-1.245)0.015a0.895 (0.694-1.155)0.393
LMR (> 1.1)0.888 (0.798-0.989)0.030a0.922 (0.771-1.101)0.368
NMR (> 11.5)0.992 (0.959-1.026)0.624
ABR (> 3.7)0.938 (0.792-1.112)0.463
SII (> 496.6)1.001 (1.000-1.001)0.002a1.001 (1.000-1.002)0.146
ICPI (> 445.4)1.001 (1.000-1.002)0.001a0.894 (0.784-1.020)0.096
APRI (> 1.7)1.032 (0.866-1.229)0.726
SIRI (> 1.1)1.080 (0.972-1.200)0.001a0.949 (0.752-1.199)0.000a
Tumor size (cm) (> 1.8)1.258 (1.013-1.561)0.038a1.144 (0.887-1.475)0.229
Venous invasion type1.503 (0.819-2.759)0.1891.577 (0.566-4.391)0.383

These factors included TB (> 144.4), CA19-9 (> 235.8), SIRI (> 1.5), and tumor size (> 1.8), with hazard ratios (HR) and corresponding P values as follows: TB (HR = 1.002, P = 0.030), CA19-9 (HR = 1.000, P = 0.000), SIRI (HR = 1.868, P = 0.006), and tumor size (HR = 1.343, P = 0.001) (Table 3). In RFS, SIRI (> 1.5) was found to be a significant risk factor with a HR of 0.949 and P value of 0.000 (Table 4). Survival analysis using Kaplan-Meier method was performed to evaluate the extended outlook (Figure 3). In comparison to the high-SIRI group, the low-SIRI group exhibited notably higher 5-year OS rate and RFS rate (14.0% vs 30.0%, log-rank test, P < 0.001; 8.0% vs 14.0%, Breslow-Wilcoxon test, P = 0.006).

Figure 3
Figure 3 Kaplan-Meier curves for long-time prognosis by systemic inflammatory response index. A: Kaplan-Meier curves for overall survival by systemic inflammatory response index (SIRI); B: Kaplan-Meier curves for recurrence-free survival by SIRI. SIRI: Systemic inflammatory response index; OS: Overall survival; RFS: Recurrence-free survival.
DISCUSSION

This study is significant as it is the first to examine the relationship between preoperative SIRI, clinicopathological factors, and PD outcomes in patients with dCCA, with a larger patient cohort than previous studies[5,22-24]. In this retrospective clinical study, the researchers established predictive factors by considering numerous clinicopathological factors, as observed in previous clinical investigations[25-28]. The findings of this study revealed that SIRI functioned as an independent and definitive prognostic indicator for the 5-year OS and RFS of patients who underwent PD for dCCA.

The study found that SIRI was not linked to preoperative biliary drainage or other pathological results, except for tumor size. However, significant differences were seen in TB and CA19-9 levels. These results suggest that SIRI may offer insights into tumor progression mechanisms not captured by traditional methods. Preoperative imaging can provide an early assessment of tumor size, which may be delayed with histopathological tests[29-31]. Multiple studies have linked tumor size detected through imaging to patient prognosis. CA19-9 is a tumor biomarker that can track tumor progression and treatment effectiveness in cancer, particularly in dCCA. It is not useful for early diagnosis but is crucial for comprehensive treatment and follow-up[32]. These findings align with the results of other studies[33,34].

SIRI serves as a novel predictive tool that reflects immune response and inflammatory status. Multiple studies have shown that additional inflammatory indicators like SII[35], PLR[10] and NLR[11] are considered independent risk factors for patients with dCCA after undergoing PD. Furthermore, a recent systematic review and meta-analysis found that PLR may independently predict the prognosis of patients with dCCA[36]. These findings highlight the close correlation between preoperative inflammatory markers and tumor progression. In fact, PLR, NLR, and LMR are superior inflammatory markers for predicting tumor prognosis than individual markers, such as CRP, WBC, neutrophil, and procalcitonin based on many valuable studies[10,37,38]. Moreover, SIRI, SII, and prognostic nutritional index, which combine inflammation markers, are even more effective[35,39]. In contrast to other inflammatory markers, SIRI integrates neutrophil, lymphocyte, and monocyte counts, reflecting both inflammation and immune status. SIRI’s superior predictive power in dCCA may result from its holistic evaluation of the immune-inflammatory balance. It interprets neutrophilia as inflammation, lymphopenia as immunosuppression, and monocyte changes as tumor-related inflammation. This comprehensive perspective provides a more detailed prognosis than individual biomarkers, making SIRI a reliable predictor of dCCA outcomes. Mechanistically, a strict and copious review has indicated that neutrophils play heterogeneous and multifaceted roles either protumoral functions or antitumoral in tumor microenvironment[40]. Neutrophils discharge a sequence of enzymes that cultivate an environment conducive to tumor angiogenesis, a process that can potentially facilitate tumor growth, progression, and metastasis[41,42]. Alternatively, neutrophils can combat tumors by directly eliminating tumor cells and contributing to cellular networks that resist tumors[42]. The two-sided role of neutrophils in tumors has sparked interest in using them for cancer treatment[42,43]. Likewise, activated monocytes have been shown to have anti-tumor functions and can shape the tumor microenvironment to promote the spread of tumor cells. Antitumor functions and antigen-presenting cells activated by monocytes were concluded in many studies[44-46]. In summary, both neutrophils and monocytes demonstrate plasticity and exhibit dual effects in response to environmental stimuli. Lymphocytes, on the other hand, are well-recognized for their roles in human immune responses, including anti-infection and anti-tumor effects[47]. Various T cell subtypes like cluster of differentiation (CD) 4+ T cells and CD8+ T cells have the ability to suppress the growth, spread, and movement of tumor cells[48-50].

An elevated SIRI in dCCA can significantly influence treatment decisions and postoperative monitoring strategies. Elevated SIRI levels may indicate dysregulated immune responses and heightened tumor-related inflammation, suggesting a more aggressive disease phenotype[51,52]. This insight can direct clinicians towards enhanced postoperative surveillance, potentially requiring closer monitoring and personalized adjuvant therapies to address the increased inflammatory burden and immune dysregulation[53,54]. Furthermore, identifying elevated SIRI levels opens avenues for future research in dCCA. It is crucial to conduct prospective validation studies to validate the prognostic significance of SIRI and its impact on treatment outcomes. Exploring the mechanistic connections between inflammation and tumor progression in dCCA could offer valuable insights into the underlying pathophysiology and unveil potential therapeutic targets. Understanding these intricate relationships may pave the way for targeted interventions aimed at modulating the immune-inflammatory microenvironment to enhance patient outcomes in dCCA. The advancement of minimally invasive techniques has made laparoscopic pancreaticoduodenectomy (LPD) more beneficial than open PD due to shorter hospital stays and comparable short-term health risks. The use of preoperative inflammatory markers can further refine the benefits and indications of LPD, especially in superior mesenteric artery-first approach, mitigating postoperative complications, enhancing patient prognosis in further research[55,56].

SIRI has garnered significant attention as a comprehensive biomarker not only for predicting prognosis but also for assessing the activity of chemotherapy in advanced cancers[57-59]. Pretreatment SIRI has shown potential prognostic significance for cancer patients receiving neoadjuvant chemotherapy and has influenced treatment strategy decisions[58,60]. Doctors can more accurately evaluate the progress of patients receiving neoadjuvant chemotherapy for malignant tumors by using SIRI as an important predictive factor. This highlights the importance of considering dCCA as a systemic disease and underscores the potential of SIRI to reflect the response to chemotherapy and immunotherapy in patients within a multidisciplinary setting.

There are various constraints to this study that need to be taken into account. Firstly, the study was conducted retrospectively at a single center, potentially restricting the applicability of the results. Secondly, preoperative inflammatory biomarkers, including SIRI, can exhibit fluctuations due to internal homeostasis, which may weaken their clinical value if dynamic monitoring is not conducted. Next, although this study adjusted for a variety of clinicopathological characteristics in multivariate analyses, there remain a number of potential confounders that may affect inflammatory biomarker levels or patient prognosis, such as comorbidities, preoperative treatment affecting inflammation, or postoperative complications. Common complications of dCCA, such as biliary tract infection, obstructive jaundice, liver dysfunction, and cholelithiasis, can all serve as confounding factors significantly affecting the predictive value of SIRI[61,62]. Biliary tract infection exacerbates the inflammatory state by releasing pro-inflammatory cytokines and activating immune cells, impacting the accuracy of preoperative inflammatory markers[62]. Obstructive jaundice is a hallmark of advanced dCCA, leading to bile stasis and increased bile pressure, creating a local inflammatory environment within the bile ducts. This local inflammation can trigger systemic inflammation, thereby affecting preoperative inflammatory markers[63]. Furthermore, due to impaired liver function and disrupted bile flow, the synthesis and regulation of inflammatory mediators are compromised, also influencing the accuracy of preoperative inflammatory markers[61]. Concurrent cholelithiasis further escalates inflammation and complicates the interpretation of preoperative inflammatory indicators[64-66]. Medications administered to dCCA patients, particularly those with anti-inflammatory or immunomodulatory properties, can modulate the systemic inflammatory response. These medications may suppress or enhance inflammatory pathways, leading to alterations in preoperative inflammatory marker levels[67,68]. Consequently, the predictive capacity of these markers for prognosis may be influenced by the concurrent use of such medications. Postoperative complications in dCCA patients primarily encompass pancreatic fistula, abdominal infections, hemorrhage, bile leakage, and delayed gastric emptying[69]. While the occurrence of postoperative complications does not directly impact the preoperative inflammatory markers, it significantly influences the patient’s prognosis[69,70]. In severe instances, these complications can precipitate early postoperative mortality. Henceforth, forthcoming research should incorporate the inclusion of dynamic monitoring of preoperative clinical indicators. Meanwhile, LPD combined with preoperative inflammatory markers will further reduce postoperative complications. Future studies need to confirm whether combining LPD with preoperative inflammatory markers can further reduce postoperative complications. Last but not least, long-term follow-up is necessary to assess the predictive value of SIRI and other factors, but follow-up bias may occur over time. In essence, further research with larger samples and collaboration between multiple centers is needed to confirm these findings and overcome study limitations.

CONCLUSION

To summarize, the preoperative SIRI can predict recurrence and long-term survival in dCCA patients, helping guide treatment decisions for better outcomes.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Ke JJ S-Editor: Fan M L-Editor: A P-Editor: Chen YX

References
1.  Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, Nakamura Y, White R, Smits AM, Bos JL. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319:525-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4616]  [Cited by in F6Publishing: 4396]  [Article Influence: 122.1]  [Reference Citation Analysis (0)]
2.  Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet. 2014;383:2168-2179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1072]  [Cited by in F6Publishing: 1246]  [Article Influence: 124.6]  [Reference Citation Analysis (0)]
3.  Blechacz B, Komuta M, Roskams T, Gores GJ. Clinical diagnosis and staging of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol. 2011;8:512-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in F6Publishing: 475]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
4.  Loeffler MA, Hu J, Kirchner M, Wei X, Xiao Y, Albrecht T, De La Torre C, Sticht C, Banales JM, Vogel MN, Pathil-Warth A, Mehrabi A, Hoffmann K, Rupp C, Köhler B, Springfeld C, Schirmacher P, Ji J, Roessler S, Goeppert B. miRNA profiling of biliary intraepithelial neoplasia reveals stepwise tumorigenesis in distal cholangiocarcinoma via the miR-451a/ATF2 axis. J Pathol. 2020;252:239-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
5.  Cummings M, Merone L, Keeble C, Burland L, Grzelinski M, Sutton K, Begum N, Thacoor A, Green B, Sarveswaran J, Hutson R, Orsi NM. Preoperative neutrophil:lymphocyte and platelet:lymphocyte ratios predict endometrial cancer survival. Br J Cancer. 2015;113:311-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 93]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
6.  Cong R, Kong F, Ma J, Li Q, Wu Q, Ma X. Combination of preoperative neutrophil-lymphocyte ratio, platelet-lymphocyte ratio and monocyte-lymphocyte ratio: a superior prognostic factor of endometrial cancer. BMC Cancer. 2020;20:464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
7.  Mungan İ, Dicle ÇB, Bektaş Ş, Sarı S, Yamanyar S, Çavuş M, Turan S, Bostancı EB. Does the preoperative platelet-to-lymphocyte ratio and neutrophil-to-lymphocyte ratio predict morbidity after gastrectomy for gastric cancer? Mil Med Res. 2020;7:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
8.  Nøst TH, Alcala K, Urbarova I, Byrne KS, Guida F, Sandanger TM, Johansson M. Systemic inflammation markers and cancer incidence in the UK Biobank. Eur J Epidemiol. 2021;36:841-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 182]  [Article Influence: 60.7]  [Reference Citation Analysis (0)]
9.  Savioli F, Morrow ES, Dolan RD, Romics L, Lannigan A, Edwards J, McMillan DC. Prognostic role of preoperative circulating systemic inflammatory response markers in primary breast cancer: meta-analysis. Br J Surg. 2022;109:1206-1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
10.  Hoshimoto S, Hishinuma S, Shirakawa H, Tomikawa M, Ozawa I, Ogata Y. Association of Preoperative Platelet-to-Lymphocyte Ratio with Poor Outcome in Patients with Distal Cholangiocarcinoma. Oncology. 2019;96:290-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
11.  Kumamoto Y, Kaizu T, Tajima H, Nishizawa N, Ei S, Igarashi K, Watanabe M. Neutrophil-to-lymphocyte ratio as a predictor of postoperative morbidity in patients with distal cholangiocarcinoma. Mol Clin Oncol. 2018;9:362-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 15]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
12.  Xu YX, Wang YB, Tan YL, Xi C, Xu XZ. Prognostic value of pretreatment albumin to bilirubin ratio in patients with hepatocellular cancer: A meta-analysis. Medicine (Baltimore). 2019;98:e14027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
13.  Hiraoka A, Kumada T, Kudo M, Hirooka M, Tsuji K, Itobayashi E, Kariyama K, Ishikawa T, Tajiri K, Ochi H, Tada T, Toyoda H, Nouso K, Joko K, Kawasaki H, Hiasa Y, Michitaka K; Real-Life Practice Experts for HCC (RELPEC) Study Group and HCC 48 Group (hepatocellular carcinoma experts from 48 clinics). Albumin-Bilirubin (ALBI) Grade as Part of the Evidence-Based Clinical Practice Guideline for HCC of the Japan Society of Hepatology: A Comparison with the Liver Damage and Child-Pugh Classifications. Liver Cancer. 2017;6:204-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 142]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
14.  Chan SL, Wong LL, Chan KA, Chow C, Tong JH, Yip TC, Wong GL, Chong CC, Liu PH, Chu CM, Wong VW, To KF, Reeves HL, Chan AW. Development of a Novel Inflammation-Based Index for Hepatocellular Carcinoma. Liver Cancer. 2020;9:167-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
15.  Hirahara N, Matsubara T, Kaji S, Hayashi H, Sasaki Y, Kawakami K, Hyakudomi R, Yamamoto T, Tajima Y. Novel inflammation-combined prognostic index to predict survival outcomes in patients with gastric cancer. Oncotarget. 2023;14:71-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
16.  Mobarak L, Omran D, Nabeel MM, Zakaria Z. Fibro markers for prediction of hepatocellular carcinoma in Egyptian patients with chronic liver disease. J Med Virol. 2017;89:1062-1068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
17.  Geng Y, Qi Q, Sun M, Chen H, Wang P, Chen Z. Prognostic nutritional index predicts survival and correlates with systemic inflammatory response in advanced pancreatic cancer. Eur J Surg Oncol. 2015;41:1508-1514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
18.  Bai X, Feng L. Correlation between Prognostic Nutritional Index, Glasgow Prognostic Score, Systemic Inflammatory Response, and TNM Staging in Colorectal Cancer Patients. Nutr Cancer. 2020;72:1170-1177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
19.  Laird BJ, Fallon M, Hjermstad MJ, Tuck S, Kaasa S, Klepstad P, McMillan DC. Quality of Life in Patients With Advanced Cancer: Differential Association With Performance Status and Systemic Inflammatory Response. J Clin Oncol. 2016;34:2769-2775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 110]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
20.  Sa-Ngiamwibool P, Aphivatanasiri C, Sangkhamanon S, Intarawichian P, Kunprom W, Thanee M, Prajumwongs P, Loilome W, Khuntikeo N, Titapun A, Jareanrat A, Thanasukarn V, Srisuk T, Luvira V, Eurboonyanun K, Promsorn J, Wee A, Koonmee S. Modification of the AJCC/UICC 8th edition staging system for intrahepatic cholangiocarcinoma: proposal for an alternative staging system from cholangiocarcinoma-prevalent Northeast Thailand. HPB (Oxford). 2022;24:1944-1956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
21.  Vogel A, Bridgewater J, Edeline J, Kelley RK, Klümpen HJ, Malka D, Primrose JN, Rimassa L, Stenzinger A, Valle JW, Ducreux M; ESMO Guidelines Committee. Biliary tract cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2023;34:127-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 137]  [Article Influence: 137.0]  [Reference Citation Analysis (0)]
22.  Bao Y, Wang Y, Li X, Pan M, Zhang H, Cheng Z, Wang X. Prognostic significance of platelet-to-lymphocyte ratio in urothelial carcinoma patients: a meta-analysis. Cancer Cell Int. 2019;19:315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
23.  Zhong X, Su T, Yang Y, Ye L, Jiang L, Qi Y, Xie J, Jiang Y, Zhou W, Zhang C, Wu L, Zhu H, Ning G, Wang W. Platelet-Lymphocyte and Neutrophil-Lymphocyte Ratios Are Prognostic Markers for Pheochromocytomas and Paragangliomas. J Clin Endocrinol Metab. 2023;108:2230-2239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
24.  Zhou K, Cao J, Lin H, Liang L, Shen Z, Wang L, Peng Z, Mei J. Prognostic role of the platelet to lymphocyte ratio (PLR) in the clinical outcomes of patients with advanced lung cancer receiving immunotherapy: A systematic review and meta-analysis. Front Oncol. 2022;12:962173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
25.  Woo SM, Ryu JK, Lee SH, Yoo JW, Park JK, Kim YT, Jang JY, Kim SW, Kang GH, Yoon YB. Recurrence and prognostic factors of ampullary carcinoma after radical resection: comparison with distal extrahepatic cholangiocarcinoma. Ann Surg Oncol. 2007;14:3195-3201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 88]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
26.  Maeta T, Ebata T, Hayashi E, Kawahara T, Mizuno S, Matsumoto N, Ohta S, Nagino M; Nagoya Surgical Oncology Group. Pancreatoduodenectomy with portal vein resection for distal cholangiocarcinoma. Br J Surg. 2017;104:1549-1557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
27.  van der Gaag NA, Kloek JJ, de Bakker JK, Musters B, Geskus RB, Busch ORC, Bosma A, Gouma DJ, van Gulik TM. Survival analysis and prognostic nomogram for patients undergoing resection of extrahepatic cholangiocarcinoma. Ann Oncol. 2012;23:2642-2649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
28.  Bird N, Elmasry M, Jones R, Elniel M, Kelly M, Palmer D, Fenwick S, Poston G, Malik H. Role of staging laparoscopy in the stratification of patients with perihilar cholangiocarcinoma. Br J Surg. 2017;104:418-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
29.  Sun XM, Pang F, Zhuang SM, Xie LE, Zhong QY, Liu TR. Tumor size rather than the thyroid invasion affects the prognosis of parathyroid carcinoma without lymph node or distant metastasis. Eur Arch Otorhinolaryngol. 2022;279:4587-4594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
30.  Gong H, Zhou L, Wu C, Hsueh CY, Tao L, Wu H, Zhang M, Chen X, Li C, Zhou J. Preoperative tracheotomy as reflection of tumor size impacting oncologic outcomes of patients with advanced stage glottic carcinoma. Eur Arch Otorhinolaryngol. 2021;278:4943-4950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
31.  Zhang W, Liu Z, Chen J, Dong S, Cen B, Zheng S, Xu X. A preoperative model for predicting microvascular invasion and assisting in prognostic stratification in liver transplantation for HCC regarding empirical criteria. Transl Oncol. 2021;14:101200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
32.  Boyd LNC, Ali M, Kam L, Puik JR, Rodrigues SMF, Zwart ES, Daams F, Zonderhuis BM, Meijer LL, Le Large TYS, Giovannetti E, van Laarhoven HWM, Kazemier G. The Diagnostic Value of the CA19-9 and Bilirubin Ratio in Patients with Pancreatic Cancer, Distal Bile Duct Cancer and Benign Periampullary Diseases, a Novel Approach. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
33.  Le Large TYS, Meijer LL, Paleckyte R, Boyd LNC, Kok B, Wurdinger T, Schelfhorst T, Piersma SR, Pham TV, van Grieken NCT, Zonderhuis BM, Daams F, van Laarhoven HWM, Bijlsma MF, Jimenez CR, Giovannetti E, Kazemier G. Combined Expression of Plasma Thrombospondin-2 and CA19-9 for Diagnosis of Pancreatic Cancer and Distal Cholangiocarcinoma: A Proteome Approach. Oncologist. 2020;25:e634-e643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
34.  Bolm L, Petrova E, Weitz J, Rückert F, Wittel UA, Makowiec F, Lapshyn H, Bronsert P, Rau BM, Khatkov IE, Bausch D, Keck T, Wellner UF, Distler M. Prognostic relevance of preoperative bilirubin-adjusted serum carbohydrate antigen 19-9 in a multicenter subset analysis of 179 patients with distal cholangiocarcinoma. HPB (Oxford). 2019;21:1513-1519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
35.  Terasaki F, Sugiura T, Okamura Y, Ito T, Yamamoto Y, Ashida R, Ohgi K, Uesaka K. Systemic immune-inflammation index as a prognostic marker for distal cholangiocarcinoma. Surg Today. 2021;51:1602-1609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
36.  Liu D, Czigany Z, Heij LR, Bouwense SAW, van Dam R, Lang SA, Ulmer TF, Neumann UP, Bednarsch J. The Value of Platelet-to-Lymphocyte Ratio as a Prognostic Marker in Cholangiocarcinoma: A Systematic Review and Meta-Analysis. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
37.  Ji F, Kang Q, Wang L, Liu L, Ke Y, Zhu Y, Zhang N, Xiong S, Li Y, Zou H. Prognostic significance of the neutrophil-to-lymphocyte ratio with distal cholangiocarcinoma patients. Medicine (Baltimore). 2020;99:e22827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
38.  Oh C, Kim HJ, Song SH, Park EK, Hur YH, Koh YS, Cho CK. The prognostic value of the lymph node ratio in patients with distal cholangiocarcinoma after curative intended surgery: A single-center retrospective study. Ann Hepatobiliary Pancreat Surg. 2022;26:168-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  He Y, Liu H, Ma Y, Li J, Zhang J, Ren Y, Dong C, Bai B, Zhang Y, Lin Y, Yue P, Meng W. Preoperative prognostic nutritional index predicts short-term complications after radical resection of distal cholangiocarcinoma. Front Surg. 2022;9:1091534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
40.  Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol. 2022;22:173-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 262]  [Article Influence: 131.0]  [Reference Citation Analysis (0)]
41.  Houghton AM, Rzymkiewicz DM, Ji H, Gregory AD, Egea EE, Metz HE, Stolz DB, Land SR, Marconcini LA, Kliment CR, Jenkins KM, Beaulieu KA, Mouded M, Frank SJ, Wong KK, Shapiro SD. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat Med. 2010;16:219-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 457]  [Cited by in F6Publishing: 546]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
42.  Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer. 2020;20:485-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 537]  [Article Influence: 134.3]  [Reference Citation Analysis (0)]
43.  Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol. 2019;16:601-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 353]  [Article Influence: 70.6]  [Reference Citation Analysis (0)]
44.  Ugel S, Canè S, De Sanctis F, Bronte V. Monocytes in the Tumor Microenvironment. Annu Rev Pathol. 2021;16:93-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 116]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
45.  Gärtner K, Battke C, Dünzkofer J, Hüls C, von Neubeck B, Kellner MK, Fiestas E, Fackler S, Lang S, Zeidler R. Tumor-derived extracellular vesicles activate primary monocytes. Cancer Med. 2018;7:2013-2020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
46.  Lebegge E, Arnouk SM, Bardet PMR, Kiss M, Raes G, Van Ginderachter JA. Innate Immune Defense Mechanisms by Myeloid Cells That Hamper Cancer Immunotherapy. Front Immunol. 2020;11:1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Chen Y, Tian Z. Innate lymphocytes: pathogenesis and therapeutic targets of liver diseases and cancer. Cell Mol Immunol. 2021;18:57-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
48.  Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13:759-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1198]  [Cited by in F6Publishing: 1383]  [Article Influence: 125.7]  [Reference Citation Analysis (0)]
49.  Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6912]  [Cited by in F6Publishing: 7905]  [Article Influence: 494.1]  [Reference Citation Analysis (0)]
50.  van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20:218-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 755]  [Article Influence: 188.8]  [Reference Citation Analysis (0)]
51.  Zhao M, Duan X, Mi L, Shi J, Li N, Yin X, Han X, Wang J, Han G, Hou J, Yin F. Prognosis of hepatocellular carcinoma and its association with immune cells using systemic inflammatory response index. Future Oncol. 2022;18:2269-2288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
52.  Prijovic N, Acimovic M, Santric V, Stankovic B, Nikic P, Vukovic I, Soldatovic I, Nale D, Kovacevic L, Nale P, Marinkovic A, Babic U. Predictive Value of Inflammatory and Nutritional Indexes in the Pathology of Bladder Cancer Patients Treated with Radical Cystectomy. Curr Oncol. 2023;30:2582-2597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
53.  Li L, Li Z, Feng X, Yang Z, Jin N, Zhu L, Zang X, Xing L, Ren Y, Zhang H. Predictive value of systemic inflammatory response-related indices for survival in tongue cancer. Oral Dis. 2024;30:187-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
54.  Ye K, Xiao M, Li Z, He K, Wang J, Zhu L, Xiong W, Zhong Z, Tang Y. Preoperative systemic inflammation response index is an independent prognostic marker for BCG immunotherapy in patients with non-muscle-invasive bladder cancer. Cancer Med. 2023;12:4206-4217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
55.  Wang M, Li D, Chen R, Huang X, Li J, Liu Y, Liu J, Cheng W, Chen X, Zhao W, Li J, Tan Z, Huang H, Li D, Zhu F, Qin T, Ma J, Yu G, Zhou B, Zheng S, Tang Y, Han W, Meng L, Ke J, Feng F, Chen B, Yin X, Chen W, Ma H, Xu J, Liu Y, Lin R, Dong Y, Yu Y, Liu J, Zhang H, Qin R; Minimally Invasive Treatment Group in the Pancreatic Disease Branch of China's International Exchange and Promotion Association for Medicine and Healthcare (MITG-P-CPAM). Laparoscopic versus open pancreatoduodenectomy for pancreatic or periampullary tumours: a multicentre, open-label, randomised controlled trial. Lancet Gastroenterol Hepatol. 2021;6:438-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 121]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
56.  Ke J, Liu F, Ke J, Cai H, Liu Y, Ji B. Application of the superior mesenteric artery-first approach in laparoscopic pancreatoduodenectomy: A literature review. Heliyon. 2024;10:e27500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
57.  Chen L, Kong X, Wang Z, Wang X, Fang Y, Wang J. Pre-treatment systemic immune-inflammation index is a useful prognostic indicator in patients with breast cancer undergoing neoadjuvant chemotherapy. J Cell Mol Med. 2020;24:2993-3021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
58.  Chen L, Kong X, Wang Z, Wang X, Fang Y, Wang J. Pretreatment Systemic Inflammation Response Index in Patients with Breast Cancer Treated with Neoadjuvant Chemotherapy as a Useful Prognostic Indicator. Cancer Manag Res. 2020;12:1543-1567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
59.  Chen L, Yan Y, Zhu L, Cong X, Li S, Song S, Song H, Xue Y. Systemic immune-inflammation index as a useful prognostic indicator predicts survival in patients with advanced gastric cancer treated with neoadjuvant chemotherapy. Cancer Manag Res. 2017;9:849-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 53]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
60.  Jiang C, Zhang S, Qiao K, Xiu Y, Yu X, Huang Y. The Pretreatment Systemic Inflammation Response Index as a Useful Prognostic Factor is Better Than Lymphocyte to Monocyte Ratio in Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Clin Breast Cancer. 2022;22:424-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
61.  Razumilava N, Gores GJ. Classification, diagnosis, and management of cholangiocarcinoma. Clin Gastroenterol Hepatol. 2013;11:13-21.e1; quiz e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 191]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
62.  Brindley PJ, Bachini M, Ilyas SI, Khan SA, Loukas A, Sirica AE, Teh BT, Wongkham S, Gores GJ. Cholangiocarcinoma. Nat Rev Dis Primers. 2021;7:65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 298]  [Article Influence: 99.3]  [Reference Citation Analysis (0)]
63.  Ho CS, Warkentin AE. Evidence-based decompression in malignant biliary obstruction. Korean J Radiol. 2012;13 Suppl 1:S56-S61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
64.  Li Z, Chu J, Su F, Ding X, Zhang Y, Dou L, Liu Y, Ke Y, Liu X, Liu Y, Wang G, Wang L, He S. Characteristics of bile microbiota in cholelithiasis, perihilar cholangiocarcinoma, distal cholangiocarcinoma, and pancreatic cancer. Am J Transl Res. 2022;14:2962-2971.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Nishimura M, Naka S, Hanazawa K, Tani T, Fukami M, Okada S, Fujiyama Y. Cholangiocarcinoma in the distal bile duct: a probable etiologic association with choledocholithiasis. Dig Dis Sci. 2005;50:2153-2158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
66.  Chen B, Fu SW, Lu L, Zhao H. A Preliminary Study of Biliary Microbiota in Patients with Bile Duct Stones or Distal Cholangiocarcinoma. Biomed Res Int. 2019;2019:1092563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
67.  Lavu S, Therneau TM, Harmsen WS, Mara KC, Wongjarupong N, Hassan M, Ali HA, Antwi S, Giama NH, Miyabe K, Roberts LR. Effect of Statins on the Risk of Extrahepatic Cholangiocarcinoma. Hepatology. 2020;72:1298-1309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
68.  Burr NE, Talboys RJ, Savva S, Clark A, Phillips M, Metcalfe M, Dennison A, Robinson R, Lewis MP, Rhodes M, Rushbrook S, Hart AR. Aspirin may prevent cholangiocarcinoma: a case-control study from the United kingdom. Dig Dis Sci. 2014;59:1567-1572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
69.  Kim SH, Lee B, Hwang HK, Lee JS, Han HS, Lee WJ, Yoon YS, Kang CM. Comparison of postoperative complications and long-term oncological outcomes in minimally invasive versus open pancreatoduodenectomy for distal cholangiocarcinoma: A propensity score-matched analysis. J Hepatobiliary Pancreat Sci. 2022;29:329-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
70.  Macfie R, Berger Y, Liu H, Li T, Imtiaz S, Ang C, Sarpel U, Hiotis S, Labow D, Golas B, Cohen NA. Major Postoperative Complications Limit Adjuvant Therapy Administration in Patients Undergoing Pancreatoduodenectomy for Distal Cholangiocarcinoma or Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol. 2023;30:5027-5034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]