Next Article in Journal
Electrochemical Biosensors in the Diagnosis of Acute and Chronic Leukemias
Next Article in Special Issue
Isolated Pancreatic Metastases of Renal Cell Carcinoma—Clinical Particularities and Seed and Soil Hypothesis
Previous Article in Journal
Cell-Population Dynamics in Diffuse Gliomas during Gliomagenesis and Its Impact on Patient Survival
Previous Article in Special Issue
Clockwise Anterior-to-Posterior—Double Isolation (CAP-DI) Approach for Portal Lymphadenectomy in Biliary Tract Cancer: Technique, Yield, and Outcomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effect of Perioperative Blood Transfusions and Infectious Complications on Inflammatory Activation and Long-Term Survival Following Gastric Cancer Resection

1
Service of Surgery, Hospital Universitari Mútua Terrassa, 08221 Terrassa, Spain
2
Department of Surgery, Hospital Universitari de Bellvitge, L’Hospitalet del Llobregat, 08037 Barcelona, Spain
3
Service of Internal Medicine, Hospital de Sant Joan Despí Moisès Broggi, 08970 Sant Joan Despí, Spain
4
Service of Surgery, Hospital Universitario de Navarra, 31008 Pamplona, Spain
5
Service of Surgery, Hospital Universitari Arnau de Vilanova, 25198 Lleida, Spain
6
Service of Surgery, Hospital Universitari Josep Trueta, 17007 Girona, Spain
7
Service of Surgery, Hospital de Sant Joan Despí Moisès Broggi, 08970 Sant Joan Despí, Spain
8
Service of Surgery, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
9
Service of Surgery, Consorci Corporació Sanitària Parc Taulí de Sabadell, 08208 Sabadell, Spain
10
Service of Surgery, Hospital General de Granollers, 08402 Granollers, Spain
11
Service of Surgery, Hospital Universitari de Tarragona, Joan XXIII, 43005 Tarragona, Spain
12
Service of Surgery, Hospital Universitari de Vic, 08500 Vic, Spain
13
Service of Surgery, Hospital de Mataró, Consorci Sanitari del Maresme, 08304 Mataró, Spain
14
Section of Gastrointestinal Surgery, Hospital Universitario del Mar, Universitat Autònoma de Barcelona, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Noelia Puértolas and Javier Osorio contributed equally to the study and share first authorship.
Cancers 2023, 15(1), 144; https://doi.org/10.3390/cancers15010144
Submission received: 6 December 2022 / Accepted: 23 December 2022 / Published: 26 December 2022
(This article belongs to the Special Issue Diagnosis and Therapeutic Management of Gastrointestinal Cancers)

Abstract

:

Simple Summary

Both postoperative complications and perioperative blood transfusions have been separately related to worse prognosis after gastrectomy for patients with gastric cancer. This multicenter cohort study aims to evaluate their synergic effect on inflammatory activation and prognosis. Patients were classified into four groups based on their perioperative course: one, no blood transfusion and no infectious complication; two, blood transfusion; three, infectious complication; four, both transfusion and infectious complication. The analysis shows that perioperative blood transfusion and infectious complications have a synergic effect creating a pro-inflammatory activation that favors tumor recurrence. These findings reinforce the need of promoting restrictive policies of transfusion for patients undergoing gastrectomies implementing patients blood management programs.

Abstract

Background: The aim of this study was to evaluate the impact of perioperative blood transfusion and infectious complications on postoperative changes of inflammatory markers, as well as on disease-free survival (DFS) in patients undergoing curative gastric cancer resection. Methods: Multicenter cohort study in all patients undergoing gastric cancer resection with curative intent. Patients were classified into four groups based on their perioperative course: one, no blood transfusion and no infectious complication; two, blood transfusion; three, infectious complication; four, both transfusion and infectious complication. Neutrophil-to-lymphocyte ratio (NLR) was determined at diagnosis, immediately before surgery, and 10 days after surgery. A multivariate Cox regression model was used to analyze the relationship of perioperative group and dynamic changes of NLR with disease-free survival. Results: 282 patients were included, 181 in group one, 23 in group two, 55 in group three, and 23 in group four. Postoperative NLR changes showed progressive increase in the four groups. Univariate analysis showed that NLR change > 2.6 had a significant association with DFS (HR 1.55; 95% CI 1.06–2.26; p = 0.025), which was maintained in multivariate analysis (HR 1.67; 95% CI 1.14–2.46; p = 0.009). Perioperative classification was an independent predictor of DFS, with a progressive difference from group one: group two, HR 0.80 (95% CI: 0.40–1.61; p = 0.540); group three, HR 1.42 (95% CI: 0.88–2.30; p = 0.148), group four, HR 2.85 (95% CI: 1.64–4.95; p = 0.046). Conclusions: Combination of perioperative blood transfusion and infectious complications following gastric cancer surgery was related to greater NLR increase and poorer DFS. These findings suggest that perioperative blood transfusion and infectious complications may have a synergic effect creating a pro-inflammatory activation that favors tumor recurrence.

1. Introduction

Gastric cancer (GC) is one of the most common malignancies worldwide and the fourth leading cause of cancer-related death [1]. Surgery remains the main treatment for GC. Oncologic gastrectomies are complex procedures associated with high morbidity rates, ranging from 20 to 45% in Western countries [2]. Moreover, patients undergoing GC resection are at high risk of receiving blood transfusion in the perioperative period due to a high prevalence of anemia [3]. Perioperative blood transfusion leads to a higher incidence of postoperative complications, especially infectious [4]. Both postoperative complications and perioperative blood transfusions have been separately related to worse prognosis in patients with GC, probably due to postoperative cellular immune suppression and inflammatory activation [5,6,7,8]. However, there is a lack of information available to understand if the effect of transfusions and infections on inflammatory activation and tumor recurrence is independent and additive, or mutually related and synergic.
In recent years, there has been increasing concern regarding the association between the systemic inflammatory response markers and survival in patients with various types of cancer [9,10,11,12,13]. Alterations in neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio have been related to higher tumor recurrence and worse long-term survival [14,15,16,17]. These inflammatory markers have mostly been determined before surgery, but some recent studies have shown that postoperative values permit a better prognostic prediction [18,19,20]. However, this pro-inflammatory activation following GC surgery has not been evaluated in relation to two potential causes: postoperative infectious complications and perioperative blood transfusions.
Patient blood management (PBM) programs include different evidence-based interventions addressed to maintain patients’ own blood mass and avoid unnecessary transfusions [21,22]. The Spanish EURECCA Esophagogastric Cancer Group recently evaluated the clinical impact of the implementation of a PBM program in gastric cancer surgery, proving a reduction in transfusion rate, infectious complications, and postoperative 90-day mortality [23]. The current prospective study aimed to investigate the dynamic changes of inflammatory markers (NLR) following curative GC surgery, their association with perioperative blood transfusion and postoperative infectious complications, and their impact on disease-free survival.

2. Materials and Methods

2.1. Study Design and Participants

A prospective multicenter study was conducted in consecutive patients undergoing elective GC resection with curative intent in 12 hospitals of the Spanish EURECCA Esophagogastric Cancer Group between January 2017 and December 2018 (ClinicalTrials.gov NCT04286984). The study was approved by the ethics committee of the Institutional Review Board of University Hospital Mutua Terrassa, and written informed consent was obtained from all patients. The study was carried out according to the guidelines of the Declaration of Helsinki. The paper has been reported in line with the STROCSS criteria [24].
Patients with non-epithelial tumors, distant metastases, evidence at diagnosis of pre-existing infections or inflammatory conditions (such as vasculitis or rheumatoid arthritis), or who were not willing to sign the informed consent, were excluded.

2.2. Data Collection

Clinicopathological data and follow-up status for all patients were collected from a maintained database, which was common to all institutions. Ninety variables with detailed definitions were continuously collected from each patient by the reference surgeon at each institution. Validation of data registration (period 2014–2018) in the EURECCA dataset was recently performed, revealing 97% completeness and 95% accuracy rates [25].
For each patient, recorded data included: demographics (age, sex); category of the American Society of Anesthesiologists (ASA) physical status classification system; body mass index (BMI); Eastern Cooperative Oncology Group (ECOG) performance status [26]; percentage of unintended weight loss 6 months before surgery; Charlson comorbidity index score [27], (categorized as 0–2, and ≥3); tumor location; pTNM stage (8th edition, UICC) [28]; neoadjuvant treatment; type of gastrectomy (distal subtotal or total); extension of lymphadenectomy according to the Japanese Gastric Cancer Association Classification [29]; surgical approach (open or minimally invasive); radicality, and associated multivisceral resection.
Blood samples were collected at diagnosis, immediately before and 10 days after surgery, or at the time of hospital discharge if it occurred earlier. Perioperative blood transfusion, postoperative complications, neutrophil and lymphocyte count, 30-day hospital readmission, tumor recurrence, and 90-day mortality were recorded.

2.3. Outcomes and Definitions

The primary endpoint of the study was to evaluate the effect of infectious complications and perioperative transfusion on disease-free survival (DFS). The secondary outcome was to evaluate their association with perioperative changes in NLR, establishing a cut-off for the change that could show an impact on DFS.
Postoperative infectious complications occurring within the first 30 days after surgery were defined according to the Gastrectomy Complications Consensus Group (GCCG) [30] and graded with the Clavien-Dindo classification [31]. Intra-abdominal infections included anastomotic leakage (defined as full thickness gastrointestinal defect involving the anastomosis), duodenal stump fistula (full thickness duodenal defect or abscess close duodenal stump), pancreatic fistula (drain output of any measurable volume of fluid with an amylase level > 3 times the upper limit of institutional normal serum amylase activity, associated with relevant symptomatology), and intra-abdominal abscess (other postoperative abnormal fluid from drainage and/or abdominal collections without gastrointestinal leaks preventing drainage removal and/or requiring treatment). Other infections were either gastrointestinal, respiratory, urinary, or other, with both symptoms and germ isolation.
Perioperative blood transfusion was defined as transfusion of allogenic red blood cells from 30 days before surgery until hospital discharge after surgery.
In order to study both the separated and associated effects of transfusions and infectious complications on inflammatory activation and long-term outcomes, four groups of patients were defined according to their perioperative course: group one: no perioperative blood transfusion and no infectious complication; group two: perioperative blood transfusion; group three: postoperative infectious complication; group four: both blood transfusion and infectious complication.
Disease-free survival was defined as the time from the date of surgery to recurrence of tumor or death. The latest follow-up date was March 2021.

2.4. Statistical Analysis

Descriptive statistics were used to summarize population characteristics. To compare the demographic and clinical profile between the 4 groups, a Chi-square test was used for categorical variables, and an ANOVA or a Kruskall–Wallis test (according to variable distribution) to compare continuous variables. NLR changes were evaluated with the ANOVA test. The paired-sample t test was used to determine statistically significant changes in pre- and postoperative levels of inflammatory markers. NLR change was calculated by subtracting the preoperative NLR value from the postoperative one. To dichotomize the NLR change, the R package survMisc was used to calculate the optimal cut-off point for a continuous variable when a Cox regression model is used. The Contal–O’Quigley method was used to choose the best cut-off point. The Kaplan–Meier method was used to estimate survival probabilities of the 4 perioperative groups and the 2 groups defined by the NLR change. A multivariate analysis was used to assess the effect of variates on DFS. A Cox regression model was used to identify the potentially causal effect of group on NLR and DFS. Adjustment variables were entered into a multivariate Cox regression model according to their clinical relevance. Results were expressed as hazard ratios (HRs) with 95% confidence intervals (CIs). The SPSS software package (SPSS, Chicago, IL, USA) was used to manage data and perform statistical analysis.

3. Results

3.1. Baseline Variables

A total of 384 patients underwent GC resection with curative intent in the 12 participating hospitals between January 2017 and December 2018. After exclusion of 26 patients with metastatic disease detected at surgery, 45 patients with definitive histology of non-epithelial tumor, and 31 not having the three blood samples, 282 patients were included for final analysis. Fifty patients (17.7%) received perioperative blood transfusion. A total of 88 postoperative infectious complications were developed in 81 (28.7%) patients. Depending on their perioperative evolution, patients were grouped as follows: group one (no transfusion nor infectious complication): n = 181; group two (blood transfusion): n = 23; group three (infectious complication), n = 55; group four (both transfusion and infectious complication), n = 23. Characteristics of these four groups are summarized in Table 1. Types of postoperative complications are detailed in Table 2. Blood transfusions were associated with anemia, and cardiac and renal pathologies; infectious complications were significantly more frequent in patients who underwent total gastrectomies. Both transfusion and infectious complications were related with nodal involvement. The rest of the basal variables did not show significant differences between the four groups of patients.

3.2. Dynamic Changes of Inflammatory Markers

Postoperative NLR change showed a progressive increase in the four groups, ranging from +1.5 in group one to +7.7 in group four; global difference among groups was significant (p < 0.001), as well as between groups one and three (p < 0.001) and groups one and four (p < 0.001) (Figure 1).

3.3. Effect of Postoperative Infectious Complications and Blood Transfusion on DFS

The median follow-up was 27.2 months (range 14.6–39.6). At the end of the study, 100 (35.5%) patients had died. Among the 182 patients who survived, 19 had a GC recurrence. DFS showed a progressive difference from group one to group four, as shown in Figure 2. Kaplan–Meier analysis’ log-rank test was significant (p = 0.0013); pairwise comparisons showed a significant difference between group one and four (p = 0.0004), with the rest of the differences being non-significant. In univariate analysis, the perioperative groups, the ECOG score, percentage of weight loss, surgical radicality, and pT and pN categories were found to be associated with poor DFS (Table 3). In multivariate analysis, independent prognostic factors were the perioperative group, the ECOG score, surgical radicality, and pN.

3.4. Effect of Inflammatory Changes on DFS

The best cut-off value for the NLR change after surgery was 2.6. As shown in Figure 3, patients with NLR change > 2.6 had worse long-term prognosis. In univariate analysis, NLR change > 2.6 had a statistically significant association with poorer DFS (HR 1.55; 95% CI 1.06–2.26; p = 0.025), which was maintained in multivariate analysis (HR 1.67; 95% CI 1.14–2.46; p = 0.009) (Table 3).
Table 3. Univariate and multivariate analyses of clinicopathological variables in relation to disease-free survival.
Table 3. Univariate and multivariate analyses of clinicopathological variables in relation to disease-free survival.
Clinicopathological FeaturesUnivariate Analysis
HR (95% CI)
p RatioMultivariate Analysis
HR (95% CI)
p Ratio
Perioperative classification, (%):
1: Trans(−)/Inf(−)Ref.Ref.Ref.Ref.
2: Trans(+)/Inf(−)1.36 [0.70; 2.67]0.3640.80 [0.40; 1.61]0.540
3: Trans(−)/Inf(+)1.51 [0.94; 2.42]0.0861.42 [0.88; 2.30]0.148
4: Trans(+)/Inf(+)2.85 [1.64; 4.95]<0.0011.77 [1.01–3.11]0.046
NLR difference
<2.6Ref.Ref.Ref.Ref.
≥2.61.55 [1.06; 2.26]0.0251.67 [1.14; 2.46]0.009
Weight loss
0–5%Ref.Ref. (34.1%)
6–10%1.66 [1.08; 2.55]0.022
>10%1.99 [1.20; 3.30]0.008 (55.6%)
Radicality
R0Ref.Ref. (37.6%)Ref.Ref.
R1–R22.32 [1.38; 3.89]0.001(70.8%)2.15 [1.26; 3.69]0.005
pN a
N0Ref.Ref. (19.7%)Ref.Ref.
≥N13.41 [2.17; 5.36]<0.001 (56.2%)2.89 [1.75; 4.76]<0.001
pT a
≤T2Ref.Ref. (51.6%)Ref.Ref.
>T30.47 [0.31; 0.70]<0.001 (25.6%)0.87 [0.54; 1.40]0.561
ECOG b
ECOG 0Ref.Ref. (25.8%)Ref.Ref.
ECOG ≥ 12.49 [1.59; 3.92]<0.001 (48.1%)2.04 [1.29; 3.24]0.002
ASA
ASA I/IIRef.Ref. (39.0%)
ASA III/IV1.25 [0.86; 1.81]0.242 (41.8%)
ASA, American Society of Anesthesiologists; CI, confidence interval; HR, hazard ratio; NLR (neutrophil-to-lymphocyte ratio); a According to 8th edition of the International Union Against Cancer tumor node metastasis staging system [27]; b Performance status according to Eastern Cooperative Oncology Group (ECOG) [25].

4. Discussion

This prospective cohort study of patients undergoing GC resection with curative intent demonstrated a synergic effect of both perioperative blood transfusion and infectious complications on long-term prognosis. Previous studies have separately studied the impact of complications and blood transfusion following GC resection on DFS, with heterogeneous results: some studies support a negative effect of anastomotic leakage, postoperative infection, or intra-abdominal infection in prognosis [32,33,34,35], while others did not find that relation [36]; moreover, the adverse prognostic effect of perioperative blood transfusion has also been confirmed in many studies [37,38,39,40], including some systematic reviews and meta-analyses [41,42,43,44], while other studies did not find this association [45,46,47]. To date, only two studies, one of patients with colorectal cancer [48], and another with locally advanced gastric cancer from Xiao et al. [49] (with no determination of inflammatory markers), have analyzed the combined effect of perioperative blood transfusion and postoperative infectious complications, suggesting their additive negative association with worse cancer specific survival. In univariate analysis of the present study, perioperative transfusions and infections could also seem to have an additive effect on DFS, as the sum of their HR is like the one deriving from their combination. However, looking at the results of multivariate analysis, it seems more likely to think of a synergetic effect of both variables, which would potentiate their individual effect on DFS. If we consider that both variables are also causally related, as perioperative blood transfusion can also favor postoperative infections, our findings support the need for promoting PBM policies in GC surgery, in order to minimize unnecessary transfusions [50].
Inflammatory ratios showed a postoperative increase that was significantly greater when perioperative blood transfusion and infectious complications occurred simultaneously. In addition, NLR postoperative change > 2.6 showed a good predictive accuracy for determining DFS. This finding could have some clinical implications, as patients with a higher postoperative NLR increase may potentially benefit from adjuvant treatment and/or a closer long-term follow-up. Various studies have confirmed the prognostic usefulness of preoperative inflammatory markers in patients with GC, with different cut-off points [15,16,51,52,53]. Those studies focused only on the initial values and not on the dynamic changes after surgery. In contrast, other authors have reported that postoperative inflammatory markers, especially NLR, behaved as better prognostic predictors [18,19,20]. A recent study retrospectively analyzed the dynamic changes of inflammatory markers in patients who underwent radical gastrectomy, showing a significantly improved prognostic accuracy by incorporating the post-12-month lymphocyte–monocyte ratio in the TNM staging system [19]. However, in all those studies, postoperative inflammatory activation was not related to its most probable causes: perioperative blood transfusion and postoperative infectious complications.
This study has some limitations, including its reduced sample size, particularly in perioperative groups two and four. Prospective studies with more patients will help determine the relationship of postoperative adverse events with inflammatory activation and prognosis with greater precision. In addition, the lack of previous consensus on the optimal cut-off value for inflammatory markers has made comparison of results difficult. Due to the growing evidence of the prognostic relevance of inflammatory markers, an effort is needed to establish their optimal categories in order to assist in clinical decision making. Additionally, heterogeneity in the chronological definition of perioperative blood transfusion in the literature adds difficulty to benchmarking.

5. Conclusions

In conclusions, this study shows that blood transfusion and infectious complications may have a synergic impact on short-term increase in NLR and poorer long-term outcomes in patients undergoing gastric cancer surgery. As infectious complications are favored by perioperative transfusions, these findings reinforce the need of promoting PBM policies in order to minimize pro-inflammatory activation immediately after surgery that may favor tumor recurrence.

Author Contributions

N.P., J.O. and C.J. were the principal investigators and contributed to design of the study, data collection, data interpretation and writing of the manuscript. G.G., A.A. and A.L. contributed to PBM protocol design and implementation. C.M., M.S., E.A., E.G., C.O., N.P., J.M., M.G. and L.P. contributed to data collection and critical review of the manuscript. M.P. contributed to design of the study, data collection and manuscript correction. All authors have read and agreed to the published version of the manuscript.

Funding

The project IPAT has been awarded with a research grant from the Catalan Society of Surgery (Spain) 2017-1.

Institutional Review Board Statement

The study protocol was approved by the Clinical Research Ethics Committee of the leading and collaborating centers (Ethics and Clinical Investigation Committee of the University Hospital Mutua Terrassa, code EO/1631, date of approval 24/05/16.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors thank all the professionals who have participated in the registration and analysis of the data. This study is part of a doctoral programme of the Department of Surgery at Universitat Autònoma de Barcelona.

Conflicts of Interest

J.O. has received honoraria for lectures from Vifor Pharma España SL, but not for this work. C.J. has received honoraria for lectures/consultancies from Vifor Pharma España SL, Bial, and Zambon, but not for this work. For the remaining authors, there are no conflicts of interest to be declared.

References

  1. Machlowska, J.; Baj, J.; Sitarz, M.; Sitarz, R.M. Gastric Cancer: Epidemiology, Risk Factors, Classification, Genomic Characteristics and Treatment Strategies. Int. J. Mol. Sci. 2020, 21, 4012. [Google Scholar] [CrossRef] [PubMed]
  2. Baiocchi, G.L.; Giacopuzzi, S.; Reim, D.; Piessen, G.; da Costa, P.M.; Reynolds, J.V.M.; Meyer, H.-J.; Morgagni, P.; Gockel, I.M.; Santos, L.L.; et al. Incidence and Grading of Complications After Gastrectomy for Cancer Using the GASTRODATA Registry: A European Retrospective Observational Study. Ann. Surg. 2020, 272, 807–813. [Google Scholar] [CrossRef] [PubMed]
  3. Pirker, R.; Wiesenberger, K.; Minar, W. Cancer-related anemia. Clinical relevance and treatment strategies. Am. J. Cancer 2005, 4, 233–245. [Google Scholar] [CrossRef]
  4. Xiao, H.; Quan, H.; Pan, S.; Yin, B.; Luo, W.; Huang, G.; Ouyang, Y. Impact of peri-operative blood transfusion on post-operative infections after radical gastrectomy for gastric cancer: A propensity score matching analysis focusing on the timing, amount of transfusion and role of leukocyte depletion. J. Cancer Res. Clin. Oncol. 2018, 144, 1143–1154. [Google Scholar] [CrossRef] [Green Version]
  5. Sun, C.; Wang, Y.; Yao, H.; Hu, Z. Allogeneic blood transfusion and the prognosis of gastric cancer patients: Systematic review and meta-analysis. Int. J. Surg. 2015, 13, 102–110. [Google Scholar] [CrossRef] [PubMed]
  6. Squires, M.H.; Kooby, D.A.; Poultsides, G.A.; Weber, S.M.; Bloomston, M.; Fields, R.C.; Pawlik, T.M.; Votanopoulos, K.I.; Schmidt, C.R.; Ejaz, A.; et al. Effect of Perioperative Transfusion on Recurrence and Survival after Gastric Cancer Resection: A 7-Institution Analysis of 765 Patients from the US Gastric Cancer Collaborative. J. Am. Coll. Surg. 2015, 221, 767–777. [Google Scholar] [CrossRef]
  7. Yuan, P.; Wu, Z.; Li, Z.; Bu, Z.; Wu, A.; Wu, X.; Zhang, L.; Shi, J.; Ji, J. Impact of postoperative major complications on long-term survival after radical resection of gastric cancer. BMC Cancer 2019, 19, 1–9. [Google Scholar] [CrossRef] [Green Version]
  8. Snyder, G.L.; Greenberg, S. Effect of anaesthetic technique and other perioperative factors on cancer recurrence. Br. J. Anaesth. 2010, 105, 106–115. [Google Scholar] [CrossRef] [Green Version]
  9. Cata, J.; Wang, H.; Gottumukkala, V.; Reuben, J.; Sessler, D. Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions. Br. J. Anaesth. 2013, 110, 690–701. [Google Scholar] [CrossRef] [Green Version]
  10. Hirahara, N.; Matsubara, T.; Mizota, Y.; Ishibashi, S.; Tajima, Y. Prognostic value of preoperative inflammatory response biomarkers in patients with esophageal cancer who undergo a curative thoracoscopic esophagectomy. BMC Surg. 2016, 16, 1–12. [Google Scholar] [CrossRef]
  11. Qu, J.; Qu, X.; Li, Z.; Zhang, J.; Teng, Y.; Jin, B.; Zhao, M.; Yu, P.; Wang, Z.; Liu, Y. Role of patient-, tumor- and systemic inflammatory response–related factors in predicting survival of patients with node-negative gastric cancer. Tumor Biol. 2017, 39, 101042831769837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yang, R.; Chang, Q.; Meng, X.; Gao, N.; Wang, W. Prognostic value of Systemic immune-inflammation index in cancer: A meta-analysis. J. Cancer 2018, 9, 3295–3302. [Google Scholar] [CrossRef] [PubMed]
  13. Zheng, L.; Zou, K.; Yang, C.; Chen, F.; Guo, T.; Xiong, B. Inflammation-based indexes and clinicopathologic features are strong predictive values of preoperative circulating tumor cell detection in gastric cancer patients. Clin. Transl. Oncol. 2017, 19, 1125–1132. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, X.; Huang, Y.; Feng, J.; Liu, J. Prognostic significance of neutrophil-to-lymphocyte ratio in esophageal cancer: A meta-analysis. OncoTargets Ther. 2015, 8, 789–794. [Google Scholar] [CrossRef] [Green Version]
  15. Sun, X.; Liu, X.; Liu, J.; Chen, S.; Xu, D.; Li, W.; Zhan, Y.; Li, Y.; Chen, Y.; Zhou, Z. Preoperative neutrophil-to-lymphocyte ratio plus platelet-to-lymphocyte ratio in predicting survival for patients with stage I–II gastric cancer. Chin. J. Cancer 2016, 35, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kim, E.Y.; Lee, J.W.; Yoo, H.M.; Park, C.H.; Song, K.Y. The Platelet-to-Lymphocyte Ratio Versus Neutrophil-to-Lymphocyte Ratio: Which is Better as a Prognostic Factor in Gastric Cancer? Ann. Surg. Oncol. 2015, 22, 4363–4370. [Google Scholar] [CrossRef]
  17. Pan, Q.-X.; Su, Z.-J.; Zhang, J.-H.; Wang, C.-R.; Ke, S.-Y. A comparison of the prognostic value of preoperative inflammation-based scores and TNM stage in patients with gastric cancer. OncoTargets Ther. 2015, 8, 1375–1385. [Google Scholar] [CrossRef] [Green Version]
  18. Min, K.-W.; Kwon, M.J.; Kim, D.-H.; Son, B.K.; Kim, E.-K.; Oh, Y.H.; Wi, Y.C. Persistent elevation of postoperative neutrophil-to-lymphocyte ratio: A better predictor of survival in gastric cancer than elevated preoperative neutrophil-to-lymphocyte ratio. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  19. Lin, J.-X.; Wang, Z.-K.; Huang, Y.-Q.; Xie, J.-W.; Wang, J.-B.; Lu, J.; Chen, Q.-Y.; Lin, M.; Tu, R.-H.; Huang, Z.-N.; et al. Dynamic Changes in Pre- and Postoperative Levels of Inflammatory Markers and Their Effects on the Prognosis of Patients with Gastric Cancer. J. Gastrointest. Surg. 2020, 25, 387–396. [Google Scholar] [CrossRef] [Green Version]
  20. Kim, E.Y.; Song, K.Y. The preoperative and the postoperative neutrophil-to-lymphocyte ratios both predict prognosis in gastric cancer patients. World J. Surg. Oncol. 2020, 18, 1–8. [Google Scholar] [CrossRef]
  21. Thomson, A.; Farmer, S.; Hofmann, A.; Isbister, J.; Shander, A. Patient blood management—A new paradigm for transfusion medicine? ISBT Sci. Ser. 2009, 4, 423–435. [Google Scholar] [CrossRef] [PubMed]
  22. Shander, A.; Hofmann, A.; Isbister, J.; Van Aken, H. Patient blood management—The new frontier. Best Pract. Res. Clin. Anaesthesiol. 2013, 27, 5–10. [Google Scholar] [CrossRef] [PubMed]
  23. Osorio, J.; Jericó, C.; Miranda, C.; Santamaría, M.; Artigau, E.; Galofré, G.; Garsot, E.; Luna, A.; Puértolas, N.; Aldeano, A.; et al. Improved postoperative outcomes and reduced transfusion rates after implementation of a Patient Blood Management program in gastric cancer surgery. Eur. J. Surg. Oncol. (EJSO) 2020, 47, 1449–1457. [Google Scholar] [CrossRef] [PubMed]
  24. Agha, R.; Abdall-Razak, A.; Crossley, E.; Dowlut, N.; Iosifidis, C.; Mathew, G.; Beamishaj; Bashashati, M.; Millham, F.H.; Orgill, D.P.; et al. STROCSS 2019 Guideline: Strengthening the reporting of cohort studies in surgery. Int. J. Surg. 2019, 72, 156–165. [Google Scholar] [CrossRef]
  25. Cero, M.D.; Rodríguez-Santiago, J.; Miró, M.; Castro, S.; Miranda, C.; Santamaría, M.; Gobbini, Y.; Garsot, E.; Pujadas, M.; Luna, A.; et al. Evaluation of data quality in the Spanish EURECCA Esophagogastric Cancer Registry. Eur. J. Surg. Oncol. (EJSO) 2021, 47, 3081–3087. [Google Scholar] [CrossRef]
  26. Kelly, C.M.; Shahrokni, A. Moving beyond Karnofsky and ECOG Performance Status Assessments with New Technologies. J. Oncol. 2016, 2016, 1–13. [Google Scholar] [CrossRef] [Green Version]
  27. Charlson, M.E.; Pompei, P.; Ales, K.L.; MacKenzie, C.R. A new method of classifying prognostic comorbidity in longitudinal studies: Development and validation. J. Chronic Dis. 1987, 40, 373–383. [Google Scholar] [CrossRef]
  28. In, H.; Solsky, I.; Palis, B.; Langdon-Embry, M.; A Ajani, J.; Sano, T. Validation of the 8th Edition of the AJCC TNM Staging System for Gastric Cancer using the National Cancer Database. Ann. Surg. Oncol. 2017, 24, 3683–3691. [Google Scholar] [CrossRef]
  29. Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer 2017, 20, 1–19. [Google Scholar] [CrossRef] [Green Version]
  30. Baiocchi, G.L.; Giacopuzzi, S.; Marrelli, D.; Reim, D.; Piessen, G.; da Costa, P.M.; Reynolds, J.V.; Meyer, H.-J.; Morgagni, P.; Gockel, I.; et al. International consensus on a complications list after gastrectomy for cancer. Gastric Cancer 2018, 22, 172–189. [Google Scholar] [CrossRef]
  31. Clavien, P.A.; Barkun, J.; de Oliveira, M.L.; Vauthey, J.N.; Dindo, D.; Schulick, R.D.; de Santibañes, E.; Pekolj, J.; Slankamenac, K.; Bassi, C.; et al. The Clavien-Dindo Classification of Surgical Complications: Five-year experience. Ann. Surg. 2009, 250, 187–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tsujimoto, H.; Ichikura, T.; Ono, S.; Sugasawa, H.; Hiraki, S.; Sakamoto, N.; Yaguchi, Y.; Yoshida, K.; Matsumoto, Y.; Hase, K. Impact of Postoperative Infection on Long-Term Survival After Potentially Curative Resection for Gastric Cancer. Ann. Surg. Oncol. 2008, 16, 311–318. [Google Scholar] [CrossRef] [PubMed]
  33. Yoo, H.M.; Lee, H.H.; Shim, J.H.; Jeon, H.M.; Park, C.H.; Song, K.Y. Negative impact of leakage on survival of patients undergoing curative resection for advanced gastric cancer. J. Surg. Oncol. 2011, 104, 734–740. [Google Scholar] [CrossRef] [PubMed]
  34. Li, Q.-G. Impact of postoperative complications on long-term survival after radical resection for gastric cancer. World J. Gastroenterol. 2013, 19, 4060–4065. [Google Scholar] [CrossRef] [PubMed]
  35. Tokunaga, M.; Kurokawa, Y.; Machida, R.; Sato, Y.; Takiguchi, S.; Doki, Y.; Yabusaki, H.; Watanabe, M.; Hato, S.; Nakamori, M.; et al. Impact of postoperative complications on survival outcomes in patients with gastric cancer: Exploratory analysis of a randomized controlled JCOG1001 trial. Gastric Cancer 2020, 24, 214–223. [Google Scholar] [CrossRef]
  36. Climent, M.; Hidalgo, N.; Vidal, Ó.; Puig, S.; Iglesias, M.; Cuatrecasas, M.; Ramón, J.; García-Albéniz, X.; Grande, L.; Pera, M. Postoperative complications do not impact on recurrence and survival after curative resection of gastric cancer. Eur. J. Surg. Oncol. (EJSO) 2015, 42, 132–139. [Google Scholar] [CrossRef] [Green Version]
  37. Nakanishi, K.; Kanda, M.; Kodera, Y. Long-lasting discussion: Adverse effects of intraoperative blood loss and allogeneic transfusion on prognosis of patients with gastric cancer. World J. Gastroenterol. 2019, 25, 2743–2751. [Google Scholar] [CrossRef]
  38. Mizuno, A.; Kanda, M.; Kobayashi, D.; Tanaka, C.; Iwata, N.; Yamada, S.; Fujii, T.; Nakayama, G.; Sugimoto, H.; Koike, M.; et al. Adverse Effects of Intraoperative Blood Loss on Long-Term Outcomes after Curative Gastrectomy of Patients with Stage II/III Gastric Cancer. Dig. Surg. 2016, 33, 121–128. [Google Scholar] [CrossRef]
  39. Ito, Y.; Kanda, M.; Ito, S.; Mochizuki, Y.; Teramoto, H.; Ishigure, K.; Murai, T.; Asada, T.; Ishiyama, A.; Matsushita, H.; et al. Intraoperative Blood Loss is Associated with Shortened Postoperative Survival of Patients with Stage II/III Gastric Cancer: Analysis of a Multi-institutional Dataset. World J. Surg. 2018, 43, 870–877. [Google Scholar] [CrossRef]
  40. Hsu, F.-K.; Chang, W.-K.; Lin, K.-J.; Liu, C.-Y.; Fang, W.-L.; Chang, K.-Y. The Associations between Perioperative Blood Transfusion and Long-Term Outcomes after Stomach Cancer Surgery. Cancers 2021, 13, 5438. [Google Scholar] [CrossRef]
  41. Li, L.; Zhu, D.; Chen, X.; Huang, Y.; Ouyang, M.; Zhang, W. Perioperative Allogenenic Blood Transfusion is Associated With Worse Clinical Outcome for Patients Undergoing Gastric Carcinoma Surgery. Medicine 2015, 94, e1574. [Google Scholar] [CrossRef] [PubMed]
  42. Agnes, A.; Lirosi, M.C.; Panunzi, S.; Santocchi, P.; Persiani, R.; D’Ugo, D. The prognostic role of perioperative allogeneic blood transfusions in gastric cancer patients undergoing curative resection: A systematic review and meta-analysis of non-randomized, adjusted studies. Eur. J. Surg. Oncol. (EJSO) 2018, 44, 404–419. [Google Scholar] [CrossRef] [PubMed]
  43. Zuckerman, J.; Coburn, N.; Callum, J.; Mahar, A.L.; Acuña, S.A.; Guttman, M.P.; Zuk, V.; Lin, Y.; Turgeon, A.F.; Martel, G.; et al. Association of perioperative red blood cell transfusions with all-cause and cancer-specific death in patients undergoing surgery for gastrointestinal cancer: Long-term outcomes from a population-based cohort. Surgery 2021, 170, 870–879. [Google Scholar] [CrossRef] [PubMed]
  44. Petrelli, F.; Ghidini, M.; Ghidini, A.; Sgroi, G.; Vavassori, I.; Petrò, D.; Cabiddu, M.; Aiolfi, A.; Bonitta, G.; Zaniboni, A.; et al. Red blood cell transfusions and the survival in patients with cancer undergoing curative surgery: A systematic review and meta-analysis. Surg. Today 2021, 51, 1535–1557. [Google Scholar] [CrossRef]
  45. Liang, Y.; Guo, H.; Deng, J.; Wang, B.; Ding, X.; Wang, X.; Zhang, L.; Liang, H. Impact of intraoperative blood loss on survival after curative resection for gastric cancer. World J. Gastroenterol. 2013, 19, 5542–5550. [Google Scholar] [CrossRef]
  46. Pacelli, F.; Rosa, F.; Marrelli, D.; Pedrazzani, C.; Bossola, M.; Zoccali, M.B.; Marchet, A.; Di Cosmo, M.; Roata, C.; Graziosi, L.; et al. Do Perioperative Blood Transfusions Influence Prognosis of Gastric Cancer Patients? Analysis of 927 Patients and Interactions with Splenectomy. Ann. Surg. Oncol. 2011, 18, 1615–1623. [Google Scholar] [CrossRef]
  47. Rausei, S.; Ruspi, L.; Galli, F.; Tirotta, F.; Inversini, D.; Frattini, F.; Chiappa, C.; Rovera, F.; Boni, L.; Dionigi, G.; et al. Peri-operative blood transfusion in gastric cancer surgery: Prognostic or confounding factor? Int. J. Surg. 2013, 11, S100–S103. [Google Scholar] [CrossRef] [Green Version]
  48. Danish Ranx05 Colorectal Cancer Study Group; Mynster, T.; Christensen, I.J.; Moesgaard, F.; Nielsen, H.J. Effects of the combination of blood transfusion and postoperative infectious complications on prognosis after surgery for colorectal cancer. Br. J. Surg. 2000, 87, 1553–1562. [Google Scholar] [CrossRef]
  49. Xiao, H.; Xiao, Y.; Chen, P.; Quan, H.; Luo, J.; Huang, G. Association Among Blood Transfusion, Postoperative Infectious Complications, and Cancer-Specific Survival in Patients with Stage II/III Gastric Cancer After Radical Gastrectomy: Emphasizing Benefit from Adjuvant Chemotherapy. Ann. Surg. Oncol. 2020, 28, 2394–2404. [Google Scholar] [CrossRef]
  50. Jericó, C.; Osorio, J.; García-Erce, J.A.; Pera, M. Patient Blood Management strategies for iron deficiency anemia management in gastric cancer. Eur. J. Gastroenterol. Hepatol. 2019, 31, 547–548. [Google Scholar] [CrossRef]
  51. Mungan, I.; Dicle, B.; Bektaş, Ş.; Sarı, S.; Yamanyar, S.; Çavuş, M.; Turan, S.; Bostancı, E.B. Does the preoperative platelet-to-lymphocyte ratio and neutrophil-to-lymphocyte ratio predict morbidity after gastrectomy for gastric cancer? Mil. Med. Res. 2020, 7, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Inaoka, K.; Kanda, M.; Uda, H.; Tanaka, Y.; Tanaka, C.; Kobayashi, D.; Takami, H.; Iwata, N.; Hayashi, M.; Niwa, Y.; et al. Clinical utility of the platelet-lymphocyte ratio as a predictor of postoperative complications after radical gastrectomy for clinical T2-4 gastric cancer. World J. Gastroenterol. 2017, 23, 2519–2526. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Y.; Lu, J.-J.; Du, Y.-P.; Feng, C.-X.; Wang, L.-Q.; Chen, M.-B. Prognostic value of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in gastric cancer. Medicine 2018, 97, e0144. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Perioperative neutrophil-to-lymphocyte ratio (NLR) changes classified in four groups depending on their perioperative course. Values are expressed as mean and standard deviation.
Figure 1. Perioperative neutrophil-to-lymphocyte ratio (NLR) changes classified in four groups depending on their perioperative course. Values are expressed as mean and standard deviation.
Cancers 15 00144 g001aCancers 15 00144 g001b
Figure 2. Kaplan–Meier analysis of probability of disease-free survival according to the perioperative classification.
Figure 2. Kaplan–Meier analysis of probability of disease-free survival according to the perioperative classification.
Cancers 15 00144 g002
Figure 3. Kaplan–Meier analysis of probability of disease-free survival according to the NLR change cut-off.
Figure 3. Kaplan–Meier analysis of probability of disease-free survival according to the NLR change cut-off.
Cancers 15 00144 g003
Table 1. Demographics, comorbidities; analytical, surgical, and pathology characteristics in the study population, classified in four groups depending on their perioperative course.
Table 1. Demographics, comorbidities; analytical, surgical, and pathology characteristics in the study population, classified in four groups depending on their perioperative course.
VariablesTransfusion (−)
Infection (−)
Transfusion (+)
Infection (−)
Transfusion (−)
Infection (+)
Transfusion (+)
Infection (+)
p Values
N = 181N = 23N = 55N = 23
Age, Mean (SD)68.4 (11.5)76.0(9.32)70.8 (12.3)74.4(8.27)0.004
Sex, N (%): 0.059
  Male118 (65.2%)9 (39.1%)39 (70.9%)15 (65.2%)
  Female63 (34.8%)14(60.9%)16 (29.1%)8 (34.8%)
ASA score, N (%): 0.237
  ASA I7 (3.87%)0 (0.00%)0 (0.00%)0 (0.00%)
  ASA II85 (47.0%)7 (30.4%)28 (50.9%)9 (39.1%)
  ASA III85 (47.0%)15(65.2%)27 (49.1%)12 (52.2%)
  ASA IV4 (2.21%)1 (4.35%)0 (0.00%)2 (8.70%)
ECOG c, N (%): 0.027
  ECOG 070 (38.7%)4 (17.4%)20 (36.4%)3 (13.0%)
  ECOG ≥ 1111 (61.3%)19(82.6%)35 (63.6%)20 (87.0%)
Charlson score a, N (%): 0.376
  0–298 (54.1%)10(43.5%)25 (45.5%)9 (39.1%)
  ≥383 (45.9%)13(56.5%)30 (54.5%)14 (60.9%)
Ischemic heart, N (%): disease, N (%)6 (3.31%)3 (13.0%)1 (1.82%)2 (8.70%)0.076
CHF, N (%)5 (2.76%)3 (13.0%)1 (1.82%)3 (13.0%)0.017
DM complic., N (%)26 (14.4%)4 (17.4%)13 (23.6%)7 (30.4%)0.138
DM no complic., N (%)12 (6.63%)2 (8.70%)3 (5.45%)2 (8.70%)0.840
COPD, N (%)22 (12.2%)3 (13.0%)12 (21.8%)4 (17.4%)0.298
Renal failure, N (%)5 (2.76%)1 (4.35%)1 (1.82%)4 (17.4%)0.019
Vasc. Dis., N (%)20 (11.0%)5 (21.7%)9 (16.4%)2 (8.70%)0.350
Weight loss, N (%): 0.174
  0–5%120 (66.3%)17(73.9%)36 (65.5%)12 (52.2%)
  6–10%41 (22.7%)1 (4.35%)11 (20.0%)8 (34.8%)
  >10%20 (11.0%)5 (21.7%)8 (14.5%)3 (13.0%)
Hb (g/dL), Mean (SD)12.0 (2.63)10.4(2.26)11.8 (2.57)10.8 (2.99)0.012
Tumor location, N (%): 0.060
  Upper third15 (8.29%)1 (4.35%)10 (18.2%)4 (17.4%)
  Middle third67 (37.0%)6 (26.1%)21 (38.2%)7 (30.4%)
  Lower third97 (53.6%)16 (69.6%)23 (41.8%)12 (52.2%)
  Entire2 (1.10%)0 (0.00%)1 (1.82%)0 (0.00%)
Neoadjuvancy, N (%)70 (38.7%)4 (17.4%)25 (45.5%)10 (43.5%)0.128
Gastrectomy, N (%): 0.006
  Distal subtotal117 (64.6%)18(78.3%)23 (41.8%)13 (56.5%)
  Total64 (35.4%)5 (21.7%)32 (58.2%)10 (43.5%)
Access, N (%): 0.269
  Open88 (48.6%)14(60.9%)23 (41.8%)8 (34.8%)
  Laparoscopic93 (51.4%)9 (39.1%)32 (58.2%)15 (65.2%)
pT b, N (%): 0.179
  ≤T298 (54.1%)17(73.9%)30 (54.5%)16 (69.6%)
  >T383 (45.9%)6 (26.1%)25 (45.5%)7 (30.4%)
pN b, N (%): 0.008
  N091 (50.3%)6 (26.1%)20 (36.4%)5 (21.7%)
  ≥N190 (49.7%)17(73.9%)35 (63.6%)18 (78.3%)
Node count, Mean (SD)27.6 (15.8)27.7(16.3)26.5 (13.5)27.7 (9.69)0.972
Radicality, N (%): 0.881
  R0164 (90.6%)21(91.3%)52 (94.5%)21 (91.3%)
  R1–R217 (9.39%)2 (8.70%)3 (5.45%)2 (8.70%)
ASA, American Society of Anesthesiologists; CHF, congestive heart failure; DM, diabetes mellitus; COPD, chronic obstructive pulmonary disease; Vasc. Dis., vascular disease; SD, standard deviation. a According to Charlson Comorbidity Index [26]; b According to 8th edition of the International Union Against Cancer tumor node metastasis staging system [27]; c Performance status according to Eastern Cooperative Oncology Group (ECOG) [25].
Table 2. Type of postoperative infectious complication, as defined according to the Gastrectomy Complications Consensus Group (GCCG) [29]. A total of 88 postoperative infectious complications in 81 patients.
Table 2. Type of postoperative infectious complication, as defined according to the Gastrectomy Complications Consensus Group (GCCG) [29]. A total of 88 postoperative infectious complications in 81 patients.
lnfectious Complications an (%) b
Gastrointestinal complications
Anastomotic leak24 (8.3%)
Duodenal stump leak7 (2.4%)
Pancreatic fistula2 (0.7%)
Abdominal collection17 (5.5%)
Clostridium infection1 (0.3%)
Wound infection7 (2.4%)
Pneumonia13 (4.5%)
Catheter infection10 (3.4%)
Urinary infection7 (2.4%)
a There were seven patients with two postoperative infectious complications; b % is given above total of patients included in the study.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Puértolas, N.; Osorio, J.; Jericó, C.; Miranda, C.; Santamaría, M.; Artigau, E.; Galofré, G.; Garsot, E.; Luna, A.; Aldeano, A.; et al. Effect of Perioperative Blood Transfusions and Infectious Complications on Inflammatory Activation and Long-Term Survival Following Gastric Cancer Resection. Cancers 2023, 15, 144. https://doi.org/10.3390/cancers15010144

AMA Style

Puértolas N, Osorio J, Jericó C, Miranda C, Santamaría M, Artigau E, Galofré G, Garsot E, Luna A, Aldeano A, et al. Effect of Perioperative Blood Transfusions and Infectious Complications on Inflammatory Activation and Long-Term Survival Following Gastric Cancer Resection. Cancers. 2023; 15(1):144. https://doi.org/10.3390/cancers15010144

Chicago/Turabian Style

Puértolas, Noelia, Javier Osorio, Carlos Jericó, Coro Miranda, Maite Santamaría, Eva Artigau, Gonzalo Galofré, Elisenda Garsot, Alexis Luna, Aurora Aldeano, and et al. 2023. "Effect of Perioperative Blood Transfusions and Infectious Complications on Inflammatory Activation and Long-Term Survival Following Gastric Cancer Resection" Cancers 15, no. 1: 144. https://doi.org/10.3390/cancers15010144

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop