Retrospective Study Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. Apr 27, 2023; 15(4): 643-654
Published online Apr 27, 2023. doi: 10.4240/wjgs.v15.i4.643
Risk factors for blood transfusion and its prognostic implications in curative gastrectomy for gastric cancer
Lucas Eiki Kawakami, Pedro Barzan Bonomi, Marina Alessandra Pereira, Ulysses Ribeiro Jr, Bruno Zilberstein, Luiz Augusto Carneiro-D'Albuquerque, Marcus Fernando Kodama Pertille Ramos, Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01249000, Brazil
Fabrício Oliveira Carvalho, Luciana Ribeiro Sampaio, Blood Transfusion Unit, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01249000, Brazil
ORCID number: Lucas Eiki Kawakami (0000-0003-3666-0062); Pedro Barzan Bonomi (0000-0001-9540-3512); Marina Alessandra Pereira (0000-0002-6865-0988); Fabrício Oliveira Carvalho (0000-0002-6262-9424); Ulysses Ribeiro Jr (0000-0003-1711-7347); Bruno Zilberstein (0000-0002-1809-8558); Luciana Ribeiro Sampaio (0000-0003-4888-3546); Luiz Augusto Carneiro-D'Albuquerque (0000-0001-7607-7168); Marcus Fernando Kodama Pertille Ramos (0000-0003-0200-7858).
Author contributions: Kawakami LE and Bonomi PB contributed to data retrieval, critical analysis, and manuscript drafting; Pereira MA contributed to data retrieval, critical analysis, statistical analysis, and manuscript review; Carvalho FO and Sampaio LR contributed to data retrieval and manuscript review; Ribeiro Jr. U, Zilberstein B, and Carneiro-D'Albuquerque LA contributed to critical analysis and manuscript review; Ramos MFKP contributed to study design, data retrieval, critical analysis, and manuscript drafting.
Institutional review board statement: The study was approved by the Hospital Ethics Committee and registered online (https://plataformabrasil.saude.gov.br; CAAE: 59337222.7.0000.0068).
Informed consent statement: Informed consent was waived by the local ethics committee, given the study's retrospective nature.
Conflict-of-interest statement: The authors declare no conflict of interest for this article.
Data sharing statement: No additional data are available.
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: Marcus Fernando Kodama Pertille Ramos, PhD, Doctor, Surgical Oncologist, Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo, 251, Sao Paulo 01249000, Brazil. marcus.kodama@hc.fm.usp.br
Received: December 23, 2022
Peer-review started: December 23, 2022
First decision: February 3, 2023
Revised: February 12, 2023
Accepted: March 30, 2023
Article in press: March 30, 2023
Published online: April 27, 2023

Abstract
BACKGROUND

Gastric cancer (GC) is still a prevalent neoplasm around the world and its main treatment modality is surgical resection. The need for perioperative blood transfusions is frequent, and there is a long-lasting debate regarding its impact on survival.

AIM

To evaluate the factors related to the risk of receiving red blood cell (RBC) transfusion and its influence on surgical and survival outcomes of patients with GC.

METHODS

Patients who underwent curative resection for primary gastric adenocarcinoma at our Institute between 2009 and 2021 were retrospectively evaluated. Clinicopathological and surgical characteristics data were collected. The patients were divided into transfusion and non-transfusion groups for analysis.

RESULTS

A total of 718 patients were included, and 189 (26.3%) patients received perioperative RBC transfusion (23 intraoperatively, 133 postoperatively, and 33 in both periods). Patients in the RBC transfusions group were older (P < 0.001), and had more comorbidities (P = 0.014), American Society of Anesthesiologists classification III/IV (P < 0.001), and lower preoperative hemoglobin (P < 0.001) and albumin levels (P < 0.001). Larger tumors (P < 0.001) and advanced tumor node metastasis stage (P < 0.001) were also associated with the RBC transfusion group. The rates of postoperative complications (POC) and 30-d and 90-d mortality were significantly higher in the RBC transfusion group than in the non-transfusion group. Lower hemoglobin and albumin levels, total gastrectomy, open surgery, and the occurrence of POC were factors associated with the RBC transfusion. Survival analysis demonstrated that the RBC transfusions group had worse disease-free survival (DFS) and overall survival (OS) compared with patients who did not receive transfusion (P < 0.001 for both). In multivariate analysis, RBC transfusion, major POC, pT3/T4 category, pN+, D1 lymphadenectomy, and total gastrectomy were independent risk factors related to worse DFS and OS.

CONCLUSION

Perioperative RBC transfusion is associated with worse clinical conditions and more advanced tumors. Further, it is an independent factor related to worse survival in the curative intent gastrectomy setting.

Key Words: Stomach neoplasms, Blood transfusion, Red blood cells, Postoperative complications, Survival, Prognosis

Core Tip: This is a retrospective study to investigate the association of perioperative red blood cell (RBC) transfusion with surgical and survival outcomes in patients with gastric cancer. Our findings demonstrated that patients who received RBC transfusion had poorer preoperative clinical conditions and more aggressive tumors, and were submitted to more invasive procedures. The rates of postoperative complications and 30-d and 90-d mortality were also significantly higher in patients who received RBC transfusions compared to those who did. Further, receiving an RBC transfusion was an independent factor associated with worse survival.



INTRODUCTION

In 2020, gastric cancer (GC) was the fifth most diagnosed neoplasm and the fourth cause of death by neoplasms[1]. Although its incidence and mortality rates have decreased in the last two decades, in 2025, GC will be accountable for more than one million cases and eight hundred thousand deaths[2]. GC is frequently associated with perioperative blood loss, whether by its biological behavior or its most important treatment, radical gastrectomy. Therefore, anemia and blood transfusion in the perioperative setting are a common concern[3,4].

The Transfusion Requirements in Critical Care trial (1999) was the first study to show worse outcomes related to excessive use of blood components in critical care patients, and since then, more restrictive use of transfusions has been recommended[5]. In the last ten years, surgeons and oncologists have studied the continuous pro-inflammatory status triggered by surgical tissue damage, postoperative complications (POC), and blood transfusions[6]. This effort confirmed the association between transfusion and higher recurrence rates in colorectal, pancreatic, and biliary tract cancers[6-8]. However, the current literature seems to struggle to find an answer for the impact of blood components on the outcomes of curative intent treatment in GC. The debate on how blood transfusion impacts survival and POC in GC is a complex topic, given the heterogeneity of results found in recent years. One side supported blood transfusion as an independently associated risk factor for inferior results; the other concluded that using blood components is a confounding factor for the worse prognosis since patients needing transfusions presented unfavorable clinical conditions previous to the surgical procedure and more advanced tumors at pathological staging compared to patients who did not receive transfusions[9-11].

Thus, this study aimed to evaluate the influence of perioperative red blood cell (RBC) transfusion on surgical and survival outcomes of patients with GC. We also examined the factors related to the risk of receiving a blood transfusion.

MATERIALS AND METHODS
Patient selection and study design

This is a retrospective cohort of patients with GC who underwent gastrectomy with curative intent in an oncological reference center from February 2009 to December 2021. Non-adenocarcinoma tumors (lymphoma, gastrointestinal stromal tumor, and neuroendocrine tumors) were excluded, as well as palliative surgery, diagnostic laparoscopy, previous hematological disorders, and patients with synchronic neoplasms.

Data collection and definitions

The following clinical variables were evaluated: Age, sex, preoperative body mass index (BMI), neutrophil-lymphocyte ratio (NLR), hemoglobin, albumin level, and performance status based on the American Society of Anesthesiologists (ASA) classification. Charlson-Deyo comorbidity index (CCI) was used to measure comorbidities without including age and GC as comorbidity[12]. Tumor node metastasis (TNM) staging was determined according to the 8th edition of the American Joint Committee on Cancer[13].

Experienced surgeons performed surgical procedures. The surgical approach (open or laparoscopic) was carried out based on the surgeon's decision after a multidisciplinary meeting composed of the oncology, surgery, radiology, and pathology departments. The extension of gastric and lymph node (LN) dissection followed the recommendations of the Japanese Gastric Cancer Association (JGCA)[14]. The classification proposed by Baiocchi et al[15] was employed to define intraoperative complications. Intraoperative blood loss was measured in milliliters, and the length of the surgical procedure was assessed in minutes.

For analysis, the patients were divided into two groups: Patients who received an RBC transfusion and those who did not. In addition to the RBC transfusion, we also describe the transfusion of platelet concentrate (PC) and fresh frozen plasma (FFP). Regarding the moment in which the administration occurred, the following periods were considered: Intraoperative and postoperative (until the 30th day).

POC were graded according to Clavien-Dindo's classification. Clavien III to V was considered major complications[16]. Mortality at 30 and 90 d after the surgical procedure was also assessed. Adjuvant or perioperative platin-based chemotherapy was administered according to clinical indications (T3, T4, and regional LN metastasis)[17].

Surgical and oncological teams performed postoperative follow-up medical appointments every 3 mo in the first year and every 6 mo in the following years. The attending clinician assigned to each case determined recurrence based on laboratory tests, CT, or endoscopy reports. Lost to follow-up was defined as an absence for more than 12 mo in follow-up visits.

We obtained all data by reviewing the patient’s medical chart and blood center system. The hospital ethics committee approved the study (CAAE: 59337222.7.0000.0068) and it was registered online in the national research projects database (www.plataformabrasil.org.br).

Statistical analysis

The Chi-square test was used to compare categorical variables between the two groups, and the t-test or Mann-Whitney test for continuous variables. Univariate and multivariate binary regression analyses were used to identify risk factors for receiving perioperative RBC transfusion. Odds ratios (ORs) with 95%CI were calculated.

Survival was estimated using the Kaplan-Meier method, and the log-rank test was used to identify differences between the survival curves. The Cox proportional hazards model was used to identify risk factors independently associated with survival outcomes. The results are reported as hazard ratios (HRs) with 95%CIs. Disease-free survival (DFS) was calculated from the date of surgery to recurrence or death from any cause. Overall survival (OS) was the duration between the date of surgery to death. All patients alive were censored at the date of the last follow-up. All tests were two-sided, and P < 0.05 was considered statistically significant. Statistical analyses were performed using SPSS software, version 20.0 (SPSS Inc, Chicago, IL).

RESULTS
Population description

During the selected period, 718 patients underwent radical gastrectomy with curative intent. Among them, 189 (26.3%) patients received perioperative RBC transfusion (RBC transfusion group). The remaining 529 (73.6%) patients who did not receive any perioperative RBC formed the non-transfusion group.

Among 189 patients who received RBC transfusion, 23 underwent transfusion in the intraoperative period, 133 in the postoperative period, and 33 in both periods (intra and postoperative). Besides RBC transfusion, FFP transfusions occurred in 12 patients (6.4%) and 2 patients (1.1%) also received PC transfusion.

Patients in the RBC transfusion group had older age (P < 0.001), higher CCI (P = 0.014), ASA III/IV score (P < 0.001), and lower BMI (P = 0.016) compared with patients who did not receive a transfusion. Higher NLR (P = 0.010) and lower preoperative hemoglobin (P < 0.001) and albumin levels (P < 0.001) were also associated with the RBC transfusions group. There was no difference regarding preoperative chemotherapy between the groups (P = 0.095). Complete clinical characteristics are demonstrated in Table 1.

Table 1 Clinical characteristics of patients according to perioperative red blood cell transfusion.
Variable
Non-transfusion
Red blood cell transfusion
P value
n = 529 (%)
n = 189 (%)
Sex0.318
    Female215 (40.6)69 (36.5)
    Male314 (59.4)120 (63.5)
Age (yr), mean ± SD61.3 ± 12.465.6 ± 11.6< 0.001
Body mass index (kg/m²), mean ± SD24.6 ± 4.623.7 ± 11.60.019
Hemoglobin (g/dL), mean ± SD12.5 ± 2.110.8 ± 2.2< 0.001
Albumin (g/dL), mean ± SD4.0 ± 0.63.7 ± 0.7< 0.001
Neutrophil-lymphocyte ratio, mean ± SD2.65 ± 2.773.26 ± 2.750.010
American Society of Anesthesiologists< 0.001
    I/II404 (76.4)11 (58.7)
    III/IV125 (23.6)78 (41.3)
Charlson-Deyo comorbidity index0.014
    0360 (68.1)110 (58.2)
    ≥ 1169 (31.9)79 (41.8)
Preoperative chemotherapy0.095
    No237 (44.8)98 (51.9)
    Yes292 (55.2)91 (48.1)

Regarding surgical procedures and postoperative features demonstrated in Table 2, total gastrectomy (P < 0.001) and open surgery (P < 0.001) were more frequent in the RBC transfusion group. There was no difference regarding the duration of surgery (P = 0.636), intraoperative complications (P = 0.209), and intraoperative blood loss (P = 0.186) between the two groups. Length of hospital stay was higher in the transfusion group (10.4 d vs 21.6 d, P < 0.001). Considering the postoperative outcomes, the rates of POC (P < 0.001) and mortality at 30 and 90 d were significantly higher in the transfusion group (P < 0.001).

Table 2 Surgical and postoperative characteristics of patients according to perioperative red blood cell transfusion.
Variable
Non-transfusion
Red blood cell transfusion
P value
n = 529 (%)
n >= 189 (%)
Type of resection< 0.001
    Subtotal337 (63.7)91 (48.1)
    Total192 (36.3)98 (51.9)
Surgical access< 0.001
    Open 396 (74.9)168 (88.9)
    Minimally invasive133 (25.1)21 (11.1)
Type of lymphadenectomy< 0.001
    D1104 (19.7)72 (38.1)
    D2425 (80.3)117 (61.9)
Operation time (min), mean ± SD243.5 ± 74.3246.6 ± 77.30.636
Intraoperative blood loss (mL), mean ± SD299.7 ± 336.7342.0 ± 362.60.186
Intraoperative complications0.209
    No508 (96.0)178 (94.2)
    Yes21 (4.0)11 (5.8)
Length of postoperative stay (d), mean ± SD10.4 ± 7.221.6 ± 17.4< 0.001
Postoperative complications (Clavien-Dindo)< 0.001
    0/I/II (minor)488 (92.2)112 (59.3)
    III/IV/V (major)41 (7.8)77 (40.7)
Adjuvant chemotherapy0.001
    No287 (54.3)129 (68.3)
    Yes242 (45.7)60 (31.7)
Mortality
    30-d8 (1.5)19 (10.1)< 0.001
    90-d16 (3.1)36 (19.0)< 0.001

The pathological characteristics of the two groups are shown in Table 3. Larger tumor size (P < 0.001), intestinal Lauren type (P = 0.002), pT3/T4 (P < 0.001), and advanced pathological TNM (pTNM) stages (P < 0.001) were more frequent in the RBC transfusion group. The presence of lymphatic (P = 0.027), vascular (P = 0.017), and perineural (P = 0.001) invasions was also associated with the transfusion group.

Table 3 Pathological characteristics of patients according to perioperative red blood cell transfusion.
Variable
Non-transfusion
Red blood cell transfusion
P value
n >= 529 (%)
n = 189 (%)
Tumor size (cm), mean ± SD4.3 ± 2.66.0 ± 3.6< 0.001
Tumor location0.039
    Lower327 (61.8)95 (50.3)
    Middle130 (24.6)60 (31.7)
    Upper64 (12.2)29 (15.3)
    Diffuse8 (1.5)5 (2.6)
Lauren histologic type10.002
    Intestinal276 (52.6)124 (66.0)
    Diffuse/mixed249 (47.4)64 (34.0)
Histological differentiation10.056
    Well/moderate 245 (46.7)103 (54.8)
    Poor 280 (53.3)85 (45.2)
Lymphatic invasion0.027
    No293 (55.4)87 (46.0)
    Yes236 (44.6)102 (54.0)
Vascular invasion0.017
    No364 (68.8)112 (59.3)
    Yes165 (31.2)77 (40.7)
Perineural invasion0.001
    No304 (57.5)83 (43.9)
    Yes225 (42.5)106 (56.1)
pT status < 0.001
    pT1/T2248 (46.9)54 (28.6)
    pT3/T4281 (53.1)135 (71.4)
Lymph nodes harvested, mean ± SD41.5 ± 19.439.2 ± 19.50.175
pN status 0.126
    pN0244 (46.1)75 (39.7)
    pN+285 (53.9)114 (60.3)
pTNM stage< 0.001
    I/II329 (62.2)86 (45.5)
    III/IV200 (37.8)103 (54.5)

In multivariate analysis, low preoperative hemoglobin (P < 0.001), low albumin (P = 0.017), total gastrectomy (P = 0.011), open surgical access (P = 0.034), and occurrence of major POC (P < 0.001) were independent factors associated to a higher risk of receiving perioperative RBC transfusions (Table 4).

Table 4 Univariate and multivariate analyses of factors associated with risk of receiving perioperative red blood cell transfusion.
Variable
Univariate
Multivariate
OR
95%CI
P value
OR
95%CI
P value
Male vs female1.190.85-1.680.319
Age ≥ 65 yr vs < 65 yr1.481.06-2.070.021.20.79-1.810.394
Charlson ≥ 1 vs 01.531.09-2.150.0151.220.76-1.970.402
ASA III/IV vs I/II2.271.60-3.23< 0.0011.180,72-1.940.507
HB < 11 g/dL vs ≥ 11 g/dL4.73.30-6.70< 0.0014.322.76-6.78< 0.001
ALB < 3.5 g/dL vs ≥ 3.5 g/dL3.492.29-5.31< 0.0011.861.12-3.090.017
Total gastrectomy vs distal1.891.35-2.65< 0.0011.711.13-2.600.011
Open surgery vs MIS2.691.64-4.41< 0.0011.911.05-3.470.034
Major POC vs non/minor POC8.185.32-12.59< 0.0018.835.28-14.79< 0.001
Intraoperative intercurrence vs none1.490.71-3.160.293
Survival analysis

The median follow-up time for the entire cohort of cases was 35.6 mo. During the follow-up period, 174 patients had disease recurrence and 261 died.

Patients who received perioperative RBC transfusions had worse DFS and OS than the non-transfusion group (P < 0.001) (Figure 1). The median DFS and OS for the RBC transfusion group were 19.5 and 35.8 mo, respectively.

Figure 1
Figure 1 Disease-free survival and overall survival of patients according to perioperative red blood cell transfusion. RBC: Red blood cell.

In multivariate analysis, total gastrectomy, more advanced pT stage, LN metastasis, D1 Lymphadenectomy, the occurrence of POC, and perioperative RBC transfusion were independent factors associated with worse DFS (Table 5).

Table 5 Univariate and multivariate analyses of factors associated with disease-free and overall survival.

Univariate
Multivariate
HR
95%CI
P value
HR
95%CI
P value
Disease-free survival
    Male vs female1.270.99-1.620.051
    Age ≥ 65 yr vs < 65 yr1.250.99-1.570.059
    Charlson ≥ 1 vs 01.331.05-1.680.0181.220.93-1.600.159
    ASA III/IV vs I/II1.831.44-2.33< 0.0011.240.93-1.660.149
    Total gastrectomy vs distal1.771.41-2.33< 0.0011.451.14-1.840.002
    D1 vs D21.611.25-2.07< 0.0011.41.06-1.850.017
    pT3/T4 vs pT1/T22.872.19-3.76< 0.0012.051.50-2.79< 0.001
    pN+ vs pN02.812.17-3.65< 0.0011.971.47-2.65< 0.001
    Major POC vs non/minor POC2.862.19-3.73< 0.0012.151.62-2.86< 0.001
    Non-CMT vs received CMT10.79-1.260.995
    Perioperative RBC transfusion vs non2.391.88-3.02< 0.0011.491.14-1.940.003
Overall survival
    Male vs female1.250.97-1.610.084
    Age ≥ 65 yr vs < 65 yr1.411.11-1.800.0061.240.95-1.610.113
    Charlson ≥ 1 vs 01.351.06-1.730.0171.110.83-1.490.473
    ASA III/IV vs I/II2.021.57-2.61< 0.0011.361.01-1.850.048
    Total gastrectomy vs distal1.651.29-2.10< 0.0011.331.03-1.710.027
    Lymphadenectomy D1 vs D21.811.39-2.35< 0.0011.511.11-2.050.008
    pT3/T4 vs pT1/T22.922.19-3.90< 0.0012.191.57-3.06< 0.001
    pN+ vs pN02.72.06-3.56< 0.0011.871.37-2.56< 0.001
    Major POC vs non/minor POC3.332.53-4.38< 0.0012.651.97-3.56< 0.001
    Non-CMT vs received CMT1.050.83-1.340.678
    Perioperative RBC transfusion vs non2.331.81-2.99< 0.0011.341.02-1.770.038

ASA, type of gastrectomy, lymphadenectomy, pT, pN, POC, and perioperative RBC transfusion were factors significantly associated with OS in the multivariate model (Table 5).

Perioperative RBC transfusion remained an independently associated risk factor for both DFS [hazard ratio (HR) = 1.49, 95%CI: 1.14-1.94, P = 0.003] and OS (HR = 1.34, 95%CI: 1.02-1.77, P = 0.038).

DISCUSSION

During the progression of GC, cachexia and uncontrolled tumor bleeding may induce severe anemia leading to life-threatening conditions and worse clinical outcomes. In this scenario, perioperative RBC transfusion is indicated to improve performance and decrease morbidity in the postoperative period[4,18,19]. On the other hand, recent advances in immunology have questioned the role of immunosuppression triggered by transfusion and its impact on tumor recurrence in gastrointestinal tract neoplasms[20-22].

In our retrospective cohort composed of 718 patients, perioperative RBC transfusions were related to worse DFS and OS. It is crucial to recognize expressive baseline differences between patients who received RBC transfusions and those who did not. Patients in the transfusion group were older and presented more unfavorable clinical conditions. Further, a higher frequency of total gastrectomy, open surgery, and advanced tumors was observed in the transfusion group. Other studies also reported the same heterogeneity noted in the analyzed population[9-11,23]. Even though, after multivariate analysis, we found that perioperative RBC transfusion was an independent factor related to recurrence and survival.

Although there is a vast amount of literature investigating the impact of blood components on the oncologic prognosis of GC patients, the current data still present discordant results[11,24,25]. This situation will probably be extended since it is difficult to perform a randomized controlled trial (RCT) in this scenario since in many situations the need for transfusion is a life-threatening condition. Further, most meta-analyses stress that current studies lack high-quality data[26]. In the recent retrospective studies that found no impact of RBC transfusions on long-term survival, some of them applied propensity-score matched analysis; however, preoperative hemoglobin and intraoperative blood loss remain as factors that could not be matched between groups[11,27]. Despite the knowledge of the relevance of anemia in the perioperative setting, conflicting data persist around the impact of intraoperative blood loss on OS and DFS of GC patients[28-30]. Grasso et al[31] carried out prospective studies comparing different hemoglobin threshold values for the indication of transfusion may be an alternative to define this issue.

The current hypothesis that explains the biological association between blood components and poorer oncological outcomes is that transfusion-related immunomodulation (TRIM) acts as a propagating factor for the TH2 immune response, favoring a pro-tumoral environment through inhibition of interleukin (IL)-2 and stimulation of suppressor T cells, allowing tumor spread and recurrence[20-22]. The recent application of immunotherapy in gastrointestinal tract cancers provided additional data by demonstrating that TRIM could be acting as an opponent and negatively impacting its effectiveness and survival[32]. Another important topic related to the immune response is the data provided by Lange et al[33] that showed no difference in using leukocyte depletion in long-term survival, underlining that specific constituents of allogeneic blood may mediate the TRIM effect. This same result was detected when RBC transfusions were applied to other neoplasms[34].

Preoperative hemoglobin and albumin were independent factors associated with RBC transfusions. Since GC causes feeding and bleeding disturbances, aggressive protocols for improving hematologic and nutritional preoperative status must be paramount in clinical compensation ahead of surgical treatment. Current data support those policies in clinical and financial terms since Jericó et al[35] demonstrated reduced direct and indirect spent resources, lower hospital length of stay, and readmissions succeeding radical gastrectomy[36].

Interestingly, D1 lymphadenectomy was associated with worse DFS and OS. D2 lymphadenectomy is considered a more aggressive procedure and the standard in GC treatment. However, even though 42% of our population were composed of patients with more advanced stages III/IV, some of them did not have the clinical conditions to perform D2 lymphadenectomy. So, the employment of D1 lymphadenectomy rarely was an oncological indication as proposed in the 2018 JGCA guideline for GC treatment[14]. It was mainly indicated for patients with unfavorable medical conditions to reduce POC and mortality, as previously reported by our service[37].

Open surgical access was associated with the transfusion group. Minimally invasive surgery causes less tissue damage to the abdominal wall with reports of less intraoperative blood loss on several RCTs[38-40]. However, intraoperative blood loss, although higher in the transfusion group, did not show a significant difference between our groups. Intraoperative blood loss is a variable that is difficult to measure in clinical practice. The retrospective nature of the study also makes accurate measurement difficult. Despite this possible bias, we also found that there was no difference between the groups in the occurrence of intraoperative complications, a variable that is very well documented. Baiocchi et al[15] reported a low 2% incidence of intraoperative complications in GC surgery. In our study, a 4.45% incidence of intraoperative complications was reported, represented mainly by intraoperative bleeding. Those numbers indicate adequate documentation of the intraoperative complications in our medical reports and eventually, intraoperative blood loss did not differ between the groups. Therefore, our best efforts should focus on better patient perioperative management to avoid RBC transfusion[41].

Regarding POC, the multivariate analysis indicated that major POC presented the highest OR related to RBC transfusion among the eight selected variables. It must also be emphasized that the transfusion in the postoperative period was more frequent than in the pre and intraoperative periods. The importance of POC was already stressed in 2020 through a meta-analysis evaluating their repercussions on GC survival[42]. A plausible reason for cancer recurrence is the pro-inflammatory state caused by surgical trauma, where IL-6 suppresses the specific and non-specific immune responses. This mechanism could be synergically associated with IL-2 suppression caused by TRIM since a retrospective analysis found a signature of cytokines (including IL-2 and IL-6) and angiogenic factors associated with poor DFS and OS[43]. Further, POCs may prevent patients to return to the intended oncological treatment, a known prognostic factor[44].

The performance of retrospective studies has some limitations inherent to its design. Despite the relevant number of patients included for a Western center, the numerous variables evaluated are confounding factors for the adequate definition of the association between RBC transfusion and prognosis. We chose multivariate logistic regression to adjust the potential bias of covariates. Ultimately, perioperative transfusion of RBC was an independent prognostic factor together with known prognostic factors such as pTNM stage, demonstrating a good accuracy of the performed analyses. As another limitation, we must point out that our data were collected over 13 years, so variations and advances in oncological treatments and surgical techniques may cause additional heterogeneity.

CONCLUSION

In GC patients undergoing curative surgeries, poor clinical status, more extensive surgical procedures, and advanced tumor stages are common features in patients receiving RBC transfusions. In addition to being associated with higher rates of POC and mortality, receiving an RBC transfusion proves to be an independent factor associated with worse survival.

ARTICLE HIGHLIGHTS
Research background

Anemia and intraoperative blood loss are frequent issues in gastric cancer (GC) surgical treatment. The current literature still debates the impact of perioperative blood transfusion on GC survival.

Research motivation

Red blood cell (RBC) transfusions are sometimes required for patients undergoing surgery for GC. However, the prognostic impact of perioperative RBC transfusion in GC is controversial.

Research objectives

We analyzed the influence of RBC transfusions on the prognosis of patients with gastric adenocarcinoma undergoing gastrectomy with curative intention.

Research methods

We retrospectively evaluated all GC patients who underwent gastrectomy between 2009 and 2021. Patients were divided into transfusion group and non-transfusion group for analysis. RBC transfusions that occurred intraoperatively and postoperatively within 30 d were considered.

Research results

A total of 718 patients were included, and 189 (26.3%) patients received RBC transfusions. Patients who received transfusions had unfavorable clinical and pathological characteristics, and underwent more extensive surgical procedures. Patients who received RBC transfusions had worse survival compared to those who did not. In multivariate analysis, receiving an RCB transfusion was an independent factor associated with poor disease-free survival (DFS) and overall survival (OS).

Research conclusions

Even though the patients who receive RCB transfusion have worse clinical conditions, we found that perioperative transfusion represents an independent factor associated with poor prognosis, with worse DFS and OS.

Research perspectives

The application of blood component transfusion in randomized clinical trials presents ethical limitations; however, the current design of retrospective studies still interferes with controlling confounding factors. With this study, we endorse a favorable position for increasing preoperative and postoperative care to avoid RBC transfusion. Further, our findings provide additional data for future meta-analysis.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Brazil

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Kinami S, Japan; Manojlovic N, Serbia S-Editor: Zhang H L-Editor: Wang TQ P-Editor: Zhang H

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 46550]  [Article Influence: 15516.7]  [Reference Citation Analysis (47)]
2.  Ilic M, Ilic I. Epidemiology of stomach cancer. World J Gastroenterol. 2022;28:1187-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 48]  [Cited by in F6Publishing: 110]  [Article Influence: 55.0]  [Reference Citation Analysis (19)]
3.  Kouyoumdjian A, Trepanier M, Al Shehhi R, Cools-Lartigue J, Ferri LE, Lee L, Mueller CL. The Effect of Preoperative Anemia and Perioperative Transfusion on Surgical Outcomes After Gastrectomy for Gastric Cancer. J Surg Res. 2021;259:523-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
4.  Huang XZ, Yang YC, Chen Y, Wu CC, Lin RF, Wang ZN, Zhang X. Preoperative Anemia or Low Hemoglobin Predicts Poor Prognosis in Gastric Cancer Patients: A Meta-Analysis. Dis Markers. 2019;2019:7606128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
5.  Hébert PC, Wells G, Blajchman MA, Marshall J, Martin C, Pagliarello G, Tweeddale M, Schweitzer I, Yetisir E. A multicenter, randomized, controlled clinical trial of transfusion requirements in critical care. Transfusion Requirements in Critical Care Investigators, Canadian Critical Care Trials Group. N Engl J Med. 1999;340:409-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3699]  [Cited by in F6Publishing: 3264]  [Article Influence: 130.6]  [Reference Citation Analysis (0)]
6.  McSorley ST, Tham A, Dolan RD, Steele CW, Ramsingh J, Roxburgh C, Horgan PG, McMillan DC. Perioperative Blood Transfusion is Associated with Postoperative Systemic Inflammatory Response and Poorer Outcomes Following Surgery for Colorectal Cancer. Ann Surg Oncol. 2020;27:833-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 37]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
7.  Kanda T, Wakiya T, Ishido K, Kimura N, Nagase H, Kubota S, Fujita H, Hagiwara Y, Hakamada K. Intraoperative Allogeneic Red Blood Cell Transfusion Negatively Influences Prognosis After Radical Surgery for Pancreatic Cancer: A Propensity Score Matching Analysis. Pancreas. 2021;50:1314-1325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
8.  Wang Q, Du T, Lu C. Perioperative blood transfusion and the clinical outcomes of patients undergoing cholangiocarcinoma surgery: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2016;28:1233-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
9.  Liu X, Ma M, Huang H, Wang Y. Effect of perioperative blood transfusion on prognosis of patients with gastric cancer: a retrospective analysis of a single center database. BMC Cancer. 2018;18:649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
10.  Xu D, Fang X, Li Y, Zhang Z, Li Q. Perioperative blood transfusion is one of the factors that affect the prognosis of gastric cancer. J BUON. 2018;23:672-677.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Xiao H, Liu W, Quan H, Ouyang Y. Peri-Operative Blood Transfusion Does Not Influence Overall and Disease-Free Survival After Radical Gastrectomy for Stage II/III Gastric Cancer: a Propensity Score Matching Analysis. J Gastrointest Surg. 2018;22:1489-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
12.  Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40:373-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32099]  [Cited by in F6Publishing: 35393]  [Article Influence: 956.6]  [Reference Citation Analysis (0)]
13.  Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM.   AJCC Cancer Staging Manual (8th edition). Springer International Publishing: American Joint Commission on Cancer, 2017.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2018 (5th edition). Gastric Cancer. 2021;24:1-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 735]  [Cited by in F6Publishing: 1129]  [Article Influence: 376.3]  [Reference Citation Analysis (0)]
15.  Baiocchi GL, Giacopuzzi S, Marrelli D, Reim D, Piessen G, Matos da Costa P, Reynolds JV, Meyer HJ, Morgagni P, Gockel I, Lara Santos L, Jensen LS, Murphy T, Preston SR, Ter-Ovanesov M, Fumagalli Romario U, Degiuli M, Kielan W, Mönig S, Kołodziejczyk P, Polkowski W, Hardwick R, Pera M, Johansson J, Schneider PM, de Steur WO, Gisbertz SS, Hartgrink H, van Sandick JW, Portolani N, Hölscher AH, Botticini M, Roviello F, Mariette C, Allum W, De Manzoni G. International consensus on a complications list after gastrectomy for cancer. Gastric Cancer. 2019;22:172-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
16.  Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240:205-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18532]  [Cited by in F6Publishing: 22292]  [Article Influence: 1114.6]  [Reference Citation Analysis (0)]
17.  Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, Vogel A, Smyth EC; ESMO Guidelines Committee. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33:1005-1020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 309]  [Article Influence: 154.5]  [Reference Citation Analysis (0)]
18.  Tang GH, Hart R, Sholzberg M, Brezden-Masley C. Iron deficiency anemia in gastric cancer: a Canadian retrospective review. Eur J Gastroenterol Hepatol. 2018;30:1497-1501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
19.  Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers. 2018;4:17105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 644]  [Cited by in F6Publishing: 818]  [Article Influence: 136.3]  [Reference Citation Analysis (0)]
20.  Youssef LA, Spitalnik SL. Transfusion-related immunomodulation: a reappraisal. Curr Opin Hematol. 2017;24:551-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 60]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
21.  Cata JP, Wang H, Gottumukkala V, Reuben J, Sessler DI. Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions. Br J Anaesth. 2013;110:690-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 306]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
22.  Vamvakas EC, Blajchman MA. Transfusion-related immunomodulation (TRIM): an update. Blood Rev. 2007;21:327-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in F6Publishing: 470]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
23.  Elmi M, Mahar A, Kagedan D, Law CH, Karanicolas PJ, Lin Y, Callum J, Coburn NG, Hallet J. The impact of blood transfusion on perioperative outcomes following gastric cancer resection: an analysis of the American College of Surgeons National Surgical Quality Improvement Program database. Can J Surg. 2016;59:322-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
24.  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. 2021;28:2394-2404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
25.  Song JH, Shin HJ, Lee S, Park SH, Cho M, Kim YM, Hyung WJ, Kim HI. No detrimental effect of perioperative blood transfusion on recurrence in 2905 stage II/III gastric cancer patients: A propensity-score matching analysis. Eur J Surg Oncol. 2022;48:2132-2140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
26.  Agnes A, Lirosi MC, 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. 2018;44:404-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (1)]
27.  Hsu FK, Chang WK, Lin KJ, Liu CY, Fang WL, Chang KY. The Associations between Perioperative Blood Transfusion and Long-Term Outcomes after Stomach Cancer Surgery. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
28.  Wen ZL, Xiao DC, Zhou X. Does Intraoperative Blood Loss Affect the Short-Term Outcomes and Prognosis of Gastric Cancer Patients After Gastrectomy? Front Surg. 2022;9:924444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
29.  Zhao B, Huang X, Lu H, Zhang J, Luo R, Xu H, Huang B. Intraoperative blood loss does not independently affect the survival outcome of gastric cancer patients who underwent curative resection. Clin Transl Oncol. 2019;21:1197-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
30.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
31.  Grasso M, Pacella G, Sangiuliano N, De Palma M, Puzziello A. Gastric cancer surgery: clinical outcomes and prognosis are influenced by perioperative blood transfusions. Updates Surg. 2019;71:439-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
32.  Mispelbaum R, Hattenhauer ST, Brossart P, Heine A. Red blood cell transfusions impact response rates to immunotherapy in patients with solid malignant tumors. Front Immunol. 2022;13:976011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
33.  Lange MM, van Hilten JA, van de Watering LM, Bijnen BA, Roumen RM, Putter H, Brand A, van de Velde CJ; cooperative clinical investigators of the Cancer Recurrence And Blood Transfusion (CRAB) study and the Transfusion Associated Complications = Transfusion Induced Complications? (TACTIC) study. Leucocyte depletion of perioperative blood transfusion does not affect long-term survival and recurrence in patients with gastrointestinal cancer. Br J Surg. 2009;96:734-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
34.  Geng Y, Liu L. Impact of Allogeneic Leukocyte-Depleted Red Blood Cell Transfusion on Inflammatory Response and Blood Coagulation in Patients with Recurrence of Colon Cancer after Operation. Evid Based Complement Alternat Med. 2021;2021:6957569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
35.  Jericó C, Puértolas N, Osorio J, Miranda C, Santamaría M, Artigau E, Galofré G, Garsot E, Luna A, Aldeano A, Olona C, Pulido L, Pera M; Spanish EURECCA Esophagogastric Cancer Group. Cost analysis of a patient blood management program for patients undergoing gastric cancer surgery. Eur J Surg Oncol. 2023;49:293-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
36.  Osorio J, Jericó C, Miranda C, Santamaría M, Artigau E, Galofré G, Garsot E, Luna A, Puértolas N, Aldeano A, Olona C, Molinas J, Feliu J, Videla S, Tebe C, Pera M. Improved postoperative outcomes and reduced transfusion rates after implementation of a Patient Blood Management program in gastric cancer surgery. Eur J Surg Oncol. 2021;47:1449-1457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
37.  Ramos MFKP, Pereira MA, Dias AR, Yagi OK, Zaidan EP, Ribeiro-Júnior U, Zilberstein B, Cecconello I. Surgical outcomes of gastrectomy with D1 lymph node dissection performed for patients with unfavorable clinical conditions. Eur J Surg Oncol. 2019;45:460-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
38.  Hyung WJ, Yang HK, Park YK, Lee HJ, An JY, Kim W, Kim HI, Kim HH, Ryu SW, Hur H, Kim MC, Kong SH, Cho GS, Kim JJ, Park DJ, Ryu KW, Kim YW, Kim JW, Lee JH, Han SU; Korean Laparoendoscopic Gastrointestinal Surgery Study Group. Long-Term Outcomes of Laparoscopic Distal Gastrectomy for Locally Advanced Gastric Cancer: The KLASS-02-RCT Randomized Clinical Trial. J Clin Oncol. 2020;38:3304-3313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 188]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
39.  Yu J, Huang C, Sun Y, Su X, Cao H, Hu J, Wang K, Suo J, Tao K, He X, Wei H, Ying M, Hu W, Du X, Hu Y, Liu H, Zheng C, Li P, Xie J, Liu F, Li Z, Zhao G, Yang K, Liu C, Li H, Chen P, Ji J, Li G; Chinese Laparoscopic Gastrointestinal Surgery Study (CLASS) Group. Effect of Laparoscopic vs Open Distal Gastrectomy on 3-Year Disease-Free Survival in Patients With Locally Advanced Gastric Cancer: The CLASS-01 Randomized Clinical Trial. JAMA. 2019;321:1983-1992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 436]  [Article Influence: 87.2]  [Reference Citation Analysis (1)]
40.  Yang Y, Chen Y, Hu Y, Feng Y, Mao Q, Xue W. Outcomes of laparoscopic versus open total gastrectomy with D2 lymphadenectomy for gastric cancer: a systematic review and meta-analysis. Eur J Med Res. 2022;27:124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
41.  Huang H, Cao M. Development and validation of a nomogram to predict intraoperative blood transfusion for gastric cancer surgery. Transfus Med. 2021;31:250-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
42.  Li J, Zhang Y, Hu DM, Gong TP, Xu R, Gao J. Impact of postoperative complications on long-term outcomes of patients following surgery for gastric cancer: A systematic review and meta-analysis of 64 follow-up studies. Asian J Surg. 2020;43:719-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
43.  Ock CY, Nam AR, Bang JH, Kim TY, Lee KH, Han SW, Im SA, Bang YJ, Oh DY. Signature of cytokines and angiogenic factors (CAFs) defines a clinically distinct subgroup of gastric cancer. Gastric Cancer. 2017;20:164-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
44.  Ramos MFKP, de Castria TB, Pereira MA, Dias AR, Antonacio FF, Zilberstein B, Hoff PMG, Ribeiro Jr U, Cecconello I. Return to Intended Oncologic Treatment (RIOT) in Resected Gastric Cancer Patients. J Gastrointest Surg. 2020;24:19-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]