Skip to main content
Scientific Reports logoLink to Scientific Reports
. 2021 Apr 27;11:9038. doi: 10.1038/s41598-021-88254-z

Geriatric nutritional risk index predicts prognosis in hepatocellular carcinoma after hepatectomy: a propensity score matching analysis

Hiroki Kanno 1,, Yuichi Goto 1, Shin Sasaki 1, Shogo Fukutomi 1, Toru Hisaka 1, Fumihiko Fujita 1, Yoshito Akagi 1, Koji Okuda 1
PMCID: PMC8079680  PMID: 33907232

Abstract

The geriatric nutritional risk index (GNRI) is widely used for nutritional assessment in older inpatients and is associated with postoperative complications and cancer prognosis. We investigated the use of GNRI to predict long-term outcomes in hepatocellular carcinoma of all etiologies after hepatectomy. Overall, 346 patients were examined after propensity score matching. We dichotomized the GNRI score into high GNRI (> 98: N = 173) and low GNRI (≤ 98: N = 173) and evaluated recurrence-free survival (RFS) and overall survival (OS) between both groups. Clinicopathological characteristics between the low- and high-GNRI groups were similar after propensity score matching except for the components of the GNRI score (body mass index and serum albumin level), Child–Pugh score (comprising serum albumin level), and preoperative alpha-fetoprotein level (p < 0.0001, p < 0.0001, p = 0.0030, and p = 0.0007, respectively). High GNRI was associated with significantly better RFS and OS (p = 0.0003 and p = 0.0211, respectively; log-rank test). Multivariate analysis revealed that GNRI is an independent prognostic factor of RFS and OS (low vs. high; hazard ratio [HR], 1.8284; 95% confidence interval [CI] 1.3598–2.4586; p < 0.0001, and HR, 1.5452; 95% CI 1.0345–2.3079; p = 0.0335, respectively). GNRI is an objective, inexpensive, and easily calculated assessment tool for nutritional status and can predict prognosis of hepatocellular carcinoma after hepatectomy.

Subject terms: Cancer, Gastroenterology

Introduction

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide1. Despite improvements in therapeutic options such as surgery, ablation, transarterial chemoembolization, and molecular targeted agents, the outcome of HCC is unsatisfactory owing to the high recurrence rate. Chronic hepatitis or liver cirrhosis is usually present in the background of HCC and may result in malnutrition in these patients. Some studies have reported that malnutrition negatively impacts postoperative complications and cancer prognosis2.

Inflammation, malnutrition, and cancer have a close relationship with each other. Malnutrition causes the deterioration of tumor immunity, leading to postoperative complications and cancer progression3,4. Furthermore, cancer-related systemic inflammation induces catabolism of patients, which causes metastasis5,6. Some inflammatory indices—neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, and the modified Glasgow prognostic score—and several nutritional indices—the prognostic nutritional index, controlling nutritional status score (CONUT), sarcopenia, and geriatric nutritional index (GNRI)—can predict postoperative complications and long-term outcomes of several cancers79. These scores are also useful for evaluating postoperative complications and prognosis of HCC1012.

The GNRI was proposed by Bouillanne et al. in 200513 and has been widely used for nutritional assessment in older inpatients with cardiovascular diseases, chronic renal failure with hemodialysis, and chronic liver disease1417. The GNRI is calculated using serum albumin concentration, body height, and body weight, and patients are classified into four nutritional risk grades. Recently, some researchers have reported that the GNRI is associated with postoperative complications and cancer prognosis18,19. However, only one report has described this relationship in HCC, and this study was limited to patients aged 65 years or older who were positive for the hepatitis B virus20. However, the median age of onset of HCC is around 65 years old, and hepatitis virus C is the major cause of HCC in the developed countries. Therefore, we investigated the use of the GNRI to predict long-term outcomes in HCC patients, including those younger than 65 years of age with all underlying liver diseases.

Materials and methods

Patients

Six hundred and forty-eight consecutive patients who underwent hepatectomy for initial HCC at Kurume University between 2008 and 2018 were retrospectively reviewed. Of these, 120 were excluded due to the following reasons: 107 underwent preoperative treatment such as transcatheter arterial infusion, transarterial chemoembolization, and/or ablation; six did not have histologically confirmed HCC (HCC + combined hepatocellular-cholangiocellular carcinoma, necrosis, etc.); three had insufficient data; two did not achieve curative resection; one had another synchronous malignancy; and one had inferior vena cava tumor thrombus. Therefore, 528 patients were finally enrolled for analyses before propensity score matching (PSM). This study was approved by the Research Ethics Committee of Kurume University (No. 20236) and was conducted according to the tenets of the Declaration of Helsinki. The need for informed consent was waived owing to the retrospective nature of the study.

Data collection and calculation of the GNRI scores

Clinical and pathological data were collected from the patients’ medical records. Blood samples and physical data were obtained within 1 week before surgery. Calculation of the GNRI score was as follow: GNRI = (1.489 × serum albumin concentration [g/L]) + (41.7 × present/ideal body weight [kg]). When the present body weight exceeded the ideal body weight, “present/ideal body weight” was set to 1. Pathological diagnosis was performed by two pathologists in accordance with the rules of the Liver Cancer Study Group of Japan.

Cutoff value of the GNRI score

The GNRI was categorized into four risk groups: no risk (GNRI: > 98), low risk (GNRI: 92 to 98), moderate risk (GNRI: 82 to < 92), and major risk (GNRI: < 82). We set the cutoff value to ≤ 98 or > 98, which was in line with other studies18,21. The distribution of the GNRI score in the present study is shown in Supplementary Fig. S1.

Surgical procedure

The type of surgery was comprehensively planned according to the Child–Pugh score, liver damage, platelet count, liver stiffness, extent of cirrhosis, presence of esophageal varix and splenomegaly, tumor number, tumor location, and patients’ comorbidities. If poor liver function with Child–Pugh score C, elevated bilirubin level, or low platelet count was observed and/or portal hypertension was suspected before or during operation, we did not perform the surgery. Major hepatectomy was defined as lobectomy or more, and minor hepatectomy was defined as sectionectomy or less. Surgical procedures were performed as follows. Liver mobilization was performed in most cases, if necessary. The Pringle maneuver was used in most cases according to the surgeon’s preference. Parenchymal transection was carried out using an ultrasonic coagulation dissector and/or clamp crushing methods. Sizable vasculature was ligated tightly.

Postoperative follow-up

In the postoperative follow-up, patients were examined using ultrasonography and tumor markers (alpha-fetoprotein [AFP] and/or protein induced by vitamin K absence or antagonist-II [PIVKA-II]) at least every 3 months after surgery. Additionally, computed tomography (CT) and/or magnetic resonance imaging (MRI) were performed at least every 6 months in the patients with a high risk of recurrence. If recurrence was suspected, further examinations such as gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid-enhanced MRI or contrast-enhanced ultrasonography were conducted. Recurrence-free survival (RFS) was defined as the time from surgery to recurrence or death. Overall survival (OS) was defined as the time from surgery to death.

Statistical analyses

The clinicopathological characteristics were compared using the chi-square test for categorical variables and the Mann–Whitney U test for continuous variables. PSM was performed to overcome selection bias. The propensity score was calculated using a logistic regression model. Covariates entered into the PSM model were as follow: age, sex, underlying liver disease, total bilirubin level, prothrombin time, platelet count, AFP level, duration of surgery, estimated blood loss, surgical procedure, tumor diameter, tumor number, tumor differentiation, microvascular invasion (MVI), and histological fibrosis grade. PSM was performed using a 1:1 matching method with a caliper width of 0.2, and the area under the curve calculated from the receiver operating characteristics curve was 0.71734 (p < 0.0001). Survival curves were created using the Kaplan–Meier method and compared using the log-rank test. A Cox proportional hazards model was used for univariate and multivariate analyses, and hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. All statistical analyses were performed using JMP Pro, version 15 (SAS Institute, Cary, NC). A p value < 0.05 was considered statistically significant.

Ethics approval

This study was approved by the Research Ethics Committee of Kurume University (Approval number: No. 20236) and was conducted in accordance with the Declaration of Helsinki. The need for informed consent was waived owing to the retrospective nature of the study.

Results

Patient characteristics

Clinicopathological features between the low- and high-GNRI groups in the entire cohort before PSM are summarized in Supplementary Table S1. Age, body mass index (BMI), underlying liver disease, total bilirubin level, serum albumin concentrations, prothrombin time, AFP, Child–Pugh score, duration of surgery, and tumor number were significantly different between the two groups (p = 0.0002, p < 0.0001, p < 0.0001, p = 0.0150, p < 0.0001, p = 0.0005, p = 0.0001, p = 0.0004, p = 0.0126, and p = 0.0286, respectively). After matching, almost all variables were adjusted except for the components of the GNRI score (BMI and serum albumin level), Child–Pugh score (consisting serum albumin level), and preoperative AFP level (p < 0.0001, p < 0.0001, p = 0.0030, and p = 0.0007, respectively) (Table 1).

Table 1.

Clinicopathological characteristics of the patients in the low- and high-GNRI groups after propensity score matching.

Characteristics (n = 346) GNRI low (n = 173) % GNRI high (n = 173) % Total P value
Age (years), median (range) 73 (43–87) 72 (33–89) 0.4644
Sex 0.4163
 Male 125 72.3 128 74.0 253
 Female 48 27.7 45 26.0 93
BMI, median (IQR) 21.7 (19.3–24.45) 23.5 (21.75–25.65)  < 0.0001
Underlying liver disease 0.7165
 HBV 14 8.1 9 5.2 23
 HCV 108 62.4 112 64.7 220
 B + C 4 2.3 3 1.7 7
 NonBnonC 47 27.2 49 28.3 96
Excessive alcohol consumption* 0.2486
 Yes 35 20.2 44 25.4 79
 No 138 79.8 129 74.6 267
DM 0.3641
 Yes 55 31.8 63 36.4 118
 No 118 68.2 110 63.6 228
T.bil (mg/dL), median (IQR) 0.71 (0.57–0.85) 0.73 (0.59–0.90) 0.3933
Albumin (g/dL), median (IQR) 3.63 (3.41–3.77) 4.18 (3.98–4.38)  < 0.0001
PT (%), median (IQR) 91 (80.5–100) 91 (82.0–100) 0.5820
Platelet (× 104/L), median (IQR) 13.7 (10.2–17.9) 14.9 (11.5–18.2) 0.1466
AFP (ng/mL), median (IQR) 16.9 (5.4–101.25) 6.6 (3.8–33.7) 0.0007
Child–Pugh 0.0030
 A 163 94.2 172 99.4 335
 B 10 5.8 1 0.6 11
Surgical procedure
 Major 54 31.2 57 32.9 111 0.7297
 Minor 119 68.8 116 67.1 235
Duration of surgery (min), median (IQR) 354 (289–434) 350 (273–449) 0.7790
Estimated blood loss (ml), median (IQR) 385 (175–678) 330 (78–818) 0.1785
Tumor diameter (mm), median (IQR) 27 (19.5–39) 26 (20–40) 0.9118
Tumor number 0.6896
 Solitary 139 80.3 136 78.6 275
 Multiple 34 19.7 37 21.4 71
Differentiation 0.6680
 Well/mod 142 82.1 145 83.8 287
 Poor 31 17.9 28 16.2 59
Microvascular invasion 1.0000
 Yes 93 53.8 93 53.8 186
 No 80 46.2 80 46.2 160
Histological fibrosis grade 0.7466
 F0-2 80 46.2 83 48.0 163
 F3-4 93 53.8 90 52.0 183
GNRI, median (IQR) 94.0 (90.2–96.4) 102.7 (100.2–105.7)  < 0.0001

*Excessive alcohol consumption is defined as > 28 g/day ethanol in men and > 14 g/day in women.

AFP: alpha-fetoprotein, BMI: body mass index, DM: diabetes mellitus, GNRI: geriatric nutritional risk index, IQR: interquartile range, PT: prothrombin time, T.bil: total bilirubin.

Comparison of survival between the low- and high-GNRI groups

The RFS and OS curves of the two groups are shown in Fig. 1. A high GNRI was associated with significantly better RFS and OS (p = 0.0003 and p = 0.0211, respectively).

Figure 1.

Figure 1

Kaplan–Meier curves of RFS and OS in the low- and high-GNRI groups. RFS, recurrence-free survival; OS, overall survival, GNRI, geriatric nutritional risk index.

Univariate and multivariate analyses for RFS and OS

The results of the univariate and multivariate analyses of RFS and OS are shown in Tables 2 and 3. Univariate analysis of RFS demonstrated that underlying liver disease (Viral vs. Non-viral; HR, 1.4269; 95% CI 1.0195–1.9970; p = 0.0325), AFP level (5 > vs. ≥ 5 ng/mL; HR, 0.5440; 95% CI 0.3864–0.7657; p = 0.0005), tumor number (solitary vs. multiple; HR, 0.5588; 95% CI 0.4053–0.7706; p = 0.0004), tumor differentiation (well/mod vs. poor; HR, 0.6494; 95% CI 0.4535–0.9298; p = 0.0184), MVI (− vs. + ; HR, 0.5099; 95% CI 0.3809–0.6825; p < 0.0001), histological fibrosis grade (F0-2 vs. F3-4; HR, 0.6963; 95% CI 0.5229–0.9272; p = 0.0133), and GNRI (low vs. high; HR, 1.6809; 95% CI 1.2645–2.2346; p = 0.0003) were the prognostic factors. On multivariate analysis, underlying liver disease (Viral vs. Non-viral; HR, 1.4519; 95% CI 1.0265–2.0535; p = 0.0351), tumor number (solitary vs. multiple; HR, 0.5645; 95% CI 0.4033–0.7903; p = 0.0009), MVI (− vs. + ; HR, 0.4957; 95% CI 0.3637–0.6757; p < 0.0001), and GNRI (low vs. high; HR, 1.8284; 95% CI 1.3598–2.4586; p < 0.0001) were the independent prognostic factors. Univariate analysis of OS showed that tumor differentiation (well/mod vs. poor; HR, 0.5099; 95% CI 0.3227–0.8059; p = 0.0039), MVI (− vs. + ; HR, 0.4837; 95% CI 0.3206–0.7300; p = 0.0005), and GNRI (low vs. high; HR, 1.5935; 95% CI 1.0674–2.3789; p = 0.0227) were the prognostic factors. On multivariate analysis, MVI (− vs. + ; HR, 0.5464; 95% CI 0.3553–0.8403; p = 0.0059), and GNRI (low vs. high; HR, 1.5452; 95% CI 1.0345–2.3079; p = 0.0335) were the independent prognostic factors.

Table 2.

Univariate and multivariate analyses of recurrence-free survival.

RFS Univariate analysis Multivariate analysis
OR 95% CI P value OR 95% CI P value
Age (75 > vs. ≥ 75) 1.0433 0.7797–1.3959 0.7752
Sex (Male vs. Female) 1.2893 0.9260–1.7951 0.1241
Viral vs. Non-viral 1.4269 1.0195–1.9970 0.0325 1.4519 1.0265–2.0535 0.0351
DM (− vs. +) 0.9823 0.7308–1.3205 0.9060
AFP (5 > vs. ≥ 5) 0.5440 0.3864–0.7657 0.0005 0.7886 0.5486–1.1337 0.1998
Surgical procedure (Minor vs. Major) 0.9406 0.6971–1.2690 0.6884
Duration of surgery (300 > vs. ≥ 300) 0.8277 0.6246–1.0968 0.1880
Estimated blood loss (500 > vs. ≥ 500) 0.8957 0.6719–1.1941 0.4529
Tumor diameter (20 ≥ vs. > 20) 0.7635 0.5628–1.0358 0.0829
Tumor number (Solitary vs. Multiple) 0.5588 0.4053–0.7706 0.0004 0.5645 0.4033–0.7903 0.0009
Differentiation (well/mod vs. poor) 0.6494 0.4535–0.9298 0.0184 0.8895 0.6070–1.3035 0.5483
Microvascular invasion (− vs. +) 0.5099 0.3809–0.6825  < 0.0001 0.4957 0.3637–0.6757  < 0.0001
Histological fibrosis grade (F0-2 vs. F3-4) 0.6963 0.5229–0.9272 0.0133 0.7918 0.5879–1.0665 0.1245
GNRI (98 > vs. ≥ 98) 1.6809 1.2645–2.2346 0.0003 1.8284 1.3598–2.4586  < 0.0001

AFP: alpha-fetoprotein, CI: confidence interval, DM: diabetes mellitus, GNRI: geriatric nutritional risk index, OR: odds ratio, RFS: recurrence-free survival.

Table 3.

Univariate and multivariate analyses of overall survival.

OS Univariate analysis Multivariate analysis
OR 95% CI P value OR 95% CI P value
Age (75 > vs. ≥ 75) 0.7049 0.4785–1.0384 0.0769
Sex (Male vs. Female) 0.9949 0.6410–1.5442 0.9818
Viral vs. Non-viral 0.9748 0.6244–1.5219 0.9106
DM (− vs. +) 1.0914 0.7212–1.6516 0.6791
AFP (5 > vs. ≥ 5) 0.6767 0.4255–1.0763 0.0990
Surgical procedure (Minor vs. Major) 0.7704 0.5173–1.1473 0.1992
Duration of surgery (300 > vs. ≥ 300) 0.8989 0.6096–1.3255 0.5907
Estimated blood loss (500 > vs. ≥ 500) 0.7651 0.5190–1.1279 0.1763
Tumor diameter (20 ≥ vs. > 20) 0.7332 0.4772–1.1267 0.1569
Tumor number (Solitary vs. Multiple) 0.7841 0.4996–1.2306 0.2902
Differentiation (well/mod vs. poor) 0.5099 0.3227–0.8059 0.0039 0.6449 0.3996–1.0407 0.0724
Microvascular invasion (− vs. +) 0.4837 0.3206–0.7300 0.0005 0.5464 0.3553–0.8403 0.0059
Histological fibrosis grade (F0-2 vs. F3-4) 0.7846 0.5309–1.1596 0.2235
GNRI (98 > vs. ≥ 98) 1.5935 1.0674–2.3789 0.0227 1.5452 1.0345–2.3079 0.0335

AFP: alpha-fetoprotein, CI: confidence interval, DM: diabetes mellitus, GNRI: geriatric nutritional risk index, OR: odds ratio, OS: overall survival.

Discussion

In the present study, we evaluated the relationship between the GNRI score and the long-term outcomes of HCC after hepatectomy. In the entire cohort, the clinicopathological characteristics were different between the two groups; therefore, PSM was applied to overcome that bias. After PSM, the high-GNRI group demonstrated significantly better RFS and OS according to the log-rank test, and a high GNRI was an independent prognostic factor of better RFS and OS in multivariate analyses. Thus far, only one study has mentioned the relationship between GNRI and the prognosis of HCC. Li et al. reported that a low GNRI resulted in a worse prognosis in HCC; however, their study was limited to hepatitis B virus-associated HCC and patients aged 65 years or older20. To the best of our knowledge, this is the first study that evaluated the relationship between GNRI and the long-term outcomes of HCC to include patients younger than 65 years and whole underlying liver disease in HCC.

Some researchers have demonstrated that immune nutritional score could predict postoperative complications10,18,20,22. Li et al. reported that preoperative low GNRI score in the HCC cohort was associated with postoperative liver failure grade A and B (defined by the International Study Group of Liver Surgery) and severe complications. Additionally, Li et al. revealed that an early postoperative low CONUT score was associated with Clavien-Dindo grade III-V complications after hepatectomy. Preoperative risk assessment will become increasingly important owing to an increasingly aging population globally.

Many studies have described the usefulness of sarcopenia with respect to postoperative complications and cancer prognosis23,24. Although sarcopenia is a powerful tool to evaluate nutritional status, the measurements are complex. Measurements of calf circumference, 6-m walking time, handgrip strength, 5-times sit-to-standing time, and/or the area of the iliopsoas muscle at the third lumbar vertebra level on CT, etc., are required. Child–Pugh score is also a very useful prognostic assessment tool irrespective of surgery25. Vitale et al. reported that in Child–Pugh score B patients, survival after surgery was similar with that after locoregional therapies26. However, in some institutes, including our own, non-surgical therapies such as ablation and transarterial chemoembolization are preferred in cases with Child–Pugh score B. Indeed, the rate of Child–Pugh score B cases was only 3% in the present study. Therefore, we believe another risk assessment tool is warranted. The GNRI is an objective, inexpensive, and readily available assessment tool of nutritional status. Only serum albumin level, body height, and weight are needed to calculate the GNRI. Shoji et al. reported a correlation between the area of the iliopsoas muscle at the third lumbar vertebra level and the GNRI score27. Besides, Cereda and Vanotti indicated that the GNRI was associated with mid-upper arm muscle circumference, arm muscle area, handgrip strength, and handgrip strength/arm muscle area28. Therefore, the GNRI may be a useful tool for predicting postoperative complications and prognosis, especially in patients in whom evaluating sarcopenia is challenging and in institutes that lack the appropriate measuring equipment.

Serum albumin concentration is the main screening tool for immune nutritional status. Hypoalbuminemia induces an impaired immune response, and immunity has a strong influence on cancer prognosis29,30. Additionally, a low albumin level is associated with elevated inflammatory cytokines such as tumor necrosis factor-alpha, interleukin-1, and interleukin-6, which may lead to the progression of HCC31,32. Therefore, a low GNRI may reflect impaired tumor immunity which may cause cancer progression.

BMI is also an immune nutritional index, and several recent studies have mentioned an association between BMI and the response to immune checkpoint inhibitors33,34. A high BMI was reported to be associated with improved survival in patients treated with immune checkpoint inhibitors. Cortellini et al. reported that adipose tissue could activate cytotoxic T-cells and decrease regulatory T-cells. Thus, BMI might influence host immunity. The GNRI is based on serum albumin concentration and BMI, and high values for these components can positively influence host immunity which may improve the cancer prognosis.

Our results indicated that preoperative malnutrition causes worse outcomes after hepatectomy in HCC. Interventions aimed at preoperative nutritional status will improve not only short-term outcomes but also cancer prognosis. Several studies have demonstrated that perioperative nutritional support improves morbidity and prognosis3537. Therefore, we believe that perioperative immune nutritional support should be performed intensively for patients who are planned to undergo hepatectomy.

The present study has some study limitations. First, there is a potential risk of selection bias owing to the single-center, retrospective design. Prospective, multi-institutional studies are needed to validate our results. Second, postoperative complications were not assessed in the present study. Third, differences in patient characteristics between the two groups were not completely overcome. Child–Pugh score and AFP level were significantly different between the two groups even after PSM, which could influence prognosis. Additionally, the follow-up protocol was not standardized, which could make RFS data less powerful. Finally, the evaluation of resectability and the preselection of suitable candidates for surgery are quite complex. Even if liver function is not well preserved, surgery is sometimes performed in cases where the tumor is near the liver surface. In contrast, if the tumor is located around the hepatic hilum or the root of the hepatic veins, we sometimes hesitate to perform surgery even in cases with preserved liver function.

In conclusion, the GNRI is an objective, inexpensive, and easily calculated assessment tool for nutritional status. Our findings suggest that GNRI can be a useful predictor of survival in HCC after hepatectomy. Perioperative nutritional support might improve cancer survival.

Supplementary Information

Supplementary Information (203.1KB, docx)

Acknowledgements

We thank Editage for editing the manuscript (https://www.editage.com).

Author contributions

K.H. designed the study and developed the methodology. K.H., G.Y., S.S., and F.S. collected the data. K.H. analyzed and interpreted the data. K. H. and O.K. wrote and reviewed the main manuscript text. F.F., H.T., A.Y., and O.K. supervised the study.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's note

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

Supplementary Information

The online version contains supplementary material available at 10.1038/s41598-021-88254-z.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J. Clin. 2019;69:7–34. doi: 10.3322/caac.21551. [DOI] [PubMed] [Google Scholar]
  • 2.Kawaguchi T, et al. Changes in prognostic factors for patients with hepatocellular carcinoma underwent transarterial chemoembolization with the transition of the time: Child–Pugh class, Albumin-Bilirubin grade, and then. Ann. Transl. Med. 2020;8:1045. doi: 10.21037/atm-2020-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Márton S, et al. Kinetics of inflammatory markers following cancer-related bowel and liver resection. Ups. J. Med. Sci. 2011;116:124–128. doi: 10.3109/03009734.2010.519446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Krzystek-Korpacka M, et al. Acute-phase response proteins are related to cachexia and accelerated angiogenesis in gastroesophageal cancers. Clin. Chem. Lab. Med. 2008;46:359–364. doi: 10.1515/CCLM.2008.089. [DOI] [PubMed] [Google Scholar]
  • 5.Barao K, Abe Vicente Cavagnari M, Silva Fucuta P, Manoukian Forones N. 2017 Association between nutrition status and survival in elderly patients with colorectal cancer. Nutr. Clin. Pract. 2017;32:658–663. doi: 10.1177/0884533617706894. [DOI] [PubMed] [Google Scholar]
  • 6.Norman K, Pichard C, Lochs H, Pirlich M. Prognostic impact of disease-related malnutrition. Clin. Nutr. 2008;27:5–15. doi: 10.1016/j.clnu.2007.10.007. [DOI] [PubMed] [Google Scholar]
  • 7.Sasaki H, Nagano S, Komiya S, Taniguchi N, Setoguchi T. Validation of different nutritional assessment tools in predicting prognosis of patients with soft tissue spindle-cell sarcomas. Nutrients. 2018;10:765. doi: 10.3390/nu10060765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Matsukawa T, et al. Validation and comparison of prognostic values of GNRI, PNI, and CONUT in newly diagnosed diffuse large B cell lymphoma. Ann. Hematol. 2020;99:2859–2868. doi: 10.1007/s00277-020-04262-5. [DOI] [PubMed] [Google Scholar]
  • 9.Takamizawa Y, et al. Nutritional and inflammatory measures predict survival of patients with stage IV colorectal cancer. BMC Cancer. 2020;20:1092. doi: 10.1186/s12885-020-07560-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Harimoto N, et al. Prognostic significance of preoperative controlling nutritional status (CONUT) score in patients undergoing hepatic resection for hepatocellular carcinoma: A multi-institutional study. Ann. Surg Oncol. 2018;25:3316–3323. doi: 10.1245/s10434-018-6672-6. [DOI] [PubMed] [Google Scholar]
  • 11.Li L, et al. Early postoperative controlling nutritional status (CONUT) score is associated with complication III-V after hepatectomy in hepatocellular carcinoma: A retrospective cohort study of 1,334 patients. Sci. Rep. 2018;8:13406. doi: 10.1038/s41598-018-31714-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Imai D, et al. Prognostic nutritional index is superior as a predictor of prognosis among various inflammation-based prognostic scores in patients with hepatocellular carcinoma after curative resection. Hepatol. Res. 2020;50:101–109. doi: 10.1111/hepr.13431. [DOI] [PubMed] [Google Scholar]
  • 13.Bouillanne O, et al. Geriatric Nutritional Risk Index: A new index for evaluating at-risk elderly medical patients. Am J. Clin Nutr. 2005;82:777–783. doi: 10.1093/ajcn/82.4.777. [DOI] [PubMed] [Google Scholar]
  • 14.Narumi T, et al. The prognostic importance of objective nutritional indexes in patients with chronic heart failure. J. Cardiol. 2013;62:307–313. doi: 10.1016/j.jjcc.2013.05.007. [DOI] [PubMed] [Google Scholar]
  • 15.Kinugasa Y, et al. Geriatric nutritional risk index predicts functional dependency and mortality in patients with heart failure with preserved ejection fraction. Circ. J. 2013;77:705–711. doi: 10.1253/circj.CJ-12-1091. [DOI] [PubMed] [Google Scholar]
  • 16.Yamada K, et al. Simplified nutritional screening tools for patients on maintenance hemodialysis. Am J. Clin Nutr. 2008;87:106–113. doi: 10.1093/ajcn/87.1.106. [DOI] [PubMed] [Google Scholar]
  • 17.Taniguchi E, et al. Nutritional assessments for ordinary medical care in patients with chronic liver disease. Hepatol. Res. 2013;43:192–199. doi: 10.1111/j.1872-034X.2012.01055.x. [DOI] [PubMed] [Google Scholar]
  • 18.Sasaki M, et al. The Geriatric Nutritional Risk Index predicts postoperative complications and prognosis in elderly patients with colorectal cancer after curative surgery. Sci. Rep. 2020;10:10744. doi: 10.1038/s41598-020-67285-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kim YN, et al. Comparison of postoperative nutritional status after distal gastrectomy for gastric cancer using three reconstructive methods: A multicenter study of over 1300 patients. J. Gastrointest Surg. 2020;24:1482–1488. doi: 10.1007/s11605-019-04301-1. [DOI] [PubMed] [Google Scholar]
  • 20.Li L, et al. Geriatric nutritional risk index predicts prognosis after hepatectomy in elderly patients with hepatitis B virus-related hepatocellular carcinoma. Sci. Rep. 2018;8:12561. doi: 10.1038/s41598-018-30906-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Miyake H, Tei H, Fujisawa M. Geriatric nutrition risk index is an important predictor of cancer-specific survival, but not recurrence-free survival, in patients undergoing surgical resection for non-metastatic renal cell carcinoma. Curr. Urol. 2017;10:26–31. doi: 10.1159/000447147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Li L, et al. Early postoperative controlling nutritional status (CONUT) score is associated with complication III–V after hepatectomy in hepatocellular carcinoma: A retrospective cohort study of 1,334 patients. Sci. Rep. 2018;8:13406. doi: 10.1038/s41598-018-31714-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KCH. Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res. 2009;15:6973–6979. doi: 10.1158/1078-0432.CCR-09-1525. [DOI] [PubMed] [Google Scholar]
  • 24.Harimoto N, et al. Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Br. J. Surg. 2013;100:1523–1530. doi: 10.1002/bjs.9258. [DOI] [PubMed] [Google Scholar]
  • 25.Granito A, Bolondi L. Non-transplant therapies for patients with hepatocellular carcinoma and Chil-Pugh-Turcotte class B cirrhosis. Lancet Oncol. 2017;18:e101–e112. doi: 10.1016/S1470-2045(16)30569-1. [DOI] [PubMed] [Google Scholar]
  • 26.Vitale A, et al. Survival benefit of liver resection for patients with hepatocellular carcinoma across different Barcelona Clinic Liver Cancer stages: A multicentre study. J. Hepatol. 2015;62:617–624. doi: 10.1016/j.jhep.2014.10.037. [DOI] [PubMed] [Google Scholar]
  • 27.Shoji F, et al. Relationship between preoperative sarcopenia status and immuno-nutritional parameters in patients with early-stage non-small cell lung cancer. Anticancer Res. 2017;37:6997–7003. doi: 10.21873/anticanres.12168. [DOI] [PubMed] [Google Scholar]
  • 28.Cereda E, Vanotti A. The new Geriatric Nutritional Risk Index is a good predictor of muscle dysfunction in institutionalized older patients. Clin Nutr. 2007;26:78–83. doi: 10.1016/j.clnu.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 29.Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 30.Kanno H, et al. Differences in the immunosurveillance pattern associated with DNA mismatch repair status between right-sided and left-sided colorectal cancer. Cancer Sci. 2020;111:3032–3044. doi: 10.1111/cas.14495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yeh SS, Schuster MW. Geriatric cachexia: The role of cytokines. Am. J. Clin Nutr. 1999;70:183–197. doi: 10.1093/ajcn.70.2.183. [DOI] [PubMed] [Google Scholar]
  • 32.Kotler DP. Cachexia—Review. Ann. Intern. Med. 2000;133:622–634. doi: 10.7326/0003-4819-133-8-200010170-00015. [DOI] [PubMed] [Google Scholar]
  • 33.Ichihara E, et al. The impact of body mass index on the efficacy of anti-PD-1/PD-L1 antibodies in patients with non-small cell lung cancer. Lung Cancer. 2020;139:140–145. doi: 10.1016/j.lungcan.2019.11.011. [DOI] [PubMed] [Google Scholar]
  • 34.Cortellini A, et al. A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: when overweight becomes favorable. J. Immunother Cancer. 2019;7:57. doi: 10.1186/s40425-019-0527-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhao Y, Qin H, Wu Y, Xiang B. Enhanced recovery after surgery program reduces length of hospital stay and complications in liver resection: A PRISMA-compliant systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore) 2017;96:7628. doi: 10.1097/MD.0000000000007628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Okabayashi T, et al. Effects of branched-chain amino acids-enriched nutrient support for patients undergoing liver resection for hepatocellular carcinoma. J. Gastroenterol. Hepatol. 2008;23:1869–1873. doi: 10.1111/j.1440-1746.2008.05504.x. [DOI] [PubMed] [Google Scholar]
  • 37.Gustafsson UO, Oppelstrup H, Thorell A. Adherence to the ERAS protocol is associated with 5-year survival after colorectal cancer surgery: A retrospective cohort study. World J. Surg. 2016;40:1741–1747. doi: 10.1007/s00268-016-3460-y. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Information (203.1KB, docx)

Articles from Scientific Reports are provided here courtesy of Nature Publishing Group

RESOURCES