Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Prognostic value of postoperative decrease in serum albumin on surgically resected early-stage non-small cell lung carcinoma: A multicenter retrospective study

  • Fumihiko Kinoshita,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Tetsuzo Tagawa ,

    Roles Conceptualization, Data curation, Formal analysis, Project administration, Supervision, Validation, Visualization, Writing – review & editing

    t_tagawa@surg2.med.kyushu-u.ac.jp

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Takanori Yamashita,

    Roles Conceptualization, Data curation, Methodology

    Affiliation Medical Information Center, Kyushu University Hospital, Fukuoka, Japan

  • Tomoyoshi Takenaka,

    Roles Formal analysis, Project administration, Supervision, Validation

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Taichi Matsubara,

    Roles Data curation, Investigation

    Affiliation Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan

  • Gouji Toyokawa,

    Roles Data curation, Investigation

    Affiliation Department of Thoracic Surgery, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan

  • Kazuki Takada,

    Roles Data curation, Formal analysis, Methodology

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Taro Oba,

    Roles Data curation, Formal analysis

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Atsushi Osoegawa,

    Roles Data curation, Formal analysis

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

  • Koji Yamazaki,

    Roles Supervision

    Affiliation Department of Thoracic Surgery, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan

  • Mitsuhiro Takenoyama,

    Roles Supervision

    Affiliation Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan

  • Mototsugu Shimokawa,

    Roles Formal analysis, Investigation, Methodology, Supervision, Validation

    Affiliation Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan

  • Naoki Nakashima,

    Roles Supervision, Writing – review & editing

    Affiliation Medical Information Center, Kyushu University Hospital, Fukuoka, Japan

  • Masaki Mori

    Roles Supervision, Writing – review & editing

    Affiliation Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Abstract

Background

Preoperative nutritional status is an important host-related prognostic factor for non-small cell lung carcinoma (NSCLC); however, the significance of postoperative changes in nutritional status remains unclear. This study aimed to elucidate the significance of postoperative decreases in serum albumin (ΔAlb) on the outcomes of early-stage NSCLC.

Methods

We analyzed 443 training cohort (TC) and 642 validation cohort (VC) patients with pStage IA NSCLC who underwent surgery and did not recur within 1 year. We measured preoperative serum albumin levels (preAlb) and postoperative levels 1 year after surgery (postAlb), and calculated ΔAlb as (preAlb − postAlb)/preAlb × 100%. A cutoff value of 11% for ΔAlb was defined on the basis of the receiver operating characteristic curve for the TC.

Results

Patients were divided into ΔAlb-Decreased and ΔAlb-Stable groups, including 100 (22.6%) and 343 (77.4%) in the TC, and 58 (9.0%) and 584 (90.1%) in the VC. ΔAlb-Decreased was associated with male sex (p = 0.0490), smoking (p = 0.0156), and non-adenocarcinoma (p<0.0001) in the TC, and pT1b (p = 0.0169) and non-adenocarcinoma (p = 0.0251) in the VC. Multivariable analysis identified ΔAlb as an independent prognostic factor for disease-free survival (DFS) and overall survival (OS) in both cohorts (VC: DFS, HR = 1.9, 95%CI: 1.10–3.15, p = 0.0197; OS, HR = 2.0, 95%CI: 1.13–3.45, p = 0.0173). Moreover, subgroup analysis demonstrated that the prognostic value of ΔAlb was consistent for age, sex, smoking history, surgical procedure, and histological type.

Conclusion

We demonstrated a negative impact of postoperative decrease of the serum albumin on the prognosis of patients with early-stage NSCLC. Postoperative changes in nutritional status might be important in NSCLC outcomes.

Introduction

The preoperative nutritional status of patients, reflected by factors such as serum albumin [1], Controlling Nutritional Status score [2,3], geriatric nutritional risk index [4], prognostic nutritional index [5,6], Glasgow prognostic score [7], body mass index [8,9], and skeletal muscle area (SMA) [1013], has attracted much attention as an important host-related prognostic factor in patients with non-small cell lung carcinoma (NSCLC).

A previous study showed that a postoperative decrease in serum albumin (ΔAlb) 1 day after surgery was associated with postoperative pulmonary complications in patients with NSCLC [14]. However, the prognostic significance of ΔAlb on the long-term outcomes of NSCLC patients after surgery remains unclear.

We previously showed that a decrease in SMA 1 year after surgery was associated with a poor prognosis of NSCLC [12,13]. Furthermore, a decrease in SMA correlated with exacerbation of nutritional indices, and postoperative deterioration of nutritional status was related to a poor prognosis of NSCLC [13].

In this study, we analyzed the clinical significance of ΔAlb at 1 year after surgery, as a convenient marker of nutritional status, on the long-term outcomes of patients with pathological stage (pStage) IA NSCLC.

Patients and methods

Study patients

This study was reviewed and approved by our institutional review boards (Kyushu University Hospital [KUH], IRB No. 2019–232; Kyushu Cancer Center [KCC], IRB No. 2019–56; Kyushu Medical Center [KMC], IRB No. 19D152). Regarding participant consent, the informed consent for this research was waived by the ethics committee.

This study included 1210 Japanese patients with pStage IA NSCLC who underwent surgical resection between January 2003 and December 2014 at KUH (545 patients) and between January 2009 and December 2014 at KCC (324 patients) and KMC (341 patients). Of these, 59 patients who recurred within 1 year after surgery were excluded (KUH, 44 patients; KCC, six patients; KMC, nine patients). Furthermore, because of incomplete resection, pretreatment before surgery, or lack of data, 66 patients were also excluded (KUH, 58 patients; KCC, three patients; KMC, five patients). Patients who received chemotherapy within 1 year after surgery including adjuvant chemotherapy were also excluded from this study. Finally, 1085 patients were enrolled in this study (KUH, 443 patients; KCC, 315 patients; KMC, 327 patients). We first examined patients from KUH as a training cohort (TC). Subsequently, we validated our findings in an independent validation cohort (VC) composed of patients from KCC and KMC. A total of 443 and 642 patients were enrolled in the TC and VC, respectively. The consort diagram of patient selection was shown in Fig 1.

thumbnail
Fig 1. Consort diagram of patient selection.

pStage, pathological stage; NSCLC, non-small cell lung carcinoma.

https://doi.org/10.1371/journal.pone.0256894.g001

Clinicopathological characteristics, disease-free survival (DFS), and overall survival (OS) were analyzed retrospectively. Clinicopathological characteristics included age, sex, smoking history, pulmonary comorbidities, surgical procedure, pathological T status (pT), histological type, vascular invasion, and lymphatic invasion. Pulmonary comorbidities included interstitial pneumonia, chronic obstructive pulmonary disease, or asthma. The pStage was defined according to the 7th edition of TNM classification [15]. The clinical information and follow-up data were obtained from the patients’ medical records.

Evaluation of serum albumin

Preoperative serum albumin levels (preAlb) were measured before surgery and postoperative serum albumin levels (postAlb) at the closest point to 12 months at 9–15 months after surgery. For measuring serum albumin levels, all of three institutions used the modified bromocresol purple method. ΔAlb was calculated as (preAlb − postAlb)/preAlb × 100%, as in previous reports [14,16,17].

Statistical analysis

All statistical analyses were performed using JMP pro 14.0 software (SAS Institute). Patient characteristics were analyzed by Pearson’s χ2 test. DFS was defined as the time between surgery and the date of last follow-up, recurrence, or death; OS was the time between surgery and the date of last follow-up or death. Survival curves were estimated by the Kaplan–Meier method with the log-rank test. Hazard ratios for positive risk factors were estimated by Cox proportional hazards regression analysis with backwards elimination. As for multivariable analysis, after applying multivariable analysis to all factors used in univariable analysis at the same time, we eliminated less important factors with high p-values one by one and reduced factors so that only factors with p<0.05 were included. All results were considered statistically significant at p<0.05.

Results

Patient characteristics

A total of 1085 patients with pStage IA NSCLC who underwent complete surgical resection and did not recur within 1 year were enrolled in this study. We classified 443 patients from KUH as the TC, and 642 patients from KCC and KMC as the VC. The detailed clinicopathological characteristics of the two cohorts are shown in S1 Table. In the TC, 214 (48.3%), 223 (50.3%), and 232 (52.4%) patients were of older age (≥70 years), male, and smoked, respectively. The median follow-up period was 5.47 years (range, 1.01–15.97 years). In terms of the VC, 279 (43.5%), 283 (44.1%), and 295 (46.0%) patients were of older age (≥70 years), male, and smoked, respectively, and the median follow-up period was 5.75 years (range, 1.36–10.59 years).

In terms of serum albumin, the median preAlb and ΔAlb in the TC were 4.3 g/dL (range, 3.1 to 5.3 g/dL) and 4.8% (range, -29.2 to 79.2%), respectively. In the VC, the median preAlb and ΔAlb in the TC were 4.3 g/dL (range, 3.0 to 5.4 g/dL) and 2.4% (range, -30.0 to 43.9%), respectively. The histograms of distributions of preAlb and ΔAlb in each cohort were shown in S1 Fig.

Clinicopathological characteristics associated with ΔAlb

The cutoff value of ΔAlb was set as 11% based on the receiver operating characteristic curve (ROC) curve for 5-year OS in the TC (Fig 2). One hundred (22.6%) patients in the TC and 58 (9.0%) patients in the VC were classified as ΔAlb-Decreased (ΔAlb ≥11%), respectively. The association between clinicopathological characteristics and ΔAlb is shown in Table 1. ΔAlb-Decreased in the TC was significantly associated with male sex (p = 0.0490), smoking (p = 0.0156), and non-adenocarcinoma (p<0.0001). In the VC, ΔAlb-Decreased was significantly associated with pT1b (p = 0.0169) and non-adenocarcinoma (p = 0.0251).

thumbnail
Fig 2. Receiver operating characteristic curve (ROC) of training cohort (TC).

ROC curves for postoperative decrease in serum albumin (ΔAlb) in the TC. AUC: Area under the curve.

https://doi.org/10.1371/journal.pone.0256894.g002

thumbnail
Table 1. Associations of clinicopathological characteristics with ΔAlb.

https://doi.org/10.1371/journal.pone.0256894.t001

Survival analysis according to ΔAlb

We evaluated the difference in survival between the ΔAlb-Decreased and ΔAlb-Stable groups by Kaplan–Meier analysis. DFS and OS of the ΔAlb-Decreased group in the TC were significantly poorer compared with the ΔAlb-Stable group (5-year DFS, 77.2% versus 94.8%, p<0.0001; 5-year OS, 84.1% versus 98.2%, p<0.0001) (Fig 3A and 3B). Furthermore, in the VC, DFS and OS were also significantly worse in the ΔAlb-Decreased group compared with the ΔAlb-Stable group (5-year DFS, 78.0% versus 87.7%, p = 0.0015; 5-year OS, 83.4% versus 90.6%, p = 0.0010) (Fig 3C and 3D).

thumbnail
Fig 3. Survival analysis according to postoperative decrease in serum albumin (ΔAlb).

Disease-free survival and overall survival of the ΔAlb-Decreased and ΔAlb-Stable groups in the training cohort (A, B) and in the validation cohort (C, D).

https://doi.org/10.1371/journal.pone.0256894.g003

Survival analysis according to preAlb

The cutoff value of preAlb was set as 4.1 g/dL based on the ROC curve for 5-year OS in the TC (S2 Fig). The clinicopathological characteristics according to preAlb are shown in S2 Table, and preAlb-Low was significantly associated with poor prognosis in both the TC and VC (S3 Fig).

Univariable and multivariable analyses of survival

We performed univariable and multivariable analyses of survival. In the TC, multivariable analysis identified smoker (p = 0.0233), lobectomy (p = 0.0175), adenocarcinoma (p = 0.0187), lymphatic invasion (p = 0.0180), and ΔAlb-Decreased (p = 0.0003) as independent prognostic factors for DFS, and lobectomy (p = 0.0015), adenocarcinoma (p = 0.0004), ΔAlb-Decreased (p<0.0001), and preAlb-Low (p = 0.0010) as independent factors for OS (S3 Table). In the VC, multivariable analysis identified older age (p<0.0001), male sex (p<0.0001), pulmonary comorbidity (p = 0.0017), and ΔAlb-Decreased (p = 0.0186) as independent prognostic factors for DFS, and older age (p<0.0001), male sex (p = 0.0001), pulmonary comorbidity (p = 0.0006), lobectomy (p = 0.0247), and ΔAlb-Decreased (p = 0.0145) as independent prognostic factors for OS (Table 2).

thumbnail
Table 2. Univariable and multivariable analyses of disease-free survival and overall survival in the validation cohort.

https://doi.org/10.1371/journal.pone.0256894.t002

Furthermore, a forest plot of the subgroup analysis demonstrated that the prognostic value of ΔAlb was almost consistent across different subgroups in both the TC (S4 Fig) and VC (Fig 4).

thumbnail
Fig 4. Forest plot of subgroup analysis for postoperative decrease in serum albumin (ΔAlb) in the validation cohort (VC).

Hazard ratios of ΔAlb for (A) disease-free survival and (B) overall survival in the VC. HR: Hazard ratio, CI: Confidence interval.

https://doi.org/10.1371/journal.pone.0256894.g004

Combined survival analysis of ΔAlb and preAlb

To elucidate the prognostic significance of ΔAlb in more detail, we performed a combined analysis of ΔAlb and preAlb. Patients were categorized into the following four groups: Alb-Low-Decreased (preAlb <4.1g/dL and ΔAlb ≥11%), Alb-Low-Stable (preAlb <4.1g/dL and ΔAlb <11%), Alb-High-Decreased (preAlb ≥4.1g/dL and ΔAlb ≥11%), and Alb-High-Stable (preAlb ≥4.1g/dL and ΔAlb <11%). DFS and OS differed significantly among the four groups in both the TC (p<0.0001 and p<0.0001, respectively) and VC (p = 0.0006 and p = 0.0001, respectively) (Fig 5A and 5B). The 5-year DFS rates in the Alb-Low-Decreased, Alb-Low-Stable, Alb-High-Decreased, and Alb-High-Stable groups were 48.3%, 92.8%, 82.4%, and 95.3% in the TC, respectively, and 68.6%, 81.4%, 79.2%, and 89.2% in the VC, respectively (Fig 5C and 5D). The 5-year OS rates were 54.3%, 93.9%, 89.3%, and 99.4% in the TC, respectively, and 68.6%, 86.0%, 85.3%, and 91.5% in the VC, respectively. The Alb-Low-Decreased group had the worst prognosis for DFS and OS; however, DFS and OS of the Alb-Low-Stable group were not significantly worse compared with the Alb-High-Decreased group and the Alb-High-Stable group in both cohorts.

thumbnail
Fig 5. Combined survival analysis of preoperative serum albumin levels (preAlb) and postoperative decrease in serum albumin (ΔAlb).

Disease-free survival and overall survival of the Alb-Low-Decreased, Alb-Low-Stable, Alb-High-Decreased, and Alb-High-Stable groups in the training cohort (A, B) and in the validation cohort (C, D).

https://doi.org/10.1371/journal.pone.0256894.g005

Discussion

The present study demonstrated the prognostic impact of ΔAlb on the long-term outcomes of patients with pStage IA NSCLC. Moreover, our subgroup analysis demonstrated that the prognostic value of ΔAlb was consistent for age, sex, smoking history, surgical procedure, and histological type. In addition, combined survival analysis of ΔAlb and preAlb suggested that, even if nutritional status was poor before surgery, patients with maintained nutritional status had a better prognosis.

Serum albumin is one of the most convenient and important indicators for assessing nutritional status, and previous reports have identified preAlb as an essential prognostic factor for several cancers, including NSCLC [1,18]. Nutritional status is an important determinant of the immune response, and decreased albumin levels may be associated with an impaired antitumor immune response [19,20]. In addition, low levels of serum albumin reflect systemic inflammation, which is significantly associated with cancer progression [2123]. Serum albumin is also an endogenous antioxidant [24,25]. Although the association between endogenous antioxidants and cancer progression remains unclear, a previous study demonstrated that loss of albumin resulted in a plasma redox imbalance and promoted cancer growth and metastasis [25]. These findings may help to explain the association between serum albumin and the prognosis of NSCLC.

We previously showed the prognostic significance of postoperative changes in nutritional status in patients with NSCLC by analyzing the postoperative decrease in SMA 1 year after surgery [12,13]. Despite its usefulness, evaluation of SMA requires complicated analysis of computed tomography results. In contrast, serum albumin can be conveniently measured by simple blood testing, suggesting that this may be a suitable marker for monitoring the nutritional condition of patients.

In the cancer-bearing state, it is well known that albumin consumption by cancer cells causes albumin depletion [26]. Therefore, when considering the significance of ΔAlb, recurrence within 1 year must be a confounding factor in the association between ΔAlb and the outcomes of NSCLC. To eliminate the influence of recurrence within 1 year, this study excluded patients who recurred within 1 year; then, the prognostic impact of ΔAlb seemed to be independent from recurrence within 1 year. However, in patients with early-stage NSCLC who underwent complete resection, postoperative albumin depletion might reflect a potential cancer recurrence before an apparent cancer recurrence is identified by tests such as computed tomography and tumor markers. Several studies to detect circulating cancer cells [2730] and circulating tumor DNA [3133] in blood are underway to investigate potential cancer existence and metastasis in NSCLC. Our results might be supportive to these studies, and it seems useful to investigate changes in serum albumin along with techniques for detection of micro cancer cells. Although adjuvant chemotherapy after complete resection is not currently recommended for pStage IA NSCLC, future improvements in techniques for detecting micro cancer cells, including decreased albumin, might allow the selection of cases that would benefit from adjuvant chemotherapy.

Our combined survival analysis of preAlb and ΔAlb clarified the clinical significance of ΔAlb in terms of the outcomes of patients with NSCLC. In particular, the prognosis of patients in the Alb-Low-Stable group was not significantly worse compared with those of the Alb-High-Decreased and the Alb-High-Stable groups. This result suggested that, even if the preoperative nutritional status was poor, the prognosis of patients with a maintained nutritional status was favorable. From another point of view, even in the preAlb-High group, the prognosis of the Alb-High-Decreased group, in which serum albumin levels were decreased after surgery, was worse than that of the Alb-High-Stable group, and equal to or worse than that of the Alb-Low-Stable group.

Several drugs, including megestrol acetate, ghrelin agonists, and anti-myostatin peptides, are expected to improve cancer cachexia, in addition to nutritional support and physical exercise [3437]. Recently, anamorelin, which is an orally active ghrelin receptor agonist, has been shown the usefulness for improving cancer cachexia [3840]. Although there has been no direct evaluation of serum albumin levels, blood sampling data on nutrition, such as prealbumin, have been shown to improve with oral anamorelin [38], and we expect that serum albumin levels may also improve by anamorelin. In addition, in the perioperative period of NSCLC, enteral nutrition in addition to active rehabilitation has been shown to improve albumin levels [41], and a randomized controlled trial has been conducted to determine the changes in nutritional status when enteral nutrition is added to perioperative rehabilitation of NSCLC [42]. Therefore, we consider that these strategies might be applied to NSCLC patients after surgery to maintain their nutritional status, leading to improved outcomes.

This study had several limitations. This was a retrospective study; however, the study cohort was relatively large, and 1085 patients were enrolled. We excluded patients who recurred or died within 1 year, and the prognostic impact of preAlb was therefore probably underestimated. In addition, we analyzed ΔAlb at 1 year after surgery in this study, and ΔAlb at 1 year was significantly associated with the subsequent prognosis of early-stage NSCLC; however, to further clarify how ΔAlb is related to the recurrence and prognosis of NSCLC, more detailed examinations of changes in serum albumin over time using serum albumin levels at other time points, such as 1 month, 3 months, and 6 months after surgery, may be required. Furthermore, serum albumin can be influenced by various diseases as well as cancer, such as heart, liver, and renal disease, and care is therefore needed in interpreting the meaning of serum albumin levels in clinical practice. In addition, postoperative smoking status might be one of the related factors with the postoperative nutritional changes of the patients; however, we have not been able to collect the data on postoperative smoking status and evaluate the influence of postoperative smoking status in this study. There were several limitations in pathological information, such as the TNM classification and adenocarcinoma subtypes; then, it seems necessary to perform the analysis using 8th edition or detailed adenocarcinoma subtypes in the future.

In conclusion, the results of this study demonstrated that ΔAlb was significantly associated with the prognosis of early-stage NSCLC. Therefore, postoperative changes in nutritional status might be important for the outcomes of NSCLC.

Supporting information

S1 Fig. The histograms of distributions of preoperative serum albumin levels (preAlb) and postoperative decrease in serum albumin (ΔAlb).

The histograms of distributions of preAlb and ΔAlb in the training cohort (A, B) and in the validation cohort (C, D).

https://doi.org/10.1371/journal.pone.0256894.s001

(TIF)

S2 Fig. Receiver operating characteristic curve (ROC) of training cohort (TC).

ROC curves for preoperative serum albumin levels (preAlb) in the TC. AUC: area under the curve.

https://doi.org/10.1371/journal.pone.0256894.s002

(TIF)

S3 Fig. Survival analysis according to preoperative serum albumin levels (preAlb).

Disease-free survival and overall survival of the preAlb-Low and preAlb-High groups in the training cohort (A, B) and in the validation cohort (C, D).

https://doi.org/10.1371/journal.pone.0256894.s003

(TIF)

S4 Fig. Forest plot of subgroup analysis for postoperative decrease in serum albumin (ΔAlb) in the training cohort (TC).

Hazard ratios of ΔAlb for (A) disease-free survival and (B) overall survival in the TC. HR: hazard ratio, CI: confidence interval.

https://doi.org/10.1371/journal.pone.0256894.s004

(TIF)

S1 Table. Clinicopathological characteristics of patients with pStage IA non-small cell lung carcinoma.

https://doi.org/10.1371/journal.pone.0256894.s005

(DOCX)

S2 Table. Associations of clinicopathological characteristics with preAlb.

https://doi.org/10.1371/journal.pone.0256894.s006

(DOCX)

S3 Table. Univariable and multivariable analyses of disease-free survival and overall survival in the training cohort.

https://doi.org/10.1371/journal.pone.0256894.s007

(DOCX)

Acknowledgments

We thank Takashi Kinoshita from the Medical Information Center, Kyushu University Hospital, for invaluable help with the data collection. We also thank H. Nikki March, PhD, and Susan Furness, PhD, from Edanz Group (https://en-author-services.edanz.com/ac) for editing a draft of this manuscript.

References

  1. 1. Miura K, Hamanaka K, Koizumi T, et al. (2017) Clinical significance of preoperative serum albumin level for prognosis in surgically resected patients with non-small cell lung cancer: Comparative study of normal lung, emphysema, and pulmonary fibrosis. Lung Cancer. 111:88–95. pmid:28838406
  2. 2. Shoji F, Haratake N, Akamine T, et al. (2017) The preoperative controlling nutritional status score predicts survival after curative surgery in patients with pathological stage i non-small cell lung cancer. Anticancer Res. 37:741–748. pmid:28179325
  3. 3. Toyokawa G, Kozuma Y, Matsubara T, et al. (2017) Prognostic impact of controlling nutritional status score in resected lung squamous cell carcinoma. J Thorac Dis. 9:2942–2951. pmid:29221266
  4. 4. Shoji F, Matsubara T, Kozuma Y, et al. (2017) Preoperative Geriatric Nutritional Risk Index: A predictive and prognostic factor in patients with pathological stage I non-small cell lung cancer. Surg Oncol. 26:483–488. pmid:29113668
  5. 5. Okada S, Shimada J, Kato D, et al. (2017) Clinical significance of prognostic nutritional index after surgical treatment in lung cancer. Ann Thorac Surg. 104:296–302. pmid:28433217
  6. 6. Li D, Yuan X, Liu J, et al. (2018) Prognostic value of prognostic nutritional index in lung cancer: A meta-analysis. J Thorac Dis. 10:5298–5307. pmid:30416777
  7. 7. Yotsukura M, Ohtsuka T, Kaseda K, et al. (2016) Value of the Glasgow Prognostic Score as a prognostic factor in resectable non–small cell lung cancer. J Thorac Oncol. 11:1311–1318. pmid:27234603
  8. 8. Morel H, Raynard B, d’Arlhac M, et al. (2018) Prediagnosis weight loss, a stronger factor than BMI, to predict survival in patients with lung cancer. Lung Cancer. 126:55–63. pmid:30527193
  9. 9. Shepshelovich D, Xu W, Lu L, et al. (2019) Body mass index (BMI), BMI change, and overall survival in patients with SCLC and NSCLC: A pooled analysis of the International Lung Cancer Consortium. J Thorac Oncol. 14:1594–1607. pmid:31163278
  10. 10. Martin L, Birdsell L, MacDonald N, et al. (2013) Cancer cachexia in the age of obesity: Skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 31:1539–1547. pmid:23530101
  11. 11. Buentzel J, Heinz J, Bleckmann A, et al. (2019) Sarcopenia as prognostic factor in lung cancer patients: A systematic review and meta-analysis. Anticancer Res. 39:4603–4612. pmid:31519557
  12. 12. Takamori S, Toyokawa G, Okamoto T, et al. (2018) Clinical impact and risk factors for skeletal muscle loss after complete resection of early non-small cell lung cancer. Ann Surg Oncol. 25:1229–1236. pmid:29327178
  13. 13. Takamori S, Tagawa T, Toyokawa G, et al. (2020) Prognostic impact of postoperative skeletal muscle decrease in non-small cell lung cancer. Ann Thorac Surg. 109:914–920. pmid:31655044
  14. 14. Li P, Li J, Lai Y, et al. (2018) Perioperative changes of serum albumin are a predictor of postoperative pulmonary complications in lung cancer patients: A retrospective cohort study. J Thorac Dis. 10:5755–5763. pmid:30505483
  15. 15. Goldstraw P, Crowley J, Chansky K, et al. (2007) The IASLC lung cancer staging project: Proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM classification of malignant tumours. J Thorac Oncol. 2:706–714. pmid:17762336
  16. 16. Liu ZJ, Ge XL, Ai SC, et al. (2017) Postoperative decrease of serum albumin predicts short-term complications in patients undergoing gastric cancer resection. World J Gastroenterol. 23:4978–4985. pmid:28785152
  17. 17. Spolverato G, Kim Y, Ejaz A, et al. (2015) Effect of relative decrease in blood hemoglobin concentrations on postoperative morbidity in patients who undergo major gastrointestinal surgery. JAMA Surg. 150:949–956. pmid:26222497
  18. 18. Gupta D, Lis CG. (2010) Pretreatment serum albumin as a predictor of cancer survival: A systematic review of the epidemiological literature. Nutr J. 9:1–16. pmid:20056003
  19. 19. Walrand S, Moreau K, Caldefie F, et al. (2001) Specific and nonspecific immune responses to fasting and refeeding differ in healthy young adult and elderly persons. Am J Clin Nutr. 74:670–678. pmid:11684537
  20. 20. Marcos A, Nova E, Montero A. (2003) Changes in the immune system are conditioned by nutrition. Eur J Clin Nutr. 57:S66–S69. pmid:12947457
  21. 21. Coussens LM, Werb Z. Coussens LM, et al. (2002) Inflammation and cancer. Nature. 420:860–867. pmid:12490959
  22. 22. Mantovani A, Allavena P, Sica A, et al. (2008) Cancer-related inflammation. Nature. 454:436–444. pmid:18650914
  23. 23. Allin KH, Bojesen SE, Nordestgaard BG. (2016) Inflammatory biomarkers and risk of cancer in 84,000 individuals from the general population. Int J Cancer. 139:1493–1500. pmid:27194008
  24. 24. Kühn T, Sookthai D, Graf ME, et al. (2017) Albumin, bilirubin, uric acid and cancer risk: Results from a prospective population-based study. Br J Cancer. 117:1572–1579. pmid:28898231
  25. 25. Inoue M, Nakashima R, Enomoto M, et al. (2018) Plasma redox imbalance caused by albumin oxidation promotes lung-predominant NETosis and pulmonary cancer metastasis. Nat Commun. 9:1–11. pmid:29317637
  26. 26. Finicle BT, Jayashankar V, Edinger AL. (2018) Nutrient scavenging in cancer. Nat Rev Cancer. 18:619–633. pmid:30097614
  27. 27. Ichimura H, Nawa T, Yamamoto Y, et al. (2020) Detection of circulating tumor cells in patients with lung cancer using metallic micro-cavity array filter: A pilot study. Mol Clin Oncol. 12:278–283. pmid:32064107
  28. 28. Dandachi N, Tiran V, Lindenmann J, et al. (2017) Frequency and clinical impact of preoperative circulating tumor cells in resectable non-metastatic lung adenocarcinomas. Lung Cancer. 113:152–157. pmid:29110843
  29. 29. Sawabata N, Nakamura T, Kawaguchi T, et al. (2020) Circulating tumor cells detected only after surgery for non-small cell lung cancer: Is it a predictor of recurrence? J Thorac Dis. 12:4623–4632. pmid:33145035
  30. 30. Tamminga M, de Wit S, van de Wauwer C, et al. (2020) Analysis of released circulating tumor cells during surgery for non-small cell lung cancer. Clin Cancer Res. 26:1656–1666. pmid:31772122
  31. 31. Liang W, Zhao Y, Huang W, et al. (2019) Non-invasive diagnosis of early-stage lung cancer using high-throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA). Theranostics. 9:2056–2070. pmid:31037156
  32. 32. Chen K, Zhao H, Shi Y, et al. (2019) Perioperative dynamic changes in circulating tumor DNA in patients with lung cancer (Dynamic). Clin Cancer Res. 25:7058–7067. pmid:31439586
  33. 33. Chae YK, Oh MS. (2019) Detection of Minimal Residual Disease Using ctDNA in Lung Cancer: Current Evidence and Future Directions. J Thorac Oncol. 14:16–24. pmid:30296486
  34. 34. Argilés JM, López-Soriano FJ, Stemmler B, et al. (2017) Novel targeted therapies for cancer cachexia. Biochem J. 474:2663–2678. pmid:28751550
  35. 35. Sadeghi M, Keshavarz-Fathi M, Baracos V, et al. (2018) Cancer cachexia: Diagnosis, assessment, and treatment. Crit Rev Oncol Hematol. 127:91–104. pmid:29891116
  36. 36. Argilés JM, López-Soriano FJ, Stemmler B, et al. (2019) Therapeutic strategies against cancer cachexia. Eur J Transl Myol. 29:4–13. pmid:31019661
  37. 37. Del Fabbro E. (2019) Combination therapy in cachexia. Ann Palliat Med. 8:59–66. pmid:30180745
  38. 38. Katakami N, Uchino J, Yokoyama T, et al. (2018) Anamorelin (ONO-7643) for the treatment of patients with non-small cell lung cancer and cachexia: Results from a randomized, double-blind, placebo-controlled, multicenter study of Japanese patients (ONO-7643-04). Cancer. 124:606–616. pmid:29205286
  39. 39. Takayama K, Katakami N, Yokoyama T, et al. (2016) Anamorelin (ONO-7643) in Japanese patients with non-small cell lung cancer and cachexia: results of a randomized phase 2 trial. Support Care Cancer. 24:3495–505. pmid:27005463
  40. 40. Temel JS, Abernethy AP, Currow DC, et al. (2016) Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol. 17:519–531. pmid:26906526
  41. 41. Ding Q, Chen W, Gu Y, et al. (2020) Accelerated rehabilitation combined with enteral nutrition in the management of lung cancer surgery patients. Asia Pac J Clin Nutr. 29:274–279. pmid:32674235
  42. 42. Ji X, Ding H. (2020) The efficacy of enteral nutrition combined with accelerated rehabilitation in non-small cell lung cancer surgery: A randomized controlled trial protocol. Medicine (Baltimore). 99:e23382. pmid:33235112