Skip to main content

Advertisement

Log in

Comparison of Inflammation-Based Prognostic Scores Associated with the Prognostic Impact of Adenocarcinoma of Esophagogastric Junction and Upper Gastric Cancer

  • Thoracic Oncology
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

Several inflammation-based prognostic scores have a prognostic value in patients with various cancers. This study investigated the prognostic value of various inflammation-based prognostic scores in patients who underwent a surgery for adenocarcinoma of the esophagogastric junction (AEG) and upper gastric cancer (UGC).

Methods

We reviewed data of 206 patients who underwent surgery for AEG and UGC. We calculated neutrophil–lymphocyte ratio (NLR), platelet–lymphocyte ratio (PLR), Glasgow Prognostic Score (GPS), modified GPS (mGPS), C-reactive protein (CRP)/albumin (Alb) ratio, prognostic index (PI), and prognostic nutritional index (PNI) and analyzed the relationship between these biomarkers and postoperative prognosis.

Results

In multivariate analyses for overall survival, mGPS (P = 0.0337, hazard ratio [HR] = 5.211), PI (P = 0.0002, HR = 21.20), and PNI (P < 0.0001, HR = 6.907) were identified as independent predictive factors. A multivariate analysis for recurrence-free survival showed that only PI (P = 0.0006, HR = 11.89) and PNI (P = 0.0002, HR = 4.972) were independent predictive factors among the above-mentioned inflammation-based prognostic scores.

Conclusions

In various inflammation-based prognostic scores, PI and PNI were more strongly associated with poor prognosis in patients who underwent surgery for AEG and UGC.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.

    Article  Google Scholar 

  2. Lu J, Zheng CH, Cao LL, et al. The effectiveness of the 8th American joint committee on cancer TNM classification in the prognosis evaluation of gastric cancer patients: a comparative study between the 7th and 8th editions. Eur J Surg Oncol. 2017;43(12):2349–56.

    Article  Google Scholar 

  3. Jomrich G, Hollenstein M, John M, et al. The modified Glasgow prognostic score is an independent prognostic indicator in neoadjuvantly treated adenocarcinoma of the esophagogastric junction. Oncotarget. 2018;9(6):6968–76.

    Article  Google Scholar 

  4. Park JH, Watt DG, Roxburgh CS, et al. Colorectal cancer, systemic inflammation, and outcome: Staging the tumor and staging the host. Ann Surg. 2016;263:326–36.

    Article  Google Scholar 

  5. Roxburgh CS, McMillan DC. Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol. 2010;6:149–63.

    Article  CAS  Google Scholar 

  6. Kudou K, Saeki H, Nakashima Y, et al. Postoperative skeletal muscle loss predicts poor prognosis of adenocarcinoma of upper stomach and esophagogastric junction. World J Surg. 2018; https://doi.org/10.1007/s00268-018-4873-6.

    Article  PubMed  Google Scholar 

  7. Tamandl D, Paireder M, Asari R, et al. Markers of sarcopenia quantified by computed tomography predict adverse long-term outcome in patients with resected oesophageal or gastro-oesophageal junction cancer. Eur Radiol. 2016;26:1359–67.

    Article  Google Scholar 

  8. Vayrynen JP, Tuomisto A, Klintrup K, et al. Detailed analysis of inflammatory cell infiltration in colorectal cancer. Br J Cancer. 2013;109:1839–47.

    Article  CAS  Google Scholar 

  9. Miyamoto Y, Baba Y, Sakamoto Y, et al. Sarcopenia is a negative prognostic factor after curative resection of colorectal cancer. Ann Surg Oncol. 2015;22:2663–8.

    Article  Google Scholar 

  10. Kudou K, Saeki H, Nakashima Y, et al. Prognostic significance of sarcopenia in patients with esophagogastric junction cancer or upper gastric cancer. Ann Surg Oncol. 2017;24(7):1804–10.

    Article  Google Scholar 

  11. Kinoshita A, Onoda H, Imai N, et al. Comparison of the prognostic value of inflammation-based prognostic scores in patients with hepatocellular carcinoma. Br J Cancer. 2012;107(6):988–93.

    Article  CAS  Google Scholar 

  12. Ignacio de Ulibarri J, Gonzalez-Madrono A, et al. CONUT: a tool for controlling nutritional status. First validation in a hospital population. Nutr Hosp. 2005;20:38–45.

  13. Yamanaka T, Matsumoto S, Teramukai S, et al. The baseline ratio of neutrophils to lymphocytes is associated with patient prognosis in advanced gastric cancer. Oncology. 2007;73(3–4):215–20.

    Article  Google Scholar 

  14. Pan QX, Su ZJ, Zhang JH, et al. A comparison of the prognostic value of preoperative inflammation-based scores and TNM stage in patients with gastric cancer. Onco Targets Ther. 2015;8:1375–85.

    Article  Google Scholar 

  15. Kuroda D, Sawayama H, Kurashige J, et al. Controlling nutritional status (CONUT) score is a prognostic marker for gastric cancer patients after curative resection. Gastric Cancer. 2017; https://doi.org/10.1007/s10120-017-0744-3.

    Article  PubMed  Google Scholar 

  16. Melling N, Gruning A, Tachezy M, et al. Glasgow Prognostic Score may be a prognostic index for overall and perioperative survival in gastric cancer without perioperative treatment. Surgery. 2016;159:1548–56.

    Article  Google Scholar 

  17. Yu X, Wen Y, Lin Y, et al. The value of preoperative Glasgow Prognostic Score and the C-reactive protein to albumin ratio as prognostic factors for long-term survival in pathological T1N0 esophageal squamous cell carcinoma. J Cancer. 2018;9(5):807–15.

    Article  Google Scholar 

  18. Gao GD, Sun B, Wang XB, et al. Neutrophil to lymphocyte ratio as prognostic indicator for patients with esophageal squamous cell cancer. Int J Biol Markers. 2017;32(4):e409–14.

    Article  Google Scholar 

  19. Hirahara N, Tajima Y, Fujii Y, et al. Prognostic nutritional index as a predictor of survival in resectable gastric cancer patients with normal preoperative serum carcinoembryonic antigen levels: a propensity score matching analysis. BMC Cancer. 2018;18(1):285.

    Article  Google Scholar 

  20. Iseki Y, Shibutani M, Maeda K, et al. Impact of the preoperative controlling nutritional status (CONUT) score on the survival after curative surgery for colorectal cancer. PLoS One. 2015;10(7):e0132488.

    Article  Google Scholar 

  21. Lu C, Gao P, Yang Y, et al. Prognostic evaluation of platelet to lymphocyte ratio in patients with colorectal cancer. Oncotarget. 2017;8(49):86287–95.

    Article  Google Scholar 

  22. Kudou K, Saeki H, Nakashima Y, et al. C-reactive protein/albumin ratio is a poor prognostic factor of esophagogastric junction and upper gastric cancer. J Gastroenterol Hepatol. 2018; https://doi.org/10.1111/jgh.14442.

    Article  PubMed  Google Scholar 

  23. Siewert JR, Stein HJ. Carcinoma of the cardia: Carcinoma of the gastroesophageal junction-classification, pathology and extent of resection. Dis Esophagus. 1996;9:173–82.

    Article  Google Scholar 

  24. Han WX, Chen ZM, Wei ZJ, et al. Preoperative pre-albumin predicts prognosis of patients after gastrectomy for adenocarcinoma of esophagogastric junction. World J Surg Oncol. 2016;14(1):279.

    Article  Google Scholar 

  25. Zhang L, Su Y, Chen Z, et al. The prognostic value of preoperative inflammation-based prognostic scores and nutritional status for overall survival in resected patients with nonmetastatic Siewert type II/III adenocarcinoma of esophagogastric junction. Medicine (Baltimore). 2017;96(30):e7647.

    Article  Google Scholar 

  26. Toyokawa T, Muguruma K, Tamura T, et al. Comparison of the prognostic impact and combination of preoperative inflammation-based and/or nutritional markers in patients with stage II gastric cancer. Oncotarget. 2018;9(50):29351–64.

    Article  Google Scholar 

  27. Wen J, Bedford M, Begum R, et al. The value of inflammation based prognostic scores in patients undergoing surgical resection for oesophageal and gastric carcinoma. J Surg Oncol. 2018; 117(8):1697–707.

    Article  Google Scholar 

  28. Aggarwal BB, Gehlot P. Inflammation and cancer: how friendly is the relationship for cancer patients? Curr Opin Pharmacol. 2009;9(4):351–69.

    Article  CAS  Google Scholar 

  29. Crumley AB, Stuart RC, McKernan M, et al. Is hypoalbuminemia an independent prognostic factor in patients with gastric cancer? World J Surg. 2010;34(10):2393–8.

    Article  Google Scholar 

  30. Lindenmann J, Fink-Neuboeck N, Koesslbacher M, et al. The influence of elevated levels of C-reactive protein and hypoalbuminemia on survival in patients with advanced inoperable esophageal cancer undergoing palliative treatment. J Surg Oncol. 2014;110:645–50.

    Article  CAS  Google Scholar 

  31. Espinosa E, Feliu J, Zamora P, et al. Serum albumin and other prognostic factors related to response and survival in patients with advanced non–small cell lung cancer. Lung Cancer. 1995;12(1–2):67–76.

    Article  CAS  Google Scholar 

  32. Asher V., Lee J., Bali A. Preoperative serum albumin is an independent prognostic predictor of survival in ovarian cancer. Med Oncol. 2012;29(3):2005–9.

    Article  CAS  Google Scholar 

  33. Borda F, Borda A, Jimenez J, et al. Predictive value of pre-treatment hypoalbuminemia in prognosis of resected colorectal cancer. Gastroenterol Hepatol. 2014;37(5):289–95.

    Article  Google Scholar 

  34. Ray-Coquard I, Cropet C, Van Glabbeke M, et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009;69:5383–91.

    Article  CAS  Google Scholar 

  35. Aoi W, Naito Y, Takagi T, et al. A novel myokine, secreted protein acidic and rich in cysteine (SPARC), suppresses colon tumorigenesis via regular exercise. Gut. 2013;62(6):882–9.

    Article  CAS  Google Scholar 

  36. Hojman P, Dethlefsen C, Brandt C, et al. Exercise-induced muscle-derived cytokines inhibit mammary cancer cell growth. Am J Phys. Endocrinol Metab. 2011;301(3):E504–10.

    Article  CAS  Google Scholar 

Download references

Acknowledgment

The authors thank Editage Group (https://www.editage.jp/) for editing the draft of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kensuke Kudou MD, PhD.

Ethics declarations

Disclosure

The authors declare that they have no conflict of interest.

Ethical Standards

All procedures followed were in accordance with the ethical standards of the responsible committee on human experimentation (institutional and national) and with the Helsinki Declaration of 1964 and later versions.

Informed Consent

Informed consent to be included in the study, or the equivalent, was obtained from all patients.

Additional information

Publisher's Note

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

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

10434_2020_8821_MOESM1_ESM.tif

Supplementary Fig. 1. Comparison of the predictive ability of seven inflammation-based prognostic scores—NLR, PLR, GPS, mGPS, CRP/Alb ratio, PI, and PNI—by receiver operating characteristic (ROC) curve analyses. NLR neutrophil–lymphocyte ratio; PLR platelet-lymphocyte ratio; GPS Glasgow prognostic score; mGPS modified Glasgow prognostic score; CRP C-reactive protein; Alb albumin; PI prognostic index; PNI prognostic nutritional index (TIFF 29297 kb)

10434_2020_8821_MOESM2_ESM.tif

Supplementary Fig. 2 Comparison of the predictive ability of seven inflammation-based prognostic scores—NLR, PLR, GPS, mGPS, CRP/Alb ratio, PI, and PNI—by time-dependent receiver operating characteristic (ROC) curve analyses for (a) recurrence-free survival and (b) overall survival. NLR neutrophil–lymphocyte ratio; PLR platelet-lymphocyte ratio; GPS Glasgow prognostic score; mGPS modified Glasgow prognostic score; CRP C-reactive protein; Alb albumin; PI prognostic index; PNI prognostic nutritional index (TIFF 29297 kb)

10434_2020_8821_MOESM3_ESM.tif

Supplementary Fig. 3 Postoperative survival in patients with AEG and UGC based on prognostic index (PI). The Kaplan–Meier method was performed separately by tumor location. (a) Recurrence-free and (b) overall survival in patients with AEG. (c) Recurrence-free and (d) overall survival in patients with UGC. Kaplan–Meier analyses were performed according to the same criteria as in Fig. 1. AEG adenocarcinoma of esophagogastric junction; UGC upper gastric cancer; PI prognostic index(TIFF 29297 kb)

10434_2020_8821_MOESM4_ESM.tif

Supplementary Fig. 4 Postoperative survival in patients with AEG based on prognostic index (PI). The Kaplan–Meier method was performed separately by tumor location according to Siewert classification. (a) Recurrence-free and (b) overall survival in patients with Siewert type I. (c) Recurrence-free and (d) overall survival in patients with Siewert type II. (e) Recurrence-free and (f) overall survival in patients with Siewert type III. Kaplan–Meier analyses were performed according to the same criteria as in Fig. 1. AEG adenocarcinoma of esophagogastric junction; PI prognostic index (TIFF 29297 kb)

10434_2020_8821_MOESM5_ESM.tif

Supplementary Fig. 5 Postoperative survival in patients with AEG and UGC based on prognostic nutritional index (PNI). The Kaplan–Meier method was performed separately by tumor location. (a) Recurrence-free and (b) overall survival in patients with AEG. (c) Recurrence-free and (d) overall survival in patients with UGC. Kaplan–Meier analyses were performed according to the same criteria as in Fig. 2. AEG adenocarcinoma of esophagogastric junction; UGC upper gastric cancer; PNI prognostic nutritional index (TIFF 29297 kb)

10434_2020_8821_MOESM6_ESM.tif

Supplementary Fig. 6 Postoperative survival in patients with AEG based on prognostic nutritional index (PNI). The Kaplan–Meier method was performed separately by tumor location according to Siewert classification. (a) Recurrence-free and (b) overall survival in patients with Siewert type I. (c) Recurrence-free and (d) overall survival in patients with Siewert type II. (e) Recurrence-free and (f) overall survival in patients with Siewert type III. Kaplan–Meier analyses were performed according to the same criteria as in Fig. 2. AEG adenocarcinoma of esophagogastric junction; PNI prognostic nutritional index (TIFF 29297 kb)

Supplementary material 7 (DOCX 28 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kudou, K., Nakashima, Y., Haruta, Y. et al. Comparison of Inflammation-Based Prognostic Scores Associated with the Prognostic Impact of Adenocarcinoma of Esophagogastric Junction and Upper Gastric Cancer. Ann Surg Oncol 28, 2059–2067 (2021). https://doi.org/10.1245/s10434-020-08821-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-020-08821-y

Navigation