Next Article in Journal
Dissecting Tumor-Immune Microenvironment in Breast Cancer at a Spatial and Multiplex Resolution
Next Article in Special Issue
Can We Reboot the Role of Intraperitoneal Chemotherapy in the Treatment for Gastric Cancer with Peritoneal Carcinomatosis?: A Retrospective Cohort Study Regarding Minimally Invasive Surgery Conjoined with Intraperitoneal plus Systemic Chemotherapy
Previous Article in Journal
Development and Functional Characterization of a Versatile Radio-/Immunotheranostic Tool for Prostate Cancer Management
Previous Article in Special Issue
Characterization of Total RNA, CD44, FASN, and PTEN mRNAs from Extracellular Vesicles as Biomarkers in Gastric Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Value of Systemic Inflammatory Response Markers in Patients Undergoing Neoadjuvant Chemotherapy and Gastrectomy for Advanced Gastric Cancer in the Eastern European Population

by
Agnieszka Pikuła
1,†,
Magdalena Skórzewska
1,†,
Zuzanna Pelc
1,
Radosław Mlak
2,
Katarzyna Gęca
1,
Katarzyna Sędłak
1,
Bogumiła Ciseł
1,
Magdalena Kwietniewska
1,
Karol Rawicz-Pruszyński
1,* and
Wojciech P. Polkowski
1
1
Department of Surgical Oncology, Medical University of Lublin, Radziwiłłowska 13 St., 20-080 Lublin, Poland
2
Department of Human Physiology, Medical University of Lublin, Radziwiłłowska 11 St., 20-080 Lublin, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(8), 1997; https://doi.org/10.3390/cancers14081997
Submission received: 24 February 2022 / Revised: 1 April 2022 / Accepted: 12 April 2022 / Published: 14 April 2022
(This article belongs to the Special Issue Advanced Gastric Cancer)

Abstract

:

Simple Summary

This study aimed to verify the prognostic value of neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and platelet-to-lymphocyte ratio (PLR) in GC patients undergoing neoadjuvant chemotherapy (NAC) and gastrectomy. Elevated NLR and PLR prior to NAC were associated with significantly higher risk of death (mOS: 36 vs. 87 months; HR = 2.21; p = 0.0255 and mOS: 30 vs. 87 months; HR = 2.89; p = 0.0034, respectively). Additionally, a significantly higher risk of death was observed in patients with elevated NLR after NAC (mOS: 35 vs. 87 months; HR = 1.94; p = 0.0368). Selected systemic inflammatory response markers (NLR, PLR) are significant prognostic factors in patients with advanced GC treated with NAC and gastrectomy, as shown in the Eastern European population.

Abstract

The prognostic value of the systemic inflammatory response markers, namely neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and platelet-to-lymphocyte ratio (PLR) has not yet been clarified in patients undergoing neoadjuvant chemotherapy (NAC) and gastrectomy for advanced gastric cancer (GC) in the Eastern European population. This study aimed to verify the prognostic value of NLR, PLR, and LMR in GC patients undergoing multimodal treatment. One hundred six GC patients undergoing NAC and gastrectomy between 2012 and 2020 were included. Analysed blood samples were obtained prior to NAC (pre-NAC group) and before surgical treatment (post-NAC group). To evaluate the prognostic value of the NLR, LMR, and PLR, univariable and multivariable overall survival (OS) analyses were performed. In the pre-NAC group, elevated NLR and PLR were associated with significantly higher risk of death (mOS: 36 vs. 87 months; HR = 2.21; p = 0.0255 and mOS: 30 vs. 87 months; HR = 2.89; p = 0.0034, respectively). Additionally, a significantly higher risk of death was observed in patients with elevated NLR in the post-NAC group (mOS: 35 vs. 87 months; HR = 1.94; p = 0.0368). Selected systemic inflammatory response markers (NLR, PLR) are significant prognostic factors in patients with advanced GC treated with NAC and gastrectomy, as shown in the Eastern European population.

1. Introduction

Gastric cancer (GC) remains a considerable burden of cancer-related mortality worldwide. However, the development of new biomarkers may improve its non-favourable outcome [1].
GC patients should undergo multidisciplinary treatment, including endoscopy, surgery, chemotherapy, and radiation therapy. In Europe, neoadjuvant chemotherapy (NAC) with a platinum and fluoropyrimidine combination is recommended for at least stage IB resectable GC patients [2]. The FLOT4 regimen (5-Fluorouracil, Leucovorin, Oxaliplatin, Docetaksel) is currently the preferred regimen for NAC, demonstrating improved survival in patients with resectable GC when compared to ECX/ECF (Epirubicin, Cisplatin, Capecitabine or 5-Fluorouracil) [3,4,5].
Personalised therapy in GC depends on the appropriate staging. Despite visible progress in computed tomography (CT) performed in specialised centres, the inadequacy of evaluating the tumour stage is a critical issue in clinical practice [6,7,8]. Staging laparoscopy enables the diagnosis of peritoneal dissemination with improved sensitivity and specificity compared to CT [7]; however, assessment of the nodal stage remains difficult [9]. There is a need for additional, cost-effective and readily available prognostic and predictive factors to select patients who may require prolonged NAC. Systemic inflammatory markers are of special interest since they are strongly correlated with the progression and response to cancer treatment [10]. These include blood count parameters, such as neutrophil, lymphocyte, monocyte and platelet number, as well as their combinations: neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and the platelet-to-lymphocyte ratio [PLR] [11]. Among complete blood count derived markers, anaemia is recognised as an independent prognostic factor in numerous malignancies [12]. As the results of currently available studies are inconclusive, interpretation of NLR, PLR and LMR in routine practice is equivocal. Importantly, available literature provides data solely from the Asian population [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27]. Exploratory analysis of the REAL-2 trial revealed that a high NLR value had a significant, independent negative prognostic effect. Although the study was conducted in the Western population, it should be noted that most patients had inoperable or metastatic oesophagogastric cancer [27].
To the best of our knowledge, this is the first study that aimed to verify the prognostic value of NLR, PLR and LMR in GC patients undergoing multimodal treatment in the Eastern European population.

2. Materials and Methods

After receiving institutional review board approval (KE—0254/331/2018), we collected data from the database of patients diagnosed with GC treated with gastrectomy from 1 July 2012 to 31 July 2020 at the Department of Oncological Surgery of the Medical University of Lublin, Poland. The initial date of patients’ recruitment was set due to the standardisation of the NAC with 5-Fluorouracil and platinum derivatives, reflecting the current evidence-based clinical guidelines for GC [2,28,29]. The preoperative staging, evaluation of the patient’s general condition and treatment plan were carried out by the multidisciplinary team. A ypTNM stage of the disease was established according to the 8th edition of the American Joint Committee on Cancer (AJCC) [30]. In the present study, we included patients with histologically confirmed GC who underwent multimodal treatment based on NAC and surgical resection. We excluded patients in clinical stages I and IV, without NAC or gastrectomy and with adjuvant radiotherapy. One hundred six patients were eligible for the final analysis. The flow chart of the study is shown in Figure 1.

2.1. Neoadjuvant Chemotherapy (NAC)

The majority of the study group (95%) received treatment based on a combination of platinum and fluoropyrimidine derivatives. The EOX regimen included epirubicin 50 mg/m2 on the 1st day of the cycle, 130 mg/m2 oxaliplatin on the 1st day of the cycle and 625 mg/m2 of capecitabine twice a day from days 1–21; cycles were repeated every 21 days (30). Three cycles EOX before surgery and three cycles after surgery. The FLOT-4 regimen consisted of docetaxel at 50 mg/m2 on day 1, oxaliplatin at 85 mg/m2 on day 1, leucovorin at 200 mg/m2 on day 1 and 5-fluorouracil at 2600 mg/m2 on day 1 of the cycle, cycles were repeated every 14 days [4]. After 4–5 week time intervals, patients were scheduled for surgical treatment.

2.2. Complete Blood Count Based Inflammatory Response Markers

Blood samples used for analysis were obtained one day before administration of the first NAC cycle (pre-NAC group) and one day before surgical treatment (post-NAC group). The NLR was calculated by dividing the absolute number of neutrophils by the absolute number of lymphocytes in the peripheral blood. The PLR was calculated by dividing the absolute number of neutrophils by the absolute number of lymphocytes in the peripheral blood. The LMR was calculated by dividing the absolute number of lymphocytes by the absolute number of peripheral blood monocytes.

2.3. Statistical Analysis

The main outcome measured was overall survival (OS). Survival data were obtained from medical records of both hospitalisation and follow-up visits. Additionally, data from the National Health Fund were analysed, as well as personal telephone information obtained from patients and/or their families during the COVID pandemic. For this research, the observation was finalised on 1 August 2021. Statistical analysis of the data was performed using MedCalc v.15.8 (MedCalc Software, Oostende, Belgium). To assess the normality of the data distribution, the D’Agostino-Pearson test was used. Since all continuous variables had non-normal data distribution, the median was used to measure data concentration and the data spread was presented by the interquartile range and minimum-maximum range. Categorised and dichotomised variables were expressed as numbers and percentages. As an objective method of cut-off establishment, the Receiver operating characteristic (ROC) analysis was used. Determined cut-offs for pre-treatment NLR, PLR and LMR were ≤1.76, ≤132.34 and >2.75, respectively, whereas these values for the preoperative phase of POC, a day before gastrectomy were ≤1.94, ≤151.06 and >2.81, respectively. OS was defined as the time from the date of surgery to the date of the patient’s death or the date of the last follow-up. The log-rank test was used to calculate the proportional hazard ratio in univariable OS analysis (the Kaplan–Meier estimation method was used for the generation of survival curves), whereas Cox logistic regression models were used in multivariable OS analysis. Comparisons of studied ratios depending on demographic and clinical variables were performed with the use of the Mann–Whitney U test (if 2 independent groups were compared) or ANOVA Kruskal–Wallis test (if more than 2 independent groups were compared). In all analyses, we used two-tailed p-tests and results with a p-value below 0.05 were statistically significant.

3. Results

3.1. Characteristics of the Research Group

One hundred six patients were included. The majority of the study group was male (61.3%). The median age of patients was 61 years (range 38–76 years). Patients with middle (39.6%) or upper (34%) GC were predominant. According to Lauren’s classification, the intestinal type was predominant (53.8%). Patients in the ypT4 stage accounted for 13.2% of patients. Nearly half of the patients (49.5%) were node-positive (ypN1-N3b). None of the patients had distant metastases. Fifty-five patients (52%) were treated with chemotherapy according to the EOX regimen (33), whereas 32 patients (30%) according to the FLOT-4 regimen [4]. The median number of preoperative chemotherapy cycles was 4 and the postoperative cycles were 3.5. Detailed data on the characteristics of patients in terms of demographic and clinical variables are presented in Table 1.

3.2. Survival Analysis

3.2.1. Univariate Analysis

The relationship between clinical, demographic variables and systemic inflammatory response markers and the OS are presented in Table 2 and Table 3.
Following demographic and clinical variables were associated with a significantly higher risk of death: anatomical location in esophagogastric junction (EGJ) (mOS: 11 vs. 87 months; HR = 4.70; p = 0.0040), lymph node metastases (N1-N3b) (mOS: 29 months vs. NR; HR = 4.43; p < 0.0001), grade G3 (mOS: 33 months vs. NR; HR = 2.77; p = 0.0038) and no pathological tumour response to NAC (TRG3, TRG4) (mOS: 35 months vs. NR; HR = 6.42; p = 0.0003). On the other hand, the intestinal type was associated with a significantly lower risk of death (mOS: 87 vs. 35 months; HR = 0.51; p = 0.0394). Elevated NLR in the pre-NAC group was associated with a significantly higher risk of death (mOS: 36 vs. 87 months; HR = 2.21; p = 0.0255; Figure 2).
Similarly, elevated PLR in the pre-NAC group was associated with a significantly higher risk of death (mOS: 30 vs. 87 months; HR = 2.89; p = 0.0034; Figure 3).
Moreover, high NLR in the post-NAC group was associated with a significantly higher risk of death (mOS: 35 vs. 87 months; HR = 1.94; p = 0.0368; Figure 4).

3.2.2. Multivariate Analysis

Multivariate analysis confirmed the independent, unfavourable prognostic value of anatomical location (EGJ) (HR = 6.63; p = 0.0042), positive lymph nodes (HR = 3.94; p = 0.0009), grade G3 (HR = 2.66; p = 0.0304) and no pathological tumour response to NAC (TRG3, TRG4; HR = 5.92, p = 0.0066).
Moreover, in multivariate analysis, elevated NLR in the pre-NAC group was associated with a non-significantly higher risk of death (HR = 1.29; p = 0.5263). Whereas elevated PLR in the pre-NAC group was associated with a significantly higher risk of death (HR = 2.75; p = 0.0143). Elevated NLR in the post-NAC group was associated with a significantly higher risk of death (HR = 1.97; p = 0.0414).

3.3. Relationship of Demographic and Clinical Variables with the Systemic Inflammatory Response Markers

The age of patients was the only factor related to one of the systemic inflammatory response markers. Patients under 61 years old (median, Table 1) had significantly lower LMR compared to patients over 61 years old (Me: 3.58 vs. 2.87; p = 0.0188). There were no statistically significant relationships between other demographic and clinical variables and the systemic inflammatory response markers.

4. Discussion

This study aimed to verify the prognostic value of the systemic inflammatory response markers in patients with advanced GC treated with NAC and gastrectomy. PLR in the pre-NAC group and NLR in the post-NAC group were independent prognostic factors in patients undergoing multimodal treatment. Although tumour-related neutrophils are significant factors in promoting angiogenesis, tumour growth or metastasis, the exact correlation between higher NLR values and poor prognosis remains unclear [10,31]. Recently, Asian research confirmed PLR to be an independent prognostic factor for OS also in patients undergoing adjuvant chemoradiotherapy after radical gastrectomy [32]. Moreover, the prognostic model, including systemic inflammation, was more precise than the model without systemic inflammation.
Various original studies and meta-analyses have evaluated systemic inflammatory response markers in GC patients, as shown in Table 4.
Overviewed studies are mostly retrospective, published between 2020–2021 and conducted in the Far East (China and Japan). Moreover, all patients were treated with NAC followed by surgical resection. Notably, the rate of NAC administration in the Asian GC population is rather low when compared with the West. A meta-analysis of 28,929 patients from 51 international cohorts (10 from Europe and the USA and 41 from Asia), evaluating the role of PLR in GC patients treated with surgery and chemotherapy, concluded high PLR is associated with a shorter OS in regard to more frequent involvement of the serosa, lymph node metastases and a higher rate of stage III/IV [25]. Elevated PLR had a high prognostic impact only in the Asian population. Furthermore, considering different treatment methods, high PLR was a significant predictor of shorter OS in patients undergoing surgery alone. However, it was of no prognostic value in patients receiving chemotherapy or combined treatment [25]. Another meta-analysis of 28 studies confirmed the association between high PLR values and unfavourable OS [26]. Twenty-one studies were conducted in Asia and seven in Europe/America. Patients with elevated PLR had significantly shorter OS.
In patients undergoing gastrectomy with curative intent, increasing evidence underlines that systemic inflammatory response, triggered by higher neutrophil and platelet levels, worsens the prognosis [11]. Mohri et al. concluded that evaluation of NLR prior to the treatment remains the crucial host-related parameter affecting prognosis in patients with GC [33], possibly due to intracellular instability through atypical cell proliferation leading to further deterioration of the cell’s environment [34]. Our study confirms the prognostic value of these markers in the Caucasian population. Elevated NLR and PLR values were associated with a higher risk of death.
A Chinese study aimed to verify the prognostic relationship between NAC and conversion rates of NLR and PLR in patients with unresectable and metastatic GC. Patients with low inflammatory response markers rates after NAC had significantly longer progression-free survival when compared to patients with sustained high-NLR/PLR [35]. In our study, patients with poor or no response to NAC (Tumour Regression Grade 3 and 4) had a significantly higher risk of death in both univariable and multivariable analyses.
Limitations of our study include its retrospective and single-centre nature and relatively small sample size. A prospective multi-centre study with a large sample size would overcome these shortcomings and possibly validate our current findings.

5. Conclusions

Selected systemic inflammatory response markers (NLR, PLR) are significant prognostic factors in patients with advanced GC treated with NAC and gastrectomy, as shown in the Eastern European population.

Author Contributions

Conceptualisation, A.P., M.S., Z.P. and K.R.-P.; methodology, R.M., K.S. and K.G.; software, R.M., K.S. and K.G.; validation, W.P.P., K.R.-P., M.K. and Z.P.; formal analysis, A.P., M.S., K.R.-P. and R.M.; investigation, B.C., M.S., A.P. and Z.P.; data curation, K.S., K.G., M.K. and B.C.; writing—original draft preparation, A.P., M.S., Z.P. and K.R.-P.; writing—review and editing, K.R.-P., W.P.P. and M.S.; visualisation, R.M., K.G. and K.S.; supervision, W.P.P.; project administration, A.P., M.S., Z.P., R.M., K.G., K.S., B.C., M.K., K.R.-P. and W.P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted after obtaining institutional review board approval (Bioethical Committee of Medical University of Lublin, Ethic Code: KE—0254/331/2018).

Informed Consent Statement

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

Data Availability Statement

Data available on special request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Smyth, E.C.; Verheij, M.; Allum, W.; Cunningham, D.; Cervantes, A.; Arnold, D.; Committee, E.G. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, v38–v49. [Google Scholar] [CrossRef]
  3. Cherny, N.I.; Dafni, U.; Bogaerts, J.; Latino, N.J.; Pentheroudakis, G.; Douillard, J.-Y.; Tabernero, J.; Zielinski, C.; Piccart, M.J.; de Vries, E.G.E. ESMO-Magnitude of Clinical Benefit Scale version 1.1. Ann. Oncol. 2017, 28, 2340–2366. [Google Scholar] [CrossRef] [PubMed]
  4. Al-Batran, S.-E.; Hofheinz, R.D.; Pauligk, C.; Kopp, H.-G.; Haag, G.M.; Luley, K.B.; Meiler, J.; Homann, N.; Lorenzen, S.; Schmalenberg, H.; et al. Histopathological regression after neoadjuvant docetaxel, oxaliplatin, fluorouracil, and leucovorin versus epirubicin, cisplatin, and fluorouracil or capecitabine in patients with resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4-AIO): Results from the phase 2 part of a multicentre, open-label, randomised phase 2/3 trial. Lancet Oncol. 2016, 17, 1697–1708. [Google Scholar] [CrossRef] [PubMed]
  5. Al-Batran, S.-E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.-G.; Mayer, F.; Haag, G.M.; Luley, K.; et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): A randomised, phase 2/3 trial. Lancet 2019, 393, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, C.-Y.; Hsu, J.-S.; Wu, D.-C.; Kang, W.-Y.; Hsieh, J.-S.; Jaw, T.-S.; Wu, M.-T.; Liu, G.-C. Gastric Cancer: Preoperative Local Staging with 3D Multi-Detector Row CT—Correlation with Surgical and Histopathologic Results. Radiology 2007, 242, 472–482. [Google Scholar] [CrossRef] [PubMed]
  7. Leake, P.-A.; Cardoso, R.; Seevaratnam, R.; Lourenco, L.; Helyer, L.; Mahar, A.; Law, C.; Coburn, N.G. A systematic review of the accuracy and indications for diagnostic laparoscopy prior to curative-intent resection of gastric cancer. Gastric Cancer 2012, 15 (Suppl. 1), S38–S47. [Google Scholar] [CrossRef] [Green Version]
  8. Fairweather, M.; Jajoo, K.; Sainani, N.; Bertagnolli, M.M.; Wang, J. Accuracy of EUS and CT imaging in preoperative gastric cancer staging. J. Surg. Oncol. 2015, 111, 1016–1020. [Google Scholar] [CrossRef] [PubMed]
  9. Kwee, R.M.; Kwee, T.C. Imaging in assessing lymph node status in gastric cancer. Gastric Cancer 2009, 12, 6–22. [Google Scholar] [CrossRef]
  10. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  11. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
  12. Caro, J.J.; Salas, M.; Ward, A.; Goss, G. Anemia as an independent prognostic factor for survival in patients with cancer: A systemic, quantitative review. Cancer 2001, 91, 2214–2221. [Google Scholar] [CrossRef]
  13. Yin, X.; Fang, T.; Wang, Y.; Wang, Y.; Zhang, D.; Li, C.; Xue, Y. Prognostic significance of serum inflammation indexes in different Lauren classification of gastric cancer. Cancer Med. 2021, 10, 1103–1119. [Google Scholar] [CrossRef]
  14. Sun, X.-J.; Ai, L.; Feng, Y.-C. The Value of ABO Blood Group and Complete Blood Count for the Prognosis Analysis of Gastric Cancer Patients. OncoTargets Ther. 2020, 13, 4627–4633. [Google Scholar] [CrossRef] [PubMed]
  15. Lin, J.-X.; Lin, J.-P.; Xie, J.-W.; Wang, J.-B.; Lu, J.; Chen, Q.-Y.; Cao, L.-L.; Lin, M.; Tu, R.; Zheng, C.-H.; et al. Complete blood count-based inflammatory score (CBCS) is a novel prognostic marker for gastric cancer patients after curative resection. BMC Cancer 2020, 20, 11. [Google Scholar] [CrossRef]
  16. Lin, J.-X.; Wang, Z.-K.; Huang, Y.-Q.; Xie, J.-W.; Wang, J.-B.; Lu, J.; Chen, Q.-Y.; Lin, M.; Tu, R.-H.; Huang, Z.-N.; et al. Dynamic Changes in Pre- and Postoperative Levels of Inflammatory Markers and Their Effects on the Prognosis of Patients with Gastric Cancer. J. Gastrointest. Surg. 2021, 25, 387–396. [Google Scholar] [CrossRef] [Green Version]
  17. Xu, B.-B.; Xu, Y.; Lu, J.; Wu, Y.; Wang, J.-B.; Lin, J.-X.; Xie, J.-W.; Li, P.; Zheng, C.-H.; Huang, A.-M.; et al. Prognostic significance of combined Lymphocyte-monocyte Ratio and Tumor-associated Macrophages in Gastric Cancer Patients after Radical Resection. J. Cancer 2020, 11, 5078–5087. [Google Scholar] [CrossRef]
  18. Nakamura, N.; Kinami, S.; Tomita, Y.; Miyata, T.; Fujita, H.; Takamura, H.; Ueda, N.; Kosaka, T. The neutrophil/lymphocyte ratio as a predictor of successful conversion surgery for stage IV gastric cancer: A retrospective study. BMC Cancer 2020, 20, 363. [Google Scholar] [CrossRef]
  19. Gu, L.; Wang, M.; Cui, X.; Mo, J.; Yuan, L.; Mao, F.; Zhang, K.; Ng, D.M.; Chen, P.; Wang, D. Clinical significance of peripheral blood-derived inflammation markers in advanced gastric cancer after radical resection. BMC Surg. 2020, 20, 219. [Google Scholar] [CrossRef]
  20. Kudou, K.; Nakashima, Y.; Haruta, Y.; Nambara, S.; Tsuda, Y.; Kusumoto, E.; Ando, K.; Kimura, Y.; Hashimoto, K.; Yoshinaga, K.; 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. 2021, 28, 2059–2067. [Google Scholar] [CrossRef]
  21. Hirahara, N.; Matsubara, T.; Fujii, Y.; Kaji, S.; Kawabata, Y.; Hyakudomi, R.; Yamamoto, T.; Taniura, T.; Tajima, Y. Comparison of the prognostic value of immunoinflammation-based biomarkers in patients with gastric cancer. Oncotarget 2020, 11, 2625–2635. [Google Scholar] [CrossRef] [PubMed]
  22. Toyokawa, T.; Muguruma, K.; Yoshii, M.; Tamura, T.; Sakurai, K.; Kubo, N.; Tanaka, H.; Lee, S.; Yashiro, M.; Ohira, M. Clinical significance of prognostic inflammation-based and/or nutritional markers in patients with stage III gastric cancer. BMC Cancer 2020, 20, 517. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, Z.; Ge, H.; Miao, Z.; Shao, S.; Shi, H.; Dong, C. Dynamic Changes in the Systemic Inflammation Response Index Predict the Outcome of Resectable Gastric Cancer Patients. Front. Oncol. 2021, 11, 577043. [Google Scholar] [CrossRef] [PubMed]
  24. Ohe, Y.; Fushida, S.; Yamaguchi, T.; Kinoshita, J.; Saito, H.; Okamoto, K.; Nakamura, K.; Tajima, H.; Ninomiya, I.; Ohta, T. Peripheral Blood Platelet-Lymphocyte Ratio Is Good Predictor of Chemosensitivity and Prognosis in Gastric Cancer Patients. Cancer Manag. Res. 2020, 12, 1303–1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zhang, X.; Zhao, W.; Yu, Y.; Qi, X.; Song, L.; Zhang, C.; Li, G.; Yang, L. Clinicopathological and prognostic significance of platelet-lymphocyte ratio (PLR) in gastric cancer: An updated meta-analysis. World J. Surg. Oncol. 2020, 18, 191. [Google Scholar] [CrossRef] [PubMed]
  26. Cao, W.; Yao, X.; Cen, D.; Zhi, Y.; Zhu, N.; Xu, L. The prognostic role of platelet-to-lymphocyte ratio on overall survival in gastric cancer: A systematic review and meta-analysis. BMC Gastroenterol. 2020, 20, 16. [Google Scholar] [CrossRef] [Green Version]
  27. Grenader, T.; Waddell, T.; Peckitt, C.; Oates, J.; Starling, N.; Cunningham, D.; Bridgewater, J. Prognostic value of neutrophil-to-lymphocyte ratio in advanced oesophago-gastric cancer: Exploratory analysis of the REAL-2 trial. Ann. Oncol. 2016, 27, 687–692. [Google Scholar] [CrossRef]
  28. Cunningham, D.; Starling, N.; Rao, S.; Iveson, T.; Nicolson, M.; Coxon, F.; Middleton, G.; Daniel, F.; Oates, J.; Norman, A.R. Capecitabine and Oxaliplatin for Advanced Esophagogastric Cancer. N Engl. J. Med. 2008, 358, 36–46. [Google Scholar] [CrossRef] [Green Version]
  29. Ychou, M.; Boige, V.; Pignon, J.-P.; Conroy, T.; Bouché, O.; Lebreton, G.; Ducourtieux, M.; Bedenne, L.; Fabre, J.-M.; Saint-Aubert, B.; et al. Perioperative Chemotherapy Compared with Surgery Alone for Resectable Gastroesophageal Adenocarcinoma: An FNCLCC and FFCD Multicenter Phase III Trial. J. Clin. Oncol. 2011, 29, 1715–1721. [Google Scholar] [CrossRef]
  30. In, H.; Solsky, I.; Palis, B.; Langdon-Embry, M.; Ajani, J.A.; Sano, T. Validation of the 8th Edition of the AJCC TNM Staging System for Gastric Cancer using the National Cancer Database. Ann. Surg. Oncol. 2017, 24, 3683–3691. [Google Scholar] [CrossRef]
  31. McMillan, D.C. Systemic inflammation, nutritional status and survival in patients with cancer. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 223–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Li, Y.; Wang, W.-B.; Yang, L.; Wang, Q.-Y.; Dai, J.; Xia, L.; Peng, J.; Zhou, F.-X.; Wei, Y.-C.; Shi, H.-P. The combination of body composition conditions and systemic inflammatory markers has prognostic value for patients with gastric cancer treated with adjuvant chemoradiotherapy. Nutrition 2022, 93, 111464. [Google Scholar] [CrossRef] [PubMed]
  33. Mohri, Y.; Tanaka, K.; Ohi, M.; Yokoe, T.; Miki, C.; Kusunoki, M. Prognostic Significance of Host- and Tumor-Related Factors in Patients with Gastric Cancer. World J. Surg. 2010, 34, 285–290. [Google Scholar] [CrossRef] [PubMed]
  34. Ma, Y.; Zhang, J.; Chen, X. Lymphocyte-to-Monocyte Ratio is Associated with the Poor Prognosis of Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Cancer Manag. Res. 2021, 13, 1571–1580. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, F.; Liu, Z.-Y.; Xia, Y.-Y.; Zhou, C.; Shen, X.-M.; Li, X.-L.; Han, S.-G.; Zheng, Y.; Mao, Z.-Q.; Gong, F.-R.; et al. Changes in neutrophil/lymphocyte and platelet/lymphocyte ratios after chemotherapy correlate with chemotherapy response and prediction of prognosis in patients with unresectable gastric cancer. Oncol. Lett. 2015, 10, 3411–3418. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow chart of the study. (GC—gastric cancer; M1—distant metastases; N—number of patients).
Figure 1. Flow chart of the study. (GC—gastric cancer; M1—distant metastases; N—number of patients).
Cancers 14 01997 g001
Figure 2. Kaplan–Meier graph representing survival probability in relation to the NLR (pre-NAC group).
Figure 2. Kaplan–Meier graph representing survival probability in relation to the NLR (pre-NAC group).
Cancers 14 01997 g002
Figure 3. Kaplan–Meier graph representing survival probability in relation to the PLR (pre-NAC group).
Figure 3. Kaplan–Meier graph representing survival probability in relation to the PLR (pre-NAC group).
Cancers 14 01997 g003
Figure 4. Kaplan–Meier graph representing survival probability in relation to the NLR (post-NAC group).
Figure 4. Kaplan–Meier graph representing survival probability in relation to the NLR (post-NAC group).
Cancers 14 01997 g004
Table 1. Demographic and clinical characteristics of the study group.
Table 1. Demographic and clinical characteristics of the study group.
VariableNo. of Patients n = 106 (%)
Sex
Female41 (38.7%)
Male65 (61.3%)
Age (years)
Median61
Min–Max38–76
Anatomical localisation of gastric cancer
Esophagogastric junction4 (3.8%)
Upper36 (34.0%)
Middle42 (39.6%)
Lower24 (22.6%)
Lauren’s histological type
Intestinal56 (53.8%)
Diffuse14 (13.5%)
Mixed34 (32.7%)
Missing data (n = 2)
ypT
T06 (5.7%)
T1a5 (4.7%)
T1b9 (8.5%)
T229 (27.4%)
T343 (40.6%)
T4a12 (11.3%)
T4b2 (1.9%)
ypN
N053 (50.5%)
N114 (13.3%)
N217 (16.2%)
N3a10 (9.5%)
N3b11 (10.5%)
Missing data (n = 1)
ypM
M0106 (100%)
Grading
G16 (6.1%)
G243 (43.9%)
G349 (50.5%)
Missing data (n = 8)
No. of neoadjuvant chemotherapy cycles
Median4
Min–Max1–6
Adjuvant chemotherapy compliance
Yes101 (95.3%)
No5 (4.7%)
Tumour regression grade
17 (6.7%)
228 (26.7%)
348 (45.7%)
422 (21.0%)
Missing data (n = 1)
Table 2. Relationship between the systemic inflammatory response markers on overall survival.
Table 2. Relationship between the systemic inflammatory response markers on overall survival.
VariablePre-NAC GroupPost-NAC Group
UnivariateMultivariate #UnivariateMultivariate #
mOSp
HR [95%CI]
p
HR [95%CI]
mOSp
HR [95%CI]
p
HR [95%CI]
NLR
Low87.000.0255 *0.526387.000.0368 *0.0414 *
High36.002.21 [1.16–4.22]1.29 [0.59–2.80]35.001.94 [1.02–3.70]1.97 [1.03–3.76]
PLR
Low87.000.0034 *0.0143 *87.000.58720.8887
High30.002.89 [1.49–5.60]2.75 [1.23–6.13]67.001.21 [0.59–2.49]0.95 [0.44–2.03]
LMR
Low-0.74950.531287.000.80350.6845
High67.000.89 [0.43–1.86]1.28 [0.59–2.76]62.000.92 [0.47–1.79]1.16 [0.57–2.37]
CI—confidence interval, HR—hazard ratio, LMR—lymphocyte to monocyte ratio, mOS—median overall survival, POC- perioperative chemotherapy, NLR—neutrophil to lymphocyte ratio, PLR—platelet to lymphocyte ratio. *—statistically significant results. #—in multivariate analysis, the results were adjusted for all statistically significant results of univariate analysis (including both Table 2 and Table 3).
Table 3. Influence of selected demographic and clinical variables on overall survival.
Table 3. Influence of selected demographic and clinical variables on overall survival.
VariableUnivariableMultivariable #
mOSp
HR [95%CI]
p
HR [95%CI]
Sex
Female620.46740.5751
Male870.78 [0.40–1.54]0.81 [0.39–1.67]
Age (years)
< Median870.96840.4542
>=Median621.01 [0.533–1.91]1.31 [0.5–2.63]
Anatomical localisation
Oesophagogastric junction110.0040 *0.0042 *
Non–junctional874.70 [0.43–51.84]6.43 [1.81–22.80]
Lauren’s histological type
Intestinal870.0394 *0.756
Diffuse, Mixed350.51 [0.27–0.98]0.88 [0.39–1.97]
ypT
T0–T3870.48980.5101
T4a–T4b621.35 [0.51–3.58]1.38 [0.53–3.60]
ypN
N0NR<0.0001 *0.0009 *
N1–N3b294.43 [2.33–8.44]3.94 [1.76–8.84]
Grade
G1–G2 NR0.0038 *0.0304 *
G3332.77 [1.45–5.27]2.66 [1.10–6.44]
Tumour regression grade
1–2NR0.0003 *0.0066 *
3–4356.42 [3.30–12.48]5.92 [1.65–21.25]
CI—confidence interval, HR—hazard ratio, NR—not reached, mOS—median overall survival, *—statistically significant results. #—in multivariable analysis, the results were adjusted for all statistically significant results of univariable analysis (including Table 2).
Table 4. Baseline characteristics of the selected studies in comparison to the results of the present study.
Table 4. Baseline characteristics of the selected studies in comparison to the results of the present study.
StudyPublication YearCountryStudy DesignTreatment StrategyStageSample SizeSignificant Prognostic Blood Markers
Yin [13]2021ChinaretrospectiveSurgery
AC when applicable
I, II, III852SII, PLR
Sun [14]2020Chinaretrospective--488NLR, PLT, PDW, ABO blood group
Lin [15]2020ChinaretrospectiveSurgery
AC when applicable
I, II, III1810LMR, Hg,
Lin [16]2021ChinaretrospectiveSurgery
AC when applicable
I, II, III2257LMR
Xu [17]2020ChinaprospectiveSurgery
AC when applicable
I, II, III438LMR
Nakamura [18]2020JapanretrospectiveNAC
SurgeryAC
IV50NLR, albumin levels
Gu [19]2020ChinaprospectiveSurgery
AC
II, III598PLR
Kudou [20]2020JapanretrospectiveSurgery
AC when applicable
I, II, III206GPS, PI and PNI
Hirahara [21]2020JapanretrospectiveSurgery
AC when applicable
I, II, III412SII
Toyokawa [22]2020JapanretrospectiveSurgery
AC
III225CAR, PLR
Liu [23]2021ChinaretrospectiveSurgery
AC when applicable
I, II, III442SIRI
Ohe [24]2020JapanretrospectiveNAC
Surgery AC
III, IV41PLR
present study-PolandretrospectiveNAC
Surgery
II, III106NLR, PLR
AC—adjuvant chemotherapy, NAC—neoadjuvant chemotherapy (NAC + AC = POC), SII—systemic immune-inflammation index, SII = N × P/L (N = Neutrophil count, L = Lymphocyte count and P = Platelet count), PDW—platelet distribution width, PI—prognostic index, PI ranges based on the CRP level and the white blood cell count, PNI—prognostic nutritional index, PNI was calculated using the following formula: 10 × serum albumin (g/dL) + 0.005 × total lymphocyte count (per mm3), GPS—Glasgow prognostic score, GPS ranges based on CRP and albumin level, SIRI—systemic inflammation response index, SIRI = neutrophil count × monocyte count/lymphocyte count, CAR—C-reactive protein/ albumin ratio.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pikuła, A.; Skórzewska, M.; Pelc, Z.; Mlak, R.; Gęca, K.; Sędłak, K.; Ciseł, B.; Kwietniewska, M.; Rawicz-Pruszyński, K.; Polkowski, W.P. Prognostic Value of Systemic Inflammatory Response Markers in Patients Undergoing Neoadjuvant Chemotherapy and Gastrectomy for Advanced Gastric Cancer in the Eastern European Population. Cancers 2022, 14, 1997. https://doi.org/10.3390/cancers14081997

AMA Style

Pikuła A, Skórzewska M, Pelc Z, Mlak R, Gęca K, Sędłak K, Ciseł B, Kwietniewska M, Rawicz-Pruszyński K, Polkowski WP. Prognostic Value of Systemic Inflammatory Response Markers in Patients Undergoing Neoadjuvant Chemotherapy and Gastrectomy for Advanced Gastric Cancer in the Eastern European Population. Cancers. 2022; 14(8):1997. https://doi.org/10.3390/cancers14081997

Chicago/Turabian Style

Pikuła, Agnieszka, Magdalena Skórzewska, Zuzanna Pelc, Radosław Mlak, Katarzyna Gęca, Katarzyna Sędłak, Bogumiła Ciseł, Magdalena Kwietniewska, Karol Rawicz-Pruszyński, and Wojciech P. Polkowski. 2022. "Prognostic Value of Systemic Inflammatory Response Markers in Patients Undergoing Neoadjuvant Chemotherapy and Gastrectomy for Advanced Gastric Cancer in the Eastern European Population" Cancers 14, no. 8: 1997. https://doi.org/10.3390/cancers14081997

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

Article Metrics

Back to TopTop