Skip to main content
  • Research article
  • Open access
  • Published:

Increased neutrophil–lymphocyte ratio predicts recurrence in patients with well-differentiated pancreatic neuroendocrine neoplasm based on the 2017 World Health Organization classification

Abstract

Background

The prognostic values of inflammation-based markers in well-differentiated pancreatic neuroendocrine neoplasms, diagnosed according to the new 2017 World Health Organization classification, have remained unclear. Therefore, we assessed the ability to predict the recurrence of such markers after curative resection in patients with these neoplasms.

Methods

Circulating/systemic neutrophil–lymphocyte, monocyte–lymphocyte, platelet–lymphocyte, and platelet–white cell ratios were evaluated in 120 patients who underwent curative resection for well-differentiated pancreatic neuroendocrine neoplasms without synchronous distant metastasis between 2001 and 2018. Recurrence-free-survival and overall survival were compared using Kaplan–Meier analysis and log-rank tests. Univariate or multivariate analyses, using a Cox proportional hazards model, were used to calculate hazard ratios with 95% confidence intervals.

Results

Univariate analysis demonstrated that preoperative neutrophil–lymphocyte ratio, tumor size, European Neuroendocrine Tumor Society TMN classification, 2017 World Health Organization classification, and venous invasion were associated with recurrence. The optimal preoperative neutrophil–lymphocyte ratio cut-off value was 2.62, based on receiver operating characteristic curve analysis. In multivariate analysis, a higher preoperative neutrophil–lymphocyte ratio (HR = 3.49 95% CI 1.05–11.7; P = 0.042) and 2017 World Health Organization classification (HR = 8.81, 95% CI 1.46–168.2; P = 0.015) were independent recurrence predictors.

Conclusions

The circulating/systemic neutrophil–lymphocyte ratio is a useful and convenient preoperative prognostic marker of recurrence in patients with well-differentiated pancreatic neuroendocrine neoplasm based on the 2017 World Health Organization classification.

Peer Review reports

Background

Pancreatic neuroendocrine neoplasm (PanNEN) is a biologically heterogeneous and relatively rare malignancy, with an incidence rate of approximately 5 cases per 1 million person-years, which accounts for 1–2% of primary pancreatic neoplasms [1]. In recent years, the incidence of PanNEN detected clinically has significantly increased because of the advances in imaging modalities during the past few decades [2]. When the disease is clinically detected before it becomes symptomatic, the lesions are typically localized, increasing the possibility of curative resection and improving prognosis [3]. Although surgical resection is currently the only curative treatment for PanNEN [4], recurrence could occur at intervals, and therefore, reoperation for recurrent lesions may occasionally be required. Reoperation for distant metastases can lead to excellent long-term survival [5]. Even if unresectable metastases occur, novel targeted drugs, such as the multiple tyrosine kinase inhibitor sunitinib and the mTOR inhibitor everolimus, have been approved and registered for antiproliferative therapy for well-differentiated PanNEN [6, 7]. Therefore, it is essential to identify recurrence earlier. For this reason, indicators that could predict recurrence after surgery are required for the optimal management of PanNEN.

Several studies, however, have demonstrated that tumorigenesis and clinical manifestations of well-differentiated PanNEN are distinctively different from poorly differentiated PanNEN (neuroendocrine carcinoma; NEC), and thus, the determinants of treatment should be considered separately [8, 9]. The prognosis in NEC is poor [10], and the National Comprehensive Cancer Network (NCCN) guidelines recommend platinum-based systemic chemotherapy for patients with NEC [6]. For this reason, the 2017 World Health Organization (WHO) introduced significant changes to the classification of PanNEN. Of note, a new category of well-differentiated neoplasms, neuroendocrine tumors G3 (NET-G3), was introduced, and these are distinct from poorly differentiated NEC-G3 [11, 12].

Recently, systemic immune-inflammatory markers have been reported as factors that influence the outcomes of treatments, such as surgery, and the efficacy of chemotherapy in patients with various types of malignancies [13,14,15,16,17,18]. However, the ability of systemic immune-inflammatory markers to predict prognosis in patients with sole, well-differentiated PanNEN, based on the 2017 WHO classification [11], other than NEC or synchronous distant metastasis, has remained unknown.

In this study, we sought to evaluate whether systemic immune-inflammatory markers can be preoperative prognostic factors for predicting recurrence and overall survival after curative resection in patients with well-differentiated PanNEN based on the new 2017 WHO classification.

Materials and methods

Patients

We analyzed 132 consecutive cases who underwent surgery for primary, histologically confirmed PanNEN at the Department of Surgery, Tohoku University Hospital, between 2001 and 2018. Eight patients with synchronous hepatic metastasis during the surgery, two patients with NEC, one patient not suitable for curative resection, and one patient with an active infection at blood sampling were excluded from the study. Finally, 120 patients with well-differentiated PanNEN were enrolled in this study. Patient characteristics (age, sex), perioperative factors (serum albumin, hormonal secretion, tumor location, clinical stage), pathological findings (2017 WHO classification, tumor size, lymph node metastasis, surgical margin status, lymphovascular invasion), and prognosis were investigated retrospectively. Histopathological findings were assessed by experienced pathologists (FF, and HS). For all the patients, visual assessment ‘‘eyeballing calculation’’ was performed to assess Ki-67 index. TNM staging was adopted according to the European Neuroendocrine Tumor Society (ENETS) classification [19], and the new 2017 WHO classification of NET by the gastro-entero-pancreatic (GEP) system was used for histopathological classification [11]. Peripheral blood routine tests were performed within 14 days before surgery, according to our internal institutional policy. The serum neutrophil–lymphocyte ratio (NLR) was calculated as the number of neutrophils divided by the number of lymphocytes. The monocyte–lymphocyte ratio (MLR), platelet–lymphocyte ratio (PLR), and platelet–white cell ratio (PWR) were calculated in the same manner.

Clinical follow-up

Postoperative follow-up evaluation included physical examinations, laboratory tests, and enhanced computed tomography (chest and abdominal cavity), once every 6 months. There were no patients who received neoadjuvant or adjuvant therapies in this cohort. Treatment after recurrence was determined by the available evidence at the time of surgery and based on the patient’s condition.

Statistical analysis and software

Recurrence-free-survival (RFS) and overall survival (OS) were calculated from the date of surgery to the date of recurrence, the date of death from any cause, or the date of last follow-up. To determine the appropriate cut-off values, we used receiver operating characteristic (ROC) curves and determined the area under the curve (AUC). Differences between groups were determined using t-tests in the case of normally distributed variables or by the Wilcoxon rank-sum test in the case of abnormally distributed variables for examining differences in continuous variable distributions, and Pearson’s chi-square tests for categorical variables. RFS probabilities were compared for various categories of interest using the Kaplan–Meier method with the log-rank test.

Prognostic factors were assessed with univariate and multivariate analyses, using Cox’s proportional hazards model. Hazard ratios (HR) with 95% confidence intervals (CIs) were calculated. P < 0.05 was considered to indicate statistical significance.

All statistical analyses were performed using the JMP Pro 14.2.0 statistical software (SAS Institute Inc., Cary, NC, USA) and GraphPad Prism Version 8.4.2 (GraphPad Software, San Diego, CA, USA).

Ethics approval

This study was approved by the ethics committee of the Tohoku University Hospital (Approval No. 2020-1-322). It was performed in adherence to the tenets of the Declaration of Helsinki and its later amendments. The need to obtain written informed consent was waived due to the retrospective nature of the study.

Results

Characteristics of patients with resected well-differentiated PanNEN

The demographic and clinicopathological features of the 120 patients who underwent curative resection of PanNEN are shown in Table 1. The median age was 60 years (range 12–88 years), and the median follow-up period in all patients was 64 months (range, 6–185 months). There were no perioperative deaths. The pathological findings (based on the 2017 WHO classification) were NET-G1 in 73 patients, NET-G2 in 45, and NET-G3 in 2. The median tumor size was 14.5 mm (range 4–168 mm). Pathology investigations confirmed lymph node metastasis in 18 patients (15.0%).

Table 1 Clinicopathological characteristics of 120 patients with well-differentiated PanNEN

Clinicopathological features associated with recurrence and NLR

Postoperative recurrences were observed in 12 cases (10%). The sites of recurrence were in the liver in 10 patients, the para-aortic lymph node in 1, and the lung in 1. The 5- and 10-year RFS rates for the entire cohort were 92.0% and 78.7%, respectively. Three patients died due to PanNEN, 10 patients died due to other diseases, and the remaining 107 patients were alive at the end of the surveillance period. Thus, the 5- and 10-year disease-specific survival rates were 100% and 92.5%, respectively. The NLR was significantly higher in patients with recurrence than in those without recurrence (median NLR: 2.40 vs 1.90, P = 0.001), while the MLR, PLR, and PWR were not statistically significantly different between those with and those without recurrence (Fig. 1).

Fig. 1
figure 1

Distribution of the inflammation-based markers in PanNENs. The NLR was significantly higher in patients with recurrence than in those without recurrence, while the MLR, PLR, and PWR were not statistically different between those with and those without recurrence

An ROC curve was used to determine the cut-off value associated with postoperative recurrence. Each cut-off value of NLR and tumor size was defined as the highest log-rank statistic of any threshold. The optimal cut-off values for preoperative NLR and tumor size were 2.62 mm and 25 mm, respectively (Additional file 1: Fig. S1.). The Ki-67 index was statistically higher in patients with high NLR (≥ 2.62) than in patients with low NLR (< 2.62) (mean: 5.46 vs 3.14, P = 0.042). In contrast, age, sex, albumin, surgical margin status, clinical stage, 2017 WHO classification, tumor functionality, tumor size, tumor location, lymph node metastasis, and lymphovascular invasion were not associated with NLR status (Table 2). The recurrence rate was 33.3% and 31.0% in 18 patients with a high NLR (≥ 2.62) and 29 patients with larger tumors (≥ 25 mm), respectively.

Table 2 Relationship between NLR and clinicopathological characteristics (n = 120)

Comparison of clinical variables in relationship to RFS after curative resection

The results of the univariate and multivariate analyses for each of the clinicopathological variables are shown in Table 3. According to univariate analysis, the recurrence risk was about six times higher in patients with a high NLR than in those with a low NLR (95% CI 1.81–18.5, P = 0.004). Additionally, the TMN clinical-stage, 2017 WHO classification G2/3, tumor size, and venous invasion were also significantly predictive factors for recurrence (P < 0.05 for all). In contrast, age, sex, albumin, surgical margin status, hormonal function, tumor location, lymph node metastasis, and lymphatic invasion were not significant predictors of recurrence. Moreover, in multivariate analysis, higher NLR (HR = 3.49, 95% CI 1.05–11.7, P = 0.042) and 2017 WHO classification G2/3 (HR = 8.81, 95% CI 1.46–168.2, P = 0.015) were independent predictive factors for recurrence. A higher NLR showed a significant correlation with shorter RFS (median RFS duration, 117.8 months, P < 0.001) (Fig. 2a) and poor OS (median OS duration, 95.2 months, P = 0.032) after curative resection (Fig. 2b).

Table 3 Prognostic factors for recurrence-free-survival in 120 patients with well-differentiated PanNEN
Fig. 2
figure 2

Recurrence-free-survival and overall survival for PanNENs stratified by NLR. A higher NLR showed a significant correlation with shorter RFS (median RFS duration, 117.8 months, P < 0.001) (a) and poor OS (median OS duration, 95.2 months, P = 0.032) after curative resection (b)

Subgroup analyses of the hormonal function associated with the NLR

We then focused on the usefulness of the NLR for the classification of functional and nonfunctional PanNEN. We confirmed a strong association between NLR and RFS, especially in nonfunctional PanNEN (HR 4.95; 95% CI 1.430–20.1; P = 0.002) (Table 4). Additionally, a higher NLR was significantly associated with a shorter RFS in nonfunctional PanNEN (P = 0.009) (Fig. 3a). Contrary to nonfunctional PanNEN, NLR was not associated with RFS in functional PanNEN (P = 0.094) (Fig. 3b).

Table 4 Subgroup analysis for recurrence-free-survival according to neutrophil–lymphocyte ratio
Fig. 3
figure 3

Recurrence-free-survival for nonfunctional and functional PanNENs stratified by NLR. A higher NLR was significantly associated with a shorter RFS in nonfunctional PanNEN (a). Contrary to nonfunctional PanNEN, NLR was not associated with RFS in functional PanNEN (b)

Discussion

The current study demonstrated that elevated preoperative NLR and 2017 WHO classification independently predicted recurrence in patients with well-differentiated PanNEN after curative surgery. No previous studies have demonstrated that increased NLR serves as an independent prognostic factor in patients with nonmetastatic well-differentiated PanNEN, as defined by the 2017 WHO classification. This may be of potential clinical benefit in these patients. Furthermore, we observed that elevated preoperative NLR was predictive of a significantly shorter RFS in nonfunctional PanNEN patients.

Previously, PanNEN with lymph node metastasis, a higher Ki-67 index, and a higher 2010 WHO grade were reported to be associated with a significantly higher risk of recurrence [20, 21]. In contrast, a large international cohort study showed that the ENETs TNM classification was superior to the Union for International Cancer Control/American Joint Committee on Cancer/WHO staging system and could more accurately predict the clinical outcome of patients [22]. We revealed that the ENETs TNM classification was related to RFS in univariate analysis but not in multivariate analysis. A possible explanation for this finding is that we assessed the patients with curatively resected PanNEN and excluded metastatic stage IV patients in our present study. Clinically, the preoperative Ki-67 index obtained by fine-needle aspiration biopsy is less accurate due to intra-tumoral heterogeneity [23], which highlights the requirement of preoperative non-invasive prognostic indicators, such as inflammation-based markers. Preoperative precise assessment of recurrence risk of the patients allows clinically more relevant selection of an optimal surgical strategy, such as enucleation or further lymph node dissection.

In terms of systemic inflammation-based markers in PanNEN, preoperative NLR and PLR have been reported to be useful for predicting lymph node metastasis or recurrence [24,25,26,27,28]. However, these studies included a moderate number of patients with distant metastatic stage IV or poorly differentiated PanNEN (NEC), as defined as 2010 WHO NET-G3. Generally, poorly differentiated PanNEN has substantial distant metastases and a distinctly poor prognosis [29]. The 2010 WHO classification of NET-G3 included both well-differentiated and poorly differentiated PanNEN, resulting in a morphologically and biologically heterogeneous population [30]. Consequently, the 2017 WHO classification of NET-G3 was recategorized as only well-differentiated PanNEN, distinctively different from NEC. Indeed, the median RFS (6.7 months) and median OS (15.3 months) of surgically resected NEC were markedly shorter than in well-differentiated PanNEN in our institute (Additional file 1: Fig. S2.). Furthermore, the value of NLR for NEC was significantly higher than that in patients with well-differentiated PanNEN (Additional file 1: Fig. S3.). In our present study, we assessed the efficacy of NLR to predict recurrence in well-differentiated PanNEN, other than NEC or distant metastasis, based on the 2017 WHO classification. Hence, more prolonged RFS and OS were detected in our present study than in the previously reported ones [24,25,26,27,28].

NLR was recently reported to be associated with tumor progression in several human malignancies [14,15,16,17,18]. In addition, NLR could serve as a predictive marker in patients with not only PanNEN but also gastrointestinal NEN [27]. We previously reported that the NLR was a useful diagnostic marker for predicting intraductal papillary mucinous neoplasm with high-grade dysplasia/invasive carcinoma to differentiate low-grade dysplasia [31]. Previous studies reported that a high NLR was significantly consistent with accumulation of tumor infiltrating CD66b neutrophils or CD163+ macrophages in patients with PanNEN and pancreatic cancer, which results in poor RFS and OS [28, 32]. In general, neutrophils are markers of acute inflammation and could possibly promote tumor development and progression by providing an adequate tumor microenvironment via the production of cytokines and chemokines [33]. In addition, an increased number of lymphocytes play a crucial role in the host’s anticancer immune response; thus, lymphocytosis is generally associated with a better prognosis and a more favorable response to chemotherapy or radiation therapy in a variety of cancers [34]. Therefore, in cancer patients, peripheral blood neutrophilia and lymphopenia may reflect a weak anticancer reaction and worse clinical outcomes [35].

Regardless of the histological findings, there are hormonally functional and nonfunctional phenotypes in PanNEN. According to an epidemiological survey, the number of PanNEN patients has increased rapidly. In particular, hormonally nonfunctional PanNEN was most prevalent and increased significantly [36, 37]. To the best of our knowledge, no previous studies have demonstrated possible roles of NLR as a prognostic factor for RFS in distinct categories of nonfunctional and functional PanNEN. A high NLR has also been proven to be a risk factor of recurrence in nonfunctional PanNEN. In contrast, the NLR was statistically unrelated to RFS in functional PanNEN. One reason for this might be that we analyzed a relatively small number of the patients with hormonally functional PanNEN; only three patients in this subgroup had recurrence during the follow-up period.

Surveillance at shorter intervals might be required in patients with nonfunctional well-differentiated PanNEN with a high NLR and 2017 WHO G2/G3 classification to detect recurrence earlier after surgery. Furthermore, almost all well-differentiated PanNENs express somatostatin receptors; hence, somatostatin receptor scintigraphy should be considered in PanNEN patients with a high NLR to help identify distant metastases that could be missed by computed tomography or positron emission tomography before and after surgery [38,39,40]. Well-differentiated PanNEN with the risk factors described above may receive clinical benefits by adjuvant treatments such as somatostatin analogs after surgery. However, there is no current evidence or clinical indication for adjuvant therapy, and further studies that focus on these high-risk groups are required.

This study had some limitations. First, it was a retrospective review of a single, high-volume institution in the field of pancreatic tumors. Several studies reported the NLR cutoff values as a prognostic factor, but the cutoff values were different across these studies due to the variation in disease stage or heterogeneity of the patient population. Therefore, we need to perform the external validation study in a large population using a nationwide clinical database or multi-center trial to confirm our findings in the future. Second, although consecutive patients were enrolled, they were collected over a relatively long period, during which treatment strategies changed reasonably.

Conclusion

In summary, the results of our present study clearly demonstrated that the NLR could serve as a useful preoperative marker of clinical recurrence risks after the surgery. It is considered a convenient screening tool for the host immune response and should be incorporated into preoperative workups in the clinical management of well-differentiated PanNEN patients.

Availability of data and materials

The datasets used and analyzed during this study are available from the corresponding author upon reasonable request.

Abbreviations

PanNEN:

Pancreatic neuroendocrine neoplasm

NLR:

Neutrophil–lymphocyte

MLR:

Monocyte–lymphocyte ratio

PLR:

Platelet–lymphocyte

PWR:

Platelet–white blood cell ratio

RFS:

Recurrence-free-survival

OS:

Overall survival

References

  1. Oberg K, Eriksson B. Endocrine tumours of the pancreas. Best Pract Res Clin Gastroenterol. 2005;19(5):753–81.

    Article  Google Scholar 

  2. Cives M, Strosberg J. An update on gastroenteropancreatic neuroendocrine tumors. Oncology (Williston Park). 2014;28(9):749–56.

    Google Scholar 

  3. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135(5):1469–92.

    Article  CAS  Google Scholar 

  4. Doi R. Determinants of surgical resection for pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015;22(8):610–7.

    Article  Google Scholar 

  5. Fendrich V, Langer P, Celik I, Bartsch DK, Zielke A, Ramaswamy A, et al. An aggressive surgical approach leads to long-term survival in patients with pancreatic endocrine tumors. Ann Surg. 2006;244(6):845–51; discussion 852–43.

    Article  Google Scholar 

  6. Shah MH, Goldner WS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, et al. NCCN Guidelines insights: neuroendocrine and adrenal tumors, Version 2.2018. J Natl Compr Canc Netw. 2018;16(6):693–702.

    Article  CAS  Google Scholar 

  7. Frilling A, Modlin IM, Kidd M, Russell C, Breitenstein S, Salem R, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15(1):e8-21.

    Article  Google Scholar 

  8. Hijioka S, Hosoda W, Matsuo K, Ueno M, Furukawa M, Yoshitomi H, et al. Rb loss and KRAS mutation are predictors of the response to platinum-based chemotherapy in pancreatic neuroendocrine neoplasm with Grade 3: a Japanese multicenter pancreatic NEN-G3 study. Clin Cancer Res. 2017;23(16):4625–32.

    Article  CAS  Google Scholar 

  9. Singhi AD, Klimstra DS. Well-differentiated pancreatic neuroendocrine tumours (PanNETs) and poorly differentiated pancreatic neuroendocrine carcinomas (PanNECs): concepts, issues and a practical diagnostic approach to high-grade (G3) cases. Histopathology. 2018;72(1):168–77.

    Article  Google Scholar 

  10. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013;24(1):152–60.

    Article  CAS  Google Scholar 

  11. Lloyd ROR, Kloppel G, et al. WHO classification of tumours of endocrine organs. 4th ed. Lyon: International Agency for Research on Cancer Press; 2017.

    Google Scholar 

  12. Scoazec JY, Couvelard A. Classification of pancreatic neuroendocrine tumours: changes made in the 2017 WHO classification of tumours of endocrine organs and perspectives for the future. Ann Pathol. 2017;37(6):444–56.

    Article  Google Scholar 

  13. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–7.

    Article  CAS  Google Scholar 

  14. Ohira M, Yoshizumi T, Yugawa K, Kosai-Fujimoto Y, Inokuchi S, Motomura T, et al. Association of inflammatory biomarkers with long-term outcomes after curative surgery for mass-forming intrahepatic cholangiocarcinoma. Surg Today. 2020;50(4):379–88.

    Article  CAS  Google Scholar 

  15. Migita K, Matsumoto S, Wakatsuki K, Ito M, Kunishige T, Nakade H, et al. The prognostic significance of inflammation-based markers in patients with recurrent gastric cancer. Surg Today. 2018;48(3):282–91.

    Article  CAS  Google Scholar 

  16. Cananzi FCM, Ruspi L, Quagliuolo VL. Preoperative monocyte-to-lymphocyte ratio predicts recurrence in gastrointestinal stromal tumors. J Surg Oncol. 2019;119(7):1026.

    Article  Google Scholar 

  17. Kawai M, Hirono S, Okada KI, Miyazawa M, Shimizu A, Kitahata Y, et al. Low lymphocyte monocyte ratio after neoadjuvant therapy predicts poor survival after pancreatectomy in patients with borderline resectable pancreatic cancer. Surgery. 2019;165(6):1151–60.

    Article  Google Scholar 

  18. Chen F, Lin L, Yan L, Qiu Y, Cai L, He B. Preoperative neutrophil-to-lymphocyte ratio predicts the prognosis of oral squamous cell carcinoma: a large-sample prospective study. J Oral Maxillofac Surg. 2017;75(6):1275–82.

    Article  Google Scholar 

  19. Rindi G, Kloppel G, Alhman H, Caplin M, Couvelard A, de Herder WW, et al. TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006;449(4):395–401.

    Article  CAS  Google Scholar 

  20. Genc CG, Falconi M, Partelli S, Muffatti F, van Eeden S, Doglioni C, et al. Recurrence of pancreatic neuroendocrine tumors and survival predicted by Ki67. Ann Surg Oncol. 2018;25(8):2467–74.

    Article  CAS  Google Scholar 

  21. Song KB, Kim SC, Kim JH, Hong SM, Park KM, Hwang DW, et al. Prognostic factors in 151 patients with surgically resected non-functioning pancreatic neuroendocrine tumours. ANZ J Surg. 2016;86(7–8):563–7.

    Article  Google Scholar 

  22. Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L, Buchler MW, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764–77.

    Article  CAS  Google Scholar 

  23. Rebours V, Cordova J, Couvelard A, Fabre M, Palazzo L, Vullierme MP, et al. Can pancreatic neuroendocrine tumour biopsy accurately determine pathological characteristics? Dig Liver Dis. 2015;47(11):973–7.

    Article  Google Scholar 

  24. Zou J, Li Q, Kou F, Zhu Y, Lu M, Li J, et al. Prognostic value of inflammation-based markers in advanced or metastatic neuroendocrine tumours. Curr Oncol. 2019;26(1):e30–8.

    Article  CAS  Google Scholar 

  25. Zhou B, Zhan C, Wu J, Liu J, Zhou J, Zheng S. Prognostic significance of preoperative neutrophil-to-lymphocyte ratio in surgically resectable pancreatic neuroendocrine tumors. Med Sci Monit. 2017;23:5574–88.

    Article  CAS  Google Scholar 

  26. Tong Z, Liu L, Zheng Y, Jiang W, Zhao P, Fang W, et al. Predictive value of preoperative peripheral blood neutrophil/lymphocyte ratio for lymph node metastasis in patients of resectable pancreatic neuroendocrine tumors: a nomogram-based study. World J Surg Oncol. 2017;15(1):108.

    Article  Google Scholar 

  27. Salman T, Kazaz SN, Varol U, Oflazoglu U, Unek IT, Kucukzeybek Y, et al. Prognostic value of the pretreatment neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio for patients with neuroendocrine tumors: an Izmir oncology group study. Chemotherapy. 2016;61(6):281–6.

    Article  CAS  Google Scholar 

  28. Harimoto N, Hoshino K, Muranushi R, Hagiwara K, Yamanaka T, Ishii N, et al. Prognostic significance of neutrophil-lymphocyte ratio in resectable pancreatic neuroendocrine tumors with special reference to tumor-associated macrophages. Pancreatology. 2019;19(6):897–902.

    Article  CAS  Google Scholar 

  29. Strosberg JR, Coppola D, Klimstra DS, Phan AT, Kulke MH, Wiseman GA, et al. The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high-grade) extrapulmonary neuroendocrine carcinomas. Pancreas. 2010;39(6):799–800.

    Article  Google Scholar 

  30. Basturk O, Yang Z, Tang LH, Hruban RH, Adsay V, McCall CM, et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol. 2015;39(5):683–90.

    Article  Google Scholar 

  31. Hata T, Mizuma M, Motoi F, Ishida M, Morikawa T, Takadate T, et al. Diagnostic and prognostic impact of neutrophil-to-lymphocyte ratio for intraductal papillary mucinous neoplasms of the pancreas with high-grade dysplasia and associated invasive carcinoma. Pancreas. 2019;48(1):99–106.

    Article  Google Scholar 

  32. Takakura K, Ito Z, Suka M, Kanai T, Matsumoto Y, Odahara S, et al. Comprehensive assessment of the prognosis of pancreatic cancer: peripheral blood neutrophil–lymphocyte ratio and immunohistochemical analyses of the tumour site. Scand J Gastroenterol. 2016;51(5):610–7.

    Article  CAS  Google Scholar 

  33. Gregory AD, Houghton AM. Tumor-associated neutrophils: new targets for cancer therapy. Cancer Res. 2011;71(7):2411–6.

    Article  CAS  Google Scholar 

  34. Barnes TA, Amir E. HYPE or HOPE: the prognostic value of infiltrating immune cells in cancer. Br J Cancer. 2017;117(4):451–60.

    Article  CAS  Google Scholar 

  35. Halazun KJ, Hardy MA, Rana AA, Woodland DCT, Luyten EJ, Mahadev S, et al. Negative impact of neutrophil–lymphocyte ratio on outcome after liver transplantation for hepatocellular carcinoma. Ann Surg. 2009;250(1):141–51.

    Article  Google Scholar 

  36. Ito T, Lee L, Hijioka M, Kawabe K, Kato M, Nakamura K, et al. The up-to-date review of epidemiological pancreatic neuroendocrine tumors in Japan. J Hepatobiliary Pancreat Sci. 2015;22(8):574–7.

    Article  Google Scholar 

  37. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26(18):3063–72.

    Article  Google Scholar 

  38. Treglia G, Castaldi P, Rindi G, Giordano A, Rufini V. Diagnostic performance of Gallium-68 somatostatin receptor PET and PET/CT in patients with thoracic and gastroenteropancreatic neuroendocrine tumours: a meta-analysis. Endocrine. 2012;42(1):80–7.

    Article  CAS  Google Scholar 

  39. Sadowski SM, Neychev V, Millo C, Shih J, Nilubol N, Herscovitch P, et al. Prospective study of 68Ga-DOTATATE positron emission tomography/computed tomography for detecting gastro-entero-pancreatic neuroendocrine tumors and unknown primary sites. J Clin Oncol. 2016;34(6):588–96.

    Article  CAS  Google Scholar 

  40. Matsumoto T, Okabe H, Yamashita YI, Yusa T, Itoyama R, Nakao Y, et al. Clinical role of fludeoxyglucose (18F) positron emission tomography/computed tomography ((18)F-FDG PET/CT) in patients with pancreatic neuroendocrine tumors. Surg Today. 2019;49(1):21–6.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

No funding supported this work.

Author information

Authors and Affiliations

Authors

Contributions

TM: protocol/project development, data collection and management, and manuscript writing. HO, TA, and SA: protocol/project development, data collection and management. TM, TK, and MU: protocol/project development, management, and manuscript editing. TH, TT, SM, and KA: data collection and management. KK and KM: data collection. MI and MM: data analysis and manuscript editing. FF and HS: histopathological analysis and assessment. All authors read and approved final manuscript.

Corresponding author

Correspondence to Hideo Ohtsuka.

Ethics declarations

Ethics approval and consent to participate

All procedures performed in studies involving human participants were in accordance with the ethical standards of the Institutional Research Committee and with the 1964 Helsinki Declaration and its later amendments or with comparable ethical standards. This study was approved by the Ethics Committee of the Tohoku University Hospital (institutional review board approval No. 2019-1-303). The Ethics Committee of the Tohoku University Hospital waived the need for informed consent because of the of the study’s retrospective nature.

Consent for publication

Not applicable.

Competing interests

All authors have no conflicts of interest and no financial disclosures.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Fig. S1.

ROC curve for the NLR and tumor size in well-differentiated PanNENs. The ROC curve illustrated that NLR has an AUC of 0.664 (95% CI 0.464–0.864) and tumor size has an AUC of 0.801 (95% CI 0.675–0.945). Arrows indicate optimal cut-off values. Fig. S2. Recurrence-free-survival and overall survival for PanNENs stratified by the 2017 WHO classification. Recurrence-free-survival and overall survival of neuroendocrine carcinoma (NEC) were significantly shorter than those of well-differentiated PanNENs (NET G1-3). Fig. S3. Distribution of NLR in PanNENs stratified by the 2017 WHO classification. The value of NLR in patients with NEC was significantly higher than that in patients with well-differentiated PanNEN.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miura, T., Ohtsuka, H., Aoki, T. et al. Increased neutrophil–lymphocyte ratio predicts recurrence in patients with well-differentiated pancreatic neuroendocrine neoplasm based on the 2017 World Health Organization classification. BMC Surg 21, 176 (2021). https://doi.org/10.1186/s12893-021-01178-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12893-021-01178-3

Keywords