Next Article in Journal
Comprehensive Analysis of Disease Pathology in Immunocompetent and Immunocompromised Hosts following Pulmonary SARS-CoV-2 Infection
Next Article in Special Issue
SOX9 Protein in Pancreatic Cancer Regulates Multiple Cellular Networks in a Cell-Specific Manner
Previous Article in Journal
COVID-19 and the Importance of Being Prepared: A Multidisciplinary Strategy for the Discovery of Antivirals to Combat Pandemics
Previous Article in Special Issue
Prognostic Implications of Portal Venous Circulating Tumor Cells in Resectable Pancreatic Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Development of Nomograms for Predicting Prognosis of Pancreatic Cancer after Pancreatectomy: A Multicenter Study

1
Division of Hepatobiliary-Pancreatic Surgery, Department of Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea
2
Biomedical Statistics Center, Samsung Medical Center, Research Institute for Future Medicine, Seoul 06351, Korea
3
Department of Surgery, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul 03181, Korea
4
Department of Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, College of Medicine, Seoul National University, Seoul 07061, Korea
5
Department of Surgery, College of Medicine, Ilsan Paik Hospital, Inje University, Goyang 10380, Korea
6
Department of Surgery, College of Medicine, Ajou University Hospital, Ajou University, Suwon 16499, Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(6), 1341; https://doi.org/10.3390/biomedicines10061341
Submission received: 4 May 2022 / Revised: 31 May 2022 / Accepted: 2 June 2022 / Published: 7 June 2022
(This article belongs to the Special Issue Pancreatic Cancer: From Mechanisms to Therapeutic Approaches)

Abstract

:
Surgical resection is the only curative treatment for pancreatic ductal adenocarcinoma (PDAC). Currently, the TNM classification system is considered the standard for predicting prognosis after surgery. However, the prognostic accuracy of the system remains limited. This study aimed to develop new predictive nomograms for resected PDAC. The clinicopathological data of patients who underwent surgery for PDAC between 2006 and 2015 at five major institutions were retrospectively reviewed; 885 patients were included in the analysis. Cox regression analysis was performed to investigate prognostic factors for recurrence and survival, and statistically significant factors were used for creating nomograms. The nomogram for predicting recurrence-free survival included nine factors: sarcopenic obesity, elevated carbohydrate antigen 19–9, platelet-to-lymphocyte ratio, preoperatively-identified arterial abutment, estimated blood loss (EBL), tumor differentiation, size, lymph node ratio, and tumor necrosis. The nomogram for predicting overall survival included 10 variables: age, underlying liver disease, chronic kidney disease, preoperatively found portal vein invasion, portal vein resection, EBL, tumor differentiation, size, lymph node metastasis, and tumor necrosis. The time-dependent area under the receiver operating characteristic curve for both nomograms exceeded 0.70. Nomograms were developed for predicting survival after resection of PDAC, and the platforms showed fair predictive performance. These new comprehensive nomograms provide information on disease status and are useful for determining further treatment for PDAC patients.

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal malignancy, with a poor overall survival rate of approximately 10% [1]. The only curative treatment is surgical resection. As reported in recent studies, five-year survival for patients with resected PDAC has increased up to 17% [2]. The most widely accepted method for predicting survival outcomes after surgery is the American Joint Committee on Cancer (AJCC) system, consisting of tumor extent (T stage), lymph node status (N stage), and distant metastasis (M stage) (TNM staging) [3]. However, even the most recent AJCC system showed limited prognostic accuracy in previous multi-center validation studies [4,5]. In addition, because this system was largely based on a Western population, the validity of the system for general population is unclear.
Numerous attempts have been made to identify prognostic factors for PDAC apart from several well-known factors such as TNM stage and carbohydrate antigen 19–9 (CA 19–9). For preoperative factors, inflammatory markers such as neutrophil-to-lymphocyte (NLR) ratio have been studied as prognosticators [6,7]. Additionally, the prognostic values of nutritional status including sarcopenia are under assessment [8]. Intraoperatively, estimated blood loss (EBL) and transfusion have been reported as independent risk factors for survival [9]. In terms of pathologic features, lymph node ratio (LNR) and resection margin status are considered potentially associated with outcomes of patients with resected PDAC [2,10,11].
Some recent studies tried to develop platforms for predicting the prognosis of PDAC. The authors proposed nomograms predicting recurrence or survival, and the C-indices ranged from 0.64 to 0.73 [12,13]. These nomograms could be useful in easily calculating the probability of survival, but up-to-date variables were not included in the analyses. In the present study, we aimed to develop a new comprehensive nomogram with more prognosticators, which could estimate the probability of survival more accurately in patients with PDAC after surgical resection.

2. Materials and Methods

2.1. Data Collection

From January 2006 to December 2015, 963 patients were diagnosed with PDAC and underwent pancreatectomy at five different centers: Samsung Medical Center, Kangbuk Samsung Hospital, Seoul National University Boramae Medical Center, Ilsan Paik Hospital, and Ajou University Hospital. Patients’ medical records, including data for recurrence, were retrospectively reviewed. Patients with missing values or lacking recurrence data were excluded. Finally, a total of 885 patients were included in the analyses. This study was approved by the Institutional Review Board of Samsung Medical Center (Seoul, Korea, approval number: 2020–11–133).

2.2. Clinical Variables for Analysis

The demographic data and preoperative laboratory results, including CA 19–9, were collected. NLR and platelet-to-lymphocyte ratio (PLR) were calculated and included in the analysis as inflammatory markers. Preoperative computed tomography (CT) scans taken within 2 weeks of surgery were used to evaluate and diagnose sarcopenia. The diagnostic cut-off values for sarcopenia were based on a cohort study from a Korean national institution [14]: skeletal muscle index (SMI = skeletal muscle area at L3/height2) < 50.18 cm2/m2 for males and SMI < 38.63 cm2/m2 for females. Sarcopenic obesity (SO) was defined as visceral fat area (VFA)/SMI ≥ 2.5 according to our previous study on the effect of SO in patients with pancreatic cancer [8,15]. The preoperative relationship between tumors and the following major vessels were also evaluated: common hepatic artery (CHA), superior mesenteric artery (SMA), portal vein (PV), and superior mesenteric vein (SMV).
In terms of operation-related factors, types of operation including combined vascular resection, EBL, and the need for red blood cell transfusions were reviewed. In the final pathology reports, the size of tumor, lymph node (LN) status, and resection margin status were reported. Revised R1 resection refers to the existence of tumor within 1 mm of the resection margins and was included in R1. LNR was calculated as the number of metastatic LNs divided by the number of harvested LNs.
The optimal cut-off points were selected using log-rank test statistics to maximize the difference between low and high groups for the following variable: NLR, PLR, tumor size, and EBL, and the cut-off values were determined to be 2, 90, 2 cm, and 500 mL, respectively.
Determining cancer recurrence was based on abdomino-pelvic CT scans and CA 19–9 levels during postoperative surveillance. An additional chest CT or positron emission tomography (PET) scan was performed to confirm recurrent tumors. Recurrence-free survival (RFS) was measured by the time from surgery to the date of recurrence or last follow-up. Overall survival (OS) was defined as the time between surgery and death from any cause, by 30 June 2021.

2.3. Statistical Analysis

The demographic and clinicopathological data were presented as mean ± standard deviation (SD) for continuous variables, and frequency with percentile for categorical variables. Cox regression analysis was performed to assess risk factors for RFS and OS, and hazard ratios (HRs) were reported with 95% confidence intervals (CIs). The final model was determined after applying the backward selection method for variables with p < 0.05 in the multivariable model. To assess the discrimination of model, the C-index (Harrell’s concordance statistic) and the receiver operating characteristic (ROC) curve with time-dependent area under the curve (AUC) were used. Standard error (SE) and 95% CIs were presented. The bootstrap procedure with the number of 2000 was employed for internal validation of prediction models. Based on the bootstrap, the bias-corrected (overfitting-corrected) C-indices were obtained, and calibration curves were plotted with predicted and observed values. The results were considered statistically significant when two-sided p-values were <0.05. All statistical analyses were conducted using SAS software, version 9.4 (SAS Institute Inc., Cary, NC, USA.) and R software, version 4.0.5 (R Project for Statistical Computing).

3. Results

The demographic and clinicopathological data of the development cohort are summarized in Table 1. The mean age of the patients at operation was 63.1 years, and 58.8% were male. There were 99 (11.2%) sarcopenic patients and 285 (32.2%) with SO. On preoperative imaging, 166 (18.8%) patients showed arterial abutment and 260 (29.4%) patients showed PV/SMV abutment or invasion. Pancreatoduodenectomy, left-sided pancreatectomy, and total pancreatectomy were performed in 585 (66.1%), 295 (33.3%), and 5 (0.6%) patients, respectively. R0 resection was accomplished in 671 (75.8%) patients.
Table 2 shows risk factor analysis for RFS. In multivariable analysis, SO (HR: 1.22, 95% CI: 1.03–1.44, p = 0.020), elevated CA 19–9 (HR: 1.28, 95% CI: 1.07–1.53, p = 0.006), PLR (HR: 1.34, 95% CI: 1.07–1.68, p = 0.010), and CHA/SMA abutment (HR: 1.31, 95% CI: 1.08–1.59, p = 0.007) were preoperative factors associated with RFS. In addition, EBL (HR: 1.51, 95% CI: 1.28–1.78, p < 0.001), moderate and poor differentiation (HR: 1.43, 95% CI: 1.07–1.90, p = 0.015, HR: 1.83, 95% CI: 1.35–2.49, p < 0.001, respectively), tumor size (HR: 1.67, 95% CI: 1.33–2.09, p < 0.001), LN ratio (HR: 1.14, 95% CI: 1.08–1.20, p < 0.001), and tumor necrosis (HR: 1.56, 95% CI: 1.27–1.93, p < 0.001) were related to RFS.
In risk factor analysis for OS (Table 3), age at operation (HR: 1.16, 95% CI: 1.07–1.25, p < 0.001), underlying liver and chronic kidney disease (HR: 1.58, 95% CI: 1.12–2.22, p = 0.009 and HR: 3.62, 95% CI: 1.58–8.28, p = 0.002, respectively), preoperative PV/SMV invasion (HR: 1.74, 95% CI: 1.32–2.29, p < 0.001), PV/SMV resection (HR: 1.38, 95% CI: 1.10–1.72, p = 0.005), and EBL (HR: 1.36, 95% CI: 1.16–1.59, p < 0.001) were clinically significant factors in multivariable analysis. In pathologic features, moderate and poor differentiation (HR: 1.34, 95% CI: 1.01–1.78, p = 0.040 and HR: 2.20, 95% CI: 1.64–2.96, p < 0.001, respectively), tumor size (HR: 1.53, 95% CI: 1.24–1.89, p < 0.001), LN metastasis (HR: 1.69, 95% CI: 1.42–1.99, p < 0.001), and tumor necrosis (HR: 1.67, 95% CI: 1.37–2.04, p < 0.001) were related factors.
New nomograms predicting probabilities of RFS and OS at 2 years and 5 years after surgery were established (Figure 1), using the variables identified from the multivariable Cox’s proportional hazard models: sarcopenic obesity; elevated carbohydrate antigen 19–9; platelet-to-lymphocyte ratio; preoperatively-identified arterial abutment; estimated blood loss (EBL); tumor differentiation, size, lymph node ratio, and tumor necrosis for RFS models; age at operation; underlying liver disease; chronic kidney disease; preoperatively found portal vein invasion; portal vein resection; EBL; and tumor differentiation, size, lymph node metastasis, and tumor necrosis for OS models. Points were assigned to each variable considering the HR from the Cox model. The time-dependent AUCs were as follows: 0.742 (95% CI: 0.703–0.781, SE: 0.020) and 0.765 (95% CI: 0.717–0.813, SE: 0.025) for two-year and five-year RFS model, 0.723 (95% CI: 0.686–0.760, SE: 0.019) and 0.732 (95% CI: 0.699–0.767, SE: 0.017) for two-year and five-year OS model. Calibration plots of the models through 2000 bootstrap resamples were constructed to show the agreement between the predicted and actual probabilities (Figure 2). The bias-corrected two-year and fove-year AUCs were 0.737 (95% CI: 0.701–0.773) and 0.760 (95% CI: 0.717–0.803) for the RFS model and 0.721 (95% CI: 0.688–0.754) and 0.729 (95% CI: 0.693–0.766) for the OS model, respectively. Lastly, a user-friendly website containing calculators was built, in which users can obtain the survival probabilities automatically calculated by simply filling in the blanks (Samsung Medical Center—All rights reserved, © 2022. URL: http://pdacprognosis.smchbp.org/, accessed on 30 March 2022) (Figure 3).

4. Discussion

In the present study, we proposed nomograms for predicting RFS and OS after resection of PDAC. These new nomograms included recently evaluated diverse risk factors as well as conventional factors, which could be easily obtained from patient perioperative data. The predictive power of the nomograms was fair, with the AUC values exceeding 0.70. Additionally, the web calculators based on the nomograms have been established, providing a user-friendly interface for potential users.
During the process of investigating risk factors for composing new nomograms, some novel prognostic factors showed association with survival of PDAC patients. Among preoperative variables, PLR was significantly associated with RFS. Reportedly, chronic inflammation plays an important role in carcinogenesis of various GI tract malignancies as well as in PDAC [16,17]. Several studies have proposed possible links between PDAC and numerous inflammatory parameters, but the findings were somewhat inconsistent. Elevated NLR was shown to predict a poor prognosis in some studies [6]; however, only PLR was a significant risk factor in other studies [7]. In our previous single institutional study, both NLR and PLR were associated with the OS of patients with resected PDAC [2]. In the present study based on multi-center data, only PLR was significantly associated with RFS. Other parameters, such as lymphocyte-to-monocyte ratio, advanced lung cancer inflammation index, and prognostic nutritional index, which are readily available hematologic markers, should also be investigated. Further studies are needed in which the utility of several inflammatory markers is evaluated and the appropriate cut-off values identified to predict recurrence or survival.
Another significant preoperative factor related to RFS was SO. Sarcopenia has been an important issue for the last decade because many reports have suggested its relation to various adverse outcomes such as poor physical performance and even mortality [18,19]. Particularly for patients undergoing surgery for GI tract malignancies, sarcopenia is a risk factor for postoperative complications or poor survival [8,20]. In this study, SO rather than sarcopenia was an independent risk factor for RFS. SO is an indicator that reflects excessive body fat mass as well as low muscle mass, of which the pathogenesis involves diverse factors including both lifestyle and medical factors [15]. SO is a potentially adjustable risk factor that should be taken into consideration. Although only preoperative status was measured in the present study, sarcopenia or SO can change postoperatively because body composition is altered due to physiologic stress from surgery. Further in-depth analysis on the relationship between SO and oncologic outcomes would be helpful to emphasize the importance of SO to patients undergoing surgery for PDAC. In terms of diagnosis, several organizations including the European Working Group for Sarcopenia in Older People (EWGSOP) and the Asian Working Group for Sarcopenia (AWGS) have suggested different diagnostic criteria for sarcopenia and SO. In addition, various diagnostic tools for sarcopenia are being used, mostly dual-energy X-ray absorptiometry (DXA) or bioelectrical impedance analysis (BIA). In our study, we used CT scans for measuring body composition considering that every patient with a scheduled surgery takes routine preoperative CT. Recently, handgrip strength or cross-sectional area of bicep brachii muscle were suggested as assessment tools for sarcopenia [14,21]. The diagnostic concordance between the various tools has not been investigated, which may have led to inconsistency among studies in which the role of sarcopenia or SO on postoperative outcomes was investigated. In our institution, a prospective study using the various above-mentioned tools is planned to identify optimal diagnostic methods and clinical implications of sarcopenia-related markers in patients with PDAC.
Among pathological features, an important prognostic factor for resected PDAC is LN status. The N stage in the current TNM staging system refers only to the number of metastatic LNs. In this regard, concerns exist in terms of stage migration because the number of metastatic nodes could be affected by the number of harvested nodes. In a previous study, the total number of LNs obtained was reported to have a strong influence on the survival of PDAC patients [22]. The authors proposed that a minimum of 15 nodes be resected to gain survival benefits. The International Study Group on Pancreatic Surgery (ISGPS) also suggested that at least 12 or 15 LNs should be retrieved for accurate staging [23]. Extended lymphadenectomy, particularly in pancreatoduodenectomy, is not recommended due to its morbidity without obvious survival benefits [24]. Consequently, LN ratio could be more feasible to reflect the nodal status. LNR was identified as a prognostic factor for resected PDAC in several previous studies [10,11], and these findings are consistent with the results of the present study, which showed LNR was an independent risk factor for RFS. However, because consensus has not been reached regarding the optimal cut-off value for LNR, a numerical value was applied in our nomogram. Many studies have suggested potentially relevant cut-off values; however, the median number of harvested LNs was inconsistent among the studies. To effectively utilize the nodal status of PDAC, further analytical approaches using well-organized pathological data should be considered.
Regarding the variables included in the final models, the components were different between the RFS model and the OS model. In the nomogram for predicting OS, age at operation as well as underlying liver and chronic kidney diseases are included. Because OS refers to all-cause mortality, the factors which can affect the performance of patients are potentially associated with OS. These factors might also have influenced the completion of adjuvant therapy after surgery. The results of the present study could be a basis for future studies investigating the clinical implications of common chronic diseases and related parameters in patients undergoing pancreatectomy for PDAC. Similarly, the oncologic or survival benefit of PV/SMV resection (PV/SMVR), mostly performed with PD, has been a topic of debate, and this study showed that PV/SMVR was associated with poor OS. This result may indicate that venous resection is accompanied by increased morbidity with no actual survival benefit, as demonstrated in several previous studies [25,26]. Conversely, in a recent study, propensity score matching analysis showed that PV/SMVR might be a reasonable option for patients with PV/SMV involvement to achieve survival outcomes comparable to patients without PV/SMV involvement without increasing severe postoperative complications [27]. Further investigation is necessary to verify the actual survival benefit and safety of PV/SMVR in patients undergoing pancreatectomy.
In terms of predictability, the AUC values of our nomograms were >0.7, surpassing the predictive power of some previous prediction systems. First, van Roessel et al. performed external validation of the recent TNM staging system using an international multicenter cohort [5]. The results indicated the possibility for further improvement with a C-index value of 0.57. An original nomogram study was conducted in 2004 with prospectively collected single-institutional data, and the nomogram was developed to predict disease-specific survival at 1, 2, and 3 years after surgery [12]. The bootstrap-corrected C-index was 0.64, showing better discriminative power than contemporary TNM staging, but still limited predictability compared with our nomograms. Meanwhile, in 2013, there was a pilot study proposing a prediction platform for resected PDAC based on artificial intelligence technique [28]. Although the number of the subjects was only 84, the possibility that artificial intelligence might offer better prediction than conventional Cox regression models was demonstrated. Many studies are being performed to compare the predictive ability between nomogram and machine learning techniques using a database of several types of cancer [29,30]. In our institution, a combinations of various analytical techniques is being investigated to improve the predictive platforms.
The present study has several limitations. First, the study was based on a retrospectively reviewed database collected from multiple institutions. There are many possible sources of heterogeneity, including differences in diagnostic protocols, perioperative management, surgical techniques, and physicians who reviewed the medical records. In addition, selection bias might have affected the study because some patients with insufficient data regarding recurrence were excluded. However, this study has several strengths. We used a large database with several up-to-date variables, such as inflammatory markers and sarcopenia. Some variables were identified as independent prognostic factors and were included in the new models. With these prognosticators, nomograms were proposed that provide intuitive visualization of the predictive power of each factor. Furthermore, using the nomograms, a website was constructed where potential users can easily calculate the survival probability of a patient. Notably, our new platforms showed fair predictive ability, with AUC values exceeding 0.7. The development of another platform using artificial intelligence, which could have an advantage over conventional statistics in terms of data imputation, is planned at our institution. We are also planning to perform external validation in order to verify the utility of the new platforms for the general population.
In conclusion, new nomograms predicting prognosis of PDAC after surgical resection were proposed. These new platforms could offer a patient considerable insight into the disease status and act as a reference for further treatment. A future study with more extensive data and artificial intelligence technique is planned to improve the predictive performance of the platforms.

Author Contributions

Conceptualization, S.J.Y., J.K., C.-S.L., Y.C.S., W.J., S.H.S., J.S.H. and I.W.H.; methodology, S.J.Y., B.P. and I.W.H.; formal analysis, all authors; investigation, S.J.Y., B.P. and I.W.H.; writing—original draft preparation, S.J.Y., B.P. and I.W.H.; writing—review and editing, all authors; supervision, J.K., C.-S.L., Y.C.S., W.J., S.H.S., J.S.H. and I.W.H.; funding acquisition, I.W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Samsung Medical Center, Grant #SMO1210421.

Institutional Review Board Statement

This study was approved by the Institutional Review Board of Samsung Medical Center (Seoul, Korea, approval number: 2020–11-133).

Informed Consent Statement

Our Institutional Review Board of Samsung Medical Center waived the need for written informed consent from the participants because the research involved no more than minimal risk to subjects, and there was no reason to assume rejection of agreement.

Data Availability Statement

Research data are not shared.

Acknowledgments

The authors would like to thank Hyemin Kim (Data Manager, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine) for help with data collection.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PDACpancreatic ductal adenocarcinoma
AJCCthe American Joint Committee on Cancer
CA 19–9carbohydrate antigen 19–9
NLRneutrophil-to-lymphocyte
EBLestimated blood loss
LNRlymph node ratio
PLRplatelet-to-lymphocyte ratio
CTcomputed tomography
SMIskeletal muscle index
SOSarcopenic obesity
VFAvisceral fat area
CHAcommon hepatic artery
SMAsuperior mesenteric artery
PVportal vein
SMVsuperior mesenteric vein
LNlymph node
PETpositron emission tomography
RFSrecurrence-free survival
OSoverall survival
SDstandard deviation
HRhazard ratio
CIconfidence interval
ROCcurve, receiver operating characteristic curve
AUCarea under the curve
SEstandard error

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Yoon, S.J.; Shin, S.H.; Yoon, S.K.; Jung, J.H.; You, Y.; Han, I.W.; Choi, D.W.; Heo, J.S. Appraisal of 5-year recurrence-free survival after surgery in pancreatic ductal adenocarcinoma. J. Hepatobiliary Pancreat. Sci. 2021, 28, 287–296. [Google Scholar] [CrossRef] [PubMed]
  3. Amin, M.B.; Edge, S.; Greene, F.; Byrd, D.R.; Brookland, R.K.; Washington, M.K.; Gershenwald, J.E.; Compton, C.C.; Hess, K.R.; Sullivan, D.C.; et al. AJCC Cancer Staging Manual, 8th ed.; Springer: New York, NY, USA, 2017. [Google Scholar]
  4. Allen, P.J.; Kuk, D.; Castillo, C.F.; Basturk, O.; Wolfgang, C.L.; Cameron, J.L.; Lillemoe, K.D.; Ferrone, C.R.; Morales-Oyarvide, V.; He, J.; et al. Multi-institutional Validation Study of the American Joint Commission on Cancer (8th Edition) Changes for T and N Staging in Patients with Pancreatic Adenocarcinoma. Ann. Surg. 2017, 265, 185–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Van Roessel, S.; Kasumova, G.G.; Verheij, J.; Najarian, R.M.; Maggino, L.; de Pastena, M.; Malleo, G.; Marchegiani, G.; Salvia, R.; Ng, S.C.; et al. International Validation of the Eighth Edition of the American Joint Committee on Cancer (AJCC) TNM Staging System in Patients with Resected Pancreatic Cancer. JAMA Surg. 2018, 153, e183617. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, J.J.; Hu, Z.G.; Shi, W.X.; Deng, T.; He, S.Q.; Yuan, S.G. Prognostic significance of neutrophil to lymphocyte ratio in pancreatic cancer: A meta-analysis. World J. Gastroenterol. 2015, 21, 2807–2815. [Google Scholar] [CrossRef] [PubMed]
  7. Hoshimoto, S.; Hishinuma, S.; Shirakawa, H.; Tomikawa, M.; Ozawa, I.; Ogata, Y. Validation and clinical usefulness of pre- and postoperative systemic inflammatory parameters as prognostic markers in patients with potentially resectable pancreatic cancer. Pancreatology 2020, 20, 239–246. [Google Scholar] [CrossRef]
  8. Ryu, Y.; Shin, S.H.; Kim, J.H.; Jeong, W.K.; Park, D.J.; Kim, N.; Heo, J.S.; Choi, D.W.; Han, I.W. The effects of sarcopenia and sarcopenic obesity after pancreaticoduodenectomy in patients with pancreatic head cancer. HPB 2020, 22, 1782–1792. [Google Scholar] [CrossRef]
  9. Tamagawa, H.; Aoyama, T.; Yamamoto, N.; Kamiya, M.; Murakawa, M.; Atsumi, Y.; Numata, M.; Kazama, K.; Hara, K.; Yukawa, N.; et al. The Impact of Intraoperative Blood Loss on the Survival of Patients with Stage II/III Pancreatic Cancer. In Vivo 2020, 34, 1469–1474. [Google Scholar] [CrossRef]
  10. Aoyama, T.; Yamamoto, N.; Kamiya, M.; Murakawa, M.; Tamagawa, H.; Sawazaki, S.; Numata, M.; Shiozawa, M.; Kobayashi, S.; Ueno, M.; et al. The Lymph Node Ratio Is an Independent Prognostic Factor in Pancreatic Cancer Patients Who Receive Curative Resection Followed by Adjuvant Chemotherapy. Anticancer Res. 2018, 38, 4877–4882. [Google Scholar] [CrossRef]
  11. Yamamoto, Y.; Ikoma, H.; Morimura, R.; Konishi, H.; Murayama, Y.; Komatsu, S.; Shiozaki, A.; Kuriu, Y.; Kubota, T.; Nakanishi, M.; et al. The clinical impact of the lymph node ratio as a prognostic factor after resection of pancreatic cancer. Anticancer Res. 2014, 34, 2389–2394. [Google Scholar]
  12. Brennan, M.F.; Kattan, M.W.; Klimstra, D.; Conlon, K. Prognostic nomogram for patients undergoing resection for adenocarcinoma of the pancreas. Ann. Surg. 2004, 240, 293–298. [Google Scholar] [CrossRef]
  13. Song, W.; Miao, D.L.; Chen, L. Nomogram for predicting survival in patients with pancreatic cancer. Onco. Targets Ther. 2018, 11, 539–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Moon, J.H.; Kim, K.M.; Kim, J.H.; Moon, J.H.; Choi, S.H.; Lim, S.; Lim, J.Y.; Kim, K.W.; Park, K.S.; Jang, H.C. Predictive Values of the New Sarcopenia Index by the Foundation for the National Institutes of Health Sarcopenia Project for Mortality among Older Korean Adults. PLoS ONE 2016, 11, e0166344. [Google Scholar] [CrossRef] [PubMed]
  15. Prado, C.M.; Wells, J.C.; Smith, S.R.; Stephan, B.C.; Siervo, M. Sarcopenic obesity: A Critical appraisal of the current evidence. Clin. Nutr. 2012, 31, 583–601. [Google Scholar] [CrossRef] [PubMed]
  16. Storz, P.; Crawford, H.C. Carcinogenesis of Pancreatic Ductal Adenocarcinoma. Gastroenterology 2020, 158, 2072–2081. [Google Scholar] [CrossRef] [PubMed]
  17. Shibutani, M.; Maeda, K.; Nagahara, H.; Ohtani, H.; Iseki, Y.; Ikeya, T.; Sugano, K.; Hirakawa, K. The prognostic significance of a postoperative systemic inflammatory response in patients with colorectal cancer. World J. Surg. Oncol. 2015, 13, 194. [Google Scholar] [CrossRef] [Green Version]
  18. Da Silva Alexandre, T.; de Oliveira Duarte, Y.A.; Ferreira Santos, J.L.; Wong, R.; Lebrão, M.L. Sarcopenia according to the european working group on sarcopenia in older people (EWGSOP) versus Dynapenia as a risk factor for disability in the elderly. J. Nutr. Health Aging 2014, 18, 547–553. [Google Scholar] [CrossRef]
  19. Vetrano, D.L.; Landi, F.; Volpato, S.; Corsonello, A.; Meloni, E.; Bernabei, R.; Onder, G. Association of sarcopenia with short- and long-term mortality in older adults admitted to acute care wards: Results from the CRIME study. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 1154–1161. [Google Scholar] [CrossRef] [Green Version]
  20. Elliott, J.A.; Doyle, S.L.; Murphy, C.F.; King, S.; Guinan, E.M.; Beddy, P.; Ravi, N.; Reynolds, J.V. Sarcopenia: Prevalence, and Impact on Operative and Oncologic Outcomes in the Multimodal Management of Locally Advanced Esophageal Cancer. Ann. Surg. 2017, 266, 822–830. [Google Scholar] [CrossRef]
  21. Li, S.; Li, H.; Hu, Y.; Zhu, S.; Xu, Z.; Zhang, Q.; Yang, Y.; Wang, Z.; Xu, J. Ultrasound for Measuring the Cross-Sectional Area of Biceps Brachii Muscle in Sarcopenia. Int. J. Med. Sci. 2020, 17, 2947–2953. [Google Scholar] [CrossRef]
  22. Schwarz, R.E.; Smith, D.D. Extent of lymph node retrieval and pancreatic cancer survival: Information from a large US population database. Ann. Surg. Oncol. 2006, 13, 1189–1200. [Google Scholar] [CrossRef] [PubMed]
  23. Tol, J.A.; Gouma, D.J.; Bassi, C.; Dervenis, C.; Montorsi, M.; Adham, M.; Andrén-Sandberg, A.; Asbun, H.J.; Bockhorn, M.; Büchler, M.W.; et al. Definition of a standard lymphadenectomy in surgery for pancreatic ductal adenocarcinoma: A consensus statement by the International Study Group on Pancreatic Surgery (ISGPS). Surgery 2014, 156, 591–600. [Google Scholar] [CrossRef] [PubMed]
  24. Pedrazzoli, S.; DiCarlo, V.; Dionigi, R.; Mosca, F.; Pederzoli, P.; Pasquali, C.; Klöppel, G.; Dhaene, K.; Michelassi, F. Standard versus extended lymphadenectomy associated with pancreatoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas: A multicenter, prospective, randomized study. Lymphadenectomy Study Group. Ann. Surg. 1998, 228, 508–517. [Google Scholar] [CrossRef] [PubMed]
  25. Worni, M.; Castleberry, A.W.; Clary, B.M.; Gloor, B.; Carvalho, E.; Jacobs, D.O.; Pietrobon, R.; Scarborough, J.E.; White, R.R. Concomitant vascular reconstruction during pancreatectomy for malignant disease: A propensity score-adjusted, population-based trend analysis involving 10,206 patients. JAMA Surg. 2013, 148, 331–338. [Google Scholar] [CrossRef] [Green Version]
  26. Roch, A.M.; House, M.G.; Cioffi, J.; Ceppa, E.P.; Zyromski, N.J.; Nakeeb, A.; Schmidt, C.M. Significance of Portal Vein Invasion and Extent of Invasion in Patients Undergoing Pancreatoduodenectomy for Pancreatic Adenocarcinoma. J. Gastrointest. Surg. 2016, 20, 479–487. [Google Scholar] [CrossRef]
  27. Xie, Z.B.; Li, J.; Gu, J.C.; Jin, C.; Zou, C.F.; Fu, D.L. Pancreatoduodenectomy with portal vein resection favors the survival time of patients with pancreatic ductal adenocarcinoma: A propensity score matching analysis. Oncol. Lett. 2019, 18, 4563–4572. [Google Scholar] [CrossRef] [Green Version]
  28. Ansari, D.; Nilsson, J.; Andersson, R.; Regnér, S.; Tingstedt, B.; Andersson, B. Artificial neural networks predict survival from pancreatic cancer after radical surgery. Am. J. Surg. 2013, 205, 1–7. [Google Scholar] [CrossRef]
  29. Alabi, R.O.; Mäkitie, A.A.; Pirinen, M.; Elmusrati, M.; Leivo, I.; Almangush, A. Comparison of nomogram with machine learning techniques for prediction of overall survival in patients with tongue cancer. Int. J. Med. Inform. 2021, 145, 104313. [Google Scholar] [CrossRef]
  30. Chen, S.; Jiang, L.; Zhang, E.; Hu, S.; Wang, T.; Gao, F.; Zhang, N.; Wang, X.; Zheng, J. A Novel Nomogram Based on Machine Learning-Pathomics Signature and Neutrophil to Lymphocyte Ratio for Survival Prediction of Bladder Cancer Patients. Front. Oncol. 2021, 11, 703033. [Google Scholar] [CrossRef]
Figure 1. The new nomograms predicting recurrence-free survival (RFS) and overall survival (OS) of pancreatic cancer patients after surgery. (A) The nomogram for recurrence-free survival; (B) The nomogram for overall survival.
Figure 1. The new nomograms predicting recurrence-free survival (RFS) and overall survival (OS) of pancreatic cancer patients after surgery. (A) The nomogram for recurrence-free survival; (B) The nomogram for overall survival.
Biomedicines 10 01341 g001
Figure 2. Calibration plots of internal validation for the new nomograms. (A) The plot of the nomogram predicting recurrence-free survival; (B) The plot of the nomogram predicting overall survival.
Figure 2. Calibration plots of internal validation for the new nomograms. (A) The plot of the nomogram predicting recurrence-free survival; (B) The plot of the nomogram predicting overall survival.
Biomedicines 10 01341 g002
Figure 3. The website for calculating survival probability (http://pdacprognosis.smchbp.org, accessed on 30 March 2022). (A) The page for calculating recurrence-free survival; (B) The page for calculating overall survival.
Figure 3. The website for calculating survival probability (http://pdacprognosis.smchbp.org, accessed on 30 March 2022). (A) The page for calculating recurrence-free survival; (B) The page for calculating overall survival.
Biomedicines 10 01341 g003
Table 1. Demographic and clinicopathological data of the development cohort (n = 885).
Table 1. Demographic and clinicopathological data of the development cohort (n = 885).
VariableN (%) or Mean (±SD)VariableN (%) or Mean (±SD)
Age at operation63.1 (±10.3)Operation
Sex, male520 (58.8%)Pancreatoduodenectomy585 (66.1%)
BMI22.9 (±3.1)Left-sided pancreatectomy295 (33.3%)
ASA score Total pancreatectomy5 (0.6%)
I176 (19.9%)Combined PV/SMV resection144 (16.3%)
II632 (71.4%)EBL > 500 mL301 (34.0%)
III77 (8.7%)Intraop. RBC transfusion149 (16.8%)
Underlying disease
Cardiovascular371 (41.9%)Tumor differentiation
Respiratory66 (7.5%)Well85 (9.6%)
Liver41 (4.6%)Moderate521 (58.9%)
CKD6 (0.7%)Poor/Undifferentiated279 (31.5%)
DM347 (39.2%)Tumor size, cm3.14 (±1.3)
Neoadjuvant treatment33 (3.7%)LN metastasis562 (63.5%)
Sarcopenia, yes99 (11.2%)LN ratio0.1 (±0.15)
Sarcopenic obesity, yes285 (32.2%)Tumor necrosis, yes149 (16.8%)
Preop. elevated CA 19–9 a617 (69.7%)R0 resection671 (75.8%)
Preop. serum albumin4.0 (±0.5)
NLR > 2451 (51.0%)Length of stay, days15.1 (±11.9)
PLR > 90739 (83.5%)Major complication, yes d158 (17.9%)
Preop. CHA/SMA abutment b166 (18.8%)Adjuvant chemotherapy508 (57.4%)
Preop. PV/SMV abutment b181 (20.5%)
Preop. PV/SMV invasion c79 (8.9%)
Abbreviations: N, number; SD, standard deviation; BMI, body mass index; ASA, American Society of Anesthesiologists; CKD, chronic kidney disease; DM, diabetes mellitus; Preop., preoperative; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; CHA, common hepatic artery; SMA, superior mesenteric artery; PV, portal vein; SMV, superior mesenteric vein; EBL, estimated blood loss; Intraop., intraoperative; RBC, red blood cell; LN, lymph node. a CA 19–9 ≥ 35 U/mL. b Abutment: abutment or contact of tumor ≤ 180°. c Invasion: contact of tumor > 180° or invasion. d Clavien–Dindo grade ≥ 3.
Table 2. Cox proportional hazard model for recurrence-free survival (n = 885).
Table 2. Cox proportional hazard model for recurrence-free survival (n = 885).
VariableUnivariable AnalysisMultivariable Analysis
HR95% CIpHR95% CIp
Age at operation0.960.89–1.030.261
BMI0.990.97–1.020.560
ASA score (ref. I)
II1.050.86–1.280.634
III0.890.63–1.240.479
Underlying liver disease1.280.89–1.830.186
Underlying CKD1.770.73–4.260.205
Underlying DM1.050.89–1.230.563
Sarcopenia1.030.80–1.320.821
Sarcopenic obesity1.221.03–1.430.0201.221.03–1.440.020
Preop. Albumin0.850.71–1.010.056
Preop. elevated CA 19–9 a1.461.23–1.74<0.0011.281.07–1.530.006
NLR > 21.191.01–1.390.033
PLR > 901.431.14–1.780.0021.341.07–1.680.010
Preop. CHA/SMA abutment b1.301.07–1.580.0071.311.08–1.590.007
Preop. PV/SMV abutment b1.070.88–1.310.475
Preop. PV/SMV invasion c1.441.10–1.890.009
Operation type (ref. PD)
DP0.880.74–1.040.132
TP1.830.76–4.420.181
PV/SMV resection1.220.99–1.510.062
EBL > 500 mL1.551.31–1.82<0.0011.511.28–1.78<0.001
RBC transfusion1.381.13–1.700.002
Major complication (CD ≥ 3)1.050.85–1.290.669
Differentiation (ref. Well)
Moderate1.381.04–1.830.0271.431.07–1.900.015
Poor/Undifferentiated1.861.38–2.50<0.0011.831.35–2.49<0.001
Tumor size > 2 cm1.911.53–2.39<0.0011.671.33–2.09<0.001
LN metastasis1.531.30–1.81<0.001
LN ratio1.161.10–1.22<0.0011.141.08–1.20<0.001
Tumor necrosis1.761.44–2.16<0.0011.561.27–1.93<0.001
R1 resection (including rR1)1.140.95–1.370.165
Abbreviations: HR, hazard ratio; CI, confidence interval; P, p-value; BMI, body mass index; ASA, American Society of Anesthesiologists; CKD, chronic kidney disease; DM, diabetes mellitus; SO, sarcopenic obesity; Preop., preoperative; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; CHA, common hepatic artery; SMA, superior mesenteric artery; PV, portal vein; SMV, superior mesenteric vein; PD, pancreatoduodenectomy; EBL, estimated blood loss; Intraop., intraoperative; RBC, red blood cell; CD, Clavien–Dindo; LN, lymph node; LNR, lymph node ratio. a CA 19–9 ≥ 35 U/mL. b Abutment: abutment or contact of tumor ≤ 180°. c Invasion: contact of tumor > 180° or invasion.
Table 3. Cox proportional hazard model for overall survival (n = 885).
Table 3. Cox proportional hazard model for overall survival (n = 885).
VariableUnivariable AnalysisMultivariable Analysis
HR95% CIpHR95% CIp
Age at operation1.070.99–1.150.0881.161.07–1.25<0.001
BMI0.970.95–1.000.031
ASA score (ref. I)
II1.190.98–1.440.076
III1.000.73–1.370.992
Underlying liver disease1.400.99–1.960.0551.581.12–2.220.009
Underlying CKD2.251.01–5.030.0493.621.58–8.280.002
Underlying DM1.171.01–1.370.040
Sarcopenia1.070.84–1.350.605
Sarcopenic obesity1.060.90–1.240.499
Preop. Albumin0.920.78–1.080.300
Preop. elevated CA 19–9 a1.271.08–1.510.005
NLR > 21.090.94–1.270.244
PLR > 901.180.96–1.450.113
Preop. CHA/SMA abutment b1.201.00–1.450.052
Preop. PV/SMV abutment b1.251.04–1.510.0161.160.95–1.410.152
Preop. PV/SMV invasion c1.991.56–2.56< 0.0011.741.32–2.29<0.001
Operation type (ref. PD)
DP0.870.74–1.020.092
TP0.800.30–2.130.649
PV/SMV resection1.651.36–2.00<0.0011.381.10–1.720.005
EBL > 500 cc1.381.18–1.61<0.0011.361.16–1.59<0.001
RBC transfusion1.090.89–1.330.421
Major complication (CD ≥ 3)1.060.88–1.290.536
Differentiation (ref. Well)
Moderate1.421.08–1.880.0131.341.01–1.780.040
Poor/Undifferentiated2.341.75–3.12<0.0012.201.64–2.96<0.001
Tumor size > 2 cm1.731.40–2.13<0.0011.531.24–1.89<0.001
LN metastasis1.731.47–2.04<0.0011.691.42–1.99<0.001
LN ratio1.101.05–1.15<0.001
Tumor necrosis1.901.57–2.30<0.0011.671.37–2.04<0.001
R1 resection (including rR1)1.201.01–1.430.038
Abbreviations: HR, hazard ratio; CI, confidence interval; P, p-value; BMI, body mass index; ASA, American Society of Anesthesiologists; CKD, chronic kidney disease; DM, diabetes mellitus; SO, sarcopenic obesity; Preop., preoperative; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; CHA, common hepatic artery; SMA, superior mesenteric artery; PV, portal vein; SMV, superior mesenteric vein; PD, pancreatoduodenectomy; EBL, estimated blood loss; Intraop., intraoperative; RBC, red blood cell; CD, Clavien–Dindo; LN, lymph node; LNR, lymph node ratio. a CA 19–9 ≥ 35 U/mL. b Abutment: abutment or contact of tumor ≤ 180°. c Invasion: contact of tumor > 180° or invasion.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yoon, S.J.; Park, B.; Kwon, J.; Lim, C.-S.; Shin, Y.C.; Jung, W.; Shin, S.H.; Heo, J.S.; Han, I.W. Development of Nomograms for Predicting Prognosis of Pancreatic Cancer after Pancreatectomy: A Multicenter Study. Biomedicines 2022, 10, 1341. https://doi.org/10.3390/biomedicines10061341

AMA Style

Yoon SJ, Park B, Kwon J, Lim C-S, Shin YC, Jung W, Shin SH, Heo JS, Han IW. Development of Nomograms for Predicting Prognosis of Pancreatic Cancer after Pancreatectomy: A Multicenter Study. Biomedicines. 2022; 10(6):1341. https://doi.org/10.3390/biomedicines10061341

Chicago/Turabian Style

Yoon, So Jeong, Boram Park, Jaewoo Kwon, Chang-Sup Lim, Yong Chan Shin, Woohyun Jung, Sang Hyun Shin, Jin Seok Heo, and In Woong Han. 2022. "Development of Nomograms for Predicting Prognosis of Pancreatic Cancer after Pancreatectomy: A Multicenter Study" Biomedicines 10, no. 6: 1341. https://doi.org/10.3390/biomedicines10061341

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