Next Article in Journal
Online Adaptive MRI-Guided Radiotherapy for Primary Tumor and Lymph Node Boosting in Rectal Cancer
Next Article in Special Issue
Prognosis of Pancreatic Cancer Based on Resectability: A Single Center Experience
Previous Article in Journal
The Diagnostic and Therapeutic Role of snoRNA and lincRNA in Bladder Cancer
Previous Article in Special Issue
Optimizing First-Line Chemotherapy in Metastatic Pancreatic Cancer: Efficacy of FOLFIRINOX versus Nab-Paclitaxel Plus Gemcitabine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Real-World Data Validation of NAPOLI-1 Nomogram for the Prediction of Overall Survival in Metastatic Pancreatic Cancer

1
National Institute of Cancer Research, National Health Research Institutes, Tainan 704016, Taiwan
2
Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
3
Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704017, Taiwan
4
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
5
Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
6
School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
7
Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan
8
School of Medicine, College of Medicine, China Medical University, Taichung 404328, Taiwan
9
Cancer Center, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan
10
Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
11
Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
12
Department of Oncology, National Taiwan University Hospital, Taipei 100229, Taiwan
13
Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
14
College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
15
Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan
16
Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
17
Department of Surgery, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402306, Taiwan
18
PharmaEngine, Inc., Taipei 104511, Taiwan
19
Division of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan
20
Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
21
Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
22
Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(4), 1008; https://doi.org/10.3390/cancers15041008
Submission received: 30 November 2022 / Revised: 20 January 2023 / Accepted: 31 January 2023 / Published: 5 February 2023
(This article belongs to the Special Issue Advanced Pancreatic Cancer)

Abstract

:

Simple Summary

The nomogram derived from the pivotal phase III NAPOLI-1 study could predict the overall survival in gemcitabine-refractory metastatic pancreatic cancer treated with liposomal irinotecan plus fluorouracil and leucovorin. However, the NAPOLI-1 nomogram has not been validated in a real-world setting and therefore the applicability of the NAPOLI-1 nomogram in daily practice remains unknown. In the current study, we validated the NAPOLI-1 nomogram in a multicenter real-world cohort and confirmed that the NAPOLI-1 nomogram could predict the prognosis of gemcitabine-refractory metastatic pancreatic cancer in daily practice and may help clinical decision making. We further found that the relative dose intensity at 6 weeks was an independent prognostic factor beyond the NAPOLI-1 nomogram, which highlighted the importance of optimal dose delivery regardless of the baseline condition.

Abstract

Background: The nomogram derived from the pivotal phase III NAPOLI-1 study demonstrated a significant ability to predict median overall survival (OS) in gemcitabine-refractory metastatic pancreatic ductal adenocarcinoma (PDAC) treated with liposomal irinotecan plus fluorouracil and leucovorin (nal-IRI+5-FU/LV). However, the NAPOLI-1 nomogram has not been validated in a real-world setting and therefore the applicability of the NAPOLI-1 nomogram in daily practice remains unknown. This study aims to evaluate the NAPOLI-1 nomogram in a multicenter real-world cohort. Methods: The NAPOLI-1 nomogram was applied to a previously established cohort of metastatic PDAC patients treated with nal-IRI+5-FU/LV in nine participating centers in Taiwan. Patients were divided into three risk groups according to the NAPOLI-1 nomogram. The survival impact of relative dose intensity at 6 weeks (RDI at 6 weeks) in different risk groups was also investigated. Results: Of the 473 included patients, the median OSs of patients classified as low (n = 156), medium (n = 186), and high (n = 131) risk were 10.9, 6.3, and 4.3 months, respectively (p < 0.0001). The survival impact of RDI at 6 weeks remained significant after stratification by risk groups, adjustment with Cox regression, inverse probability weighting, or propensity score matching. Conclusions: Our results support the usefulness of the NAPOLI-1 nomogram for risk stratification in gemcitabine-refractory metastatic PDAC treated with nal-IRI+5-FU/LV in daily practice. We further showed that the RDI at 6 weeks is an independent prognostic factor beyond the NAPOLI-1 nomogram.

1. Introduction

Liposomal irinotecan plus 5-fluorouracil/leucovorin (nal-IRI+5-FU/LV) has been shown to improve overall survival (OS) compared to 5-FU/LV alone (median OS 6.1 months vs. 4.2 months, hazard ratio 0.67, p = 0.012) in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) who have failed to respond to gemcitabine-based chemotherapy (NAPOLI-1 Study) [1]. Based on the NAPOLI-1 study, nal-IRI+5-FU/LV was approved by the United States Food and Drug Administration (FDA) in October 2015 and to date, nal-IRI+5-FU/LV remains the only approved regimen in patients with gemcitabine-refractory metastatic PDAC. Several studies using real-world data (RWD) assessing the effectiveness of nal-IRI+5-FU/LV in metastatic PDAC have been reported but their clinical outcomes are barely comparable due to the heterogeneity in study populations and different practice patterns. For example, the proportion of Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≥ 2 ranged from 0 to 27.3% in the published real-world data, while 1.9% to 69.6% of patients had a reduced starting dose and 20.5% to 50% of patients required dose modification [2,3,4,5,6,7,8,9,10,11,12,13,14,15]. Such heterogenicity among studies resulted in heterogeneous treatment outcomes, i.e., an objective response rate of 2.9–19.2%, median progression-free survival (PFS) of 2.0–4.5 months, and median overall survival of 4.3–9.4 months.
A nomogram, usually derived from complicated models, is a user-friendly tool to help physicians quickly and easily understand the prognosis of a patient and aid clinical decision-making [16,17,18]. A well-established nomogram will not only help clinicians with risk stratification and decision-making but may also serve as a comparison benchmark to compare different studies. The NAPOLI-1 study-derived nomogram consisted of eight independent dichotomized parameters identified in multivariate Cox regression for overall survival in the NAPOLI-1 population, including baseline Karnofsky performance score, baseline albumin level, baseline neutrophil–lymphocyte ratio, the presence of liver metastasis, baseline CA19-9, stage IV at diagnosis, body mass index (BMI) and receiving nal-IRI+5-FU/LV combination treatment [1,19]. Based on the NALOPI-1 nomogram, a risk score ranging from 0 to 680 was calculated for each patient. The NAPOLI-1 population was then divided into three risk tertiles, low risk, intermediate risk, and high risk, with cut-off point scores of >370, 260–370, and <260, respectively. The NAPOLI-1 nomogram demonstrated significant discriminatory power to predict overall survival. Patients in high-, intermediate-, and low-risk groups had a median OS of 2.9, 5.3, and 8.5 months, respectively. However, the NAPOLI-1 nomogram was not validated in a real-world setting and therefore the applicability of the NAPOLI-1 nomogram in daily practice remains uncertain.
The per-protocol (PP) analysis of the NAPOLI-1 study defined the PP population as relative dose intensity at 6 weeks (RDI at 6 weeks) ≥ 80% and the median OS of the PP population was significantly better than that of non-PP populations (8.9 vs. 4.4 months, respectively) [20]. Our previous real-world multicenter study further set another cut-point of 60% and divided patients into three groups based on their RDI at 6 weeks: >80%, 60–80%, and <60%. We found that RDI at 6 weeks was an independent prognostic factor of OS even after adjustment of covariates by multivariable Cox regression [21]. In this study, we validate the NAPOLI-1 nomogram in the same large real-world multicenter cohort and further address how RDI at 6 weeks impacts the survival of patients in different risk groups.

2. Materials and Methods

A real-world multicenter cohort of patients with pancreatic cancer treated with nal-IRI+5-FU/LV has been previously described [21]. The inclusion and exclusion criteria of the current study were set to accord with the NAPOLI-1 population. The inclusion criteria included (1) prior use of gemcitabine; (2) presence of metastatic disease while receiving nal-IRI+5-FU/LV; patients with an initial diagnosis of American Joint Committee on Cancer (AJCC) stage I-III are eligible if they developed metastasis after gemcitabine-based treatment; and (3) Karnofsky performance score ≥ 70 (equivalent to ECOG performance score 0–1) while receiving nal-IRI+5-FU/LV. Patients with the following conditions were excluded: (1) using nal-IRI without 5-FU or in combination with S1; (2) receiving nal-IRI+5-FU/LV plus other agents; and (3) receiving nal-IRI+5-FU/LV before the reimbursement in August 2018. This retrospective study was approved by the Institutional Review Board (IRB) with a waiver of informed consent and followed the Declaration of Helsinki. The IRB approval numbers of each participating institute were as follows: Chang Gung Memorial Hospital, 202100783B0; China Medical University Hospital, CMUH109-REC2-176; Chung Shan Medical University Hospital, CS2-21095; National Cheng Kung University Hospital, A-ER-109-477; National Taiwan University Hospital, 201911042RINC; Kaohsiung Medical University Hospital, KMUHIRB-E(I)-20210150; Taipei Veterans General Hospital, 2021-08-001AC; and Tri-Service General Hospital, B202105057.
Patient baseline characteristics, outcomes, and adverse events were extracted from electronic medical records. Tumor response was evaluated by computed tomography (CT) or magnetic resonance imaging (MRI) every 8–12 weeks at the physician’s discretion. Common Terminology Criteria for Adverse Events (CTCAE) version 4.0.3 was used to evaluate the adverse events. The risk score of each patient was calculated from a previously published NAPOLI-1 nomogram consisting of eight dichotomized parameters: baseline Karnofsky performance score ≥ 90 vs. <90 (equivalent to ECOG PS 0 vs. ≥1); baseline albumin ≥ 4 g/dL vs. <4 g/dL; neutrophil–lymphocyte ratio ≤ 5 vs. >5; absence vs. presence of liver metastasis; baseline CA19-9 ≤ 1542 IU/mL vs. >1542 IU/mL; stage I to III vs. stage IV at diagnosis; body mass index (BMI) > 25 kg/m2 vs. ≤25 kg/m2; nal-IRI+5-FU/LV vs. nal-IRI or 5-FU/LV monotherapy (all patients in our study received nal-IRI+5-FU/LV) [19]. Patients were then classified as low (score points > 370), intermediate (score points 260–370) and high risk (score points < 260) according to the original NAPOLI-1 nomogram report.
Descriptive statistics are presented as medians or percentages. The difference in proportions between groups was compared by Fisher’s exact test. The Kolmogorov–Smirnov test was used to check the normality of data distribution.
The reverse Kaplan–Meier method was used to estimate the median duration of follow-up. The definition of PFS was the time between the beginning of nal-IRI+5-FU/LV to the date of either radiological or clinical progression, death, intolerance, loss to follow-up, or data cut-off. OS was defined as the interval between the beginning of nal-IRI+5-FU/LV and death, loss to follow-up, or data cut-off. Intolerance, loss to follow-up, or data cut-off will be censored in PFS data while the loss to follow-up or data cut-off will be censored in OS data. The Kaplan–Meier method was applied for the calculation of PFS and OS while the log-rank test was used for survival comparison between groups.
Three parameters in the NAPOLI-1 nomogram, albumin, neutrophil–lymphocyte ratio, and baseline CA19-9, were not available in some patients and were imputed by using the R multivariate imputation by chained equation (MICE) package [22]. The area under the time-dependent receiver operating characteristic curve (AUC) with inverse probability of censoring weighting (IPCW) technique was estimated to evaluate the prediction accuracy of the NAPOLI-1 nomogram. In terms of investigating the effect of cumulative dose, Cox regression was used to estimate the hazard ratio (HR) and 95% CI with the nomogram score as an adjusted covariate. Propensity score weighting and matching methods were also applied to further balance the distribution of nomogram scores among cumulative dose groups. The inverse probability of weights (IPW) was computed from the generalized propensity scores (GPSs) by using multinomial logistic regression with cumulative dose groups as the outcome variable and the nomogram as the covariate. The 3 groups were matched at 1:1:1 with the same nomogram score. When more than one participant had the same score, patients were randomly selected. We also conducted these statistical analyses on patients without any missing values (n = 264) as a sensitivity analysis. All variables with p < 0.05 were statistically significant. All statistical analyses were performed using R version 4.0.5 (R Core Team, Vienna, Austria) and SAS version 9.4 (SAS Institute Inc., Cary, NC, USA).

3. Results

3.1. Patient Demographics

A total of 696 patients treated with nal-IRI+5-FU/LV were identified from 9 participating centers in Taiwan, as previously described [21]. Among them, 523 patients met the inclusion criteria. After excluding the 50 patients whose treatment had a combination that included oxaliplatin (n = 19), immunotherapy (n = 4) or other agents (n = 3), or whose treatment used S1 instead of 5-FU (n = 3) and those who received nal-IRI before reimbursement in Aug. 2018 (n = 21), finally, 473 patients fulfilled the inclusion and exclusion criteria and were included in the current study (Figure 1).
Using the NAPOLI-1 nomogram for risk classification, 156, 186, and 131 patients were classified as belonging to low, intermediate, and high-risk groups, respectively. As classification parameters, four baseline characteristics in the nomogram including disease stage at diagnosis, baseline albumin, CA 19.9, and liver metastasis were different among the three risk groups (Table 1). Otherwise, all remaining baseline characteristics were comparable between low, intermediate, and high-risk groups.
As of the data cutoff on 31 December 2020, the median duration of follow-up was 13.1 months (interquartile range, IQR 7.0–20.6 months). The median PFS and OS in low-, intermediate-, and high-risk groups were significantly different. The median PFSs in the corresponding risk groups were 4.8 (95% CI, 3.5–6.8), 3.2 (95% CI, 2.7–4.7), and 2.3 (95% CI, 2.0–2.8) months, respectively (log-rank p < 0.0001, Figure 2A); while the median OSs were 10.9 (95% CI, 9.3–12.5), 6.3 (95% CI, 5.7–7.3) and 4.3 (95% CI, 3.8–5.5) months, respectively (log-rank p < 0.0001, Figure 2B).
In the sensitivity analysis which excluded 209 patients with missing values in the nomogram, the distribution of NAPOLI-1 nomogram risk classification, median PFS, and median OS of the remaining 264 patients without missing value imputation were similar to those of the entire cohort (Supplementary Figure S1).

3.2. Model Performance

In the current study, the AUCs of the NAPOLI-1 nomogram for median OS prediction range from 0.698 to 0.738 (Figure 3A,C). Most of the parameters used in the NAPOLI-1 nomogram remain significant in the multivariable analysis for the current RWD population (Supplementary Table S1). Another multivariable analysis was performed to further explore whether any factor other than the NAPOLI-1 nomogram had a significant impact on survival by selecting a covariate with a p-value < 0.05 in univariate analysis. RDI at 6 weeks was found to be the most significant factor beyond the NAPOLI-1 nomogram (Supplementary Table S2). Adding RDI at 6 weeks to the NAPOLI-1 nomogram improved the AUC mainly at 3 months and 6 months, which reached values of 0.822 and 0.731, respectively (Figure 3B,D). In the sensitivity analysis, the AUC of 264 patients without missing value imputation was similar to that of the entire cohort (Supplementary Figure S2).

3.3. Relative Dose Intensity at 6 Weeks Is an Independent Prognostic Factor

The distribution of cumulative dose during the first 6 weeks was similar among different risk groups (Figure 4A). The distribution of nomogram risk scores was similar among the three dose intensity groups (Figure 4B). Linear regression demonstrated no statistically significant correlation between nomogram risk score and 6-week cumulative dose (Spearman’s correlation coefficient = 0.041, p = 0.37) (Figure 4C). All results were consistent in that RDI at 6 weeks was independent of the baseline condition.
The survival of patients with different RDI at 6 weeks was significantly different across three NAPOLI-1 nomogram risk groups (Figure 5A–C). The survival difference remained significant after adjustment with nomogram risk score, age, and gender by Cox regression analysis (<60% vs. >80%, HR 1.975, 95% CI 1.461–2.670) (Table 2). The survival difference among patients with different RDI at 6 weeks remained significant in the inverse probability of weighting survival analysis (<60% vs. >80%, HR 1.800, 95% CI 1.547–2.095, Figure 5D) or after 1:1:1 three-group propensity score matching by nomogram risk score (<60% vs. >80%, HR 3.091, 95% CI 1.643–5.815, Figure 5E).
In the sensitivity analysis, the dose distribution, survival difference before and after covariate adjustment, stratification, weighting, and matching in the 264 patients without missing value imputation were quite similar to those of the entire cohort (Supplementary Figures S3 and S4, Table S3).

3.4. Real-World Safety Profile

The most common ≥ grade 3 hematological toxicities were neutropenia (23.3%) and anemia (19.5%); while the most common non-hematological toxicities were hypokalemia (12.7%) and increased bilirubin (7.4%) (Table 3). There was no significant difference in safety profiles among the three different risk groups.

4. Discussion

Our study is the first study to evaluate and validate the NAPOLI-1 nomogram in a real-world setting and support its use as a risk stratification tool, which will not only help physicians and patients to make decisions but can also be implemented in the comparison of results among parallel studies, including real-world data and clinical trials.
In the per-protocol (PP) analysis of the NAPOLI-1 study, patients in PP populations (defined as RDI at 6 weeks ≥ 80%) had significantly better median OS as compared to non-PP populations (8.9 vs. 4.4 months, respectively) [20]. One may argue that the longer survival of patients with higher RDI at 6 weeks might result from better baseline condition as reflected by the higher percentage of patients with Karnofsky performance score ≥ 90 in PP than in non-PP populations (62.1% and 49%, respectively). In our previous study, we showed that RDI at 6 weeks remained an important prognostic factor after confounder adjustment [21]. In the current study, there was no significant correlation between NAPOLI-1 nomogram risk score, an indicator of baseline condition, and RDI at 6 weeks (Figure 3C). In addition, RDI at 6 weeks significantly impacted the survival of different risk groups (Figure 5A–C), even after weighting (Figure 4D), matching (Figure 4E), and Cox regression adjustment (Table 2). Through different methodologies, the results highlighted the importance of dose delivery regardless of the baseline condition.
In the post hoc analysis of the NAPOLI-1 study, the median OS of Asian patients receiving nal-IRI+5-FU/LV was 8.9 months compared to the 6.1 months of the entire nal-IRI+5-FU/LV-treated population [23]. Of note, the median OS of patients with a low and intermediate risk score in the current study was numerically better than that of patients with nal-IRI+5-FU/LV in the NAPOLI-1 study (10.9 vs. 9.0 months and 6.3 vs. 5.4 months, respectively), but similar survival for patients with a high risk score (4.3 vs. 4.3 months) [19]. In a recent population pharmacokinetic study, Asian patients had a significantly higher average concentration of un-encapsulated SN38 (uSN38 Cavg), a pharmacokinetic parameter associated with prolonged OS, compared to Caucasians in the nal-IRI+5-FU/LV arm of the NAPOLI-1 study [24]. Interestingly, the uSN38 concentration was similar in Caucasian patients with and without UGT1A1*28/*28 homozygosity, but significantly higher in Asian patients with UGT1A1*6/*6 homozygosity or UGT1A1*6/*28 compound heterozygosity than in those with UGT1A1*6 single heterozygosity or wild-type individuals [24,25]. Since the allele frequency of UGT1A1*6 only ranged from 15–30% with UGT1A1*6 homozygosity or compound heterozygosity in 9–10% of the Asian population, the mechanisms for the ethnic differences in nal-IRI pharmacokinetics and survival after nal-IRI+5-FU/LV are likely multifactorial and warrant further investigation.
This study has some limitations. First, three parameters in the nomogram, baseline albumin, neutrophil–lymphocyte ratio, and baseline CA 19-9 level were not available in all patients and imputed by the MICE procedure. The handling of missing values is a huge discipline and each method had its advantages and disadvantages. Originally proposed by Donald Rubin in 1987, the multiple imputation procedure has been greatly improved and become more widely used even in clinical trials published in The Lancet and The New England Journal of Medicine [26,27,28]. However, the results from the sensitivity analyses of the 264 patients without missing values were consistent with all study results, which justifies the imputation procedure. Second, in the original NAPOLI-1 nomogram, nal-IRI+5-FU/LV treatment was one of the eight dichotomized parameters. The inclusion of such a parameter can be redundant if the goal of the study was intra-study risk categorization for a patient cohort with homogenous treatment. On the other hand, it is reasonable to do so if inter-studies survival outcome comparison is the main theme, such as RWD versus clinical trial results in this study. Third, although all baseline characteristics other than ethnicity were comparable between the current study and NAPOLI-1, the practice pattern was very different in that more patients in our cohort had prior exposure to fluorouracil-containing treatment than did participants in NAPOLI-1 (77.4% vs. 42.7%, respectively). Due to the delayed reimbursement of nab-paclitaxel and FOLFIRINOX, the gemcitabine/fluorouracil-based triplet, including the GOFL regimen (gemcitabine, oxaliplatin, 5-FU, and leucovorin) and SLOG regimen (S1, leucovorin, oxaliplatin, and gemcitabine), were among the most commonly used first-line regimens for advanced pancreatic cancer in Taiwan before 2020 [29,30,31,32,33,34]. Although the current study suggests previous more aggressive gemcitabine-based treatments, such as GOFL and SLOG, might not impact the therapeutic efficacy of second-line nal-IRI+5-FU/LV, the issue still deserves further exploration with NAPOLI-1 nomogram risk group-stratified comparison.

5. Conclusions

In the current study, we employed several strategies including stratification, multivariable adjustment, weighting, and matching methods. The results were consistent among these methods, which indicates the robustness of our findings in using the NAPOLI-1 nomogram as a measurable confounding factor. Our study not only validated the NAPOLI-1 nomogram for risk stratification in a real-world setting but also identified RDI at 6 weeks as an independent prognostic factor beyond the NAPOLI-1 nomogram.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15041008/s1, Figure S1: Sensitivity analysis of survival in patients without missing values in the NAPOLI-1 nomogram (n = 264); Figure S2: Sensitivity analysis of model performance in patients without missing values in the NAPOLI-1 nomogram (n = 264); Figure S3: Sensitivity analysis of dose distribution in patients without missing values in the NAPOLI-1 nomogram (n = 264); Figure S4: Sensitivity analysis of survival in patients without missing values in the NAPOLI-1 nomogram (n = 264). Table S1: Multivariable Cox regression of factors in the NAPOLI-1 nomogram (n = 473); Table S2: Multivariable Cox regression of other covariates with NAPOLI-1 nomogram risk score (n =473); Table S3: Model in patients without missing values (n = 264).

Author Contributions

Conceptualization, N.-J.C. and Y.-S.S.; methodology, Y.-H.Y.; software, Y.-Y.S. and Y.-H.Y.; validation, N.-J.C., C.-J.Y., C.-F.C., J.-S.C. and Y.-Y.C.; formal analysis, Y.-H.Y.; resources, C.-J.Y., C.-F.C., J.-S.C., Y.-Y.C. and S.-C.C. (Sz-Chi Chiu); data curation, L.-Y.B., S.-C.C. (Shih-Chang Chuang), S.-H.Y., W.-C.C., T.-J.C., D.-C.C. and C.-M.P.; writing—original draft preparation, Y.-Y.S., N.-J.C. and Y.-H.Y.; writing—review and editing, C.-P.L., Y.-S.S. and L.-T.C.; project administration, Y.-S.S. and L.-T.C. 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 in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of Chang Gung Memorial Hospital, 202100783B0; China Medical University Hospital, CMUH109-REC2-176; Chung Shan Medical University Hospital, CS2-21095; National Cheng Kung University Hospital, A-ER-109-477; National Taiwan University Hospital, 201911042RINC; Kaohsiung Medical University Hospital, KMUHIRB-E(I)-20210150; Taipei Veterans General Hospital, 2021-08-001AC; and Tri-Service General Hospital, B202105057.

Informed Consent Statement

Patient consent was waived due to the retrospective nature of the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

Sz-Chi Chiu is employed by PharmaEngine, Inc. The remaining authors declare no conflict of interest.

References

  1. Wang-Gillam, A.; Li, C.-P.; Bodoky, G.; Dean, A.; Shan, Y.-S.; Jameson, G.; Macarulla, T.; Lee, K.-H.; Cunningham, D.; Blanc, J.F.; et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): A global, randomised, open-label, phase 3 trial. Lancet 2016, 387, 545–557. [Google Scholar] [CrossRef] [PubMed]
  2. Miki, M.; Fujimori, N.; Ueda, K.; Lee, L.; Murakami, M.; Takamatsu, Y.; Shimokawa, Y.; Niina, Y.; Oono, T.; Hisano, T.; et al. Treatment Effect and Safety of Nanoliposomal Irinotecan with Fluorouracil and Folinic Acid after Gemcitabine-Based Therapy in Patients with Advanced Pancreatic Cancer: A Multicenter, Prospective Observational Study. J. Clin. Med. 2022, 11, 5084. [Google Scholar] [CrossRef] [PubMed]
  3. Glassman, D.C.; Palmaira, R.L.; Covington, C.M.; Desai, A.M.; Ku, G.Y.; Li, J.; Harding, J.J.; Varghese, A.M.; O’Reilly, E.M.; Yu, K.H. Nanoliposomal irinotecan with fluorouracil for the treatment of advanced pancreatic cancer, a single institution experience. BMC Cancer 2018, 18, 693. [Google Scholar] [CrossRef] [PubMed]
  4. Kieler, M.; Unseld, M.; Bianconi, D.; Scheithauer, W.; Prager, G.W. A real-world analysis of second-line treatment options in pancreatic cancer: Liposomal-irinotecan plus 5-fluorouracil and folinic acid. Ther. Adv. Med. Oncol. 2019, 11, 1758835919853196. [Google Scholar] [CrossRef]
  5. Yoo, C.; Im, H.-S.; Kim, K.-P.; Oh, D.-Y.; Lee, K.-H.; Chon, H.J.; Kim, J.H.; Kang, M.; Kim, I.; Lee, G.J.; et al. Real-world efficacy and safety of liposomal irinotecan plus fluorouracil/leucovorin in patients with metastatic pancreatic adenocarcinoma: A study by the Korean Cancer Study Group. Ther. Adv. Med. Oncol. 2019, 11, 1758835919871126. [Google Scholar] [CrossRef]
  6. Tossey, J.C.; Reardon, J.; VanDeusen, J.B.; Noonan, A.M.; Porter, K.; Arango, M.J. Comparison of conventional versus liposomal irinotecan in combination with fluorouracil for advanced pancreatic cancer: A single-institution experience. Med. Oncol. 2019, 36, 87. [Google Scholar] [CrossRef]
  7. Barzi, A.; Miksad, R.; Surinach, A.; Corvino, F.A.; Wang, S.; Torres, A.Z.; Mamlouk, K.; Pulgar, S.; Valderrama, A.; Bekaii-Saab, T.; et al. Real-World Dosing Patterns and Outcomes of Patients With Metastatic Pancreatic Cancer Treated With a Liposomal Irinotecan Regimen in the United States. Pancreas 2020, 49, 193–200. [Google Scholar] [CrossRef]
  8. Su, Y.-Y.; Chiang, N.-J.; Tsai, H.-J.; Yen, C.-J.; Shan, Y.-S.; Chen, L.-T. The Impact of Liposomal Irinotecan on the Treatment of Advanced Pancreatic Adenocarcinoma: Real-World Experience in a Taiwanese Cohort. Sci. Rep. 2020, 10, 7420. [Google Scholar] [CrossRef]
  9. Kim, G.P.; Surinach, A.; Corvino, F.A.; Cockrum, P.; Belanger, B.; Abushahin, L. Real-world outcomes associated with liposomal irinotecan dose reductions in metastatic pancreatic ductal adenocarcinoma. Futur. Oncol. 2021, 17, 675–688. [Google Scholar] [CrossRef]
  10. Kasi, A.; McGinnis, T.; Naik, G.; Handa, S.; Williams, G.; Paluri, R. Efficacy and tolerability of the combination of nano-liposomal irinotecan and 5-fluorouracil/leucovorin in advanced pancreatic adenocarcinoma: Post-approval clinic experience. J. Gastrointest. Oncol. 2021, 12, 464–473. [Google Scholar] [CrossRef]
  11. Park, H.; Kang, B.; Chon, H.; Im, H.-S.; Lee, C.-K.; Kim, I.; Kang, M.; Hwang, J.; Bae, W.; Cheon, J.; et al. Liposomal irinotecan plus fluorouracil/leucovorin versus FOLFIRINOX as the second-line chemotherapy for patients with metastatic pancreatic cancer: A multicenter retrospective study of the Korean Cancer Study Group (KCSG). ESMO Open 2021, 6, 100049. [Google Scholar] [CrossRef] [PubMed]
  12. Bang, K.; Cheon, J.; Jeong, J.H.; Im, H.-S.; Kim, K.-P.; Ryoo, B.-Y.; Yoo, C. Clinical outcomes of liposomal irinotecan plus fluorouracil/leucovorin for metastatic pancreatic adenocarcinoma in patients previously treated with conventional irinotecan-containing chemotherapy. Ther. Adv. Med. Oncol. 2021, 13, 17588359211003053. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, K.H.; Hendifar, A.E.; Alese, O.B.; Draper, A.; Abdelrahim, M.; Burns, E.; Khan, G.; Cockrum, P.; Bhak, R.H.; Nguyen, C.; et al. Clinical Outcomes Among Patients With Metastatic Pancreatic Ductal Adenocarcinoma Treated With Liposomal Irinotecan. Front. Oncol. 2021, 11, 678070. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, S.-H.; Chiang, N.-J.; Chiu, S.-C.; Chou, W.-C.; Bai, L.-Y.; Li, C.-P.; Su, Y.-Y.; Chiu, T.-J.; Chuang, S.-C.; Peng, C.-M.; et al. The impact of spleen volume on the survival of metastatic pancreatic adenocarcinoma patients receiving nanoliposomal irinotecan. Am. J. Cancer Res. 2022, 12, 1884–1898. [Google Scholar]
  15. Yu, H.-Y.; Lee, C.-Y.; Lin, L.-G.; Chao, Y.; Li, C.-P. Nanoliposomal irinotecan with 5-fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy: A real-world experience. J. Chin. Med. Assoc. 2022, 85, 42–50. [Google Scholar] [CrossRef]
  16. Iasonos, A.; Schrag, D.; Raj, G.V.; Panageas, K.S. How to build and interpret a nomogram for cancer prognosis. J. Clin. Oncol. 2008, 26, 1364–1370. [Google Scholar] [CrossRef]
  17. Balachandran, V.P.; Gonen, M.; Smith, J.J.; DeMatteo, R.P. Nomograms in oncology: More than meets the eye. Lancet Oncol. 2015, 16, e173–e180. [Google Scholar] [CrossRef]
  18. Hsu, C.; Liu, K.; Chang, P.; Chen, P.; Hung, C.; Hsueh, S.; Yeh, K.; Chen, Y.; Lu, C.; Hung, Y.; et al. Development and validation of a prognostic nomogram to predict survival in patients with advanced pancreatic cancer receiving second-line palliative chemotherapy. J. Gastroenterol. Hepatol. 2020, 35, 1694–1703. [Google Scholar] [CrossRef]
  19. Chen, L.-T.; Macarulla, T.; Blanc, J.-F.; Mirakhur, B.; de Jong, F.A.; Belanger, B.; Bekaii-Saab, T.; Siveke, J.T. Nomogram for Predicting Survival in Patients Treated with Liposomal Irinotecan Plus Fluorouracil and Leucovorin in Metastatic Pancreatic Cancer. Cancers 2019, 11, 1068. [Google Scholar] [CrossRef]
  20. Chen, L.-T.; Siveke, J.T.; Wang-Gillam, A.; Li, C.-P.; Bodoky, G.; Dean, A.P.; Shan, Y.-S.; Jameson, G.S.; Macarulla, T.; Lee, K.-H.; et al. Survival with nal-IRI (liposomal irinotecan) plus 5-fluorouracil and leucovorin versus 5-fluorouracil and leucovorin in per-protocol and non-per-protocol populations of NAPOLI-1: Expanded analysis of a global phase 3 trial. Eur. J. Cancer 2018, 105, 71–78. [Google Scholar] [CrossRef] [PubMed]
  21. Su, Y.-Y.; Chiang, N.-J.; Li, C.-P.; Yen, C.-J.; Yang, S.-H.; Chou, W.-C.; Chen, J.-S.; Chiu, T.-J.; Chen, Y.-Y.; Chuang, S.-C.; et al. Dosing Pattern and Early Cumulative Dose of Liposomal Irinotecan in Metastatic Pancreatic Cancer: A Real-World Multicenter Study. Front. Oncol. 2022, 12, 800842. [Google Scholar] [CrossRef] [PubMed]
  22. van Buuren, S.; Groothuis-Oudshoorn, K. Mice: Multivariate Imputation by Chained Equations in R. J. Stat. Softw. 2011, 45, 1–67. [Google Scholar] [CrossRef]
  23. Bang, Y.; Li, C.; Lee, K.; Chiu, C.; Park, J.O.; Shan, Y.; Kim, J.S.; Chen, J.; Shim, H.; Rau, K.; et al. Liposomal irinotecan in metastatic pancreatic adenocarcinoma in Asian patients: Subgroup analysis of the NAPOLI-1 study. Cancer Sci. 2020, 111, 513–527. [Google Scholar] [CrossRef]
  24. Adiwijaya, B.; Kim, J.; Lang, I.; Csõszi, T.; Cubillo, A.; Chen, J.-S.; Wong, M.; Park, J.; Rau, K.; Melichar, B.; et al. Population Pharmacokinetics of Liposomal Irinotecan in Patients With Cancer. Clin. Pharmacol. Ther. 2017, 102, 997–1005. [Google Scholar] [CrossRef]
  25. Su, Y.-Y.; Chiang, N.-J.; Chang, J.; Wang, Y.-W.; Shen, B.-N.; Li, Y.-J.; Hwang, D.-Y.; Shan, Y.-S.; Chen, L.-T. The association between UGT1A1 polymorphisms and treatment toxicities of liposomal irinotecan. ESMO Open 2022, 8, 100746. [Google Scholar] [CrossRef] [PubMed]
  26. Rubin, D.B. Multiple Imputation After 18+ Years. J. Am. Stat. Assoc. 1996, 91, 473–489. [Google Scholar] [CrossRef]
  27. Rezvan, P.H.; Lee, K.J.; Simpson, J.A. The rise of multiple imputation: A review of the reporting and implementation of the method in medical research. BMC Med. Res. Methodol. 2015, 15, 30. [Google Scholar] [CrossRef]
  28. Tan, P.-T.; Cro, S.; Van Vogt, E.; Szigeti, M.; Cornelius, V.R. A review of the use of controlled multiple imputation in randomised controlled trials with missing outcome data. BMC Med. Res. Methodol. 2021, 21, 72. [Google Scholar] [CrossRef]
  29. Ch’Ang, H.-J.; Wang, C.-C.; Cheng, A.-L.; Hsu, C.; Lu, Y.-S.; Chang, M.-C.; Lin, J.-T.; Wang, H.-P.; Shiah, H.-S.; Liu, T.-W.; et al. Phase I study of biweekly gemcitabine followed by oxaliplatin and simplified 48-h infusion of fluorouracil/leucovorin for advanced pancreatic cancer. J. Gastroenterol. Hepatol. 2006, 21, 874–879. [Google Scholar] [CrossRef]
  30. Ch’Ang, H.-J.; Huang, C.-L.; Wang, H.-P.; Shiah, H.-S.; Chang, M.-C.; Jan, C.-M.; Chen, J.-S.; Tien, Y.-W.; Lin, J.-T.; Cheng, A.-L.; et al. Phase II study of biweekly gemcitabine followed by oxaliplatin and simplified 48-h infusion of 5-fluorouracil/leucovorin (GOFL) in advanced pancreatic cancer. Cancer Chemother. Pharmacol. 2009, 64, 1173–1179. [Google Scholar] [CrossRef] [PubMed]
  31. Chiang, N.-J.; Tsai, K.K.; Hsiao, C.-F.; Yang, S.-H.; Hsiao, H.-H.; Shen, W.-C.; Hsu, C.; Lin, Y.-L.; Chen, J.-S.; Shan, Y.-S.; et al. A multicenter, phase I/II trial of biweekly S-1, leucovorin, oxaliplatin and gemcitabine in metastatic pancreatic adenocarcinoma-TCOG T1211 study. Eur. J. Cancer 2020, 124, 123–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Su, Y.-Y.; Chiu, Y.-F.; Li, C.-P.; Yang, S.-H.; Lin, J.; Lin, S.-J.; Chang, P.-Y.; Chiang, N.-J.; Shan, Y.-S.; Ch’Ang, H.-J.; et al. A phase II randomised trial of induction chemotherapy followed by concurrent chemoradiotherapy in locally advanced pancreatic cancer: The Taiwan Cooperative Oncology Group T2212 study. Br. J. Cancer 2022, 126, 1018–1026. [Google Scholar] [CrossRef] [PubMed]
  33. Chiang, N.-J.; Shan, Y.-S.; Bai, L.-Y.; Li, C.-P.; Chen, J.-S.; Yang, S.-H.; Kuo, Y.-C.; Chao, Y.; Hsieh, Y.-Y.; Kao, H.-F.; et al. TCOG T5217 trial: A phase II randomized study of SLOG versus modified FOLFIRINOX as the first-line treatment in locally advanced or metastatic pancreatic ductal adenocarcinoma. J. Clin. Oncol. 2021, 39, 4143. [Google Scholar] [CrossRef]
  34. Su, Y.-Y.; Ting, Y.-L.; Wang, C.-J.; Chao, Y.-J.; Liao, T.-K.; Su, P.-J.; Chiang, N.-J.; Liao, I.-C.; Yu, Y.-T.; Liu, Y.-S.; et al. Improved survival with induction chemotherapy and conversion surgery in locally advanced unresectable pancreatic cancer: A single institution experience. Am. J. Cancer Res. 2022, 12, 2189–2202. [Google Scholar]
Figure 1. Study schema.
Figure 1. Study schema.
Cancers 15 01008 g001
Figure 2. Survival analysis. Kaplan–Meier plot of progression-free survival (A) and overall survival (B) of the entire cohort and different risk groups.
Figure 2. Survival analysis. Kaplan–Meier plot of progression-free survival (A) and overall survival (B) of the entire cohort and different risk groups.
Cancers 15 01008 g002
Figure 3. Model Performance. The area under the receiver operating characteristic curve (AUC) at 3, 6, 9, 12 and 15 months of the NAPOLI-1 nomogram (A) and of the NAPOLI-1 nomogram with relative dose intensity at 6 weeks (B). Time-dependent AUC of the NAPOLI-1 nomogram (C) and of the NAPOLI-1 nomogram with relative dose intensity at 6 weeks (D).
Figure 3. Model Performance. The area under the receiver operating characteristic curve (AUC) at 3, 6, 9, 12 and 15 months of the NAPOLI-1 nomogram (A) and of the NAPOLI-1 nomogram with relative dose intensity at 6 weeks (B). Time-dependent AUC of the NAPOLI-1 nomogram (C) and of the NAPOLI-1 nomogram with relative dose intensity at 6 weeks (D).
Cancers 15 01008 g003
Figure 4. Dose distribution. (A) Distribution of cumulative dose at 6 weeks in different NAPOLI-1 nomogram risk groups. (B) Distribution of NAPOLI-1 nomogram risk score in different groups of relative dose intensity at 6 weeks. (C) Spearman correlation of nomogram risk score and cumulative dose at 6 weeks. Each dot represents one patient.
Figure 4. Dose distribution. (A) Distribution of cumulative dose at 6 weeks in different NAPOLI-1 nomogram risk groups. (B) Distribution of NAPOLI-1 nomogram risk score in different groups of relative dose intensity at 6 weeks. (C) Spearman correlation of nomogram risk score and cumulative dose at 6 weeks. Each dot represents one patient.
Cancers 15 01008 g004
Figure 5. Overall survival. Kaplan–Meier plot in low-risk group (A), medium-risk group (B), and high-risk group (C). (D) Overall survival weighted by inverse probability of weights (IPW) in different cumulative dose groups. (E) Overall survival after 1:1:1 three-group matching by NAPOLI-1 nomogram risk score in different dose intensity groups (n = 53 in each group).
Figure 5. Overall survival. Kaplan–Meier plot in low-risk group (A), medium-risk group (B), and high-risk group (C). (D) Overall survival weighted by inverse probability of weights (IPW) in different cumulative dose groups. (E) Overall survival after 1:1:1 three-group matching by NAPOLI-1 nomogram risk score in different dose intensity groups (n = 53 in each group).
Cancers 15 01008 g005
Table 1. Baseline characteristics in different risk groups.
Table 1. Baseline characteristics in different risk groups.
Risk GroupLowIntermediateHighOverall
(n = 156)(n = 186)(n = 131)(n = 473)
Gender
Female67 (42.9%)83 (44.6%)48 (36.6%)198 (41.9%)
Male89 (57.1%)103 (55.4%)83 (63.4%)275 (58.1%)
Age, median (range)62.5 (27–82)63.0 (34–86)63 (33–86)63 (27–86)
Disease stage at diagnosis
Stage I-III108 (69.2%)40 (21.5%)6 (4.6%)154 (32.6%)
Stage IV48 (30.8%)146 (78.5%)125 (95.4%)319 (67.4%)
Primary tumor location
Head90 (57.7%)95 (51.1%)64 (48.9%)249 (52.6%)
Body34 (21.8%)52 (28.0%)22 (16.8%)108 (22.8%)
Tail24 (15.4%)32 (17.2%)37 (28.2%)93 (19.7%)
Body + Tail7 (4.5%)6 (3.2%)7 (5.3%)20 (4.2%)
Head + Body or Tail1 (0.6%)1 (0.5%)1 (0.8%)3 (0.6%)
Albumin
<445 (28.8%)69 (37.1%)65 (49.6%)179 (37.8%)
≥455 (35.3%)37 (19.9%)22 (16.8%)114 (24.1%)
Not checked56 (35.9%)80 (43.0%)44 (33.6%)180 (38.1%)
Number of metastatic sites
1117 (75.0%)90 (48.4%)47 (35.9%)254 (53.7%)
234 (21.8%)66 (35.5%)41 (31.3%)141 (29.8%)
35 (3.2%)27 (14.5%)33 (25.2%)65 (13.7%)
≥40 (0%)3 (1.6%)10 (7.6%)13 (2.7%)
Site of metastasis
Liver69 (44.2%)125 (67.2%)125 (95.4%)319 (67.4%)
Lung35 (22.4%)47 (25.3%)40 (30.5%)122 (25.8%)
Peritoneum38 (24.4%)55 (29.6%)37 (28.2%)130 (27.5%)
CA-19.9
<40 U/mL37 (23.7%)26 (14.0%)6 (4.6%)69 (14.6%)
≥40 U/mL109 (69.9%)138 (74.2%)99 (75.6%)346 (73.2%)
Not checked10 (6.4%)22 (11.8%)26 (19.8%)58 (12.3%)
Prior treatment
Gemcitabine-containing156 (100%)186 (100%)131 (100%)473 (100%)
Fluorouracil-containing112 (71.8%)150 (80.6%)104 (79.4%)366 (77.4%)
Irinotecan-containing16 (10.3%)28 (15.1%)20 (15.3%)64 (13.5%)
Platinum-containing59 (37.8%)86 (46.2%)73 (55.7%)218 (46.1%)
Taxane-containing47 (30.1%)56 (30.1%)41 (31.3%)144 (30.4%)
Prior lines of systemic treatment †
02 (1.3%)5 (2.7%)0 (0%)7 (1.5%)
1101 (64.7%)110 (59.1%)84 (64.1%)295 (62.4%)
≥253 (34.0%)71 (38.2%)47 (35.9%)171 (36.2%)
Operation history
No surgery74 (47.4%)119 (64.0%)88 (67.2%)281 (59.4%)
Whipple operation26 (16.7%)34 (18.3%)17 (13.0%)77 (16.3%)
Distal pancreatectomy20 (12.8%)13 (7.0%)9 (6.9%)42 (8.9%)
Total pancreatectomy3 (1.9%)4 (2.2%)3 (2.3%)10 (2.1%)
Other procedures33 (21.2%)16 (8.6%)14 (10.7%)63 (13.3%)
Interval between the last therapy and nal-IRI+5-FU/LV
Median (IQR)0.754 (0.475–1.28)0.672 (0.459–1.08)0.689 (0.459–1.11)0.689 (0.459–1.15)
Not recorded21 (13.5%)28 (15.1%)16 (12.2%)65 (13.7%)
IQR: interquartile range; † 0 indicated only adjuvant gemcitabine without other systemic treatment before nal-IRI+5-FU/LV.
Table 2. NAPOLI-1 nomogram and multivariable Cox regression in the overall population (n = 473).
Table 2. NAPOLI-1 nomogram and multivariable Cox regression in the overall population (n = 473).
Model 1
NAPOLI-1 Nomogram
Model 2
NAPOLI-1 Nomogram and Cumulative Dose
ParameterHR (95%CI)p-ValueParameterHR (95%CI)p-Value
Nomogram risk score0.996 (0.995–0.997)<0.0001Nomogram risk score0.996 (0.995–0.997)<0.0001
Age1.009 (0.996–1.023)0.1607RDI6-week > 80%Reference-
Gender: male1.406 (1.118–1.766)0.003460–80%1.050 (0.777–1.419)0.9148
<60%1.975 (1.461–2.670)<0.0001
Age1.008 (0.994–1.021)0.2786
Gender: male1.287 (1.022–1.620)0.0317
RDI: relative dose intensity.
Table 3. Adverse effects in different NAPOLI-1 nomogram risk groups.
Table 3. Adverse effects in different NAPOLI-1 nomogram risk groups.
Risk GroupLowIntermediate
(n = 186)
HighOverall
(n = 156)(n = 131)(n = 473)
Neutropenia
All-grade69 (44.2%)76 (40.9%)52 (39.7%)197 (41.6%)
≥grade 336 (23.1%)44 (23.7%)30 (22.9%)110 (23.3%)
Febrile neutropenia4 (2.6%)7 (3.8%)5 (3.8%)16 (3.4%)
Not recorded4 (2.1%)2 (1.2%)3 (2.4%)9 (1.9%)
Anemia
All-grade83 (53.2%)121 (65.1%)97 (74.0%)301(63.6%)
≥grade 323 (14.7%)37 (19.9%)32 (24.4%)92 (19.5%)
Not recorded2 (1.1%)2 (1.2%)2 (1.6%)6 (1.3%)
Thrombocytopenia
All-grade26 (16.7%)47 (25.3%)42 (32.1%)115 (24.3%)
≥grade 35 (3.2%)10 (5.4%)9 (6.9%)24 (5.1%)
Not recorded3 (1.6%)1 (0.6%)2 (1.6%)6 (1.3%)
AST or ALT increased
All-grade43 (27.6%)61 (32.8%)40 (30.5%)144 (30.4%)
≥grade 36 (3.8%)7 (3.8%)0 (0%)13 (2.7%)
Not recorded40 (21.4%)44 (27.2%)33 (26.6%)117 (24.7%)
Blood bilirubin increased
All-grade20 (12.8%)47 (25.3%)35 (26.7%)102 (21.6%)
≥grade 310 (6.4%)15 (8.1%)10 (7.6%)35 (7.4%)
Not recorded23 (12.3%)8 (4.9%)9 (7.3%)40 (8.5%)
Creatinine increased
All-grade28 (17.9%)30 (16.1%)22 (16.8%)80 (16.9%)
≥grade 30 (0%)2 (1.1%)1 (0.8%)3 (0.6%)
Not recorded16 (8.6%)3 (1.9%)6 (4.8%)25 (5.3%)
Hypokalemia
All-grade44 (28.2%)61 (32.8%)47 (35.9%)152 (32.1%)
≥grade 321 (13.5%)21 (11.3%)18 (13.7%)60 (12.7%)
Not recorded42 (22.5%)34 (21.0%)29 (23.4%)105 (22.2%)
Fatigue
All-grade64 (41.0%)84 (45.2%)63 (48.1%)211 (44.6%)
≥ grade 31 (0.6%)5 (2.7%)2 (1.5%)8 (1.7%)
Not recorded12 (6.4%)13 (8.0%)10 (8.1%)35 (7.4%)
Vomiting
All grade62 (39.7%)78 (41.9%)46 (35.1%)186 (39.3%)
≥grade 35 (3.2%)8 (4.3%)2 (1.5%)15 (3.2%)
Not recorded3 (1.6%)2 (1.2%)4 (3.2%)9 (1.9%)
Diarrhea
All-grade53 (34.0%)55 (29.6%)34 (26.0%)142 (30.0%)
≥grade 33 (1.9%)5 (2.7%)5 (3.8%)13 (2.7%)
Not recorded5 (2.7%)3 (1.9%)5 (4.0%)13 (2.7%)
Hypoalbuminemia
All-grade29 (18.6%)50 (26.9%)44 (33.6%)123 (26.0%)
≥grade 31 (0.6%)5 (2.7%)0 (0%)6 (1.3%)
Not recorded28 (15.0%)22 (13.6%)15 (12.1%)65 (13.7%)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Su, Y.-Y.; Chiang, N.-J.; Yang, Y.-H.; Yen, C.-J.; Bai, L.-Y.; Chiu, C.-F.; Chuang, S.-C.; Yang, S.-H.; Chou, W.-C.; Chen, J.-S.; et al. Real-World Data Validation of NAPOLI-1 Nomogram for the Prediction of Overall Survival in Metastatic Pancreatic Cancer. Cancers 2023, 15, 1008. https://doi.org/10.3390/cancers15041008

AMA Style

Su Y-Y, Chiang N-J, Yang Y-H, Yen C-J, Bai L-Y, Chiu C-F, Chuang S-C, Yang S-H, Chou W-C, Chen J-S, et al. Real-World Data Validation of NAPOLI-1 Nomogram for the Prediction of Overall Survival in Metastatic Pancreatic Cancer. Cancers. 2023; 15(4):1008. https://doi.org/10.3390/cancers15041008

Chicago/Turabian Style

Su, Yung-Yeh, Nai-Jung Chiang, Yi-Hsin Yang, Chia-Jui Yen, Li-Yuan Bai, Chang-Fang Chiu, Shih-Chang Chuang, Shih-Hung Yang, Wen-Chi Chou, Jen-Shi Chen, and et al. 2023. "Real-World Data Validation of NAPOLI-1 Nomogram for the Prediction of Overall Survival in Metastatic Pancreatic Cancer" Cancers 15, no. 4: 1008. https://doi.org/10.3390/cancers15041008

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