Abstract

Background

In advanced pancreatic ductal adenocarcinoma (aPDAC), there is no consensual strategy for second-line chemotherapy (L2). Better discrimination of overall survival (OS) may help clinical decision-making. We aimed to predict OS from the beginning of L2 and to assess the benefit from chemotherapy among the identified risk groups.

Methods

Analyses were derived from all consecutive aPDAC patients treated at Besancon University Hospital, Besancon, France, between January 2003 and December 2013 (n = 462). The association of 50 parameters with OS was evaluated using univariate and multivariable Cox analyses. Based on the final model, a prognostic nomogram and score were developed and externally validated. Patients in the external validation cohort who received L2 (n = 163) were treated at three French institutions between January 2010 and April 2016. All statistical tests were two-sided.

Results

In the development cohort, 395 patients (85.5%) were eligible for L2, of which 261 (66.1%) were treated. Age, smoking status, liver metastases, performance status, pain, jaundice, ascites, duration of first-line, and type of L2 regimen were identified as independent prognostic factors for OS in L2. The score determined three groups with median OS of 11.3 months (95% confidence interval [CI] = 9.1 to 12.9 months), 3.6 months (95% CI = 2.6 to 4.7 months), and 1.4 months (95% CI = 1.2 to 1.7 months), for low-, intermediate-, and high-risk groups, respectively (P < .001). By applying the score in the population eligible for L2 but untreated, the chemotherapy benefit was statistically significant across all groups, but with a magnitude of the effect decreased statistically significantly from low- to high-risk groups (P = .001 for treatment and risk groups interaction term). The ability of the score to discriminate OS was confirmed in the external validation cohort.

Conclusions

This prognostic nomogram and score in patients with aPDAC can accurately predict OS before administration of L2 and may help clinicians in their therapeutic decisions.

Pancreatic ductal adenocarcinoma (PDAC) is the fourth cause of cancer-related death in developed countries (1), and it is expected to become the second leading cause of cancer death in 2030 (2). PDAC has the poorest prognostic among digestive malignancies with a five-year survival rate of 5% to 7%, with no important change in death rate between 1997 and 2013 (3). Surgical resection of localized PDAC is the only treatment that can provide prolonged survival. However, diagnosis is made at an advanced stage in the vast majority of cases (>80%). The median overall survival (OS) is about nine to 15 months in patients with locally advanced PDAC, and six to nine months in those with metastatic disease (4,5).

Advanced PDAC (aPDAC) remains a challenging, noncurable disease. Gemcitabine chemotherapy has been the only standard of care in this setting for more than a decade, until 2010. Major progress has been achieved in the landscape of aPDAC management during the five past years with the approval of two active cytotoxic combinations: the FOLFIRINOX (5-fluorouracil [5-FU], irinotecan, and oxaliplatin) and the gemcitabine plus nab-paclitaxel regimens. These regimens were shown to be superior to standard gemcitabine as first-line chemotherapy (L1) in patients with metastatic PDAC, yielding median OS of 11.1 months (95% confidence interval [CI] = 9.0 to 13.1 months) and 8.5 months (95% CI = 7.89 to 9.53 months), respectively (6,7). Currently, clinical practice guidelines for L1 are well established (8,9).

Beyond L1, about half of patients with aPDAC remain in good general condition, and thus may receive subsequent line(s) of chemotherapy (10). Combinations of 5-FU with oxaliplatin (11) or with nanoliposomal irinotecan (12) have shown interesting activity after failure of gemcitabine and can be proposed as second-line chemotherapy (L2) (8). Nevertheless, not all patients seem to benefit from L2 treatment, and L2 has to be discussed on an individual basis in terms of risk benefit for each patient. Of note, evidence to guide patient selection for L2 after progression under FOLFIRINOX or gemcitabine plus nab-paclitaxel regimen is scarce (13), and there is no validated strategy to date.

Age, Eastern Cooperative Oncology Group performance status (ECOG PS), and duration of L1 are so-called “pragmatic parameters,” which are frequently used in multidisciplinary meetings to estimate the potential benefit of L2, but the level of evidence is low (14,15). Thus, identification of reliable factors for patient prognostic stratification is warranted to improve therapeutic decision-making in this setting. However, there is no well-validated and widely accepted prognostic model for application in routine practice or clinical trial for L2 in aPDAC. In this context, it is urgent to develop useful prognostic tools that may help to estimate patient OS in order to guide clinicians’ decision for L2 administration and to optimize future clinical trial design.

In this prospective population-based cohort study, we aimed to develop and validate a prognostic nomogram and score to predict OS in patients with aPDAC who received L2 in routine clinical practice using a broad spectrum of parameters. Then, we assessed the benefit from chemotherapy across risk groups identified by this score.

Methods

Patients

All consecutive patients with histologically proven aPDAC (ie, metastatic, locally advanced, or recurrent after surgery) who were treated at Besancon University Hospital, Besancon, France, between January 2003 and December 2013 were involved in the development cohort. Patients were considered eligible for medical evaluation of L2 indication if they had received one previous line of chemotherapy, including adjuvant chemotherapy in the case of early postoperative tumor relapse (ie, within six months after the last administration of this treatment) and if they were not dead during L1. The external validation cohort included consecutive patients with aPDAC who received L2 with the same inclusion criteria in three other French institutions (Institute Mutualiste Montsouris, Henri Mondor and Reims University Hospitals) between January 2010 and April 2016. The main procedures for both cohorts are specified in the Supplementary Methods (available online). The database was registered and declared to the National French Commission for bioinformatics data and patient liberty (CNIL; No. of CNIL declaration: 1906173 v 0). The study is in accordance with standard procedures in France, with approval from the relevant institutional review boards. A general informed consent was signed by all patients with cancer at the time of their first visit in the Department of Medical Oncology. This consent allows use of their clinical and biological data in the cohort study. No additional specific informed consent for this study was necessary.

Demographics, cancer history, pathological, clinical, biological, and radiological (tumor response according to Response Evaluation Criteria in Solid Tumors [RECIST] v1.1 criteria) parameters at the beginning of L2 (or at the end of L1 for patients who did not receive L2), as well as treatment outcomes, were retrospectively collected from medical records.

Statistical Analysis

Median value (interquartile range) and frequency (percentage) were provided for the description of continuous and categorical variables, respectively. Medians and proportions were compared using Student’s t test and chi-square test (or Fisher’s exact test, if appropriate), respectively.

OS was calculated from the date of progression under L1 (or the date of first administration of L2 if no progression was observed under L1) to the date of death from any cause. Survival data were censored at the last follow-up. Progression-free survival (PFS) was calculated from the date of progression under L1 (or the date of first administration of L2 if no progression was observed under L1) to the date of progression or death from any cause, or the date of the last follow-up, at which point data were censored. OS and PFS were estimated using the Kaplan-Meier method and described using median or rate at specific time points with 95% confidence intervals. Follow-up duration was calculated using a reverse Kaplan-Meier estimation when feasible (16).

Cox proportional hazard models were performed to estimate hazard ratio (HR) and 95% confidence interval for factors associated with OS. The association of 50 baseline parameters with OS was first assessed using univariate Cox analyses, and then parameters with P values of less than .05 were entered into a final multivariable Cox regression model, after considering collinearity among variables with a correlation matrix. When used continuously in the Cox model, a potential nonlinear relationship between predictors and OS was first investigated using the fractional polynomials method to determine the best transformation for continuous variables (17–19) and validated by the restricted cubic splines method with graphical evaluation. The assumption of proportionality was checked by plotting log-minus-log survival curves and by cumulative martingale process plots.

Sensitivity analyses to explore the reliability of the univariate analysis and the robustness of the final multivariable model were performed with a stratified and a frailty approach by using a random component for the hazard function based on the L2 regimen, and with a full-model, a backward, a stepwise, and a forward procedure.

Accuracy of the final model was verified regarding two parameters: discrimination and calibration. The predictive value and the discrimination ability of the final model were assessed with the Harrell’s concordance index (C-index) (20). Random samples of the population were used to derive 95% confidence interval bootstrap percentile for the C-statistic. Calibration was assessed by visual examination of calibration plot. Internal validation of the final model was performed with a bootstrap sample procedure.

Improvement in discrimination ability of our final model compared with the classical approach (“pragmatic parameters”: age, ECOG PS, and duration of L1) was evaluated with the C-index. We used continuous net reclassification improvement (cNRI) (21) and integrated discrimination improvement (IDI) (22) to quantify the performance in risk reclassification of adding others parameters of the final model to the classical approach at six months after the beginning of L2.

The final model was used to establish a nomogram allowing the estimation of median and individual post-L1 OS probabilities at six, 12, 18, and 24 months. At a population level, a prognostic score was constructed with nomogram total points. To give a reasonable spread of risk, we chose to distinguish three prognostic groups according to their risk score levels, which were identified based on cut-points determined following the Cox method (23). Patients’ baseline key characteristics were compared between prognostic risk groups using Fisher’s exact test and the Kruskal-Wallis test for categorical and quantitative parameters, respectively. The discrimination abilities of the scores produced were assessed with the C-index by considering risk group classifications.

The prognostic score developed in patients who received L2 was applied to patients who did not receive L2 in order to assess the benefit from treatment in each group identified.

Another multivariable analysis was performed by including relevant biological data recorded but not initially selected in the multivariable analysis process because of their high rate of missing data.

The prognostic score discrimination ability was confirmed in an external validation cohort and evaluated with the C-index. To identify risk groups and determine their survival profile, the same development cohort–derived risk predictive algorithm was applied. A clinical benefit–centered accuracy of the final model was evaluated by a decision curve analysis (24) for both cohorts.

All analyses were performed using SAS version 9.4 (SAS Institute, Cary NC) and R software version 2.15.2 (R Development Core Team, Vienna, Austria; http://www.r-project.org). P values of less than .05 were considered statistically significant, and all tests were two-sided. Details on the interpretation of important statistical concepts are given in the Supplementary Methods (available online).

Results

Population-Based Prospective Cohort

Among 462 patients who had received L1 for aPDAC, 395 (85.5%) were eligible for medical evaluation of L2 indication; 261 (66.1%) of them actually received L2 and were included in the development cohort while 134 (33.9%) patients were not treated (Figure 1). Patient characteristics in the L2-treated and -untreated groups were statistically significantly different for age, ECOG PS, and duration of L1 (Table 1), corresponding to the three “pragmatic parameters.”

Table 1.

Patients characteristics according to second-line chemotherapy administration in the development cohort

CharacteristicsPopulation eligible for second-line chemotherapy (n = 395)Second-line chemotherapy administration (n = 261)No second-line chemotherapy administration (n = 134)P*
Demographic parameters
 Age, median (IQR), y67.2 (60.8–74.9)65.8 (60.3–72.9)71.5 (63.2–76.9)<.001
 Sex, No. (%).38
  Male233 (59.0)158 (60.5)75 (56.0)
  Female162 (41.0)103 (39.5)59 (44.0)
 Smoking status, No. (%).30
  Never smoker or former smoker296 (75.3)200 (76.9)96 (72.2)
  Current smoker97 (24.7)60 (23.1)37 (27.8)
  Missing211
Pathologic parameters at diagnosis of cancer
 Primary tumor site, No. (%).25
  Head237 (60.9)152 (58.9)85 (64.9)
  Body and/or tail152 (39.1)106 (41.1)46 (35.1)
  Missing633
 Tumor stage, No. (%).42
  Localized92 (23.3)66 (25.3)26 (19.4)
  Locally advanced96 (24.3)61 (23.4)35 (26.1)
  Metastatic207 (52.4)134 (51.3)73 (54.5)
Tumor extension at the beginning of L2
 No. of metastatic sites, No. (%).87
  0–1244 (61.8)162 (62.1)82 (61.2)
  ≥2151 (38.2)99 (37.9)52 (38.8)
 Liver metastases, No. (%).35
  No167 (42.2)106 (40.6)61 (45.5)
  Yes228 (57.7)155 (59.4)73 (55.5)
 Peritoneal metastases, No. (%).65
  No242 (61.3)162 (62.1)80 (59.7)
  Yes153 (38.7)99 (37.9)54 (40.3)
 Lung metastases, No. (%).43
  No304 (77.0)204 (78.2)100 (74.6)
  Yes91 (23.0)57 (21.8)34 (25.4)
 Bone metastases, No. (%).14
  No377 (95.4)252 (96.6)125 (93.3)
  Yes18 (4.6)9 (3.4)9 (6.7)
Clinical parameters at the beginning of L2
 Performance status (WHO), No. (%)<.001
  052 (15.5)47 (18.7)5 (6.0)
  1126 (37.6)118 (47.0)8 (9.5)
  ≥ 2157 (46.9)86 (34.3)71 (84.5)
  Missing601050
 Body mass index, No. (%), kg/m2.02
  Normal weight (18.5–25)225 (59.5)167 (64.2)58 (49.1)
  Underweight (<18.5)66 (17.5)39 (15.0)27 (22.9)
  Overweight (25–30) and obesity (≥30)87 (23.0)54 (20.8)33 (28.0)
  Missing17116
 Pain, No. (%).51
  No165 (48.7)119 (47.6)46 (51.7)
  Yes174 (51.3)131 (52.4)43 (48.3)
  Missing561145
 Jaundice, No. (%).07
  No280 (82.6)212 (84.8)68 (76.4)
  Yes59 (17.4)38 (15.2)21 (23.6)
  Missing561145
 Ascites, No. (%).92
  No274 (80.4)202 (80.5)72 (80.0)
  Yes67 (19.6)49 (19.5)18 (20.0)
  Missing541045
Previous treatment at the beginning of L2
 Primary tumor resection, No. (%).02
  Yes111 (28.3)83 (31.8)28 (21.6)
  No284 (71.7)178 (68.2)106 (78.4)
First-line chemotherapy
 Type of L1 regimen, No. (%).009
  5-FU ± oxaliplatin and/or irinotecan99 (25.1)76 (29.1)23 (17.2)
  Gemcitabine based296 (74.9)185 (70.9)111 (82.8)
 RECIST best response, No. (%).14
  Complete or partial response or stability223 (65.2)170 (67.5)53 (58.9)
  Progression disease119 (34.8)82 (32.5)37 (41.1)
  Missing53944
 Duration of L1, median (IQR), mo4.7 (2.3–8.1)5.7 (2.9–8.7)2.5 (0.8–5.8)<.001
 Reason for discontinuation, No. (%).94
  Progression disease265 (67.1)176 (67.4)89 (66.4)
  Toxicity22 (5.6)15 (5.8)7 (5.2)
  Other108 (27.3)70 (26.8)38 (28.4)
Median follow-up time (95% CI), mo§25.6 (14.6 to 101.0)
CharacteristicsPopulation eligible for second-line chemotherapy (n = 395)Second-line chemotherapy administration (n = 261)No second-line chemotherapy administration (n = 134)P*
Demographic parameters
 Age, median (IQR), y67.2 (60.8–74.9)65.8 (60.3–72.9)71.5 (63.2–76.9)<.001
 Sex, No. (%).38
  Male233 (59.0)158 (60.5)75 (56.0)
  Female162 (41.0)103 (39.5)59 (44.0)
 Smoking status, No. (%).30
  Never smoker or former smoker296 (75.3)200 (76.9)96 (72.2)
  Current smoker97 (24.7)60 (23.1)37 (27.8)
  Missing211
Pathologic parameters at diagnosis of cancer
 Primary tumor site, No. (%).25
  Head237 (60.9)152 (58.9)85 (64.9)
  Body and/or tail152 (39.1)106 (41.1)46 (35.1)
  Missing633
 Tumor stage, No. (%).42
  Localized92 (23.3)66 (25.3)26 (19.4)
  Locally advanced96 (24.3)61 (23.4)35 (26.1)
  Metastatic207 (52.4)134 (51.3)73 (54.5)
Tumor extension at the beginning of L2
 No. of metastatic sites, No. (%).87
  0–1244 (61.8)162 (62.1)82 (61.2)
  ≥2151 (38.2)99 (37.9)52 (38.8)
 Liver metastases, No. (%).35
  No167 (42.2)106 (40.6)61 (45.5)
  Yes228 (57.7)155 (59.4)73 (55.5)
 Peritoneal metastases, No. (%).65
  No242 (61.3)162 (62.1)80 (59.7)
  Yes153 (38.7)99 (37.9)54 (40.3)
 Lung metastases, No. (%).43
  No304 (77.0)204 (78.2)100 (74.6)
  Yes91 (23.0)57 (21.8)34 (25.4)
 Bone metastases, No. (%).14
  No377 (95.4)252 (96.6)125 (93.3)
  Yes18 (4.6)9 (3.4)9 (6.7)
Clinical parameters at the beginning of L2
 Performance status (WHO), No. (%)<.001
  052 (15.5)47 (18.7)5 (6.0)
  1126 (37.6)118 (47.0)8 (9.5)
  ≥ 2157 (46.9)86 (34.3)71 (84.5)
  Missing601050
 Body mass index, No. (%), kg/m2.02
  Normal weight (18.5–25)225 (59.5)167 (64.2)58 (49.1)
  Underweight (<18.5)66 (17.5)39 (15.0)27 (22.9)
  Overweight (25–30) and obesity (≥30)87 (23.0)54 (20.8)33 (28.0)
  Missing17116
 Pain, No. (%).51
  No165 (48.7)119 (47.6)46 (51.7)
  Yes174 (51.3)131 (52.4)43 (48.3)
  Missing561145
 Jaundice, No. (%).07
  No280 (82.6)212 (84.8)68 (76.4)
  Yes59 (17.4)38 (15.2)21 (23.6)
  Missing561145
 Ascites, No. (%).92
  No274 (80.4)202 (80.5)72 (80.0)
  Yes67 (19.6)49 (19.5)18 (20.0)
  Missing541045
Previous treatment at the beginning of L2
 Primary tumor resection, No. (%).02
  Yes111 (28.3)83 (31.8)28 (21.6)
  No284 (71.7)178 (68.2)106 (78.4)
First-line chemotherapy
 Type of L1 regimen, No. (%).009
  5-FU ± oxaliplatin and/or irinotecan99 (25.1)76 (29.1)23 (17.2)
  Gemcitabine based296 (74.9)185 (70.9)111 (82.8)
 RECIST best response, No. (%).14
  Complete or partial response or stability223 (65.2)170 (67.5)53 (58.9)
  Progression disease119 (34.8)82 (32.5)37 (41.1)
  Missing53944
 Duration of L1, median (IQR), mo4.7 (2.3–8.1)5.7 (2.9–8.7)2.5 (0.8–5.8)<.001
 Reason for discontinuation, No. (%).94
  Progression disease265 (67.1)176 (67.4)89 (66.4)
  Toxicity22 (5.6)15 (5.8)7 (5.2)
  Other108 (27.3)70 (26.8)38 (28.4)
Median follow-up time (95% CI), mo§25.6 (14.6 to 101.0)
*

χ2 tests or Fisher’s exact tests used to compare proportions, and Wilcoxon tests used to compare continuous variables between the groups with or without second-line chemotherapy administration. All statistical tests were two-sided. 5-FU = 5-fluorouracil; IQR = interquartile range; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Corresponding to prescription of morphine.

All patients were followed until death (maximum time observed = 126.1 months) except 28 censored patients with a median follow-up equal to 11.7 months.

§

All patients were followed until death (maximum time observed = 126.1 months) except 14 censored patients with a median follow-up equal to 14.1 months.

Table 1.

Patients characteristics according to second-line chemotherapy administration in the development cohort

CharacteristicsPopulation eligible for second-line chemotherapy (n = 395)Second-line chemotherapy administration (n = 261)No second-line chemotherapy administration (n = 134)P*
Demographic parameters
 Age, median (IQR), y67.2 (60.8–74.9)65.8 (60.3–72.9)71.5 (63.2–76.9)<.001
 Sex, No. (%).38
  Male233 (59.0)158 (60.5)75 (56.0)
  Female162 (41.0)103 (39.5)59 (44.0)
 Smoking status, No. (%).30
  Never smoker or former smoker296 (75.3)200 (76.9)96 (72.2)
  Current smoker97 (24.7)60 (23.1)37 (27.8)
  Missing211
Pathologic parameters at diagnosis of cancer
 Primary tumor site, No. (%).25
  Head237 (60.9)152 (58.9)85 (64.9)
  Body and/or tail152 (39.1)106 (41.1)46 (35.1)
  Missing633
 Tumor stage, No. (%).42
  Localized92 (23.3)66 (25.3)26 (19.4)
  Locally advanced96 (24.3)61 (23.4)35 (26.1)
  Metastatic207 (52.4)134 (51.3)73 (54.5)
Tumor extension at the beginning of L2
 No. of metastatic sites, No. (%).87
  0–1244 (61.8)162 (62.1)82 (61.2)
  ≥2151 (38.2)99 (37.9)52 (38.8)
 Liver metastases, No. (%).35
  No167 (42.2)106 (40.6)61 (45.5)
  Yes228 (57.7)155 (59.4)73 (55.5)
 Peritoneal metastases, No. (%).65
  No242 (61.3)162 (62.1)80 (59.7)
  Yes153 (38.7)99 (37.9)54 (40.3)
 Lung metastases, No. (%).43
  No304 (77.0)204 (78.2)100 (74.6)
  Yes91 (23.0)57 (21.8)34 (25.4)
 Bone metastases, No. (%).14
  No377 (95.4)252 (96.6)125 (93.3)
  Yes18 (4.6)9 (3.4)9 (6.7)
Clinical parameters at the beginning of L2
 Performance status (WHO), No. (%)<.001
  052 (15.5)47 (18.7)5 (6.0)
  1126 (37.6)118 (47.0)8 (9.5)
  ≥ 2157 (46.9)86 (34.3)71 (84.5)
  Missing601050
 Body mass index, No. (%), kg/m2.02
  Normal weight (18.5–25)225 (59.5)167 (64.2)58 (49.1)
  Underweight (<18.5)66 (17.5)39 (15.0)27 (22.9)
  Overweight (25–30) and obesity (≥30)87 (23.0)54 (20.8)33 (28.0)
  Missing17116
 Pain, No. (%).51
  No165 (48.7)119 (47.6)46 (51.7)
  Yes174 (51.3)131 (52.4)43 (48.3)
  Missing561145
 Jaundice, No. (%).07
  No280 (82.6)212 (84.8)68 (76.4)
  Yes59 (17.4)38 (15.2)21 (23.6)
  Missing561145
 Ascites, No. (%).92
  No274 (80.4)202 (80.5)72 (80.0)
  Yes67 (19.6)49 (19.5)18 (20.0)
  Missing541045
Previous treatment at the beginning of L2
 Primary tumor resection, No. (%).02
  Yes111 (28.3)83 (31.8)28 (21.6)
  No284 (71.7)178 (68.2)106 (78.4)
First-line chemotherapy
 Type of L1 regimen, No. (%).009
  5-FU ± oxaliplatin and/or irinotecan99 (25.1)76 (29.1)23 (17.2)
  Gemcitabine based296 (74.9)185 (70.9)111 (82.8)
 RECIST best response, No. (%).14
  Complete or partial response or stability223 (65.2)170 (67.5)53 (58.9)
  Progression disease119 (34.8)82 (32.5)37 (41.1)
  Missing53944
 Duration of L1, median (IQR), mo4.7 (2.3–8.1)5.7 (2.9–8.7)2.5 (0.8–5.8)<.001
 Reason for discontinuation, No. (%).94
  Progression disease265 (67.1)176 (67.4)89 (66.4)
  Toxicity22 (5.6)15 (5.8)7 (5.2)
  Other108 (27.3)70 (26.8)38 (28.4)
Median follow-up time (95% CI), mo§25.6 (14.6 to 101.0)
CharacteristicsPopulation eligible for second-line chemotherapy (n = 395)Second-line chemotherapy administration (n = 261)No second-line chemotherapy administration (n = 134)P*
Demographic parameters
 Age, median (IQR), y67.2 (60.8–74.9)65.8 (60.3–72.9)71.5 (63.2–76.9)<.001
 Sex, No. (%).38
  Male233 (59.0)158 (60.5)75 (56.0)
  Female162 (41.0)103 (39.5)59 (44.0)
 Smoking status, No. (%).30
  Never smoker or former smoker296 (75.3)200 (76.9)96 (72.2)
  Current smoker97 (24.7)60 (23.1)37 (27.8)
  Missing211
Pathologic parameters at diagnosis of cancer
 Primary tumor site, No. (%).25
  Head237 (60.9)152 (58.9)85 (64.9)
  Body and/or tail152 (39.1)106 (41.1)46 (35.1)
  Missing633
 Tumor stage, No. (%).42
  Localized92 (23.3)66 (25.3)26 (19.4)
  Locally advanced96 (24.3)61 (23.4)35 (26.1)
  Metastatic207 (52.4)134 (51.3)73 (54.5)
Tumor extension at the beginning of L2
 No. of metastatic sites, No. (%).87
  0–1244 (61.8)162 (62.1)82 (61.2)
  ≥2151 (38.2)99 (37.9)52 (38.8)
 Liver metastases, No. (%).35
  No167 (42.2)106 (40.6)61 (45.5)
  Yes228 (57.7)155 (59.4)73 (55.5)
 Peritoneal metastases, No. (%).65
  No242 (61.3)162 (62.1)80 (59.7)
  Yes153 (38.7)99 (37.9)54 (40.3)
 Lung metastases, No. (%).43
  No304 (77.0)204 (78.2)100 (74.6)
  Yes91 (23.0)57 (21.8)34 (25.4)
 Bone metastases, No. (%).14
  No377 (95.4)252 (96.6)125 (93.3)
  Yes18 (4.6)9 (3.4)9 (6.7)
Clinical parameters at the beginning of L2
 Performance status (WHO), No. (%)<.001
  052 (15.5)47 (18.7)5 (6.0)
  1126 (37.6)118 (47.0)8 (9.5)
  ≥ 2157 (46.9)86 (34.3)71 (84.5)
  Missing601050
 Body mass index, No. (%), kg/m2.02
  Normal weight (18.5–25)225 (59.5)167 (64.2)58 (49.1)
  Underweight (<18.5)66 (17.5)39 (15.0)27 (22.9)
  Overweight (25–30) and obesity (≥30)87 (23.0)54 (20.8)33 (28.0)
  Missing17116
 Pain, No. (%).51
  No165 (48.7)119 (47.6)46 (51.7)
  Yes174 (51.3)131 (52.4)43 (48.3)
  Missing561145
 Jaundice, No. (%).07
  No280 (82.6)212 (84.8)68 (76.4)
  Yes59 (17.4)38 (15.2)21 (23.6)
  Missing561145
 Ascites, No. (%).92
  No274 (80.4)202 (80.5)72 (80.0)
  Yes67 (19.6)49 (19.5)18 (20.0)
  Missing541045
Previous treatment at the beginning of L2
 Primary tumor resection, No. (%).02
  Yes111 (28.3)83 (31.8)28 (21.6)
  No284 (71.7)178 (68.2)106 (78.4)
First-line chemotherapy
 Type of L1 regimen, No. (%).009
  5-FU ± oxaliplatin and/or irinotecan99 (25.1)76 (29.1)23 (17.2)
  Gemcitabine based296 (74.9)185 (70.9)111 (82.8)
 RECIST best response, No. (%).14
  Complete or partial response or stability223 (65.2)170 (67.5)53 (58.9)
  Progression disease119 (34.8)82 (32.5)37 (41.1)
  Missing53944
 Duration of L1, median (IQR), mo4.7 (2.3–8.1)5.7 (2.9–8.7)2.5 (0.8–5.8)<.001
 Reason for discontinuation, No. (%).94
  Progression disease265 (67.1)176 (67.4)89 (66.4)
  Toxicity22 (5.6)15 (5.8)7 (5.2)
  Other108 (27.3)70 (26.8)38 (28.4)
Median follow-up time (95% CI), mo§25.6 (14.6 to 101.0)
*

χ2 tests or Fisher’s exact tests used to compare proportions, and Wilcoxon tests used to compare continuous variables between the groups with or without second-line chemotherapy administration. All statistical tests were two-sided. 5-FU = 5-fluorouracil; IQR = interquartile range; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Corresponding to prescription of morphine.

All patients were followed until death (maximum time observed = 126.1 months) except 28 censored patients with a median follow-up equal to 11.7 months.

§

All patients were followed until death (maximum time observed = 126.1 months) except 14 censored patients with a median follow-up equal to 14.1 months.

Figure 1.

Flow chart. *Almost half of patients (44.1%) in L2 received a third line or more (up to seven lines). 5-FU = 5-fluorouracil; L1 = first-line chemotherapy; L2 = second-line chemotherapy.

In the external validation cohort, 163 patients with aPDAC who received L2 treatment were analyzed, and 126 events (deaths) were observed. The two cohorts displayed similar patient characteristics, except for the type of L1 and L2 regimens (Supplementary Table 1, available online).

Determinants of OS in Patients Receiving L2

In univariate Cox analysis, we identified 27 parameters as prognostics factors for OS with P values of less than .05 (Table 2). The main analyses did not include biological parameters because of the high rate of missing data in some of them. A correlation matrix was used to detect statistically significant correlations between investigated parameters (Supplementary Figure 1, available online). When a correlation was identified, only the most clinically relevant variable was considered in the multivariable model. The multivariable Cox analysis showed nine independent risk factors for OS: age, smoking status, liver metastases, ECOG PS, pain, jaundice, ascites, duration of L1, and type of L2 regimen (Table 3). The transformations used for continuous variables are summarized in Supplementary Figure 2 (available online).

Table 2.

Prognostics factors associated with overall survival in univariate analysis

ParametersNo. of patientsNo. of eventsHR (95% CI)P*
Demographic parameters
 Age, y2612471.02 (1.00 to 1.03).04
 Sex
  Male1581521
  Female103950.80 (0.62 to 1.04).07
 Smoking status
  Never smoker or former smoker2001871
  Current smoker60591.53 (1.14 to 2.07).005
  Missing11
 Personal history of cancer
  No2101971
  Yes49481.16 (0.84 to 1.59).37
  Missing22
 Family history of cancer
  No1461381
  Yes1131070.85 (0.66 to 1.10).22
  Missing20
 Family history of pancreatic cancer
  No2402271
  Yes19181.18 (0.73 to 1.91).51
  Missing22
Pathologic parameters at diagnosis of cancer
 Primary tumor site
  Head1521421
  Body and/or tail1061021.25 (0.97 to 1.62).09
  Missing33
 Primary tumor size, mm2422291.01 (1.00 to 1.01).21
  Missing1918
 Histological grade
  Well or moderately differentiated100921
  Poorly differentiated or undifferentiated28261.33 (0.86 to 2.07).20
  Missing133129
 Tumor stage
  Localized66611
  Locally advanced61571.15 (0.80 to 1.65)
  Metastatic1341291.80 (1.32 to 2.45)<.001
Tumor extension at the beginning of L2
 No. of metastatic sites
  0–11621521
  ≥299951.69 (1.31 to 2.19)<.001
 Lymph node metastases
  No2402301
  Yes21170.80 (0.49 to 1.31).37
 Liver metastases
  No1061001
  Yes1551471.74 (1.35 to 2.26)<.001
 Peritoneal metastases
  No1621481
  Yes99991.61 (1.24 to 2.08)<.001
 Lung metastases
  No2041931
  Yes57540.93 (0.69 to 1.26).63
 Bone metastases
  No2522391
  Yes980.86 (0.42 to 1.74).67
 Other metastases
  No2572431
  Yes441.19 (0.44 to 3.22).73
 Isolated lung metastases
  No2482351
  Yes13120.52 (0.29 to 0.93).03
 Performance status (WHO)
  047401
  11181151.97 (1.37 to 2.85)
  ≥286834.62 (3.11 to 6.85)<.001
  Missing109
 Body mass index, kg/m22602470.98 (0.95 to 1.02).36
  Missing10
 Body mass index, kg/m2
  Normal weight (18.5–25)1671601
  Underweight (<18.5)39361.18 (0.82 to 1.70)
  Overweight (25–30) and obesity (≥30)54510.94 (0.68 to 1.29).56
  Missing10
 Pain
  No1191091
  Yes1311281.89 (1.45 to 2.45)<.001
  Missing1110
 Jaundice
  No2122001
  Yes38372.68 (1.87 to 3.85)<.001
  Missing1110
 Ascites
  No2021901
  Yes49482.14 (1.55 to 2.95)<.001
  Missing109
Biological parameters at the beginning of L2
 Hemoglobin, g/dL2332230.82 (0.75 to 0.90)<.001
  Missing2824
 Neutrophils, mm3 (square root value)2192101.02 (1.01 to 1.02)<.001
  Missing4237
 Lymphocytes, mm3 (inverse transformation value)2122042.28 ×10134 (1.336 ×1030 to 3.88 ×10238).01
  Missing4943
 Neutrophil-to-lymphocyte ratio (square root value)2102021.67 (1.42 to 1.96)<.001
  Missing5145
 Platelets, mm3 (log value)2302201.53 (0.87 to 2.69).14
  Missing3127
 Creatinine, µmol/L (log value)2472340.79 (0.26 to 2.36).67
  Missing1413
 Total bilirubin, µmol/L (square root value)2182091.15 (1.09 to 1.22)<.001
  Missing4338
 Albumin, g/L1611550.94 (0.91 to 0.96)<.001
  Missing10092
 CA19-9, UI/mL (square root value)2041951.01 (1.01 to 1.01)<.001
  Missing5752
 CEA, ng/mL (log value)1591501.86 (1.47 to 2.34)<.001
  Missing10297
Previous treatment at the beginning of L2
 Primary tumor resection
  Yes83761
  No1781711.56 (1.20 to 2.07).001
 Adjuvant chemotherapy
  Yes67611
  No1941861.59 (1.18 to 2.13).002
 Radiotherapy
  Yes17151
  No2442321.11 (0.66 to 1.88).69
 Neo-adjuvant chemotherapy
  Yes331
  No2582441.01 (0.32 to 3.15).99
 Biliary stent
  Yes83781
  No1771680.84 (0.64 to 1.10).21
  Missing11
 Duodenal stent
  Yes19181
  No2412280.82 (0.50 to 1.32).41
  Missing11
 Alcohol celiac plexus
  Yes22211
  No2382250.75 (0.48 to 1.17).20
  Missing11
First-line chemotherapy
 Type of L1 regimen
  5-FU ± oxaliplatin and/or irinotecan76691
  Gemcitabine based1851781.11 (0.84 to 1.47).46
 No. of cures2612470.99 (0.98 to 1.00).01
 RECIST best response
  Complete or partial response or stability1701611
  Progression disease82771.38 (1.05 to 1.81).02
  Missing99
 Duration of L1 (log value), mo2612470.55 (0.39 to 0.78)<.001
 Toxicity of grade 3 or 4
  No1981891
  Yes63580.83 (0.62 to 1.12).22
 Type of toxicity
  Digestive871
  Neurological17140.96 (0.39 to 2.39)
  Skin442.29 (0.65 to 8.05)
  Hematology20201.73 (0.72 to 4.12)
  Other14131.70 (0.67 to 4.32).30
  Missing198189
 Reason for discontinuation
  Other70611
  Toxicity15151.27 (0.72 to 2.24)
  Progression disease1761711.58 (1.17 to 2.11)<.01
 Locoregional progression
  No2071961
  Yes54510.94 (0.69 to 1.28).69
 Metastatic progression
  No67611
  Yes1941861.44 (1.07 to 1.93).02
Second-line chemotherapy
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan2041921
  Gemcitabine based57551.49 (1.10 to 2.02).01
ParametersNo. of patientsNo. of eventsHR (95% CI)P*
Demographic parameters
 Age, y2612471.02 (1.00 to 1.03).04
 Sex
  Male1581521
  Female103950.80 (0.62 to 1.04).07
 Smoking status
  Never smoker or former smoker2001871
  Current smoker60591.53 (1.14 to 2.07).005
  Missing11
 Personal history of cancer
  No2101971
  Yes49481.16 (0.84 to 1.59).37
  Missing22
 Family history of cancer
  No1461381
  Yes1131070.85 (0.66 to 1.10).22
  Missing20
 Family history of pancreatic cancer
  No2402271
  Yes19181.18 (0.73 to 1.91).51
  Missing22
Pathologic parameters at diagnosis of cancer
 Primary tumor site
  Head1521421
  Body and/or tail1061021.25 (0.97 to 1.62).09
  Missing33
 Primary tumor size, mm2422291.01 (1.00 to 1.01).21
  Missing1918
 Histological grade
  Well or moderately differentiated100921
  Poorly differentiated or undifferentiated28261.33 (0.86 to 2.07).20
  Missing133129
 Tumor stage
  Localized66611
  Locally advanced61571.15 (0.80 to 1.65)
  Metastatic1341291.80 (1.32 to 2.45)<.001
Tumor extension at the beginning of L2
 No. of metastatic sites
  0–11621521
  ≥299951.69 (1.31 to 2.19)<.001
 Lymph node metastases
  No2402301
  Yes21170.80 (0.49 to 1.31).37
 Liver metastases
  No1061001
  Yes1551471.74 (1.35 to 2.26)<.001
 Peritoneal metastases
  No1621481
  Yes99991.61 (1.24 to 2.08)<.001
 Lung metastases
  No2041931
  Yes57540.93 (0.69 to 1.26).63
 Bone metastases
  No2522391
  Yes980.86 (0.42 to 1.74).67
 Other metastases
  No2572431
  Yes441.19 (0.44 to 3.22).73
 Isolated lung metastases
  No2482351
  Yes13120.52 (0.29 to 0.93).03
 Performance status (WHO)
  047401
  11181151.97 (1.37 to 2.85)
  ≥286834.62 (3.11 to 6.85)<.001
  Missing109
 Body mass index, kg/m22602470.98 (0.95 to 1.02).36
  Missing10
 Body mass index, kg/m2
  Normal weight (18.5–25)1671601
  Underweight (<18.5)39361.18 (0.82 to 1.70)
  Overweight (25–30) and obesity (≥30)54510.94 (0.68 to 1.29).56
  Missing10
 Pain
  No1191091
  Yes1311281.89 (1.45 to 2.45)<.001
  Missing1110
 Jaundice
  No2122001
  Yes38372.68 (1.87 to 3.85)<.001
  Missing1110
 Ascites
  No2021901
  Yes49482.14 (1.55 to 2.95)<.001
  Missing109
Biological parameters at the beginning of L2
 Hemoglobin, g/dL2332230.82 (0.75 to 0.90)<.001
  Missing2824
 Neutrophils, mm3 (square root value)2192101.02 (1.01 to 1.02)<.001
  Missing4237
 Lymphocytes, mm3 (inverse transformation value)2122042.28 ×10134 (1.336 ×1030 to 3.88 ×10238).01
  Missing4943
 Neutrophil-to-lymphocyte ratio (square root value)2102021.67 (1.42 to 1.96)<.001
  Missing5145
 Platelets, mm3 (log value)2302201.53 (0.87 to 2.69).14
  Missing3127
 Creatinine, µmol/L (log value)2472340.79 (0.26 to 2.36).67
  Missing1413
 Total bilirubin, µmol/L (square root value)2182091.15 (1.09 to 1.22)<.001
  Missing4338
 Albumin, g/L1611550.94 (0.91 to 0.96)<.001
  Missing10092
 CA19-9, UI/mL (square root value)2041951.01 (1.01 to 1.01)<.001
  Missing5752
 CEA, ng/mL (log value)1591501.86 (1.47 to 2.34)<.001
  Missing10297
Previous treatment at the beginning of L2
 Primary tumor resection
  Yes83761
  No1781711.56 (1.20 to 2.07).001
 Adjuvant chemotherapy
  Yes67611
  No1941861.59 (1.18 to 2.13).002
 Radiotherapy
  Yes17151
  No2442321.11 (0.66 to 1.88).69
 Neo-adjuvant chemotherapy
  Yes331
  No2582441.01 (0.32 to 3.15).99
 Biliary stent
  Yes83781
  No1771680.84 (0.64 to 1.10).21
  Missing11
 Duodenal stent
  Yes19181
  No2412280.82 (0.50 to 1.32).41
  Missing11
 Alcohol celiac plexus
  Yes22211
  No2382250.75 (0.48 to 1.17).20
  Missing11
First-line chemotherapy
 Type of L1 regimen
  5-FU ± oxaliplatin and/or irinotecan76691
  Gemcitabine based1851781.11 (0.84 to 1.47).46
 No. of cures2612470.99 (0.98 to 1.00).01
 RECIST best response
  Complete or partial response or stability1701611
  Progression disease82771.38 (1.05 to 1.81).02
  Missing99
 Duration of L1 (log value), mo2612470.55 (0.39 to 0.78)<.001
 Toxicity of grade 3 or 4
  No1981891
  Yes63580.83 (0.62 to 1.12).22
 Type of toxicity
  Digestive871
  Neurological17140.96 (0.39 to 2.39)
  Skin442.29 (0.65 to 8.05)
  Hematology20201.73 (0.72 to 4.12)
  Other14131.70 (0.67 to 4.32).30
  Missing198189
 Reason for discontinuation
  Other70611
  Toxicity15151.27 (0.72 to 2.24)
  Progression disease1761711.58 (1.17 to 2.11)<.01
 Locoregional progression
  No2071961
  Yes54510.94 (0.69 to 1.28).69
 Metastatic progression
  No67611
  Yes1941861.44 (1.07 to 1.93).02
Second-line chemotherapy
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan2041921
  Gemcitabine based57551.49 (1.10 to 2.02).01
*

Cox proportional hazard models used to estimate association of the parameters with overall survival. Values of P < .05 were considered statistically significant, and all tests were two-sided. 5-FU = 5-fluorouracil; CA 19-9 = carbohydrate antigen 19-9; CEA = carcinoembryonic antigen; HR = hazard ratio; CI = confidence interval; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Corresponding to prescription of morphine.

Table 2.

Prognostics factors associated with overall survival in univariate analysis

ParametersNo. of patientsNo. of eventsHR (95% CI)P*
Demographic parameters
 Age, y2612471.02 (1.00 to 1.03).04
 Sex
  Male1581521
  Female103950.80 (0.62 to 1.04).07
 Smoking status
  Never smoker or former smoker2001871
  Current smoker60591.53 (1.14 to 2.07).005
  Missing11
 Personal history of cancer
  No2101971
  Yes49481.16 (0.84 to 1.59).37
  Missing22
 Family history of cancer
  No1461381
  Yes1131070.85 (0.66 to 1.10).22
  Missing20
 Family history of pancreatic cancer
  No2402271
  Yes19181.18 (0.73 to 1.91).51
  Missing22
Pathologic parameters at diagnosis of cancer
 Primary tumor site
  Head1521421
  Body and/or tail1061021.25 (0.97 to 1.62).09
  Missing33
 Primary tumor size, mm2422291.01 (1.00 to 1.01).21
  Missing1918
 Histological grade
  Well or moderately differentiated100921
  Poorly differentiated or undifferentiated28261.33 (0.86 to 2.07).20
  Missing133129
 Tumor stage
  Localized66611
  Locally advanced61571.15 (0.80 to 1.65)
  Metastatic1341291.80 (1.32 to 2.45)<.001
Tumor extension at the beginning of L2
 No. of metastatic sites
  0–11621521
  ≥299951.69 (1.31 to 2.19)<.001
 Lymph node metastases
  No2402301
  Yes21170.80 (0.49 to 1.31).37
 Liver metastases
  No1061001
  Yes1551471.74 (1.35 to 2.26)<.001
 Peritoneal metastases
  No1621481
  Yes99991.61 (1.24 to 2.08)<.001
 Lung metastases
  No2041931
  Yes57540.93 (0.69 to 1.26).63
 Bone metastases
  No2522391
  Yes980.86 (0.42 to 1.74).67
 Other metastases
  No2572431
  Yes441.19 (0.44 to 3.22).73
 Isolated lung metastases
  No2482351
  Yes13120.52 (0.29 to 0.93).03
 Performance status (WHO)
  047401
  11181151.97 (1.37 to 2.85)
  ≥286834.62 (3.11 to 6.85)<.001
  Missing109
 Body mass index, kg/m22602470.98 (0.95 to 1.02).36
  Missing10
 Body mass index, kg/m2
  Normal weight (18.5–25)1671601
  Underweight (<18.5)39361.18 (0.82 to 1.70)
  Overweight (25–30) and obesity (≥30)54510.94 (0.68 to 1.29).56
  Missing10
 Pain
  No1191091
  Yes1311281.89 (1.45 to 2.45)<.001
  Missing1110
 Jaundice
  No2122001
  Yes38372.68 (1.87 to 3.85)<.001
  Missing1110
 Ascites
  No2021901
  Yes49482.14 (1.55 to 2.95)<.001
  Missing109
Biological parameters at the beginning of L2
 Hemoglobin, g/dL2332230.82 (0.75 to 0.90)<.001
  Missing2824
 Neutrophils, mm3 (square root value)2192101.02 (1.01 to 1.02)<.001
  Missing4237
 Lymphocytes, mm3 (inverse transformation value)2122042.28 ×10134 (1.336 ×1030 to 3.88 ×10238).01
  Missing4943
 Neutrophil-to-lymphocyte ratio (square root value)2102021.67 (1.42 to 1.96)<.001
  Missing5145
 Platelets, mm3 (log value)2302201.53 (0.87 to 2.69).14
  Missing3127
 Creatinine, µmol/L (log value)2472340.79 (0.26 to 2.36).67
  Missing1413
 Total bilirubin, µmol/L (square root value)2182091.15 (1.09 to 1.22)<.001
  Missing4338
 Albumin, g/L1611550.94 (0.91 to 0.96)<.001
  Missing10092
 CA19-9, UI/mL (square root value)2041951.01 (1.01 to 1.01)<.001
  Missing5752
 CEA, ng/mL (log value)1591501.86 (1.47 to 2.34)<.001
  Missing10297
Previous treatment at the beginning of L2
 Primary tumor resection
  Yes83761
  No1781711.56 (1.20 to 2.07).001
 Adjuvant chemotherapy
  Yes67611
  No1941861.59 (1.18 to 2.13).002
 Radiotherapy
  Yes17151
  No2442321.11 (0.66 to 1.88).69
 Neo-adjuvant chemotherapy
  Yes331
  No2582441.01 (0.32 to 3.15).99
 Biliary stent
  Yes83781
  No1771680.84 (0.64 to 1.10).21
  Missing11
 Duodenal stent
  Yes19181
  No2412280.82 (0.50 to 1.32).41
  Missing11
 Alcohol celiac plexus
  Yes22211
  No2382250.75 (0.48 to 1.17).20
  Missing11
First-line chemotherapy
 Type of L1 regimen
  5-FU ± oxaliplatin and/or irinotecan76691
  Gemcitabine based1851781.11 (0.84 to 1.47).46
 No. of cures2612470.99 (0.98 to 1.00).01
 RECIST best response
  Complete or partial response or stability1701611
  Progression disease82771.38 (1.05 to 1.81).02
  Missing99
 Duration of L1 (log value), mo2612470.55 (0.39 to 0.78)<.001
 Toxicity of grade 3 or 4
  No1981891
  Yes63580.83 (0.62 to 1.12).22
 Type of toxicity
  Digestive871
  Neurological17140.96 (0.39 to 2.39)
  Skin442.29 (0.65 to 8.05)
  Hematology20201.73 (0.72 to 4.12)
  Other14131.70 (0.67 to 4.32).30
  Missing198189
 Reason for discontinuation
  Other70611
  Toxicity15151.27 (0.72 to 2.24)
  Progression disease1761711.58 (1.17 to 2.11)<.01
 Locoregional progression
  No2071961
  Yes54510.94 (0.69 to 1.28).69
 Metastatic progression
  No67611
  Yes1941861.44 (1.07 to 1.93).02
Second-line chemotherapy
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan2041921
  Gemcitabine based57551.49 (1.10 to 2.02).01
ParametersNo. of patientsNo. of eventsHR (95% CI)P*
Demographic parameters
 Age, y2612471.02 (1.00 to 1.03).04
 Sex
  Male1581521
  Female103950.80 (0.62 to 1.04).07
 Smoking status
  Never smoker or former smoker2001871
  Current smoker60591.53 (1.14 to 2.07).005
  Missing11
 Personal history of cancer
  No2101971
  Yes49481.16 (0.84 to 1.59).37
  Missing22
 Family history of cancer
  No1461381
  Yes1131070.85 (0.66 to 1.10).22
  Missing20
 Family history of pancreatic cancer
  No2402271
  Yes19181.18 (0.73 to 1.91).51
  Missing22
Pathologic parameters at diagnosis of cancer
 Primary tumor site
  Head1521421
  Body and/or tail1061021.25 (0.97 to 1.62).09
  Missing33
 Primary tumor size, mm2422291.01 (1.00 to 1.01).21
  Missing1918
 Histological grade
  Well or moderately differentiated100921
  Poorly differentiated or undifferentiated28261.33 (0.86 to 2.07).20
  Missing133129
 Tumor stage
  Localized66611
  Locally advanced61571.15 (0.80 to 1.65)
  Metastatic1341291.80 (1.32 to 2.45)<.001
Tumor extension at the beginning of L2
 No. of metastatic sites
  0–11621521
  ≥299951.69 (1.31 to 2.19)<.001
 Lymph node metastases
  No2402301
  Yes21170.80 (0.49 to 1.31).37
 Liver metastases
  No1061001
  Yes1551471.74 (1.35 to 2.26)<.001
 Peritoneal metastases
  No1621481
  Yes99991.61 (1.24 to 2.08)<.001
 Lung metastases
  No2041931
  Yes57540.93 (0.69 to 1.26).63
 Bone metastases
  No2522391
  Yes980.86 (0.42 to 1.74).67
 Other metastases
  No2572431
  Yes441.19 (0.44 to 3.22).73
 Isolated lung metastases
  No2482351
  Yes13120.52 (0.29 to 0.93).03
 Performance status (WHO)
  047401
  11181151.97 (1.37 to 2.85)
  ≥286834.62 (3.11 to 6.85)<.001
  Missing109
 Body mass index, kg/m22602470.98 (0.95 to 1.02).36
  Missing10
 Body mass index, kg/m2
  Normal weight (18.5–25)1671601
  Underweight (<18.5)39361.18 (0.82 to 1.70)
  Overweight (25–30) and obesity (≥30)54510.94 (0.68 to 1.29).56
  Missing10
 Pain
  No1191091
  Yes1311281.89 (1.45 to 2.45)<.001
  Missing1110
 Jaundice
  No2122001
  Yes38372.68 (1.87 to 3.85)<.001
  Missing1110
 Ascites
  No2021901
  Yes49482.14 (1.55 to 2.95)<.001
  Missing109
Biological parameters at the beginning of L2
 Hemoglobin, g/dL2332230.82 (0.75 to 0.90)<.001
  Missing2824
 Neutrophils, mm3 (square root value)2192101.02 (1.01 to 1.02)<.001
  Missing4237
 Lymphocytes, mm3 (inverse transformation value)2122042.28 ×10134 (1.336 ×1030 to 3.88 ×10238).01
  Missing4943
 Neutrophil-to-lymphocyte ratio (square root value)2102021.67 (1.42 to 1.96)<.001
  Missing5145
 Platelets, mm3 (log value)2302201.53 (0.87 to 2.69).14
  Missing3127
 Creatinine, µmol/L (log value)2472340.79 (0.26 to 2.36).67
  Missing1413
 Total bilirubin, µmol/L (square root value)2182091.15 (1.09 to 1.22)<.001
  Missing4338
 Albumin, g/L1611550.94 (0.91 to 0.96)<.001
  Missing10092
 CA19-9, UI/mL (square root value)2041951.01 (1.01 to 1.01)<.001
  Missing5752
 CEA, ng/mL (log value)1591501.86 (1.47 to 2.34)<.001
  Missing10297
Previous treatment at the beginning of L2
 Primary tumor resection
  Yes83761
  No1781711.56 (1.20 to 2.07).001
 Adjuvant chemotherapy
  Yes67611
  No1941861.59 (1.18 to 2.13).002
 Radiotherapy
  Yes17151
  No2442321.11 (0.66 to 1.88).69
 Neo-adjuvant chemotherapy
  Yes331
  No2582441.01 (0.32 to 3.15).99
 Biliary stent
  Yes83781
  No1771680.84 (0.64 to 1.10).21
  Missing11
 Duodenal stent
  Yes19181
  No2412280.82 (0.50 to 1.32).41
  Missing11
 Alcohol celiac plexus
  Yes22211
  No2382250.75 (0.48 to 1.17).20
  Missing11
First-line chemotherapy
 Type of L1 regimen
  5-FU ± oxaliplatin and/or irinotecan76691
  Gemcitabine based1851781.11 (0.84 to 1.47).46
 No. of cures2612470.99 (0.98 to 1.00).01
 RECIST best response
  Complete or partial response or stability1701611
  Progression disease82771.38 (1.05 to 1.81).02
  Missing99
 Duration of L1 (log value), mo2612470.55 (0.39 to 0.78)<.001
 Toxicity of grade 3 or 4
  No1981891
  Yes63580.83 (0.62 to 1.12).22
 Type of toxicity
  Digestive871
  Neurological17140.96 (0.39 to 2.39)
  Skin442.29 (0.65 to 8.05)
  Hematology20201.73 (0.72 to 4.12)
  Other14131.70 (0.67 to 4.32).30
  Missing198189
 Reason for discontinuation
  Other70611
  Toxicity15151.27 (0.72 to 2.24)
  Progression disease1761711.58 (1.17 to 2.11)<.01
 Locoregional progression
  No2071961
  Yes54510.94 (0.69 to 1.28).69
 Metastatic progression
  No67611
  Yes1941861.44 (1.07 to 1.93).02
Second-line chemotherapy
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan2041921
  Gemcitabine based57551.49 (1.10 to 2.02).01
*

Cox proportional hazard models used to estimate association of the parameters with overall survival. Values of P < .05 were considered statistically significant, and all tests were two-sided. 5-FU = 5-fluorouracil; CA 19-9 = carbohydrate antigen 19-9; CEA = carcinoembryonic antigen; HR = hazard ratio; CI = confidence interval; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Corresponding to prescription of morphine.

Table 3.

Prognostics factors associated with overall survival in multivariable analysis (n = 248)*

ParametersNo. of patientsNo. of eventsHR (95% CI)PInternal validation BCA HR 95%βMax scores in nomogram
Demographic parameters
 Age, y2482351.02 (1.00 to 1.04).021.00 to 1.040.0192155
 Smoking status
  Never smoker or former smoker1901781
  Current smoker58571.50 (1.09 to 2.05).011.02 to 2.140.3994621
Tumor extension at the beginning of L2
 Liver metastases
  No99931
  Yes1491422.15 (1.62 to 2.85)<.0011.48 to 2.940.7650240
Clinical parameters at the beginning of L2
 Performance status (WHO)
  047401
  11171141.61 (1.07 to 2.37)1.01 to 2.390.4787925
  ≥284813.06 (1.94 to 4.82)<.0011.75 to 4.721.1167158
 Pain
  No1181081
  Yes1301271.41 (1.06 to 1.88).020.99 to 1.850.3444218
 Jaundice
  No2101981
  Yes38371.96 (1.33 to 2.89)<.0011.19 to 2.980.6719235
 Ascites
  No2001881
  Yes48471.79 (1.26 to 2.54)<.0011.21 to 2.760.5833530
Treatment
 Duration of L1 (log value), mo2482350.48 (0.33 to 0.70)<.0010.33 to 0.81−0.73868100
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan1951841
  Gemcitabine based53511.77 (1.27 to 2.46)<.0011.18 to 2.490.5682730
ParametersNo. of patientsNo. of eventsHR (95% CI)PInternal validation BCA HR 95%βMax scores in nomogram
Demographic parameters
 Age, y2482351.02 (1.00 to 1.04).021.00 to 1.040.0192155
 Smoking status
  Never smoker or former smoker1901781
  Current smoker58571.50 (1.09 to 2.05).011.02 to 2.140.3994621
Tumor extension at the beginning of L2
 Liver metastases
  No99931
  Yes1491422.15 (1.62 to 2.85)<.0011.48 to 2.940.7650240
Clinical parameters at the beginning of L2
 Performance status (WHO)
  047401
  11171141.61 (1.07 to 2.37)1.01 to 2.390.4787925
  ≥284813.06 (1.94 to 4.82)<.0011.75 to 4.721.1167158
 Pain
  No1181081
  Yes1301271.41 (1.06 to 1.88).020.99 to 1.850.3444218
 Jaundice
  No2101981
  Yes38371.96 (1.33 to 2.89)<.0011.19 to 2.980.6719235
 Ascites
  No2001881
  Yes48471.79 (1.26 to 2.54)<.0011.21 to 2.760.5833530
Treatment
 Duration of L1 (log value), mo2482350.48 (0.33 to 0.70)<.0010.33 to 0.81−0.73868100
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan1951841
  Gemcitabine based53511.77 (1.27 to 2.46)<.0011.18 to 2.490.5682730
*

The final multivariable Cox model was obtained by entering risks factors from the univariate model that achieved P ≤ .05 as the thresholds in a single multivariable proportional hazards model. 5-FU = 5-fluorouracil; HR = hazard ratio; CI = confidence interval; BCA = accelerated bootstrap confidence interval; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Cox proportional hazard models used to estimate association of the parameters with overall survival. Values of P < .05 were considered statistically significant, and all tests were two-sided.

Corresponding to prescription of morphine.

Table 3.

Prognostics factors associated with overall survival in multivariable analysis (n = 248)*

ParametersNo. of patientsNo. of eventsHR (95% CI)PInternal validation BCA HR 95%βMax scores in nomogram
Demographic parameters
 Age, y2482351.02 (1.00 to 1.04).021.00 to 1.040.0192155
 Smoking status
  Never smoker or former smoker1901781
  Current smoker58571.50 (1.09 to 2.05).011.02 to 2.140.3994621
Tumor extension at the beginning of L2
 Liver metastases
  No99931
  Yes1491422.15 (1.62 to 2.85)<.0011.48 to 2.940.7650240
Clinical parameters at the beginning of L2
 Performance status (WHO)
  047401
  11171141.61 (1.07 to 2.37)1.01 to 2.390.4787925
  ≥284813.06 (1.94 to 4.82)<.0011.75 to 4.721.1167158
 Pain
  No1181081
  Yes1301271.41 (1.06 to 1.88).020.99 to 1.850.3444218
 Jaundice
  No2101981
  Yes38371.96 (1.33 to 2.89)<.0011.19 to 2.980.6719235
 Ascites
  No2001881
  Yes48471.79 (1.26 to 2.54)<.0011.21 to 2.760.5833530
Treatment
 Duration of L1 (log value), mo2482350.48 (0.33 to 0.70)<.0010.33 to 0.81−0.73868100
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan1951841
  Gemcitabine based53511.77 (1.27 to 2.46)<.0011.18 to 2.490.5682730
ParametersNo. of patientsNo. of eventsHR (95% CI)PInternal validation BCA HR 95%βMax scores in nomogram
Demographic parameters
 Age, y2482351.02 (1.00 to 1.04).021.00 to 1.040.0192155
 Smoking status
  Never smoker or former smoker1901781
  Current smoker58571.50 (1.09 to 2.05).011.02 to 2.140.3994621
Tumor extension at the beginning of L2
 Liver metastases
  No99931
  Yes1491422.15 (1.62 to 2.85)<.0011.48 to 2.940.7650240
Clinical parameters at the beginning of L2
 Performance status (WHO)
  047401
  11171141.61 (1.07 to 2.37)1.01 to 2.390.4787925
  ≥284813.06 (1.94 to 4.82)<.0011.75 to 4.721.1167158
 Pain
  No1181081
  Yes1301271.41 (1.06 to 1.88).020.99 to 1.850.3444218
 Jaundice
  No2101981
  Yes38371.96 (1.33 to 2.89)<.0011.19 to 2.980.6719235
 Ascites
  No2001881
  Yes48471.79 (1.26 to 2.54)<.0011.21 to 2.760.5833530
Treatment
 Duration of L1 (log value), mo2482350.48 (0.33 to 0.70)<.0010.33 to 0.81−0.73868100
 Type of L2 regimen
  5-FU ± oxaliplatin and/or irinotecan1951841
  Gemcitabine based53511.77 (1.27 to 2.46)<.0011.18 to 2.490.5682730
*

The final multivariable Cox model was obtained by entering risks factors from the univariate model that achieved P ≤ .05 as the thresholds in a single multivariable proportional hazards model. 5-FU = 5-fluorouracil; HR = hazard ratio; CI = confidence interval; BCA = accelerated bootstrap confidence interval; L1 = first-line chemotherapy; L2 = second-line chemotherapy; WHO = World Health Organization.

Cox proportional hazard models used to estimate association of the parameters with overall survival. Values of P < .05 were considered statistically significant, and all tests were two-sided.

Corresponding to prescription of morphine.

In the sensitivity analyses, associations remained unchanged in the univariate and multivariable analyses (Supplementary Tables 2–7, available online).

Performance Assessment and Internal Validation of the Final Model

The multivariable model exhibited good discrimination ability (C-index = 0.75, 95% CI = 0.72 to 0.78). The calibration plots showed an optimal agreement between model prediction and actual observation for predicting OS probability at six, 12, 18, and 24 months (Supplementary Figure 3, available online). In the internal validation, uncertainties around hazard ratio measured with a bootstrapping procedure reflected the robustness of the final model (Table 3).

Assessment in OS Discrimination Ability of the Final Model Compared With the Classical Approach

The input of six parameters identified in the final model (smoking status, liver metastases, pain, jaundice, ascites, and type of L2 regimen) in addition to those frequently used in multidisciplinary meetings (“pragmatic parameters”: age, ECOG PS, and duration of L1) (Supplementary Table 8, available online) statistically significantly improved its discrimination capacity because the C-statistic increased from 0.70 to 0.75 (difference = 0.05, 95% CI = 0.02 to 0.08).

Similarly, their addition adequately reclassified patients at lower risk of death and those at higher risk, as reported by a cNRI of 0.79 (95% CI = 0.56 to 1.0, P < .001) at six months (Supplementary Figure 4, available online), with an IDI of 0.14 (95% CI = 0.10 to 0.18, P < .001) (Supplementary Figure 5, available online).

Prognostic Nomogram and Score for OS

A nomogram integrating all statistically significant independent factors for OS was built (Figure 2). The prognostic score was based on the total number of points obtained from the nomogram. This prognostic score assumed a normal distribution (Supplementary Figure 6, available online). Based on the Cox method, patients were categorized into three risk groups (low, intermediate, and high) with median OS of 11.3 months (95% CI = 9.1 to 12.9 months), 3.6 months (95% CI = 2.6 to 4.7 months), and 1.4 months (95% CI = 1.2 to 1.7 months), respectively (P < .001) (Figure 3A). Approaches with two risk groups (high and low risk) are presented in Supplementary Figure 7 (available online). Patient characteristics in each risk group are described in Supplementary Table 9 (available online).

Figure 2.

Prognostic nomogram to predict individual overall survival probability at the beginning of second-line chemotherapy in patients with advanced pancreatic ductal adenocarcinoma. First, the points associated with each of the nine prognostic factors are obtained via upward vertical translation of the patient’s variable value to the line labeled “Points.” Next, the points are summed and the corresponding total number is reported as a dot on the line labeled “Total points.” A vertical line is then drawn downward from the total point dot to obtain the overall survival prediction at the intersection with the “6-,” “8-,” “12-,” and “24-month survival probability” and “median survival time” lines. An online, web-based, smartphone-compatible application was developed that provides individualized survival estimates from the nomogram: http://www.umqvc.org/en/tool/proscap.html. *The value in gray at the right end of the line corresponds to the maximum number of points for each factor. 5-FU = 5-fluorouracil; L1 = first-line chemotherapy; L2 = second-line chemotherapy.

Figure 3.

Kaplan-Meier curves of overall survival for three risk groups in the development cohort (A) and in the external validation cohort (B). An online, web-based, smartphone-compatible application was developed that provides risk group classification for overall survival from the prognostic score: http://www.umqvc.org/en/tool/proscap.html. Values of the log-rank test P < .05 were considered statistically significant, and all tests were two-sided. CI = confidence interval.

This prognostic score was applied to patients who did not receive an L2, after assigning to each patient a type of regimen according to the chemotherapy that could have been proposed in multidisciplinary meeting. Similarly, patients were categorized into the same three risk groups (low, intermediate, and high risk) previously identified with statistically significantly different prognostic profiles (Supplementary Figure 8, available online). Then, the risk groups were compared according to L2 administration or not (Supplementary Figure 9, available online) and described in Supplementary Table 10 (available online). The benefit of L2 was higher in better prognostic groups with a statistically significant interaction term between risk groups and L2 administration (P = .01) (Supplementary Tables 11 and 12, available online).

PFS Analysis

PFS analysis was investigated in the OS risk groups identified (low, intermediate, and high). Interestingly, the discriminative ability of the three-group model was confirmed in L2 PFS analysis (Supplementary Figure 10, available online). Approaches for the two risk groups are presented in Supplementary Figure 11 (available online).

Clinico-biological Model

The multivariable model with biological parameters was established based on patients for whom the 11 parameters were available, including two independent biological factors: neutrophil-to-lymphocyte ratio (NLR) and carbohydrate antigen 19-9 (CA19-9) (Supplementary Table 13, available online). Supplementary analyses are specified in the Supplementary Materials and provided in Supplementary Tables 14 and 15 (stratified and random approaches and multiple imputations, respectively, available online).

External Validation of the Prognostic Score

Information for the nine baseline parameters that were required for the score calculation was available for 162 (99%) patients from the external validation cohort. As expected, the external validation cohort differed from the development cohort (Supplementary Table 1, available online).

The good discrimination ability of the final model developed in the main analysis was externally confirmed (C-index = 0.63, 95% CI = 0.58 to 0.69). We identified the same risk groups with OS applying to Kaplan-Meier curves (P < .001) (Figure 3B).

Clinical Benefit Analysis

The clinical benefit–centered accuracy of the final model was confirmed by a decision curve analysis in both cohorts (Figure 4). The decision curve analysis shows a threshold of risk of death at six months at which decisions will cause greater benefit for true positives and decision of false positives will be reduced. The net benefit for decisions based on our final multivariable model for threshold values greater than 20% (Figure 4A) and 40% (Figure 4B) is better than considering patients on the same level of risk. Here the decision is defined by the possibility of not treating a patient by L2. In this context, the clinician must ensure that the patient is at high risk of death at six months and minimize false positives, that is, patients who are still alive at six months who must be treated. Overall, the decision curve shows that the net benefit for decision based on our final multivariable model is of interest, especially for thresholds greater than 0.5, the area of interest.

Figure 4.

Decision curves to plot the net benefit achieved by making clinical decisions based on the final multivariable model risk of death predictions at six months in the development cohort (A) and in the external validation cohort (B).

Discussion

PDAC is a highly aggressive cancer, and chemotherapy remains the cornerstone of advanced disease therapy. While the clinical indications for L1 are well described and consensual (8), decision-making regarding L2 is a much more controversial issue. OS improvement with L2 has been reported in patients with gemcitabine-refractory PDAC (25,26). Nevertheless, the analysis of 34 clinical trials comparing L2 with best supportive care showed that the benefit remains modest and limited to selected patients (15). Moreover, because of a lack of high-level evidence, no guidelines are available for L2 after FOLFIRINOX (6) or gemcitabine plus nab-paclitaxel L1 regimens (7).

A prognostic model is a useful tool for clinical management by predicting patient life expectancy. The purpose of implementing such a scoring system, using easily measurable clinical parameters, is to guide clinical decision-making in routine practice and to propose patient risk stratification for clinical trials. Currently, no validated prognostic model is available to estimate OS in patients with aPDAC at the beginning of L2. Some scores have been proposed but failed to complete validation process because of small sample sizes with insufficient statistical power (27,28). More recently, Sinn et al. (14) developed a score based on prognostic factors including ECOG-PS, CA19-9, and duration of L1, but the performance and internal validation of the final model were not assessed.

From a large population-based study, we established a new straightforward prognostic model. We identified prognostic factors for survival of patients with aPDAC who received L2 and derived a prognostic model based on nine easily available clinical parameters. Of note, the administration of a 5-FU ± oxaliplatin and/or irinotecan regimen in L2 is statistically significantly associated with longer overall survival than a gemcitabine- based regimen, regardless of the type of L1 initially received. However, the aim of this study was not to compare treatment response, and the superiority of the 5-FU ± oxaliplatin and/or irinotecan regimen is probably multifactorial, involving patient-, tumor-, and treatment-related factors, and should be considered an exploratory finding warranting evaluation in further dedicated studies. Smoking status is a factor associated with an increased risk of PDAC (29), and here we demonstrate that it is also a negative prognostic factor (30). In the clinico-biological model, two parameters were identified, CA19-9 and NLR, whose relevance was previously demonstrated in the L1 setting (31,32). We developed a robust prognostic nomogram for individual estimation of patient survival. In addition, the prognostic score allows classifying patients into three risk groups. Both tools are available in an open access version on a smartphone-compatible website (http://www.umqvc.org/en/tool/proscap.html).

Our results highlight considerable heterogeneity in survival in patients with PDAC who receive L2, and our prognostic score may help to refine patient selection for L2. Even if “pragmatic parameters” are already frequently used for decision-making in multidisciplinary meeting, our final model improves the discrimination ability compared with this classical approach. Interestingly, the risk groups identified based on OS matched with the risk groups for PFS. Therefore, through the PFS analysis, the prognostic value of our scoring system was confirmed independently from the “background noise” of the eventual following lines of chemotherapy for both patient-related (OS) and tumor-related (PFS) outcomes. Overall, we confirmed the survival benefit of L2 for all patients with aPDAC, although a decreasing clinical benefit was observed from low- to high-risk groups. In addition to strengthening decision-making for clinicians, these tools may be beneficial for better selection of patients for treatment, for stratified random assignment to ensure well-balanced treatment groups, and for a potential optimization of clinical trial design. The development of risk-adapted strategies for aPDAC second-line management in the future could be also considered in the different risk groups identified by the score.

Our study has several strengths. We evaluated a large population including patients treated with FOLFIRINOX regimen in both cohorts. Parameters used in the model are clinically relevant, simple to collect for clinicians, and consistent with previous studies. We built our model in a rigorous methodological framework, using a recent recommended checklist for nomograms (33) and providing transparent reporting of the multivariable model as suggested in the TRIPOD statement (34). Moreover, discrimination, calibration, and internal validation demonstrated the satisfactory performance and validity of the model. Our prognostic score was externally replicated in routine clinical practice in three other institutions. The reproducibility of the results observed in the development cohort in a clinically different external validation cohort reinforce their generalizability. The clinical benefit–centered accuracy of the final model was confirmed by the decision curve analysis.

The inclusion period in our study was long; meanwhile, clinical practices have changed with the approval of the FOLFIRINOX and gemcitabine plus nab-paclitaxel chemotherapy. This highlights the reliability of our model in the era of these new treatment regimens and not only in gemcitabine-refractory patients. One limitation of our model is the heterogeneity in L1 treatment regimens. It would be of interest to assess similar prognostic tools in homogenous patient groups for L1. Another limitation of the study is the absence of patients treated with nab-paclitaxel in the development cohort, as well as the presence of only six patients who received this treatment in the validation cohort. An external validation in different countries will definitively confirm the worldwide relevance of the model. Moreover, the predictive value of the score in terms of the benefit of L2 was not explored in the external validation cohort. Furthermore, in the development cohort, the untreated patient group was too small and with a high level of missing data to propose a specific tool in this population. Additional variables, particularly albumin (28,35) or serum C-reactive protein levels (27,36), could not be evaluated in our study because of the retrospective nature of the data collection, with a high rate of missing data, and may be relevant to assess in further studies. Accumulating evidence for the prognostic value of health-related quality of life (HRQoL) for OS prediction in metastatic pancreatic cancer (37,38) and in other tumor types (39–41) suggests that HRQoL should also be evaluated in the future models for aPDAC. The less discriminatory ability of the model in the external validation cohort may be improved by additional inputs in the future (biological data, HRQoL).

In conclusion, we propose a novel OS prediction model based on nine key independent clinical prognostic factors at the beginning of L2 in patients with aPDAC. Our study highlights considerable heterogeneity in the survival of patients who receive L2. We identify three distinct risk groups in which chemotherapy was found to be statistically significantly beneficial but with unequal benefit magnitude across risk groups. This study allowed us to develop and externally validate a prognostic model for OS and provide functional tools at individual (nomogram) and population (score) levels. These tools can accurately predict OS before administration of L2 and thereby improve risk stratification and patient selection for routine clinical decision-making and future clinical trials.

Notes

We thank Florian Limousin (graphic designer and independent web developer in Pearlweb society) for the web development application support.

Author’s contributions: conception and design: AV, AM, FB, CB, DV; collection and assembly of data: all authors; data analysis and interpretation: AV, CN, CB, DV; manuscript writing: all authors; final approval of manuscript: all authors.

The authors declare no potential conflicts of interest.

References

1

Siegel
RL
,
Miller
KD
,
Jemal
A.
Cancer statistics, 2016
.
CA Cancer J Clin.
2016
;
66
1
:
7
30
.

2

Rahib
L
,
Smith
BD
,
Aizenberg
R
,
Rosenzweig
AB
,
Fleshman
JM
,
Matrisian
LM.
Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States
.
Cancer Res.
2014
;
74
11
:
2913
2921
.

3

National Cancer Institute
. SEER Cancer Statistics Review, 1975–2013. http://seer.cancer.gov/csr/1975_2013/. Accessed June 13, 2016.

4

Ryan
DP
,
Hong
TS
,
Bardeesy
N.
Pancreatic adenocarcinoma
.
N Engl J Med.
2014
;
371
11
:
1039
1049
.

5

Neuzillet
C
,
Tijeras-Raballand
A
,
Bourget
P
, et al. 
State of the art and future directions of pancreatic ductal adenocarcinoma therapy
.
Pharmacol Ther.
2015
;
155
:
80
104
.

6

Conroy
T
,
Desseigne
F
,
Ychou
M
, et al. 
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med.
2011
;
364
19
:
1817
1825
.

7

Von Hoff
DD
,
Ervin
T
,
Arena
FP
, et al. 
Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine
.
N Engl J Med.
2013
;
369
18
:
1691
1703
.

8

Ducreux
M
,
Cuhna
AS
,
Caramella
C
, et al. 
Cancer of the pancreas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up
.
Ann Oncol.
2015
;
26
(
suppl 5
):
v56
v68
.

9

Sohal
DPS
,
Mangu
PB
,
Khorana
AA
, et al. 
Metastatic pancreatic cancer: American Society of Clinical Oncology clinical practice guideline
.
J Clin Oncol.
2016
;
34
23
:
2784
2796
.

10

Walker
EJ.
Beyond first-line chemotherapy for advanced pancreatic cancer: An expanding array of therapeutic options?
World J Gastroenterol.
2014
;
20
9
:
2224
2236
.

11

Oettle
H
,
Riess
H
,
Stieler
JM
, et al. 
Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: Outcomes from the CONKO-003 trial
.
J Clin Oncol.
2014
;
32
23
:
2423
2429
.

12

Wang-Gillam
A
,
Li
C-P
,
Bodoky
G
, 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
10018
:
545
557
.

13

Portal
A
,
Pernot
S
,
Tougeron
D
, et al. 
Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: An AGEO prospective multicentre cohort
.
Br J Cancer.
2015
;
113
7
:
989
995
.

14

Sinn
M
,
Dälken
L
,
Striefler
JK
, et al. 
Second-line treatment in pancreatic cancer patients: Who profits?—Results from the CONKO study group
.
Pancreas.
2016
;
45
4
:
601
605
.

15

Rahma
OE
,
Duffy
A
,
Liewehr
DJ
,
Steinberg
SM
,
Greten
TF.
Second-line treatment in advanced pancreatic cancer: A comprehensive analysis of published clinical trials
.
Ann Oncol.
2013
;
24
8
:
1972
1979
.

16

Schemper
M
,
Smith
TL.
A note on quantifying follow-up in studies of failure time
.
Control Clin Trials.
1996
;
17
4
:
343
346
.

17

Royston
P
,
Altman
DG.
Regression using fractional polynomials of continuous covariates: Parsimonious parametric modelling
.
Appl Stat.
1994
;
43
3
:
429
.

18

Royston
P
,
Sauerbrei
W.
Multivariable Model-Building: A Pragmatic Approach to Regression Anaylsis Based on Fractional Polynomials for Modelling Continuous Variables.
Wiley;
2008
.

19

Moons
KGM
,
Altman
DG
,
Reitsma
JB
, et al. 
Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD): Explanation and elaboration
.
Ann Intern Med.
2015
;
162
1
:
W1
.

20

Harrell
FE
,
Lee
KL
,
Mark
DB.
Multivariable prognostic models: Issues in developing models, evaluating assumptions and adequacy, and mesuaring and reducing errors
.
Stat Med.
1996
;
15
4
:
361
387
.

21

Pencina
MJ
,
Steyerberg
EW
,
D’Agostino
RB.
Net reclassification index at event rate: Properties and relationships: Net Reclassification Index at Event Rate
.
Stat Med.
2016
. http://doi.wiley.com/10.1002/sim.7041 (accessed November 1, 2016).

22

Pencina
MJ
,
D’Agostino
RB
,
Vasan
RS.
Statistical methods for assessment of added usefulness of new biomarkers
.
Clin Chem Lab Med.
2010
;
48
12
:
1703
1711
.

23

Cox
DR.
Note on grouping
.
J Am Stat Assoc.
1957
;
52
280
:
543
547
.

24

Fitzgerald
M
,
Saville
BR
,
Lewis
RJ.
Decision curve analysis
.
JAMA.
2015
;
313
4
:
409
.

25

Pelzer
U
,
Schwaner
I
,
Stieler
J
, et al. 
Best supportive care (BSC) versus oxaliplatin, folinic acid and 5-fluorouracil (OFF) plus BSC in patients for second-line advanced pancreatic cancer: A phase III-study from the German CONKO-study group
.
Eur J Cancer.
2011
;
47
11
:
1676
1681
.

26

Chiorean
EG
,
Von Hoff
DD
,
Tabernero
J
, et al. 
Second-line therapy after nab-paclitaxel plus gemcitabine or after gemcitabine for patients with metastatic pancreatic cancer
.
Br J Cancer.
2016
;
115
2
:
188
194
.

27

Nakachi
K
,
Furuse
J
,
Ishii
H
,
Suzuki
E
,
Yoshino
M.
Prognostic factors in patients with gemcitabine-refractory pancreatic cancer
.
Jpn J Clin Oncol.
2007
;
37
2
:
114
120
.

28

Kim
ST
,
Choi
YJ
,
Park
KH
, et al. 
A prognostic model to identify patients with advanced pancreas adenocarcinoma who could benefit from second-line chemotherapy
.
Clin Oncol.
2012
;
24
2
:
105
111
.

29

Bosetti
C
,
Lucenteforte
E
,
Silverman
DT
, et al. 
Cigarette smoking and pancreatic cancer: An analysis from the International Pancreatic Cancer Case-Control Consortium (Panc4)
.
Ann Oncol.
2012
;
23
7
:
1880
1888
.

30

Kawaguchi
C
,
Sho
M
,
Tanaka
T
, et al. 
Impact of smoking on pancreatic cancer patients receiving current chemotherapy:
Pancreas.
2015
;
44
7
:
1155
1160
.

31

Goldstein
D
,
El-Maraghi
RH
,
Hammel
P
, et al. 
nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: Long-term survival from a phase III trial
.
J Natl Cancer Inst.
2015
;
107
2
:dju413.

32

Park
HS
,
Lee
HS
,
Park
JS
, et al. 
Prognostic scoring index for patients with metastatic pancreatic adenocarcinoma
.
Cancer Res Treat.
2016
;
48
4
:
1253
1263
.

33

Balachandran
VP
,
Gonen
M
,
Smith
JJ
,
DeMatteo
RP.
Nomograms in oncology: More than meets the eye
.
Lancet Oncol.
2015
;
16
4
:
e173
e180
.

34

Collins
GS
,
Reitsma
JB
,
Altman
DG
,
Moons
KGM.
Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD): The TRIPOD statement
.
Br J Cancer.
2015
;
112
2
:
251
259
.

35

Vernerey
D
,
Huguet
F
,
Vienot
A
, et al. 
Prognostic nomogram and score to predict overall survival in locally advanced untreated pancreatic cancer (PROLAP)
.
Br J Cancer.
2016
;
115
3
:
281
289
.

36

Hurwitz
HI
,
Uppal
N
,
Wagner
SA
, et al. 
Randomized, double-blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed
.
J Clin Oncol.
2015
;
33
34
:
4039
4047
.

37

Gourgou-Bourgade
S
,
Bascoul-Mollevi
C
,
Desseigne
F
, et al. 
Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: Results from the PRODIGE 4/ACCORD 11 randomized trial
.
J Clin Oncol.
2013
;
31
1
:
23
29
.

38

Diouf
M
,
Filleron
T
,
Pointet
A-L
, et al. 
Prognostic value of health-related quality of life in patients with metastatic pancreatic adenocarcinoma: A random forest methodology
.
Qual Life Res.
2016
;
25
7
:
1713
1723
.

39

Diouf
M
,
Chibaudel
B
,
Filleron
T
, et al. 
Could baseline health-related quality of life (QoL) predict overall survival in metastatic colorectal cancer? The results of the GERCOR OPTIMOX 1 study
.
Health Qual Life Outcomes.
2014
;
12
1
:
69
.

40

Fiteni
F
,
Vernerey
D
,
Bonnetain
F
, et al. 
Prognostic value of health-related quality of life for overall survival in elderly non-small-cell lung cancer patients
.
Eur J Cancer.
2016
;
52
:
120
128
.

41

Paquette
B
,
Vernerey
D
,
Chauffert
B
, et al. 
Prognostic value of health-related quality of life for death risk stratification in patients with unresectable glioblastoma
.
Cancer Med.
2016
;
5
8
:
1753
1764
.

Author notes

*

Authors contributed equally to this work.

Supplementary data