Background:

IL6 and YKL-40 (also known as chitinase 3-like 1 protein, CHI3L1) are produced by pancreatic cancer cells and macrophages and activate inflammation. C-reactive protein (CRP) is synthesized mainly in hepatic cells and primarily stimulated by IL6. The aim of this study was to determine the prognostic value of combined detection of serum IL6, YKL-40, and CRP in patients with pancreatic cancer receiving palliative chemotherapy.

Methods:

In all, 592 patients with unresectable pancreatic cancer from five hospitals in Denmark were included in the BIOPAC biomarker study between 2008 and 2017. Pretreatment and longitudinal serum values of IL6 and YKL-40 were determined. Baseline CRP and CA19-9 values were available for the whole cohort. Patients were dichotomized as low versus high using cutoffs for IL6 of >4.92 pg/mL, YKL-40 of >95% age-corrected percentile, and CRP of >10 mg/L. The main outcome was overall survival.

Results:

Combined elevations of serum IL6, YKL-40, and CRP were associated with worse survival in contrast to an isolated high concentration of a single marker. Serum IL6, YKL-40, and CRP were higher in patients with advanced stage disease and in patients with poor performance status. Higher IL6 and YKL-40 levels at the time of tumor progression and serum IL6 measured over time were associated with shorter overall survival.

Conclusions:

Combined high baseline serum levels of IL6, YKL-40, and CRP are associated with poor survival.

Impact:

Assessment of systemic inflammation via measurements of IL6, YKL-40, and CRP may be important for pancreatic cancer prognostication.

Pancreatic cancer is characterized by lack of early symptoms, advanced stage at the time of diagnosis, resistance to chemotherapy, and a disadvantageous tumor microenvironment due to an excess of stroma, poor vascularity, and a high grade of inflammation and immunosuppression. Pancreatic cancer's aggressive biology, genetic complexity, rapid clinical deterioration, and absence of reliable robust biomarkers yield poor survival (1). Serum cancer antigen 19-9 (CA 19-9) is the only recommended biomarker in the routine management of pancreatic cancer; however, it is limited in use due to low sensitivity and specificity. No prognostic biomarkers have yet been definitively validated in clinical practice in pancreatic cancer.

Tumor-promoting inflammation, which was first hypothesized by Rudolf Virchow in 1863 (2) and later framed into one of the classical hallmarks of the cancer concept (3), is a critical component of pancreatic cancer development and progression and markedly contributes to tumor aggressiveness and chemotherapy resistance. Activation of the systemic inflammatory response and elevated inflammatory markers, for example, C-reactive protein (CRP) and albumin (Glasgow Prognostic Score), white blood cell counts (WBC) especially absolute neutrophil count (ANC), platelet counts, and neutrophil lymphocyte and platelet lymphocyte ratios correlate with poor survival (4–6).

Risk of pancreatic cancer is associated with a variety of inflammatory conditions including smoking, high body mass index (BMI), chronic pancreatitis, heavy alcohol use, inflammatory bowel disease, and long-standing diabetes mellitus (7). Activation of inflammation is largely dependent on IL6 (8, 9), which is one of the proinflammatory cytokines secreted by activated macrophages, lymphocytes, and endothelial and tumor cells (10, 11). IL6 cytokine is a multifunctional signaling protein that facilitates the inflammation cascade and key pathways and processes within the respective tumor microenvironment, among others such as regulation of stromal desmoplasia, promotion of tumor-induced immunosuppression, and facilitation of metastasis (11, 12). Debilitating features (e.g., lack of appetite, weight loss, fatigue, anemia, fever, pain, inactivity, negative mood, cachexia), which characterize behavior in many patients with pancreatic cancer, are also partly associated with IL6 cytokine activation (13, 14). IL6 levels are elevated in the majority of patients with metastatic cancer (15). However, prospectively tested IL6 and CRP were not associated with pancreatic cancer risk (16, 17).

Another marker with a role in inflammation is YKL-40 (also known as chitinase 3-like 1 protein, CHI3L1), a member of the mammalian chitinase-like proteins, and is a highly conserved glycoprotein. YKL-40 is mainly produced by cancer cells, macrophages, and neutrophils (18–21). YKL-40 elevation is exponentially age dependent (22), possibly due to an increase in age-related inflammation. YKL-40 is elevated in patients with inflammatory disorders (22) and also in subgroups of patients with many cancer types, including up to 80% in patients with pancreatic cancer (19). Furthermore, YKL-40 regulates VEGF and angiogenesis (23), cell proliferation, and differentiation (24); re-modulates extracellular matrix (21); activates Akt signaling (25); protects against apoptosis (26); and promotes metastases (27) and tumor progression (18, 20, 21, 28, 29).

Because of unfavorable prognosis of pancreatic cancer, the lack of effective treatment, and biomarkers to predict disease outcome along with association with inflammation, we explored the prognostic value of the combined detection of serum IL6, YKL-40, and CRP as potential prognostic marker in patients with pancreatic cancer receiving palliative chemotherapy.

Study population and blood samples

We analyzed 1,899 serum samples from 592 consecutive patients with unresectable pancreatic cancer undergoing palliative chemotherapy who were included in the BIOPAC study (NCT03311776) from five hospitals in Denmark in the period from July 1, 2008 until December 31, 2017. Blood samples and clinical data at the time of inclusion were collected prospectively. Blood samples were collected at the initiation of palliative chemotherapy, before the second cycle, and at each timepoint of CT evaluation during treatment until cancer progression. Samples were processed according to the nationally approved standard operating procedures for blood samples (www.herlevhospital.dk/BIOPAC; ref. 15s). IL6 and YKL-40 analyses were performed prospectively and blinded to clinical data. WBC, ANC, and platelet counts as well as the majority of CRP determinations were obtained as part of routine care. CA 19-9 as the most frequently used biomarker for pancreatic cancer was measured (Siemens) in all included patients.

Serum IL6 analysis

Pretreatment and longitudinal (before the second cycle and at timepoint of first CT evaluation) serum concentrations of IL6 were determined by a commercially available human IL6 high-sensitive ELISA (Quantikine HS600B, R&D Systems) in accordance with the manufacturer's instructions. The minimal detectable limit for IL6 was 0.01 pg/mL. The intra-assay coefficient of variation (CV) was ≤8% and the interassay CV was ≤11% (30). Patients were grouped as having low or high values dichotomized using a cutoff for IL6 of >4.92 pg/mL, the 95th percentile in healthy blood donors (30).

Serum YKL-40 analysis

Pretreatment and longitudinal (before the second cycle and at timepoint of first CT evaluation) serum concentrations of YKL-40 were determined by a commercial ELISA (Quidel). The minimal detectable limit for YKL-40 was 10 μg/L. The intra-assay CV was 5% and the interassay CV was <6% (19). Elevated serum YKL-40 was defined as higher than the age-corrected 95th percentile (22).

CRP analysis

In the majority of patients, pretreatment serum CRP concentration was determined as a part of routine blood tests according to the manufacturer's instructions, using a previously validated Sentinel CRP Ultra ready-to-use, liquid assay reagent by an immunoturbidimetric method on a fully automated chemistry analyzer [Kit-test SENTINEL CRP Ultra (UD), 11508 UD-2.0/02 2015/09/23]. The measurement range for CRP is 0.3–640 mg/L, with an intra-assay CV of 3% and an interassay CV of <15%. Elevated serum CRP was conventionally defined as higher than 10 mg/L (4–6).

Statistical analysis

Results are reported in accordance with the REMARK (Reporting Recommendations for Tumor Marker Prognostic Studies) guidelines (31). Descriptive statistics were performed to describe patient demographics and baseline clinical characteristics. Spearman's correlation coefficient rank test was used to examine the inter-relationships between the median serum levels of IL6, YKL-40, and CRP and the relation of these levels to CA19-9, WBC, ANC, platelet counts, bilirubin, alkaline phosphatase (ALP), and alanine transaminase (ALT).

The outcome was 3-year overall survival (OS); thus, the patients were followed up from date of inclusion to the BIOPAC study until death from any cause, or the cohort censor date (August 3, 2018), or 3 years after date of inclusion, whichever came first. The levels of baseline serum IL6, YKL-40, and CRP were modeled by the dichotomized value using the actual cut-off point of the biomarker as described under “Materials and Methods” section. Graphical presentation using Kaplan–Meier estimates of OS was shown by grouping patients according to low versus high serum IL6, YKL-40, and CRP levels and with inclusion of the combined values of these markers. HRs and 95% confidence intervals (CI) were estimated with Cox proportional hazards regression. Variables with statistically significant association on univariate analysis were included in multivariable models.

In addition, longitudinally measured biomarkers (IL6 and YKL-40) before the second cycle and at timepoint of first CT evaluation were analyzed as time-dependent variables in a Cox model adjusted for the significant terms from the baseline analysis. Each biomarker was analyzed by including the log2-transformed baseline value as well as the log2-transformed ratio between latest value and the baseline value corresponding to HRs representing the effect of doubling the baseline value.

Finally, to evaluate the biomarkers ability to predict early death (<90 days), analysis by the ROC curve by the area under the curve (AUC) was performed. In addition, the associations between biomarkers and early death were analyzed using logistic regression.

Statistical analyses were conducted using the latest version of R (version 3.2.4). In all analyses, the significance level was set to 5%.

Characteristics of study population

Between July 2008 and December 2017, a total of 592 consecutive patients with unresectable pancreatic cancer undergoing palliative chemotherapy were registered in the BIOPAC database. The 592 patients represent 20% of all patients with unresectable pancreatic cancer treated with chemotherapy during this period in Denmark. The median follow-up was 6.7 months, with no loss to follow-up.

Patient and disease characteristics, treatment modalities, and baseline blood test results are given in Table 1. The median age at inclusion was 68 years (interquartile range, 62–73 years). The cohort consisted of more men (53%) than women (47%). The majority of patients (303/51.2%) were classified with an Eastern Cooperative Oncology Group (ECOG) Performance Status (PS) of 1, while 200 (33.8%) and 88 (14.9%) were registered with a PS 0 and 2, respectively. According to the WHO classification of BMI subgroups, 51 patients (8.6%) were underweight (BMI < 18.5), 341 patients (57.6%) were of normal weight BMI from 18.5 to 25), and 177 patients (29.9%) were overweight (BMI > 25). The majority of patients (324/54.7%) had lost more than 5% of their body weight before inclusion in the study. Most patients were diagnosed with stage IV cancer (72.3%), 82.4% of whom had liver metastases, and 37.3% had peritoneal carcinomatosis. All patients were given first-line chemotherapy as follows: gemcitabine n = 413 (69.8%), FOLFIRINOX n = 113 (19.1%), gemcitabine + nab-Paclitaxel n = 52 (8.8%), or capecitabine-containing regimens n = 14 (2.4%). One hundred forty-eight patients (25%) received second-line chemotherapy.

Table 1.

Clinical characteristics and baseline blood test results in 592 patients with unresectable pancreatic cancer.

CharacteristicNumber (%)
Median age, (IQR) years 68.00 (62.00–73.00) 
 71–89 years 226 (38.2) 
Gender, female 278 (47.0) 
ECOG PS 
 0 200 (33.8) 
 1 303 (51.2) 
 2 88 (14.9) 
 3 1 (0.2) 
BMI 
 Normal weight 341 (57.6) 
 Underweight 51 (8.6) 
 Overweight 177 (29.9) 
 NA 23 (3.9) 
Weight loss 
 <2% 116 (19.6) 
 ≥2% 327 (55.2) 
 NA 149 (25.2) 
Diabetes 139 (23.5) 
Smoking 
 No 161 (27.2) 
 Yes 362 (61.1) 
 NA 69 (11.7) 
Alcohol 
 No 389 (65.7) 
 Yes 132 (22.3) 
 NA 71 (12.0) 
Stage 
 II and III 162 (27.4) 
 IV 428 (72.3) 
 NA 2 (0.3) 
Number of metastatic sites 
 0 163 (27.5) 
 1 302 (51.0) 
 2 77 (13.0) 
 3+ 12 (2.0) 
 NA 38 (6.4) 
Metastatic sites 
 Liver 488 (82.4) 
 Peritoneum 221 (37.3) 
Chemotherapy type, first-line 
 Gemcitabine 413 (69.8) 
 Platinum-based combination 113 (19.1) 
 Gemcitabine plus nab-paclitaxel 52 (8.8) 
 Capecitabine-based combination 14 (2.4) 
Median CA19-9 (IQR) U/mL 1,000.00 (132.00–7,285.00) 
 <59 × ULN 325 (54.9) 
 ≥59 × ULN 215 (36.3) 
 NA 52 (8.8) 
Median WBC (IQR; × 109/L) 8.70 (6.80–10.80) 
Median ANC (IQR; × 109/L) 6.20 (4.68–8.10) 
Median platelets (IQR; × 109/L) 301 (232–392) 
lL6 median (IQR) pg/mL 6.45 (3.20–14.85) 
 >4.92 (high) 355 (60.0) 
 ≤4.92 (low) 237 (40.0) 
CRP median (IQR) mg/L 15.65 (3.61–48.18) 
 >10 (high) 339 (57.3) 
 ≤10 (low) 253 (42.7) 
YKL-40 median (IQR) μg/L 138.00 (78.50–233.50) 
 >95th age percentile (high) 224 (37.8) 
 ≤95th age percentile (low) 367 (62.0) 
Low IL6, YKL-40, and CRP (type 0) 152 (25.7) 
High IL6 only (type I) 39 (6.6) 
High YKL-40 only (type I) 29 (4.9) 
High CRP only (type I) 55 (9.3) 
High IL6 and YKL-40, or IL6 and CRP, or YKL-40 and CRP (type II) 154 (26.0) 
High IL6, YKL-40, and CRP (type III) 162 (27.4) 
CharacteristicNumber (%)
Median age, (IQR) years 68.00 (62.00–73.00) 
 71–89 years 226 (38.2) 
Gender, female 278 (47.0) 
ECOG PS 
 0 200 (33.8) 
 1 303 (51.2) 
 2 88 (14.9) 
 3 1 (0.2) 
BMI 
 Normal weight 341 (57.6) 
 Underweight 51 (8.6) 
 Overweight 177 (29.9) 
 NA 23 (3.9) 
Weight loss 
 <2% 116 (19.6) 
 ≥2% 327 (55.2) 
 NA 149 (25.2) 
Diabetes 139 (23.5) 
Smoking 
 No 161 (27.2) 
 Yes 362 (61.1) 
 NA 69 (11.7) 
Alcohol 
 No 389 (65.7) 
 Yes 132 (22.3) 
 NA 71 (12.0) 
Stage 
 II and III 162 (27.4) 
 IV 428 (72.3) 
 NA 2 (0.3) 
Number of metastatic sites 
 0 163 (27.5) 
 1 302 (51.0) 
 2 77 (13.0) 
 3+ 12 (2.0) 
 NA 38 (6.4) 
Metastatic sites 
 Liver 488 (82.4) 
 Peritoneum 221 (37.3) 
Chemotherapy type, first-line 
 Gemcitabine 413 (69.8) 
 Platinum-based combination 113 (19.1) 
 Gemcitabine plus nab-paclitaxel 52 (8.8) 
 Capecitabine-based combination 14 (2.4) 
Median CA19-9 (IQR) U/mL 1,000.00 (132.00–7,285.00) 
 <59 × ULN 325 (54.9) 
 ≥59 × ULN 215 (36.3) 
 NA 52 (8.8) 
Median WBC (IQR; × 109/L) 8.70 (6.80–10.80) 
Median ANC (IQR; × 109/L) 6.20 (4.68–8.10) 
Median platelets (IQR; × 109/L) 301 (232–392) 
lL6 median (IQR) pg/mL 6.45 (3.20–14.85) 
 >4.92 (high) 355 (60.0) 
 ≤4.92 (low) 237 (40.0) 
CRP median (IQR) mg/L 15.65 (3.61–48.18) 
 >10 (high) 339 (57.3) 
 ≤10 (low) 253 (42.7) 
YKL-40 median (IQR) μg/L 138.00 (78.50–233.50) 
 >95th age percentile (high) 224 (37.8) 
 ≤95th age percentile (low) 367 (62.0) 
Low IL6, YKL-40, and CRP (type 0) 152 (25.7) 
High IL6 only (type I) 39 (6.6) 
High YKL-40 only (type I) 29 (4.9) 
High CRP only (type I) 55 (9.3) 
High IL6 and YKL-40, or IL6 and CRP, or YKL-40 and CRP (type II) 154 (26.0) 
High IL6, YKL-40, and CRP (type III) 162 (27.4) 

Note: Baseline serum values of IL6, CRP, and YKL-40 were dichotomized using cutoffs for IL6 of >4.92 pg/mL, CRP mg/L of >10, and YKL-40 of >95% age-corrected percentile as reference. Patients with low values of IL6, YKL-40, and CRP (type 0); high value of one (type I) or two biomarkers (type II); or high values of all three biomarkers (type III). The baseline concentrations of IL6, CRP, and YKL-40 were available for all patients except one with a missing YKL-40 value.

Abbreviations: IQR, interquartile range; NA, not available; ULN, upper limit of normal.

One hundred sixty-two (27.4%) patients had concomitant high values of serum IL6, CRP, and YKL at the time of inclusion (type III). High values of two biomarkers were observed in 154 (26.0%) patients (type II). One hundred twenty-three (20.8%) patients, including 39 (high IL6), 55 (high CRP), and 29 (high YKL-40) patients had an isolated high concentration of a single marker (type I). One hundred fifty-two (25.7%) patients had low serum values of all three biomarkers (type 0). Slightly higher prevalence of patients with stage II–III and PS 0 was observed in this group (Supplementary Table S1). More than 50% of the patients had high IL6 or CRP levels based on the described cutoffs at baseline and 38% of patients had high YKL-40.

Comparison of median serum IL6, CRP, and YKL-40 by stage, PS, and presence of liver metastases and peritoneal carcinomatosis

IL6, CRP, and YKL-40 levels increased in accordance with increasing stage and worsening PS (Table 2). In contrast to CRP [8.86 mg/L (3.00–21.50) and 17.00 mg/L (4.00–47.94), P = 0.01 for patients without versus with metastases, respectively], no difference was found in IL6 and YKL-40 values stratified by presence of liver metastases. Baseline levels of IL6, CRP, and YKL-40 for patients with peritoneal carcinomatosis were lower compared with those diagnosed with liver metastasis.

Table 2.

Baseline median serum levels of IL6, CRP, and YKL-40 stratified by stage, performance status, and presence of liver metastases.

Baseline serum IL6, CRP, and YKL-40 stratified by stage
StageII and IIIIVP
Number of patients 162 428  
IL6 median (IQR) pg/mL 4.90 (2.60–9.30) 7.45 (3.40–17.92) <0.001 
CRP median (IQR) mg/L 7.50 (3.00–30.00) 19.00 (5.00–59.50) <0.001 
YKL-40 median (IQR) μg/L 107.00 (67.25–173.50) 151.00 (85.00–255.00) <0.001 
Baseline serum IL6, CRP, and YKL-40 stratified by PS 
PS 0 1 2 3 P 
Number of patients 200 303 88  
IL6 median (IQR) pg/mL 5.60 (2.40–11.72) 6.90 (3.45–14.00) 10.50 (4.20–21.27) 44.00 (44.00–44.00) <0.001 
CRP median (IQR) mg/L 11.00 (3.00–38.25) 18.00 (4.00–49.00) 27.89 (7.90–76.27) 177.00 (177.00–177.00) 0.001 
YKL-40 median (IQR) μg/L 109.00 (67.75–202.75) 148.00 (85.25–232.25) 167.00 (100.00–299.25) 1,474.00 (1,474.00–1,474.00) <0.001 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of liver metastases 
Liver metastases No Yes P 
Number of patients 66 488  
IL6 median (IQR) pg/mL 6.25 (2.62–12.70) 6.40 (3.20–14.33) 0.339 
CRP median (IQR) mg/L 8.86 (3.00–21.50) 17.00 (4.00–47.94) 0.011 
YKL-40 median (IQR) μg/L 144.00 (82.25–225.00) 137.00 (79.00–228.00) 0.870 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of peritoneal carcinomatosis 
Peritoneal carcinomatosis No Yes P 
Number of patients 333 221  
IL6 median (IQR) pg/mL 7.40 (3.50–18.00) 5.00 (2.60–10.30) <0.001 
CRP median (IQR) mg/L 21.00 (5.00–63.37) 10.00 (3.00–32.00) <0.001 
YKL-40 median (IQR) μg/L 148.50 (84.00–255.25) 125.00 (72.00–186.00) 0.003 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of metastasis in: liver versus peritoneum versus both versus other 
PS Liver Peritoneum Both Other P 
Number of patients 304 37 184 29  
IL6 median (IQR) pg/mL 7.50 (3.50–18.00) 6.40 (2.70–13.00) 5.00 (2.60–9.93) 6.10 (2.40–11.00) <0.001 
CRP median (IQR) mg/L 23.00 (6.00–65.24) 9.00 (3.00–24.00) 10.00 (3.00–32.55) 8.71 (3.00–18.36) <0.001 
YKL-40 median (IQR) μg/L 146.00 (84.50–262.50) 132.00 (83.00–201.00) 119.00 (70.50–185.25) 206.00 (74.00–227.00) 0.025 
Baseline serum IL6, CRP, and YKL-40 stratified by stage
StageII and IIIIVP
Number of patients 162 428  
IL6 median (IQR) pg/mL 4.90 (2.60–9.30) 7.45 (3.40–17.92) <0.001 
CRP median (IQR) mg/L 7.50 (3.00–30.00) 19.00 (5.00–59.50) <0.001 
YKL-40 median (IQR) μg/L 107.00 (67.25–173.50) 151.00 (85.00–255.00) <0.001 
Baseline serum IL6, CRP, and YKL-40 stratified by PS 
PS 0 1 2 3 P 
Number of patients 200 303 88  
IL6 median (IQR) pg/mL 5.60 (2.40–11.72) 6.90 (3.45–14.00) 10.50 (4.20–21.27) 44.00 (44.00–44.00) <0.001 
CRP median (IQR) mg/L 11.00 (3.00–38.25) 18.00 (4.00–49.00) 27.89 (7.90–76.27) 177.00 (177.00–177.00) 0.001 
YKL-40 median (IQR) μg/L 109.00 (67.75–202.75) 148.00 (85.25–232.25) 167.00 (100.00–299.25) 1,474.00 (1,474.00–1,474.00) <0.001 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of liver metastases 
Liver metastases No Yes P 
Number of patients 66 488  
IL6 median (IQR) pg/mL 6.25 (2.62–12.70) 6.40 (3.20–14.33) 0.339 
CRP median (IQR) mg/L 8.86 (3.00–21.50) 17.00 (4.00–47.94) 0.011 
YKL-40 median (IQR) μg/L 144.00 (82.25–225.00) 137.00 (79.00–228.00) 0.870 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of peritoneal carcinomatosis 
Peritoneal carcinomatosis No Yes P 
Number of patients 333 221  
IL6 median (IQR) pg/mL 7.40 (3.50–18.00) 5.00 (2.60–10.30) <0.001 
CRP median (IQR) mg/L 21.00 (5.00–63.37) 10.00 (3.00–32.00) <0.001 
YKL-40 median (IQR) μg/L 148.50 (84.00–255.25) 125.00 (72.00–186.00) 0.003 
Baseline serum IL6, CRP, and YKL-40 stratified by presence of metastasis in: liver versus peritoneum versus both versus other 
PS Liver Peritoneum Both Other P 
Number of patients 304 37 184 29  
IL6 median (IQR) pg/mL 7.50 (3.50–18.00) 6.40 (2.70–13.00) 5.00 (2.60–9.93) 6.10 (2.40–11.00) <0.001 
CRP median (IQR) mg/L 23.00 (6.00–65.24) 9.00 (3.00–24.00) 10.00 (3.00–32.55) 8.71 (3.00–18.36) <0.001 
YKL-40 median (IQR) μg/L 146.00 (84.50–262.50) 132.00 (83.00–201.00) 119.00 (70.50–185.25) 206.00 (74.00–227.00) 0.025 

Abbreviation: IQR, interquartile range.

Relationships between serum IL6, CRP, and YKL-40 and CA19-9, liver serum parameters, and inflammatory biomarkers

IL6, CRP, and YKL-40 were all correlated with each other and with the other examined inflammatory biomarkers (Table 3). IL6 and CRP but not YKL-40 were positively correlated with CA19-9. Serum levels of liver parameters were correlated with all three biomarkers, except for IL6 and YKL-40 in relation to ALT.

Table 3.

Spearman correlation coefficients between baseline IL6, CRP, and YKL-40 and CA19-9, WBC, ANC, platelets, and liver serum parameters.

IL6CRPYKL-40
IL6 — Rs = 0.69; P < 0.01 Rs = 0.42; P < 0.01 
CRP Rs = 0.69; P < 0.01 — Rs = 0.41; P < 0.01 
YKL-40 Rs = 0.42; P < 0.01 Rs = 0.41; P < 0.01 — 
CA19-9 Rs = 0.25; P < 0.01 Rs = 0.23; P < 0.01 Rs = 0.07; P = 0.12 
WBC Rs = 0.50; P < 0.01 Rs = 0.46; P < 0.01 Rs = 0.29; P < 0.01 
ANC Rs = 0.50; P < 0.01 Rs = 0.47; P < 0.01 Rs = 0.29; P < 0.01 
Platelets Rs = 0.20; P < 0.01 Rs = 0.23; P < 0.01 Rs = 0.14; P < 0.01 
Alkaline phosphatase Rs = 0.41; P < 0.01 Rs = 0.48; P < 0.01 Rs = 0.26; P < 0.01 
Bilirubin Rs = 0.18; P < 0.01 Rs = 0.20; P < 0.01 Rs = 0.10; P = 0.02 
Alanine transaminase Rs = 0.06; P = 0.16 Rs = 0.16; P < 0.01 Rs = 0.04; P = 0.32 
IL6CRPYKL-40
IL6 — Rs = 0.69; P < 0.01 Rs = 0.42; P < 0.01 
CRP Rs = 0.69; P < 0.01 — Rs = 0.41; P < 0.01 
YKL-40 Rs = 0.42; P < 0.01 Rs = 0.41; P < 0.01 — 
CA19-9 Rs = 0.25; P < 0.01 Rs = 0.23; P < 0.01 Rs = 0.07; P = 0.12 
WBC Rs = 0.50; P < 0.01 Rs = 0.46; P < 0.01 Rs = 0.29; P < 0.01 
ANC Rs = 0.50; P < 0.01 Rs = 0.47; P < 0.01 Rs = 0.29; P < 0.01 
Platelets Rs = 0.20; P < 0.01 Rs = 0.23; P < 0.01 Rs = 0.14; P < 0.01 
Alkaline phosphatase Rs = 0.41; P < 0.01 Rs = 0.48; P < 0.01 Rs = 0.26; P < 0.01 
Bilirubin Rs = 0.18; P < 0.01 Rs = 0.20; P < 0.01 Rs = 0.10; P = 0.02 
Alanine transaminase Rs = 0.06; P = 0.16 Rs = 0.16; P < 0.01 Rs = 0.04; P = 0.32 

Abbreviation: Rs, correlation coefficient from Spearman rank test.

Univariate and multivariate analyses

In survival analyses, patients with combined high serum values of two or three biomarkers including IL6, YKL-40, and CRP had worse OS compared with patients with low values or with an isolated high concentration of one biomarker (Table 4). When IL6, YKL-40, or CRP were analyzed separately, patients with serum levels above cutoff had increased overall mortality in a univariate Cox model and reduced survival in Kaplan–Meier plots (Fig. 1).

Table 4.

Univariate and multivariate HRs for OS according to clinical characteristics and baseline serum values of IL6, CRP, and YKL-40, dichotomized using cutoffs for IL6 of >4.92 pg/mL, CRP mg/L of >10, and YKL-40 of >95% age-corrected percentile as reference.

Univariate analysisMultivariate analysis
VariablesHR (95% CI)PHR (95% CI)P
Age >70 years 1.14 (0.96–1.35) 0.13 – – 
Sex (female) 0.95 (0.80–1.12) 0.52 – – 
PS 1 1.59 (1.32–1.92) <0.01 1.20 (0.92–1.55) 0.18 
PS 2 2.67 (2.06–3.46) <0.01 3.11 (2.12–4.56) <0.01 
BMI (underweight) 1.30 (0.96–1.75) 0.09 – – 
BMI (overweight) 1.12 (0.93–1.35) 0.22 – – 
Weight loss ≥2% 1.28 (1.03–1.60) 0.03 1.17 (0.89–1.53) 0.25 
Diabetes 0.89 (0.73–1.08) 0.25 – – 
Smoking 1.02 (0.85–1.24) 0.82 – – 
Alcohol 0.89 (0.73–1.09) 0.26 – – 
Stage II + III 0.58 (0.48–0.70) <0.01 a a 
Number of metastatic sites (1) 1.60 (1.31–1.95) <0.01 1.21 (0.77–1.89) 0.41 
Number of metastatic sites (2) 1.78 (1.34–2.35) <0.01 1.40 (0.83–2.37) 0.20 
Number of metastatic sites (3+) 3.93 (2.17–7.12) <0.01 2.31 (1.03–5.22) 0.04 
Liver metastasis 1.09 (0.83–1.41) 0.54 – – 
Peritoneum metastasis 0.60 (0.51–0.72) <0.01 0.73 (0.49–1.07) 0.10 
Platinum-based combination 0.53 (0.43–0.66) <0.01 0.55 (0.42–0.74) <0.01 
Gemcitabine plus nab-paclitaxel 0.55 (0.39–0.76) <0.01 0.44 (0.29–0.67) <0.01 
Capecitabine-based combination 0.66 (0.39–1.13) 0.13 – – 
CA19-9 U/mL ≥59 x ULN 1.54 (1.29–1.84) <0.01 1.27 (0.99–1.63) 0.06 
WBC × 109/L (per unit increase) 1.11 (1.09–1.14) <0.01 0.94 (0.84–1.05) 0.29 
ANC × 109/L (per unit increase) 1.14 (1.11–1.17) <0.01 1.13 (0.99–1.29) 0.08 
Platelets × 109/L (per unit increase) 1.00 (1.00–1.00) 0.04 1.00 (1.00–1.00) 0.29 
Low IL6, YKL-40, and CRP (Type 0) 1.00 (Ref.) Ref. Ref. Ref. 
High IL6 only (Type I) 1.08 (0.75–1.55) 0.69 – – 
High CRP only (Type I) 1.41 (1.03–1.94) 0.03 1.17 (0.77–1.78) 0.46 
High YKL-40 only (Type I) 1.33 (0.89–1.99) 0.16 – – 
High IL6 and YKL-40 or IL6 and CRP or YKL-40 and CRP (Type II) 1.94 (1.54–2.45) <0.01 1.41 (1.02–1.93) 0.04 
High IL6, YKL-40, and CRP (Type III) 2.93 (2.32–3.69) <0.01 1.87 (1.34–2.62) <0.01 
Univariate analysisMultivariate analysis
VariablesHR (95% CI)PHR (95% CI)P
Age >70 years 1.14 (0.96–1.35) 0.13 – – 
Sex (female) 0.95 (0.80–1.12) 0.52 – – 
PS 1 1.59 (1.32–1.92) <0.01 1.20 (0.92–1.55) 0.18 
PS 2 2.67 (2.06–3.46) <0.01 3.11 (2.12–4.56) <0.01 
BMI (underweight) 1.30 (0.96–1.75) 0.09 – – 
BMI (overweight) 1.12 (0.93–1.35) 0.22 – – 
Weight loss ≥2% 1.28 (1.03–1.60) 0.03 1.17 (0.89–1.53) 0.25 
Diabetes 0.89 (0.73–1.08) 0.25 – – 
Smoking 1.02 (0.85–1.24) 0.82 – – 
Alcohol 0.89 (0.73–1.09) 0.26 – – 
Stage II + III 0.58 (0.48–0.70) <0.01 a a 
Number of metastatic sites (1) 1.60 (1.31–1.95) <0.01 1.21 (0.77–1.89) 0.41 
Number of metastatic sites (2) 1.78 (1.34–2.35) <0.01 1.40 (0.83–2.37) 0.20 
Number of metastatic sites (3+) 3.93 (2.17–7.12) <0.01 2.31 (1.03–5.22) 0.04 
Liver metastasis 1.09 (0.83–1.41) 0.54 – – 
Peritoneum metastasis 0.60 (0.51–0.72) <0.01 0.73 (0.49–1.07) 0.10 
Platinum-based combination 0.53 (0.43–0.66) <0.01 0.55 (0.42–0.74) <0.01 
Gemcitabine plus nab-paclitaxel 0.55 (0.39–0.76) <0.01 0.44 (0.29–0.67) <0.01 
Capecitabine-based combination 0.66 (0.39–1.13) 0.13 – – 
CA19-9 U/mL ≥59 x ULN 1.54 (1.29–1.84) <0.01 1.27 (0.99–1.63) 0.06 
WBC × 109/L (per unit increase) 1.11 (1.09–1.14) <0.01 0.94 (0.84–1.05) 0.29 
ANC × 109/L (per unit increase) 1.14 (1.11–1.17) <0.01 1.13 (0.99–1.29) 0.08 
Platelets × 109/L (per unit increase) 1.00 (1.00–1.00) 0.04 1.00 (1.00–1.00) 0.29 
Low IL6, YKL-40, and CRP (Type 0) 1.00 (Ref.) Ref. Ref. Ref. 
High IL6 only (Type I) 1.08 (0.75–1.55) 0.69 – – 
High CRP only (Type I) 1.41 (1.03–1.94) 0.03 1.17 (0.77–1.78) 0.46 
High YKL-40 only (Type I) 1.33 (0.89–1.99) 0.16 – – 
High IL6 and YKL-40 or IL6 and CRP or YKL-40 and CRP (Type II) 1.94 (1.54–2.45) <0.01 1.41 (1.02–1.93) 0.04 
High IL6, YKL-40, and CRP (Type III) 2.93 (2.32–3.69) <0.01 1.87 (1.34–2.62) <0.01 

Note: Patients with low values of IL6, YKL-40, and CRP (type 0); high value of one (type I) or two biomarkers (type II); or high values of all three biomarkers (type III). The significant terms are included in the multivariate analyses.

Abbreviation: ULN, upper limit of normal.

aBecause both number of metastasis and stage are significant, only number of metastasis is included.

Figure 1.

Kaplan–Meier survival curves showing baseline serum IL6 (A), YKL-40 (B), and CRP (C) and combined IL6, YKL-40, and CRP (D), dichotomized as high and low using cutoffs of >4.92 pg/mL, >95th age percentile, and >10 mg/L, respectively. In the combined model, Kaplan–Meier curves show patients with low values of IL6, YKL-40, and CRP (type 0), high value of one (type I) or two biomarkers (type II), or high values of all three biomarkers (type III).

Figure 1.

Kaplan–Meier survival curves showing baseline serum IL6 (A), YKL-40 (B), and CRP (C) and combined IL6, YKL-40, and CRP (D), dichotomized as high and low using cutoffs of >4.92 pg/mL, >95th age percentile, and >10 mg/L, respectively. In the combined model, Kaplan–Meier curves show patients with low values of IL6, YKL-40, and CRP (type 0), high value of one (type I) or two biomarkers (type II), or high values of all three biomarkers (type III).

Close modal

Mortality was increased in the patients with higher PS; substantial weight loss; higher stage; number of metastatic sites; increased baseline CA 19.9, WBC, ANC, platelet counts, and capecitabine-containing regimens. Combination chemotherapy with either FOLFIRINOX or gemcitabine + nab-Paclitaxel was associated with better survival compared with gemcitabine alone. The following exposures were not associated with mortality: age, sex, BMI, history of diabetes, smoking, alcohol consumption, and presence of liver or peritoneum metastases.

Significant variables in univariate analysis were subsequently analyzed in a multivariate model. Combined high values of two or three of the biomarkers IL6, YKL-40, and CRP remained independently associated with a poorer OS (Table 4).

In addition, we performed ROC curve analysis to describe the ability of the three biomarkers compared with CA19-9 to predict early death, defined as survival duration <90 days from the time of diagnosis (Fig. 2). For baseline serum IL6, CRP, YKL-40, and CA19-9, the area under the ROC curve was 0.74, 0.72, 0.69, and 0.68, respectively (Supplementary Table S2). The AUC for combined high values of IL6, CRP, YKL-40 was higher (AUC = 0.86; 95% CI, 81.4–89.7) compared with the high value of each marker separately. Notably, combining the values of these three markers with CA19-9 did not increase the AUC further.

Figure 2.

ROC curve of single biomarker, IL6, CRP, and YKL-40, and combined marker detection in predicting early death, defined as survival duration <90 days from the time of diagnosis.

Figure 2.

ROC curve of single biomarker, IL6, CRP, and YKL-40, and combined marker detection in predicting early death, defined as survival duration <90 days from the time of diagnosis.

Close modal

In contrast to an isolated high value of one biomarker, having more than one biomarker was associated with early death in the univariate analyses along with PS 1–2, stage IV cancer, higher baseline CA 19.9, WBC, and ANC (Supplementary Table S3). Combined high baseline values of IL6, YKL-40, and CRP remained independently associated with early death after further adjustment for significant covariates in multivariate analysis. Patients with high levels of two or three biomarkers were at 6.3-fold increased risk of early death compared with patients without increased levels of IL6, CRP, and YKL-40.

Changes in serum IL6 and YKL-40 during first-line chemotherapy

Longitudinal serum levels of IL6 and YKL-40 were available in 398 patients before the second cycle of chemotherapy and in 260 patients at the time of first CT evaluation. This analysis showed that IL6 values, both the baseline log2-transformed value and the log2-transformed ratio between current value and the baseline value, were associated with OS (HR: 1.34; 95% CI, 1.21–1.47 and HR: 1.40; 95% CI, 1.29–1.52, respectively; Supplementary Table S4). YKL-40 levels in a time-varying model were not prognostic for survival.

Finally, the biomarkers were dichotomized based on the ≥25% versus <25% increase of IL6 and YKL-40 levels measured before the second cycle of chemotherapy and prior to first CT evaluation (Supplementary Fig. S1). For IL6, a significantly shorter OS was observed among patients with an increase of at least 25% from baseline level (P = 0.012). In addition, we analyzed IL6 and YKL-40 concentrations 281 patients with available samples collected at the time of cancer progression as determined by CT. We found statistically significant associations between survival and serum IL6 and YKL-40 levels, also after adjustment (Supplementary Table S5).

In this large cohort of patients with pancreatic cancer undergoing palliative chemotherapy, we found that combined high serum levels of IL6, YKL-40, and CRP were associated with a worse OS, independently of other prognostic covariates. The majority of patients had concomitant high serum values of all or at least two of the biomarkers IL6, YKL-40, and CRP and a minority was registered with either low serum values or an isolated high value of a single marker. Higher values of IL6, YKL-40, or CRP were seen in patients with worse PS and higher stage. High levels of at least two biomarkers were associated with increased 90-day mortality. Accordingly, high serum IL6 and YKL-40 values at the time of cancer progression were negatively correlated with survival. Moreover, patients with an increase in IL6 greater than 25% 2–3 months after start of palliative chemotherapy had worse outcomes. These findings support the notion that the inflammatory response is a complex systemic reaction and is associated with poor outcome in patients with advanced pancreatic cancer.

Pancreatic cancer is one of the most aggressive of all malignant tumors (7), and only minor improvements in treatment have taken place over the past 20 years (32). Almost one-third of the BIOPAC cohort patients die within 3 months after start of palliative chemotherapy (Supplementary Fig. S2). Although pancreatic cancer implicates a rich pool of genetic alterations, no biomarker has yet been definitively validated for use in the clinical setting for prognostic, predictive, or diagnostic applications. In fact, the complex heterogeneity of the cancer at the molecular level may complicate identification of a single reliable biomarker that can be readily measured or utilized. Application of CA19-9, a serum-based protein biomarker, which is the most extensively validated and commonly used in daily clinical practice, is restricted because of false-positive elevations in other benign conditions, as well as not being detectable in individuals with a Lewis antigen–negative phenotype. CA19-9 above the median of 59× upper limit of normal (ULN) U/mL was reported to be associated with the poorest prognosis among a large group of patients with advanced pancreatic cancer (33, 34). However, the impact of high CA19-9 in our study was dependent on other covariates. Indeed, a PS of 2 was confirmed to be the strongest negative factor in all analyses, as expected (34).

Our findings should be interpreted in the context of the increased understanding of the interaction between cancer and activation of systemic inflammation (35), which can be measured by CRP, a nonspecific acute-phase protein synthesized in the liver in response to stimulation mainly from IL6 (35). The discovery of IL6-mediated protumor inflammation (7, 8), which is one of the players in the immune cell traffic (36, 37) and activation, provides further evidence of communication between pancreatic cancer–induced immunosuppression and inflammation (38). Although our findings can be placed in the context of immune system communication, the specific details of this process and applicable therapeutic targeting, as they relate to IL6-induced signaling, are yet to be determined. Our results indicating that high IL6, CRP, and YKL-40 levels are associated with increased mortality in the extended cohort of patients with pancreatic cancer undergoing palliative chemotherapy are consistent with previous studies (15) and in line with the known association between pancreatic cancer and IL6-induced inflammation (39). Individual and combined higher levels of CRP, IL6, and macrophage-inhibitory cytokine-1 were associated with significant reductions in overall mortality in a large prospective study of 446 patients with pancreatic cancer (40). Inflammation plays an important role in cancer progression (3, 37, 41). IL6 is an immunomodulating cytokine with pleiotropic properties and is produced by many cell types (10, 11). Several reports have highlighted the integral role of IL6 in facilitating tumor-promoting processes including inflammation (8, 9, 42). IL6-mediated inflammation is thought to be a component of tumor-induced immunosuppression (11, 12) and resistance to immunotherapy (38, 43). Greater activation of the systemic inflammatory response and high plasma IL6 levels have been reported in the majority of patients with advanced pancreatic cancer (5, 6, 13, 15). Preclinical pancreatic cancer models have clearly shown that the anti-IL6 receptor antibody, tocilizumab, in combination with chemotherapy reduces tumor growth, number of distant metastases, and local recurrence rate (44). Furthermore, blockade of IL6-regulated signaling pathways has been shown to inhibit liver metastasis (45), immunomodulate suppressive microenvironment, and enhance the efficacy of anti-programmed death-1 ligand 1 checkpoint inhibitor therapy (38, 43). Targeting of IL6 in combination with chemotherapy is actively under investigation in patients with advanced pancreatic cancer (NCT02767557).

In addition, our findings that YKL-40 had a negative prognostic value and that it was correlated with other inflammation markers are consistent with previous studies (15, 46). YKL-40 regulates key pathways and processes within the respective tumor microenvironment, including inflammation, angiogenesis, cell proliferation, differentiation, and remodeling of the extracellular matrix and thus promotes tumor progression (29, 47). Ablation of YKL-40 expression promotes CTL activation and, thus, antitumor immunity (18). YKL-40 may be a new therapeutic target for patients with cancer, because antibodies against YKL-40 are able to inhibit tumor angiogenesis and cancer progression (18, 23, 28). Tumor-associated macrophage-secreted YKL-40 could promote metastasis via activation of MAPK signaling (27). Previous studies characterize the majority of patients with pancreatic cancer as having high plasma YKL-40 (19). Further studies in different populations are needed to confirm our results.

The primary limitation of this study is that the patients with pancreatic cancer in the BIOPAC study represent only 20% of all newly diagnosed cases of pancreatic cancer in Denmark according to Danish Cancer Registry. The biomarkers of interests were prespecified in the BIOPAC project from the beginning and analyzed prospectively. However, due to the way clinical data were obtained from the patient files, recall bias was an inevitable potential source of bias due to the differences in the accuracy or completeness of the recollections retrieved from the patient files, although our records were thoroughly quality-checked by two separate investigators. The analyzed cohort in this study was restricted to patients treated in a palliative setting, thus survival prediction is limited by generally short survival in this population and cannot be directly applied to patients with resectable pancreatic cancer, or patients with much longer survival. Further longitudinal prospective studies (5) are required before inflammatory markers might be used routinely in the management of pancreatic cancer across all stages (5).

In conclusion, our findings suggest that combined high baseline serum levels of IL6, YKL-40, and CRP are associated with poor prognosis in patients with unresectable pancreatic cancer. A high concentration of IL6 measured before chemotherapy and over time is a strong independent prognostic biomarker in patients with unresectable pancreatic cancer. Detection of inflammation markers including IL6, YKL-40, and CRP is likely to help clinicians prognosticate outcomes in patients with pancreatic cancer. Further investigation and development of anti-inflammation therapeutic strategies in patients with pancreatic cancer are warranted.

No potential conflicts of interest were disclosed.

Conception and design: I.M. Chen, C. Dehlendorff, B.V. Jensen, P. Pfeiffer, J.S. Johansen, N.H. Holländer

Development of methodology: I.M. Chen, P. Pfeiffer, J.S. Johansen, N.H. Holländer

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): I.M. Chen, B.V. Jensen, S.E. Bojesen, P. Pfeiffer, J.K. Bjerregaard, S.E. Nielsen, F. Andersen, L.S. Rasmussen, J.S. Johansen, N.H. Holländer

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): I.M. Chen, C. Dehlendorff, B.V. Jensen, S.E. Bojesen, P. Pfeiffer, J.K. Bjerregaard, S.E. Nielsen, J.S. Johansen, N.H. Holländer

Writing, review, and/or revision of the manuscript: I.M. Chen, A.Z. Johansen, C. Dehlendorff, B.V. Jensen, S.E. Bojesen, P. Pfeiffer, S.E. Nielsen, F. Andersen, M.K. Yilmaz, L.S. Rasmussen, J.S. Johansen, N.H. Holländer

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): I.M. Chen, A.Z. Johansen, B.V. Jensen, S.E. Bojesen, L.S. Rasmussen, J.S. Johansen, N.H. Holländer

Study supervision: I.M. Chen, B.V. Jensen, J.S. Johansen

The authors thank the biomedical laboratory scientists Charlotte Falk, Vibeke H. Holm, Dorte B. Nielsen, Helle B. Sørensen, Ulla Kjærulff-Hansen, Teresa Rozenfeld, and Mie B. Krüger for their assistance. The authors acknowledge the nurses Hanne M. Michaelsen, Lars M. Larsen, Lene T. Voxen, Finn Hejlesen, Signe Gamborg Nielsen, and Jette L. Hansen; the physician Dan Calatayud; and the medical students Camilla Palmquist and Karin Bagni for their contribution to inclusions of patients, data registration, or sample collection.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Kleeff
J
,
Korc
M
,
Apte
M
,
La Vecchia
C
,
Johnson
CD
,
Biankin
AV
, et al
Pancreatic cancer
.
Nat Rev Dis Primers
2016
;
21
:
16022
.
2.
Balkwill
F
,
Mantovani
A
. 
Inflammation and cancer: back to Virchow?
Lancet
2001
;
357
:
539
45
.
3.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
4.
Proctor
MJ
,
Morrison
DS
,
Talwar
D
,
Balmer
SM
,
O'Reilly
DS
,
Foulis
AK
, et al
An inflammation-based prognostic score (mGPS) predicts cancer survival independent of tumour site: a Glasgow Inflammation Outcome Study
.
Br J Cancer
2011
;
104
:
726
34
.
5.
Szkandera
J
,
Stotz
M
,
Absenger
G
,
Stojakovic
T
,
Samonigg
H
,
Kornprat
P
, et al
Validation of C-reactive protein levels as a prognostic indicator for survival in a large cohort of pancreatic cancer patients
.
Br J Cancer
2014
;
110
:
183
8
.
6.
Wang
DS
,
Luo
HY
,
Qiu
MZ
,
Wang
ZQ
,
Zhang
DS
,
Wang
FH
, et al
Comparison of the prognostic values of various inflammation based factors in patients with pancreatic cancer
.
Med Oncol
2012
;
29
:
3092
100
.
7.
Ryan
DP
,
Hong
TS
,
Bardeesy
N
. 
Pancreatic adenocarcinoma
.
N Engl J Med
2014
;
371
:
2140
1
.
8.
Bromberg
J
,
Wang
TC
. 
Inflammation and cancer: IL-6 and STAT3 complete the link
.
Cancer Cell
2009
;
15
:
79
80
.
9.
Scheller
J
,
Ohnesorge
N
,
Rose-John
S
. 
Interleukin-6 trans-signalling in chronic inflammation and cancer
.
Scand J Immunol
2006
;
63
:
321
9
.
10.
Johnson
DE
,
O'Keefe
RA
,
Grandis
JR
. 
Targeting the IL-6/JAK/STAT3 signalling axis in cancer
.
Nat Rev Clin Oncol
2018
;
15
:
234
48
.
11.
Kumari
N
,
Dwarakanath
BS
,
Das
A
,
Bhatt
AN
. 
Role of interleukin-6 in cancer progression and therapeutic resistance
.
Tumour Biol
2016
;
37
:
11553
72
.
12.
Guo
Y
,
Xu
F
,
Lu
T
,
Duan
Z
,
Zhang
Z
. 
Interleukin-6 signaling pathway in targeted therapy for cancer
.
Cancer Treat Rev
2012
;
38
:
904
10
.
13.
Miura
T
,
Mitsunaga
S
,
Ikeda
M
,
Shimizu
S
,
Ohno
I
,
Takahashi
H
, et al
Characterization of patients with advanced pancreatic cancer and high serum interleukin-6 levels
.
Pancreas
2015
;
44
:
756
63
.
14.
Strassmann
G
,
Fong
M
,
Kenney
JS
,
Jacob
CO
. 
Evidence for the involvement of interleukin 6 in experimental cancer cachexia
.
J Clin Invest
1992
;
89
:
1681
4
.
15.
Schultz
NA
,
Christensen
IJ
,
Werner
J
,
Giese
N
,
Jensen
BV
,
Larsen
O
, et al
Diagnostic and prognostic impact of circulating YKL-40, IL-6, and CA 19.9 in patients with pancreatic cancer
.
PLoS One
2013
;
8
:
e67059
.
16.
Grote
VA
,
Kaaks
R
,
Nieters
A
,
Tjønneland
A
,
Halkjær
J
,
Overvad
K
, et al
Inflammation marker and risk of pancreatic cancer: a nested case-control study within the EPIC cohort
.
Br J Cancer
2012
;
106
:
1866
74
.
17.
Bao
Y
,
Giovannucci
EL
,
Kraft
P
,
Qian
ZR
,
Wu
C
,
Ogino
S
, et al
Inflammatory plasma markers and pancreatic cancer risk: a prospective study of five U.S. cohorts
.
Cancer Epidemiol Biomarkers Prev
2013
;
22
:
855
61
.
18.
Kim
DH
,
Park
HJ
,
Lim
S
,
Koo
JH
,
Lee
HG
,
Choi
JO
, et al
Regulation of chitinase-3-like-1 in T cell elicits Th1 and cytotoxic responses to inhibit lung metastasis
.
Nat Commun
2018
;
9
:
503
.
19.
Johansen
JS
,
Schultz
NA
,
Jensen
BV
. 
Plasma YKL-40: a potential new cancer biomarker?
Future Oncol
2009
;
5
:
1065
82
.
20.
Libreros
S
,
Iragavarapu-Charyulu
V
. 
YKL-40/CHI3L1 drives inflammation on the road of tumor progression
.
J Leukoc Biol
2015
;
98
:
931
6
.
21.
Lee
CG
,
Da Silva
CA
,
Dela Cruz
CS
,
Ahangari
F
,
Ma
B
,
Kang
MJ
, et al
Role of chitin and chitinase/chitinase-like proteins in inflammation, tissue remodeling, and injury
.
Annu Rev Physiol
2011
;
73
:
479
501
.
22.
Bojesen
SE
,
Johansen
JS
,
Nordestgaard
BG
. 
Plasma YKL-40 levels in healthy subjects from the general population
.
Clin Chim Acta
2011
;
412
:
709
12
.
23.
Faibish
M
,
Francescone
R
,
Bentley
B
,
Yan
W
,
Shao
R
. 
A YKL-40-neutralizing antibody blocks tumor angiogenesis and progression: a potential therapeutic agent in cancers
.
Mol Cancer Ther
2011
;
10
:
742
51
.
24.
Low
D
,
Subramaniam
R
,
Lin
L
,
Aomatsu
T
,
Mizoguchi
A
,
Ng
A
, et al
Chitinase 3-like 1 induces survival and proliferation of intestinal epithelial cells during chronic inflammation and colitis-associated cancer by regulating S100A9
.
Oncotarget
2015
;
6
:
36535
50
.
25.
Shao
R
,
Hamel
K
,
Petersen
L
,
Cao
QJ
,
Arenas
RB
,
Bigelow
C
, et al
YKL-40, a secreted glycoprotein, promotes tumor angiogenesis
.
Oncogene
2009
;
28
:
4456
68
.
26.
Lee
CG
,
Hartl
D
,
Lee
GR
,
Koller
B
,
Matsuura
H
,
Da Silva
CA
, et al
Role of breast regression protein 39 (BRP-39)/chitinase 3-like-1 in Th2 and IL-13-induced tissue responses and apoptosis
.
J Exp Med
2009
;
206
:
1149
66
.
27.
Chen
Y
,
Zhang
S
,
Wang
Q
,
Zhang
X
. 
Tumor-recruited M2 macrophages promote gastric and breast cancer metastasis via M2 macrophage-secreted CHI3L1 protein
.
J Hematol Oncol
2017
;
10
:
36
.
28.
Libreros
S
,
Garcia-Areas
R
,
Shibata
Y
,
Carrio
R
,
Torroella-Kouri
M
,
Iragavarapu-Charyulu
V
. 
Induction of proinflammatory mediators by CHI3L1 is reduced by chitin treatment: decreased tumor metastasis in a breast cancer model
.
Int J Cancer
2012
;
131
:
377
86
.
29.
Hamilton
G
,
Rath
B
. 
Circulating tumor cell interactions with macrophages: implications for biology and treatment
.
Transl Lung Cancer Res
2017
;
6
:
418
30
.
30.
Knudsen
LS
,
Christensen
IJ
,
Lottenburger
T
,
Svendsen
MN
,
Nielsen
HJ
,
Nielsen
L
, et al
Pre-analytical and biological variability in circulating interleukin 6 in healthy subjects and patients with rheumatoid arthritis
.
Biomarkers
2008
;
13
59
78
.
31.
Altman
DG
,
McShane
LM
,
Sauerbrei
W
,
Taube
SE
. 
Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration
.
PLoS Med
2012
;
9
:
e1001216
.
32.
Golan
T
,
Sella
T
,
Margalit
O
,
Amit
U
,
Halpern
N
,
Aderka
D
, et al
Short- and long-term survival in metastatic pancreatic adenocarcinoma, 1993–2013
.
J Natl Compr Canc Netw
2017
;
15
:
1022
7
.
33.
Hess
V
,
Glimelius
B
,
Grawe
P
,
Dietrich
D
,
Bodoky
G
,
Ruhstaller
T
, et al
CA 19-9 tumour-marker response to chemotherapy in patients with advanced pancreatic cancer enrolled in a randomised controlled trial
.
Lancet Oncol
2008
;
9
:
132
8
.
34.
Tabernero
J
,
Chiorean
EG
,
Infante
JR
,
Hingorani
SR
,
Ganju
V
,
Weekes
C
, et al
Prognostic factors of survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer
.
Oncologist
2015
;
20
:
143
50
.
35.
Rincon
M
. 
Interleukin-6: from an inflammatory marker to a target for inflammatory diseases
.
Trends Immunol
2012
;
33
:
571
7
.
36.
Hunter
CA
,
Jones
SA
. 
IL-6 as a keystone cytokine in health and disease
.
Nat Immunol
2015
;
16
:
448
57
.
37.
Grivennikov
SI
,
Greten
FR
,
Karin
M
. 
Immunity, inflammation, and cancer
.
Cell
2010
;
140
:
883
99
.
38.
Mace
TA
,
Shakya
R
,
Pitarresi
JR
,
Swanson
B
,
McQuinn
CW
,
Loftus
S
, et al
IL-6 and PD-L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer
.
Gut
2018
;
67
:
320
32
.
39.
Lesina
M
,
Kurkowski
MU
,
Ludes
K
,
Rose-John
S
,
Treiber
M
,
Klöppel
G
, et al
Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer
.
Cancer Cell
2011
;
19
:
456
69
.
40.
Babic
A
,
Schnure
N
,
Neupane
NP
,
Zaman
MM
,
Rifai
N
,
Welch
MW
, et al
Plasma inflammatory cytokines and survival of pancreatic cancer patients
.
Clin Transl Gastroenterol
2018
;
9
:
145
57
.
41.
Rakoff-Nahoum
S
. 
Why cancer and inflammation?
Yale J Biol Med
2006
;
79
:
123
30
.
42.
Jones
SA
,
Jenkins
BJ
. 
Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer
.
Nat Rev Immunol
2018
;
18
:
773
89
.
43.
Liu
H
,
Shen
J
,
Lu
K
. 
IL-6 and PD-L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model
.
Biochem Biophys Res Commun
2017
;
486
:
239
44
.
44.
Goumas
FA
,
Holmer
R
,
Egberts
JH
,
Gontarewicz
A
,
Heneweer
C
,
Geisen
U
, et al
Inhibition of IL-6 signaling significantly reduces primary tumor growth and recurrencies in orthotopic xenograft models of pancreatic cancer
.
Int J Cancer
2015
;
137
:
1035
46
.
45.
Lee
JW
,
Stone
ML
,
Porrett
PM
,
Thomas
SK
,
Komar
CA
,
Li
JH
, et al
Hepatocytes direct the formation of a pro-metastatic niche in the liver
.
Nature
2019
;
567
:
249
52
.
46.
Johansen
JS
,
Bojesen
SE
,
Mylin
AK
,
Frikke-Schmidt
R
,
Price
PA
,
Nordestgaard
BG
. 
Elevated plasma YKL-40 predicts increased risk of gastrointestinal cancer and decreased survival after any cancer diagnosis in the general population
.
J Clin Oncol
2009
;
27
:
572
8
.
47.
Yeo
IJ
,
Lee
CK
,
Han
SB
,
Yun
J
,
Hong
JT
. 
Roles of chi tinase 3-like 1 in the development of cancer, neurodegenerative diseases, and inflammatory diseases
.
Pharmacol Ther
2019
;
26
:
107394
.

Supplementary data