Purpose:

Pancreatic cancer is one of the most lethal solid tumors, mainly because of its intrinsic chemoresistance. We identified TAK1 as a central hub sustaining this resistance. Nanoliposomal irinotecan (nal-IRI) is a novel treatment for metastatic gemcitabine-refractory pancreatic cancer. We endeavored to identify circulating markers for TAK1 activation predicting chemoresistance in this setting.

Experimental Design:

In vivo activity of nal-IRI was validated in an orthotopic nude murine model expressing TAK1-specific shRNA. Plasma concentration of 20 different cytokines were measured by a multiplex xMAP/Luminex technology in patients prospectively enrolled to receive nal-IRI plus 5-fluorouracil/leucovorin (5-FU/LV). The optimal cutoff thresholds able to significantly predict patients' outcome were obtained on the basis of the maximization of the Youden's statistics.

Results:

Differential expression profiling revealed the gene coding for IL8 as the most significantly downregulated in shTAK1 pancreatic cancer cell lines. Mice bearing shTAK1 tumors had significantly lower plasma levels of IL8 and experienced a significant reduction in tumor growth if treated with nal-IRI, whereas those bearing TAK1-proficient tumors were resistant to this agent. In a discovery cohort of 77 patients, IL8 was the circulating factor most significantly correlated with survival (plasma levels lower vs higher than cutoff: mPFS 3.4 months vs 2.8 months; hazard ratio [HR], 2.55; 95% CI, 1.39–4.67; P = 0.0017; median overall survival 8.9 months vs 5.3 months; HR, 3.51; 95% CI, 0.84–6.68; P = 4.9e−05). These results were confirmed in a validation cohort of 50 patients.

Conclusions:

Our study identified IL8 as the most significant circulating factor for TAK1 pathway activation and candidates IL8 as a potential predictive biomarker of resistance to nal-IRI in gemcitabine-refractory patients with pancreatic cancer.

Translational Relevance

The poor prognosis of patients with pancreatic cancer could be mainly attributed to its intrinsic resistance to the currently approved classic chemotherapeutic treatments. In the last decade, we contributed to this field by demonstrating that TAK1 pathway has one of the most essential roles in pancreatic cancer treatment resistance. Nanoliposomal irinotecan (nal-IRI) is a novel standard treatment for metastatic gemcitabine-refractory pancreatic cancer. However, the exceeding cost-effectiveness ratio limited the recommendation for marketing authorization of this regimens and, thus, the identification of biomarkers for patients' selection remains a significant priority. Here, we demonstrated that the chemokine interleukin-8 is the circulating factor most significantly regulated by TAK1 activation. By demonstrating that interleukin-8 could be, indeed, a useful biomarker for identifying patients that could take the larger advantage by nal-IRI, our findings cover one of the most significant unmet need in the selection of second- and further lines of treatment for pancreatic cancer.

Pancreatic cancer remains one of the most lethal and poorly understood human malignancies and will continue to be a major unsolved health problem in the 21st century (1). Pancreatic cancer has still the lowest 5-year relative survival rate among solid tumors at 7%, and is projected to become the second leading cause of cancer-related death by 2030 in Western countries (2, 3). The poor prognosis for patients with pancreatic cancer could be mainly attributed to the limited efficacy of currently approved classic chemotherapeutic treatments (4).

We recently identified the serine/threonine kinase TGFβ-activated kinase 1 (TAK1) as a central hub integrating the most relevant signals from various cytokines (5, 6) and sustaining, in turn, resistance to chemotherapeutic treatments through the activation of different transcription factors, including AP-1, NF-κB (7, 8), and YAP/TAZ (9). In particular, we demonstrated that targeting the kinase activity of TAK1 dramatically led to a proapoptotic phenotype and, in turn, to a significantly higher sensitivity to chemotherapy and radiotherapy in pancreatic (10) and esophageal carcinoma (11).

Nanoliposomal irinotecan (nal-IRI; Onivyde; MM-398) is a liposomal formulation of the topoisomerase I inhibitor irinotecan. Efficacy of nal-IRI was recently demonstrated in the NAPOLI-1 study, a phase III, randomized, open-label, multicenter study that tested nal-IRI monotherapy or nal-IRI plus 5-fluorouracil (5-FU) and leucovorin (LV) versus 5-FU/LV alone in patients with metastatic pancreatic cancer previously treated with gemcitabine-based therapy (12). nal-IRI plus 5-FU/LV led to a significant improvement in median overall survival (mOS) over 5-FU/LV alone (13, 14), becoming a novel standard of care for second-line treatment of patients with metastatic pancreatic cancer (15).

We aimed this study at identifying circulating markers of TAK1 pathway activation, which could potentially serve as resistance biomarkers in this clinical setting.

Cell cultures and reagents

AsPC1, Panc1, and MDA-Panc28 human pancreatic cancer cell lines were purchased from the ATCC. MDA-Panc28 cell line was a kind gift by Dr. Paul J. Chiao. Panc1, AsPC1, and MDA-Panc28 pancreatic cancer cell lines silenced for the expression of TAK1 were established as described in ref. 10. All cell lines were cultured as monolayers at 37°C, 5% CO2 in high glucose DMEM (catalog No. 41966-029; Life Technologies), supplemented with 10% heat-inactivated FBS (catalog No. 10270-106; Life Technologies), 2 mmol/L L-glutamine (catalog No. BE17-605E; Lonza), 100 IU/mL penicillin and 100 μg/mL streptomycin (catalog No. 15140-122; Life Technologies), and cultured for maximum 1 month. Cell lines were authenticated by DNA fingerprinting at the genomic core facility at Wayne State University (2009) and routinely tested for mycoplasma presence using the MycoAlert Mycoplasma Detection Kit (catalog No. LT07-118; Lonza).

Gene expression and pathway analyses

Differences in gene expression between control and TAK1-silenced cells (GEO accession No. GSE137265) were examined by using Illumina Human 48k gene chips (D-103-0204; Illumina) as previously described in ref. 9.

Protein extraction and Western blotting

Western blot analyses were performed as described previously (16). Briefly, total protein extracts were prepared by lysing cells in radioimmunoprecipitation assay buffer [50 mmol/L Tris HCl (pH 8), 150 mmol/L NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% SDS]. All protein extracts were quantified by BCA Protein Assay Kit (23225; Thermo Fisher Scientific) and equal amounts (20–50 μg of protein extract) were loaded onto SDS-PAGE (4–20%) and transferred to polyvinylidene fluoride membranes (Immobilon-P, catalog No. IPVH00010; Millipore). Immunoblots were performed using the indicated antibodies. Anti-TAK1 antibody (ab109526, 1:1,000) was purchased from Abcam. Secondary anti-mouse and anti-rabbit antibodies were purchased from Jackson Immunoresearch. All antibodies were diluted in 5% nonfat dry milk dissolved in Tris-buffered saline/0.1% Tween-20. Immunoreactive proteins were visualized with Immobilion Western Kit (catalog No. WBKLS0500; EMD MIllipore) according to the manufacturer's instructions. Images were acquired using ImageQuant LAS 4000 mini (GE Healthcare Life Sciences).

Nude mouse orthotopic xenograft models

Five-week-old female athymic nude mice (Crl:CD1-Foxn1nu, CDNSSFE05S) were purchased from Charles River. University of Verona Animal Ethic Committee approved this study. Subconfluent cultures of pancreatic cancer cells were collected using 0.05% trypsin-EDTA (GIBCO, ref. 25300-054), that was inhibited with 10% FBS in DMEM. Tumor cells were resuspended in a solution of 1:1 Matrigel:PBS at 1.0 × 104 cells/μL concentration (Matrigel Matrix Growth Factor, 356230; BD Biosciences). Orthotopic injection of pancreatic cancer cells was performed as described previously (17).

All mice were weighed weekly and observed for tumors becoming palpable. When a tumor mass reached the diameter of 10 mm became palpable and measurable with a standard caliper. Tumor diameter was assessed with a Vernier caliper, and tumor volume (mm3) was calculated as d2 × D/2, wherein d and D represent the shortest and longest diameters, respectively. Bulky disease was considered present when tumor burden was prominent in the mouse abdomen (tumor volume, >2,000 mm3). When at least three of five mice in a treatment group presented with bulky disease, the median survival duration for that group was considered to be reached. At the median survival duration of the control groups, all mice of control and treatment groups were euthanized by deep terminal anesthesia and blood and tumor samples were collected. Differences in mean weight were considered for the effects of treatments between different groups.

Immunohistochemistry

Immunostaining was performed in formalin-fixed paraffin-embedded tissue sections using the polymeric HRP staining system (UltraTek HRP Anti-Polyvalent DAB Staining System, AMF080; Histoline) as described ref. 18. Citrate pH 6.0 was used as antigen retrieval buffer. Primary monoclonal antibodies to IL8 (ab106350, 1:500) or to CD68 (ab31630; Abcam), or polyclonal antibody to CES2 (orb214832; Biorbyt) were incubated in a humified chamber at 4°C overnight. Signal detection was performed incubating sections with UltraTek Anti-Polyvalent and UltraTek HRP for 10 minutes at room temperature (UltraTek HRP Anti-Polyvalent DAB Staining System, AMF080; Histoline). Section were counterstained with Mayer's hematoxylin and slides were mounted using Entellan Neo Rapid nonaqueous mounting medium (Merck Millipore).

Patients and samples analysis

Patients enrolled in this study had advanced histologically or cytologically confirmed pancreatic adenocarcinoma and were previously treated with gemcitabine-based therapy, received in localized or metastatic setting. Other eligibility criteria included adequate hepatic, renal, and hematologic function, an ECOG performance status ≤2, and no other clinically significant disorders. Eligible patients were prospectively included in an Expanded Access Program to receive nal-IRI 80 mg/m2 intravenously over 90 minutes, followed by LV 400 mg/m2 intravenously over 30 minutes and 5-FU 2,400 mg/m2 intravenously over 46 hours, every 2 weeks. We analyzed patients of two different cohorts on the basis of site of enrollment. Patients from the University Hospital Trust in Verona comprised the discovery cohort (n = 77), and those from Veneto Institute of Oncology (IOV-IRCCS) in Padua comprised the validation cohort (n = 50). At baseline, before first dose of nal-IRI, peripheral blood samples were collected using EDTA-containing tubes. All data were anonymized assigning a progressive number to every patient and the corresponding blood sample. Plasma aliquots were isolated from the blood samples by centrifugation with 1,600 rcf for 10 minutes and subsequently with 3,000 g for 10 minutes. Plasma samples were stored at −80°C. Written informed consent was obtained from all subjects. The study was conducted in accordance with the Declaration of Helsinki and approved by local ethics committee. Tumor response was assessed according to RECIST 1.1. Disease was assessed by CT (or MRI) every 12 weeks until disease progression.

Multiplex cytokines profiling

Plasma concentration of IL1β, IL2, IL4, IL5, IL6, IL8, IL12p70, IL13, IL18, eotaxin, GM-CSF, IFNγ, IP-10 (CXCL10), monocyte chemoattractant protein (MCP1; CCL2), macrophage inflammatory protein 1 α (MIP-1a; CCL3), MIP1-β (CCL4), TNFα, Rantes, SDF1α, GROα were measured by using a 20-plex Luminex Kit (Life Technologies, catalog No. EPX200-12173-901). All Luminex assays were performed according to the instructions provided by the manufacturer (Bio-Rad Laboratories). Median fluorescence intensities were collected on a Luminex-200 instrument, using Bio-Plex Manager software version 6.2. Standard curves for each cytokine were generated using the premixed lyophilized standards provided in the kits. Cytokine concentrations in samples were determined from the standard curve using a 5-point regression to transform mean fluorescence intensities into concentrations.

Statistical analysis

Provided the exploratory design of our study, the sample size was not predetermined. Results are shown as means and SDs. Survival curves were estimated using the Kaplan–Meier method and compared by log-rank test. Univariate and multivariate analyses of median progression-free survival (mPFS) and mOS, with stepwise variable selection, were conducted by Cox's proportional hazard regression models. Multivariate analysis was conducted using the clinical–pathologic variables with a P value <0.05 and the strongest significant molecular variables in univariate analysis (P value <0.01). Fisher exact test was used for pairwise comparisons of objective response.

The optimal cutoff thresholds for soluble biomarkers were obtained on the basis of the maximization of the Youden's statistics (J = sensitivity + specificity + 1; ref. 19) using an R-based software as described in ref. 20. Statistical analyses were performed using SPSS 24.0 statistical software (SPSS, Inc.), GraphPad Prism software program (version 6.0; GraphPad Software), and the statistical language R.

Identification of IL8 as the most significant TAK1-regulated secreted protein in nal-IRI–resistant pancreatic cancer in vivo models

To identify novel circulating markers of TAK1-pathway activation, which could serve as biomarkers for chemoresistance, we compared gene expression profiles in AsPC1, Panc1, and MDA-Panc28 pancreatic cancer cell lines transduced with lentivirus expressing TAK1-specific shRNA (shTAK1) or scramble sequence (shctrl) as control by microarray analysis (Fig. 1A). We analyzed data of the genes with significantly different levels of expression by using the Ingenuity Pathway Analysis software program, which identifies groups of genes associated with specific molecular pathways. We found a significant association of the genes in the TAK1-proficient pancreatic cancer cells lines with the IL8 signaling pathway (Fig. 1B), and we identified CXCL8, the gene coding for IL8, as the most significant downregulated gene coding for secreted protein in all shTAK1 pancreatic cancer cell lines as compared with their respective scramble controls (Fig. 1C). We confirmed a significant downregulation of CXCL8 mRNA in all shTAK1 pancreatic cancer cell lines (Fig. 1D) and a significant reduction in the IL8 expression in conditional medium from shTAK1 cell lines (Fig. 1E).

Figure 1.

Identification of IL8 as the most significant TAK1-regulated secreted protein. A, Western blot analysis for the expression of TAK1 in AsPc-1, PANC-1, and MDAPanc-28 pancreatic cancer cells transduced with lentiviruses expressing TAK1-specific small hairpin RNA (shTAK1) or a scramble sequence as control (shctrl). β-Actin was used as loading control. B, Selection of relevant biological processes and secreted protein genes by using global transcript profiling. IL8 signaling was enriched among genes differentially expressed in pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl by ingenuity pathway analyses (IPA) software. The X-axis represents the log (10) P value for enrichment, with the threshold drawn at P = 0.05. C, Levels of the most significantly downregulated (blue) and upregulated (red) genes coding for secreted proteins in pancreatic cancer cells transduced with lentiviruses expressing shTAK1 (P28T, AST, P1T) versus those expressing a shctrl (P28, AS, P1). D, Histogram shows normalized mRNA levels of IL8/β-actin in the indicated pancreatic cancer cell lines. Means and SDs are shown, ***P < 0.001, as determined by unpaired Student t test. E, Histogram shows IL8 protein levels in the conditioned media of the indicated pancreatic cancer cell lines. Means and SDs are shown, ***, P < 0.001, as determined by unpaired Student t test.

Figure 1.

Identification of IL8 as the most significant TAK1-regulated secreted protein. A, Western blot analysis for the expression of TAK1 in AsPc-1, PANC-1, and MDAPanc-28 pancreatic cancer cells transduced with lentiviruses expressing TAK1-specific small hairpin RNA (shTAK1) or a scramble sequence as control (shctrl). β-Actin was used as loading control. B, Selection of relevant biological processes and secreted protein genes by using global transcript profiling. IL8 signaling was enriched among genes differentially expressed in pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl by ingenuity pathway analyses (IPA) software. The X-axis represents the log (10) P value for enrichment, with the threshold drawn at P = 0.05. C, Levels of the most significantly downregulated (blue) and upregulated (red) genes coding for secreted proteins in pancreatic cancer cells transduced with lentiviruses expressing shTAK1 (P28T, AST, P1T) versus those expressing a shctrl (P28, AS, P1). D, Histogram shows normalized mRNA levels of IL8/β-actin in the indicated pancreatic cancer cell lines. Means and SDs are shown, ***P < 0.001, as determined by unpaired Student t test. E, Histogram shows IL8 protein levels in the conditioned media of the indicated pancreatic cancer cell lines. Means and SDs are shown, ***, P < 0.001, as determined by unpaired Student t test.

Close modal

To measure whether the expression of tumor TAK1 could correlate in vivo with circulating levels of IL8, and to demonstrate that TAK1 kinase is relevant to sustain resistance to nal-IRI, we used an orthotopic xenograft nude mouse model. AsPC1shctrl and AsPC1shTAK1 orthotopic tumor-bearing mice were randomly assigned (n = 5) to receive nal-IRI 10 mg/kg i.p. once a week or its intraperitoneal vehicle as control. At median survival duration of mice in the respective control groups, all of the mice were sacrificed and tumors and blood samples collected for analyses. We measured a significant downregulation for IL8 expression in tumor specimens excised from mice bearing shTAK1 AsPc1 tumors if compared with that in control tumors (Fig. 2A). Consistently, we demonstrated significantly lower plasma levels of IL8 in mice bearing TAK1 knockdown AsPc1 tumors than in those bearing TAK1-proficient tumors (Fig. 2B).

Figure 2.

TAK1 sustains high circulating levels of IL8 and resistance to nal-IRI- in pancreatic cancer in vivo models. A, IHC analysis. Serial paraffin sections from tumors excised from mice orthotopically inoculated with pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl were stained with antibodies to TAK1 and IL8. B, Histogram shows IL8 plasmatic concentration in mice inoculated with pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl. Means and SDs (n = 3) are shown, *, P < 0.05, as determined by unpaired Student t test. C, Antitumor activity of nal-IRI in vivo in AsPc-1 expressing shctrl pancreatic tumor orthotopic xenografts (n = 10, five mice per group). When at least three of five mice in a treatment group presented with bulky disease (tumor volume, >2,000 mm3), the median survival duration for that group was considered to be reached. At the median survival duration of the control group, all mice of control and treatment groups were euthanized by deep terminal anesthesia and blood and tumor samples were collected. Differences in weight were considered for the effects of treatments between different groups. Box-and-whiskers plot shows median and 25th to 75th percentiles of tumors weight in each group. P value not significant (>0.05), as determined by unpaired Student t test. D, Antitumor activity of nal-IRI in vivo in AsPc-1 expressing shTAK1 pancreatic tumor orthotopic xenografts (n = 10, five mice per group). Analysis conducted as described above. Box-and-whiskers plot shows median and 25th to 75th percentiles of tumors weight in each group. P value = 0.0091 as determined by unpaired Student t test.

Figure 2.

TAK1 sustains high circulating levels of IL8 and resistance to nal-IRI- in pancreatic cancer in vivo models. A, IHC analysis. Serial paraffin sections from tumors excised from mice orthotopically inoculated with pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl were stained with antibodies to TAK1 and IL8. B, Histogram shows IL8 plasmatic concentration in mice inoculated with pancreatic cancer cells transduced with lentiviruses expressing shTAK1 or a shctrl. Means and SDs (n = 3) are shown, *, P < 0.05, as determined by unpaired Student t test. C, Antitumor activity of nal-IRI in vivo in AsPc-1 expressing shctrl pancreatic tumor orthotopic xenografts (n = 10, five mice per group). When at least three of five mice in a treatment group presented with bulky disease (tumor volume, >2,000 mm3), the median survival duration for that group was considered to be reached. At the median survival duration of the control group, all mice of control and treatment groups were euthanized by deep terminal anesthesia and blood and tumor samples were collected. Differences in weight were considered for the effects of treatments between different groups. Box-and-whiskers plot shows median and 25th to 75th percentiles of tumors weight in each group. P value not significant (>0.05), as determined by unpaired Student t test. D, Antitumor activity of nal-IRI in vivo in AsPc-1 expressing shTAK1 pancreatic tumor orthotopic xenografts (n = 10, five mice per group). Analysis conducted as described above. Box-and-whiskers plot shows median and 25th to 75th percentiles of tumors weight in each group. P value = 0.0091 as determined by unpaired Student t test.

Close modal

Most importantly, mice bearing TAK1-proficient AsPc1 tumors treated with nal-IRI had tumor growth rates comparable with those in vehicle-treated control mice [tumor weight mean ± SEM (g) = 0.94 ± 0.15 vs 1.615 ± 0.305, P value = 0.0973, unpaired t test; Fig. 2C]. Conversely, mice bearing TAK1 knockdown AsPc1 tumors treated with nal-IRI experienced a statistically significant reduction in tumor growth if compared with their respective vehicle-treated controls [mean ± SEM (g) = 0.5288 ± 0.1974 vs 3.036 ± 0.7067, P value = 0.0091, unpaired t test; Fig. 2D]. We ruled out the possibility that different levels of IL8 could modulate the infiltration of tumor-associated macrophages (TAMs), and the local conversion of nal-IRI in SN-38 by measuring similarly minimal levels (less than 1 cell/field) of CD68+ cells and carboxylesterase-2 in tumor specimens excised from mice bearing shTAK1 AsPc1 tumors if compared with that in control tumors (Supplementary Fig. S1).

Altogether, these data demonstrate that IL8 is the most relevant secreted marker in vitro and in vivo for the activation of the TAK1 pathway that is responsible for a tumor cells autonomous resistance of pancreatic cancer to nal-IRI.

IL8 is the most significant predictive marker of survival in patients with metastatic pancreatic cancer treated with nal-IRI plus 5-FU/LV

To demonstrate the clinical relevance of IL8 in predicting resistance to nal-IRI, we initially collected plasma at baseline in a discovery cohort of 77 patients with gemcitabine-refractory metastatic pancreatic cancer that were prospectively enrolled between November 2016 and June 2018 within an Expanded Access Program to receive nal-IRI plus 5-FU/LV as second or further line therapy. Compliance with REMARK guidelines is reported in Supplementary Table S1. Patients' characteristics are shown in Supplementary Table S2. In this discovery cohort, median age was 64 years and 52% were male. Most of them had a normal ECOG performance status (58.4%). Half of the patients had tumors in the head of pancreas (52%). The majority of patients had increased levels of Ca19.9 (83.1%). More than one third of patients had two or more site of metastasis, and liver was the most common secondary site (70.1%). 76.7% of patients received nal-IRI + 5-FU/LV as second-line, 22.0% as third-line, and one patient (1.2%) as fourth-line treatment for metastatic disease (Supplementary Table S2). After a median follow-up of 26 months, the mPFS was 3.3 months (95% CI, 3.040–3.560) and the mOS was 7.4 months (95% CI, 8.309–12.251) for the overall population (Supplementary Figs. S2A and S2B).

Thus, we measured the plasma concentration of a panel of 20 different cytokines, chemokines, and growth factors. The optimal cutoff thresholds able to significantly predict patients' outcomes were evaluated for each cytokine (Table 1). This analysis confirmed IL8 as the circulating factor most significantly able to predict mPFS and mOS in this discovery cohort of patients. The mPFS was 3.4 months compared with 2.8 months (HR, 2.55; 95% CI, 1.39–4.67; P = 0.0017) (Fig. 3A), and the mOS was 8.9 months compared with 5.3 months (HR, 3.51; 95% CI, 1.84–6.68; P = 4.9e−05; Fig. 3B) for patients with plasma levels of IL8 at baseline lower versus higher than cutoff of 16.68 pg/mL, respectively (Supplementary Figs. S3A–S3F). Furthermore, in those 15 patients with plasma levels of IL8 at baseline higher than cutoff, we measured only one (6.7%) stable disease (SD), and no partial (PR) or complete responses. On the contrary, among those 62 patients with plasma levels of IL8 at baseline lower than cutoff, we measured 14 (23%) SD, 5 (8.1%) PR, and 1 (1.7%) CR, counting for a disease control rate of 32.7% (Supplementary Table S3).

Table 1.

Plasmatic cytokines able to significantly predict PFS and OS in the discovery cohort population.

PFSOS
Median concentration (pg/mL)RangeCutoff (pg/mL)HR (95% CI)PCutoff (pg/mL)HR (95% CI)P
Chemokines 
IL8 5.96 54.67 16.68 2.55 (1.39–4.66) 0.0017 16.68 3.51 (1.84–6.68) 4.9e-05 
MCP-1 55.27 20.41 237.7 60.02 2.21 (1.35–3.62) 0.0013 58.07 2.38 (1.46–3.89) 0.00038 
IP-10 19.56 9.73 46.85 25.49 2.11 (1.24–3.6) 0.005 25.35 2.52 (1.47–4.34) 0.00055 
MIP-1β 36.24 20.29 121.8 56.21 2.51 (1.3–4.85) 0.0046 56.21 2.39 (1.23–4–65) 0.0079 
SDF-1α 258 94.29 647.6 334 2.38 (1.24–4.55) 0.0069 334 2.06 (1.09–3.88) 0.023 
RANTES 139.31 36.67 255.1 — — — — — — 
Eotaxin 23.07 12.25 56.84 — — — — — — 
GROAα 102.91 — — — — — — 
MIP-1α 1.79 54.33 — — — — — — 
TH
IL2 12.93 31.27 — — — 6.775 0.5 (0.27–0.93) 0.026 
IL18 31.55 136.71 54 2.14 (1.21–3.8) 0.0076 25.21 2 (1.2–3.33) 0.0065 
IL12p70 4.28 72.33 — — — 1.755 0.53 (0.32–0.86) 0.0098 
IFNγ 24.53 137.69 44.27 2.62 (1.4–4.88) 0.0017 20.32 1.88 (1.11–3.2) 0.018 
TNFα 11.75 2.46 47.04 — — — — — — 
TH
IL4 218.39 6.195 2.43 (1.2–4.88) 0.01 — — — 
IL5 7.23 44.52 — — — — — — 
IL6 0.5 193.81 — — — 6.175 2 1.21–3.3 0.0061 
IL13 123.7 7.93 2.31 (1.17–4.59) 0.014 — — — 
Other cytokines and growth factors 
IL1β 13.13 — — — — — — 
GM-CSF 102.98 — — — — — — 
PFSOS
Median concentration (pg/mL)RangeCutoff (pg/mL)HR (95% CI)PCutoff (pg/mL)HR (95% CI)P
Chemokines 
IL8 5.96 54.67 16.68 2.55 (1.39–4.66) 0.0017 16.68 3.51 (1.84–6.68) 4.9e-05 
MCP-1 55.27 20.41 237.7 60.02 2.21 (1.35–3.62) 0.0013 58.07 2.38 (1.46–3.89) 0.00038 
IP-10 19.56 9.73 46.85 25.49 2.11 (1.24–3.6) 0.005 25.35 2.52 (1.47–4.34) 0.00055 
MIP-1β 36.24 20.29 121.8 56.21 2.51 (1.3–4.85) 0.0046 56.21 2.39 (1.23–4–65) 0.0079 
SDF-1α 258 94.29 647.6 334 2.38 (1.24–4.55) 0.0069 334 2.06 (1.09–3.88) 0.023 
RANTES 139.31 36.67 255.1 — — — — — — 
Eotaxin 23.07 12.25 56.84 — — — — — — 
GROAα 102.91 — — — — — — 
MIP-1α 1.79 54.33 — — — — — — 
TH
IL2 12.93 31.27 — — — 6.775 0.5 (0.27–0.93) 0.026 
IL18 31.55 136.71 54 2.14 (1.21–3.8) 0.0076 25.21 2 (1.2–3.33) 0.0065 
IL12p70 4.28 72.33 — — — 1.755 0.53 (0.32–0.86) 0.0098 
IFNγ 24.53 137.69 44.27 2.62 (1.4–4.88) 0.0017 20.32 1.88 (1.11–3.2) 0.018 
TNFα 11.75 2.46 47.04 — — — — — — 
TH
IL4 218.39 6.195 2.43 (1.2–4.88) 0.01 — — — 
IL5 7.23 44.52 — — — — — — 
IL6 0.5 193.81 — — — 6.175 2 1.21–3.3 0.0061 
IL13 123.7 7.93 2.31 (1.17–4.59) 0.014 — — — 
Other cytokines and growth factors 
IL1β 13.13 — — — — — — 
GM-CSF 102.98 — — — — — — 

Note: Median concentration, range, cutoff identified, hazard ratio (HR) and 95% CI, and P value are reported.

Figure 3.

Kaplan–Meier analysis of PFS (A) and OS (B), according to cut-off value of IL8 in the discovery cohort. A, In the discovery cohort, the median PFS was 3.4 months in low IL8 group, as compared with 2.8 months in high IL8 group. B, The median OS was 8.9 months in low IL8 group, as compared with 5.3 months in high IL8 group.

Figure 3.

Kaplan–Meier analysis of PFS (A) and OS (B), according to cut-off value of IL8 in the discovery cohort. A, In the discovery cohort, the median PFS was 3.4 months in low IL8 group, as compared with 2.8 months in high IL8 group. B, The median OS was 8.9 months in low IL8 group, as compared with 5.3 months in high IL8 group.

Close modal

Different circulating factors and cytokines with levels higher than cutoff were also significantly associated with either a shorter patients' mPFS and mOS, including the chemokines MCP-1, IP-10, MIP-1β, and SDF-1α, and the TH1 cytokines IL18 and INFγ. The same associations were not proven for the other cytokines (Table 1; Supplementary Figs. S4–S11).

Univariate analysis of correlation between IL8 plasma levels, clinical features, and survival outcomes is shown in Table 2. Among the clinical parameters analyzed, patients who previously underwent surgery had a significantly longer mOS (HR, 0.387; P = 0.012) and mPFS (HR, 0.437; P = 0.017) compared with unresectable patients. Furthermore, patients with a normal ECOG PS had a longer mPFS compared with patients with ECOG PS 1-2 (HR, 1.884; P = 0.010). To confirm the independence of the predictive value for IL8 plasma levels, we performed a multivariate analysis including IL8 and those clinical features significant at univariate analysis (P < 0.05). This analysis revealed IL8 as an independent predictor of PFS and OS in the discovery cohort of patients (Table 2).

Table 2.

Univariate and multivariate analyses of clinical characteristics and IL8 levels influencing PFS and OS of patients in the discovery cohort.

OS
UnivariateMultivariate
95% CI95% CI
VariablesHRLowerUpperPHRLowerUpperP
PS ECOG 0 vs 1–2 1.170 0.721 1.900 0.526     
Primary tumor location Head vs body/tail 0.924 0.567 1.508 0.753     
Liver metastases No vs yes 1.517 0.871 2.640 0.141     
CA19.9 (U/mL) <40 vs ≥40 1.575 0.716 3.466 0.259     
N/L ratio ≤5 vs >5 1.368 0.696 2.689 0.363     
IL8 Low vs high 3.406 1.790 6.481 0.000     
PFS 
  Univariate Multivariate 
  95% CI 95% CI 
Variables HR Lower Upper P HR Lower Upper P 
PS ECOG 0 vs 1–2 1.884 1.164 3.049 0.010 2.216 1.346 3.649 0.002 
Primary tumor location Head vs body/tail 0.850 0.529 1.366 0.502     
Liver metastases No vs yes 0.925 0.538 1.591 0.778     
CA19.9 (U/mL) <40 vs ≥40 1.832 0.828 4.057 0.135     
N/L ratio ≤5 vs >5 1.233 0.672 2.264 0.499     
IL8 Low vs high 2.505 1.367 4.590 0.003 3.090 1.649 5.791 0.000 
OS
UnivariateMultivariate
95% CI95% CI
VariablesHRLowerUpperPHRLowerUpperP
PS ECOG 0 vs 1–2 1.170 0.721 1.900 0.526     
Primary tumor location Head vs body/tail 0.924 0.567 1.508 0.753     
Liver metastases No vs yes 1.517 0.871 2.640 0.141     
CA19.9 (U/mL) <40 vs ≥40 1.575 0.716 3.466 0.259     
N/L ratio ≤5 vs >5 1.368 0.696 2.689 0.363     
IL8 Low vs high 3.406 1.790 6.481 0.000     
PFS 
  Univariate Multivariate 
  95% CI 95% CI 
Variables HR Lower Upper P HR Lower Upper P 
PS ECOG 0 vs 1–2 1.884 1.164 3.049 0.010 2.216 1.346 3.649 0.002 
Primary tumor location Head vs body/tail 0.850 0.529 1.366 0.502     
Liver metastases No vs yes 0.925 0.538 1.591 0.778     
CA19.9 (U/mL) <40 vs ≥40 1.832 0.828 4.057 0.135     
N/L ratio ≤5 vs >5 1.233 0.672 2.264 0.499     
IL8 Low vs high 2.505 1.367 4.590 0.003 3.090 1.649 5.791 0.000 

To validate the negative predictive value of IL8 in patients treated with nal-IRI plus 5-FU/LV, we analyzed the association of IL8 levels and clinical outcomes in an external validation cohort of patients treated in a different center. Patients' characteristics are shown in Supplementary Table S4. The mPFS was 3.3 months (95% CI, 4.504-7.804) and the mOS was 7.4 months (95% CI, 8.309–12.251) for the overall population (Supplementary Figs. S12A and S12B). In this validation cohort, we confirmed IL8 as significantly associated with mPFS and mOS. The mPFS was 4.6 months compared with 1.8 months (HR, 2.84; 95% CI, 1.41–5.72; P = 0.0023; Fig. 4A), and the mOS was 9.7 months compared with 5.4 months (HR, 2.32; 95% CI, 1.18–4.56; P = 0.012; Fig. 4B) for patients with serum levels of IL8 at baseline lower versus higher than cutoff, respectively (Supplementary Fig. S13).

Figure 4.

Kaplan–Meier analysis of PFS (A) and OS (B), according to cut-off value of IL8 in the validation cohort. A, In the validation cohort, the median PFS was 4.6 months in low IL8 group, as compared with 1.8 months in high IL8 group. B, The median OS was 9.7 months in low IL8 group, as compared with 5.4 months in high IL8 group. IL8 cut off levels (pg/mL), HR and P value are indicated.

Figure 4.

Kaplan–Meier analysis of PFS (A) and OS (B), according to cut-off value of IL8 in the validation cohort. A, In the validation cohort, the median PFS was 4.6 months in low IL8 group, as compared with 1.8 months in high IL8 group. B, The median OS was 9.7 months in low IL8 group, as compared with 5.4 months in high IL8 group. IL8 cut off levels (pg/mL), HR and P value are indicated.

Close modal

In this study, we sought to identify circulating biomarkers useful to estimate the activation of TAK1 pathway, one of the most relevant molecular mechanisms responsible for the resistance of pancreatic cancer to the proapoptotic activity of chemotherapeutic treatments (5). We demonstrated that the chemokine IL8 is the secreted factor most significantly regulated by TAK1 activation. Most importantly, we demonstrated for the first time that IL8 could be, indeed, a useful biomarker to predict resistance to the novel nanotechnologic agent nal-IRI in two large discovery and external validation cohorts of patient affected by gemcitabine-refractory advanced pancreatic cancer.

The most appropriate systemic regimen for patients with advanced pancreatic cancer progressing under a first-line gemcitabine-containing regimen largely remains a matter of debate. Two randomized phase III trials that explored the role of oxaliplatin-based chemotherapy in patients progressing to a first-line single-agent gemcitabine therapy reached conflicting results. No randomized phase III trials explored standard irinotecan-containing regimens in this setting (21, 22) To date, NAPOLI-1 remains the largest phase III study in second-line metastatic pancreatic cancer (12). This trial led to the regulatory approval of nal-IRI in the United States in 2015 for the treatment of metastatic pancreatic cancer in combination with 5-FU/LV after progression on gemcitabine-based therapy. Nonetheless, the lack of randomized trials comparing nal-IRI plus 5-FU/LV to standard irinotecan-containing regimens such as FOLFIRI, and the exceeding cost-effectiveness ratio per quality-adjusted life year gained for nal-IRI plus 5-FU/LV limited the recommendation for marketing authorization of this regimen in several European countries and in United Kingdom (23). In this regard, the identification of biomarkers for the selection of those patients that could gain the largest advantage by this regimen remains a significant priority. Here, we demonstrated that IL8 is the most significant independent factor for PFS and OS in two different cohorts of patients among a series of cytokines in patients with advanced pancreatic cancer receiving nal-IRI plus 5-FU/LV treatment, representing a potential biomarker for patients' selection and, in turn, for potentially improving clinical-effectiveness evidence and cost-effectiveness results for this regimen.

IL8 is a chemokine mainly produced by tumor cells, and its main functions include angiogenesis, survival signaling for cancer stem cells, and attraction of myeloid-derived suppressor cells (24). We recently discovered IL8 as part of a proinflammatory cytokine signature inducing resistance to antiangiogenic drugs in patients with pancreatic and colorectal cancer through the recruitment of myeloid CD11b+ cells (25–27). Previous studies demonstrated a central role for TAK1 in sustaining IL1-induced transcription and mRNA stabilization, as well as expression of endogenous IL8 (28). TAK1 is also part of an intracellular signaling axis responsible for Death receptor 3-mediated expression of IL8 (29). More recently, an autocrine loop between IL8 and TAK1 has been demonstrated given that IL8 can activate TAK1/NF-κB signaling via the CXCR2 receptor in models of ovarian cancer (30). In this study, we demonstrated that IL8 is the secreted factor most significantly regulated by TAK1 activation, one of the most potent mediators of chemoresistance in pancreatic cancer through the activation of AP-1 and NF-κB transcription factors. AP-1 and NF-κB are, indeed, recognized as potent transcription factors cooperating for the induction of IL8 in human pancreatic cancer cells by hypoxia (31).

The stable nanoliposome formulation of nal-IRI has been developed to improve the therapeutic index of irinotecan throughout an active accumulation and conversion in TAMs, allowing for a more efficient delivery of its active metabolite SN-38 within the tumor microenvironment. However, the effect of tumor derived IL8 on TAMs in murine models has been only limitedly explored (32) as an homologue of human CXCL8 gene is completely absent from the genome of rodents (33). Previous studies in mice carrying a bacterial artificial chromosome encompassing the entire human CXCL8 gene showed an increased mobilization of immature CD11b+Gr-1+ myeloid cells (34). In our study, we ruled out a putative mechanistic association between the infiltration and the carboxylesterase activity of TAMs and IL8 levels as cause for the lack of activity of nal-IRI in pancreatic cancer models. The activation of transcription factors, including AP-1, NF-κB, and YAP/TAZ (7–9), remains the most relevant mechanism for the treatment resistance sustained by TAK1, and IL8 is the most reliable circulating factor to measure the activation of this pathway.

This study, however, had one limitation in lacking a control cohort including untreated patients with pancreatic cancer to distinguish between a merely prognostic or a predictive value for TAK1-regulated IL8. Although its relevant mechanistic role in cancer pathogenesis, a prognostic value for IL8 in pancreatic cancer has been, however, excluded by several previous studies. In one of the initial series including healthy volunteers and patients with pancreatic cancer with resectable, locally advanced or metastatic disease, IL8 levels were higher in patients with cancer than in healthy controls but were not associated with survival differences (35). More recently, we conducted the most comprehensive profiling of cytokines in the largest prospective cohort of patients with resectable pancreatic cancer to date and IL8 had no prognostic value in this cohort of patients (36). Excluded a prognostic role for IL8, we acknowledge that its negative predictive value could be although extended also to other cytotoxic agents different from nal-IRI that induce in pancreatic cancer cells a proapoptotic stimuli by which a TAK1 activation mirrored by IL8 could protect.

In conclusion, we identified for the first time IL8 as the most significant circulating biomarker for predicting chemoresistance sustained by TAK1 pathway in pancreatic cancer. The expression of this chemokine could be useful to select those patients with pancreatic cancer that could benefit from the novel nanotechnologic agent nal-IRI in the larger extent.

S. Lonardi reports grants and personal fees from Amgen, personal fees from Bayer and Merck Serono and personal fees from Lilly, Servier, Roche, and BMS outside the submitted work. D. Melisi receives research funding by Servier and had a consulting role with Shire. No potential conflicts of interest were disclosed by the other authors.

V. Merz: Conceptualization, data curation, formal analysis, validation, investigation, methodology, writing-original draft, writing-review and editing. C. Zecchetto: Investigation. R. Santoro: Conceptualization, data curation, formal analysis, investigation, methodology, writing-original draft, writing-review and editing. F. Simionato: Investigation. F. Sabbadini: Investigation. D. Mangiameli: Investigation. G. Piro: Investigation. A. Cavaliere: Investigation. M. Deiana: Investigation. M.T. Valenti: Resources. D. Bazan: Resources and investigation. V. Fedele: Investigation. S. Lonardi: Resources and investigation. D. Melisi: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, validation, methodology, writing-original draft, writing-review and editing.

This work was supported by the Investigator Grants nos. 19111 and 23719 and 5 × 1000 Grant No. 12182 through the Associazione Italiana per la Ricerca sul Cancro (AIRC), by the Ricerca Finalizzata 2016 grant GR-2016-02361134 through the Italian Ministry of Health, and by the “Nastro Viola” and “Voglio il Massimo” associations of patients' donations to D. Melisi. Part of the work was performed at the Laboratorio Universitario di Ricerca Medica (LURM) Research Center, University of Verona. We thank Dr. Hayley Louise Salt for data entry and administrative support.

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.
Melisi
D
,
Calvetti
L
,
Frizziero
M
,
Tortora
G
. 
Pancreatic cancer: systemic combination therapies for a heterogeneous disease
.
Curr Pharm Des
2014
;
20
:
6660
9
.
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
:
2913
21
.
3.
Collaborators
GBDPC
. 
The global, regional, and national burden of pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017
.
Lancet Gastroenterol Hepatol
2019
;
4
:
934
47
.
4.
Tamburrino
A
,
Piro
G
,
Carbone
C
,
Tortora
G
,
Melisi
D
. 
Mechanisms of resistance to chemotherapeutic and anti-angiogenic drugs as novel targets for pancreatic cancer therapy
.
Frontiers in pharmacology
2013
;
4
:
56
.
5.
Santoro
R
,
Carbone
C
,
Piro
G
,
Chiao
PJ
,
Melisi
D
. 
TAK-ing aim at chemoresistance: the emerging role of MAP3K7 as a target for cancer therapy
.
Drug Resist Updat
2017
;
33–35
:
36
42
.
6.
Zhuang
Z
,
Ju
HQ
,
Aguilar
M
,
Gocho
T
,
Li
H
,
Iida
T
, et al
IL1 receptor antagonist inhibits pancreatic cancer growth by abrogating NF-kappaB activation
.
Clin Cancer Res
2016
;
22
:
1432
44
.
7.
Carbone
C
,
Melisi
D
. 
NF-kappaB as a target for pancreatic cancer therapy
.
Expert Opin Ther Targets
2012
;
2
:
S1
10
.
8.
Melisi
D
,
Chiao
PJ
. 
NF-kappa B as a target for cancer therapy
.
Expert Opin Ther Targets
2007
;
11
:
133
44
.
9.
Santoro
R
,
Zanotto
M
,
Simionato
F
,
Zecchetto
C
,
Merz
V
,
Cavallini
C
, et al
Modulating TAK1 expression inhibits YAP and TAZ oncogenic functions in pancreatic cancer
.
Mol Cancer Ther
2019
;
19
:
247
57
.
10.
Melisi
D
,
Xia
Q
,
Paradiso
G
,
Ling
J
,
Moccia
T
,
Carbone
C
, et al
Modulation of pancreatic cancer chemoresistance by inhibition of TAK1
.
J Natl Cancer Inst
2011
;
103
:
1190
204
.
11.
Piro
G
,
Giacopuzzi
S
,
Bencivenga
M
,
Carbone
C
,
Verlato
G
,
Frizziero
M
, et al
TAK1-regulated expression of BIRC3 predicts resistance to preoperative chemoradiotherapy in oesophageal adenocarcinoma patients
.
Br J Cancer
2015
;
113
:
878
85
.
12.
Wang-Gillam
A
,
Li
CP
,
Bodoky
G
,
Dean
A
,
Shan
YS
,
Jameson
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
:
545
57
.
13.
Hubner
RA
,
Cubillo
A
,
Blanc
JF
,
Melisi
D
,
Von Hoff
DD
,
Wang-Gillam
A
, et al
Quality of life in metastatic pancreatic cancer patients receiving liposomal irinotecan plus 5-fluorouracil and leucovorin
.
Eur J Cancer
2019
;
106
:
24
33
.
14.
Pelzer
U
,
Blanc
JF
,
Melisi
D
,
Cubillo
A
,
Von Hoff
DD
,
Wang-Gillam
A
, et al
Quality-adjusted survival with combination nal-IRI+5-FU/LV vs. 5-FU/LV alone in metastatic pancreatic cancer patients previously treated with gemcitabine-based therapy: a Q-TWiST analysis
.
Br J Cancer
2017
;
116
:
1247
53
.
15.
Aprile
G
,
Negri
FV
,
Giuliani
F
,
De Carlo
E
,
Melisi
D
,
Simionato
F
, et al
Second-line chemotherapy for advanced pancreatic cancer: which is the best option?
Crit Rev Oncol Hematol
2017
;
115
:
1
12
.
16.
Dalla Pozza
E
,
Dando
I
,
Biondani
G
,
Brandi
J
,
Costanzo
C
,
Zoratti
E
, et al
Pancreatic ductal adenocarcinoma cell lines display a plastic ability to bidirectionally convert into cancer stem cells
.
Int J Oncol
2015
;
46
:
1099
108
.
17.
Melisi
D
,
Ossovskaya
V
,
Zhu
C
,
Rosa
R
,
Ling
J
,
Dougherty
PM
, et al
Oral poly(ADP-ribose) polymerase-1 inhibitor BSI-401 has antitumor activity and synergizes with oxaliplatin against pancreatic cancer, preventing acute neurotoxicity
.
Clin Cancer Res
2009
;
15
:
6367
77
.
18.
Carbone
C
,
Piro
G
,
Fassan
M
,
Tamburrino
A
,
Mina
MM
,
Zanotto
M
, et al
An angiopoietin-like protein 2 autocrine signaling promotes EMT during pancreatic ductal carcinogenesis
.
Oncotarget
2015
;
6
:
13822
34
.
19.
Youden
WJ
. 
Index for rating diagnostic tests
.
Cancer
1950
;
3
:
32
5
.
20.
Budczies
J
,
Klauschen
F
,
Sinn
BV
,
Gyorffy
B
,
Schmitt
WD
,
Darb-Esfahani
S
, et al
Cutoff Finder: a comprehensive and straightforward Web application enabling rapid biomarker cutoff optimization
.
PLoS One
2012
;
7
:
e51862
.
21.
Oettle
H
,
Riess
H
,
Stieler
JM
,
Heil
G
,
Schwaner
I
,
Seraphin
J
, 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
:
2423
9
.
22.
Gill
S
,
Ko
YJ
,
Cripps
C
,
Beaudoin
A
,
Dhesy-Thind
S
,
Zulfiqar
M
, et al
PANCREOX: a randomized phase III study of fluorouracil/leucovorin with or without oxaliplatin for second-line advanced pancreatic cancer in patients who have received gemcitabine-based chemotherapy
.
J Clin Oncol
2016
;
34
:
3914
20
.
23.
Fleeman
N
,
Abdulla
A
,
Bagust
A
,
Beale
S
,
Richardson
M
,
Stainthorpe
A
, et al
Pegylated liposomal irinotecan hydrochloride trihydrate for treating pancreatic cancer after gemcitabine: an evidence review group perspective of a NICE single technology appraisal
.
Pharmacoeconomics
2018
;
36
:
289
99
.
24.
Alfaro
C
,
Sanmamed
MF
,
Rodriguez-Ruiz
ME
,
Teijeira
A
,
Onate
C
,
Gonzalez
A
, et al
Interleukin-8 in cancer pathogenesis, treatment and follow-up
.
Cancer Treat Rev
2017
;
60
:
24
31
.
25.
Carbone
C
,
Moccia
T
,
Zhu
C
,
Paradiso
G
,
Budillon
A
,
Chiao
PJ
, et al
Anti-VEGF treatment-resistant pancreatic cancers secrete proinflammatory factors that contribute to malignant progression by inducing an EMT cell phenotype
.
Clin Cancer Res
2011
;
17
:
5822
32
.
26.
Carbone
C
,
Tamburrino
A
,
Piro
G
,
Boschi
F
,
Cataldo
I
,
Zanotto
M
, et al
Combined inhibition of IL1, CXCR1/2, and TGFbeta signaling pathways modulates in-vivo resistance to anti-VEGF treatment
.
Anticancer Drugs
2016
;
27
:
29
40
.
27.
Carbone
C
,
Piro
G
,
Simionato
F
,
Ligorio
F
,
Cremolini
C
,
Loupakis
F
, et al
Homeobox B9 mediates resistance to anti-VEGF therapy in colorectal cancer patients
.
Clin Cancer Res
2017
;
23
:
4312
22
.
28.
Holtmann
H
,
Enninga
J
,
Kalble
S
,
Thiefes
A
,
Dorrie
A
,
Broemer
M
, et al
The MAPK kinase kinase TAK1 plays a central role in coupling the interleukin-1 receptor to both transcriptional and RNA-targeted mechanisms of gene regulation
.
J Biol Chem
2001
;
276
:
3508
16
.
29.
Su
WB
,
Chang
YH
,
Lin
WW
,
Hsieh
SL
. 
Differential regulation of interleukin-8 gene transcription by death receptor 3 (DR3) and type I TNF receptor (TNFRI)
.
Exp Cell Res
2006
;
312
:
266
77
.
30.
Yung
MM
,
Tang
HW
,
Cai
PC
,
Leung
TH
,
Ngu
SF
,
Chan
KK
, et al
GRO-alpha and IL-8 enhance ovarian cancer metastatic potential via the CXCR2-mediated TAK1/NFkappaB signaling cascade
.
Theranostics
2018
;
8
:
1270
85
.
31.
Shi
Q
,
Le
X
,
Abbruzzese
JL
,
Wang
B
,
Mujaida
N
,
Matsushima
K
, et al
Cooperation between transcription factor AP-1 and NF-kappaB in the induction of interleukin-8 in human pancreatic adenocarcinoma cells by hypoxia
.
J Interferon Cytokine Res
1999
;
19
:
1363
71
.
32.
Xiao
P
,
Long
X
,
Zhang
L
,
Ye
Y
,
Guo
J
,
Liu
P
, et al
Neurotensin/IL-8 pathway orchestrates local inflammatory response and tumor invasion by inducing M2 polarization of Tumor-Associated macrophages and epithelial-mesenchymal transition of hepatocellular carcinoma cells
.
Oncoimmunology
2018
;
7
:
e1440166
.
33.
Lee
J
,
Cacalano
G
,
Camerato
T
,
Toy
K
,
Moore
MW
,
Wood
WI
. 
Chemokine binding and activities mediated by the mouse IL-8 receptor
.
J Immunol
1995
;
155
:
2158
64
.
34.
Asfaha
S
,
Dubeykovskiy
AN
,
Tomita
H
,
Yang
X
,
Stokes
S
,
Shibata
W
, et al
Mice that express human interleukin-8 have increased mobilization of immature myeloid cells, which exacerbates inflammation and accelerates colon carcinogenesis
.
Gastroenterology
2013
;
144
:
155
66
.
35.
Ebrahimi
B
,
Tucker
SL
,
Li
D
,
Abbruzzese
JL
,
Kurzrock
R
. 
Cytokines in pancreatic carcinoma: correlation with phenotypic characteristics and prognosis
.
Cancer
2004
;
101
:
2727
36
.
36.
Piro
G
,
Simionato
F
,
Carbone
C
,
Frizziero
M
,
Malleo
G
,
Zanini
S
, et al
A circulating TH2 cytokines profile predicts survival in patients with resectable pancreatic adenocarcinoma
.
Oncoimmunology
2017
;
6
:
e1322242
.

Supplementary data