Skip to main content
Erschienen in: BMC Cancer 1/2020

Open Access 01.12.2020 | Research article

Interferon-beta enhances sensitivity to gemcitabine in pancreatic cancer

verfasst von: Amber Blaauboer, Stephanie Booy, Peter M. van Koetsveld, Bas Karels, Fadime Dogan, Suzanne van Zwienen, Casper H. J. van Eijck, Leo J. Hofland

Erschienen in: BMC Cancer | Ausgabe 1/2020

Abstract

Background

Adjuvant gemcitabine for pancreatic cancer has limited efficacy in the clinical setting. Impaired drug metabolism is associated with treatment resistance. We aimed to evaluate the chemosensitising effect of interferon-beta (IFN-β).

Methods

BxPC-3, CFPAC-1, and Panc-1 cells were pre-treated with IFN-β followed by gemcitabine monotherapy. The effect on cell growth, colony formation, and cell cycle was determined. RT-qPCR was used to measure gene expression. BxPC-3 cells were used in a heterotopic subcutaneous mouse model.

Results

IFN-β increased sensitivity to gemcitabine (4-, 7.7-, and 1.7-fold EC50 decrease in BxPC-3, CFPAC-1, and Panc-1, respectively; all P < 0.001). Findings were confirmed when assessing colony formation. The percentage of cells in the S-phase was significantly increased after IFN-β treatment only in BxPC-3 and CFPAC-1 by 12 and 7%, respectively (p < 0.001 and p < 0.05, respectively). Thereby, IFN-β upregulated expression of the drug transporters SLC28A1 in BxPC-3 (252%) and SLC28A3 in BxPC-3 (127%) and CFPAC-1 (223%) (all p < 0.001). In vivo, combination therapy reduced tumor volume with 45% (P = 0.01). Both ex vivo and in vivo data demonstrate a significant reduction in the number of proliferating cells, whereas apoptosis was increased.

Conclusions

For the first time, we validated the chemosensitising effects of IFN-β when combined with gemcitabine in vitro, ex vivo, and in vivo. This was driven by cell cycle modulation and associated with an upregulation of genes involving intracellular uptake of gemcitabine. The use of IFN-β in combination with gemcitabine seems promising in patients with pancreatic cancer and needs to be further explored.
Hinweise
Amber Blaauboer and Stephanie Booy contributed equally to this work.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12885-020-07420-0.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
dFdC
2′,2′-difluoro 2′-deoxycytidine
dFdU
2′2’ difluorodeoxyuridine
β-actin
Bèta-actin
CDA
Cytidine deaminase
CMPK1
Cytidine monophosphate kinase 1
dCK
Deoxycytidine kinase
DCTD
Deoxycytidylate deaminase
EF
Efficiency factor
EGF-R
Epidermal growth factor receptor
GEM
Gemcitabine
GUSB
Glucuronidase bèta
EC50
Half maximal effective concentration
hCNT1
Human concentrative nucleoside transporters 1
hCNT3
Human concentrative nucleoside transporters 3
hENT1
Human equilibrative nucleoside transporter 1
HPRT1
7 hypoxanthine-guanine phosphoribosyl transferase 1
IFN-β
Interferon-beta
IFIT1
Interferon induced protein with tetratricopeptide repeats 1
ISGs
Interferon stimulated genes
i.p.
Intraperitoneal
Mx1
MX dynamin like GTPase 1
MxA
Myxovirus resistant protein A
NF-κB
Nuclear factor kappa-beta
NME1
Nucleoside diphosphate kinase A
NT5E
5′-nucleotidases
OAS1A
2′-5′ oligoadenylate synthetase 1a
PE
Plating efficiency
SLC28A1
Solute carrier family 28 member 1
SLC28A3
Solute carrier family 28 member 3
SLC29A1
Solute carrier family 29 member 1

Background

Pancreatic cancer is a highly malignant disease, with an estimated 5-years survival of 9% [1]. At diagnosis, approximately 15% of the patients have resectable disease (stage I or II), which indicates surgery followed by systemic therapy [2]. The CONKO-001 trial is the first randomized controlled trial in pancreatic cancer that reported longer overall survival (OS) rates in patients treated with adjuvant gemcitabine compared with observation alone. However, the median OS with this regimen is still only 22 months [3]. Recently, Conroy et al. demonstrated a significant survival benefit with modified FOLFIRINOX compared to gemcitabine alone (PRODIGE-24 trial) in highly selected patients [4]. In addition, new chemotherapeutic strategies in combination with gemcitabine are currently explored and also improved the prognosis of pancreatic cancer [4, 5]. However, these new regimens are associated with severe toxicity in comparison with gemcitabine alone. Since the majority of patients are elderly or have serious comorbidity, gemcitabine is often the only therapeutic option. The resistance of this chemotherapeutic is still a major impediment of successful systemic treatment for patients diagnosed with pancreatic cancer.
The limited efficacy of gemcitabine has been associated with impaired drug metabolism, hindering its intracellular uptake and activation [6]. Gemcitabine, 2′,2′-difluoro 2′-deoxycytidine (dFdC), is a hydrophilic pro-drug that requires cellular uptake and intracellular phosphorylation. Several transporters have been identified, but its major uptake is through hENT1, hCNT1, and hCNT3 (encoded by SLC29A1, SLC28A1, and SLC28A3, respectively) [7]. Expression levels of these transporters correlate with gemcitabine sensitivity and OS in pancreatic cancer, making them good predictive markers [8, 9].
Once inside the cell, gemcitabine requires a serial of phosphorylation by multiple kinases to become pharmacologically active. The cytotoxic activity of gemcitabine is a result of several inhibitory actions on DNA synthesis. Incorporation into new DNA strands as the cell replicates is the most likely mechanism by which gemcitabine causes cell death. This incorporation creates an irreparable error (masked chain termination) that leads to inhibition of DNA synthesis, and thus apoptosis.
Deoxycytidine kinase (dCK) catalyzes the initial and rate-limiting monophosphorylation of gemcitabine. However, the majority (~ 90%) of intracellular gemcitabine is directly inactivated by deamination by cytidine deaminase (CDA) to form 2′2’ difluorodeoxyuridine (dFdU), which is subsequently degraded and excreted out of the cell [7]. Thus, deficiency in dCK is a major contributor to gemcitabine resistance, while upregulation of CDA has been shown to confer gemcitabine resistance in pancreatic cancer [10, 11].
Prior studies have proposed a potential role for type I IFNs (IFN-α and -β) in combination with gemcitabine in the treatment of pancreatic cancer. IFN-α and -β were originally identified as immunomodulatory cytokines, due to their antiviral activity. Further characterization of their biological effect revealed a wide range of potential anti-tumor effects, e.g. inhibition of cell proliferation, induction of apoptosis, and cell cycle arrest. Importantly, type I IFNs have been shown to sensitize cancer cells to chemo- and radiotherapy [1214].
Both IFN-α and -β interact with the type I IFN receptor complex, which initiates the JAK/STAT downstream pathway, resulting in subsequent transcription of interferon stimulated genes (ISGs). These ISGs encode for numerous proteins that initiates different IFN activities, including anti-tumor effects, immunoregulatory effects, and other host effects [15].
While prior research has primarily focused on the anti-tumor activities of IFN-α, studies have reported that the direct anti-tumor effects of IFN-β are much stronger, and elicited at much lower concentrations, as compared to IFN-α [16, 17]. However, there are no animal or clinical studies that investigated the use of concomitant adjuvant IFN-β therapy in the treatment of pancreatic cancer.
In the present study, we aimed to further explore the potential chemosensitising effect of IFN-β and studied the mechanism of action in three human pancreatic cancer cells lines. For the first time, we demonstrate the interaction between IFN-β and the expression of genes involved in gemcitabine transport and metabolism. In addition, we evaluated the effects of IFN-β and gemcitabine in a pancreatic cancer xenograft tissue slice model. Finally, we confirmed the anti-tumor effect of IFN-β in combination with gemcitabine in a heterotopic pancreatic cancer mouse model.

Methods

Cell culture and compounds

Three human pancreatic cancer cell lines (BxPC-3, CFPAC-1, and Panc-1) were used and obtained from American Type Culture Collection (Rockville, MD, USA). Short tandem repeat profiling using a Powerplex Kit (Promega, Leiden, the Netherlands) of the cells gave consistent results with the ATCC database. Cells were confirmed as mycoplasma-free. Culture conditions were described in detail previously [18]. Human recombinant IFN-β-1a (Rebif, Rockland, MA, USA) and gemcitabine (Sigma-Aldrich, Zwijndrecht, the Netherlands) stock solutions, prepared in H2O, were stored at 4 °C. After trypsinization, cells were plated at the appropriate density in order to obtain 80% confluency at the end of the experiment. The next day, incubations with the indicated compounds were initiated and control cells were vehicle treated. For seven-day experiments, both drug compounds and medium were refreshed after 3 days. All cell culture experiments were carried out at least twice in quadruplicate.

Cell proliferation assay

Treatment effects were assessed on DNA amount as measure of cell number. After treatment, media were removed and plates were stored at − 20 °C until DNA measurement. Measurement of total DNA was performed with the bisbenzimide fluorescent dye (Hoechst 33258, Sigma-Aldrich, Zwijndrecht, the Netherlands) as previously described [19].

Colony-forming assay

Plates were coated with 1 mL poly-L-lysine (10 μg/mL), where after 1500, 200, or 300 cells were plated in a 6 wells plate for BxPC-3, CFPAC-1, and Panc-1, respectively. After 1 day, drug treatment was initiated. After treatment, media were removed and refreshed without drugs. When colonies contained at least 50 cells (2 weeks for all cell lines), cells were washed and stained with haematoxylin. Amount and size of colonies was measured using MultiImage light cabinet (Alpha Innotech) and the ImageJ software. Plating efficiency (PE) was calculated as the mean number of colonies/number of plated cells for control cultures not exposed to drugs. Surviving fraction was calculated as the mean number of colonies/(number of inoculated cells x PE).

Cell cycle analysis

After treatment, cells were harvested, washed with NaCl, fixed with ice-cold 70% EtOH, and stored at − 20 °C until analysis. Analyses were performed using the Muse® Cell Cycle Assay Kit utilizing Muse™ Cell Analyser (Merck Millipore, Amsterdam, the Netherlands).

Real-time quantitative PCR

Total RNA was isolated using the High Pure RNA Purification Kit (Roche). Yield and purity were assessed with the nanodrop. cDNA was synthesized from mRNA template by reversed transcription. To synthesize cDNA, 500 ng mRNA template was added to 40 μL Super RT buffer (Thermofisher Scientific, the Netherlands) containing 40 nmol dNTP, 20 U RNA’sin, 15 ng oligo-dT, 4 U Super RT. After 1-h incubation at 40 °C, cDNA was five times diluted. 7.5 μL TaqMan Universal PCR Master Mix (Applied Biosystems) was mixed with concentrations of the used primers and probes, with 5 μL cDNA template. The RT-PCR reaction was performed using the 7900HT Fast Real-Time PCR System. Three housekeeping genes (HPRT, β-actin, and GUSB) were used to normalize mRNA levels using the Vandesompele method (Thermo Fisher Scientific, Breda, the Netherlands) (Table S1) [20].

Animals and heterotopic injection of BxPC-3 tumor cells

Male athymic Balb/C nude mice (commercially obtained from Harlan laboratories, UK ltd) of 8 weeks old, were used and kept in a barrier facility under HEPA filtration. BxPC-3 cells (1 × 106/100 μl PBS) were subcutaneously injected at the flank after which the mice were randomized into four groups (n = 8 each). A separate group (n = 4), which did not receive any treatment, was used for tissue slice experiments. All mouse experiments were controlled by the animal welfare committee (IvD) of the Erasmus Medical Center and approved by the national central committee of animal experiments (CCD) under the protocol number 105–12-52, in accordance with the Dutch Act on Animal Experimentation and EU Directive 2010/63/EU.

Therapy and assessment of tumor size

Tumor size and body weight was measured twice weekly. Tumor volumes were calculated as (lengthxwidth)1.5x(π/6). Treatment was started when tumor volumes reached ~150mm3. Mice in the control group and IFN-β group received five times a week, on consecutive days, an i.p. injection of 100 μl of 0.9% NaCl or 1.5 × 105 IU of IFN-β. Mice in the gemcitabine group received on day 2 and 4 40 mg/kg gemcitabine i.p. Mice in the combination group recieved five times a week, on consecutive days, an injection of 1.5 × 105 IU of IFN-β i.p. and, on day 2 and 4, an 40 mg/kg gemcitabine i.p.

Necropsy procedures

Mice were sacrificed by cervical dislocation under isoflurane anesthesia after 4 weeks of treatment, when tumor volume reached 2000mm3 (1500mm3 for tumors of mice used in the tissue slice experiments), or when the wellbeing (i.e. weight loss, lethargy, tumor ulceration) of the mice could no longer be maintained.
During necropsy, tumors were resected and tumor weight and volume were measured. Tumors were divided into three parts and subsequently snap frozen in liquid nitrogen, embedded in Tissue-Tek (Sakura Finetek, Zoeterwoude, the Netherlands) for cryosectioning and fixed in freshly prepared 4% formaldehyde solution, and prepared for paraffin sectioning. Tumors were harvested, weighted and fixed in 4% formaldehyde.

Tissue slicing and slice culture

When tumors reached a volume of 1500 mm3, mice were sacrificed and necropsy was performed as described above. After resection, tumors were washed twice with Hanks’ Balanced Salt Solution (HBSS) supplemented with penicillin (1 × 105 U/L), streptomycin (1000 IU/ml) and fungizone (30 μg/ml).
After the vibrocheck (measurement of vertical deflection) was performed, tumor specimens, buffered in ice-cold HBSS, were cut, with stainless steel razor blades, into slices of 200 μm using the Leica vibrating blade microtome VT1000 S (Leica, Wetzlar, Germany). After slicing, samples were washed once more and transferred into six-well multiplates containing 5 ml of culture medium consisting of Dulbecco’s Modified Eagle’s Medium: nutrient mixture F-12 (DMEM/F-12), supplemented with penicillin (1 × 105 U/L), streptomycin (1000 IU/ml) and 10% FCS. Consecutive slices were used for the experiments (minimum of 15 slices per tumor).
Tissue culture plates were placed in a humidified incubator at 5% CO2 and continuously shaken (60 rounds/min) at 37 °C up to 4 days post slicing. Media and supplements were obtained from GIBCO Bio-cult Europe (Invitrogen, Breda, The Netherlands). After 24 h, media were refreshed and slices were incubated with IFN-β (100 IU/ml), gemcitabine (1 ng/ml), or with the combination of IFN-β and gemcitabine. After 72 h of incubation, tissue slices were harvested fixed in freshly prepared 4% formaldehyde solution and prepared (in upright position) for standard paraffin sectioning.

Immunohistochemistry

The formalin fixed and paraffin embedded sections (5 μm thick) were treated for immunohistochemistry as described previously [21]. Briefly, sections were deparaffinised and rehydrated, followed by heat induced epitope retrieval, rinsed (TRIS/Tween 0.5% (pH 8.0)) and blocked (hydrogen peroxide 3% in PBS) for 15 min before incubation with the primary antibodies for Caspase-3 and Ki-67 (both overnight at 4 °C). For negative controls, the primary antibody was omitted. The Dako Real EnVision Detection System kit (Dako Detection System, Dako Denmark, Glostrup, Denmark) was used to visualize the bound antibody after which the slides were counterstained with haematoxylin and coverslipped. The rabbit monoclonal Cleaved Caspase-3 (Asp175) antibody (cell signalling technology, Beverly, MA, USA) was used at a dilution of 1:750. The mouse monoclonal Ki-67 antibody (Dako Detection System) was used at a dilution of 1:400.

Immunohistochemical analysis

All sections were evaluated and counted by AB using CellProfiler (cell image analysis software) [22]. Apoptosis and cell proliferation were assessed by counting the total number of caspase-3 or Ki-67 positive tumor cells per high-power field (Olympus, Nikon Eclipse E400, HPF × 40 objective). For the analysis, a minimum of 3 HPF were used to evaluate cells with a positive and negative staining.

Statistical analysis

GraphPad Prism version 3.0 (GraphPad Software) was used for statistical analysis. Non-linear regression curves were used to calculate the half maximal effective concentration (EC50) on cell growth. For analysis of the combination therapy, effect of IFN-β was set on 100% and used as control. One-way ANOVA test with Tukey’s multiple comparisons test was used for comparisons among treatment groups. Regarding in vivo experiments, differences between groups were evaluated by the Mann-Whitney t-test. In all analyses, values of p < 0.05 were considered as significant. Data are indicated as mean ± SEM, unless specified otherwise.

Results

IFN-β and gemcitabine dose-dependently inhibit proliferation in human pancreatic cancer cells

To assess the growth inhibitory effect of IFN-β and gemcitabine, cells were treated with increasing concentrations (10–10.000 IU/ml) IFN-β or (0.1–5 ng/ml) gemcitabine during 3 or 7 days. Both drugs inhibited cell growth, measured as DNA amount per well, in a dose- and time dependent manner (Fig. 1a and S1).
After 7 days, IFN-β inhibited cell growth in BxPC-3 at significantly lower concentrations (EC50 62 IU/ml; 95% CI 7.4–66.4) than CFPAC-1 (302 IU/ml; 95% CI 285.6–319.5; p < 0.001) and Panc-1 (978 IU/ml; 95% CI 896.8–1067; p < 0.001). For gemcitabine, EC50 value after 7 days was lower for CFPAC-1 (0.19 ng/ml; 95% CI 0.17–0.2), compared to BxPC-3 (0.81 ng/ml; 95% CI 0.79–0.84; p < 0.001) and Panc-1 (1.0 ng/ml; 95% CI 0.98–1.02; p < 0.001).

Time-dependent upregulation of expression of interferon-stimulated genes by IFN-β

Gene expression analysis of three ISGs (IFIT1, OAS1A, and Mx1) was performed after cells were treated for 4, 12, 24 or 72 h with 100 IU/ml (BxPC-3 and CFPAC-1) or 1000 IU/ml (Panc-1) IFN-β. The average baseline expression of all three genes was higher in BxPC-3 and CFPAC-1, compared to Panc-1 (Fig. 1b, upper panel).
All genes were upregulated during IFN-β treatment in a time-dependent manner (Fig. 1b, lower panel). The strongest induction was observed in Mx1 after 72 h treatment. In BxPC-3 and CFPAC-1, Mx1 expression was already significantly upregulated after 4 h treatment with IFN-β. In Panc-1, a significant lower increase of all three genes was observed, even though cells were treated with 1000 IU/ml IFN-β.

IFN-β sensitizes pancreatic cancer cells to gemcitabine treatment

Next, we assessed the potential chemosensitising effect of IFN-β, as indicated by a fold decrease in EC50 value of gemcitabine. Cells were pre-treated for 4, 12, 24 or 72 h with 100 IU/ml (BxPC-3 and CFPAC-1) or 1000 IU/ml (Panc-1) IFN-β, followed by 3 days of increasing concentrations (0.1–5 ng/ml) gemcitabine monotherapy (Fig. 1c).
The overall response to gemcitabine was significantly stronger in IFN-β pre-treated cells compared to untreated control cells, even at time points where the effect of IFN-β on cell growth was minimal (Fig. S2).
This chemosensitising effect was most evident when cells were pre-treated for 72 h with IFN-β, as shown by a 4-, 7.7-, and 1.7-fold decrease in EC50 value in BxPC-3, CFPAC-1, and Panc-1 respectively (all P < 0.001 vs untreated control cells). Strikingly, in BxPC-3 and CFPAC-1, 4 h IFN-β pre-treatment resulted already at low concentrations gemcitabine (up to 0.1 ng/ml) in a significant higher cell growth inhibition (1.9- and 1.8-fold lower EC50 value respectively, both p < 0.001 vs untreated control cells) (Fig. 1d and Fig. S2).
An additional experiment was performed, in which cells were treated with simultaneously 100 IU/ml (BxPC-3 and CFPAC-1) or 1000 IU/ml (Panc-1) IFN-β plus increasing concentrations (0.5–5 ng/ml) gemcitabine for 3 days. In this experimental setting, the overall response of gemcitabine was not significantly stronger compared to cells without IFN-β treatment (Fig. S3).

IFN-β potentiates the anti-tumor effects of gemcitabine on colony formation

Effect on mean colony size and surviving fraction was assessed after 72 h IFN-β (100 IU/ml for BxPC-3 and CFPAC-1; 1000 IU/ml for Panc-1), after 72 h gemcitabine, and after 72 h IFN-β pre-treatment followed by 72 h gemcitabine monotherapy. Two concentrations gemcitabine were used, in which a minimal effect on cell growth was observed at monolayer culture (1 and 2.5 ng/ml for BxPC-3 and Panc-1; 0.1 and 1 ng/ml for CFPAC-1). PE (%) for BxPC-3, CFPAC-1, and Panc-1 were 19 ± 2.8, 78 ± 18, and 24 ± 7 respectively.
IFN-β reduced the mean colony size in BxPC-3 and lowered the survival fraction in CFPAC-1 (40 and 44% respectively, both p < 0.001). While no effect on survival fraction was observed, IFN-β pre-treatment enhanced the cytotoxic effect of gemcitabine in BxPC-3 (p < 0.001). Moreover, the cytostatic effect of gemcitabine was increased in CFPAC-1 and Panc-1 due to IFN-β pre-treatment (both p < 0.001) (Fig. 2).

IFN-β accumulates the proportion of cells in the S-phase

Cell cycle analysis was performed after 72 h IFN-β (100 IU/ml for BxPC-3 and CFPAC-1; 1000 IU/ml for Panc-1), after 72 h gemcitabine (1 ng/ml for CFPAC-1; 2.5 ng/ml for BxPC-3 and Panc-1), and after 72 h IFN-β pre-treatment followed by 72 h gemcitabine monotherapy.
IFN-β increased the percentage of cells in the S-phase in BxCP-3 (12%, p < 0.001) and CFPAC-1 (7%, p < 0.05) (Fig. 3). Additionally, an increase of S-phase population was observed after gemcitabine treatment in CFPAC-1 and Panc-1, which was associated with a decrease of cells in the G0/G1-phase (27 and 19% respectively, both p < 0.001). In BxPC-3, treatment with gemcitabine caused a minimal decrease of cells in the S-phase (− 4%, p < 0.01).
IFN-β pre-treatment followed by gemcitabine resulted in the strongest S-phase accumulation, with an increase of 33, 42, and 29% in BxPC-3, CFPAC-1, and Panc-1, respectively (all p < 0.001). This S-phase accumulation was associated with a strong decrease of cells in the G0/G1−phase (BxPC-3, 28%; CFPAC-1, 35%; Panc-1, 33%; all p < 0.001).

IFN-β upregulates expression of transporter genes involving the intracellular uptake of gemcitabine

As illustrated in Fig. 4a, several genes are involved in the metabolic pathway of gemcitabine. Gene expression analysis was performed of all genes after 72 h IFN-β (100 IU/ml for BxPC-3 and CFPAC-1; 1000 IU/ml for Panc-1). Baseline expression of SLC28A1 and SLC28A3 was low (< 0.0001) in BxPC-3 and CFPAC-1, and not detectable in Panc-1 (Fig. S3). IFN-β strongly increased SLC28A1 and SLC28A3 in BxPC-3 by 252 and 127%, respectively (both p < 0.001). In CFPAC-1, SLC28A3 was upregulated by 223% (p < 0.001). Remarkably, no increase in the expression of these genes was observed in Panc-1. Regarding the inactivating genes, IFN-β increased expression of CDA in BxPC-3 and Panc-1 (21 and 78% respectively, both p < 0.001). Expression of NT5E was increased in all cell lines (BxPC-3, 64% p < 0.001; CFPAC-1, 34% p < 0.001; Panc-1, 17% p < 0.01). No difference in expression of the activating genes was observed (Fig. 4b).

Ex vivo prevalidation of IFN-β mono- and combination therapy

In order to attempt to use a model that might predict treatment effects in vivo, an ex vivo precision cut tissue slice model was used. Four xenograft tumors of untreated mice were used to create the tissue slices. Slices were incubated for 72 h with 100 IU/ml IFN-β, 1 ng/ml gemcitabine, or the combination of IFN-β plus gemcitabine (Fig. 5a).
We next examined the expression of the cell proliferation marker Ki-67 and the apoptosis maker Caspase-3 in tumor tissue slices. The proportion of Ki-67 positive cells was significantly reduced after IFN-β, gemcitabine, and after the combination of IFN-β plus gemcitabine (decrease of 34, 43, and 50% respectively; all P < 0.01). Additionally, combination therapy significantly increased the number of apoptotic cells (increase of 85%, P = 0.02) (Fig. 5b and c).

In vivo validation of IFN-β mono- and combination therapy using a subcutaneous pancreatic cancer model

Finally, we examined the effects of IFN-β and gemcitabine, alone or in combination, on the growth of pancreatic tumors in nude mice. BxPC-3 cells were subcutaneously injected into male Balb/C mice. After 1 week, mice were randomized into one of four treatment arms: vehicle (NaCl), IFN-β (1.5 × 105 IU), gemcitabine (40 mg/kg) or both agents combined (Fig. 6a).
The time course of the growth of tumor volume is depicted in Fig. 6b. After 4 weeks of treatment, a significant reduction in tumor volume by 45% was found compared to untreated mice (P = 0.01). No significant weight loss was observed in any of the treatment groups, indicating that all the treatments were well tolerated (Table 1). However, in the gemcitabine arm, one mouse was found dead before start of the treatment and one mouse at the end of the total treatment cycle. Due to organ and tumor lysis, necropsy was not possible anymore.
Table 1
Treatment of subcutaneous heterotopic human pancreatic BxPC-3 tumors in nude mice
Treatment groups
Completion of treatment
Tumor volume (mm3)
Tumor weight (g)
Body weight (g)
Mean
Range
Mean
Range
Mean
Range
Control
5/8a
816
524–999
596
373–790
26
24–28
IFN-β
7/8a
660
281–1370
507
161–852
26
24–28
Gemcitabine
5/7a + b
745
402–1187
628
166–1121
25
24–27
IFN-β + gemcitabine
6/8
447c
213–572
492
161–958
26
24–28
a Number of mice that completed 4 weeks of treatment. Mice were sacrificed before the end of treatment if the wellbeing of the animal could not be maintained (in all these mice this was due to ulceration of the tumor)
b One mice died before the start of the treatment
c P < 0.05 versus control
As observed ex vivo in the tissue slice experiments, the combination of IFN-β plus gemcitabine significantly reduced the proportion of Ki-67 positive cells with 44%, while apoptosis was increased with 168% in tumor tissues compared with control group (both P < 0.05) (Fig. 6c and d).

Discussion

So far, gemcitabine has demonstrated disappointing results in patients with pancreatic cancer. More effective chemotherapeutics like FOLFIRINOX, where chemoresistance also remains a major clinical problem, is associated with frequent dose reductions, treatment-related serious adverse events, and often grade 3–4 infections. Since gemcitabine is much better tolerated with less toxicity compared to the newer chemotherapeutic agents, especially in the elderly pancreatic cancer patients, we aimed to evaluate whether the anti-tumor effect of gemcitabine could be enhanced by IFN-β and to demonstrate the mechanism of action.
First, we studied the effect of IFN-β alone and in combination with gemcitabine in three human pancreatic cancer cell lines: two IFN-β sensitive cell lines (BxPC-3 and CFPAC-1) and an IFN-β insensitive cell line (Panc-1) [16].
IFN-β strongly increased the inhibitory effects of gemcitabine in the IFN-β sensitive cell lines, which was confirmed in colony-forming assay. Both the cytotoxic and cytostatic effects of gemcitabine were significantly enhanced by IFN-β.
The increased chemosensitivity can be explained by two important observations. First, IFN-β increased cell population in the S-phase, suggesting that these cells were not able to transit into the G2/M-phase efficiently, and therefore exhibited a prolonged stay in the S-phase. As a result, DNA replication fails and cells can become more vulnerable for gemcitabine treatment. A downregulation and impaired activity of cyclins and cyclin dependent kinases was previously reported after IFN-β treatment, explaining the prolonged stay in the S-phase [23]. The active metabolites of gemcitabine, dFdCDP and dFdCTP, are also known to inhibit DNA synthesis by inhibiting ribonucleotide reductase and by DNA incorporation, respectively. Consequently, a complete inhibition of DNA synthesis is achieved, and apoptosis is induced [7]. Combination therapy with IFN-β and gemcitabine resulted in the strongest induction of cells in the S-phase in the IFN-β sensitive cell lines. In Panc-1, no difference was observed upon IFN-β treatment, which is in line with the absence of the chemosensitising effect in this cell line.
The second observation is the upregulation of gemcitabine transporters by IFN-β, resulting in potentially increased drug influx. A strong correlation was found between treatment resistance and the expression of gemcitabine transporters (hENT1, hCNT1, and hCNT3), activating enzyme (dCK), and inactivating enzyme (CDA) of gemcitabine [6, 7]. It should be emphasized, however, that we studied the effect of IFN-β on mRNA expression of gemcitabine transporters only. Further studies are necessary to demonstrate that IFN-β treatment results in increased intracellular gemcitabine levels and to confirm this mechanism of action by e.g. transporter knockdown. Nevertheless, this is the first study that evaluated the potential interaction between IFN-β and the expression of genes involved in gemcitabine transport and metabolism of gemcitabine. IFN-β strongly increased the expression of genes encoding for the hCNT1 and hCNT3 transporter in BxPC-3 and CFPAC-1. In contrast, IFN-β upregulated expression of CDA in Panc-1 cells.
The time of incubation with IFN-β appeared to be a significant parameter in our study. First, the (chemosensitising) anti-tumor effect was time-dependent. Notably, a 4 h pre-treatment with IFN-β already increased the response to gemcitabine in the IFN-β sensitive cell lines. In line with this, expression of ISGs was upregulated after 4 h and increased over time. Secondly, IFN-β did not enhance the gemcitabine response when both drugs were given simultaneously, suggesting that IFN-β needs time to sensitize tumor cells for chemotherapy.
Remarkably, there were significant differences between the IFN-β sensitive cell lines BxPC-3 and CFPAC-1 and the relative insensitive cell line Panc-1. First, the anti-tumor effects of IFN-β were less pronounced in Panc-1, even though these cells were treated with a 10-fold higher concentration (1000 IU/ml). Thereby, IFN-β pre-treatment mainly resulted in an additive anti-tumor effect in monolayer culture. Surprisingly, while IFN-β monotherapy had no effect on the colony size, it significantly enhanced the effect of gemcitabine on the colony size, suggesting a synergistic effect.
So far, there are no biomarkers for monitoring IFN activity and predicting clinical efficacy during IFN-β treatment. Booy et al. studied the correlation between the expression of the type I interferon receptor and the anti-tumor effect in a large panel of human pancreatic cancer cells. Despite the variable receptor expression among the cell lines, no significant correlation was reported regarding the maximal inhibitory effect of IFN-β [16]. Potentially, the downstream pathway of IFN can predict the response toward IFN-β treatment. In the current study, we measured the expression level of three ISGs (Mx1, IFIT1, and OAS1A), which are the functional end products of the IFN signalling pathway [24]. Therefore, their expression is induced as a result of an active IFN pathway. Consequently, expression levels of these ISGs can be assumed as a representation of activeness of the IFN pathway. At baseline, highest expression was observed in BxPC-3 and CFPAC-1, which is in line with the IFN-β sensitivity. IFN-β upregulated expression of these ISGs in all three cell lines. However, a much stronger upregulation was observed in BxPC-3 and CFPAC-1 compared to Panc-1, suggesting a less active IFN pathway in Panc-1, which might explain the lower response to IFN-β.
Expression of Mx1 was significant higher (approximately 13-fold) in the IFN-β sensitive cell lines compared to Panc-1, and strongly increased upon IFN-β treatment. The Mx1 gene encodes for the myxovirus resistant protein A (MxA) protein, which is an important antiviral factor against a wide spectrum of RNA viruses [25]. Apart from its role as a prominent antiviral protein in innate immunity, studies have indicated a role for Mx1 as a potential tumor suppressor gene. For example, deletion of Mx1 in prostate cancer is associated with a higher aggressive tendency and the expression of MxA is suppressed in a highly metastatic human prostate carcinoma cell line [26, 27]. Importantly, MxA is also employed to predict the efficacy of chemotherapy in several cancers. Knockout of Mx1 in prostate cancer cells resulted in a lower sensitivity to the chemotherapeutic agent docetaxel compared to MxA-positive cells [28]. Additionally, a study by Sistigu et al. reported a benefit of high MxA expression in patients with breast cancer receiving anthracycline-based chemotherapy [29]. Above findings indicate an important role for Mx1 in predicting the response to IFN-β treatment, as well as the potential chemosensitising effects.
So far, the effects of IFN-β, alone and combined with gemcitabine, have not been studied in animal models. By using a heterotopic subcutaneous pancreatic cancer mouse model, we are the first that confirmed the chemosensitising effect of IFN-β in vivo. Based on the response to IFN-β and the amount of IFN receptors expressed, we used the BxPC-3 cells for in vivo experiments [16].
First, we aimed to predict therapy response before start of treatment in an ex vivo tissue slice model. The advantage of this model is the ability to evaluate multiple treatments in one tumor sample. We were able to maintain viable slices up to 4 days of culture, which is in agreement with findings of two other studies [30, 31]. Promising results were observed as the combination of IFN-β plus gemcitabine significantly reduced the proportion of Ki-67 positive cells, while apoptosis was increased in tumor tissues compared with control group.
Regarding in vivo research, the most frequently used gemcitabine concentration varies between 100 mg/kg and 125 mg/kg [32, 33]. Nevertheless, based on the previously described in vitro findings, we decided to reduce the gemcitabine concentration and used a suboptimal concentration of 40 mg/kg.
As expected, given this suboptimal treatment dose, no significant decrease of tumor volume or weight was found in mice treated with gemcitabine alone. Additionally, despite the potent anti-tumor effects in vitro, no difference was found in mice treated with IFN-β alone, however, there was a clear trend towards a smaller tumor volume. Although IFN-β concentrations were not measured in this study, it may be possible that the circulating concentration of IFN-β was not sufficient. This may be related to the relatively short half-life of IFNs in the circulation [34]. In vitro, the concentration of IFN-β required to reduce cell growth to 50% in a large series of pancreatic cancer cell lines, ranged between 70 and 1000 IU/ml [16]. These concentrations are not easily reached (4–10 IU/ml after four doses of 18 MIU IFN-β at 48-h intervals in serum of human healthy volunteers after s.c. administration) [34]. Furthermore, the anti-tumor activities of type I IFNs can be limited by the activation of several survival pathways, such as the induction of the JAK2/STAT-3 pathway, the activation of nuclear factor kappa-beta (NF-κB) and the increased expression of the epidermal growth factor receptor (EGF-R). This could result in the stimulation of cell proliferation, malignant transformation and invasion, and the inhibition of apoptosis [35, 36].
After 30 days of treatment, we observed a significant synergistic effect of the combined therapy of IFN-β and gemcitabine, which was reflected by the reduction of tumor volume and, additionally, by a decreased proportion of proliferating tumor cells and increased apoptosis, confirming the results observed ex vivo.
Although heterotopic subcutaneous models are often used in cancer research, it is important to evaluate the effects of IFN-β and gemcitabine in an orthotopic model as well. Especially since type I IFNs are known to induce immunoregulatory activities and interact with the tumor microenvironment [37].
The therapeutic effectiveness of type I IFN treatment has been demonstrated in a considerable number of other malignancies, including hematologic tumors, as well as solid tumors. However, despite FDA approvals for recombinant IFN-α in a few cancers, including melanoma and renal cell carcinoma, recombinant IFN-α is not a conventional treatment for these malignancies. Long-term administration is needed to maintain therapeutic efficacy, resulting in high-grade toxicity and significant adverse side effects in patients [38].
On the other hand, IFN-β has emerged as a safer and more potent treatment compared to IFN-α. In addition to pancreatic cancer, IFN-β induced evident anti-tumor and chemosensitising effects pre-clinically in several other cancer types, e.g. hepatocellular carcinoma and breast cancer [39, 40]. Thereby, studies report chemosensitising effects with other chemotherapeutic agents, e.g. 5-FU and cisplatin, as well, suggesting that the sensitising effect of IFN-β is not only limited to gemcitabine [39, 41]. Interesting, the intracellular uptake of these drugs are also mediated by the nucleotide transporter proteins hENT1, hCNT1, and hCNT3 [42, 43].
So far, recombinant IFN-β has not yet been approved for the treatment of any cancer type and has yet to be clinically tested in pancreatic cancer. In addition, it is recommended to study the combination of IFN-β with the new developed chemotherapeutic agents such as FOLFIRINOX and Nab-Paclitaxel as well.
While IFN therapies have been around for a while, new insights in activation of the IFN pathway have resulted in novel IFN-directed cancer treatment strategies. One example is the use of IFN based conjugates, which increase the half-life time of IFN and potentially results into higher concentrations at the tumor site [44]. In addition, the PEGylated form of IFN resulted in a higher serum concentration, requiring lower and less frequent doses compared to the conventional IFNs [45]. The PEGylated form of IFN-α has already proven to be effective in the treatment of melanoma and metastatic renal cell carcinoma patients [46, 47]. Currently, the PEGylated form IFN-β is being tested in a phase III clinical trial (ADVANCE) in patients with multiple sclerosis [48, 49]. Another novel approach is the induction of type I IFN production via activation of the STING and RIG-I pathway [44]. These approaches are currently being tested in clinical trials or are in late pre-clinical development.
Although currently no improvement has been made with immunotherapy in pancreatic cancer, IFN-β might also play a crucial role in new strategies in combination with immunotherapy. Expression of Interferon-stimulated gene 15 (ISG15) is induced by IFN-β and its pathway is highly expressed in various malignancies, including pancreatic ductal adenocarcinoma. Interestingly Burke et al. demonstrated that ISG15 pathway knockdown not only reversed the KRAS-associated phenotypes of pancreatic ductal adenocarcinoma cells, such as increased proliferation and colony formation, but also decreased tumor programmed death ligand-1 (PDL-1) expression leading to increased number of CD8+ tumor-infiltrating lymphocytes [50].

Conclusions

In conclusion, to the best of our knowledge, this is the first study that determined the effects of IFN-β alone and in combination with gemcitabine on pancreatic cancer in three different experimental models. A synergistic anti-tumor effect of the combination treatment with IFN-β and gemcitabine was observed in vitro, in vivo, and ex vivo. These anti-tumor effects were already present at low concentrations of gemcitabine and involve cell cycle modulation and upregulation of gemcitabine transporters genes by IFN-β. In order to demonstrate the potent anti-tumor activities of combined gemcitabine/IFN-β therapy in the clinical setting, prospective studies are necessary.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12885-020-07420-0.

Acknowledgments

The authors thank the animal welfare takers for their help during the experiments.
All mouse experiments were controlled by the animal welfare committee (IvD) of the Erasmus Medical Center and approved by the national central committee of animal experiments (CCD) under the protocol number 105–12-52, in accordance with the Dutch Act on Animal Experimentation and EU Directive 2010/63/EU. This study was performed in accordance with the Declaration of Helsinki.
The maximum tumor size that was permitted by the ethic committee was 2000mm3. This size was not exceeded during the study.
Not applicable.

Competing interests

The authors declare that they have no competing interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
2.
Zurück zum Zitat Baulieux J, Delpero JR. [surgical treatment of pancreatic cancer: curative resections] Traitement chirurgical du cancer du pancreas: les exereses a visee curative. Ann Chir. 2000;125(7):609–17.PubMedCrossRef Baulieux J, Delpero JR. [surgical treatment of pancreatic cancer: curative resections] Traitement chirurgical du cancer du pancreas: les exereses a visee curative. Ann Chir. 2000;125(7):609–17.PubMedCrossRef
3.
Zurück zum Zitat Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. Jama. 2013;310(14):1473–81.CrossRefPubMed Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. Jama. 2013;310(14):1473–81.CrossRefPubMed
4.
Zurück zum Zitat Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic Cancer. N Engl J Med. 2018;379(25):2395–406.PubMedCrossRef Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic Cancer. N Engl J Med. 2018;379(25):2395–406.PubMedCrossRef
5.
Zurück zum Zitat Tehfe M, Dowden S, Kennecke H, El-Maraghi R, Lesperance B, Couture F, et al. Nab-paclitaxel plus gemcitabine versus gemcitabine in patients with metastatic pancreatic adenocarcinoma: Canadian subgroup analysis of the phase 3 MPACT trial. Adv Ther. 2016;33(5):747–59.PubMedPubMedCentralCrossRef Tehfe M, Dowden S, Kennecke H, El-Maraghi R, Lesperance B, Couture F, et al. Nab-paclitaxel plus gemcitabine versus gemcitabine in patients with metastatic pancreatic adenocarcinoma: Canadian subgroup analysis of the phase 3 MPACT trial. Adv Ther. 2016;33(5):747–59.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat de Sousa CL, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.CrossRef de Sousa CL, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.CrossRef
8.
Zurück zum Zitat Marechal R, Mackey JR, Lai R, Demetter P, Peeters M, Polus M, et al. Human equilibrative nucleoside transporter 1 and human concentrative nucleoside transporter 3 predict survival after adjuvant gemcitabine therapy in resected pancreatic adenocarcinoma. Clin Cancer Res. 2009;15(8):2913–9.PubMedCrossRef Marechal R, Mackey JR, Lai R, Demetter P, Peeters M, Polus M, et al. Human equilibrative nucleoside transporter 1 and human concentrative nucleoside transporter 3 predict survival after adjuvant gemcitabine therapy in resected pancreatic adenocarcinoma. Clin Cancer Res. 2009;15(8):2913–9.PubMedCrossRef
9.
Zurück zum Zitat Nakano Y, Tanno S, Koizumi K, Nishikawa T, Nakamura K, Minoguchi M, et al. Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells. Br J Cancer. 2007;96(3):457–63.PubMedPubMedCentralCrossRef Nakano Y, Tanno S, Koizumi K, Nishikawa T, Nakamura K, Minoguchi M, et al. Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells. Br J Cancer. 2007;96(3):457–63.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Ashida R, Nakata B, Shigekawa M, Mizuno N, Sawaki A, Hirakawa K, et al. Gemcitabine sensitivity-related mRNA expression in endoscopic ultrasound-guided fine-needle aspiration biopsy of unresectable pancreatic cancer. J Exp Clin Cancer Res. 2009;28:83.PubMedPubMedCentralCrossRef Ashida R, Nakata B, Shigekawa M, Mizuno N, Sawaki A, Hirakawa K, et al. Gemcitabine sensitivity-related mRNA expression in endoscopic ultrasound-guided fine-needle aspiration biopsy of unresectable pancreatic cancer. J Exp Clin Cancer Res. 2009;28:83.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Weizman N, Krelin Y, Shabtay-Orbach A, Amit M, Binenbaum Y, Wong RJ, et al. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene. 2014;33(29):3812–9.PubMedCrossRef Weizman N, Krelin Y, Shabtay-Orbach A, Amit M, Binenbaum Y, Wong RJ, et al. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene. 2014;33(29):3812–9.PubMedCrossRef
12.
Zurück zum Zitat Tomimaru Y, Eguchi H, Wada H, Tomokuni A, Kobayashi S, Marubashi S, et al. Synergistic antitumor effect of interferon-ss with gemcitabine in interferon-alpha-non-responsive pancreatic cancer cells. Int J Oncol. 2011;38(5):1237–43.PubMed Tomimaru Y, Eguchi H, Wada H, Tomokuni A, Kobayashi S, Marubashi S, et al. Synergistic antitumor effect of interferon-ss with gemcitabine in interferon-alpha-non-responsive pancreatic cancer cells. Int J Oncol. 2011;38(5):1237–43.PubMed
13.
Zurück zum Zitat Morak MJ, van Koetsveld PM, Kanaar R, Hofland LJ, van Eijck CH. Type I interferons as radiosensitisers for pancreatic cancer. Eur J Cancer. 2011;47(13):1938–45.PubMedCrossRef Morak MJ, van Koetsveld PM, Kanaar R, Hofland LJ, van Eijck CH. Type I interferons as radiosensitisers for pancreatic cancer. Eur J Cancer. 2011;47(13):1938–45.PubMedCrossRef
14.
Zurück zum Zitat Jost E, Roos WP, Kaina B, Schmidberger H. Response of pancreatic cancer cells treated with interferon-alpha or beta and co-exposed to ionising radiation. Int J Radiat Biol. 2010;86(9):732–41.PubMedCrossRef Jost E, Roos WP, Kaina B, Schmidberger H. Response of pancreatic cancer cells treated with interferon-alpha or beta and co-exposed to ionising radiation. Int J Radiat Biol. 2010;86(9):732–41.PubMedCrossRef
15.
Zurück zum Zitat De Maeyer E, De Maeyer-Guignard J. Type I interferons. Int Rev Immunol. 1998;17(1–4):53–73.PubMedCrossRef De Maeyer E, De Maeyer-Guignard J. Type I interferons. Int Rev Immunol. 1998;17(1–4):53–73.PubMedCrossRef
16.
Zurück zum Zitat Booy S, van Eijck CH, Dogan F, van Koetsveld PM, Hofland LJ. Influence of type-I interferon receptor expression level on the response to type-I Interferons in human pancreatic cancer cells. J Cell Mol Med. 2014;18(3):492–502.PubMedPubMedCentralCrossRef Booy S, van Eijck CH, Dogan F, van Koetsveld PM, Hofland LJ. Influence of type-I interferon receptor expression level on the response to type-I Interferons in human pancreatic cancer cells. J Cell Mol Med. 2014;18(3):492–502.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Vitale G, van Eijck CH, van Koetsveld Ing PM, Erdmann JI, Speel EJ, van der Wansem IK, et al. Type I interferons in the treatment of pancreatic cancer: mechanisms of action and role of related receptors. Ann Surg. 2007;246(2):259–68.PubMedPubMedCentralCrossRef Vitale G, van Eijck CH, van Koetsveld Ing PM, Erdmann JI, Speel EJ, van der Wansem IK, et al. Type I interferons in the treatment of pancreatic cancer: mechanisms of action and role of related receptors. Ann Surg. 2007;246(2):259–68.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat van Koetsveld PM, Vitale G, de Herder WW, Feelders RA, van der Wansem K, Waaijers M, et al. Potent inhibitory effects of type I interferons on human adrenocortical carcinoma cell growth. J Clin Endocrinol Metab. 2006;91(11):4537–43.PubMedCrossRef van Koetsveld PM, Vitale G, de Herder WW, Feelders RA, van der Wansem K, Waaijers M, et al. Potent inhibitory effects of type I interferons on human adrenocortical carcinoma cell growth. J Clin Endocrinol Metab. 2006;91(11):4537–43.PubMedCrossRef
19.
Zurück zum Zitat Herrera-Martinez AD, Feelders RA, de Herder WW, Castano JP, Galvez Moreno MA, Dogan F, et al. Effects of ketoconazole on ACTH-producing and non-ACTH-producing neuroendocrine tumor cells. Horm Cancer. 2019;10(2–3):107–19.PubMedCrossRef Herrera-Martinez AD, Feelders RA, de Herder WW, Castano JP, Galvez Moreno MA, Dogan F, et al. Effects of ketoconazole on ACTH-producing and non-ACTH-producing neuroendocrine tumor cells. Horm Cancer. 2019;10(2–3):107–19.PubMedCrossRef
20.
Zurück zum Zitat Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3(7):RESEARCH0034.PubMedPubMedCentralCrossRef Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3(7):RESEARCH0034.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Booy S, Hofland LJ, Waaijers AM, Croze E, van Koetsveld PM, de Vogel L, et al. Type I interferon receptor expression in human pancreatic and Periampullary Cancer tissue. Pancreas. 2015;44(1):99–105.PubMedCrossRef Booy S, Hofland LJ, Waaijers AM, Croze E, van Koetsveld PM, de Vogel L, et al. Type I interferon receptor expression in human pancreatic and Periampullary Cancer tissue. Pancreas. 2015;44(1):99–105.PubMedCrossRef
22.
Zurück zum Zitat Carpenter AE, Jones TR, Lamprecht MR, Clarke C, Kang IH, Friman O, et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 2006;7(10):R100.PubMedPubMedCentralCrossRef Carpenter AE, Jones TR, Lamprecht MR, Clarke C, Kang IH, Friman O, et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 2006;7(10):R100.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Sangfelt O, Erickson S, Castro J, Heiden T, Gustafsson A, Einhorn S, et al. Molecular mechanisms underlying interferon-alpha-induced G0/G1 arrest: CKI-mediated regulation of G1 Cdk-complexes and activation of pocket proteins. Oncogene. 1999;18(18):2798–810.PubMedCrossRef Sangfelt O, Erickson S, Castro J, Heiden T, Gustafsson A, Einhorn S, et al. Molecular mechanisms underlying interferon-alpha-induced G0/G1 arrest: CKI-mediated regulation of G1 Cdk-complexes and activation of pocket proteins. Oncogene. 1999;18(18):2798–810.PubMedCrossRef
24.
Zurück zum Zitat Samarajiwa SA, Forster S, Auchettl K, Hertzog PJ. INTERFEROME: the database of interferon regulated genes. Nucleic Acids Res. 2009;37(Database issue):D852–7.PubMedCrossRef Samarajiwa SA, Forster S, Auchettl K, Hertzog PJ. INTERFEROME: the database of interferon regulated genes. Nucleic Acids Res. 2009;37(Database issue):D852–7.PubMedCrossRef
25.
Zurück zum Zitat Haller O, Staeheli P, Schwemmle M, Kochs G. Mx GTPases: dynamin-like antiviral machines of innate immunity. Trends Microbiol. 2015;23(3):154–63.PubMedCrossRef Haller O, Staeheli P, Schwemmle M, Kochs G. Mx GTPases: dynamin-like antiviral machines of innate immunity. Trends Microbiol. 2015;23(3):154–63.PubMedCrossRef
26.
Zurück zum Zitat Demichelis F, Fall K, Perner S, Andren O, Schmidt F, Setlur SR, et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene. 2007;26(31):4596–9.PubMedCrossRef Demichelis F, Fall K, Perner S, Andren O, Schmidt F, Setlur SR, et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene. 2007;26(31):4596–9.PubMedCrossRef
27.
Zurück zum Zitat Mushinski JF, Nguyen P, Stevens LM, Khanna C, Lee S, Chung EJ, et al. Inhibition of tumor cell motility by the interferon-inducible GTPase MxA. J Biol Chem. 2009;284(22):15206–14.PubMedPubMedCentralCrossRef Mushinski JF, Nguyen P, Stevens LM, Khanna C, Lee S, Chung EJ, et al. Inhibition of tumor cell motility by the interferon-inducible GTPase MxA. J Biol Chem. 2009;284(22):15206–14.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Brown SG, Knowell AE, Hunt A, Patel D, Bhosle S, Chaudhary J. Interferon inducible antiviral MxA is inversely associated with prostate cancer and regulates cell cycle, invasion and Docetaxel induced apoptosis. Prostate. 2015;75(3):266–79.PubMedCrossRef Brown SG, Knowell AE, Hunt A, Patel D, Bhosle S, Chaudhary J. Interferon inducible antiviral MxA is inversely associated with prostate cancer and regulates cell cycle, invasion and Docetaxel induced apoptosis. Prostate. 2015;75(3):266–79.PubMedCrossRef
29.
Zurück zum Zitat Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20(11):1301–9.PubMedCrossRef Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20(11):1301–9.PubMedCrossRef
30.
Zurück zum Zitat van Geer MA, Kuhlmann KF, Bakker CT, ten Kate FJ, Oude Elferink RP, Bosma PJ. Ex-vivo evaluation of gene therapy vectors in human pancreatic (cancer) tissue slices. World J Gastroenterol. 2009;15(11):1359–66.PubMedPubMedCentralCrossRef van Geer MA, Kuhlmann KF, Bakker CT, ten Kate FJ, Oude Elferink RP, Bosma PJ. Ex-vivo evaluation of gene therapy vectors in human pancreatic (cancer) tissue slices. World J Gastroenterol. 2009;15(11):1359–66.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Rebours V, Albuquerque M, Sauvanet A, Ruszniewski P, Levy P, Paradis V, et al. Hypoxia pathways and cellular stress activate pancreatic stellate cells: development of an organotypic culture model of thick slices of normal human pancreas. PLoS One. 2013;8(9):e76229.PubMedPubMedCentralCrossRef Rebours V, Albuquerque M, Sauvanet A, Ruszniewski P, Levy P, Paradis V, et al. Hypoxia pathways and cellular stress activate pancreatic stellate cells: development of an organotypic culture model of thick slices of normal human pancreas. PLoS One. 2013;8(9):e76229.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Rajeshkumar NV, De Oliveira E, Ottenhof N, Watters J, Brooks D, Demuth T, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res. 2011;17(9):2799–806.PubMedPubMedCentralCrossRef Rajeshkumar NV, De Oliveira E, Ottenhof N, Watters J, Brooks D, Demuth T, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res. 2011;17(9):2799–806.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Awasthi N, Schwarz MA, Schwarz RE. Combination effects of bortezomib with gemcitabine and EMAP II in experimental pancreatic cancer. Cancer Biol Ther. 2010;10(1):99–107.PubMedCrossRef Awasthi N, Schwarz MA, Schwarz RE. Combination effects of bortezomib with gemcitabine and EMAP II in experimental pancreatic cancer. Cancer Biol Ther. 2010;10(1):99–107.PubMedCrossRef
34.
Zurück zum Zitat Buchwalder PA, Buclin T, Trinchard I, Munafo A, Biollaz J. Pharmacokinetics and pharmacodynamics of IFN-beta 1a in healthy volunteers. J Interf Cytokine Res. 2000;20(10):857–66.CrossRef Buchwalder PA, Buclin T, Trinchard I, Munafo A, Biollaz J. Pharmacokinetics and pharmacodynamics of IFN-beta 1a in healthy volunteers. J Interf Cytokine Res. 2000;20(10):857–66.CrossRef
35.
Zurück zum Zitat Dicitore A, Caraglia M, Gaudenzi G, Manfredi G, Amato B, Mari D, et al. Type I interferon-mediated pathway interacts with peroxisome proliferator activated receptor-gamma (PPAR-gamma): at the cross-road of pancreatic cancer cell proliferation. Biochim Biophys Acta. 2014;1845(1):42–52.PubMed Dicitore A, Caraglia M, Gaudenzi G, Manfredi G, Amato B, Mari D, et al. Type I interferon-mediated pathway interacts with peroxisome proliferator activated receptor-gamma (PPAR-gamma): at the cross-road of pancreatic cancer cell proliferation. Biochim Biophys Acta. 2014;1845(1):42–52.PubMed
36.
Zurück zum Zitat Vitale G, Zappavigna S, Marra M, Dicitore A, Meschini S, Condello M, et al. The PPAR-gamma agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-beta treated pancreatic cancer cells. Biotechnol Adv. 2012;30(1):169–84.PubMedCrossRef Vitale G, Zappavigna S, Marra M, Dicitore A, Meschini S, Condello M, et al. The PPAR-gamma agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-beta treated pancreatic cancer cells. Biotechnol Adv. 2012;30(1):169–84.PubMedCrossRef
37.
Zurück zum Zitat Parker BS, Rautela J, Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer. 2016;16(3):131–44.PubMedCrossRef Parker BS, Rautela J, Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer. 2016;16(3):131–44.PubMedCrossRef
38.
Zurück zum Zitat Quesada JR, Talpaz M, Rios A, Kurzrock R, Gutterman JU. Clinical toxicity of interferons in cancer patients: a review. J Clin Oncol. 1986;4(2):234–43.PubMedCrossRef Quesada JR, Talpaz M, Rios A, Kurzrock R, Gutterman JU. Clinical toxicity of interferons in cancer patients: a review. J Clin Oncol. 1986;4(2):234–43.PubMedCrossRef
39.
Zurück zum Zitat Ogasawara S, Yano H, Momosaki S, Akiba J, Nishida N, Kojiro S, et al. Growth inhibitory effects of IFN-beta on human liver cancer cells in vitro and in vivo. J Interf Cytokine Res. 2007;27(6):507–16.CrossRef Ogasawara S, Yano H, Momosaki S, Akiba J, Nishida N, Kojiro S, et al. Growth inhibitory effects of IFN-beta on human liver cancer cells in vitro and in vivo. J Interf Cytokine Res. 2007;27(6):507–16.CrossRef
40.
Zurück zum Zitat Doherty MR, Cheon H, Junk DJ, Vinayak S, Varadan V, Telli ML, et al. Interferon-beta represses cancer stem cell properties in triple-negative breast cancer. Proc Natl Acad Sci U S A. 2017;114(52):13792–7.PubMedPubMedCentralCrossRef Doherty MR, Cheon H, Junk DJ, Vinayak S, Varadan V, Telli ML, et al. Interferon-beta represses cancer stem cell properties in triple-negative breast cancer. Proc Natl Acad Sci U S A. 2017;114(52):13792–7.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Li Q, Kawamura K, Yang S, Okamoto S, Kobayashi H, Tada Y, et al. Interferon-β produces synergistic combinatory anti-tumor effects with cisplatin or pemetrexed on mesothelioma cells. PLoS One. 2013;8(8):e72709.PubMedPubMedCentralCrossRef Li Q, Kawamura K, Yang S, Okamoto S, Kobayashi H, Tada Y, et al. Interferon-β produces synergistic combinatory anti-tumor effects with cisplatin or pemetrexed on mesothelioma cells. PLoS One. 2013;8(8):e72709.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Pastor-Anglada M, Pérez-Torras S. Nucleoside transporter proteins as biomarkers of drug responsiveness and drug targets. Front Pharmacol. 2015;6:13.PubMedPubMedCentralCrossRef Pastor-Anglada M, Pérez-Torras S. Nucleoside transporter proteins as biomarkers of drug responsiveness and drug targets. Front Pharmacol. 2015;6:13.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Sprowl JA, Mikkelsen TS, Giovinazzo H, Sparreboom A. Contribution of tumoral and host solute carriers to clinical drug response. Drug Resist Updat. 2012;15(1–2):5–20.PubMedPubMedCentralCrossRef Sprowl JA, Mikkelsen TS, Giovinazzo H, Sparreboom A. Contribution of tumoral and host solute carriers to clinical drug response. Drug Resist Updat. 2012;15(1–2):5–20.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18(3):219–34.PubMedCrossRef Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18(3):219–34.PubMedCrossRef
45.
Zurück zum Zitat Pepinsky RB, LePage DJ, Gill A, Chakraborty A, Vaidyanathan S, Green M, et al. Improved pharmacokinetic properties of a polyethylene glycol-modified form of interferon-beta-1a with preserved in vitro bioactivity. J Pharmacol Exp Ther. 2001;297(3):1059–66.PubMed Pepinsky RB, LePage DJ, Gill A, Chakraborty A, Vaidyanathan S, Green M, et al. Improved pharmacokinetic properties of a polyethylene glycol-modified form of interferon-beta-1a with preserved in vitro bioactivity. J Pharmacol Exp Ther. 2001;297(3):1059–66.PubMed
46.
Zurück zum Zitat Eggermont AM, Suciu S, Testori A, Santinami M, Kruit WH, Marsden J, et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J Clin Oncol. 2012;30(31):3810–8.PubMedCrossRef Eggermont AM, Suciu S, Testori A, Santinami M, Kruit WH, Marsden J, et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J Clin Oncol. 2012;30(31):3810–8.PubMedCrossRef
47.
Zurück zum Zitat Motzer RJ, Rakhit A, Thompson J, Gurney H, Selby P, Figlin R, et al. Phase II trial of branched peginterferon-alpha 2a (40 kDa) for patients with advanced renal cell carcinoma. Ann Oncol. 2002;13(11):1799–805.PubMedCrossRef Motzer RJ, Rakhit A, Thompson J, Gurney H, Selby P, Figlin R, et al. Phase II trial of branched peginterferon-alpha 2a (40 kDa) for patients with advanced renal cell carcinoma. Ann Oncol. 2002;13(11):1799–805.PubMedCrossRef
48.
Zurück zum Zitat Mager DE, Neuteboom B, Jusko WJ. Pharmacokinetics and pharmacodynamics of PEGylated IFN-beta 1a following subcutaneous administration in monkeys. Pharm Res. 2005;22(1):58–61.PubMedCrossRef Mager DE, Neuteboom B, Jusko WJ. Pharmacokinetics and pharmacodynamics of PEGylated IFN-beta 1a following subcutaneous administration in monkeys. Pharm Res. 2005;22(1):58–61.PubMedCrossRef
49.
Zurück zum Zitat Hu X, Miller L, Richman S, Hitchman S, Glick G, Liu S, et al. A novel PEGylated interferon beta-1a for multiple sclerosis: safety, pharmacology, and biology. J Clin Pharmacol. 2012;52(6):798–808.PubMedCrossRef Hu X, Miller L, Richman S, Hitchman S, Glick G, Liu S, et al. A novel PEGylated interferon beta-1a for multiple sclerosis: safety, pharmacology, and biology. J Clin Pharmacol. 2012;52(6):798–808.PubMedCrossRef
50.
Zurück zum Zitat Burks J, Fleury A, Livingston S, Smith JP. ISG15 pathway knockdown reverses pancreatic cancer cell transformation and decreases murine pancreatic tumor growth via downregulation of PDL-1 expression. Cancer Immunol Immunother. 2019;68(12):2029–39.PubMedCrossRef Burks J, Fleury A, Livingston S, Smith JP. ISG15 pathway knockdown reverses pancreatic cancer cell transformation and decreases murine pancreatic tumor growth via downregulation of PDL-1 expression. Cancer Immunol Immunother. 2019;68(12):2029–39.PubMedCrossRef
Metadaten
Titel
Interferon-beta enhances sensitivity to gemcitabine in pancreatic cancer
verfasst von
Amber Blaauboer
Stephanie Booy
Peter M. van Koetsveld
Bas Karels
Fadime Dogan
Suzanne van Zwienen
Casper H. J. van Eijck
Leo J. Hofland
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2020
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07420-0

Weitere Artikel der Ausgabe 1/2020

BMC Cancer 1/2020 Zur Ausgabe

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.