Abstract
Accumulating evidence suggests a role for angiotensin-converting enzymes involving the angiotensin II-receptor 1 (AT1-R) and the cyclooxygenase pathway in carcinogenesis. The effects of ASS and enalapril were assessed in vitro and in a transgenic mouse model of pancreatic neuroendocrine neoplasms (pNENs). The effects of enalapril and ASS on proliferation and expression of the AGTR1A and its target gene vascular endothelial growth factor (Vegfa) were assessed in the neuroendocrine cell line BON1. Rip1-Tag2 mice were treated daily with either 0.6 mg/kg bodyweight of enalapril i.p., 20 mg/kg bodyweight of ASS i.p., or a vehicle in a prevention (weeks 5–12) and a survival group (week 5 till death). Tumor surface, weight of pancreatic glands, immunostaining for AT1-R and nuclear factor kappa beta (NFKB), and mice survival were analyzed. In addition, sections from human specimens of 20 insulinomas, ten gastrinomas, and 12 non-functional pNENs were evaluated for AT1-R and NFKB (NFKB1) expression and grouped according to the current WHO classification. Proliferation was significantly inhibited by enalapril and ASS in BON1 cells, with the combination being the most effective. Treatment with enalapril and ASS led to significant downregulation of known target genes Vegf and Rela at RNA level. Tumor growth was significantly inhibited by enalapril and ASS in the prevention group displayed by a reduction of tumor size (84%/67%) and number (30%/45%). Furthermore, daily treatment with enalapril and ASS prolonged the overall median survival compared with vehicle-treated Rip1-Tag2 (107 days) mice by 9 and 17 days (P=0.016 and P=0.013). The AT1-R and the inflammatory transcription factor NFKB were abolished completely upon enalapril and ASS treatment. AT1-R and NFKB expressions were observed in 80% of human pNENs. Enalapril and ASS may provide an approach for chemoprevention and treatment of pNENs.
Introduction
Pancreatic neuroendocrine neoplasms (pNENs) occur rarely with an annual incidence of 1/100 000 people, but their incidence is increasing steadily (Oberg & Eriksson 2005). pNENs present as either functional pNENs or non-functional pNENs (NF-pNENs). pNENs occur sporadically or can be associated with familial tumor syndromes such as multiple endocrine neoplasia type 1 (MEN1), von Hippel–Lindau (VHL) syndrome, or neurofibromatosis. The natural course of pNENs is highly variable. In general, a complete tumor resection improves survival in patients with pNENs. An aggressive surgical approach in patients with metastatic disease as part of a multimodal treatment often results in long-term survival (Metz & Jensen 2008). Survival is mainly determined by liver metastases, which can be targeted by several approaches such as surgery, chemoembolization, radiofrequency ablation, peptide radioreceptor therapy, or selective internal radiation therapy (Fendrich et al. 2006, Plockinger & Wiedenmann 2007). Biotherapy with somatostatin (SS) analogs is also frequently recommended for the treatment of metastatic disease achieving a tumor growth stabilization rather than inducing regression. Similar to SS analogs in midgut NENs, a tumoristatic effect may result in improved survival, but, at present, this has not been proven for pNENs (Plockinger & Wiedenmann 2007, Bajetta et al. 2009). Recently, the mTOR inhibitor everolimus and the multitargeted sunitinib have been shown to increase the overall survival in advanced neuroendocrine tumor patients in randomized controlled trials (Raymond et al. 2011, Yao et al. 2011).
The Rip1-Tag2 mouse model of pancreatic islet cell carcinoma offers a model of spontaneous multistep tumorigenesis. Transgenic mice express the oncogenic SV40 T antigen (Tag) under the control of the rat insulin gene promoter (RIP) and display different stages of tumor progression: onset of hyperproliferation, induction of angiogenesis, and formation of solid pNENs. Rip1-Tag2 mice succumb to insulinomas by the age of 14 weeks due to hypoglycemia. This multistage process suggests the sequential involvement of multiple genetic and epigenetic events in the progression from normal cells to tumors (Hanahan 1985, Folkman et al. 1989, Lopez & Hanahan 2002). As vascularization represents a key element in the progression of islet cell tumors, inhibition of tumor-induced angiogenesis offers a rational therapeutic approach. vascular endothelium-derived growth factor A (VEGFA) and platelet-derived growth factor are essential promoters of adult angiogenesis in a physiological and pathological setting and are upregulated in tumors of Rip1-Tag2 mice (Inoue et al. 2002, Bergers & Benjamin 2003, Ferrara 2004). Recently, the authors have reported new therapeutic approaches with cyclopamine and the Smo antagonist LDE225 for the treatment of pNENs using the same transgenic mouse model of islet cell tumors as presented in this study. In addition, our recent chemoprevention study in a transgenic mouse model of pancreatic cancer has revealed a delay of pancreatic intraepithelial neoplasia progression and cancer formation upon treatment with enalapril and aspirin (Fendrich et al. 2010, 2011a,b). Increasing evidence suggests a role for angiotensin-converting enzymes (ACEs) in carcinogenesis in an angiotensin II–AT1-R axis-dependent manner, involving VEGF (Asano et al. 2004, De Paepe 2009, Miyajima et al. 2009).
Population-based studies have shown that ACE inhibitor usage can be associated with reduced cancer risks, although recent studies have provided conflicting data (Lever et al. 1998, Garcia Rodriguez & Gonzalez-Perez 2004, Gonzalez-Perez et al. 2004, Teo 2011). An improved outcome can be observed in certain tumors such as pancreatic (Nakai et al. 2010) and non-small lung cancer (Wilop et al. 2009) after ACE inhibition combined with chemotherapy. Similar protective effects have been reported for the cyclooxygenase (COX) enzyme inhibitor aspirin (ASS) (Kune et al. 1988, Thun et al. 1991). As in many other types of malignant neoplasms, COX2 (PTGS2) and nuclear factor kappa beta (NFKB (NFKB1)) are overexpressed in pancreatic carcinoma and in a subset of precursor lesions (Maitra et al. 2002, Sclabas et al. 2005). Consequently, COX2 and NFKB might represent potential target for chemoprevention with selective COX2 inhibitors (Kopp & Ghosh 1994, Amaya et al. 2004). A chemopreventive strategy for pNENs is completely unexplored so far.
In this study, we evaluated antitumorigenic properties of enalapril and ASS in vitro using the human neuroendocrine cell line BON1. The effects of these drugs on tumor growth and survival were also tested in a chemopreventive setting in a transgenic mouse model of islet cell tumors. Two major targets, which are known to mediate the antitumorigenic effects of enalapril and ASS, were assessed in a set of human insulinomas, gastrinomas, and NF-pNENs.
Materials and methods
Cell culture
BON1 pancreatic carcinoid cells were grown in RPMI (Biochrom, Invitrogen) supplemented with 10% FCS (Biochrom, Berlin, Germany) and 1% penicillin/streptomycin at 37 °C in a humidified atmosphere containing 5% CO2.. Before treating, the cells were tested for mycoplasma infection using the QIAamp Viral RNA Mini Kit (Qiagen) for PCR.
Proliferation assays
For proliferation assay, BON1 cells were seeded into 96-well plates at a density of 5000 cells/well and 200 μl medium/well and were incubated overnight and, for RT-PCR, cells were seeded into 12-well-plates at a density of 80 000 cells/well and 2 ml medium/well. The following day, the cells were starved for another 24 h in a low-serum medium (RPMI with 0.5% FCS and 1% penicillin/streptomycin). Then, cells were treated with 100 μl low-serum medium and 100 μl vehicle for 24, 48, and 72 h. Vehicle concentration and combinations are listed in Supplementary Table 1, see section on supplementary data given at the end of this article. Untreated cells served as controls. Proliferation assay was carried out after 24, 48, and 72 h. To determine viable cell mass, 100 μM of 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide reagent (Thiazolyl Blue TetrazoliumBromid from Sigma) were added per well. Afterwards, cells in plates were incubated at 37 °C for 1 h. Optical density was read at an absorbance of 570/630 nm on a Wallac 1420 plate reader. All experiments were carried out in triplicate. For RT-PCR cells were treated until 48 h. Then, cells were lysed in the culture with 350 μl TRK buffer and whole RNA extraction was performed using RNA Kit PegLab. After RNA extraction, probes were transferred into cDNA with oligo-dT primer from Omniscript from Qiagen.
RT-PCR
All PCRs were carried out on a 7500 Real-Time PCR System (Applied Biosystems) over 40 cycles, with denaturation for 15 s at 95 °C and combined annealing/extension at 60 °C for 1 min, using the SYBR Green Master Mix from Applied Biosystems. Primer sequences for Vegf (Vegfa) were as follows: forward, 5′-CTC ACT TCC AGA AAC ACG ACA AA-3′ and reverse, 5′-CTG GTT GGA ACC GGC ATC T-3′. For RELA, the most abundant from of NFKB, the primer sequence was as follows: forward, 5′-CCC GAC TTG TTT GGG TGA TC-3′ and reverse, 5′-GCT CCG TCT CCA GGA GGT TAA-3′.
Mice
The generation of Rip1-Tag2 mice as a model of pancreatic islet cell carcinogenesis has been reported previously (Fendrich et al. 2011a). Mice used in this study were males and females of the Rip1-Tag2 transgenic mouse lineage bred in a C57Bl/6J background. All experiments were approved by the local committees for animal care and use. Animals were maintained in a climate-controlled environment at 22 °C, exposed to a 12 h light:12 h darkness cycle, fed on a standard laboratory chow diet and provided water ad libitum. For genotyping, genomic DNA was extracted from tail cuttings using the REDExtract-N-Amp Tissue PCR (PCR) Kit (Sigma–Aldrich). PCR was carried out for each animal, to test for the presence of Tag2. Primer sequences used were: 5′-GGA CAA ACC ACA ACT AGA ATG CAG-3′ and 5′-CAG AGC AGA ATT GTG GAG TGG-3′.
Treatment
The grouping and therapeutical algorithms are explained in Fig. 1. In the prevention group, animals were treated from 5 to 12 weeks of age and, in the survival group, from the fifth week of age until death defined by specific abort criteria in agreement with the standards for animal care. Rip1-Tag2 transgenic mice were randomly assigned to receive either i) aspirin (prevention OR survival group), ii) enalapril, or iii) vehicle. Aspirin and enalapril were dissolved in a sterile sodium chloride solution and administered to mice by an i.p. injection. Aspirin was administered once a day at a dose of 20 mg/kg bodyweight and enalapril was administered once a day at a dose of 0.6 mg/kg bodyweight, both intraperitoneally on a daily basis (Sakamoto et al. 2001). The dose of ASS is equivalent to a daily human dosage of 80–110 mg (Mahmoud et al. 1998). The equivalent human dosage of enalapril is unknown.
Necropsy
After completion of drug treatment, mice from the prevention and survival groups were killed and the pancreas was removed, weighed, and inspected for grossly visible tumors and preserved in 10% formalin solution (Sigma–Aldrich) for histology. For evaluating a change in islet cell tumor growth, four sections from each mouse were analyzed. These sections were taken at a distance of 1, 10, 20, and 40 μm from the surface of the pancreas and stained with H&E. All the islets were marked and their surface calculated computerized.
Immunostaining
Formalin-fixed and paraffin-embedded archived tumor samples, the corresponding normal tissue, and murine pancreatic glands were stained as described previously (Fendrich et al. 2008). Primary and secondary antibodies including positive and negative controls are given in Supplementary Table 1 and Supplementary Fig. 1. Briefly, slides were heated to 60 °C for 1 h, deparaffinized using xylene, and hydrated by a graded series of ethanol washes. Antigen retrieval was accomplished by microwave heating in 10 mM sodium citrate buffer, pH 6.0, for 10 min. For immunohistochemistry, endogenous peroxidase activity was quenched by 10 min incubation in 3% H2O2. Nonspecific binding was blocked with 10% serum. Sections were then probed with primary antibodies overnight at 4 °C. For immunohistochemistry, bound antibodies were detected using the avidin–biotin complex (ABC) peroxidase method (ABC Elite Kit, Vector Labs, Burlingame, CA, USA). Final staining was developed using the Sigma FAST DAB peroxidase substrate kit (Sigma). The immunohistochemistry results were scored as described previously (Fendrich et al. 2007). The AT1 and NFKB staining was categorized by the fraction of positive cells (weak, <10%; medium, >10 and <50%; and high, >50%). Lymphocytes and endothelial cells as shown in Supplementary Fig. 1 served as an internal positive control.
Patient samples
The human samples were obtained from patients with pNENs, who were treated at the authors' department. Serial sections of 20 insulinomas, 12 gastrinomas, and 12 NF-pNENs were confirmed by an experienced pathologist (A Ramaswamy) and subjected for further analyses. Clinical characteristics and WHO classification (Rindi 2010) are shown in Table 1. All patients gave their informed consent. The study followed the guidelines of the Local Ethics Committee.
Clinical characteristics and immunostaining results for AT1-R and NFKB in 44 patients with functioning and non-functioning pNENs
ID | Tumor | Age | WHO | M (yes/no) | IHC NFκB (positive/negative) | IHC AT1 (positive/negative) | Enalapril (yes/no) | Aspirin (yes/no/ND) |
---|---|---|---|---|---|---|---|---|
1 | Insulinoma (PT) | 71 | NET G1 | No | Positive | Positive | ND | ND |
2 | Insulinoma (PH) | 44 | NET G1 | No | ND | Positive | ND | ND |
3 | Insulinoma (PB) | 39 | NET G1 | No | ND | Positive | ND | ND |
4 | Insulinoma (PH) | 79 | NET G1 | No | ND | Positive | No | No |
5 | Insulinoma (PT) | 38 | NET G1 | No | Positive | Positive | No | No |
6 | Insulinoma (PH) | 55 | NET G1 | No | Negative | Positive | ND | ND |
7 | Insulinoma (PT) | 72 | NET G1 | No | Positive | Positive | ND | ND |
8 | Insulinoma (PH) | 58 | NET G1 | No | ND | Positive | No | No |
9 | Insulinoma (PB) | 30 | NET G1 | No | ND | Positive | No | No |
10 | Insulinoma (PH) | 80 | NET G1 | No | ND | Positive | ND | ND |
11 | Insulinoma (PB) | 42 | NET G1 | No | Positive | Positive | No | No |
12 | Insulinoma (PH) | 63 | NET G1 | No | Positive | Positive | No | No |
13 | Insulinoma (PB) | 64 | NET G1 | No | ND | Positive | ND | ND |
14 | Insulinoma (PH) | 31 | NET G2 | No | ND | Positive | No | No |
15 | Insulinoma (PH) | 41 | NET G1 | No | ND | Positive | No | No |
16 | Insulinoma (PT) | 33 | NET G1 | No | Negative | Negative | No | No |
17 | Insulinoma (PT) | 49 | NET G1 | No | ND | Negative | ND | ND |
18 | Insulinoma (PT) | 44 | NET G1 | No | Negative | Negative | ND | ND |
19 | Insulinoma (PB) | 67 | NET G1 | No | ND | Negative | ND | ND |
20 | Insulinoma (PH–PB) | 50 | NET G1 | No | ND | Negative | ND | ND |
21 | mGastrinoma | 36 | NET G1 | No | ND | Positive | No | No |
22 | mGastrinoma (ACTH Prod.) (LM UP) | 49 | ND | Yes (L) | ND | Negative | No | No |
23 | mGastrinoma (LM) | 47 | NET G1 | Yes (LM) | ND | Negative | ND | ND |
24 | mDG (DW pars desc.) | 70 | ND | No | ND | Positive | No | No |
25 | mDG (DW pars desc.) | 52 | ND | No | ND | Negative | No | No |
26 | mDG (DW pars sup.) | 49 | ND | No | ND | Positive | No | No |
27 | mDG (DW pars desc.) | 29 | NET G1 | No | ND | Positive | No | No |
28 | MEN mDG+mPG | 49 | NET G1 | Yes (LN) | ND | Negative | Yes (Captopril) | No |
29 | MEN mDG (DW) | 49 | NET G2 | No | Positive | Positive | No | No |
30 | mPG (PH–PB) | 32 | ND | No | Positive | Positive | No | No |
31 | MEN mDG+mPG (PH+PT) | 48 | ND | Yes (L) | Positive | Positive | No | No |
32 | NFPT (PH and PB) | 34 | NET G1 | No | Positive | Positive | ND | ND |
33 | NFPT (PH and PB) | 31 | ND | No | ND | Positive | No | No |
34 | NFPT (PH and PB) | 32 | ND | No | ND | Positive | No | No |
35 | NFPT (PH and PB) | 68 | ND | Yes (LN) | ND | Positive | Yes (Delix) | No |
36 | NFPT (PT) | 32 | NET G2 | No | ND | Positive | No | No |
37 | NFPT (PH) | 51 | ND | No | ND | Positive | No | No |
38 | NFPT (PH and PT) | 33 | NET G1 | No | ND | Positive | No | No |
39 | NFPT (PT) | 43 | NET G1 | No | Positive | Positive | No | No |
40 | NFPT (PT) | 37 | NET G1 | No | Positive | Positive | No | No |
41 | NFPT | 20 | NET G1 | No | Positive | Positive | No | No |
42 | NFPT (PB+PT) | 30 | NET G1 | No | ND | Positive | Yes (Lisinopril) | No |
43 | NFPT (LNM) | 63 | NET G2 | Yes (LN) | ND | Negative | ND | ND |
M, metastases; IHC, immunohistochemistry; AT1-R, angiotensin II-receptor 1; NFKB, nuclear factor kappa beta; NF-pNEN, non-functional pancreatic endocrine neoplasm, PH, pancreatic head; PB, pancreatic body; PT, pancreatic tail; LNM, lymph node metastasis; LM, liver metastases; LN, lymph node, ND, no data.
Statistical analysis
Survival curves were calculated using the Kaplan–Meier method. Log-rank test was applied to identify significant differences. Non-categorical parameters were analyzed by the Mann–Whitney U-test. Comparisons of more than two groups were made by one-way ANOVA with post hoc Holm–Sidak analysis for pairwise comparisons and comparisons vs control and by Kruskal–Wallis one-way ANOVA. P values <0.05 were considered statistically significant. Data were analyzed using the SPSS Software (version 14; SPSS, Inc.).
Results
Tumor growth is inhibited by enalapril and ASS in vitro
The proliferation of BON1 cells under the defined medium conditions was assessed by a standard MTT assay at 24, 48, and 72 after treatment with different doses of enalapril and ASS either alone or in combination and compared with different control groups (see also Fig. 2A, B and C). Enalapril as a single agent decreased proliferation in BON1 cells by 9–21%, which was significant after 48 and 72 h compared with non-treated and benzyl-treated cells as controls. ASS reduced the proliferation by 43, 59, and 68 h after 24, 48, and 72 h more significantly as a single agent and was only less effective than the combination with enalapril (see also Fig. 2A, B and C). RT-PCR revealed significant downregulation of VEGF and RELA upon enalapril and ASS treatment, which are known mediators of antitumorigenic properties of enalapril and ASS (see Fig. 3) in cultured BON1 cells. The combination of enalapril and ASS showed no additional effect (data not shown).
Chemoprevention with enalapril and ASS significantly reduces tumor growth
We observed development of islet cell hyperplasia, angiogenic islets, and islet cell tumors in the Rip1-Tag2 mice cohort used for our study compared with WT control mice, as described previously. The median tumor size decreased significantly upon enalapril and ASS treatment in Rip1-Tag2 mice (prevention-group) by 84% for enalapril (2 572 226 vs 15 935 703 μm2, P=0.002) and 67% for ASS (5 276 137 vs 15 935 703 μm2, P=0.001). Accordingly, the treatment resulted in a reduction of the median tumor number by 30% (9 vs 6) for enalapril and 45% for ASS (9 vs 5). Upon treatment of enalapril and ASS, the pancreas/tumor weight was analogously reduced (0.12 and 0.10 vs 0.15 g; P=0.028 and P=0.015; see also Fig. 4). In vivo enalapril and ASS treatment led to a lower Ki-67 index (1 and 17%, P<0.0001 and P<0.0001) compared with mock-treated control Rip1-Tag2 mice (29%) in the prevention group, which confirmed the in vitro results in BON1 cells.
Chemoprevention with enalapril and ASS led to loss of angiotensin II-receptor 1 and NFKB expression
Enalapril-, ASS-, and vehicle-treated Rip1-Tag2 mice (n=3 each) were stained for AT1-R and NFKB. Angiotensin II-receptor 1 (AT1-R) expression could be observed in tumor cells of all vehicle-treated control mice, whereas all enalapril- and ASS-treated mice had no AT1-R-positive tumor cells. Accordingly, NFKB expression could be observed only in tumor cells of vehicle-treated control mice, whereas enalapril- and ASS-treated mice had no NFKB-positive tumor cells (see also Fig. 5).
Chemoprevention with enalapril and ASS prolongs survival
Analyzed mice were treated with either enalapril, ASS, or vehicle from week 5 until death. Based on survival data of each group, a Kaplan–Meier survival plot was calculated (see Fig. 6). Enalapril treatment extended survival compared with survival of vehicle-treated mice by 10 days (108 vs 118 days; P=0.022) and ASS treatment prolonged the median survival by 16 days (108 vs 124 days; P=0.016) (see also Fig. 6A and B). The enalapril- and ASS-treated mice revealed a significantly lower weight of the pancreas/tumors at autopsy than the vehicle-treated control (0.15 and 0.16 vs 0.18 g; P=0.043 and P=0.039), reflecting the tumor growth inhibition.
Major reason of Rip1-Tag2 mice is hypoglycemia, which first impairs the behavior and finally leads to coma and death.
AGTR1 and NFKB expression in human sporadic and familial pNETs
To compare the AGTR1 and NFKB expression observed in the murine Rip1-Tag2 model with the situation in human sporadic and familial pNEN sections of insulinomas (n=20) and gastrinomas (n=11), and NF-pNENs (n=12) were stained for AT1-R and NFKB expression. At IHC, 15 out of 20 insulinomas, six out of 11 gastrinomas, and all NF-pNENs revealed AT1-R expression. Accordingly, five out of eight insulinomas, three out of three gastrinomas, and four out of four NF-pNENs expressed NFKB (see also Fig. 7). Negative sections at IHC for AT1-R were found in one out of three patients who were on enalapril. Clinical characteristics and classification according to the current WHO classification of analyzed patients are summarized in Table 1.
Discussion
The mortality of pNENs ranges between 5 and 70% with a 5-year survival of 30–95% and depends on clinical features and the genetic background (Jensen et al. 2012). Chemopreventive strategies may be beneficial for high-risk individuals with a hereditary predisposition for pNENs such as MEN1, VHL, or neurofibromatosis type 2. Mortality in patients with MEN1 is mainly caused by pNENs and malignant NEN of the thymus (Bartsch et al. 2005, Fendrich et al. 2006, Jensen et al. 2006, Goudet et al. 2009, 2010, Waldmann et al. 2009). Therefore, potent chemoprevention strategies may help to reduce the overall pNEN-associated mortality, but no chemopreventive strategies are known for pNENs so far.
In this study, we show for the first time that enalapril and ASS inhibit proliferation of human BON1 cells as an in vitro model for pNENs. Downregulation of VEGF and RELA, the most abundant form of NFKB, upon enalapril and ASS treatment may be held accountable for the growth inhibition and presents a known mechanism in other non-neuroendocrine tumor cell lines. NFKB orchestrates the expression of genes that encode key regulators of tumorigenesis, inflammation, and apoptosis and thus promotes tumor progression. Constitutive activation of NFKB is frequently observed in medullary thyroid cancer and NFKB protects pancreatic islet cells from tumor necrosis factor alpha-mediated cell death (Ludwig et al. 2001, Chang et al. 2003). Furthermore, activation of NFKB is a frequent event shared by differentiated and anaplastic thyroid cancer and first studies with NFKB inhibitors achieved promising results (Pacifico & Leonardi 2010). ACE inhibitors are known to reduce the expression of NFKB (Hernandez-Presa et al. 1998), while aspirin inhibits NFKB induction by decreasing the IKK2 activity through binding to IKK2 and thereby limits ATP binding. Tyrosine kinase and ERK1/2–MAP-kinase activation of the AT1-R induces VEGF expression, which mediates NFKB overexpression through the Akt pathway (Asano et al. 2004, Escobar et al. 2004, Hennessy et al. 2005). A similar mechanism was also described for the pancreatic adenocarcinoma (Arafat et al. 2007, Fendrich et al. 2010). Along with the literature, we could demonstrate in this study that enalapril and ASS significantly had an antiproliferative effect and reduced the target genes Vegf and Rela in vitro.
This observation was then confirmed by tumor growth inhibition in vivo with a decreased Ki-67 index after enalapril and ASS treatment in a transgenic mouse model of pNENs. Tumor growth inhibition was accompanied by a significantly prolonged survival. Several groups have reported that blocking the AT1-R by angiotensin receptor blockers (ARBs) can lead to reduced tumor growth and metastasis in experimental models of solid tumors (Rivera et al. 2001, Miyajima et al. 2002, Uemura et al. 2006, Rodrigues-Ferreira et al. 2012). Several animal cancer models after pharmacological ACE inhibition or AT1-R depletion revealed antitumorigenic properties (Volpert et al. 1996, Kosaka et al. 2007, Vinson et al. 2012). These observations are further reinforced by genetic models with AT1-R depletion, revealing deeper insights into the mechanisms of antiangiogenesis and growth inhibition mediated by the AT1-R (Egami et al. 2003). Neo et al. (2010) demonstrated that during the development of colorectal liver metastases, AT1-R expression increases and could be reduced by captopril, which led to a reduction in the tumor volume. Most effects of the inhibition of AT1R-driven angiogenesis are mediated through VEGF. The loss of AT1-R expression was also observed in enalapril- and ASS-treated animals compared with the vehicle control in this study. Parts of the antitumorigenic effect may be mediated by tumor-associated macrophages. A subpopulation of highly invasive breast cancer revealed a dramatic overexpression of AT1-R and raised the question of a therapeutical approach against breast cancer (Rhodes et al. 2009). The downregulation of AT1-R in islet cell tumors upon ACE inhibition may be mediated by the decreased angiotensin II levels, which are a consequence of the treatment with enalapril. This would be in the line with an induction of ATR1 in adrenal glands upon ACE inhibition, which was reported by Harrison-Bernard et al. (1999) in rats. Both the tissue-specific regulation of AT2-receptor isoforms (Sechi et al. 1996) and the complexity of multiple regulators challenge the theoretical explanation of the ablation of Atr1 in this study, which remains highly speculative.
These promising preclinical results have been challenged by a meta-analysis of randomized controlled trials, which showed a modestly higher individual risk for patients under ARBs (Sipahi et al. 2010). However, a meta-analysis of over 300 000 patients showed no increased risk for cancer, except in patients taking inhibitors of the ACEs in combination with ARBs (Bangalore et al. 2011).
Human pNENs showed robust expression of the ARB target ATR1 in the majority of analyzed pNENs in this study. This potentially points toward the AT1-R as a therapeutical target for AT1-R and ACE inhibitors for pNENs, especially in patients with an increased individual risk in a prophylactic setting. The loss of NFKB expression upon enalapril and ASS treatment in Rip1-Tag2 mice may be considered an additional potential effect.
This study found a significant antiproliferative effect of enalapril and ASS on islet cell tumors in vitro in a transgenic mouse model and revealed the target gene AT1-R to be expressed in the majority of human pNENs. Therefore, enalapril and/or ASS might be a valuable chemopreventive agent in patients with a high risk for pNENs that may attenuate the tumorigenic process rather than preventing it.
Supplementary data
This is linked to the online version of the paper at http://dx.doi.org/10.1530/ERC-14-0175.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Funding
This work was supported by the Theranostic Research Foundation Bad Berka.
References
Amaya K, Ohta T, Kitagawa H, Kayahara M, Takamura H, Fujimura T, Nishimura G, Shimizu K & Miwa K 2004 Angiotensin II activates MAP kinase and NF-κB through angiotensin II type I receptor in human pancreatic cancer cells. International Journal of Oncology 25 849–856. (doi:10.3892/ijo.25.4.849).
Arafat HA, Gong Q, Chipitsyna G, Rizvi A, Saa CT & Yeo CJ 2007 Antihypertensives as novel antineoplastics: angiotensin-I-converting enzyme inhibitors and angiotensin II type 1 receptor blockers in pancreatic ductal adenocarcinoma. Journal of the American College of Surgeons 204 996–1005.(discussion 1005–1006) (doi:10.1016/j.jamcollsurg.2007.01.067).
Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL & Reddy SA 2004 The PI3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-κB and c-Myc in pancreatic cancer cells. Oncogene 23 8571–8580. (doi:10.1038/sj.onc.1207902).
Bajetta E, Procopio G, Guadalupi V & Del Vecchio M 2009 Neuroendocrine tumors (NETs). Clinical Therapeutics 31 2060–2061. (doi:10.1016/j.clinthera.2009.09.007).
Bangalore S, Kumar S, Kjeldsen SE, Makani H, Grossman E, Wetterslev J, Gupta AK, Sever PS, Gluud C & Messerli FH 2011 Antihypertensive drugs and risk of cancer: network meta-analyses and trial sequential analyses of 324,168 participants from randomised trials. Lancet Oncology 12 65–82. (doi:10.1016/S1470-2045(10)70260-6).
Bartsch DK, Fendrich V, Langer P, Celik I, Kann PH & Rothmund M 2005 Outcome of duodenopancreatic resections in patients with multiple endocrine neoplasia type 1. Annals of Surgery 242 757–764.(discussion 764–766) (doi:10.1097/01.sla.0000189549.51913.d8).
Bergers G & Benjamin LE 2003 Tumorigenesis and the angiogenic switch. Nature Reviews. Cancer 3 401–410. (doi:10.1038/nrc1093).
Chang I, Kim S, Kim JY, Cho N, Kim YH, Kim HS, Lee MK, Kim KW & Lee MS 2003 Nuclear factor kappaB protects pancreatic β-cells from tumor necrosis factor-alpha-mediated apoptosis. Diabetes 52 1169–1175. (doi:10.2337/diabetes.52.5.1169).
De Paepe B 2009 Anti-angiogenic agents and cancer: current insights and future perspectives. Recent Patents on Anti-Cancer Drug Discovery 4 180–185. (doi:10.2174/157489209788452821).
Egami K, Murohara T, Shimada T, Sasaki K, Shintani S, Sugaya T, Ishii M, Akagi T, Ikeda H & Matsuishi T et al. 2003 Role of host angiotensin II type 1 receptor in tumor angiogenesis and growth. Journal of Clinical Investigation 112 67–75. (doi:10.1172/JCI16645).
Escobar E, Rodriguez-Reyna TS, Arrieta O & Sotelo J 2004 Angiotensin II, cell proliferation and angiogenesis regulator: biologic and therapeutic implications in cancer. Current Vascular Pharmacology 2 385–399. (doi:10.2174/1570161043385556).
Fendrich V, Langer P, Celik I, Bartsch DK, Zielke A, Ramaswamy A & Rothmund M 2006 An aggressive surgical approach leads to long-term survival in patients with pancreatic endocrine tumors. Annals of Surgery 244 845–851.(discussion 852–853) (doi:10.1097/01.sla.0000246951.21252.60).
Fendrich V, Waldmann J, Esni F, Ramaswamy A, Mullendore M, Buchholz M, Maitra A & Feldmann G 2007 Snail and Sonic Hedgehog activation in neuroendocrine tumors of the ileum. Endocrine-Related Cancer 14 865–874. (doi:10.1677/ERC-07-0108).
Fendrich V, Esni F, Garay MV, Feldmann G, Habbe N, Jensen JN, Dor Y, Stoffers D, Jensen J & Leach SD et al. 2008 Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology 135 621–631. (doi:10.1053/j.gastro.2008.04.011).
Fendrich V, Chen NM, Neef M, Waldmann J, Buchholz M, Feldmann G, Slater EP, Maitra A & Bartsch DK 2010 The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model of pancreatic cancer. Gut 59 630–637. (doi:10.1136/gut.2009.188961).
Fendrich V, Rehm J, Waldmann J, Buchholz M, Christofori G, Lauth M, Slater EP & Bartsch DK 2011a Hedgehog inhibition with cyclopamine represses tumor growth and prolongs survival in a transgenic mouse model of islet cell tumors. Annals of Surgery 253 546–552. (doi:10.1097/SLA.0b013e31820a5bbe).
Fendrich V, Wiese D, Waldmann J, Lauth M, Heverhagen AE, Rehm J & Bartsch DK 2011b Hedgehog inhibition with the orally bioavailable Smo antagonist LDE225 represses tumor growth and prolongs survival in a transgenic mouse model of islet cell neoplasms. Annals of Surgery 254 818–823.(discussion 823) (doi:10.1097/SLA.0b013e318236bc0f).
Ferrara N 2004 Vascular endothelial growth factor: basic science and clinical progress. Endocrine Reviews 25 581–611. (doi:10.1210/er.2003-0027).
Folkman J, Watson K, Ingber D & Hanahan D 1989 Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 339 58–61. (doi:10.1038/339058a0).
Garcia Rodriguez LA & Gonzalez-Perez A 2004 Risk of breast cancer among users of aspirin and other anti-inflammatory drugs. British Journal of Cancer 91 525–529. (doi:10.1038/sj.bjc.6602003).
Gonzalez-Perez A, Ronquist G & Garcia Rodriguez LA 2004 Breast cancer incidence and use of antihypertensive medication in women. Pharmacoepidemiology and Drug Safety 13 581–585. (doi:10.1002/pds.910).
Goudet P, Murat A, Cardot-Bauters C, Emy P, Baudin E, du Boullay Choplin H, Chapuis Y, Kraimps JL, Sadoul JL & Tabarin A et al. 2009 Thymic neuroendocrine tumors in multiple endocrine neoplasia type 1: a comparative study on 21 cases among a series of 761 MEN1 from the GTE (Groupe des Tumeurs Endocrines). World Journal of Surgery 33 1197–1207. (doi:10.1007/s00268-009-9980-y).
Goudet P, Murat A, Binquet C, Cardot-Bauters C, Costa A, Ruszniewski P, Niccoli P, Menegaux F, Chabrier G & Borson-Chazot F et al. 2010 Risk factors and causes of death in MEN1 disease. A GTE (Groupe d'Etude des Tumeurs Endocrines) cohort study among 758 patients. World Journal of Surgery 34 249–255. (doi:10.1007/s00268-009-0290-1).
Hanahan D 1985 Heritable formation of pancreatic β-cell tumours in transgenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature 315 115–122. (doi:10.1038/315115a0).
Harrison-Bernard LM, El-Dahr SS, O'Leary DF & Navar LG 1999 Regulation of angiotensin II type 1 receptor mRNA and protein in angiotensin II-induced hypertension. Hypertension 33 340–346. (doi:10.1161/01.HYP.33.1.340).
Hennessy BT, Smith DL, Ram PT, Lu Y & Mills GB 2005 Exploiting the PI3K/AKT pathway for cancer drug discovery. Nature Reviews. Drug Discovery 4 988–1004. (doi:10.1038/nrd1902).
Hernandez-Presa MA, Bustos C, Ortego M, Tunon J, Ortega L & Egido J 1998 ACE inhibitor quinapril reduces the arterial expression of NF-κB-dependent proinflammatory factors but not of collagen I in a rabbit model of atherosclerosis. American Journal of Pathology 153 1825–1837. (doi:10.1016/S0002-9440(10)65697-0).
Inoue M, Hager JH, Ferrara N, Gerber HP & Hanahan D 2002 VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic β cell carcinogenesis. Cancer Cell 1 193–202. (doi:10.1016/S1535-6108(02)00031-4).
Jensen RT, Niederle B, Mitry E, Ramage JK, Steinmuller T, Lewington V, Scarpa A, Sundin A, Perren A & Gross D et al. 2006 Gastrinoma (duodenal and pancreatic). Neuroendocrinology 84 173–182. (doi:10.1159/000098009).
Jensen RT, Cadiot G, Brandi ML, de Herder WW, Kaltsas G, Komminoth P, Scoazec JY, Salazar R, Sauvanet A & Kianmanesh R 2012 ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumor syndromes. Neuroendocrinology 95 98–119. (doi:10.1159/000335591).
Kopp E & Ghosh S 1994 Inhibition of NF-κB by sodium salicylate and aspirin. Science 265 956–959. (doi:10.1126/science.8052854).
Kosaka T, Miyajima A, Takayama E, Kikuchi E, Nakashima J, Ohigashi T, Asano T, Sakamoto M, Okita H & Murai M et al. 2007 Angiotensin II type 1 receptor antagonist as an angiogenic inhibitor in prostate cancer. Prostate 67 41–49. (doi:10.1002/pros.20486).
Kune GA, Kune S & Watson LF 1988 Colorectal cancer risk, chronic illnesses, operations, and medications: case control results from the Melbourne Colorectal Cancer Study. Cancer Research 48 4399–4404.
Lever AF, Hole DJ, Gillis CR, McCallum IR, McInnes GT, MacKinnon PL, Meredith PA, Murray LS, Reid JL & Robertson JW 1998 Do inhibitors of angiotensin-I-converting enzyme protect against risk of cancer? Lancet 352 179–184. (doi:10.1016/S0140-6736(98)03228-0).
Lopez T & Hanahan D 2002 Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1 339–353. (doi:10.1016/S1535-6108(02)00055-7).
Ludwig L, Kessler H, Wagner M, Hoang-Vu C, Dralle H, Adler G, Bohm BO & Schmid RM 2001 Nuclear factor-κB is constitutively active in C-cell carcinoma and required for RET-induced transformation. Cancer Research 61 4526–4535.
Mahmoud NN, Dannenberg AJ, Mestre J, Bilinski RT, Churchill MR, Martucci C, Newmark H & Bertagnolli MM 1998 Aspirin prevents tumors in a murine model of familial adenomatous polyposis. Surgery 124 225–231. (doi:10.1016/S0039-6060(98)70124-2).
Maitra A, Ashfaq R, Gunn CR, Rahman A, Yeo CJ, Sohn TA, Cameron JL, Hruban RH & Wilentz RE 2002 Cyclooxygenase 2 expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasia: an immunohistochemical analysis with automated cellular imaging. American Journal of Clinical Pathology 118 194–201. (doi:10.1309/TPG4-CK1C-9V8V-8AWC).
Metz DC & Jensen RT 2008 Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology 135 1469–1492. (doi:10.1053/j.gastro.2008.05.047).
Miyajima A, Kosaka T, Asano T, Asano T, Seta K, Kawai T & Hayakawa M 2002 Angiotensin II type I antagonist prevents pulmonary metastasis of murine renal cancer by inhibiting tumor angiogenesis. Cancer Research 62 4176–4179.
Miyajima A, Kikuchi E, Kosaka T & Oya M 2009 Angiotensin II type 1 receptor antagonist as an angiogenic inhibitor in urogenital cancer. Reviews on Recent Clinical Trials 4 75–78. (doi:10.2174/157488709788185996).
Nakai Y, Isayama H, Ijichi H, Sasaki T, Sasahira N, Hirano K, Kogure H, Kawakubo K, Yagioka H & Yashima Y et al. 2010 Inhibition of renin–angiotensin system affects prognosis of advanced pancreatic cancer receiving gemcitabine. British Journal of Cancer 103 1644–1648. (doi:10.1038/sj.bjc.6605955).
Neo JH, Ager EI, Angus PW, Zhu J, Herath CB & Christophi C 2010 Changes in the renin angiotensin system during the development of colorectal cancer liver metastases. BMC Cancer 10 134. (doi:10.1186/1471-2407-10-134).
Oberg K & Eriksson B 2005 Endocrine tumours of the pancreas. Best Practice & Research. Clinical Gastroenterology 19 753–781. (doi:10.1016/j.bpg.2005.06.002).
Pacifico F & Leonardi A 2010 Role of NF-κB in thyroid cancer. Molecular and Cellular Endocrinology 321 29–35. (doi:10.1016/j.mce.2009.10.010).
Plockinger U & Wiedenmann B 2007 Neuroendocrine tumors. Biotherapy. Best Practice & Research. Clinical Endocrinology & Metabolism 21 145–162. (doi:10.1016/j.beem.2007.01.002).
Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D & Vinik A et al. 2011 Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. New England Journal of Medicine 364 501–513. (doi:10.1056/NEJMoa1003825).
Rhodes DR, Ateeq B, Cao Q, Tomlins SA, Mehra R, Laxman B, Kalyana-Sundaram S, Lonigro RJ, Helgeson BE & Bhojani MS et al. 2009 AGTR1 overexpression defines a subset of breast cancer and confers sensitivity to losartan, an AGTR1 antagonist. PNAS 106 10284–10289. (doi:10.1073/pnas.0900351106).
Rindi G 2010 The ENETS guidelines: the new TNM classification system. Tumori 96 806–809.
Rivera E, Arrieta O, Guevara P, Duarte-Rojo A & Sotelo J 2001 AT1 receptor is present in glioma cells; its blockage reduces the growth of rat glioma. British Journal of Cancer 85 1396–1399. (doi:10.1054/bjoc.2001.2102).
Rodrigues-Ferreira S, Morel M, Reis RI, Cormier F, Baud V, Costa-Neto CM & Nahmias C 2012 A novel cellular model to study angiotensin II AT2 receptor function in breast cancer cells. International Journal of Peptides 2012 745027. (doi:10.1155/2012/745027).
Sakamoto K, Sugimoto K & Fujimura A 2001 Different potentiating effects of imidapril and enalapril on kaolin-induced writhing reaction in mice. Life Sciences 68 2415–2421. (doi:10.1016/S0024-3205(01)01033-5).
Sclabas GM, Uwagawa T, Schmidt C, Hess KR, Evans DB, Abbruzzese JL & Chiao PJ 2005 Nuclear factor κB activation is a potential target for preventing pancreatic carcinoma by aspirin. Cancer 103 2485–2490. (doi:10.1002/cncr.21075).
Sechi LA, Griffin CA, Giacchetti G, Valentin JP, Llorens-Cortes C, Corvol P & Schambelan M 1996 Tissue-specific regulation of type 1 angiotensin II receptor mRNA levels in the rat. Hypertension 28 403–408. (doi:10.1161/01.HYP.28.3.403).
Sipahi I, Debanne SM, Rowland DY, Simon DI & Fang JC 2010 Angiotensin-receptor blockade and risk of cancer: meta-analysis of randomised controlled trials. Lancet Oncology 11 627–636. (doi:10.1016/S1470-2045(10)70106-6).
Teo KK 2011 Is there an optimal combination of antihypertensives for preventing cardiovascular events? Journal of Hypertension 29 1483–1485. (doi:10.1097/HJH.0b013e32834928ff).
Thun MJ, Namboodiri MM & Heath CW Jr 1991 Aspirin use and reduced risk of fatal colon cancer. New England Journal of Medicine 325 1593–1596. (doi:10.1056/NEJM199112053252301).
Uemura H, Hasumi H, Ishiguro H, Teranishi J, Miyoshi Y & Kubota Y 2006 Renin–angiotensin system is an important factor in hormone refractory prostate cancer. Prostate 66 822–830. (doi:10.1002/pros.20407).
Vinson GP, Barker S & Puddefoot JR 2012 The renin–angiotensin system in the breast and breast cancer. Endocrine-Related Cancer 19 R1–R19. (doi:10.1530/ERC-11-0335).
Volpert OV, Ward WF, Lingen MW, Chesler L, Solt DB, Johnson MD, Molteni A, Polverini PJ & Bouck NP 1996 Captopril inhibits angiogenesis and slows the growth of experimental tumors in rats. Journal of Clinical Investigation 98 671–679. (doi:10.1172/JCI118838).
Waldmann J, Fendrich V, Habbe N, Bartsch DK, Slater EP, Kann PH, Rothmund M & Langer P 2009 Screening of patients with multiple endocrine neoplasia type 1 (MEN-1): a critical analysis of its value. World Journal of Surgery 33 1208–1218. (doi:10.1007/s00268-009-9983-8).
Wilop S, von Hobe S, Crysandt M, Esser A, Osieka R & Jost E 2009 Impact of angiotensin I converting enzyme inhibitors and angiotensin II type 1 receptor blockers on survival in patients with advanced non-small-cell lung cancer undergoing first-line platinum-based chemotherapy. Journal of Cancer Research and Clinical Oncology 135 1429–1435. (doi:10.1007/s00432-009-0587-3).
Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J & de Vries EG et al. 2011 Everolimus for advanced pancreatic neuroendocrine tumors. New England Journal of Medicine 364 514–523. (doi:10.1056/NEJMoa1009290).
(V Fendrich, C L Lopez and J Manoharan contributed equally to this work)