ArticlePDF Available

Identification of a Novel Raf-1 Pathway Activator that Inhibits Gastrointestinal Carcinoid Cell Growth

Authors:

Abstract and Figures

Carcinoids are neuroendocrine tumors (NET) that secrete hormones, including serotonin, resulting in the malignant carcinoid syndrome. In addition to the significant morbidity associated with the syndrome, carcinoids are frequently metastatic at diagnosis, and untreated mortality at 5 years exceeds 70%. Surgery is the only curative option, and the need for other therapies is clear. We have previously shown that activation of Raf-1 inhibits carcinoid cell proliferation. We investigated the ability of leflunomide (LFN), a Food and Drug Administration-approved medication for the treatment of rheumatoid arthritis, and its active metabolite teriflunomide (TFN) as a potential anti-NET treatment. LFN and TFN inhibit the in vitro proliferation of gastrointestinal carcinoid cells and induce G(2)-M phase arrest. Daily oral gavage of nude mice with subcutaneous xenografted carcinoid tumors confirms that LFN can inhibit NET growth in vivo. Treatment with TFN suppresses the cellular levels of serotonin and chromogranin A, a glycopeptide co-secreted with bioactive hormones. Additionally, TFN reduces the level of achaete-scute complex-like 1 (ASCL1), a NET marker correlated with survival. These effects are associated with the activation of the Raf-1/mitiogen-activated protein kinase kinase/extracellular signal-regulated kinase-1/2 pathway, and blockade of mitiogen-activated protein kinase kinase signaling reversed the effects of TFN on markers of the cell cycle and ASCL1 expression. In summary, LFN and TFN inhibit carcinoid cell proliferation in vitro and in vivo and alter the expression of NET markers. This compound thus represents an attractive target for further clinical investigation.
Content may be subject to copyright.
Identification of a novel Raf-1 pathway activator that inhibits
gastrointestinal carcinoid cell growth
Mackenzie R. Cook, Scott N. Pinchot, Renata Jaskula-Sztul, Jie Luo, Muthusamy
Kunnimalaiyaan, and Herbert Chen
Endocrine Surgery Research Laboratory, University of Wisconsin, and the University of Wisconsin
Carbone Cancer Center, Madison, Wisconsin 53792-7375
Abstract
Carcinoids are neuroendocrine tumors (NETs) that secrete hormones, including serotonin, resulting
in the malignant carcinoid syndrome. In addition to the significant morbidity associated with the
syndrome, carcinoids are frequently metastatic at diagnosis and untreated mortality at 5 years tops
70%. Surgery is the only curative option and the need for other therapies is clear. We have previously
shown that activation of Raf-1 inhibits carcinoid cell proliferation.
We investigated the ability of Leflunomide (LFN), an FDA approved medication for the treatment
of rheumatoid arthritis, and its active metabolite Teriflunomide (TFN) as a potential anti-NET
treatment. LFN and TFN inhibit the in vitro proliferation of gastrointestinal carcinoid cells and induce
G2/M phase arrest. Daily oral gavage of nude mice with subcutaneous xenografted carcinoid tumors
confirms that LFN can inhibit NET growth in vivo. Treatment with TFN suppresses the cellular levels
of serotonin and chromogranin A, a glycopeptide co-secreted with bioactive hormones. Additionally
TFN reduces the level of Achaete-Scute Complex-Like 1 (ASCL1), a NET marker correlated with
survival. These effects are associated with the activation of the Raf-1/MEK/ERK1/2 pathway and
blockade of MEK signaling reversed the effects of TFN on markers of the cell cycle and ASCL1
expression.
In summary, LFN and TFN inhibit carcinoid cell proliferation in vitro and in vivo and alter the
expression of NET markers. This compound thus represents an attractive target for further clinical
investigation.
Keywords
Leflunomide; Teriflunomide; Carcinoid; Raf-1; Achaete-Scute Complex-Like 1
Introduction
A compound originally reported to be effective in Rheumatoid arthritis, Leflunomide (LFN)
(Arava, SU-101) has proven to be remarkably safe in human patients (1,2). Approved by the
FDA in 1998, LFN is nearly completely converted to its main active metabolite, Teriflunomide
(TFN) in first pass metabolism (1-3). While its current indications are limited to rheumatologic
conditions, recent studies have focused on the ability of LFN and TFN to inhibit the
proliferation of a variety of human malignancies, including an in vivo model of colon cancer.
Corresponding author and requests for reprints to: Herbert Chen, H4/722 Clinical Science Center, 600 Highland Avenue, Madison,
WI 53792-7375, Office: (608) 263-1387, FAX: (608) 263-7652, Chen@surgery.wisc.edu.
The authors have no potential conflicts of interest to disclose.
NIH Public Access
Author Manuscript
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
Published in final edited form as:
Mol Cancer Ther. 2010 February ; 9(2): 429. doi:10.1158/1535-7163.MCT-09-0718.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
While capable of inducing apoptosis in some cell lines, it does not appear that this is a general
effect of either LFN or TFN, as they are also capable of inducing cell cycle arrest without
necrosis or apoptosis. This is consistent with the known safety profile of LFN in vivo (4-8).
The recent focus on oncologic applications spurred our interest in the effect of LFN and TFN
in carcinoid cancer (9).
Derived from the diffuse enterochromaffin cells of the gastrointestinal tract, carcinoid cancer
is a subtype of neuroendocrine tumors (NETs). They characteristically secrete a variety of
bioactive hormones, including serotonin, that are implicated in the malignant carcinoid
syndrome(10). These cancers also characteristically express high levels of chromogranin A
(CgA), an acidic glycopeptide co-secreted with hormones and considered a clinical marker of
disease. High serum levels of CgA have been associated with a poor clinical prognosis in
carcinoid tumors (11-13). The basic Helix-Loop-Helix transcription factor, Achaete-Scute
Complex-Like 1 (ASCL1) is similarly highly expressed in NETs. Important in the development
of normal NE cells and lost in the normal adult tissue, ASCL1 expression is associated with
poor prognosis in small cell lung cancer, a related NET (14).
Gastrointestinal carcinoids are often clinically silent until hepatic metastases are present and
the patient begins to experience the debilitating flushing, wheezing and diarrhea of the
malignant carcinoid syndrome (15). It is important to note that in addition to this significant
morbidity, the 5 year untreated mortality is approximately 70% (15-20). Surgical resection
may be potentially curative, though is difficult with metastatic disease and optimal resection
still results in significant mortality at 5 years (16). Adjuvant therapies, including chemotherapy
and radiation, have shown limited success and patients become rapidly resistant to octreotide
(15,17,19,20). The severity of carcinoid disease and the lack of effective therapies highlights
the need for targeted treatment options.
The Raf-1 pathway has been described as having anti-carcinoid effects and validated as a
potential target in the treatment of carcinoid disease. We and others have shown that humans
NETs lack phosphorylated ERK 1/2 (extracellular signal-regulated kinase 1/2), a marker of
Raf-1 pathway activation. Activation of the Raf-1/MEK/ERK1/2 pathway suppresses levels
of ASCL1 as well as CgA and serotonin. In a model of medullary thyroid cancer, a related
NET, the ability of Raf-1 pathway activation to suppress in vivo NET growth was confirmed
(21-25). While several excellent reviews of the Raf-1 pathway exist, we will briefly summarize
the important points. Active, phosphorylated, Raf-1 phosphorylates MEK1/2 (Mitogen-
activated protein kinase 1/2) and subsequently ERK 1/2 which then targets multiple key cellular
pathways (26).
In this paper we describe, for the first time, the activity of LFN and TFN in NET cells. We
show that LFN and its active metabolite, TFN, are capable of suppressing growth of human
carcinoid cells in vitro, by inducing cell cycle arrest and that this finding can be replicated in
vivo. We also show that these compounds can inhibit the cellular expression of CgA and
serotonin, compounds linked to the poor prognosis of NETs. Furthermore, we show that these
compounds suppress ASCL1, a well characterized marker of NE malignancy, at the protein
and the mRNA level through a mechanism that is predominately dependent upon the Raf-1/
MEK/ERK1/2 pathway.
Methods
Cell Culture and Treatment
Human GI carcinoid cancer cells (BON), graciously provided by Drs. B. Mark Evers and
Courtney M. Townsend, Jr. (University of Texas Medical Branch, Galveston, TX, USA), and
NCI-H727 human bronchopulmonary carcinoid tumor cells (American Type Culture
Cook et al. Page 2
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Collection, Mannassas, VA, USA) were maintained in DMEM F12 and RPMI1640 (Life
Technologies, Rockville, MD, USA), respectively, supplemented with 10% fetal bovine serum
(Sigma-Aldrich, St. Louis, MO, USA), 100 IU/mL penicillin and 100 μg/mL streptomycin
(Life Technologies) in a humidified atmosphere of 5% CO2 in air at 37°C
LFN and TFN (Calbiochem, San Diego CA) were dissolved in DMSO at 100mM and stored
at 80C. Fresh dilutions in media were prepared for each experiment and new media dilutions
were added every 48 hours for multi-day experiments. The MEK1/2 inhibitor UO126
(Promega, Madison WI) was stored as a stock solution in DMSO at 20°C and fresh dilutions
in media were prepared for each experiment.
Cells were plated and allowed to adhere overnight. Either LFN or TFN at concentrations
ranging from 0-125 μM was then added and cells incubated for 48, 96 hours or 168 hours.
When U0126 was used, cells were pretreated with 10 μM U0126 for 1 hour prior to addition
of 100 μM TFN.
Cell Proliferation Assay
Carcinoid tumor cell proliferation was measured by the 3-[4,5-dimethylthiazol-2-yl]-2,5
diphenyl tetrazolium bromide (MTT) rapid colorimetric assay as previously described (27).
Briefly, BON cells were seeded on 24-well plates and incubated overnight under standard
conditions to allow cell attachment. Cells were then treated with LFN or TFN (Calbiochem)
in quadruplicate with up to 125 μM and incubated for up to 6 days.
The MTT assay was performed by replacing the standard medium with 250 μL of serum-free
medium containing 0.5 mg/ml MTT and incubating at 37°C for 4 hours. After incubation, 750
μl of dimethyl sulfoxide (Fischer Scientific, Pittsburgh, PA) was added to each well and mixed
thoroughly. The multiwell plates were then measured at 540 nm using a spectrophotometer
(μQuant; Bio-Tek Instruments, Winooski, VT).
Cell Cycle Analysis by Flow Cytometry
BON cells were treated with TFN (0, 50, and 100 μM) for 48h. After treatment, the cells were
harvested, washed with ice cold 0.9% saline buffered with phosphate to a pH of 7.4 (1x PBS),
and viability was determined using trypan blue exclusion (Mediatech, Herndon, VA). For DNA
content analysis, cells (1 × 106) were fixed with ice cold 70% ethanol, washed with 1x PBS,
incubated with 0.2 mg/ml RNAse-A and stained with 10 μg/ml Propidium iodide (PI) staining
solution. FACS analysis was performed on a flow cytometer at 488 nm (FACSCalibur flow
cytometer; BD Biosciences), results were analyzed with ModFit LT 3.2 software (Verity,
Topsham, ME).
Tumor Xenograft Studies
Male nude (Nu/Nu) mice (Charles River Laboratories, Wilmington, MA) were injected
subcutaneously with 1×106 gastrointestinal carcinoid cells suspended in 100 μL Hanks
Balanced Salt Solution (Mediatech Inc. Manassas VA). Palpable tumors were allowed to
develop, and stratified randomization was used to assign mice to either a control or treatment
group. Mice were treated with either 35 mg/kg Leflunomide suspended in 1.5% carboxymethyl
cellulose, or 1.5 % carboxymethyl cellulose alone, by daily oral gavage. This dose was chosen
based upon previously published doses in a mouse model of colon cancer(5). Tumor size was
measured with vernier calipers every four days and tumor volume calculated using the formula,
0.52 x [(length) x (width)2], where width was defined as the shorter tumor dimension. After
28 days of treatment, animals were sacrificed. All animal care and treatment was performed
in compliance with our animal care protocol approved by the University of Wisconsin–
Madison animal care and use committee.
Cook et al. Page 3
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Immunoblot Analysis
After treatment as described, cells were washed with ice cold 1X PBS, and total protein lysates
prepared. Total protein concentration was measured with a bicinchoninic acid assay kit (Pierce
Protein Research Products, Rockford, IL). Denatured cellular extracts were resolved by
10%-12% SDS-PAGE (Invitrogen), transferred onto a nitrocellulose membrane (Bio-Rad
Laboratories, Hercules, CA) and incubated overnight in the appropriate primary antibody. The
antibody dilutions were as follows: 1:1,000 for phosphorylated ERK 1/2thr202/tyr204, Total ERK
1/2, phosphorylated MEK1/2ser217/221, phosphorylated Raf-1ser338, phosphorylated Glycogen
Synthase Kinase-3βSer9 (GSK-3β), phosphorylated AKTSer473 and cyclin B1 (Cell Signaling
Technology, Beverly, MA), 1:2000 for mammalian Achaete-Scute Homologue-1 for detection
of ASCL1 (BD PharMingen, San Diego, CA), CgA (Zymed Laboratories, San Francisco, CA),
1:1000 (Cell Signaling) and 1:10,000 for glyceraldehyde-3-phosphate dehydrogenase
(GAPDH) (Trevigen, Gaithersburg, MD). Horseradish peroxidase-conjugated goat anti-rabbit
or anti-mouse secondary antibodies (Pierce) were used depending on the source of the primary
antibody. Immunstar (Bio-Rad), or SuperSignal West Pico or Femto (Pierce) kits were used
per the manufacturer’s instructions for detection.
Serotonin Enzyme Linked Immunosorbent Assay (ELISA)
To determine serotonin levels in cellular extracts of carcinoid cells treated with TFN for 48 h,
we utilized a serotonin enzyme-linked immunosorbant assay (ELISA) kit as per the
manufacturer’s instructions (Fitzgerald, Concord, MA). The multiwell plates were then
measured at 405 nm using a spectrophotometer (μQuant; Bio-Tek Instruments, Winooski, VT).
Serotonin concentrations were calculated based on the manufacturer’s standard curve.
Quantitative real-time reverse transcriptase Polymerase Chain Reaction (qPCR)
BON cells were treated with TFN (0, 50, and 100 μM) for 48h. Total RNA was harvested using
a Qiagen RNeasy minikit (Qiagen, Valencia, CA), per the manufacturers directions. Integrity
was assured and concentration determined using a Nanodrop spectrophotometer (Nanodrop,
Wilmington DE). Exactly 2 μg of total RNA were then converted to complementary DNA
(cDNA) using the iScript cDNA synthesis kit (Biorad) according to the manufacturers
directions.
The qPCR reactions were performed on the Biorad iCycler, conditions were: 3 minutes at 95°
C, 35 cycles of: 30 seconds at 95°C, 25 seconds at 60°C and 30 seconds at 72°C followed by
1 minute at 95°C and 1 minute at 55°C. Primers were obtained from Integrated DNA
Technologies (Coralville, IA) and sequences used were: ASCL1(F: 5 TCC CCC AAC TAC
TCC AAC GAC 3, R: 5 CCC TCC CAA CGC CAC TG 3), GAPDH (F: 5 ACC TGC CAA
ATA TGA TGA C 3, R: ACC TGG TGC TCA GTG TAG 3). Results were normalized to
GAPDH from the same sample. Expression of ASCL1 calculated for each treatment using the
formula 2(Ct(GAPDH)-Ct(ASCL1)) as outlined in the iCycler Applications Guide (Biorad).
Expression was then plotted as average ± standard error of the mean (SEM).
Statistical Analysis
Densitometric analysis of Western blotting results was performed using Quantity One software
v. 4.6.3 (Biorad). Analysis of MTT growth curves was performed using a one way analysis of
variance (ANOVA) testing and Bonferroni post-hoc testing. In the analysis of the nude mouse
xenograft experiment, we utilized a Pearson’s chi-square test. Student’s t-test was utilized to
compare ELISA and qPCR results. In this work, p < 0.05 was considered statistically significant
and analyses were performed with SPSS software v. 10.0 (SPSS, Chicago, IL). Unless
specifically noted, all data are represented as mean ± SEM.
Cook et al. Page 4
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Results
Leflunomide and Teriflunomide inhibit in vitro proliferation of GI carcinoid cells
As recent studies have described attributed anti-proliferative effects to LFN and TFN, we first
sought to examine the ability of LFN and TFN to inhibit carcinoid proliferation in vitro (4,6).
BON cells were treated with increasing doses of either LFN or TFN and the MTT assay was
performed at 2, 4 and 6 days. Dose dependent, statistically significant growth inhibition was
observed after treatment with both LFN FIGURE 1A and TFN FIGURE 1B at 4 and 6 days (p
0.05 vs. control). These data suggest that LFN and its major metabolite can inhibit the in
vitro proliferation of human GI carcinoid cells.
Treatment with LFN and TFN additionally resulted in the suppression of in vitro proliferation
after 2 days of treatment, though this did not reach statistical significance in all treatment groups
(Data not shown). In total, these data suggest that LFN and TFN can inhibit in vitro proliferation
of gastrointestinal carcinoid cells.
Teriflunomide induces cell cycle arrest and suppresses in vivo carcinoid growth
TFN has been published to induce cell cycle arrest and apoptosis, thus we performed Western
blot analysis for markers of cell cycle progression and apoptosis as well as PI exclusion flow
cytometry to determine the mechanism of in vitro growth suppression (4,6).
Treatment with increasing concentrations of TFN resulted in a dose dependent induction of
cyclin B1, a marker of G2/M phase transition FIGURE 2A (28, 29). We did not observe any
cleavage of poly (ADP)-ribose polymerase (PARP), a marker of apoptosis (data not shown).
These protein results suggest that the mechanism of growth inhibition is cell cycle arrest. We
next sought to confirm this finding using PI exclusion flow cytometry. Treatment with
increasing doses of TFN for 48 hours resulted in the progressive accumulation of S-phase
products FIGURE 2B. These data, in total, suggest that arrest prior to the G2/M transition is
the mechanism of TFN induced in vitro growth suppression.
Approved by the FDA in 1998, LFN is currently in clinical use and has proven to be remarkably
safe in human patients (2,3). With this in mind, we wanted to investigate the ability of LFN,
converted to TFN in first pass metabolism, to suppress the in vivo growth of a NET xenograft
(1,30). Treatment of subcutaneous GI carcinoid xenografts with daily oral gavage of LFN
resulted in the statistically significant suppression of tumor growth after 24 days of treatment
(p < 0.02) FIGURE 2C. These data suggest that the growth inhibitory effects observed in
vitro can be replicated in vivo with oral LFN.
Leflunomide and Teriflunomide can alter the expression of neuroendocrine markers
We then looked to investigate the effect of LFN and TFN on CgA, ASCL1 and serotonin. These
markers are of particular interest as they have been associated with poor prognosis and the
malignant carcinoid syndrome (10-12,14,22,31,32). In order to determine if LFN and TFN
were able to modulate the expression of NE markers, we treated BON cells for 48 hours with
increasing doses of LFN or TFN. Treatment with 125 μM LFN resulted in the 61% suppression
of CgA and the 42% suppression of ASCL1 protein FIGURE 3A. Treatment with 125 μM TFN
resulted in the 65% suppression of CgA and the 84% suppression of ASCL1 protein FIGURE
3B. Densitometric analysis was used to compare the 125 μM dose of LFN and TFN to their
respective controls.
The ability of TFN to alter the expression of cellular serotonin, a hormone important in the
development of the carcinoid syndrome, was next investigated using an ELISA. Treatment
Cook et al. Page 5
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
with TFN reduced the cellular expression of serotonin by 77% after 48 hours of treatment (p
< 0.005) FIGURE 3C.
As the expression of ASCL1 could potentially be modulated at several levels, we next
investigated the levels of ASCL1 mRNA after treatment with TFN (33,34). Using qPCR, we
observed that treatment with 50 and 100 μM TFN for 48 hours resulted in the suppression of
ASCL1 levels by 54% and 62%, respectively (p < 0.05) FIGURE 3D.
We next sought to confirm that the above results could be generalized. Treatment of NCI-H727
bronchopulmonary carcinoid cells with increasing doses of either LFN, FIGURE 3E, or TFN,
FIGURE 3F, resulted in the dose dependent inhibition of ASCL1 protein expression, by
western blot analysis. Treatment with 100μM LFN and TFN resulted in a 43% and 71%
reduction in ASCL1 protein expression, respectively. These data suggest that the ability of
LFN and TFN to alter the expression of a neuroendocrine tumor marker associated with poor
prognosis is not limited to a single cell line.
These data, in total, suggest that LFN and TFN can suppress the expression of NET markers
that are associated with the carcinoid syndrome as well a poor survival in NETs (14,31).
Teriflunomide activates the Raf-1/MEK/ERK pathway
The Raf-1/MEK/ERK1/2 pathway has been described by our group as being an important
regulator of NET cellular proliferation and hormone production (13,23-25). A relationship
between phosphorylated ERK1/2 and the regulation of the G2/M checkpoint has additionally
been described (35,36). Given these known associations, and the effects described above, we
hypothesized that TFN was inducing Raf-1 pathway activation. To examine the status of Raf-1
signaling, BON cells were treated with increasing doses of TFN and Western blotting
performed for key components of the pathway.
A dose dependent induction of phosphorylated Raf-1 as well as phosphorylated MEK1/2 and
phosphorylated ERK 1/2 was observed in BON cells treated for 48 hours with TFN FIGURE
4A. These results suggest that the Raf-1/MEK/ERK1/2 pathway is intact and that TFN is
capable of efficiently inducing pathway activation. The levels of total ERK 1/2 remained
unchanged, suggesting that pathway activation and not translation of additional protein was
responsible for the observed results.
Suppression of ASCL1 and induction of cell cycle arrest is a direct result of Raf-1 pathway
activation
In order to show that the effects on NET cells are directly due to Raf-1/MEK/ERK pathway
activation, we used U0126, an inhibitor of phosphorylated MEK1/2, to disrupt activation of
this pathway. The induction of phosphorylated ERK 1/2 seen with 100 μM TFN can be totally
blocked by pretreatment with 10 μM U0126. This suggests that TFN induces the
phosphorylation of ERK 1/2 via MEK1/2. Importantly, the reduction of ASCL1 and induction
of cyclin B1 are similarly reversed by pretreatment with U0126 FIGURE 4B.
As the regulation of ASCL1 was shown to be at the level of mRNA, we next performed qPCR
on BON cells treated with either TFN alone or TFN after pre-treatment with UO126. While
100 μM TFN potently suppresses the level of ASCL1 mRNA, pre-treatment with U0126 is
able to block this suppression FIGURE 4C. These data suggest that TFN mediated suppression
of ASCL1 is dependent upon the Raf-1/MEK/ERK pathway at both the protein and mRNA
level. Additionally, these results suggest that TFN induced cell cycle arrest may also be
modulated by this pathway.
Cook et al. Page 6
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
In order to demonstrate that the effects on protein phosphorylation were pathway specific, we
next investigated the phosphorylation state of two other anti-carcinoid pathways, GSK-3β
FIGURE 4D and Akt FIGURE 4E (37-39). No change in the phosphorylation state of these
proteins was noted after treatment with TFN. These data suggest that the effects on protein
phosphorylation may be specific to the Raf-1/MEK/ERK1/2 pathway.
Discussion
Carcinoid tumors are the second most common source of isolated hepatic metastases, after
colorectal cancer, and carry an untreated 5 year mortality in excess of 70% (16). The most
effective chemotherapeutic combination tried to date has resulted in a less than 6% volume
response rate(40). This lack of efficacy extends to other adjuvant therapies as well as the
palliative option, octreotide, to which patients rapidly become resistant (10). The need for novel
targeted therapies is therefore clear.
We present data that suggests that LFN and TFN are novel potential therapeutic options for
the treatment of carcinoid disease. LFN and TFN are capable of suppressing in vitro cell
proliferation and TFN is shown to induce cell cycle arrest prior to the G2/M phase transition.
Additionally, in animal studies, we show that the in vitro growth inhibitory effects of LFN can
be replicated in vivo with daily oral dosing of LFN. These drugs can additionally suppress the
levels of CgA, ASCL1 and serotonin, all markers of NE malignancy. The suppression of these
three markers is an important point as serotonin is a mediator of the malignant carcinoid
syndrome and the expression of CgA and ASCL1 have been correlated with poor prognosis
(10-12,14,31,32). These effects on carcinoid cell markers appear to be mediated predominately
through the Raf-1/MEK/ERK1/2 pathway, a pathway that has been extensively studied as a
potential anti-carcinoid target (13,23-25).
The Raf-1 pathway has been traditionally thought of as a tumorigenic pathway and has been
noted to be either mutated or over-expressed in hepatocellular carcinoma, non-small cell lung
cancer, melanoma and papillary thyroid carcinoma (41,42). In tumors of NE origin, however,
there is minimal phosphorylation of ERK1/2 at baseline, suggesting that the Raf-1 pathway
does not play an essential oncogenic role. Activation of Raf-1 signaling in a gastrointestinal
carcinoid cell, with an estrogen inducible construct, results in the suppression of ASCL1, CgA
and serotonin(13,23). Additionally, in a medullary thyroid cancer xenograft model, activation
of the Raf-1 pathway resulted in the suppression of in vivo tumor growth (24). This work
suggests that Raf-1 pathway activation, if accomplished pharmacologically, could be a potent
strategy for the inhibition of carcinoid cancer and other NETs.
Our group has described ZM336372 and Tautomycetin (TTM) as two compounds that appear
to activate the Raf-1 pathway in vitro and inhibit the growth of carcinoid cancer and MTC
(25,43). Significant obstacles, however, exist between these compounds and clinical
applicability. TTM is a natural compound that must be isolated from Streptomyces
spiroverticillatus, limiting the quantity that can be produced at any one time. The exceptionally
poor solubility of ZM336372 limits its clinical utility and attempts to produce more soluble
sister compounds have met with limited success. In addition to these limitations, neither
compound has been described in vivo and thus ability to achieve necessary concentrations with
acceptable toxicity is unknown.
In contrast, we present LFN as a Raf-1 pathway activator in NETs that is FDA approved and
known to be safe in humans. Serum concentrations of TFN in human rheumatoid arthritis
patients treated with daily oral LFN are greater than 200μM, making the doses used in vitro
comparable to those attainable in humans(1,2). Additionally, peak serum TFN concentrations
in an in vivo model of oral LFN administration approach 500μM(44). Our animal data suggests
Cook et al. Page 7
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
that it is possible, even with oral dosing, to achieve blood concentrations of TFN sufficient to
slow the rate of gastrointestinal carcinoid cell proliferation. It is possible that higher doses
would result in more significant tumor inhibition. It is interesting that tumor size in the
treatment groups did not appear to diverge until after 14 days of treatment, perhaps suggesting
a role for the anti-angiogenic effects of LFN described by others (5). A larger upcoming study
designed to confirm and extend the in vivo data presented here will examine higher doses and
allow for histologic examination of the xenografted tumors.
LFN therefore, more than ZM336372 and TTM, represents a potential therapy for NETs
targeting the Raf-1 pathway. We conclude that LFN is worthy of additional study, including a
larger animal study and potentially an initial human trial.
Acknowledgments
Financial support:
Howard Hughes Medical Institute (MRC)
NIH – RO1 CA121115 (HC)
NIH – RO1 CA109053 (HC)
American College of Surgeons: George H. A. Clowes Jr. Memorial Research Career Development Award (HC)
Carcinoid Cancer Foundation Research Award (HC)
Abbreviations list
LFN Leflunomide
TFN Teriflunomide
NETs Neuroendocrine Tumors
MTC Medullary Thyroid Cancer
ASCL1 Achaete-Scute Complex-Like 1
CgA Chromogranin A
GI Gastrointestinal
PI Propidium Iodide
MTT 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide
ELISA Enzyme Linked Immunosorbent Assay
qPCR Quantitative Real-Time Polymerase Chain Reaction
ERK1/2 Extracellular regulated kinase 1/2
MEK1/2 Mitogen-activated protein kinase kinase 1/2
GSK-3βGlycogen Synthase Kinase – 3 Beta
References
1. Tallantyre E, Evangelou N, Constantinescu CS. Spotlight on teriflunomide. Int MS J Jun;2008 15(2):
62–8. [PubMed: 18782502]
2. Li EK, Tam LS, Tomlinson B. Leflunomide in the treatment of rheumatoid arthritis. Clin Ther Apr;
2004 26(4):447–59. [PubMed: 15189743]
Cook et al. Page 8
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
3. Mladenovic V, Domljan Z, Rozman B, et al. Safety and effectiveness of leflunomide in the treatment
of patients with active rheumatoid arthritis. Results of a randomized, placebo-controlled, phase II
study. Arthritis Rheum Nov;1995 38(11):1595–603. [PubMed: 7488280]
4. Baumann P, Mandl-Weber S, Volkl A, et al. Dihydroorotate dehydrogenase inhibitor A771726
(leflunomide) induces apoptosis and diminishes proliferation of multiple myeloma cells. Mol Cancer
Ther Feb;2009 8(2):366–75. [PubMed: 19174558]
5. Mall JW, Myers JA, Xu X, Saclarides TJ, Philipp AW, Pollmann C. Leflunomide reduces the
angiogenesis score and tumor growth of subcutaneously implanted colon carcinoma cells in the mouse
model. Chirurg Jul;2002 73(7):716–20. [PubMed: 12242982]
6. Ringshausen I, Oelsner M, Bogner C, Peschel C, Decker T. The immunomodulatory drug Leflunomide
inhibits cell cycle progression of B-CLL cells. Leukemia Mar;2008 22(3):635–8. [PubMed: 17805332]
7. Xu X, Williams JW, Gong H, Finnegan A, Chong AS. Two activities of the immunosuppressive
metabolite of leflunomide, A77 1726. Inhibition of pyrimidine nucleotide synthesis and protein
tyrosine phosphorylation. Biochem Pharmacol Aug 23;1996 52(4):527–34. [PubMed: 8759024]
8. Xu X, Shen J, Mall JW, et al. In vitro and in vivo antitumor activity of a novel immunomodulatory
drug, leflunomide: mechanisms of action. Biochem Pharmacol Nov 1;1999 58(9):1405–13. [PubMed:
10513984]
9. Manna SK, Aggarwal BB. Immunosuppressive leflunomide metabolite (A77 1726) blocks TNF-
dependent nuclear factor-kappa B activation and gene expression. J Immunol Feb 15;1999 162(4):
2095–102. [PubMed: 9973483]
10. Davis Z, Moertel C, McIlrath D. The malignant carcinoid syndrome. Surg Gynecol Obstet Oct;1973
137(4):637–44. [PubMed: 4730072]
11. Janson E, Holmberg L, Stridsberg M, et al. Carcinoid tumors: analysis of prognostic factors and
survival in 301 patients from a referral center. Ann Oncol Jul;1997 8(7):685–90. [PubMed: 9296223]
12. Seregni E, Ferrari L, Bajetta E, Martinetti A, Bombardieri E. Clinical significance of blood
chromogranin A measurement in neuroendocrine tumours. Ann Oncol 2001;12(Suppl 2):S69–72.
[PubMed: 11762355]
13. Sippel RS, Carpenter JE, Kunnimalaiyaan M, Chen H. The role of human achaete-scute homolog-1
in medullary thyroid cancer cells. Surgery Dec;2003 134(6):866–71. discussion 71-3. [PubMed:
14668716]
14. Jiang S, Kameya T, Asamura H, et al. hASH1 expression is closely correlated with endocrine
phenotype and differentiation extent in pulmonary neuroendocrine tumors. Mod Pathol Feb;2004 17
(2):222–9. [PubMed: 14657947]
15. Pinchot SN, Holen K, Sippel RS, Chen H. Carcinoid tumors. Oncologist Dec;2008 13(12):1255–69.
[PubMed: 19091780]
16. Chen H, Hardacre J, Uzar A, Cameron J, Choti M. Isolated liver metastases from neuroendocrine
tumors: does resection prolong survival? J Am Coll Surg Jul;1998 187(1):88–92. discussion -3.
[PubMed: 9660030]
17. Lal A, Chen H. Treatment of advanced carcinoid tumors. Curr Opin Oncol Jan;2006 18(1):9–15.
[PubMed: 16357558]
18. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer Feb 15;2003
97(4):934–59. [PubMed: 12569593]
19. Sippel RS, Chen H. Carcinoid tumors. Surg Oncol Clin N Am Jul;2006 15(3):463–78. [PubMed:
16882492]
20. Van Gompel JJ, Sippel RS, Warner TF, Chen H. Gastrointestinal carcinoid tumors: factors that predict
outcome. World J Surg Apr;2004 28(4):387–92. [PubMed: 14994141]
21. Chen H, Kunnimalaiyaan M, Van Gompel JJ. Medullary thyroid cancer: the functions of raf-1 and
human achaete-scute homologue-1. Thyroid Jun;2005 15(6):511–21. [PubMed: 16029117]
22. Kunnimalaiyaan M, Chen H. The Raf-1 pathway: a molecular target for treatment of select
neuroendocrine tumors? Anticancer Drugs Feb;2006 17(2):139–42. [PubMed: 16428931]
23. Sippel RS, Carpenter JE, Kunnimalaiyaan M, Lagerholm S, Chen H. Raf-1 activation suppresses
neuroendocrine marker and hormone levels in human gastrointestinal carcinoid cells. Am J Physiol
Gastrointest Liver Physiol Aug;2003 285(2):G245–54. [PubMed: 12851216]
Cook et al. Page 9
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
24. Vaccaro A, Chen H, Kunnimalaiyaan M. In-vivo activation of Raf-1 inhibits tumor growth and
development in a xenograft model of human medullary thyroid cancer. Anticancer Drugs Aug;2006
17(7):849–53. [PubMed: 16926634]
25. Van Gompel JJ, Kunnimalaiyaan M, Holen K, Chen H. ZM336372, a Raf-1 activator, suppresses
growth and neuroendocrine hormone levels in carcinoid tumor cells. Mol Cancer Ther Jun;2005 4
(6):910–7. [PubMed: 15956248]
26. Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene May
14;2007 26(22):3279–90. [PubMed: 17496922]
27. Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, et al. Valproic acid activates notch-1 signaling and
regulates the neuroendocrine phenotype in carcinoid cancer cells. Oncologist Aug;2007 12(8):942–
51. [PubMed: 17766653]
28. Stark G, Taylor W. Control of the G2/M transition. Mol Biotechnol Mar;2006 32(3):227–48.
[PubMed: 16632889]
29. Tyagi A, Singh R, Agarwal C, Chan D, Agarwal R. Silibinin strongly synergizes human prostate
carcinoma DU145 cells to doxorubicin-induced growth Inhibition, G2-M arrest, and apoptosis. Clin
Cancer Res Nov;2002 8(11):3512–9. [PubMed: 12429642]
30. Emery P. Leflunomide: a new DMARD for rheumatoid arthritis. Hosp Med May;2000 61(5):344–7.
[PubMed: 10953742]
31. Chen H, Biel MA, Borges MW, et al. Tissue-specific expression of human achaete-scute homologue-1
in neuroendocrine tumors: transcriptional regulation by dual inhibitory regions. Cell Growth Differ
Jun;1997 8(6):677–86. [PubMed: 9186001]
32. Tomassetti P, Migliori M, Simoni P, et al. Diagnostic value of plasma chromogranin A in
neuroendocrine tumours. Eur J Gastroenterol Hepatol Jan;2001 13(1):55–8. [PubMed: 11204811]
33. Sriuranpong V, Borges MW, Ravi RK, et al. Notch signaling induces cell cycle arrest in small cell
lung cancer cells. Cancer Res Apr 1;2001 61(7):3200–5. [PubMed: 11306509]
34. Chen H, Carson-Walter EB, Baylin SB, Nelkin BD, Ball DW. Differentiation of medullary thyroid
cancer by C-Raf-1 silences expression of the neural transcription factor human achaete-scute
homolog-1. Surgery Aug;1996 120(2):168–72. discussion 73. [PubMed: 8751579]
35. Chambard J, Lefloch R, Pouysségur J, Lenormand P. ERK implication in cell cycle regulation.
Biochim Biophys Acta Aug;2007 1773(8):1299–310. [PubMed: 17188374]
36. Nam H, Kim S, Lee M, et al. The ERK-RSK1 activation by growth factors at G2 phase delays cell
cycle progression and reduces mitotic aberrations. Cell Signal Jul;2008 20(7):1349–58. [PubMed:
18450423]
37. Adler, JT.; Hottinger, DG.; Kunnimalaiyaan, M.; Chen, H. Combination Therapy with Histone
Deacetylase Inhibitors and Lithium Chloride: A Novel Treatment for Carcinoid Tumors. 2008/11/26
ed. 2008.
38. Kunnimalaiyaan, M.; Ndiaye, M.; Chen, H. Neuroendocrine tumor cell growth inhibition by
ZM336372 through alterations in multiple signaling pathways. 2007/12/08 ed. 2007. p.
959-64.discussion -64
39. Kunnimalaiyaan, M.; Ndiaye, M.; Chen, H. Apoptosis-mediated medullary thyroid cancer growth
suppression by the PI3K inhibitor LY294002. 2006. p. 1009-14.discussion 14-5
40. Brentjens R, Saltz L. Islet cell tumors of the pancreas: the medical oncologist’s perspective. Surg
Clin North Am Jun;2001 81(3):527–42. [PubMed: 11459269]
41. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature Jun 27;2002
417(6892):949–54. [PubMed: 12068308]
42. Gollob JA, Wilhelm S, Carter C, Kelley SL. Role of Raf kinase in cancer: therapeutic potential of
targeting the Raf/MEK/ERK signal transduction pathway. Semin Oncol Aug;2006 33(4):392–406.
[PubMed: 16890795]
43. Pinchot SN, Adler JT, Luo Y, et al. Tautomycin suppresses growth and neuroendocrine hormone
markers in carcinoid cells through activation of the Raf-1 pathway. Am J Surg Mar;2009 197(3):
313–9. [PubMed: 19245907]
44. Chong, AS.; Huang, W.; Liu, W., et al. In vivo activity of leflunomide: pharmacokinetic analyses
and mechanism of immunosuppression. 1999/07/31 ed. 1999. p. 100-9.
Cook et al. Page 10
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 11
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1.
LFN and TFN inhibit the proliferation of GI carcinoid cells. BON GI carcinoid cells were
treated with the indicated concentrations of either (A) LFN or (B) TFN for up to 6 days. Cell
viability was determined by the MTT colorimetric assay, treatments were significantly
different from control at 6 days (p 0.05). Experiments were performed in quadruplicate and
data is plotted as mean ± SEM.
Cook et al. Page 12
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 13
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2.
TFN induces cell cycle arrest in vitro and suppresses in vivo proliferation of gastrointestinal
carcinoid cells. BON gastrointestinal carcinoid cells were treated with the indicated doses of
TFN and incubated for 48 hours. (A) A dose dependent accumulation of cyclin B1 was observed
by western blot, with GAPDH serving as a loading control. (B) Propidium Iodide exclusion
flow cytometry showed a dose dependent accumulation of S-phase products. Together, these
data suggest that TFN can induce in vitro cell cycle arrest prior to the G2/M transition. (C)
Daily oral gavage with 35mg/kg LFN of nude mice with subcutaneous xenografted NETs
resulted in the progressive inhibition of growth, statistically significant after 24 days (p < 0.02).
This result suggests that the observed in vitro growth suppression can be replicated in vivo.
Cook et al. Page 14
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 15
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 16
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3.
LFN and TFN suppress important markers of NE malignancy. Treatment of BON
gastrointestinal carcinoid cells with either (A) LFN or (B) TFN results in a 61% and 65%
suppression of CgA and a 42% and 84% suppression of ASCL1 protein when the highest dose
is compared to control, respectively. GAPDH is presented as a loading control and
densitometry is performed on samples normalized to their respective GAPDH and subsequently
compared to control values. (C) Treatment with TFN additionally suppresses cellular content
of serotonin after 48 hours of treatment by 77% (p < 0.005). (D) Treatment with 50 and 100
μM TFN resulted in a 54% and 62% reduction in ASCL1 mRNA, respectively (p <0.05).
Cook et al. Page 17
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Results were normalized to the GAPDH expression of the respective sample. Treatment of
NCI-H727 bronchopulmonary carcinoid cells with increasing doses of (E) LFN or (F) TFN
resulted in the 43% and 71% suppression of ASCL1 protein when the highest dose is compared
to control. Taken together, these data suggest that LFN and TFN can suppress key NET markers
implicated in the malignant carcinoid syndrome and correlated to poor prognosis, additionally
this effect may not be limited to a single cell line.
Cook et al. Page 18
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 19
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cook et al. Page 20
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4.
TFN suppresses ASCL1 via the Raf-1/MEK/ERK1/2 pathway. (A) Treatment of BON cells
for 48 hours with TFN resulted in a dose dependent increase in the level of phosphorylated
Raf-1, MEK and ERK 1/2 with no overall change in the level of total ERK. (B) Pretreatment
with the MEK1/2 inhibitor U0126 resulted in a reversal of TFN induced ASCL 1 suppression
at the protein level as well as reversal of the effects on cyclin B1, a marker of G2/M arrest.
GAPDH is presented as a loading control. (C) Similar pretreatment reversed the effects of TFN
on the level of ASCL1 mRNA, as measured by qPCR normalized to the GAPDH for the
respective sample. These data together suggest that TFN activates the Raf-1/MEK/ERK
pathway and that the suppression of ASCL1 protein and mRNA previously observed is
regulated via this pathway. Additionally, the observed cell cycle arrest may be mediated by
the same pathway. (D) No change in the phosphorylation state of Glycogen Synthase
Kinase-3β (GSK-3β) or (E) Akt was observed after carcinoid cell were treated with TFN. This
suggests that the modulation of protein phosphorylation may be specific to the Raf-1/MEK/
ERK pathway.
Cook et al. Page 21
Mol Cancer Ther. Author manuscript; available in PMC 2010 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... The FDA approved it as an immunomodulatory drug for the treatment of patients with rheumatoid arthritis (237). A number of studies reported that LFN inhibits the growth of several different cell types, including human mm, prostate cancer, NETs, breast cancer and neuroblastoma cells (238)(239)(240)(241). Hanson et al (242) showed that LFN exhibits potential therapeutic value in treating melanoma. ...
... The soluble Ephrin-A1/EphA2 system functions regulating angiogenesis in breast cancer, but similar results were found in a bladder carcinogenesis model via inhibition of the soluble Ephrin-A1/EphA2 system; Ephrin-A1 overexpression could partially reverse LFN-induced suppression of angiogenesis and subsequent tumor growth inhibition (244). Cook et al (240) showed that LFN and its natural metabolites suppress Achaete-scute homolog 1, both at the protein and mRNA level, via a mechanism that is predominately dependent upon the Raf-1/MEK/ERK1/2 pathway. Other mechanisms of action have also been considered, as described by Zhang and Chu (245). ...
Article
Full-text available
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
... The FDA approved it as an immunomodulatory drug for the treatment of patients with rheumatoid arthritis (237). A number of studies reported that LFN inhibits the growth of several different cell types, including human MM, prostate cancer, NETs, breast cancer and neuroblastoma cells (238)(239)(240)(241). Hanson et al (242) showed that LFN exhibits potential therapeutic value in treating melanoma. ...
... The soluble Ephrin-A1/EphA2 system functions regulating angiogenesis in breast cancer, but similar results were found in a bladder carcinogenesis model via inhibition of the soluble Ephrin-A1/EphA2 system; Ephrin-A1 overexpression could partially reverse LFN-induced suppression of angiogenesis and subsequent tumor growth inhibition (244). Cook et al (240) showed that LFN and its natural metabolites suppress Achaete-scute homolog 1, both at the protein and mRNA level, via a mechanism that is predominately dependent upon the Raf-1/MEK/ERK1/2 pathway. Other mechanisms of action have also been considered, as described by Zhang and Chu (245). ...
Article
Full-text available
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
... NF type 1 is classified as a RasOpathy, as Ras-MAPK signaling is affected by mutations the NF1 gene, encoding neurofibromin a RAS GTPase-activating protein [121]. [56,122] and MEK inhibitors [57,73] but instead also Raf-1 activators [107,[123][124][125] might be of interest as potential target as has been extensively reviewed by Fazio et al. [37,126]. In BON1 cells, stable transfection with an estrogen-inducible raf-1 construct (BON1-raf cells) caused an induction of MEK and ERK 1/2 signaling [127], induction of focal adhesion kinase (FAK) by a MEK/ERK1/2-dependent pathway [128] and decreased CgA expression by a MEK/ERK½/FAK-dependent pathway [127,128] and decreased cell adhesion and downregulation of beta-catenin by a MEK-dependent pathway [128,129]. ...
... However, ZM336372 in BON1 cells also inactivates GSK3 at a Raf-MEK-independent pathway [107] and ZM336372-induced reduction of CgA and ASCL1 expression has been shown to be mediated by a Raf-MEK-independent pathway [107], thus the observed Raf-1 induction might not be essential for the antitumoural effects of ZM336372. The Raf-1 activator teriflunomide has also been reported to show antiproliferative effects in BON1 cells alone [124] and in combination with pasireotide [125]. ...
Article
Full-text available
Molecular targeted therapy of advanced neuroendocrine tumours (NETs) of the gastroenteropancreatic (GEP) system currently encompasses approved therapy with the mTOR-inhibitor everolimus and the multi-tyrosinkinase inhibitor sunitinib. However clinical efficacy of these treatment strategies is limited by low objective response rates and limited progression free survival due to tumor resistance. Further novel strategies for molecular targeted therapy of NETs of the GEP-system are needed. This paper reviews preclinical research models and signaling pathways in NETs of the GEP-system. Preclinical and early clinical data on putative novel targets for molecular targeted therapy of NETs of the GEP-system are discussed, including PI3K, Akt, mTORC1/mTORC2, GSK3, c-Met, Ras-Raf-MEK-ERK, embryogenic pathways (Hedgehog, Notch, Wnt/beta-catenin, TGF-β Signaling and SMAD proteins), tumor suppressors and cell cycle regulators (p53, cyclin dependent kinases (CDK) CDK4/6, CDK inhibitor p27, retinoblastoma protein (Rb) ), heat shock protein HSP90, Aurora kinase, Src kinase family, focal adhesion kinase (FAK) and epigenetic modulation by histone deacetylase inhibitors (HDAC).
... A dose dependent cytotoxic effect of the tyrosine kinase inhibitor imatinib on these cells was reported [34]. Moreover, the immunosuppressor leflunomide and its metabolite teriflunomide have been demonstrated to be able to inhibit BON-1 cells proliferation and to induce G2/M phase arrest [26]. Lithium treatment caused a dose-dependent reduction in BON-1 growth [28] as well as sodium butyrate and hexamethylene bisacetamide [30] and all-trans-retinoic acid [25]. ...
Article
Full-text available
Carcinoid syndrome represents a debilitating paraneoplastic disease, caused by the secretion of several substances, occurring in about 10–40% of patients with well-differentiated neuroendocrine tumors (NETs). The main signs and symptoms associated with carcinoid syndrome are flushing, diarrhea, hypotension, tachycardia, bronchoconstriction, venous telangiectasia, dyspnea and fibrotic complications (mesenteric and retroperitoneal fibrosis, and carcinoid heart disease). Although there are several drugs available for the treatment of carcinoid syndrome, the lack of therapeutic response, poor tolerance or resistance to drugs are often reported. Preclinical models are indispensable tools for investigating the pathogenesis, mechanisms for tumor progression and new therapeutic approaches for cancer. This paper provides a state-of-the-art overview of in vitro and in vivo models in NETs with carcinoid syndrome, highlighting the future developments and therapeutic approaches in this field.
... However, its important role in cancer biology is thought to relate to malignant cells having a lower threshold for pyrimidine deprivation compared to non-malignant cells (129). Examples of anti-tumor mechanisms of teriflunomide and leflunomide include: down-regulation of anti-apoptotic proteins and growth factor receptors in cancer cells (62,128), interruption of cancer cell survival signaling (130,131), induction of cancer cell death (128,132), abolishment of cancer stem cells (133), and cancer cell mitochondrial disruption (132). Teriflunomide may be an attractive anti-tumor medicine due to its efficacy at lower doses compared to other medications that inhibit DNA synthesis, such as methotrexate, and may thus avoid cumulative cytotoxic damage to the body (129). ...
Article
Full-text available
Over the past two decades, the field of multiple sclerosis (MS) has been transformed by the rapidly expanding arsenal of new disease modifying therapies (DMTs). Current DMTs for MS aim to modulate innate and adaptive immune responses toward a less inflammatory phenotype. Since the immune system is also critical for identifying and eliminating malignant cells, immunosuppression from DMTs may predictably increase the risk of cancer development in MS patients. Compared with healthy controls, patients with autoimmune conditions, such as MS, may already have a higher risk of developing certain malignancies and this risk may further be magnified by DMT treatments. For those patients who develop both MS and cancer, these comorbid presentations create a challenge for clinicians on how to therapeutically address management of cancer in the context of MS autoimmunity. As there are currently no accepted guidelines for managing MS patients with prior history of or newly developed malignancy, we undertook this review to evaluate the molecular mechanisms of current DMTs and their potential for instigating and treating cancer in patients living with MS.
... Despite being marketed primarily for autoimmune disorders, leflunomide also displays anticancer effects. Several studies report anti-proliferative effects in various cell lines, including those derived from multiple myeloma (Baumann et al., 2009), non-small cell lung carcinomas (Jiang, Zhang, Li, Ling, & Jiang, 2017), neuroblastomas (Zhu, Yan, Xiang, Ding, & Cui, 2013), neuroendocrine (Cook et al., 2010), and cancer in medullary thyroids (Alhefdhi, Burke, Redlich, Kunnimalaiyaan, & Chen, 2013). Leflunomide is currently undergoing clinical trials for the treatment of multiple myeloma (NCT02509052). ...
Article
Identified as a hallmark of cancer, metabolic reprogramming allows cancer cells to rapidly proliferate, resist chemotherapies, invade, metastasize, and survive a nutrient-deprived microenvironment. Rapidly growing cells depend on sufficient concentrations of nucleotides to sustain proliferation. One enzyme essential for the de novo biosynthesis of pyrimidine-based nucleotides is dihydroorotate dehydrogenase (DHODH), a known therapeutic target for multiple diseases. Brequinar, leflunomide, and teriflunomide, all of which are potent DHODH inhibitors, have been clinically evaluated but failed to receive FDA approval for the treatment of cancer. Inhibition of DHODH depletes intracellular pyrimidine nucleotide pools and results in cell cycle arrest in S-phase, sensitization to current chemotherapies, and differentiation in neural crest cells and acute myeloid leukemia (AML). Furthermore, DHODH is a synthetic lethal susceptibility in several oncogenic backgrounds. Therefore, DHODH-targeted therapy has potential value as part of a combination therapy for the treatment of cancer. In this review, we focus on the de novo pyrimidine biosynthesis pathway as a target for cancer therapy, and in particular, DHODH. In the first part, we provide a comprehensive overview of this pathway and its regulation in cancer. We further describe the relevance of DHODH as a target for cancer therapy using bioinformatic analyses. We then explore the preclinical and clinical results of pharmacological strategies to target the de novo pyrimidine biosynthesis pathway, with an emphasis on DHODH. Finally, we discuss potential strategies to harness DHODH as a target for the treatment of cancer.
Article
Somatostatin analogs have an important role in the medical therapy of neuroendocrine tumors (NETs). Octreotide and lanreotide, both somatostatin analogs binding with high affinity for the somatostatin receptor (SSTR)2, can control symptoms in functional NETs. In addition, these compounds, because of their antiproliferative effects, can stabilize growth of well-differentiated NETs. Pasireotide is a novel multireceptor-targeted somatostatin analog with high affinity for SSTR1, 2, 3, and 5. This review provides an overview of the state of the art of pasireotide in the treatment of NETs, with the aim of addressing clinical relevance and future perspectives for this molecule in the management of NETs.
Article
Tropomyosin receptor kinase (Trk) inhibitors are investigated as a novel targeted therapy in various cancers. We investigated the in vitro effects of the pan-Trk inhibitor GNF-5837 in human neuroendocrine tumor (NET) cells. The human neuroendocrine pancreatic BON1, bronchopulmonary NCI-H727 and ileal GOT1 cell lines were treated with GNF-5837 alone and in combination with everolimus. Cell viability decreased in a time- and dose-dependent manner in GOT1 cells in response to GNF-5837 treatment, while treatment in BON1 and NCI-H727 cells showed no effect on cellular viability. Trk receptor expression determined GNF-5837 sensitivity. GNF-5837 caused downregulation of PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling, the cell cycle and increased apoptotic cell death. The combinational treatment of GNF-5837 with everolimus showed a significant enhancement in inhibition of cell viability versus single substance treatments, due to a cooperative PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation, as well as an enhanced cell cycle component downregulation. Immunohistochemical stainings for Trk receptors were performed using a tissue microarray containing 107 tumor samples of gastroenteropancreatic NETs. Immunohistochemical staining with TrkA receptor and pan-Trk receptor antibodies revealed a positive staining in pancreatic NETs in 24.2 % (8 / 33) and 33.3 % (11 / 33), respectively. We demonstrated that the pan-Trk inhibitor GNF-5837 has promising anti-tumoral properties in human NET cell lines expressing the TrkA receptor. Immunohistochemical or molecular screening for Trk expression particularly in pancreatic NETs might serve as predictive marker for molecular targeted therapy with Trk inhibitors.
Article
Leflunomide, an inhibitor of dihydroorotase dehydrogenase and thereby pyrimidine synthesis, was approved for treatment of rheumatoid arthritis in 1998. During the following years, leflunomide was used in various preclinical studies as a potential cancer treatment; at the same time, more mechanisms underlying the anticancer effect of leflunomide were identified. Thus, leflunomide has been identified as a potent anticancer drug. This article summarizes the mechanisms as well as results of leflunomide in the evolving field of cancer therapy.
Article
Full-text available
BACKGROUND Carcinoid tumors represent an unusual and complex disease spectrum with protean clinical manifestations. This compilation of several large United States-based databases comprising patients from 1950 to 1999 examines 13,715 carcinoid tumors and provides epidemiologic information regarding the natural history and evolution of the detection and diagnosis of this entity.METHODS The authors evaluated 10,878 carcinoid tumors that were identified by the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute (NCI) from 1973 to 1999 in addition to 2837 carcinoid tumors that were registered previously by two earlier NCI programs. To the authors' knowledge, this represents the largest current epidemiology series addressing carcinoid tumors to date.RESULTSSpecific trends in incidence for carcinoid tumors of certain sites were identified. Among the most recently collected subset of data, sites that demonstrated the greatest incidence of carcinoids were the gastrointestinal tract (67.5%) and the bronchopulmonary system (25.3%). Within the gastrointestinal tract, most carcinoid tumors occurred in the small intestine (41.8%), rectum (27.4%), and stomach (8.7%). For all sites, age-adjusted incidence rates were highest in black males (4.48 per 100,000 population per year). Associated noncarcinoid tumors were frequent in conjunction with small intestinal (29.0%), gastric (20.5%), colonic (20.0%), and appendiceal (18.2%) carcinoids. The highest percentages of nonlocalized lesions were noted for cecal (81.5–83.2%) and pancreatic (71.9–81.3%) carcinoids, whereas the highest percentage of localized disease was found among rectal (81.7%), gastric (67.5%), and bronchopulmonary (65.4%) carcinoids. The best 5-year survival rates were recorded for patients with rectal (88.3%), bronchopulmonary (73.5%), and appendiceal (71.0%) carcinoids; these tumors exhibited invasive growth or metastatic spread in 3.9%, 27.5%, and 38.8% of patients, respectively.CONCLUSIONS Carcinoids appear to have increased in overall incidence over the past 30 years; for some sites, this trend has been evident for nearly half a century. Recent marked increases in gastric and rectal carcinoids and a concomitant decrease in appendiceal carcinoid incidence may be due in part to varying rules of registration among the compiled databases examined in this report or to improvements in diagnostic technology; increased awareness of and about carcinoid tumors also may play a significant role. In 12.9% of all patients with carcinoid, distant metastases already were evident at the time of diagnosis; the overall 5-year survival rate for all carcinoid tumors, regardless of site, was 67.2%. These findings bring into question the widely promulgated relative benignity of carcinoid disease. Certain carcinoid tumors, such as those of the rectum, appear to be over-represented among the black and Asian populations within the United States, suggesting the role of genetics in the development of this intriguing disease. Cancer 2003;97:934–59. © 2003 American Cancer Society.DOI 10.1002/cncr.11105
Article
Full-text available
Carcinoid tumors are rare, slow-growing neuroendocrine tumors arising from the enterochromaffin cells disseminated throughout the gastrointestinal and bronchopulmonary systems. Though they have been traditionally classified based on embryologic site of origin, morphologic pattern, and silver affinity, newer classification systems have been developed to emphasize the considerable clinical and histopathologic variability of carcinoid tumors found within each embryologic site of origin. These neoplasms pose a diagnostic challenge because they are often innocuous at the time of presentation, emphasizing the need for a multidisciplinary diagnostic approach using biochemical analysis, standard cross-sectional imaging, and newer advances in nuclear medicine. Similarly, treatment of both primary and disseminated carcinoid disease reflects the need for a multidisciplinary approach, with surgery remaining the only curative modality. The prognosis for patients with these tumors is generally favorable; however, it can be quite variable and is related to the location of the primary tumor, extent of metastatic disease at initial presentation, and time of diagnosis.
Article
Full-text available
In carcinoid cell lines, the histone deacetylase (HDAC) inhibitors valproic acid (VPA) and suberoyl bis-hydroxamic acid (SBHA) activate the Notch1 pathway, whereas lithium inhibits glycogen synthase kinase-3beta (GSK-3beta). These compounds limit growth and decrease hormonal secretion in vitro. We hypothesized that lower-dose combination therapy of HDAC inhibitors and lithium chloride could achieve similar growth inhibition to that of the drugs alone. Gastrointestinal and pulmonary carcinoid cells were treated with either VPA or SBHA and lithium chloride for up to 48 hours. Western blot analysis was used to measure the effects on the Notch1 and GSK-3beta pathways and the neuroendocrine tumor marker chromogranin A (CgA). Growth was measured by a cellular proliferation assay. With lower-dose combination therapy, a decrease in CgA was observed. The HDAC inhibitors increased the amount of active Notch1 protein, whereas treatment with lithium was associated with inhibition of GSK-3beta. Moreover, growth was inhibited with lower-dose combination therapy. Treatment of carcinoid cells with either VPA or SBHA and lithium chloride suppresses the neuroendocrine marker CgA while upregulating Notch1 and inhibiting GSK-3beta. This combination effectively reduces growth. Thus, lower-dose combination therapy may be a viable therapeutic approach for carcinoid tumors.
Article
Previous studies have demonstrated that the active metabolite of leflunomide, A77 1726 [N-(4-trifluoromethylphenyl-2-cyano-3-hydroxycrotoamide)], is capable of inhibiting the activities of tyrosine kinases and dihydroorotate dehydrogenase (DHO-DHase). In the present study, we define the relative contribution of these activities to the ability of A77 1726 to inhibit proliferation of the murine leukemia cell line LSTRA. A77 1726 inhibited LSTRA cell growth and proliferation (ic50 ≈ 10–30 μM); this inhibition, however, could be reversed by the addition of exogenous uridine, suggesting that the anti-proliferative activity of A77 1726 may be due to inhibition of de novo pyrimidine nucleotide synthesis. Quantitation of nucleotide levels revealed that A77 1726, at an ic50 of about 10 μM, selectively inhibited pyrimidine nucleotide but not purine nucleotide synthesis. In vitro enzyme assays confirmed that A77 1726 directly inhibited the activity of DHO-DHase, the fourth enzyme in the de novo pathway of pyrimidine nucleotide synthesis (ic50 ≈ 220 nM). LSTRA cells overexpress p56lck and have elevated levels of tyrosine phosphorylated intracellular proteins. A77 1726 reduced the intracellular levels of tyrosine phosphorylated proteins with relatively high ic50 values ranging from 50 to 100 μM. A77 1726 also inhibited p56lck activity in LSTRA membrane preparation and immunoprecipitates; the ic50 values for inhibition of immunoprecipitated p56lck autophosphorylation and exogenous substrate histone 2B were 80 and 40 μM, respectively. The anti-tyrosine phosphorylation activity of A77 1726 was not affected by uridine. These studies therefore demonstrate the two activities of A77 1726: inhibition of pyrimidine nucleotide synthesis and interference with tyrosine phosphorylation.
Article
Objective . To assess the safety and effectiveness of leflunomide versus placebo in patients with active rheumatoid arthritis (RA) treated for 6 months. Methods . Four hundred two patients were randomly assigned to receive placebo or leflunomide at 5 mg, 10 mg, or 25 mg daily. A washout period of 6–12 weeks from prior second‐line therapy was required. Results . Statistically significant improvement in primary and secondary outcome measures, as well as by responder analyses, occurred in the 10‐mg and 25‐mg dosage groups compared to placebo. Twenty‐one patients (7.0%) in the active treatment groups withdrew due to adverse events (AEs). The incidence of AEs was higher with leflunomide than with placebo. Gastrointestinal symptoms, weight loss, allergic reactions, skin rash, and reversible alopecia were more common in the 10‐mg and 25‐mg dosage groups. The incidence of infections was similar between the treatment and placebo groupsno opportunistic infections were seen. Transient elevations in liver function studies were noted in a small number of patients. Conclusion . Leflunomide is effective in daily doses of 10 mg and 25‐mg in patients with active RA. Improved efficacy at the 25‐mg dose was associated with a higher incidence of AEs. Randomized, placebocontrolled trials using daily doses of 10 mg and 20 mg are under way in the US and Europe to confirm these positive results.
Article
Background. The inhibition of tumorangiogenesis may be of importance in the additive treatment of various cancers. Leflunomide, a drug which has been approved in Germany for the therapy of rheumatoid arthritis, inhibits the activity of several growth factors in vitro. The aim of this study was to investigate the effects of the drug on tumor angiogenesis in a nude mouse model. Materials and methods. A total of 40 nude mice were injected with human colon carcinoma cells. Following randomization in 4 groups, therapy started on day five. Group 1 was treated daily with orally administered Leflunomide (35 mg/kg) dissolved in 1.5% Carboxymethylcellulose (CMC). Group 2 served as a control group and received 1 ml CMC orally per day. The animals of group 3 were treated daily with 35 mg Leflunomide/kg KG and 500 mg Uridine/kg dissolved in 1 ml Nacl 0.9% intraperitoneally. The 4th group again served as a control group and received only 500 mg Uridine/kg intraperitoneally each day. The main outcome criterion was the angiogenesis score (AS). In addition, the tumor volume and tumor weight were also assessed. The AS was determined by immunohistochemistry using an antibody against factor VIII related antigen. Results. All animals tolerated the procedure well. In the Leflunomide and the Leflunomide/Uridine group the angiogenesis score (p<0,01), the tumor volume (p<0,01) and the tumor weight (p<0,01) were lower compared to the respective control groups. Conclusion. The administration of Leflunomide leads to a significant reduction of tumor weight and tumor volume following subcutaneous injection of human colon carcinoma cells in a nude mouse model. This could be due to the reduction of tumor angiogenesis. Following further experimental and clinical studies, Leflunomide may come to play a role in the additive treatment of colonic carcinoma.
Article
Carcinoids are neuroendocrine (NE) tumors with limited treatment options. Raf-1 pathway activation has been shown to suppress hormone production in carcinoid cells. We investigated a novel treatment for carcinoid cell growth based on pharmacologic Raf-1 activation using the compound tautomycin (TTY). Human carcinoid cells were treated with TTY for 48 hours. Western blot analysis was used to demonstrate Raf-1 pathway activation by phosphorylation of ERK1/2 and to determine the effect on NE tumor markers. Cellular growth was measured by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. Treatment with TTY resulted in dose-dependent activation of the Raf-1 pathway. Furthermore, a significant decrease in NE tumor markers was seen. Importantly, TTY inhibited carcinoid cellular growth and induced the cell-cycle inhibitors p21 and p27. TTY activates the Raf-1 pathway, limits carcinoid cell growth, and suppresses NE marker production in vitro. This new compound warrants further investigation in animal models of carcinoid cancer.
Article
Multiple myeloma is still an incurable disease; therefore, new therapeutics are urgently needed. A771726 is the active metabolite of the immunosuppressive drug leflunomide, which is currently applied in the treatment of rheumatoid arthritis, BK virus nephropathy, and cytomegaly viremia. Here, we show that dihydroorotate dehydrogenase (DHODH) is commonly expressed in multiple myeloma cell lines and primary multiple myeloma cells. The DHODH inhibitor A771726 inhibits cell growth in common myeloma cell lines at clinically achievable concentrations in a time- and dose-dependent manner. Annexin V-FITC/propidium iodide staining revealed induction of apoptosis of multiple myeloma cell lines and primary multiple myeloma cells. The 5-bromo-2'-deoxyuridine cell proliferation assay showed that inhibition of cell growth was partly due to inhibition of multiple myeloma cell proliferation. A771726 induced G(1) cell cycle arrest via modulation of cyclin D2 and pRb expression. A771726 decreased phosphorylation of protein kinase B (Akt), p70S6K, and eukaryotic translation initiation factor 4E-binding protein-1 as shown by Western blotting experiments. Furthermore, we show that the stimulatory effect of conditioned medium of HS-5 bone marrow stromal cells on multiple myeloma cell growth is completely abrogated by A771726. In addition, synergism studies revealed synergistic and additive activity of A771726 together with the genotoxic agents melphalan, treosulfan, and doxorubicin as well as with dexamethasone and bortezomib. Taken together, we show that inhibition of DHODH by A771726/leflunomide is effective in multiple myeloma. Considering the favorable toxicity profile and the great clinical experience with leflunomide in rheumatoid arthritis, this drug represents a potential new candidate for targeted therapy in multiple myeloma.
Article
Currently, licensed disease-modifying agents for multiple sclerosis (MS) all share the need for parenteral administration. Oral therapies would carry the advantage of convenience and greater acceptability. Teriflunomide is one of several oral agents currently undergoing Phase III investigation. This article describes the mode of action of teriflunomide which largely depends on inhibition of pyrimidine synthesis. We review the evidence so far on the efficacy of teriflunomide in animal models and Phase II human studies. In view of teriflunomides favourable safety profile it appears to be a promising oral alternative to interferon beta and glatiramer acetate. The ongoing Phase III investigations of teriflunomide as mono- and combination therapy are discussed.