Download Deep Insight Section Inflammatory programming and immune modulation in cancer by IDO

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Atlas of Genetics and Cytogenetics
in Oncology and Haematology
INIST-CNRS
OPEN ACCESS JOURNAL
Deep Insight Section
Inflammatory programming and immune
modulation in cancer by IDO
Courtney Smith, George C Prendergast
Lankenau Institute for Medical Research (LIMR), Wynnewood PA USA (CS), Department of Pathology,
Anatomy and Cell Biology, Jefferson Medical School and Kimmel Cancer Center, Thomas Jefferson
University, Philadelphia PA USA (GCP)
Published in Atlas Database: June 2013
Online updated version : http://AtlasGeneticsOncology.org/Deep/IDOandCancerID20121.html
DOI: 10.4267/2042/51878
This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence.
© 2013 Atlas of Genetics and Cytogenetics in Oncology and Haematology
Abstract
Immune dysregulation is one of the hallmarks of tumor growth and progression, a key event that allows for tumor
evasion of the host immune system. More recent cancer modalities are embracing combinations incorporating
immunotherapy with more traditional chemotherapy and radiotherapy. Traditional approaches are difficult to tolerate for
the patient and become less effective as tumors evolve to survive these treatments. Immunotherapy has the benefit of
reduced toxicity as it utilizes the patient's own immune system to identify and eliminate tumor cells. One mechanism
manipulated by tumors is upregulation of the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO). In this
review, we focus on the mechanism by which tumors use IDO to evade detection by T cell immunity, as well as on
novel small molecules that inhibit it as a cancer therapeutic strategy.
IDO1 is found in various cells including immune cells,
endothelial cells, fibroblast and some tumor cells
(Serafini, et al., 2006; Friberg et al., 2002; Uyttenhove
et al., 2003; Munn et al., 2004).
IDO1 is relatively well conserved between species
suggesting evolutionary importance. The primary
sequence of IDO1 is 63% identical between mouse and
human. The crystal structure of IDO1 and the resulting
site-directed mutagenesis show that both substrate
binding and the precise relationship between the
substrate and iron-bound dioxygen are necessary for
activity (Sugimoto et al., 2006). More recently, a
homolog to IDO1, termed IDO2, has been identified.
Initially, a missannotation in the human genome
database prevented the identification of IDO2. The
correction of this error revealed the 420 amino acid
open-reading frame that shares 44% sequence
homology with IDO1 (Ball et al., 2007; Metz et al.,
2007). Importantly, IDO2 contains the conserved
residues that are identified as critical for tryptophan
binding and catabolism (Sugimoto et al., 2006).
Between mouse and human, IDO2 is 73% identical at
the primary sequence. Due to the recent discovery of
IDO2, there is still much to learn about this enzyme.
While both human and murine IDO2 enzyme have been
Diversity of IDO-related
immunoregulatory enzymes
Identification of the non-hepatic tryptophan
catabolizing enzyme, indoleamine 2,3-deoxygenase
(IDO; EC 1.13.11.42; originally D-tryptophan
pyrrolase) was first reported in 1963 (Higuchi and
Hayaishi, 1967; Higuchi et al., 1963). IDO, also known
as IDO1, catalyzes the first and rate-limiting step that
converts tryptophan to N-formyl-kynurenine, a process
that utilizes oxidative cleavage of the 2,3 double bond
in the indole ring resulting in the biosynthesis of
nicotinamide adenine dinucleotide (NAD) (Takikawa,
2005). The catabolism of tryptophan can also occur
through the enzyme tryptophan 2,3-dioxygenase
(TDO2), though studies have shown that the enzymes
are not redundant. While both IDO1 and TDO2
catalyze this same reaction, beyond that the two
enzymes are structurally distinct and do not share any
significant sequence similarity. Structurally, IDO1
exists as a 41kD monomeric enzyme whereas TDO2 is
a 320kD homotetramer (Watanabe et al., 1981).
Furthermore, TDO2 and IDO1 are differentially
localized. Unlike TDO2, IDO1 is not involved in the
dietary homeostasis of tryptophan degradation. Instead
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
856
Inflammatory programming and immune modulation in cancer by IDO
regulates immunity at the level of T cells but is
regulated by or and regulated by cytokine production in
the host that is associated with the generation of a protumorigenic microenvironment. A role for IDO1 in
cancer is further suggested by the fact that many human
tumor cells themselves express IDO1 (Uyttenhove et
al., 2003; Taylor and Feng, 1991).
shown to catabolize tryptophan to kynurenine, IDO2
has a distinct pattern of expression that differs from
both IDO1 and TDO2 (Metz et al., 2012). Using total
RNAs from human tissues, full-length IDO2 was found
expressed in the placenta and brain. Interestingly,
primers located in exon 10 showed IDO2 mRNA in a
greater number of tissues including liver, small
intestine, spleen, placental, thymus, lung, brain, kidney
and colon supporting the possible existence of
additional splice isoforms and perhaps transcriptional
start or polyadenylation sites. To further complicate its
study, transcripts of IDO2 in murine tissues are
localized to liver and kidney which differs from human
tissues somewhat. Interestingly, IDO2 mRNA is
expressed in murine pre-dendritic cells and following
stimulation with IFNγ, IL-10 or lipopolysaccharide
(LPS), IDO2 protein can be detected (Metz et al.,
2012), suggesting in these specialized antigenpresenting cells of the immune system.
IDO in cancer
Treatment of cancer commonly entails surgical
resection followed by chemotherapy and radiotherapy.
The standard regimens show highly variable degrees of
success in the longer term, because of the ability of
tumor cells to escape these treatments to regenerate
primary tumor growth and more importantly seed
distant metastasis. The production of IDO in the tumor
microenvironment appears to aid in tumor growth and
metastasis. It is logical then to target IDO as a means of
slowing tumor progression. This has been the premise
of several recent studies.
Studies have revealed a pathophysiological link
between IDO1 and cancer, with increased levels of
IDO1 activity associated with a variety of different
tumors (Brandacher et al., 2006; Okamoto et al., 2005).
In a case study of ovarian cancer, overexpression of
IDO
correlated
with
poorer
survival.
Immunohistochemical staining on tumor sections were
categorized as negative or positive, with the latter
further defined as sporadic, focal or diffusely staining.
While patients with no IDO expression had greater than
5-year survival following surgery, the three
subcategories of positive IDO staining showed a 50%
survival of patients to 41, 17 and 11 months, inversely
correlated with the amount of staining (Okamoto et al.,
2005). Two mechanisms by which IDO suppresses the
local immune system are inhibition of effector T cells
or activation of Tregs (Fallarino et al., 2006; Munn and
Mellor, 2007). In the colorectal study, high IDO
expression was associated with few tumor infiltrating
CD3+ T cells. In addition to affecting the local
environment, tumor biopsies with high expression of
IDO in both colorectal and hepatocellular carcinomas
have shown greater metastasis in patients (Brandacher
et al., 2006; Pan et al., 2008). While these studies
showed IDO expressed by the tumor itself, other
clinical studies have found both stromal cells and
surrounding immune cells to be the source of IDO
overexpression. Poor survival correlated with IDOpositive eosinophils in small cell lung cancer
(Astigiano et al., 2005) while a study of melanoma
patients showed poor prognosis in patients with
detectable IDO in the dendritic cells (DC) from the
tumor draining lymph nodes (Lee et al., 2003; Munn et
al., 2004).
These clinical cancer studies are supported and
enhanced by studies in the mouse model. As shown in
the clinical studies, tumors may induce IDO1
production in neighboring cells such as antigen
presenting dendritic cells located in the tumor-draining
IDO in the immune system
The first evidence for a role of IDO in immune
regulation was observed when IDO expression was
induced following viral infection or treatment with
interferon (IFNγ), an important inflammatory cytokine
(Yoshida et al., 1979; Yoshida et al., 1981). A prior
observation that IDO is present in the urine of cancer
patients then re-surfaced and its importance reevaluated (Rose, 1967). A groundbreaking study from
Munn and Mellor in 1999 directly established IDO as
an important immune regulator (Munn et al., 1998).
This study showed that pregnant female mice treated
with 1-methyl-tryptophan (1MT), an IDO inhibitor,
caused rejection of allogeneic concepti but not
syngeneic concepti. Further studies showed that this
occurs through an MHC-restricted T cell-mediated
rejection of the allogeneic mouse concepti (Mellor et
al., 2001). These findings have been widely interpreted
to mean that the normally high levels of IDO in the
placenta are important in preventing the maternal
immune system from attacking the "foreign" fetus.
Once IDO was established as an immune modulator,
studies have focused on its role both in disease states as
well as in normal immune surveillance. The
immunosuppressive function of IDO1 manifests in
several manners. Collectively, IDO1 and its metabolites
can directly suppress T cells (Fallarino et al., 2002;
Frumento et al., 2002; Terness et al., 2002; Weber et
al., 2006) and NK cells (Della Chiesa et al., 2006) as
well as enhance local Tregs (Fallarino et al., 2003). The
protumorigenic capabilities of myeloid derived
suppressor cells (MDSCs) (Smith et al., 2012) suggest
that this population is also affected by IDO1.
Furthermore, IDO1 is produced in response to IFN-γ in
endothelial cells, fibroblasts and the immune cells
including dendritic cells and myeloid derived cells
(Taylor and Feng, 1991; Burke et al., 1995; Varga et
al., 1996; Munn et al., 1999; Hwu et al., 2000). It has
therefore been hypothesized that IDO1 not only
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Smith C, Prendergast GC
857
Inflammatory programming and immune modulation in cancer by IDO
mimicking the effects of Bin1 attenuation in human
tumor cells. Notably, the beneficial effects of Bin1
deletion to tumor growth were lost in immune deficient
or T-cell depleted mice, revealing the importance of the
immune system in mediating the primary effects of
Bin1 on tumor growth. Studies in Bin1-deficient cells
established that IDO expression was upregulated and
that the inhibitor 1MT could phenocopy the effect of
Bin1 competency. This work also indicated that Bin1
limits IDO transcription by limiting the activity of NFκB and STAT which are sufficient to support IDO
expression (Muller et al., 2004; Bild et al., 2002).
GCN2 signaling is another mechanism by which IDO
may regulate immune cell function. GCN2 is a kinase
that acts as a sensor for amino acid starvation. The
depletion of tryptophan from the microenvironment can
trip GCN2 signaling resulting in apoptosis, cell cycle
arrest and differentiation. GCN2 rapidly responds to
amino acid deprivation through the phosphorylation of
eIF-2α resulting in inhibition of translation (Bild et al.,
2006). GCN2 is activated by IDO as a result of the
tryptophan deprivation it creates. GCN2 is also a
signaling component of T cells which may be a
mechanism by which IDO regulates the immune
system and produces biological effects. Notably,
GCN2-deficient T cells are resistant to the immune
suppressive effects of IDO (Munn and Mellor, 2007;
Munn et al., 2005). GCN2 signaling switches on the
expression of stress-activated proteins that trigger
growth arrest and apoptosis as well as differentiation.
ATF4, ATF3 and CHOP/GADD153 (Harding et al.,
2000; Jiang et al., 2004; Vattem and Wek, 2004; Lu et
al., 2004; Hai et al., 1999; Wang et al., 1996; Fan et
al.,2002) have been implicated as three critical targets
of GCN2 in responding to amino acid deprivation.
Cytokines are critical for immune recognition of tumor
cells, but when they are hijacked by the tumor they
may provide a mechanism of immune escape for both
the primary tumor and distant metastases. This was
seen in Ido1-nullizygous mice that exhibited both
reduced lung tumor burden in the oncogenic KRASinduced model as well as in the metastatic 4T1
orthotopic breast cancer model. Both models showed
reduction of lung tumor burden that was directly
correlated with improved survival of Ido1-/- mice
(Smith et al., 2012). Further investigation into the
immune regulatory role revealed a reduction in the
levels of the inflammatory cytokine IL-6 in IDOdeficient mice. However, when IL-6 was restored in
these mice, the rate of metastasis was also restored to
levels of wild-type mice. Ex vivo studies of myeloid
derived suppressor cells (MDSC) from IDO-deficient
mice with 4T1 primary tumors showed an impairment
in the ability of MDSC to suppress T cell function,
compared to MDSC derived from 4T1 tumor-bearing
wild-type mice. The attenuation of IL-6 levels in IDOdeficient mice was associated with an impairment in
MDSC function, and as before restoring IL-6 overcame
the MDSC defect, allowed metastatic disease to
lymph nodes (TDLNs). 4T1 is a highly malignant
breast carcinoma-derived cell line that, following
orthotopic engraftment into the murine mammary
fatpad, forms tumors of the latter variety in which no
IDO1 expression is detectable in the primary tumor but
is found expressed at high levels in the TDLNs. The
IDO1-expressing cells in the TDLNs appear
morphologically to be plasmacytoid dendritic cells. A
similar pattern of IDO1 expression has previously been
observed in a mouse model of melanoma (Munn et al.,
2004). Importantly D-1MT treatment of 4T1-tumor
bearing mice cooperated with chemotherapy to
suppress primary tumor growth, ascribing an
immunosuppressive role of IDO1 in the TDLNs (Hou
et al., 2007).
The use of an immunogenic tumor cell line transfected
to overexpress IDO demonstrated that IDO prevents
immune surveillance from rejecting these tumors in
preimmunized mice. There was also a reduction in
tumor associated T cells. Furthermore the use of 1MT
resulted in a slowed progression of the tumor, further
implicating IDO as a tumor evasion mechanism
(Uyttenhove et al., 2003). In the MMTV-neu mouse
model of breast cancer, the synergistic benefit of
combining chemotherapy with the indoleamine-2,3dioxygenase (IDO1) inhibitor 1-methyl-D-tryptophan
was observed (Muller et al., 2005). It was shown that
the effects of D-1MT were greatly enhanced when
given in conjunction with the commonly used
chemotherapeutic agent paclitaxel. Depletion of either
CD4+ or CD8+ T-cells in these mice abolished the
benefit provided by D-1MT, indicating the importance
of T cell immunity to the antitumor response. These
studies have all led to the initiation of phase I clinical
trials testing the efficacy of 1-MT as a cancer vaccine
adjuvant.
Signaling mechanisms upstream
and downstream of IDO
The NF-κB signaling pathway has been implicated in
IDO1 signaling through the initial observation that
INDO can be induced by interferon-γ (IFN-γ) treatment
(Ozaki et al., 1988). A more detailed report showed that
BAR adapter proteins encoded by the Bin1 gene are
important mediators of NF-κB signaling to IDO. Bin1
is a suppressor of tumor growth that is poorly
expressed in tumor cells (Ge et al., 1999; Ge et al.,
2000; Tajiri et al., 2003). Using a knockout mouse for
Bin1 it was shown that there was an increase in STAT1
and NF-κB signaling leading to increased IDO (Muller
et al., 2005; Muller et al., 2004). This suggested that
under normal conditions, Bin1 acts to suppress tumor
growth by keeping levels of IDO under control.
However, loss of this regulatory gene led to increased
tumor growth as Bin1 supported T-cell mediated
immune surveillance was impaired (Muller et al.,
2005). The engraftment of c-myc+ras-transformed skin
epithelial cells in syngeneic mice resulted in limited
tumor growth if Bin1 was present than if it was deleted,
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Smith C, Prendergast GC
858
Inflammatory programming and immune modulation in cancer by IDO
progress at the rate observed in wild-type mice (Smith
et al., 2012). The implication of these results was that
IL-6 serves as a key regulator of tumor growth
downstream of IDO, a connection of potential
therapeutic value since IL-6 levels are increased in
patients with recurring tumors (Kita et al., 2011).
The mechanisms through which IDO affects tumor
growth remain only partly elucidated. One intriguing
effector pathway appears to involve the aryl
hydrocarbon receptor (AHR), discovered to be a target
receptor for kynurenine (Opitz et al., 2011). AHR was
discovered originally as the receptor for dioxin
(2,3,7,8-tetrachlorodibenzo-p-dioxin [TCDD]). Upon
binding to AHR in dendritic cell cultures, TCDD can
induce expression of both IDO1 and IDO2 suggesting
the presence of a feed-forward regulatory loop (Vogel
et al., 2008). It is postulated that induction of IDO1
through TCDD requires a combination of signaling by
AHR and RelB, the non-canonical NF-κB signaling
molecule that may work through IL-8 and AHR
following CD40 ligation (Vogel et al., 2007a; Tas et
al., 2007; Vogel et al., 2007b). Furthermore, it was
shown that TCDD treatment of mouse splenic T-cells
resulted in increased levels of FoxP3, an effect that was
abrogated in Ahr-null mice signifying that Ahr is
important in the development of Tregs (Vogel et al.,
2008). Further mechanistic connections are suggested
by observations that kynurenic acid, a byproduct of
tryptophan catabolism, also induces AHR activity and
results in IL-6 signaling (DiNatale et al., 2010).
Interestingly, IDO1-nullizygous mice show a
diminished IL-6 levels in primary lung tumors and
pulmonary metastases (Smith et al., 2012). Taken
together, studies identify AHR and IL-6 (a target of the
GCN2 pathway activated by IDO (Metz et al., 2007))
as key players in IDO signaling in cancer.
antitumor effects, recent preclinical studies in mice
have suggested that 1MT does not act directly on IDO
but rather downstream in an effector pathway leading
to mTOR control (Metz et al., 2012).
An enzymatic inhibitor of IDO termed INCB024360
has entered clinical trials. INCB024360 is a
hydrozyamidine that competitively blocks the
degradation of tryptophan to kynurenine through IDO
with an IC50 of approximately 72 nM (Liu et al.,
2010). Similarly to 1MT, treatment with INCB024360
showed attenuated tumor growth in wild-type mice but
not in immune-deficient mice (Liu et al., 2010; Koblish
et al., 2010). In both mice and dogs, INCB024360 was
given orally, resulting in a reduction of kynurenine in
the tumors, tumor draining lymph nodes and also
plasma (Koblish et al., 2010).
There was no apparent maximum tolerated dose
determined in the Phase I trials allowing it to move into
Phase II trials, where it will be tested as a monotherapy
in ovarian cancer and as a combination therapy with
ipilimumab for metastatic melanoma.
One interesting aspect of IDO inhibition is that it may
already be occurring with other cancer drugs. For
example, the paradigm targeted cancer drug Gleevec
has been found to suppress IDO expression in GIST
cells as a result of Kit inhibition (Balachandran et al.,
2011).
Another recent study has revealed that the cytotoxic
agent β-lapachone is a direct inhibitor of IDO,
postulating that this cytotoxic agent may benefit from
the additional immunological effects that derive from
its potent uncompetitive inhibitory effects on IDO1
activity.
Other drugs in use include NSAIDs that indirectly
block IDO activity as a result of COX2 inhibition
(Sayama et al., 1981).
Another effective IDO inhibitor in mouse studies is the
anti-inflammatory ethyl pyruvate, which by inhibiting
NF-kB activity blocks IDO expression and produces
robust anti-tumor responses that are both T cell and
IDO dependent (Muller et al., 2010). Taken together,
these findings point to a promising future for IDO
inhibitors as new tools for immunotherapy and
immunochemotherapy of cancer.
Clinical trials of IDO inhibitors
IDO is an appealing therapeutic target for cancer
treatment for several reasons. Structurally, it is welldefined, allowing for easier discovery of molecular
inhibitors. Furthermore, it is both structurally and
spatially distinct from the tryptophan catabolic
enzymes IDO2 and TDO2. From a clinical viewpoint,
pharmodynamic evaluations are eased by measuring
serum tryptophan and kynurenine levels. Additionally,
while immunotherapies are gaining clinical use, they
are often restricted by being either tailored to each
patient or expensive. An enzymatic inhibitor provides a
more generic and less expensive method to alter
immune recognition and elimination of tumor cells.
The potential value of targeting IDO in cancer was
further given credence by the addition of 1MT onto a
select list of the 12 immunotherapeutic agents
identified by an NCI workshop panel as having high
potential for use in cancer therapy (Koblish et al.,
2008). 1MT is a tryptophan analog that entered early
stage clinical trials in 2008 and results are expected by
the conclusion of 2013. While 1MT may provide
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Smith C, Prendergast GC
References
Higuchi K, Kuno S, Hayaishi O.. Enzymatic formation of Dkynurenine. Federation Proc. 1963; 22:243.
Higuchi K, Hayaishi O.. Enzymic formation of D-kynurenine
from
D-tryptophan.
Arch
Biochem
Biophys.
1967
May;120(2):397-403.
Rose DP.. Tryptophan metabolism in carcinoma of the breast.
Lancet. 1967 Feb 4;1(7484):239-41.
Yoshida R, Urade Y, Tokuda M, Hayaishi O.. Induction of
indoleamine 2,3-dioxygenase in mouse lung during virus
infection. Proc Natl Acad Sci U S A. 1979 Aug;76(8):4084-6.
Sayama S, Yoshida R, Oku T, Imanishi J, Kishida T, Hayaishi
O.. Inhibition of interferon-mediated induction of indoleamine
2,3-dioxygenase in mouse lung by inhibitors of prostaglandin
859
Inflammatory programming and immune modulation in cancer by IDO
biosynthesis. Proc
Dec;78(12):7327-30.
Natl
Acad
Sci
U
S
A.
1981
Smith C, Prendergast GC
Fallarino F, Grohmann U, Vacca C, Bianchi R, Orabona C,
Spreca A, Fioretti MC, Puccetti P.. T cell apoptosis by
tryptophan catabolism. Cell Death Differ. 2002 Oct;9(10):106977.
Watanabe Y, Yoshida R, Sono M, Hayaishi O..
Immunohistochemical localization of indoleamine 2,3dioxygenase in the argyrophilic cells of rabbit duodenum and
thyroid gland. J Histochem Cytochem. 1981 May;29(5):623-32.
Yoshida R, Imanishi J, Oku T, Kishida T, Hayaishi O..
Induction of pulmonary indoleamine 2,3-dioxygenase by
interferon. Proc Natl Acad Sci U S A. 1981 Jan;78(1):129-32.
Fan F, Jin S, Amundson SA, Tong T, Fan W, Zhao H, Zhu X,
Mazzacurati L, Li X, Petrik KL, Fornace AJ Jr, Rajasekaran B,
Zhan Q.. ATF3 induction following DNA damage is regulated
by distinct signaling pathways and over-expression of ATF3
protein suppresses cells growth. Oncogene. 2002 Oct
24;21(49):7488-96.
Ozaki Y, Edelstein MP, Duch DS.. Induction of indoleamine
2,3-dioxygenase: a mechanism of the antitumor activity of
interferon gamma. Proc Natl Acad Sci U S A. 1988
Feb;85(4):1242-6.
Friberg M, Jennings R, Alsarraj M, Dessureault S, Cantor A,
Extermann M, Mellor AL, Munn DH, Antonia SJ.. Indoleamine
2,3-dioxygenase contributes to tumor cell evasion of T cellmediated rejection. Int J Cancer. 2002 Sep 10;101(2):151-5.
Taylor MW, Feng GS.. Relationship between interferongamma, indoleamine 2,3-dioxygenase, and tryptophan
catabolism. FASEB J. 1991 Aug;5(11):2516-22. (REVIEW)
Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U,
Ferrara GB.. Tryptophan-derived catabolites are responsible
for inhibition of T and natural killer cell proliferation induced by
indoleamine 2,3-dioxygenase. J Exp Med. 2002 Aug
19;196(4):459-68.
Burke F, Knowles RG, East N, Balkwill FR.. The role of
indoleamine 2,3-dioxygenase in the anti-tumour activity of
human interferon-gamma in vivo. Int J Cancer. 1995 Jan
3;60(1):115-22.
Terness P, Bauer TM, Rose L, Dufter C, Watzlik A, Simon H,
Opelz G.. Inhibition of allogeneic T cell proliferation by
indoleamine 2,3-dioxygenase-expressing dendritic cells:
mediation of suppression by tryptophan metabolites. J Exp
Med. 2002 Aug 19;196(4):447-57.
Varga J, Yufit T, Hitraya E, Brown RR.. Control of extracellular
matrix degradation by interferon-gamma. The tryptophan
connection. Adv Exp Med Biol. 1996;398:143-8.
Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C,
Bianchi R, Belladonna ML, Fioretti MC, Alegre ML, Puccetti P..
Modulation of tryptophan catabolism by regulatory T cells. Nat
Immunol. 2003 Dec;4(12):1206-12. Epub 2003 Oct 26.
Wang XZ, Ron D.. Stress-induced phosphorylation and
activation of the transcription factor CHOP (GADD153) by p38
MAP Kinase. Science. 1996 May 31;272(5266):1347-9.
Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ,
Marshall B, Brown C, Mellor AL.. Prevention of allogeneic fetal
rejection by tryptophan catabolism. Science. 1998 Aug
21;281(5380):1191-3.
Lee JR, Dalton RR, Messina JL, Sharma MD, Smith DM,
Burgess RE, Mazzella F, Antonia SJ, Mellor AL, Munn DH..
Pattern of recruitment of immunoregulatory antigen-presenting
cells
in malignant melanoma.
Lab
Invest.
2003
Oct;83(10):1457-66.
Ge K, DuHadaway J, Du W, Herlyn M, Rodeck U, Prendergast
GC.. Mechanism for elimination of a tumor suppressor:
aberrant splicing of a brain-specific exon causes loss of
function of Bin1 in melanoma. Proc Natl Acad Sci U S A. 1999
Aug 17;96(17):9689-94.
Tajiri T, Liu X, Thompson PM, Tanaka S, Suita S, Zhao H,
Maris JM, Prendergast GC, Hogarty MD.. Expression of a
MYCN-interacting isoform of the tumor suppressor BIN1 is
reduced in neuroblastomas with unfavorable biological
features. Clin Cancer Res. 2003 Aug 15;9(9):3345-55.
Hai T, Wolfgang CD, Marsee DK, Allen AE, Sivaprasad U..
ATF3 and stress responses. Gene Expr. 1999;7(4-6):321-35.
(REVIEW)
Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D,
Parmentier N, Boon T, Van den Eynde BJ.. Evidence for a
tumoral immune resistance mechanism based on tryptophan
degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003
Oct;9(10):1269-74. Epub 2003 Sep 21.
Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A,
Mellor AL.. Inhibition of T cell proliferation by macrophage
tryptophan catabolism. J Exp Med. 1999 May 3;189(9):136372.
Jiang HY, Wek SA, McGrath BC, Lu D, Hai T, Harding HP,
Wang X, Ron D, Cavener DR, Wek RC.. Activating
transcription factor 3 is integral to the eukaryotic initiation factor
2 kinase stress response. Mol Cell Biol. 2004 Feb;24(3):136577.
Ge K, Minhas F, Duhadaway J, Mao NC, Wilson D,
Buccafusca R, Sakamuro D, Nelson P, Malkowicz SB,
Tomaszewski J, Prendergast GC.. Loss of heterozygosity and
tumor suppressor activity of Bin1 in prostate carcinoma. Int J
Cancer. 2000 Apr 15;86(2):155-61.
Lu PD, Harding HP, Ron D.. Translation reinitiation at
alternative open reading frames regulates gene expression in
an integrated stress response. J Cell Biol. 2004 Oct
11;167(1):27-33.
Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M,
Ron D.. Regulated translation initiation controls stress-induced
gene expression in mammalian cells. Mol Cell. 2000
Nov;6(5):1099-108.
Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E,
Prendergast GC.. Targeted deletion of the suppressor gene
bin1/amphiphysin2 accentuates the neoplastic character of
transformed mouse fibroblasts. Cancer Biol Ther. 2004
Dec;3(12):1236-42. Epub 2004 Dec 14.
Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA..
Indoleamine 2,3-dioxygenase production by human dendritic
cells results in the inhibition of T cell proliferation. J Immunol.
2000 Apr 1;164(7):3596-9.
Mellor AL, Sivakumar J, Chandler P, Smith K, Molina H, Mao
D, Munn DH.. Prevention of T cell-driven complement
activation and inflammation by tryptophan catabolism during
pregnancy. Nat Immunol. 2001 Jan;2(1):64-8.
Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ,
Messina JL, Chandler P, Koni PA, Mellor AL.. Expression of
indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in
tumor-draining lymph nodes. J Clin Invest. 2004
Jul;114(2):280-90.
Bild AH, Turkson J, Jove R.. Cytoplasmic transport of Stat3 by
receptor-mediated endocytosis. EMBO J. 2002 Jul
1;21(13):3255-63.
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Vattem KM, Wek RC.. Reinitiation involving upstream ORFs
regulates ATF4 mRNA translation in mammalian cells. Proc
860
Inflammatory programming and immune modulation in cancer by IDO
Natl Acad Sci U S A. 2004 Aug 3;101(31):11269-74. Epub
2004 Jul 26.
of indoleamine 2,3-dioxygenase in dendritic cells by
stereoisomers of 1-methyl-tryptophan correlates with antitumor
responses. Cancer Res. 2007 Jan 15;67(2):792-801.
Astigiano S, Morandi B, Costa R, Mastracci L, D'Agostino A,
Ratto GB, Melioli G, Frumento G.. Eosinophil granulocytes
account for indoleamine 2,3-dioxygenase-mediated immune
escape in human non-small cell lung cancer. Neoplasia. 2005
Apr;7(4):390-6.
Metz R, Duhadaway JB, Kamasani U, Laury-Kleintop L, Muller
AJ, Prendergast GC.. Novel tryptophan catabolic enzyme IDO2
is the preferred biochemical target of the antitumor
indoleamine 2,3-dioxygenase inhibitory compound D-1-methyltryptophan. Cancer Res. 2007 Aug 1;67(15):7082-7.
Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E,
Prendergast GC.. Inhibition of indoleamine 2,3-dioxygenase,
an immunoregulatory target of the cancer suppression gene
Bin1, potentiates cancer chemotherapy. Nat Med. 2005
Mar;11(3):312-9. Epub 2005 Feb 13.
Munn DH, Mellor AL.. Indoleamine 2,3-dioxygenase and
tumor-induced tolerance. J Clin Invest. 2007 May;117(5):114754. (REVIEW)
Tas SW, Vervoordeldonk MJ, Hajji N, Schuitemaker JH, van
der Sluijs KF, May MJ, Ghosh S, Kapsenberg ML, Tak PP, de
Jong EC.. Noncanonical NF-kappaB signaling in dendritic cells
is required for indoleamine 2,3-dioxygenase (IDO) induction
and immune regulation. Blood. 2007 Sep 1;110(5):1540-9.
Epub 2007 May 4.
Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron
D, Mellor AL.. GCN2 kinase in T cells mediates proliferative
arrest and anergy induction in response to indoleamine 2,3dioxygenase. Immunity. 2005 May;22(5):633-42.
Okamoto A, Nikaido T, Ochiai K, Takakura S, Saito M, Aoki Y,
Ishii N, Yanaihara N, Yamada K, Takikawa O, Kawaguchi R,
Isonishi S, Tanaka T, Urashima M.. Indoleamine 2,3dioxygenase serves as a marker of poor prognosis in gene
expression profiles of serous ovarian cancer cells. Clin Cancer
Res. 2005 Aug 15;11(16):6030-9.
Vogel CF, Sciullo E, Li W, Wong P, Lazennec G, Matsumura
F.. RelB, a new partner of aryl hydrocarbon receptor-mediated
transcription. Mol Endocrinol. 2007a Dec;21(12):2941-55.
Epub 2007 Sep 6.
Vogel CF, Sciullo E, Matsumura F.. Involvement of RelB in aryl
hydrocarbon receptor-mediated induction of chemokines.
Biochem Biophys Res Commun. 2007b Nov 23;363(3):722-6.
Epub 2007 Sep 19.
Takikawa O.. Biochemical and medical aspects of the
indoleamine
2,3-dioxygenase-initiated
L-tryptophan
metabolism. Biochem Biophys Res Commun. 2005 Dec
9;338(1):12-9. Epub 2005 Sep 15. (REVIEW)
Cheever MA.. Twelve immunotherapy drugs that could cure
cancers. Immunol Rev. 2008 Apr;222:357-68. doi:
10.1111/j.1600-065X.2008.00604.x. (REVIEW)
Della Chiesa M, Carlomagno S, Frumento G, Balsamo M,
Cantoni C, Conte R, Moretta L, Moretta A, Vitale M.. The
tryptophan catabolite L-kynurenine inhibits the surface
expression of NKp46- and NKG2D-activating receptors and
regulates NK-cell function. Blood. 2006 Dec 15;108(13):411825. Epub 2006 Aug 10.
Pan K, Wang H, Chen MS, Zhang HK, Weng DS, Zhou J,
Huang W, Li JJ, Song HF, Xia JC.. Expression and prognosis
role of indoleamine 2,3-dioxygenase in hepatocellular
carcinoma. J Cancer Res Clin Oncol. 2008 Nov;134(11):124753. doi: 10.1007/s00432-008-0395-1. Epub 2008 Apr 26.
Fallarino F, Grohmann U, You S, McGrath BC, Cavener DR,
Vacca C, Orabona C, Bianchi R, Belladonna ML, Volpi C,
Santamaria P, Fioretti MC, Puccetti P.. The combined effects
of tryptophan starvation and tryptophan catabolites downregulate T cell receptor zeta-chain and induce a regulatory
phenotype in naive T cells. J Immunol. 2006 Jun
1;176(11):6752-61.
Vogel CF, Goth SR, Dong B, Pessah IN, Matsumura F.. Aryl
hydrocarbon receptor signaling mediates expression of
indoleamine 2,3-dioxygenase. Biochem Biophys Res Commun.
2008 Oct 24;375(3):331-5. doi: 10.1016/j.bbrc.2008.07.156.
Epub 2008 Aug 9.
DiNatale BC, Murray IA, Schroeder JC, Flaveny CA, Lahoti TS,
Laurenzana EM, Omiecinski CJ, Perdew GH.. Kynurenic acid
is a potent endogenous aryl hydrocarbon receptor ligand that
synergistically induces interleukin-6 in the presence of
inflammatory signaling. Toxicol Sci. 2010 May;115(1):89-97.
doi: 10.1093/toxsci/kfq024. Epub 2010 Jan 27.
Serafini P, Borrello I, Bronte V.. Myeloid suppressor cells in
cancer: recruitment, phenotype, properties, and mechanisms
of immune suppression. Semin Cancer Biol. 2006
Feb;16(1):53-65. Epub 2005 Sep 15. (REVIEW)
Weber WP, Feder-Mengus C, Chiarugi A, Rosenthal R,
Reschner A, Schumacher R, Zajac P, Misteli H, Frey DM,
Oertli D, Heberer M, Spagnoli GC.. Differential effects of the
tryptophan metabolite 3-hydroxyanthranilic acid on the
proliferation of human CD8+ T cells induced by TCR triggering
or homeostatic cytokines. Eur J Immunol. 2006 Feb;36(2):296-304.
Koblish HK, Hansbury MJ, Bowman KJ, Yang G, Neilan CL,
Haley PJ, Burn TC, Waeltz P, Sparks RB, Yue EW, Combs
AP, Scherle PA, Vaddi K, Fridman JS.. Hydroxyamidine
inhibitors of indoleamine-2,3-dioxygenase potently suppress
systemic tryptophan catabolism and the growth of IDOexpressing tumors. Mol Cancer Ther. 2010 Feb;9(2):489-98.
doi: 10.1158/1535-7163.MCT-09-0628. Epub 2010 Feb 2.
Brandacher G, Perathoner A, Ladurner R, Schneeberger S,
Obrist P, Winkler C, Werner ER, Werner-Felmayer G, Weiss
HG, Gobel G, Margreiter R, Konigsrainer A, Fuchs D,
Amberger A.. Prognostic value of indoleamine 2,3-dioxygenase
expression in colorectal cancer: effect on tumor-infiltrating T
cells. Clin Cancer Res. 2006 Feb 15;12(4):1144-51.
Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, Leffet L,
Hansbury MJ, Thomas B, Rupar M, Waeltz P, Bowman KJ,
Polam P, Sparks RB, Yue EW, Li Y, Wynn R, Fridman JS,
Burn TC, Combs AP, Newton RC, Scherle PA.. Selective
inhibition of IDO1 effectively regulates mediators of antitumor
immunity. Blood. 2010 Apr 29;115(17):3520-30. doi:
10.1182/blood-2009-09-246124. Epub 2010 Mar 2.
Wek RC, Jiang HY, Anthony TG.. Coping with stress: eIF2
kinases and translational control. Biochem Soc Trans. 2006
Feb;34(Pt 1):7-11. (REVIEW)
Ball HJ, Sanchez-Perez A, Weiser S, Austin CJ, Astelbauer F,
Miu J, McQuillan JA, Stocker R, Jermiin LS, Hunt NH..
Characterization of an indoleamine 2,3-dioxygenase-like
protein found in humans and mice. Gene. 2007 Jul
1;396(1):203-13. Epub 2007 Apr 18.
Muller AJ, DuHadaway JB, Chang MY, Ramalingam A,
Sutanto-Ward E, Boulden J, Soler AP, Mandik-Nayak L,
Gilmour SK, Prendergast GC.. Non-hematopoietic expression
of IDO is integrally required for inflammatory tumor promotion.
Cancer Immunol Immunother. 2010 Nov;59(11):1655-63. doi:
10.1007/s00262-010-0891-4. Epub 2010 Jul 17.
Hou DY, Muller AJ, Sharma MD, DuHadaway J, Banerjee T,
Johnson M, Mellor AL, Prendergast GC, Munn DH.. Inhibition
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Smith C, Prendergast GC
Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM,
861
Inflammatory programming and immune modulation in cancer by IDO
Ocuin LM, Obaid H, Sorenson EC, Popow R, Ariyan C, Rossi
F, Besmer P, Guo T, Antonescu CR, Taguchi T, Yuan J,
Wolchok JD, Allison JP, DeMatteo RP.. Imatinib potentiates
antitumor T cell responses in gastrointestinal stromal tumor
through the inhibition of Ido. Nat Med. 2011 Aug
28;17(9):1094-100. doi: 10.1038/nm.2438.
Metz R, Rust S, Duhadaway JB, Mautino MR, Munn DH,
Vahanian NN, Link CJ, Prendergast GC.. IDO inhibits a
tryptophan sufficiency signal that stimulates mTOR: A novel
IDO effector pathway targeted by D-1-methyl-tryptophan.
Oncoimmunology. 2012 Dec 1;1(9):1460-1468.
Smith C, Chang MY, Parker KH, Beury DW, DuHadaway JB,
Flick HE, Boulden J, Sutanto-Ward E, Soler AP, Laury-Kleintop
LD, Mandik-Nayak L, Metz R, Ostrand-Rosenberg S,
Prendergast GC, Muller AJ.. IDO is a nodal pathogenic driver
of lung cancer and metastasis development. Cancer Discov.
2012 Aug;2(8):722-35. doi: 10.1158/2159-8290.CD-12-0014.
Epub 2012 Jul 19.
Kita H, Shiraishi Y, Watanabe K, Suda K, Ohtsuka K, Koshiishi
Y, Goya T.. Does postoperative serum interleukin-6 influence
early recurrence after curative pulmonary resection of lung
cancer? Ann Thorac Cardiovasc Surg. 2011;17(5):454-60.
Epub 2011 Jul 13.
Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump
S, Schumacher T, Jestaedt L, Schrenk D, Weller M, Jugold M,
Guillemin GJ, Miller CL, Lutz C, Radlwimmer B, Lehmann I,
von Deimling A, Wick W, Platten M.. An endogenous tumourpromoting ligand of the human aryl hydrocarbon receptor.
Nature.
2011
Oct
5;478(7368):197-203.
doi:
10.1038/nature10491.
Atlas Genet Cytogenet Oncol Haematol. 2013; 17(12)
Smith C, Prendergast GC
This article should be referenced as such:
Smith C, Prendergast GC. Inflammatory programming and
immune modulation in cancer by IDO. Atlas Genet Cytogenet
Oncol Haematol. 2013; 17(12):856-862.
862