You are on page 1of 9

Biochimica et Biophysica Acta 1813 (2011) 19781986

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / b b a m c r

Review

Akt, FoxO and regulation of apoptosis


Xinbo Zhang b,d, Naimei Tang b,d, Timothy J. Hadden a,d, Arun K. Rishi a,b,c,
a
John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI 48201, USA
b
Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
c
Department of Oncology, Wayne State University, Detroit, MI 48201, USA
d
Department of Internal Medicine, Wayne State University, Detroit, MI 48201, USA

a r t i c l e i n f o a b s t r a c t

Article history: Forkhead box O (FoxO) transcription factors are downstream targets of the serine/threonine protein kinase B
Received 7 January 2011 (PKB)/Akt. The Akt kinase regulates processes of cellular proliferation and survival. Phosphorylation of FoxOs
Received in revised form 9 March 2011 by Akt inhibits transcriptional functions of FoxOs and contributes to cell survival, growth and proliferation.
Accepted 11 March 2011
Emerging evidence suggests involvement of FoxOs in diverse intracellular signaling pathways with critical
Available online 31 March 2011
roles in a number of physiological as well as pathological conditions including cancer. The FoxO signaling is
Keywords:
regulated by their interactions with other intracellular proteins as well as their post-translational
PKB/Akt modications such as phosphorylation. FoxOs promote cell growth inhibitory and/or apoptosis signaling by
FoxO either inducing expression of multiple pro-apoptotic members of the Bcl2-family of mitochondria-targeting
Tumor suppression proteins, stimulating expression of death receptor ligands such as Fas ligand and tumor necrosis factor-related
apoptosis-inducing ligand (TRAIL), or enhancing levels of various cyclin-dependent kinase inhibitors (CDKIs).
Coupled with their ability to cross-talk with p53, FoxOs represent an important class of tumor suppressors in a
variety of cancers. This review summarizes our current understanding of mechanisms by which Akt and FoxOs
regulate cell growth and survival that in turn offers opportunities for development of novel strategies to
combat cancer. This article is part of a Special Issue entitled: P13K-AKT-FOxO axis in cancer and aging.
2011 Elsevier B.V. All rights reserved.

1. Introduction domain in the nucleus, and their nucleocytoplasmic shuttling is regulated


by their specic nuclear export and import signal sequences. Subcellular
Forkhead box transcription factors were named after the Drosophila localization and transcriptional functions of FoxOs are often regulated by
forkhead gene, and this superfamily consists of 19 subclasses of Fox genes, their post-translational modications, such as phosphorylation, acetyla-
FoxAFoxS [1,2]. The members of this superfamily share a highly tion, and ubiquitination [3].
conserved DNA-binding FOX domain. Although some of the FOX genes Protein kinase B (PKB)/Akt pathway is an essential pathway for
are ubiquitously expressed, many members are expressed in a spatial and cell survival and growth during development and carcinogenesis. Akt
temporal manner. The FOX transcription factors that belong to the other is a serine-threonine kinase that is regulated mainly following
(O) subfamily (FoxO) include only one member each in Caenorhabditis activation of the second messenger phospholipid kinase phosphati-
elegans (dauer formation-16) and Drosophila (dFoxO), and four members dylinositol 3-kinase (PI3K). In mammals three closely related iso-
(FoxO1, 3, 4, 6) in mammals [1]. The FoxO transcription factors regulate forms of Akt are encoded by distinct loci, and functional redundancy
cellular differentiation, growth, survival, cell cycle, metabolism, stress and among Akt isoforms exists as suggested by lack of developmental
tumor suppression pathways. FoxO1 (aka FKHR), FoxO3 (aka FKHRL1), defects in animals having targeted deletion of a specic isoform [4].
and FoxO4 (aka AFX) are all expressed ubiquitously in mammals. The However, in spite of the functional redundancy, only Akt 2 and 3
FoxO1 and FoxO4 transcripts however are highly expressed in adipose isoforms are often pathologically amplied in human cancers. The
tissue and skeletal muscle, respectively, while FoxO3 is abundant in PI3K/Akt signaling regulates cell proliferation and survival in part by
various tissues including brain, heart, kidney, and spleen. FoxO6 mRNA on phosphorylating FoxOs to promote their interaction with 14-3-3
the other hand is expressed predominantly in the developing and adult protein that results in nuclear exclusion and eventual ubiquitin
brain, suggesting a role in nervous system [3]. FoxO proteins activate or proteasome pathway (UPP)-dependent degradation of FoxOs [5].
repress transcription of target genes through their DNA-binding FOX
2. AKT-FoxO signaling axis
This article is part of a Special Issue entitled: P13K-AKT-FOxO axis in cancer and aging.
Corresponding author at: Room B4334, VA Medical Center, 4646 John R., Detroit, MI
The Akt kinase remains a subject of diverse investigations due
48201, USA. Tel.: + 1 313 576 4492. mainly to its critical roles in a variety of pathways and cellular processes.
E-mail address: Rishia@Karmanos.org (A.K. Rishi). Although the Akt signaling pathways are activated by receptor tyrosine

0167-4889/$ see front matter 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbamcr.2011.03.010
X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986 1979

kinases, cytokine receptors, G-protein coupled receptors, B and T cell the NF-B pathway and consequent expression of matrix metalloprotei-
receptors, and integrins, two major cellular processes, insulin metab- nase 9 in a CNK1-dependent fashion is thought to promote invasiveness
olism and cancer progression, are critically regulated by this kinase. Akt of breast and cervical cancer cells [14].
plays an important role in metabolism by targeting glycogen synthase Signaling through the insulin-like pathway thus results in
kinase 3 (GSK3) to regulate glycogen synthesis. Akt regulates cell activation of Akt, which, in turn, activates expression of pro-survival
cycle and proliferation by directly targeting CDKIs p21WAF1/CIP1 and and anti-apoptotic genes. Situations may arise that will promote an
p27KIP1, and indirectly by modulating levels of cyclin D1 and p53. Akt increase in the strength or duration of Akt-activating signals. For
regulation of cell survival however involves direct inhibition (phos- example, mutations in PTEN may decrease dephosphorylation of PIP3;
phorylation) of pro-apoptotic signals such as Bad and FoxOs. In the mutant forms of PI3K catalytic subunit may also result in increased
discussion below we summarize information on the roles of Akt in abundance of PIP3. Mutations such as these are commonly encoun-
insulin metabolism and cell growth and survival pathways associated tered in a variety of cancers and not only highlight the importance of
with carcinogenesis. the PI3K/Akt signaling cascade but relevance of the Akt activation as a
Signaling through the insulin and insulin-like growth factor (IGF) potential therapeutic target in cancer [15,16].
pathways is essential for nutrient homeostasis as well as for growth and
development. In mammals the IGF signaling system includes three
peptide hormones, insulin, IGF1 and IGF2, that are ligands for a series of 3. FOXO and suppression signaling
receptor isoforms that includes two insulin receptors (IRa and IRb), the
IGF1 receptor (IGF1r) and two receptor hybrids consisting of an IGF1r 3.1. FoxO transcription factorsbona de tumor suppressors
monomer and either an IRa or IRb monomer. There is also an IGF2 receptor
(the mannose-6-phosphate receptor) that acts as a signaling antagonist. Increasing evidence shows that members of the FoxO subfamily are
IRb predominates in insulin sensitive target tissues such as muscle, fat and pivotal for the maintenance of tissue homeostasis in organs and are
liver and binds insulin with high afnity. IRa binds both insulin and IGF-2 frequently dysregulated in cancer. The role of FoxOs in tumorigenesis
with moderate afnity and is the predominant insulin receptor isoform in was initially discovered by their involvement in chromosomal trans-
fetal tissues [6]. Upon ligand binding, the insulin or IGF receptors undergo locations in human cancers [1719]. For example, FoxO1, FoxO3A, and
tyrosine autophosphorylation, enhancing tyrosine kinase activity of the FoxO4 genes were found at chromosomal breakpoints in some forms of
receptor and propagate signals through common pathways that include leukemia such as myeloid/lymphoid or mixed lineage leukemia (MLL)
the insulin receptor substrate (IRS)/phosphatidylinositol 3-kinase (PI3K) and alveolar rhabdomyosarcoma. The FoxO1 gene was identied in the
pathway [reviewed in 7]. The IRSs are recruited to the phosphorylated studies of t(2, 13)(q35;q14) and t(1, 13)(p36;q14) chromosomal
receptor by their phosphotyrosine binding (PTB) and pleckstrin homology translocations found in rhabdomyosarcomas. The FoxO3 gene was
(PH) domains. Receptor-bound IRS proteins that are phosphorylated by found at t(6;11)(q21;q23) chromosomal translocation from an acute
action of the intrinsic receptor tyrosine kinase bind several proteins via myeloid leukemia patient, while FoxO4 was present at t(x;11)(q13;q23)
their SH2 domains. Among these is the p85 regulatory subunit of PI3K. translocation in acute lymphoblastic leukemia [20]. Further observation
Recent work by Lim et al. [8] uncovered an important role for the indicated that the MLL-FoxO4 fusion resulted in transdominant
Connector Enhancer of KSR 1 (CNK1) scaffold protein in recruitment of repression of wild-type FoxO expressed from the remaining intact allele
IRS1 to the membrane and downstream activation of the PI3K/Akt [21]. These initial ndings suggest that loss-of-function of FoxOs due to
pathway. It was shown that CNK 1 accumulates at the cell surface in gene translocations may play important roles in tumor development,
insulin-treated cells and participates in localization of cytohesins at the even though a single allele of FoxO genes may be sufcient to prevent
cell surface. The cytohesins facilitate changes in the lipid environment of tumorigenesis [22].
the cell membrane, resulting in generation of a phosphoinositide(4,5) Overexpression of FoxO inhibits tumor growth in vitro and tumor
bisphosphate (PIP2)-rich environment, essential for recruitment of IRS1 size in vivo in breast cancer, and cytoplasmic localization of FoxO
and signaling through PI3K [9]. Interaction between the IRS and both SH2 correlated with poorer survival in breast cancer patients [23,24]. In
domains of p85 activates the catalytic (p110) subunit of PI3K. Activated contrast, nuclear localization of FoxOs suspends cell cycle progression
PI3K phosphorylates its lipid substrate PIP2 to produce phosphoinositide [25], promotes apoptosis [26], and negatively regulates angiogenesis
(3,4,5)trisphosphate (PIP3), an important intracellular messenger. PIP3 [27]. The signicance of FoxO antitumoral activity is also underscored
recruits phosphatidylinositol-dependent kinase 1 (PDK1) and Akt, both by studies conducted in leukemia [28], prostate cancer [2931], and
Ser/Thr kinases, to the plasma membrane, wherein Akt is activated by glioblastoma [32]. Furthermore, triple knockout mouse models
PDK1-mediated phosphorylation. Signaling through the IRS/PI3K path- proved the tumor suppressor properties of FoxOs, as mice simulta-
way can be modulated by phospholipid phosphatases, such as SHIP2 and neously lacking the principal members of the mammalian FoxO
PTEN (phosphatase and tensin homolog deleted from chromosome ten), subfamily, FoxO1, FoxO3a, and FoxO4, are prone to develop
which mediate dephosphorylation of PIP3. It is noteworthy that recent hemangiomas and lymphoproliferative diseases [21]. Although
work has shown that CNK1 interacts with Akt, promoting cell implicated as tumor suppressors, genetic inactivation of FOXOs is
proliferation through activation of the Akt/FoxO signaling cascade [10]. rarely encountered in human cancers. Recent studies found that
FoxOs also mediate cell cycle arrest, DNA repair and apoptosis [11]. FoxO3 was deleted in carcinogen-induced human lung adenocarci-
Phosphorylation by Akt inactivates FoxOs resulting in their accumulation noma (LAC) of mice and in human non-small cell lung cancer (NSCLC)
in the cytoplasm. Thus, signaling through the IRS/PI3K/Akt cascade, by cell lines, suggesting that FoxO3 loss contributes to NSCLC pathogen-
inactivating FoxOs, results in decreased expression of negative cell-cycle esis [33,34]. FoxO3 gene was also identied as a novel target of
regulators, such as retinoblastoma-related protein p130 and CDKI deletion in human lung adenocarcinoma. Biallelic or homozygous
p27Kip1. Akt phosphorylation of FoxOs promotes cell survival since the deletion of FoxO3 was detected in 8 of 33 (24.2%) mostly early-stage
FoxOs regulate the pro-apoptotic protein TRAIL. Cell survival is also LAC of smokers [35]. FoxOs are inactivated in the majority of human
promoted by Akt phosphorylation of BAD, a component of the BAD/Bcl- cancers, owing to the overactivation of the PI3K/Akt pathway and the
XL complex, resulting in dissociation of the complex [12]. Moreover, Akt latter is due to mutations in RAS, PTEN or PI3K [36]. These
also has the capacity to phosphorylate and activate IKK a component of observations strongly dene FoxOs as bona de tumor suppressors
the upstream kinase that regulates NF-B activation [13]. Activated IKK that may be useful as therapeutic targets, although one early
phosphorylates IB, targeting it for proteasomal degradation. As a result, investigation of FoxO3 in clinical breast cancer suggested that
NF-B is activated and translocated into the nucleus, where it activates activation of FoxO3a was intriguingly associated with lymph nodal
transcription of pro-survival genes. Activation of an alternative branch of metastasis and a poor prognosis [37].
1980 X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986

3.2. FoxO-mediated suppression signaling in cell cycle binding element (SBE) and that these features are conserved in the
human and mouse genes. Further experiments showed that in response
3.2.1. FoxO-mediated regulation of CIP/KIP family to TGF, FoxO1, FoxO3a, and FoxO4 bind specically and directly to
Timely cell cycle regulation is accomplished by sequential activation Smad3 and Smad4, and this complex targets the p21WAF1/CIP1 promoter
of a family of serine-threonine kinases called cyclin dependent kinases containing the FBHE and SBE to induce cell cycle arrest at the G1/S
(CDKs). Tight CDK regulation involves CDKIs which ensure the correct transition [32,44]. Infact, FoxOs act as Smad partners to induce CDKIs
timing of CDK activation in different phases of the cell cycle [38]. p21WAF1/CIP1 and p15 as part of the cytostatic response of epithelial cells
Disruption of cell cycle control is a hallmark of cancer [39]. In particular, [45]. More recent studies also demonstrate that CDKI p21WAF1/CIP1 is an
the reduced expression of the CDKI p27KIP1 has been extensively important mediator of FoxO-induced G1 arrest and subsequent
observed in human cancers, and its low levels are often associated with a quiescence. FoxO3a inhibited acute myeloid leukemia cell proliferation
worse prognosis [40,41]. In quiescent cells, FoxOs are located within the in part by activating transcription of the Fas-L and p21WAF1/CIP1 genes,
nucleus in which they bind to the p27KIP1 promoter and initiate whereas IKK activity maintains FoxO3a in the cytoplasm and thus
transcription [25]. In response to growth factors or oncogenic signals, establish an important role of FoxO3a inactivation/nuclear exclusion in
FoxOs are phosphorylated by Akt, resulting in their nuclear export and the proliferation and survival of AML cells [46]. Furthermore, siRNA
cytoplasmic sequestration that interferes with their nuclear transcrip- knockdown of endogenous Sam68 (Src-associated in mitosis, 68 kDa), a
tional functions including p27KIP1 transcription and thus promotes cell nuclear RNA-binding protein that plays a role in cell growth, inhibited
proliferation [42]. As Akt is active in many tumors, this may represent an cell proliferation and tumorigenicity of breast cancer cells in vitro
additional mechanism to reduce p27KIP1 during tumor growth. A wealth through blocking the G1 to S phase transition. However, the anti-
of studies to date have revealed that FoxOs target a range of genes proliferative effect of silencing Sam68 on breast cancer cells was
involved in multiple cellular processes including metabolism, cell cycle, associated with up-regulation of CDKIs p21WAF1/CIP1 and p27KIP1,
apoptosis, stress resistance, DNA repair, and immune system (Fig. 1). enhanced transactivation of FoxOs, and attenuation of Akt/GSK-3
One of the main effects of FoxOs is to promote cell cycle arrest at the G1/S signaling [47]. Thus, FoxOs play a major role in G1 arrest by upregulating
boundary by up-regulating multiple cell cycle suppressor genes such as cell cycle inhibitors (p21WAF1/CIP1 and p27KIP1) and consequent
CDKI p27KIP1 [42]. In fact activation of FoxO3 is sufcient to elevate attenuation of cell cycle promoting CDKs.
p27KIP1 mRNA and protein levels, as well as to induce apoptosis [25].
FoxOs also induce cells to exit the cell cycle and enter a state of 3.2.2. FoxO-mediated regulation of INK family and cyclins
quiescence in part by upregulating levels of the retinoblastoma family FoxOs not only control cell cycle through transcription of p21WAF1/CIP1/
member p130 [43]. Consistent with a role in driving or promoting cell p27KIP1 genes, but also upregulate INK4 family of CDKIs such as p15 and
cycle arrest, FoxOs were found to be incapable of inducing G1 arrest in p19. The CDKIs of INK4 family (p16, p15, p18 and p19) specically bind to
p27KIP1/p130-decient broblasts, suggesting that p27KIP1 and p130 are and inhibit cyclin DCDK4/6 complexes, while the CIP/KIP family (p21,
both critical mediators of FoxO-dependent G1 arrest. Thus, p27KIP1 plays p27 and p57) of CDKIs bind to and inhibit the cyclin ECDK2 as well as
a crucial role in the control of cell proliferation by inhibiting the activities other cyclinCDK complexes operating throughout the cell cycle [48].
of complexes of G1 cyclins and Cdks and, as such, is an important Katayama et al. [49] have reported that p15 and p19 transcription was
candidate for therapeutic tumor suppression. associated with FoxO-mediated G1 cell-cycle arrest. Inhibition of Akt
Another FoxO target gene is CDKI p21WAF1/CIP1. Seoane et al. [32] signaling by PI3K inhibitors increased expression of CDKIs p15 and p19
demonstrated that the p21WAF1/CIP1 promoter contains both a Smad but not p16 or p18. Conversely, Akt suppresses p15 and p19 expression by
binding region as well as a consensus forkhead binding element {FBHE; Akt-dependent phosphorylation and inactivation of FoxOs. Intriguingly,
T (G/A) TT (T/G) (G/A) (T/C)} immediately upstream of the rst Smad FoxOs can also promote cell cycle arrest by repressing cyclin D1 and D2,

Fig. 1. FoxO shuttling between nucleus and cytoplasm is phosphorylation-dependent. Boxes indicate select transcriptional targets of FoxOs within different pathways.
X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986 1981

the two positive regulators of cell cycle [50,51]. Additionally, forced 4. FoxO and apoptosis
expression of FoxO1 causes a delay in the transition from G2 to M phase,
which is abrogated by overexpression of CDK1 and cyclin B1 [52]. shRNA- According to Yin Yang theory, all matter consists of two sides. Yin is
mediated downregulation of cathepsin B and uPAR results in G0/G1 arrest, the negative side, and Yang is the positive side. The coexistence of the
prominent increase of p27KIP1 levels and inhibition of p-Rb. This increased two sides in equilibrium and/or harmony is considered a key for the
expression of CDKI p27KIP1 correlates with decreased expression of overall health of a biological system. Consistent with this theory, gain-
p-PI3K, p-Akt, cyclin E, cyclin D1, cyclin D2 and increased expression of and loss-of-function models of genes in the core apoptotic pathway
FoxO3a protein [53]. Furthermore, FoxOs also regulate expression of suggest that perturbation of cellular homeostasis can be a primary
mitotic genes such as cyclin B and polo-like kinase (Plk) [54]. Since Plk pathogenic event that results in disease. Indeed, there is now
functions in mitosis and maintenance of DNA integrity, its regulation by compelling evidence that insufcient apoptosis can manifest as
FoxO further highlights an important role of FoxOs in the control of cancer or autoimmunity, whereas accelerated cell death contributes
mammalian cell cycle. Cyclin G2 is a newly identied homologue of cyclin to acute and chronic degenerative diseases [68].
G1, and in contrast to conventional cyclins, is known to induce cell cycle Factors regulating the expression levels of survival proteins are likely
arrest and is up-regulated when the cells undergo cell cycle arrest or to play a crucial role in tumor formation. For example, aberrant
apoptosis. Cyclin G2 down-regulation has been reported in papillary overactivation of PI3K/Akt signaling is a hallmark of many human
carcinomas of the thyroid [55], breast [56], oral [57], and gastric cancers cancers. One major way by which PI3K/Akt promotes cell survival is
[58]. Modur et al. [59] found that FoxO overexpression stimulated through sequestering FoxOs away from the promoters of apoptotic
expression of several genes including cyclin G2. FoxO-dependent genes. FoxOs therefore have emerged as an important effector arm
upregulation of cyclin G2 is associated with PTEN-mediated transcription of PI3K/Akt signaling by driving multiple apoptotic gene expression [3].
in an Akt-dependent as well as an Akt-independent manner [60]. Taken
together, FoxOs mediate cell cycle arrest at the G1/S and G2/M transitions, 4.1. FoxO-upregulated BH3-only proteins
two checkpoints that are critical in the cellular response to stress, by
regulation of multiple cell cycle regulators [61]. FoxO-induced cell cycle Conventionally, apoptosis occurs via two main pathways: the
arrest may allow time for repair of damaged DNA as well as for intrinsic pathway mediated by mitochondria, resulting in the activation
detoxication of cells. of caspase 9 while the extrinsic pathway is mediated by the activation of
death receptors (Fas and TNFR) and involves the activation of caspase
8 [69,70]. The BCL-2 family of proteins are well-characterized regulators
3.3. FoxO/p53 crosstalk of apoptosis and are grouped into three subfamilies based on the
number of BH (BCL-2 homology) domains they share [71]. The BH3-only
As outlined here and in previous reviews [36,62], there are many group consists of pro-apoptotic proteins including BIK, EGl-1, Bim, BMF,
parallels in the functions and modes of regulation of FoxOs and p53 [36]. NOXA, BID, BAD, BNIP3, PUMA and Beclin-1. Initial investigations
FoxOs mediate cell cycle arrest, cell death and cellular oxidative stress to indicate that the expression of constitutively nuclear forms of FoxOs
maintain metabolic stability by activating transcription of their target trigger cell death in different types of cells which involves transactiva-
genes. The tumor suppressor p53 performs similar functions to enable tion of at least two members of pro-apoptotic BH3-only proteins (Bim
DNA repair and to maintain genomic stability [36]. Cell-cycle arrest by and BNIP3) [7275]. FoxOs induce Bim expression in hematopoietic
FoxO and p53 occurs through inducing their target genes such as p16, cells deprived of growth factors [74,75]. Overexpression of a FoxO3a
p21WAF1/CIP1 and p27KIP1 [63]. FoxO and p53 orchestrate apoptosis mutant that cannot be phosphorylated by Akt results in apoptosis of
through multiple mechanisms but favor the upregulation of the pro- BCR-ABL-transformed cells [76]. Treatment with proteasome inhibitor,
apoptotic BH3-only proteins. FoxOs are known to upregulate Bim and bortezomib, also reduces the burden of BCR-ABLinduced leukemic
PUMA while PUMA is also transcriptionally upregulated by p53. The fact disease by restoration of normal FoxO3a expression, and prolonged
that FOXOs target a number of genes including p21WAF1/CIP1, GADD45, survival of BCR-ABL-transduced mice [77]. SYUNZ-16, a new derivative
WIP1 and PA26 that are also regulated by p53, suggests that FoxOs and of alkannin that induces apoptosis is associated with an increase in the
p53 likely coordinate tumor suppression signaling [3]. Indeed available nuclear accumulation of exogenous FoxO protein, and upregulation of
data support that FoxO and p53 function together to mediate the effects the mRNA expression of Bim and TRADD in liver cancer cells [78]. In
of cellular stress. In response to nutrient deprivation, FoxOs release p53- paclitaxel-sensitive breast cancer, upregulation of FoxO3a induced by
dependent repression of the Sirt1gene, allowing the upregulation of paclitaxel causes increased levels of Bim mRNA and protein, leading to
Sirt1 expression. This repression of Sirt1 seems to be mediated by direct apoptosis and thus contribute to tumor suppression by paclitaxel [79].
interaction between FoxO and p53 proteins and is independent of the PUMA is a potent regulator of mitochondrial outer membrane
presence of FoxO binding site in the Sirt1 promoter [64]. FoxO3 has also permeabilization, attributing to two distinct mechanisms which both
been found to interact with p53 in vitro in response to stress stimuli or to rely on PUMA binding to the anti-apoptotic BCL-2 proteins: de-
nutrient deprivation [65]. Intriguing examples that further show the repression and sensitization [80]. FOXO3a is responsible for the
overlapping functions of FoxO and p53 are derived from studies in transcriptional upregulation of Puma in response to cytokine or growth
transgenic mice. Mice that lack p53 develop normally, but are factor deprivation [81], indicating that FoxO-induced transcription of
remarkably predisposed to developing broad spectrum of cancers PUMA may play a role in regulation of apoptosis and stress response
including thymic lymphoma and hemangioma, and usually die from [82]. Furthermore, FoxO4 indirectly suppresses the expression of the
these malignancies within 36 months of birth [66,67]. Similarly, pro-survival Bcl-2 family member Bcl-XL by regulating expression of the
conditional triple knockout mice lacking FOXOs (FoxO1, 3a, and 4) are transcriptional repressor Bcl-6 [83]. Thus, FoxOs can induce cell death
prone to develop hemangiomas and thymic lymphoma. On the other through the intrinsic apoptotic pathway mediated by mitochondria.
hand, the individual or paired inactivation of FoxO1, FoxO3a or FoxO4
was found to cause a less severe phenotype [22]. These observations 4.2. FoxO-mediated extrinsic apoptotic pathway
suggest that FoxO and p53 play similar roles at least in suppressing
tumorigenesis by overlapping modes of target gene regulation. Overall, As described in Fig. 1, FoxO directly regulates the extrinsic
FoxO3 together with partners including p53 provide a strong tumor apoptotic pathway through enhancing the transcription of proapop-
suppressor network to protect cells from damage to the DNA and thus totic factors such as FasL and TRAIL. The FasL promoter contains three
play an important role in maintaining genomic stability to prevent FBHEs, and Foxo3a-induced apoptosis of cerebellar neurons is
clonal emergence and expansion. decreased when Fas/FasL interaction is blocked by the decoy fusion
1982 X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986

protein Fas-Fc. Overexpression of a constitutively active form of occurs mostly because of the overactivation of their inhibitory
FoxO3 triggers apoptosis in cerebellar granule neurons through the signaling [36]. This offers a wide range of possibilities for rescuing
Fas signaling cascade [84]. IKK-insensitive FoxO3a protein mutated at FoxO activity with two major strategies: 1) reactivation of FoxO and
S644 translocated into the nucleus and activated the transcription of its regulators; 2) targeting PI3K/ Akt/mTOR axis.
the Fas-L and CDKI p21WAF1/CIP1 genes. This in turn inhibited leukemic
cell proliferation and induced apoptosis [46]. In addition, over- 5.1. Reactivation of FoxO and its regulators
expression of FoxO1 and FoxO3 in the prostate cancer cells results
in apoptosis and increased expression of TRAIL. FoxO3 DNA-binding 5.1.1. Restoration of FoxO gene expression
elements exist in the TRAIL promoter, indicating that TRAIL is a direct Since a hallmark of most cancers is the inactivation of FoxO by
target of FoxO3 [59]. Exposure of human hepatic stellate cells to TRAIL PI3K-dependent phosphorylation and hyperactivation of PI3K/Akt
led to dephosphorylation and nuclear accumulation of FoxO1 and pathway that controls many biological functions such as cell
FoxO3a. Activation of FoxOs consequently promoted TRAIL-induced proliferation, survival, and insulin response, reactivation of FoxO
apoptosis in LX-2 cells [85]. Active FoxO4 antagonized deregulated may be an invaluable therapeutic approach. Kau et al. [93] have
proliferation and induced apoptosis in chronic myelogenous leuke- employed a cell-based, chemical genetics approach to search for
mia-derived cell lines. Imatinib-resistant cells underwent apoptosis compounds that trap FoxO1 in the nucleus by modulating FoxO's
after transfection with full-length TRAIL cDNA. These results suggest dynamic nucleocytoplasmic shuttling. Two classes of compounds that
that active FoxO4 can overcome imatinib resistance in CML cells via inhibit FoxO1a nuclear export were identied: (1) compounds that
TRAIL production [86]. It is worth noting that highly selective target the general nuclear transport machinery and (2) compounds
expression of FoxO target genes has been observed in different cell specic to targeting the PI3K/Akt/FoxO1a signaling pathway. They
lines and treatment, suggesting that transcriptional targets of FoxOs have shown that 19 of the compounds that promoted retention of
are tissue- and stimulus-specic. For example, HIV infection of CD4+ T FoxO1a in the nucleus were novel general protein export inhibitors.
cells leads to induction of FoxO3a, which modulates multiple target All 19 compounds blocked the nuclear export of RevGFP and FoxO1a
genes that are critical to the G0 cell cycle entry observed in the by targeting CRM1, and not by inhibition of other factors in the
infected cells and are active players in both the extrinsic and the nuclear transport machinery [93]. There were 23 small molecule
intrinsic pathways of apoptosis [87]. However, the precise mecha- inhibitors of FoxO1a that acted in the PI3K/Akt/FoxO1a signaling
nisms are unclear. Taken together, FoxO proteins regulate cell death pathway. Importantly, some of these inhibitors were able to suppress
by modulating the expression of BH3-only proteins (Bim and BNIP3) cellular proliferation of PTEN-decient 786-0 cells, even though they
and death receptor ligands (FasL and TRAIL) that function in autocrine displayed IC50 values in the micromolar range [93]. Analyzing these
and paracrine pathways. FoxO1 nuclear export inhibitors may not only yield a new class of anti-
cancer therapies, but may also provide novel regulators of the PI3-K,
4.3. PTEN/FoxO axis in apoptosis Akt and FoxO pathways.
Phosphorylation of FoxO factors by Akt triggers the rapid
The PTEN tumor suppressor was discovered by its homozygous relocalization of FoxOs from the nucleus to the cytoplasm. Akt
deletion and other mutations following mapping of the human phosphorylates FoxOs at three key regulatory sites (Thr32, Ser253,
chromosome 10q23 in cancer [88]. Sequencing of the PTEN gene in and Ser315 in the FoxO3 sequence) that are conserved from
tumors found it to be one of the most commonly mutated tumor Caenorhabditis elegans to mammals and are part of a perfect consensus
suppressors in human malignancies. Thus PTEN signaling is almost sequence for Akt phosphorylation [91]. Based on this feature of FoxOs,
universally altered in cancer [88]. Analysis of PTEN decient hematopoi- another strategy could be to mutate the three well-characterized Akt
etic system revealed that conditional ablation of PTEN drives exit from phosphorylation sites on FoxOs to alanine. This AAA FoxO mutant can
quiescence through PI3K/Akt hyperactivation, and leads to exhaustion of no longer undergo phosphorylation and will accumulate in the
normal hematopoietic stem cells while at the same time initiating acute nucleus, allowing reconstitution of FoxO activity in the PTEN null cells.
leukemia [89]. Interestingly, mice with FoxO deletions never develop Indeed, overexpression of Foxo1 or Foxo3a triple AAA mutant by
AML, suggesting that the repression of FoxO activity by Akt is not adenovirus-mediated gene transfer inhibited cell growth by inducing
sufcient for leukemogenesis, although other types of malignancies cell cycle arrest in sub-G1 phase, and subsequently apoptotic cell
develop in these mice [22,90]. This suggests that alternative or additional death, as evident by typical apoptotic morphologic changes, annexin
downstream targets of Akt are required for leukemic transformation. V-PE analysis, and caspase cascade activation in melanoma [94] and
PTEN is a major negative regulator of the PI3K-Akt pathway. In cells that endometrial cancer [95]. These data suggest that adenovirus expres-
carry the PTEN mutation, the PI3K pathway is often constitutively active, sing a FoxO triple mutant could be a useful vector for gene therapy of
leading to inactivation of endogenous FoxOs and possible initiation and cancers resistant to chemotherapy and radiotherapy induced by
progression of tumors [91]. Phosphorylation-dependent nuclear/cyto- hyperactivity of PI3K/Akt.
plasmic shuttling of FoxOs is mainly regulated by Akt and PTEN. FoxO It has been shown that the some cancer therapeutic drugs activate
phosphorylation by Akt results in sequestration of FoxOs in the FoxOs by attenuating Akt activity. In paclitaxel-sensitive MCF-7 breast
cytoplasm and inhibition of target gene transcription. FoxO dephosphor- cancer cells, up-regulation of FoxO3a by paclitaxel can cause increased
ylation leads to nuclear translocation and target gene activation [92]. levels of Bim mRNA and protein, resulting in apoptosis. Silencing of
Since PTEN is a FoxO target gene, FoxOs can enhance transcription of FoxO3a by small interfering RNA greatly reduces the induction of
PTEN to positively reinforce their biological function of tumor suppres- apoptosis caused by paclitaxel [79]. Furthermore, paclitaxel not only
sion. Therefore, restoring nuclear expression of FoxOs and their target induces FoxO3a expression but also enhances its nuclear relocation
genes may provide an important therapeutic approach for targeted through JNK-dependent inhibition of the PI3K/Akt signaling pathway,
cancer therapy. suggesting a crosstalk between the PI3K-Akt-FoxO3a and JNK signaling
pathways [96]. Phosphorylation of FoxO3a by JNK reduces the interaction
5. Targeting AKT-FoxO signaling axis between FoxO3a with 14-3-3 protein, leading to impaired nuclear export
of FoxO3a, and thereby stimulating FoxO3a activity [96]. KP372-1 (a
Based on tumor suppressor properties of FoxOs, their reactivation multiple kinase inhibitor) also inhibits pathways critical for acute myeloid
is considered an attractive anti-cancer strategy. In contrast to other leukemia survival, including PDK1/Akt and FLT3 through activation of
tumor suppressors such as p53 or PTEN, the functions of which are FoxO activity [97]. In addition, selenium (a chemopreventive agent),
abrogated by genetic or epigenetic changes, inactivation of FoxOs which signicantly reduces the incidence of prostate cancer, upregulates
X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986 1983

FoxO1 expression when compared to gene expression prole of a control phase I and II clinical trials [123].These range from inhibitors reported to
group [98]. Further analysis demonstrates that FoxO1 activation is critical act on a single class I PI3K such CAL-101, to inhibitors of multiple class I
for the anticancer effects of methylseleninic acid in prostate cancer cells PI3K isoforms such as PX-866, XL-147, and GDC-0941, and to inhibitors
[99]. Since androgens provide an Akt-independent cell survival signal in acting on multiple class I isoforms and other PIK family members such as
prostate cancer cells, FoxO transcription factors therefore are important BEZ235 and XL765 [123]. Overall, there is limited clinical benet with
nuclear targets for both Akt-dependent and -independent survival signals inhibitors of PI3K. The development of such inhibitors raises issues as to
in prostate cancer cells [100]. Thus restoring FoxO activity by different what specic member or members in the PI3K family should be inhibited
approaches can be benecial for cancer treatment and cancer prevention. to achieve maximal therapeutic benet, and can specic inhibitors be
developed with the necessary pharmacologic properties to allow them to
5.1.2. Restoration of PTEN gene expression by targeting miRNA proceed to clinical trials? The question also remains to be answered
MicroRNAs (miRNAs) are short non-protein-coding RNAs (~22 whether greater therapeutic efcacy will be obtained through the use of
nucleotide) that are known to alter gene expression at a post- inhibitors with increased specicity, or through inhibitors that act on a
transcriptional level [101,102]. Over 1200 validated human miRNAs spectrum of targets within the pathway [124].
have been discovered to date (http://www.mirbase.org), and they are To date, several types of Akt inhibitors have been investigated,
predicted to regulate one third of the human genome with involve- including phosphatidylinositol analog inhibitors, allosteric Akt kinase
ment in development and progression of many diseases [103105]. inhibitors, ATP-competitive inhibitors, and alkylphospholipids
miRNA overexpression has been shown to be indicative of invasion [125,126]. However, the use of these inhibitors is limited either by
and chemoresistance through two mechanisms. The rst is by high toxicity or by low bioavailability and stability in vivo, even
increased cell proliferation and decreased expression of programmed though several agents (e.g., perifosine, MK-2206, RX-0201, PBI-05204,
cell death 4 (PDCD4). The second mechanism is by repression of the GSK2141795) have been in various stages of clinical testing [127].
tumor suppressor PTEN in non-small cell lung cancer, which is Severe side effects of Akt inhibitors may be due to the critical role of
involved in regulation of cell growth and invasion [106]. Several Akt in multiple intracellular processes [128].
miRNAs have been shown to function as oncogenic miRNAs (onco- The most studied target in PI3K/Akt/mTOR pathway is mTOR. Four
Mirs) due to the fact that they target transcripts encoding key drugs that target mTOR have been explored for clinical use:
regulators of cell proliferation and apoptosis such as PTEN and PDCD4 rapamycin, temsirolimus, everolimus and deforolimus [129,130]. In
[107,108]. The PTEN mRNA has an unusual, about 3.3 kb long 3 patients with advanced breast cancer, these inhibitors have modest
untranslated region (UTR) that is suggestive of tight post-transcrip- anticancer activity in the range of around 10% because of collateral
tional control of PTEN expression. Adding to the complexity of PTEN effect of these inhibitors on feed-back activation of Akt and PI3K
regulation, recent evidence further indicated that the PTEN mRNA [131,132]. To date, with the exception of kidney cancer, mTOR
undergoes post-transcriptional repression/degradation by specic inhibitors have failed to inhibit tumor progression in a variety of
miRNAs. Within the PTEN 3UTR are several conserved miRNA target cancers [133,134]. Since there is a crosstalk between the estrogen
sites, and PTEN has been shown to be repressed by a variety of miRNAs receptor and the PI3K/Akt/mTOR pathway, clinical trials have
derived from miRNA precursors containing a single hairpin structure explored the combination of mTOR inhibitors and aromatase in-
such as miR-19a, 21, 22, 26a, 214, 216a, 217, 221, 222, and 494, as well hibitors. Recent data show that everolimus in combination of letrozole
as those with a polysictronic structure, such as mir-1792, mir-106b- proved to be superior over letrozole alone with a signicant response
25, mir-367-302b, and mir-221-222 [107,109116]. Emerging evi- rate (68% vs. 59%) in the patients with ER positive breast cancers
dence also indicates that aberrant expression of miRNAs is highly [130]. Although molecular targeting of tumor-specic signal trans-
linked to chemoresistance to cancer therapeutic drugs such as Her2 duction pathways is a promising strategy for discovering and
monoclonal antibody trastuzumab and the anti-estrogen tamoxifen developing novel and potent anticancer drugs, evolution of our
[106]. Therefore, antisense oligonucleotides targeted to oncoMirs knowledge of the PI3K/Akt/FoxO pathway will serve as an important
represents a potential new approach for cancer therapy as well as a guide in design of future anti-cancer agents with maximal efcacy and
solution for chemoresistance via restoration of depressed intracellular minimal side effects.
PTEN levels [115,117120].
6. Conclusions
5.2. Targeting PI3K/Akt/mTOR axis
FoxO transcription factors are emerging as key regulators of cell
Given the central role of PI3K/Akt/mTOR pathway in tumorigenesis, growth. Their ability to limit cell proliferation by promoting
its inhibition seems a powerful approach to induce anti-tumor activity. quiescence, apoptosis, or both plays a critical role in a variety of
Akt is a core player in PI3K/Akt/mTOR axis activated through physiological and pathological processes including tumor suppression.
phosphorylation of Ser-473/474 and Thr-308/309 [94]. Targeting the FoxOs are one of the major targets of the PI3K/Akt signaling in the
upstream regulators or downstream effectors of Akt is one strategy to presence of growth factors and nutrients. Activation of the PI3K
identify new targets for drug discovery. More than 100 lead compounds pathway promotes phosphorylation, and consequent nuclear exclu-
targeting multiple nodes of PI3K signaling, including PI3K, Akt, mTOR, sion of the FoxOs results in blockage of their growth inhibitory
etc., are in the preclinical drug-development pipeline so far [121,122], transcriptional functions. Since growth inhibitory functions of FoxOs
and a growing number of promising agents continually enter intensive involve activation of several proteins that include inhibitors of cell
phase I, II, and III clinical trials (http://www.clinicaltrials.gov). PTEN is cycle, retinoblastoma protein, inducers of extrinsic as well as intrinsic
the key antagonist of the PI3K/Akt pathway. Loss of PTEN in many types apoptosis, and interactions with tumor suppressors such as p53,
of cancer is associated not only with tumor development but also with restoring activities of FoxOs therefore is an excellent anti-cancer
clinical resistance to many anticancer drugs. Thus, targeting key strategy. Indeed, over-expression of phosphorylation-decient FoxO
components of PI3K/Akt/mTOR axis is a desirable means to overcome mutants has provided a proof-of-principle approach for suppression of
PTEN functional loss and consequent restoration of FoxO function. cell growth. Although FoxOs are known to function in a tissue and
Wortmannin and LY294002 are well-known PI3K inhibitors that have context-dependent manner, and functional redundancy exists among
been widely used as research tools to elucidate the role of PI3K/Akt/mTOR the FoxOs, identication of small molecule compounds that selectively
signaling in various tumor cells. Unfortunately, high cytotoxicity of both target one or multiple FoxOs and promote their nuclear retention or
inhibitors precluded their clinical application. Nonetheless, several inhibit their nuclear export nevertheless offer excellent opportunities
inhibitors of PI3K have moved through preclinical studies and into for cancer-related drug design. Furthermore, as Akt-dependent
1984 X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986

phosphorylation promotes nuclear exclusion and degradation of [27] T.T. Tang, L.A. Lasky, The forkhead transcription factor FOXO4 induces the down-
regulation of hypoxia-inducible factor 1 by a von hippel-lindau protein-
FoxOs, pharmacologic targeting of the PI3K-Akt pathway in combina- independent mechanism, J. Biol. Chem. 278 (2003) 3012530135.
tion with strategies that promote nuclear retention of FoxOs have the [28] Z. Jagani, A. Singh, R. Khosravi-Far, FoxO tumor suppressors and BCR-ABL-
potential to be superior in targeting a range of inammation- induced leukemia: a matter of evasion of apoptosis, Biochim. Biophys. Acta 1785
(2008) 6384.
associated pathologies including cancer. [29] L.C. Trotman, A. Alimonti, P.P. Scaglioni, J.A. Koutcher, C. Cordon-Cardo, P.P. Pandol,
Identication of a tumour suppressor network opposing nuclear Akt function,
Acknowledgements Nature 441 (2006) 523527.
[30] N. Kikuno, H. Shiina, S. Urakami, K. Kawamoto, H. Hirata, Y. Tanaka, R.F. Place,
D. Pookot, S. Majid, M. Igawa, R. Dahiya, Knockdown of astrocyte-elevated
Supported by the Department of Veterans Affairs Merit Review gene-1 inhibits prostate cancer progression through upregulation of FOXO3a
grant to AKR. activity, Oncogene 26 (2007) 76477655.
[31] A.N. Cornforth, J.S. Davis, E. Khanifar, K.L. Nastiuk, J.J. Krolewski, FOXO3a mediates
the androgen-dependent regulation of FLIP and contributes to TRAIL-induced
References apoptosis of LNCaP cells, Oncogene 27 (2008) 44224433.
[32] J. Seoane, H.V. Le, L. Shen, S.A. Anderson, J. Massagu, Integration of Smad and
[1] G. Tuteja, K.H. Kaestner, Forkhead transcription factors I, Cell 130 (2007) 1160. forkhead pathways in the control of neuroepithelial and glioblastoma cell
[2] G. Tuteja, K.H. Kaestner, Forkhead transcription factors II, Cell 131 (2007) 192. proliferation, Cell 117 (2004) 211223.
[3] Z. Fu, D.J. Tindall, FOXOs, cancer and regulation of apoptosis, Oncogene 27 (2008) [33] C.R. Herzog, D.C. Blake Jr., O.R. Mikse, L.S. Grigoryeva, E.L. Gundermann, FoxO3a
23122319. gene is a target of deletion in mouse lung adenocarcinoma, Oncol. Rep. 22 (2009)
[4] T.F. Franke, C.P. Hornik, L. Segev, G.A. Shostak, C. Sugimoto, PI3K/Akt and 837843.
apoptosis: size matters, Oncogene 22 (2003) 89838998. [34] D.C. Blake Jr., O.R. Mikse, W.M. Freeman, C.R. Herzog, FOXO3a elicits a pro-
[5] H. Matsuzaki, H. Daitoku, M. Hatta, K. Tanaka, A. Fukamizu, Insulin-induced apoptotic transcription program and cellular response to human lung carcinogen
phosphorylation of FKHR (Foxo1) targets to proteasomal degradation, Proc. Natl nicotine-derived nitrosaminoketone (NNK), Lung Cancer 67 (2010) 3747.
Acad. Sci. U.S.A. 100 (2003) 1128511290. [35] O.R. Mikse, D.C. Blake Jr., N.R. Jones, Y.W. Sun, S. Amin, C.J. Gallagher, P. Lazarus,
[6] G. Pandini, E. Medico, E. Conte, L. Sciacca, R. Vigneri, A. Belore, Differential gene J. Weisz, C.R. Herzog, FOXO3 encodes a carcinogen-activated transcription
expression induced by insulin and insulin-like growth factor-II through the factor frequently deleted in early-stage lung adenocarcinoma, Cancer Res. 70
insulin receptor isoform A, J. Biol. Chem. 278 (2003) 4217842189. (2010) 62056215.
[7] A. Taguchi, M.F. White, Insulin-like signaling, nutrient homeostasis, and life span, [36] T.B. Dansen, B.M. Burgering, Unravelling the tumor-suppressive functions of
Ann. Rev. Physiol. 70 (2008) 191212. FOXO proteins, Trends Cell Biol. 18 (2008) 421429.
[8] J. Lim, M. Zhou, T.D. Veenstra, D.K. Morrison, The CNK1 scaffold binds cytohesins [37] G.S. Jin, E. Kondo, T. Miyake, M. Shibata, T. Takashima, Y.X. Liu, K. Hayashi, T. Akagi,
and promotes insulin pathway signaling, Genes Dev. 24 (2010) 14961506. T. Yoshino, Expression and intracellular localization of FKHRL1 in mammary
[9] W. Kolanus, Guanine nucleotide exchange factors of the cytohesin family and gland neoplasms, Acta Med. Okayama 58 (2004) 197205.
their roles in signal transduction, Immunol. Rev. 218 (2007) 102113. [38] J. Lee, S.S. Kim, The function of p27 KIP1 during tumor development, Exp. Mol.
[10] R.D. Fritz, Z. Varga, G. Radziwill, CNK1 is a novel Akt interaction partner that Med. 41 (2009) 765771.
promotes cell proliferation through the Akt-FoxO signalling axis, Oncogene 29 [39] D.O. Morgan, Principles of CDK regulation, Nature 374 (1995) 131134.
(2010) 35753582. [40] B. Belletti, M.S. Nicoloso, M. Schiappacassi, E. Chimienti, S. Berton, F. Lovat, A.
[11] E.W.-F. Lam, R.E. Francis, M. Petkovic, FOXO transcription factors: key regulators Colombatti, G. Baldassarre, p27(kip1) functional regulation in human cancer: a
of cell fate, Biochem. Soc. Trans. 34 (2006) 722726. potential target for therapeutic designs, Curr. Med. Chem. 12 (2005)
[12] S.R. Datta, H. Dudek, X. Tao, S. Masters, H. Fu, Y. Gotoh, M.E. Greenberg, Akt 15891605.
phosphorylation of BAD couples survival signals to the cell-intrinsic death [41] M. Malumbres, M. Barbacid, To cycle or not to cycle: a critical decision in cancer,
machinery, Cell 91 (1997) 231241. Nat. Rev. Cancer 1 (2001) 222231.
[13] H.C. Dan, M.J. Cooper, P.C. Cogswell, J.A. Duncan, J.P. Ting, A.S. Baldwin, Akt-dependent [42] P.F. Dijkers, R.H. Medema, C. Pals, L. Banerji, N.S. Thomas, E.W. Lam, B.M. Burgering,
regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with IKK, J.A. Raaijmakers, J.W. Lammers, L. Koenderman, P.J. Coffer, Forkhead transcription
Genes Dev. 22 (2008) 14901500. factor FKHR-L1 modulates cytokine-dependent transcriptional regulation of p27
[14] R.D. Fritz, G. Radziwill, CNK1 promotes invasion of cancer cells through NF-B- (KIP1), Mol. Cell. Biol. 20 (2000) 91389148.
dependent signaling, Mol. Cancer Res. 8 (2010) 395406. [43] G.J. Kops, R.H. Medema, J. Glassford, M.A. Essers, P.F. Dijkers, P.J. Coffer, E.W. Lam,
[15] E. Foulstone, S. Prince, O. Zaccheo, J.L. Burns, J. Harper, C. Jacobs, D. Church, A.B. Hassan, B.M. Burgering, Control of cell cycle exit and entry by protein kinase B-regulated
Insulin-like growth factor ligands, receptors and binding proteins in cancer, J. Pathol. forkhead transcription factors, Mol. Cell. Biol. 22 (2002) 20252036.
205 (2005) 145153. [44] K.C. Arden, FoxO: linking new signaling pathways, Mol. Cell 14 (2004) 416418.
[16] M. Pollak, Insulin and insulin-like growth factor signaling in neoplasia, Nat. Rev. [45] R.R. Gomis, C. Alarcn, W. He, Q. Wang, J. Seoane, A. Lash, J. Massagu, A FoxO-
Cancer 8 (2008) 915928. Smad synexpression group in human keratinocytes, Proc. Natl Acad. Sci. U.S.A.
[17] R.J. Davis, C.M. D'Cruz, M.A. Lovell, J.A. Biegel, F.G. Barr, Fusion of PAX7 to FKHR 103 (2006) 1274712752.
by the variant t(1;13)(p36;q14) translocation in alveolar rhabdomyosarcoma, [46] N. Chapuis, S. Park, L. Leotoing, J. Tamburini, F. Verdier, V. Bardet, A.S. Green, L.
Cancer Res. 54 (1994) 28692872. Willems, F. Agou, N. Ifrah, F. Dreyfus, G. Bismuth, V. Baud, C. Lacombe, P. Mayeux,
[18] A. Borkhardt, R. Repp, O.A. Haas, T. Leis, J. Harbott, J. Kreuder, J. Hammermann, T. D. Bouscary, I{kappa}B kinase overcomes PI3K/Akt and ERK/MAPK to control
Henn, F. Lampert, Cloning and characterization of AFX, the gene that fuses to MLL in FOXO3a activity in acute myeloid leukemia, Blood 116 (2010) 42404250.
acute leukemias with a t(X;11) (q13;q23), Oncogene 14 (1997) 195202. [47] L. Song, L. Wang, Y. Li, H. Xiong, J. Wu, J. Li, M. Li, Sam68 up-regulation correlates
[19] M.J. Anderson, C.S. Viars, S. Czekay, W.K. Cavenee, K.C. Arden, Cloning and with, and its down-regulation inhibits, proliferation and tumourigenicity of
characterization of three human forkhead genes that comprise an FKHR-like breast cancer cells, J. Pathol. 222 (2010) 227237.
gene subfamily, Genomics 47 (1998) 187199. [48] J. Wesierska-Gadek, M. Maurer, N. Zulehner, O. Komina, Whether to target single or
[20] F.G. Barr, Gene fusions involving PAX and FOX family members in alveolar multiple CDKs for therapy? That is the question, J. Cell Physiol 226 (2011) 341349.
rhabdomyosarcoma, Oncogene 20 (2001) 57365746. [49] K. Katayama, A. Nakamura, Y. Sugimoto, T. Tsuruo, N. Fujita, FOXO transcription
[21] C.W. So, M.L. Cleary, MLL-AFX requires the transcriptional effector domains of factor-dependent p15(INK4b) and p19(INK4d) expression, Oncogene 27 (2008)
AFX to transform myeloid progenitors and transdominantly interfere with 16771686.
forkhead protein function, Mol. Cell. Biol. 22 (2002) 65426552. [50] S. Ramaswamy, N. Nakamura, I. Sansal, L. Bergeron, W.R. Sellers, A novel
[22] J.H. Paik, R. Kollipara, G. Chu, H. Ji, Y. Xiao, Z. Ding, L. Miao, Z. Tothova, J.W. Horner, mechanism of gene regulation and tumor suppression by the transcription factor
D.R. Carrasco, S. Jiang, D.G. Gilliland, L. Chin, W.H. Wong, D.H. Castrillon, R.A. FKHR, Cancer Cell 2 (2002) 8191.
DePinho, FoxOs are lineage-restricted redundant tumor suppressors and regulate [51] M. Schmidt, S. Fernandez de Mattos, A. van der Horst, R. Klompmaker, G.J. Kops,
endothelial cell homeostasis, Cell 128 (2007) 309323. E.W. Lam, B.M. Burgering, R.H. Medema, Cell cycle inhibition by FoxO forkhead
[23] J.Y. Yang, C.S. Zong, W. Xia, H. Yamaguchi, Q. Ding, X. Xie, J.Y. Lang, C.C. Lai, C.J. Chang, transcription factors involves downregulation of cyclin D, Mol. Cell. Biol. 22
W.C. Huang, H. Huang, H.P. Kuo, D.F. Lee, L.Y. Li, H.C. Lien, X. Cheng, K.J. Chang, C.D. (2002) 78427852.
Hsiao, F.J. Tsai, C.H. Tsai, A.A. Sahin, W.J. Muller, G.B. Mills, D. Yu, G.N. Hortobagyi, [52] P. Liu, T.P. Kao, H. Huang, CDK1 promotes cell proliferation and survival via
M.C. Hung, ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2- phosphorylation and inhibition of FOXO1 transcription factor, Oncogene 27
mediated degradation, Nat. Cell Biol. 10 (2008) 138148. (2008) 47334744.
[24] M.C. Hu, D.F. Lee, W. Xia, L.S. Golfman, F. Ou-Yang, J.Y. Yang, Y. Zhou, S. Bao, N. Handa, [53] S. Gopinath, R.R. Malla, C.S. Gondi, K. Alapati, D. Fassett, J.D. Klopfenstein, D.H. Dinh,
H. Saso, R. Kobayashi, M.C. Hung, IkappaB kinase promotes tumorigenesis through M. Gujrati, J.S. Rao, Co-depletion of cathepsin B and uPAR induces G0/G1 arrest in
inhibition of forkhead FOXO3a, Cell 16 (2004) 225237. glioma via FOXO3a mediated p27 upregulation, PLoS ONE 5 (2010) e11668.
[25] R.H. Medema, G.J. Kops, J.L. Bos, B.M. Burgering, AFX-like forkhead transcription [54] B. Alvarez, C. Martnez-A, B.M. Burgering, A.C. Carrera, Forkhead transcription
factors mediate cell-cycle regulation by Ras and PKB through p27kip1, Nature factors contribute to execution of the mitotic programme in mammals, Nature
404 (2000) 782787. 413 (2001) 744747.
[26] C. Urbich, A. Knau, S. Fichtlscherer, D.H. Walter, T. Bruhl, M. Potente, T. Brhl, [55] Y. Ito, H. Yoshida, T. Uruno, K. Nakano, Y. Takamura, A. Miya, K. Kobayashi, T.
M. Potente, W.K. Hofmann, S. de Vos, A.M. Zeiher, S. Dimmeler, FOXOdepen- Yokozawa, F. Matsuzuka, K. Kuma, A. Miyauchi, Decreased expression of cyclin G2
dent expression of the proapoptotic protein Bim: pivotal role for apoptosis is signicantly linked to the malignant transformation of papillary carcinoma of
signaling in endothelial progenitor cells, FASEB J. 19 (2005) 974976. the thyroid, Anticancer Res. 23 (2003) 23352338.
X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986 1985

[56] J. Frasor, J.M. Danes, B. Komm, K.C. Chang, C.R. Lyttle, B.S. Katzenellenbogen, [85] S.J. Park, H.Y. Sohn, J. Yoon, S.I. Park, Down-regulation of FoxO-dependent c-FLIP
Proling of estrogen up- and down-regulated gene expression in human breast expression mediates TRAIL-induced apoptosis in activated hepatic stellate cells,
cancer cells: insights into gene networks and pathways underlying estrogenic Cell. Signal. 21 (2009) 14951503.
control of proliferation and cell phenotype, Endocrinology 144 (2003) [86] S. Kikuchi, T. Nagai, M. Kunitama, K. Kirito, K. Ozawa, N. Komatsu, Active FKHRL1
45624574. overcomes imatinib resistance in chronic myelogenous leukemia-derived cell
[57] Y. Kim, S. Shintani, Y. Kohno, R. Zhang, D.T. Wong, Cyclin G2 dysregulation in lines via the production of tumor necrosis factor-related apoptosis-inducing
human oral cancer, Cancer Res. 64 (2004) 89808986. ligand, Cancer Sci. 98 (2007) 19491958.
[58] M.G. Choi, J.H. Noh, J.Y. An, S.K. Hong, S.B. Park, Y.H. Baik, K.M. Kim, T.S. Sohn, S. [87] A. Dabrowska, N. Kim, A. Aldovini, Tat-induced FOXO3a is a key mediator of apoptosis
Kim, Expression levels of cyclin G2, but not cyclin E, correlate with gastric cancer in HIV-1-infected human CD4+ T lymphocytes, J. Immunol. 181 (2008) 84608477.
progression, J. Surg. Res. 157 (2009) 168174. [88] M. Keniry, R. Parsons, The role of PTEN signaling perturbations in cancer and in
[59] V. Modur, R. Nagarajan, B.M. Evers, J. Milbrandt, FOXO proteins regulate tumor targeted therapy, Oncogene 27 (2008) 54775485.
necrosis factor-related apoptosis inducing ligand expression. Implications for [89] K. Ito, R. Bernardi, P.P. Pandol, A novel signaling network as a critical rheostat
PTEN mutation in prostate cancer, J. Biol. Chem. 277 (2002) 4792847937. for the biology and maintenance of the normal stem cell and the cancer-
[60] P.W. van Duijn, A.C. Ziel-van der Made, J.A. van der Korput, J. Trapman, PTEN- initiating cell, Curr. Opin. Genet. Dev. 19 (2009) 5159.
mediated G1 cell-cycle arrest in LNCaP prostate cancer cells is associated with [90] R. Tothova, B.J. Kollipara, B.H. Huntly, D.H. Lee, D.E. Castrillon, E.P. Cullen, S.
altered expression of cell-cycle regulators, Prostate 70 (2010) 135146. McDowell, I.R. Lazo-Kallanian, C. Williams, S.A. Sears, E. Armstrong, R.A. Passegu,
[61] Z. Tothova, D.G. Gilliland, FoxO transcription factors and stem cell homeostasis: D.G. DePinho, D.G. Gilliland, FoxOs are critical mediators of hematopoietic stem cell
insights from the hematopoietic system, Cell Stem Cell (2007) 140152. resistance to physiologic oxidative stress, Cell 128 (2007) 325339.
[62] A. van der Horst, B.M. Burgering, Stressing the role of FoxO proteins in lifespan [91] E.L. Greer, A. Brunet, FOXO transcription factors at the interface between
and disease, Nat. Rev. Mol. Cell Biol. 8 (2007) 440450. longevity and tumor suppression, Oncogene 24 (2005) 74107425.
[63] A.J. Levine, C.A. Finlay, P.W. Hinds, P53 is a tumor suppressor gene, Cell 116 [92] B.M. Burgering, G.J. Kops, Cell cycle and death control: long live Forkheads,
(2004) S67S69. Trends Biochem. Sci. 27 (2002) 352360.
[64] S. Nemoto, M.M. Fergusson, T. Finkel, Nutrient availability regulates SIRT1 [93] T.R. Kau, F. Schroeder, S. Ramaswamy, C.L. Wojciechowski, J.J. Zhao, T.M. Roberts,
through a forkhead-dependent pathway, Science 306 (2004) 21052108. J. Clardy, W.R. Sellers, P.A. Silver, A chemical genetic screen identies inhibitors
[65] A. Brunet, L.B. Sweeney, J.F. Sturgill, K.F. Chua, P.L. Greer, Y. Lin, H. Tran, S.E. Ross, of regulated nuclear export of a Forkhead transcription factor in PTEN-decient
R. Mostoslavsky, H.Y. Cohen, L.S. Hu, H.L. Cheng, M.P. Jedrychowski, S.P. Gygi, tumor cells, Cancer Cell 4 (2003) 463476.
D.A. Sinclair, F.W. Alt, M.E. Greenberg, Stress-dependent regulation of FOXO [94] J.G. Gomez-Gutierrez, V. Souza, H.Y. Hao, R. Montes de Oca-Luna, Y.B. Dong, H.S. Zhou,
transcription factors by the SIRT1 deacetylase, Science 303 (2004) 20112015. K.M. McMasters, Adenovirus-mediated gene transfer of FKHRL1 triple mutant
[66] L.A. Donehower, M. Harvey, B.L. Slagle, M.J. McArthur, C.A. Montgomery Jr., J.S. efciently induces apoptosis in melanoma cells, Cancer Biol. Ther. 5 (2006) 875883.
Butel, A. Bradley, Mice decient for p53 are developmentally normal but [95] E.C. Ward, A.V. Hoekstra, L.J. Blok, P. Hani-Moghaddam, J.R. Lurain, D.K. Singh,
susceptible to spontaneous tumours, Nature 356 (1992) 215221. B.M. Buttin, J.C. Schink, J.J. Kim, The regulation and function of the forkhead
[67] T. Jacks, L. Remington, B.O. Williams, E.M. Schmitt, S. Halachmi, R.T. Bronson, R.A. transcription factor, Forkhead box O1, is dependent on the progesterone
Weinberg, Tumor spectrum analysis in p53-mutant mice, Curr. Biol. 4 (1994) 17. receptor in endometrial carcinoma, Endocrinology 149 (2008) 19421950.
[68] M. MacFarlane, Cell death pathwayspotential therapeutic targets, Xenobiotica [96] A. Sunters, P.A. Madureira, K.M. Pomeranz, M. Aubert, J.J. Brosens, S.J. Cook, B.M.
39 (2009) 616624. Burgering, R.C. Coombes, E.W. Lam, Paclitaxel-induced nuclear translocation of
[69] F.L. Kiechle, X. Zhang, Apoptosis: biochemical aspects and clinical implications, FOXO3a in breast cancer cells is mediated byc-Jun NH2-terminal kinase and Akt,
Clin. Chim. Acta 326 (2002) 2745. Cancer Res. 66 (2006) 212220.
[70] A.K. Rishi, X. Zhang, A. Wali, Targeting of apoptosis signaling pathways and [97] Z. Zeng, I.J. Samudio, W. Zhang, Z. Estrov, H. Pelicano, D. Harris, O. Frolova, N. Hail
their mediators for cancer therapy, Pharmaceutical Perspectives of Cancer Jr., W. Chen, S.M. Kornblau, P. Huang, Y. Lu, G.B. Mills, M. Andreeff, M. Konopleva,
Therapeutics (2009) 149188. Simultaneous inhibition of PDK1/AKTand Fms-like tyrosine kinase 3 signaling by
[71] E. Lomonosova, G. Chinnadurai, BH3-only proteins in apoptosis and beyond: an a small-molecule KP372-1inducesmitochondrial dysfunction and apoptosis in
overview, Oncogene 27 (Suppl 1) (2008) S2S19. acute myelogenous leukemia, Cancer Res. 66 (2006) 37373746.
[72] P.F. Dijkers, K.U. Birkenkamp, E.W. Lam, N.S. Thomas, J.W. Lammers, L. Koenderman, [98] H. Zhang, Y. Dong, H. Zhao, J.D. Brooks, L. Hawthorn, N. Nowak, J.R. Marshall, A.C.
P.J. Coffer, FKHR-L1 can act as a critical effector of cell death induced by cytokine Gao, C. Ip, Microarray data mining for potential selenium targets in chemopre-
withdrawal: protein kinase B-enhanced cell survival through maintenance of vention of prostate cancer, Cancer Genomics Proteomics 2 (2005) 97114.
mitochondrial integrity, J. Cell Biol. 156 (2002) 531542. [99] H. Zhang, J. Fang, D. Yao, Y. Wu, C. Ip, Y. Dong, Activation of FOXO1 is critical for
[73] J. Gilley, P.J. Coffer, J. Ham, FOXO transcription factors directly activate bim gene the anticancer effect of methylseleninic acid in prostate cancer cells, Prostate 70
expression and promote apoptosis in sympathetic neurons, J. Cell Biol. 162 (2010) 12651273.
(2003) 613622. [100] P. Li, H. Lee, S. Guo, T.G. Unterman, G. Jenster, W. Bai, AKT-independent protection of
[74] P.F. Dijkers, R.H. Medema, J.W. Lammers, L. Koenderman, P.J. Coffer, Expression prostate cancer cells from apoptosis mediated through complex formation between
of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead the androgen receptor and FKHR, Mol. Cell. Biol. 23 (2003) 104118.
transcription factor FKHR-L1, Curr. Biol. 10 (2000) 12011204. [101] W. Filipowicz, S.N. Bhattacharyya, N. Sonenberg, Mechanisms of post-transcriptional
[75] M. Stahl, P.F. Dijkers, G.J. Kops, S.M. Lens, P.J. Coffer, B.M. Burgering, R.H. regulation by microRNAs: are the answers in sight? Nat. Rev. Genet. 9 (2008)
Medema, The forkhead transcription factor FoxO regulates transcription of 102114.
p27Kip1 and Bim in response to IL-2, J. Immunol. 168 (2002) 50245031. [102] D.P. Bartel, MicroRNAs: genomics, biogenesis, mechanism, and function, Cell 116
[76] A. Essa, S. Fernandez de Mattos, Y.A. Hassen, I. Soeiro, G.J. Mufti, N.S. Thomas, (2004) 281297.
R.H. Medema, E.W. Lam, Direct transcriptional regulation of Bim by FoxO3a [103] S. Grifths-Jones, H.K. Saini, S. van Dongen, A.J. Enright, miRBase: tools for
mediates STI571-induced apoptosis in Bcr-Abl-expressing cells, Oncogene 24 microRNA genomics, Nucleic Acids Res. 36 (2008) D154D158.
(2005) 23172329. [104] S. Grifths-Jones, R.J. Grocock, S. van Dongen, A. Bateman, A.J. Enright, miRBase:
[77] Z. Jagani, K. Song, J.L. Kutok, M.R. Dewar, A. Melet, T. Santos, A. Grassian, S. microRNA sequences, targets and gene nomenclature, Nucleic Acids Res. 34
Ghaffari, C. Wu, H. Yeckes-Rodin, R. Ren, K. Miller, R. Khosravi-Far, Proteasome (2006) D140D144.
inhibition causes regression of leukemia and abrogates BCR-ABL-induced [105] S. Grifths-Jones, The microRNA Registry, Nucleic Acids Res. 32 (2004)
evasion of apoptosis in part through regulation of forkhead tumor suppressors, D109D111.
Cancer Res. 69 (2009) 65466555. [106] K.E. Allen, G.J. Weiss, Resistance may not be Futile: microRNA biomarkers for
[78] R. Deng, J. Tang, B.F. Xie, G.K. Feng, Y.H. Huang, Z.C. Liu, X.F. Zhu, SYUNZ-16, a chemoresistance and potential therapeutics, Mol. Cancer Ther. 9 (2010) 31263136.
newly synthesized alkannin derivative, induces tumor cells apoptosis and [107] F. Meng, R. Henson, H. Wehbe-Janek, K. Ghoshal, S.T. Jacob, T. Patel, MicroRNA-
suppresses tumor growth through inhibition of PKB/AKT kinase activity and 21 regulates expression of the PTEN tumor suppressor gene in human
blockade of AKT/FOXO signal pathway, Int. J. Cancer 127 (2010) 220229. hepatocellular cancer, Gastroenterology 133 (2007) 647658.
[79] A. Sunters, S. Fernndez de Mattos, M. Stahl, J.J. Brosens, G. Zoumpoulidou, C.A. [108] Z. Chun-Zhi, H. Lei, Z. An-Ling, F. Yan-Chao, Y. Xiao, W. Guang-Xiu, J. Zhi-Fan, P.
Saunders, P.J. Coffer, R.H. Medema, R.C. Coombes, E.W. Lam, FoxO3a transcrip- Pei-Yu, Z. Qing-Yu, K. Chun-Sheng, MicroRNA-221 and microRNA-222 regulate
tional regulation of Bim controls apoptosis in paclitaxel-treated breast cancer gastric carcinoma cell proliferation and radioresistance by targeting PTEN, BMC
cell lines, J. Biol. Chem. 278 (2003) 4979549805. Cancer 12 (2010)8 10:367.
[80] J.E. Chipuk, D.R. Green, PUMA cooperates with direct activator proteins to [109] J.T. Huse, C. Brennan, D. Hambardzumyan, B. Wee, J. Pena, S.H. Rouhanifard, C.
promote mitochondrial outer membrane permeabilization and apoptosis, Cell Sohn-Lee, C. le Sage, R. Agami, T. Tuschl, E.C. Holland, The PTEN-regulating
Cycle 8 (2009) 26922696. microRNA miR-26a is amplied in high-grade glioma and facilitates gliomagen-
[81] H. You, M. Pellegrini, K. Tsuchihara, K. Yamamoto, G. Hacker, M. Erlacher, A. esis in vivo, Genes Dev. 23 (2009) 13271337.
Villunger, T.W. Mak, FOXO3a-dependent regulation of Puma in response to [110] M. Kato, S. Putta, M. Wang, H. Yuan, L. Lanting, I. Nair, A. Gunn, Y. Nakagawa, H.
cytokine/growth factor withdrawal, J. Exp. Med. 203 (2006) 16571663. Shimano, I. Todorov, J.J. Rossi, R. Natarajan, L. Lanting, TGF-beta activates Akt
[82] F. Chiacchiera, C. Simone, The AMPK-FoxO3A axis as a target for cancer kinase through a microRNA-dependent amplifying circuit targeting PTEN, Nat.
treatment, Cell Cycle 9 (2010) 10911096. Cell Biol. 11 (2009) 881889.
[83] T.T. Tang, D. Dowbenko, A. Jackson, L. Toney, D.A. Lewin, A.L. Dent, L.A. Lasky, The [111] C. Xiao, L. Srinivasan, D.P. Calado, H.C. Patterson, B. Zhang, J. Wang, J.M.
forkhead transcription factor AFX activates apoptosis by induction of the BCL-6 Henderson, J.L. Kutok, K. Rajewsky, Lymphoproliferative disease and autoim-
transcriptional repressor, J. Biol. Chem. 277 (2002) 1425514265. munity in mice with increased miR-17-92 expression in lymphocytes, Nat.
[84] A. Brunet, A. Bonni, M.J. Zigmond, M.Z. Lin, P. Juo, L.S. Hu, M.J. Anderson, K.C. Immunol. 9 (2008) 405414.
Arden, J. Blenis, M.E. Greenberg, Akt promotes cell survival by phosphorylating [112] H. Yang, W. Kong, L. He, J.J. Zhao, J.D. O'Donnell, J. Wang, R.M. Wenham, D.
and inhibiting a Forkhead transcription factor, Cell 96 (1999) 857868. Coppola, P.A. Kruk, S.V. Nicosia, J.Q. Cheng, MicroRNA expression proling in
1986 X. Zhang et al. / Biochimica et Biophysica Acta 1813 (2011) 19781986

human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by W.W. Ma, A.A. Adjei, Novel agents on the horizon for cancer therapy, CA Cancer
targeting PTEN, Cancer Res. 68 (2008) 425433. J. Clin. 59 (2009) 111137.
[113] N. Bar, R. Dikstein, miR-22 forms a regulatory loop in PTEN/AKT pathway and [125] S.R. Vink, W.J. van Blitterswijk, J.H. Schellens, M. Verheij, Rationale and clinical
modulates signaling kinetics, PLoS ONE 5 (2010) e10859. application of alkylphospholipid analogues in combination with radiotherapy,
[114] M.G. Pezzolesi, P. Platzer, K.A. Waite, C. Eng, Differential expression of PTEN- Cancer Treat. Rev. 33 (2007) 191202.
targeting microRNAs miR-19a and miR-21 in Cowden syndrome, Am. J. Hum. [126] S.F. Barnett, M.T. Bilodeau, C.W. Lindsley, The Akt/PKB family of protein kinases:
Genet. 82 (2008) 11411149. a review of small molecule inhibitors and progress towards target validation,
[115] L. Liu, Y. Jiang, H. Zhang, A.R. Greenlee, Z. Han, Overexpressed miR-494 down- Curr. Top. Med. Chem. 5 (2005) 109125.
regulates PTEN gene expression in cells transformed by anti-benzo(a)pyrene- [127] S.K. Pal, K. Reckamp, H. Yu, R.A. Figlin, Akt inhibitors in clinical development for
trans-7,8-dihydrodiol-9,10-epoxide, Life Sci. 86 (2010) 192198. the treatment of cancer, Expert Opin. Investig. Drugs 19 (2010) 13551366.
[116] L. He, Posttranscriptional regulation of PTEN dosage by noncoding RNAs, Sci. [128] S.B. Kondapaka, S.S. Singh, G.P. Dasmahapatra, E.A. Sausville, K.K. Roy, Perifosine,
Signal. 3 (2010) e39. a novel alkylphospholipid, inhibits protein kinase B Activation, Mol. Cancer Ther.
[117] Y. Ren, X. Zhou, M. Mei, X.B. Yuan, X.B. Yuan, L. Han, G.X. Wang, Z.F. Jia, P. Xu, 2 (2003) 10931103.
P.Y. Pu, C.S. Kang, MicroRNA-21 inhibitor sensitizes human glioblastoma cells [129] I.R. Konings, J. Verweij, E.A. Wiemer, S. Sleijfer, The applicability of mTOR
U251 (PTEN-mutant) and LN229 (PTEN-wild type) to taxol, BMC Cancer 10 inhibition in solid tumors, Curr. Cancer Drug Targets 9 (2009) 439450.
(2010) 27. [130] S. Di Cosimo, J. Baselga, Targeted therapies in breast cancer: where are we now?
[118] A. Ernst, B. Campos, J. Meier, F. Devens, F. Liesenberg, M. Wolter, G. Reifenberger, Eur. J. Cancer 44 (2008) 27812790.
C. Herold-Mende, P. Lichter, B. Radlwimmer, De-repression of CTGF via the miR- [131] E. Raymond, J. Alexandre, S. Faivre, K. Vera, E. Materman, J. Boni, C. Leister, J.
17-92 cluster upon differentiation of human glioblastoma spheroid cultures, Korth-Bradley, A. Hanauske, J.P. Armand, Safety and pharmacokinetics of
Oncogene 29 (2010) 34113422. escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR
[119] J.K. Park, E.J. Lee, C. Esau, T.D. Schmittgen, Antisense inhibition of microRNA-21 inhibitor, in patients with cancer, J. Clin. Oncol. 22 (2004) 23362347.
or -221 arrests cell cycle, induces apoptosis, and sensitizes the effects of [132] K.E. O'Reilly, F. Rojo, Q.B. She, D. Solit, G.B. Mills, D. Smith, H. Lane, F. Hofmann, D.J.
gemcitabine in pancreatic adenocarcinoma, Pancreas 38 (2009) e190e199. Hicklin, D.L. Ludwig, J. Baselga, N. Rosen, mTOR inhibition induces upstream receptor
[120] J. Li, H. Huang, L. Sun, M. Yang, C. Pan, W. Chen, D. Wu, Z. Lin, C. Zeng, Y. Yao, P. tyrosine kinase signaling and activates Akt, Cancer Res. 66 (2006) 15001508.
Zhang, E. Song, MiR-21 indicates poor prognosis in tongue squamous cell [133] G. Hudes, M. Carducci, P. Tomczak, J. Dutcher, R. Figlin, A. Kapoor, E.
carcinomas as an apoptosis inhibitor, Clin. Cancer Res. 15 (2009) 39984008. Staroslawska, J. Sosman, D. McDermott, I. Bodrogi, Z. Kovacevic, V. Lesovoy,
[121] C. Garcia-Echeverria, W.R. Sellers, Drug discovery approaches targeting the PI3K/ I.G. Schmidt-Wolf, O. Barbarash, E. Gokmen, T. O'Toole, S. Lustgarten, L. Moore,
Akt pathway in cancer, Oncogene 27 (2008) 55115526. R.J. Motzer, Global ARCC trial: temsirolimus, interferon alfa, or both for advanced
[122] S. Zhang, D. Yu, PI(3)king apart PTEN's role in cancer, Clin. Cancer Res. 16 (2010) renal-cell carcinoma, N. Engl. J. Med. 356 (2007) 22712281.
43254330. [134] R.J. Motzer, B. Escudier, S. Oudard, T.E. Hutson, C. Porta, S. Bracarda, V. Grnwald,
[123] N.T. Ihle, G. Powis, Take your PIK: phosphatidylinositol 3-kinase inhibitors race J.A. Thompson, R.A. Figlin, N. Hollaender, G. Urbanowitz, W.J. Berg, A. Kay, D.
through the clinic and toward cancer therapy, Mol. Cancer Ther. 8 (2009) 19. Lebwohl, A. Ravaud, RECORD-1 study group: efcacy of everolimus in advanced
[124] N.T. Ihle, G. Powis, Inhibitors of phosphatidylinositol-3-kinase in cancer therapy, renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III
Mol. Aspects Med. 31 (2010) 135144; trial, Lancet 372 (2008) 449456.

You might also like