You are on page 1of 10

J Chem Biol (2008) 1:27–36

DOI 10.1007/s12154-008-0003-5

REVIEW

Rapamycin and mTOR kinase inhibitors


Lisa M. Ballou & Richard Z. Lin

Received: 19 February 2008 / Accepted: 11 March 2008 / Published online: 15 May 2008
# Springer-Verlag 2008

Abstract Mammalian target of rapamycin (mTOR) is a Keywords mTOR . Rictor . Raptor . Rapamycin .
protein kinase that controls cell growth, proliferation, and Phosphatidylinositol 3-kinase . Akt
survival. mTOR signaling is often upregulated in cancer
and there is great interest in developing drugs that target
this enzyme. Rapamycin and its analogs bind to a domain Phosphatidylinositol 3-kinase-related kinases
separate from the catalytic site to block a subset of mTOR
functions. These drugs are extremely selective for mTOR Mammalian target of rapamycin (mTOR) is a serine/
and are already in clinical use for treating cancers, but they threonine protein kinase whose catalytic domain most
could potentially activate an mTOR-dependent survival closely resembles the one found in lipid kinases of the
pathway that could lead to treatment failure. By contrast, phosphatidylinositol (PI) 3-kinase (PI3K) family. PI3Ks
small molecules that compete with ATP in the catalytic site phosphorylate inositol phospholipids on the 3′-hydroxyl of
would inhibit all of the kinase-dependent functions of the inositol ring and are divided into three classes based on
mTOR without activating the survival pathway. Several structural features and substrate specificity (reviewed in
non-selective mTOR kinase inhibitors have been described Ref. [1]). Class I PI3Ks consist of four different catalytic
and here we review their chemical and cellular properties. subunits (p110α, p110β, p110δ and p110γ) that form
Further development of selective mTOR kinase inhibitors heterodimers with a variety of regulatory subunits. These
holds the promise of yielding potent anticancer drugs with a enzymes preferentially phosphorylate PI 4,5-bisphosphate
novel mechanism of action. (PI(4,5)P2) in vivo. Crystal structures of p110γ bound to
ATP and to various inhibitors have been instrumental in
determining structure-activity relationships for PI3K inhib-
L. M. Ballou : R. Z. Lin
Department of Medicine, Stony Brook University,
itors (discussed below). The structure of p110α bound to a
Stony Brook, NY 11794, USA fragment of one of its regulatory subunits was also solved
recently [2]. Class II PI3Ks (PI3K C2α, C2β and C2γ)
R. Z. Lin phosphorylate PI and PI(4)P in vitro and contain a C2
Department of Physiology and Biophysics
and the Institute of Molecular Cardiology,
homology domain at the C terminus that is not found in
Stony Brook University, other PI3Ks. The class III PI3K Vps34 only phosphorylates
Stony Brook, NY 11794, USA PI and may play a role in the regulation of mTOR by amino
acids [3]. Type III PI 4-kinases (PI4Ks) phosphorylate the
R. Z. Lin
Department of Veterans Affairs Medical Center,
4′-hydroxyl of the inositol ring and are closely related to
Northport, NY 11768, USA PI3Ks [1].
mTOR belongs to the family of PI3K-related protein
R. Z. Lin (*) kinases (PIKKs) that includes ataxia–telangiectasia mutated
Department of Medicine, Division of Hematology/Oncology,
Stony Brook University,
(ATM), ATM- and Rad3-related (ATR), DNA-dependent
Stony Brook, NY 11794-8151, USA protein kinase (DNA-PK) and suppressor of morphogenesis
e-mail: richard.lin@sunysb.edu in genitalia-1 (SMG-1; reviewed in Refs. [1, 4]). These are
28 J Chem Biol (2008) 1:27–36

large proteins (~300–500 kDa) that contain a conserved phosphorylate and suppress the activity of FOXO proteins.
kinase catalytic domain together with other regions that can These results suggest that mTORC2 is necessary for Akt-
include HEAT repeats and FAT and FATC domains. mTOR FOXO survival signaling. Additional studies using cells
also has an FRB (FKBP12/rapamycin-binding) domain that derived from these animals might shed more light on the
binds the drug rapamycin in complex with its intracellular specific cellular processes regulated by the distinct mTOR
receptor protein FKBP12. ATM, ATR, DNA-PK, and complexes. These studies will also be facilitated if small
SMG-1 are involved in DNA and mRNA surveillance and molecules that can specifically inhibit mTORC2 activity
repair pathways. By contrast, mTOR integrates growth become available.
factor-activated signals with permissive signals in the A number of different signaling pathways regulate
presence of sufficient nutrients and energy to promote cell mTORC1 activity, and the best characterized positive
growth, proliferation, and survival. mTOR signaling is effector is the growth factor/PI3K/Akt pathway (Fig. 1).
upregulated in many cancers and some benign growth or Akt plays an unusual role in mTOR signaling because it
proliferative disorders. Therefore, drugs that target mTOR acts upstream of mTORC1 and downstream of mTORC2.
activity are expected to be useful therapies for these Akt controls mTORC1 in part through tuberous sclerosis
conditions. complex (TSC) 2, a protein that has GTPase-activating
protein (GAP) activity toward Rheb, a small GTP-binding
protein related to Ras [15]. TSC2 forms a tight complex
mTOR signal transduction and cellular functions with TSC1. The GAP activity of the TSC1/TSC2 hetero-
dimer converts Rheb to an inactive GDP-bound state and
The discovery of mTOR and the understanding of its thus suppresses mTOR activity [16]. In the presence of
biological functions were greatly facilitated by the use of growth factors that activate Akt, Akt phosphorylates TSC2
rapamycin, which inhibits some of the functions of mTOR. at Ser939 and Thr1462 and inhibits its GAP activity [17].
Until recently, rapamycin sensitivity was the major criterion This allows Rheb to maintain an active GTP-bound form
used to identify mTOR-controlled events. However, it is that activates mTORC1 and increases signaling to S6K and
now known that mTOR binds to different regulatory 4EBP1.
subunits to produce complexes with distinct signaling An energy-sensing pathway also regulates mTOR activ-
functions and rapamycin sensitivity [5, 6]. The mTORC1 ity. Inhibition of mTOR in response to low intracellular
complex (containing mTOR, Raptor and mLST8) phos- energy levels is mediated by AMP-activated kinase
phorylates ribosomal protein S6 kinase (S6K) at Thr389 (AMPK) and its activator, the protein kinase LKB1 [18].
and the translation repressor 4EBP1 and is rapamycin Under conditions in which intracellular ATP is depleted and
sensitive. Biological processes regulated by mTORC1 the level of AMP is increased, AMP binds to AMPK and
include translation, ribosome biogenesis, autophagy, glu- allows LKB1 to phosphorylate Thr172 on the catalytic α
cose metabolism, and the cellular response to hypoxia [7– subunit to activate AMPK [19]. AMPK then phosphor-
9]. The mTORC2 complex (containing mTOR, Rictor, ylates TSC2 on Ser1345, which primes TSC2 for subse-
mLST8, and mSin1) phosphorylates the protein kinase Akt quent phosphorylation of Ser1341 and Ser1337 by
at Ser473 and is insensitive to rapamycin. In comparison to glycogen synthase kinase 3 [20]. Together, these modifica-
mTORC1, the biological function of mTORC2 is less clear. tions are thought to enhance the GAP activity of TSC2,
However, available evidence suggests that this mTOR inactivate Rheb, and turn off mTORC1 signaling (Fig. 1).
complex controls cell survival and organization of the actin Hypoxia and low amino acid levels also negatively
cytoskeleton. regulate mTOR activity. Signaling pathways that mediate
The physiological importance of the mTOR complexes the mTOR response to these stimuli are less well
is underscored by genetic ablation of their molecular characterized. The class III PI3K, hVps34, seems to play
components in mouse models. Mouse embryos lacking an important role in transducing the amino acid signal to
mTOR die at E5.5–6.5 days [10, 11]. Ablation of Raptor to mTOR [3, 21]. Evidence indicates that the hypoxia signal is
disrupt mTORC1 is equally lethal at approximately mediated by a pathway from REDD1 (regulated in
E6.5 days [12]. Mouse knockouts of Rictor or mSIN1 development and DNA damage responses) to TSC1/2 and
leading to disruption of mTORC2 are also embryonic lethal then to mTOR [22].
[13, 14]. Not surprisingly, embryos lacking mLST8 also do Recent studies indicate that feedback from the
not survive [12]. Although Akt Ser473 phosphorylation mTORC1/S6K pathway inhibits growth factor signaling to
was blocked in cells isolated from Rictor−/− and mSIN1−/− PI3K (Fig. 1). TSC1−/− or TSC2−/− cells have abnormally
embryos, phosphorylation of several Akt substrates was not low Akt activity due to hyperactivation of mTORC1/S6K
inhibited, with the exception of FOXO transcription factors [23–25]. Conversely, S6K1−/− cells are hypersensitive to
[12, 13]. Part of the prosurvival function of Akt is to insulin activation of PI3K signaling [26, 27]. Treatment of
J Chem Biol (2008) 1:27–36 29

Fig. 1 mTOR signaling. Growth factor activation of PI3K causes during hypoxia due to activation of TSC1/TSC2 by REDD1. Rheb-
increased production of PI 3,4,5-trisphosphate (PI(3,4,5)P3). Akt is GTP binds to and activates mTORC1. mTORC1 phosphorylates the
activated upon binding to PI(3,4,5)P3 and phosphorylation of Thr308 translation repressor 4EBP1 and S6K at Thr389, leading to its
by phosphoinositide-dependent protein kinase 1 (PDK1) and Ser473 activation. Inhibitors that target the mTOR kinase domain block both
by mTORC2. The TSC1/TSC2 complex inhibits mTORC1 by mTORC1 and mTORC2, whereas rapamycin only inhibits mTORC1.
stimulating the GTPase activity of Rheb, converting it to the inactive Upregulation of S6K in cells with hyperactive mTORC1 signaling has
GDP-bound state. Akt phosphorylation of TSC2 inhibits the TSC1/ a feedback inhibitory effect on Akt. Therefore, treatment with
TSC2 complex and allows Rheb to remain in the active GTP-bound rapamycin leading to inhibition of S6K can upregulate Akt and
form. Phosphorylation by AMPK activates TSC2 and opposes the Akt enhance cell survival. In contrast, mTOR kinase inhibitors block both
signal. In energy-poor conditions, increased AMP levels allow LKB1 S6K and Akt activation and should not enhance cell survival. It is not
to phosphorylate and activate AMPK. Decreased signaling through clear how TSC1/TSC2 and Rheb regulate the activity of mTORC2
Vps34 in the presence of low amino acid levels to inhibit mTOR [24]
might be independent of TSC1/TSC2 and Rheb. mTOR is inhibited

some cancer cells with rapamycin upregulates Akt, which considered to be an undesirable immunosuppressive effect,
can enhance survival under conditions that usually induce which subsequently led to its development as a clinically
apoptosis [23, 28–30]. As a result, there is concern that useful drug. The immunosuppressant FK506 (tacrolimus)
reactivation of Akt in tumors following rapamycin treat- and rapamycin have similar chemical structures and bind to
ment could lead to resistance to other chemotherapeutic the same intracellular receptor, FKBP12. Even though
agents. It has been proposed that small molecule inhibitors FK506 and rapamycin bind to the same protein, they have
that target the kinase domain of mTOR will display broader different mechanisms of action in cells. FK506 inhibits
antitumor activity than rapamycin because they should not T cell proliferation by blocking the Ca2+/calcineurin-
reactivate Akt [7, 31, 32]. Theoretically, a combination of dependent transcriptional activation of genes required for
rapamycin plus an inhibitor of Akt or PI3K would have the growth, whereas rapamycin interferes with growth-
same effect. promoting cytokine signaling [33]. Interleukin-2 (IL-2)
induced S6K activation in a T cell line was extremely
sensitive to rapamycin inhibition (IC50 = 0.05 nM) [34]. By
Rapamycin and rapalogs contrast, mTORC1 kinase activity in vitro was much less
sensitive to FKBP12/rapamycin [6]. The reason for this
Rapamycin (sirolimus) is a macrocyclic antibiotic produced apparent difference in mTORC1 sensitivity to FKBP12/
by the bacterium Streptomyces hygroscopicus found in the rapamycin inhibition in vivo and in vitro is not understood.
soil of Easter Island. Rapamycin was discovered as a potent In addition to rapamycin, three rapamycin analogs
antifungal agent, but it also exhibited what was at first (rapalogs) are now in use in humans (Fig. 2). CCI779
30 J Chem Biol (2008) 1:27–36

Fig. 2 Chemical structure of rapamycin, rapalogs and mTOR kinase inhibitors. Rapalogs have the indicated O-substitutions at the C-40 position
of rapamycin (underline)

(temsirolimus) is a dihydroxymethyl propionic acid ester affinity [37]. Using this information and a solution NMR
prodrug of rapamycin. This modification makes the structure of the FRB domain, Leone et al. virtually screened
compound water soluble and thus it can be administered a chemical library and discovered additional small mole-
intravenously. Upon injection, CCI779 is rapidly converted cules that bind to the FRB domain in the absence of
to rapamycin, which is probably responsible for most, if not FKBP12 [38]. However, these molecules were generally
all, of its pharmacological effects. RAD001 (everolimus) ineffective at inhibiting mTOR kinase activity [38].
has a O-(2-hydroxyethyl) chain substitution at position C- Another group used high-throughput screening to identify
40 and AP23573 (deforolimus) has a phosphine oxide 4-[6-{[(1S,2R)-2-(benzyloxy)cyclopentyl]acetyl}-4-(2-
substitution at the same position of the lactone ring of thienyl)pyridin-2-yl]-4-oxobutanoic acid (HTS-1) that
rapamycin. Rapalogs have the same mechanism of action as binds to the FRB domain in the low micromolar range
rapamycin. They bind to FKBP12 and interfere with the [39]. Unfortunately, they did not show if the compound
FRB domain of mTOR. inhibits mTOR kinase activity in vitro. Interestingly, these
Unlike kinase inhibitors that target the catalytic ATP- investigators also found that phosphatidic acid, which
binding site, these compounds are highly specific for activates mTOR [40], interacts with the FRB domain in
mTORC1. However, the exact mechanism of how the the same binding pocket as rapamycin. It remains unclear
interaction with the FRB domain leads to inhibition of why binding of phosphatidic acid activates mTOR whereas
mTORC1 remains unclear. FKBP12/rapamycin inhibits rapamycin binding has the opposite effect. Interestingly,
mTOR autophosphorylation and phosphorylation of mTOR with a mutation in the FRB domain that renders it
4EBP1 in vitro, suggesting that changes in the FRB domain rapamycin resistant exhibits decreased catalytic activity
might exert an allosteric influence on the catalytic domain [41], supporting the hypothesis that this domain modulates
[35]. It was initially thought that rapamycin interacts with the catalytic domain. Future efforts may identify novel
the FRB domain only after binding to FKBP12. The X-ray compounds that bind to this area and potently inhibit
crystal structure of the FKBP12-rapamycin-FRB complex mTOR.
shows that rapamycin occupies two different hydrophobic Rapamycin has also been proposed to inhibit mTOR by
binding pockets in FKBP12 and the FRB domain simulta- destabilizing the mTOR-Raptor complex [42]. Interestingly,
neously and brings the two proteins close together, but mTOR has been shown to be inhibited by S-trans,trans-
there are relatively few interactions between the proteins farnesyl thiosalicylic acid (FTS), a compound that resem-
[36]. Subsequently, rapamycin was shown to bind to the bles farnesylcysteine found in Ras family members and
FRB domain without FKBP12, although with a lower other proteins. FTS dislodges farnesylated Ras proteins
J Chem Biol (2008) 1:27–36 31

from cell membranes by competing for their membrane RAD001 and rapamycin are also being tested in clinical
binding sites, thus facilitating their degradation [43]. FTS studies for treatment of both hematologic and solid
inhibits mTORC1 activity in intact cells and cell extracts by malignancies. Aside from a possible direct antiproliferative
promoting the dissociation of Raptor [44]. It should be effect on tumor cells, experiments in a mouse model of
noted that although rapamycin does not exert acute effects metastatic cancer suggest that rapamycin limits the growth
on mTORC2, long-term treatment of some cell types with of solid tumors by blocking angiogenesis [50]. Intravenous
the drug has been shown to prevent mTORC2 assembly, CCI779 and AP23573 are administered on intermittent
thus inhibiting Akt [45]. This mechanism was proposed to weekly schedules. Given in this fashion, these drugs do not
account for hyperlipidemia that is commonly seen in appear to cause immunosuppression. This is surprising
patients treated with the drug and may also account for because rapalogs have long half-lives (RAD001, 24–35 h;
hyperglycemia, which is another common side-effect. CCI779, 13–25 h; AP23573, 45–74 h). In a rat model, a
A recent study provided evidence that the intracellular single dose of RAD001 suppressed mTOR signaling
protein FKBP38 is an endogenous inhibitor of mTORC1 [46]. (measured by S6K activity and 4EBP1 Thr70 phosphory-
FKBP38 bound to the FRB domain of mTOR and inhibited lation) in peripheral blood mononuclear cells for longer
its activity in vitro, similar to FKPB12/rapamycin. Further- than 72 h [51]. A possible explanation for this discrepancy
more, the FKBP12/rapamycin complex competed with is that the immunosuppressive effect of rapamycin is
FKBP38 for binding to mTOR in vitro, and cells treated mediated by other effectors that are not inhibited by the
with rapamycin had reduced association between FKBP38 drug concentrations achieved during these treatments. S6K
and mTOR [46]. Upon growth factor stimulation, Rheb– activity in blood cells or tissue might not be a good
GTP bound strongly to FKBP38 and displaced it from indicator of pharmacodynamic effectiveness because this
mTOR, thus activating the enzyme. These data raise the enzyme is exquisitely sensitive to rapamycin inhibition.
possibility that the FKBP12/rapamycin complex might Rapamycin-eluting coronary artery stents were approved
inhibit mTOR because it cannot be displaced by GTP–Rheb. for use by the U.S. FDA in 2003. Rapamycin inhibits
vascular smooth muscle cell migration and proliferation and
attenuates reocclusion of coronary arteries following an-
Clinical uses of rapamycin and rapalogs gioplasty. In a randomized, double-blind trial with 1058
patients, patients treated with rapamycin-eluting stents had
Rapamycin is an oral drug and its bioavailability is low. a restenosis rate of 8.6% vs. 21% in patients treated with a
RAD001 is also an oral compound and an often-cited standard non-drug-eluting stent [52]. It is conceivable that
rationale for its development is that it has improved the antiproliferative effect of mTOR inhibitors might be
bioavailability. However, in a clinical pharmacokinetic utilized to treat other non-malignant proliferative disorders
study, RAD001 was found to also have a relatively low such as polycystic kidney disease [53].
bioavailability (~15%) [47]. Rapamycin and RAD001 are
both approved for use as immunosuppressants for organ
transplant patients. Due to their pharmacokinetic properties, mTOR kinase inhibitors
drug level monitoring of these drugs is necessary and they
require daily dosing. Rapamycin inhibits IL-2-induced T A small molecule designed to compete with ATP in the
cell proliferation [34] that is due at least in part to a specific catalytic site of mTOR would be expected to inhibit all of
down-regulation of ribosomal protein mRNA translation the kinase-dependent functions of mTORC1 and mTORC2,
[48]. It is believed that the immunosuppressive effect of unlike rapalogs that only target mTORC1. Most if not all of
RAD001 is caused by a similar mechanism of action. Post- the non-rapalog mTOR inhibitors described to date in the
transplant malignancy is a serious complication of solid scientific literature were developed to inhibit other
organ transplantation. In contrast with calcineurin inhibitors enzymes, especially class I PI3Ks. Because PI3K regulates
such as FK506, the use of mTOR inhibitors as immuno- mTOR activity (see Fig. 1), inhibitors that target both
suppressants has the added benefit of being antitumori- enzymes are generally not useful as research tools to study
genic. Indeed, rapamycin is now the established treatment mTOR regulation or function. However, drugs that are dual
for post-transplant Kaposi’s sarcoma in patients who were PI3K/mTOR inhibitors might have a therapeutic advantage
on other immunosuppressive regimens [49]. over single-target inhibitors in certain disease settings.
CCI779 and AP23573 can be given intravenously and Wortmannin (Fig. 2) is a toxic steroidal furan that is
are being used as anticancer drugs. CCI779 was recently produced by various fungi including Penicillium wortmanni
approved by the U.S. Food and Drug Administration (FDA) [54]. The first kinase shown to be targeted by wortmannin
for treatment of renal cell carcinoma. AP23573 is in clinical was smooth muscle myosin light chain kinase (IC50 =
trials for treating a variety of cancers, including sarcomas. 0.17 μM). Inhibition was irreversible and the enzyme was
32 J Chem Biol (2008) 1:27–36

partially protected by incubation in the presence of ATP most likely by acting as low affinity ATP analogs.
[55]. Later studies exploring wortmannin inhibition of Theophylline (5 mM) strongly inhibits mTOR kinase
agonist-induced responses in neutrophils and basophils led activity in vitro and blocked insulin activation of Akt in
to the discovery that the compound is a potent inhibitor of 3T3-L1 adipocytes [73]. However, since theophylline and
class I PI3Ks (IC50 ~1–3 nM) [56, 57]. Biochemical caffeine also target class I PI3Ks with IC50 values ranging
analyses and the X-ray crystallographic structure of from 75 μM to 1 mM [74], effects on Akt cannot be
wortmannin bound to p110γ showed that the mechanism attributed solely to mTOR inhibition. Caffeine has been
of inhibition is two-fold. First, wortmannin binds with high used extensively in cell-based experiments to investigate
affinity to the ATP-binding site to block substrate binding. cell cycle checkpoints in G1/S and G2/M that are induced
Second, the ε-amino group of a lysine in the active site by DNA damage. These studies led to the discovery that
forms a covalent bond with carbon-20 of the wortmannin caffeine inhibits ATM (IC50 ~0.2 mM ), ATR (IC50 ~
furan ring to irreversibly inhibit the enzyme [58, 59]. The 1.1 mM), SMG-1 (IC50 =0.3 mM), and mTOR (IC50 ~0.2–
active site lysine (Lys802 in p110α and Lys833 in p110γ) 0.4 mM ). There is disagreement about whether DNA-PK is
is critical for catalysis and is highly conserved in lipid and also inhibited by caffeine [41, 68, 75–77]. Interestingly,
protein kinases. Wortmannin also targets class II PI3K C2β TORC1 signaling is an important target for caffeine in
(IC50 =1.6 nM), the human class III PI3K hVps34p (IC50 = yeast, and several caffeine-resistant mutants of the TOR1
2.5 nM), type III PI4Ks (IC50 =50–300 nM) and polo-like protein have been identified [78]. These proteins contain
protein kinases that regulate mitosis (IC50 =24–48 nM) [60– two mutations: one in the kinase domain that probably
64]. Not surprisingly, considering the similarity between the impairs binding to both caffeine and ATP, resulting in
PI3K and PIKK catalytic domains, wortmannin also greatly reduced kinase activity, and a second mutation in
irreversibly inhibits PIKK family members. DNA-PK is the FRB that increases the kinase activity. The double
the most sensitive (IC50 =16 nM), followed by SMG-1 mutants exhibit strong caffeine resistance and kinase
(IC50 ∼ 60 nM), ATM (IC50 ~100–150 nM), mTOR (IC50 ~ activity that is high enough to support TORC1 function in
200 nM) and ATR (IC50 =1.8 μM) [65–68]. Wortmannin vivo. By contrast, mutations in the FRB that cause
forms a covalent bond with mTOR, presumably at Lys2187 rapamycin resistance decrease TOR1 kinase activity, again
in the ATP-binding site [65]. Based on its potency and highlighting the idea that this domain regulates TOR1
selectivity, wortmannin at 100 nM has been recommended activity in multiple ways. Caffeine and theophylline also
for use in cells to assess the role of PI3K [69], although block the activity of class II PI3K C2α (IC50 ~0.4 mM) [74].
effects on mTOR at this concentration should be consid- The field of PI3K signaling was revolutionized by the
ered. The use of wortmannin as a therapeutic agent is introduction of LY294002 (2-(4-morpholinyl)-8-phenyl-
limited by its toxicity and instability in biological solutions. 4H-1-benzopyran-4-one) (Fig. 2), a synthetic PI3K inhib-
The wortmannin derivative PX-866 is more stable and less itor whose structure is based on the naturally occurring
toxic than the parent compound and exhibits antitumor flavonoid quercetin [79]. LY294002 behaves as a compet-
activity in mice, but it was reported to be inactive against itive inhibitor for the ATP-binding site and is much less
mTOR at concentrations up to 10 μM [70]. potent against class I PI3Ks than wortmannin (IC50 ~0.3–
Theophylline and caffeine are naturally occurring meth- 4 μM) [80]. However, its superior chemical stability in
ylxanthines that have pleiotropic effects on the nervous, solution has led to its widespread use as a PI3K inhibitor in
respiratory, cardiovascular, and renal systems. Caffeine is cell-based experiments, where it is usually used at concen-
the world’s most widely consumed stimulant and reaches trations of 10–50 μM. Despite its frequent description as a
plasma concentrations of ~50 μM in moderate coffee “specific” PI3K inhibitor, LY294004 blocks the activity of
drinkers. Plasma levels above 200 μM are toxic in humans. a number of different protein kinases unrelated to PI3K
Caffeine is used medically to treat apnea of prematurity and [81]. In addition, use of immobilized LY294002 showed
is a component of various headache and pain remedies [71]. that the drug binds tightly to numerous non-kinase proteins
Theophylline causes bronchodilator and antiinflammatory with diverse functions, with as yet unknown biological
responses and has long been used clinically for the consequences [80]. Class II PI3Ks (IC50 =6.9–19 μM) and
treatment of asthma and other respiratory diseases. The PI4Ks are relatively resistant to LY294002 as compared
therapeutic serum levels range from 55–111 μM, while with the class I enzymes [60, 61]. By contrast, the PIKK
concentrations >111 μM are considered to be toxic [72]. family members SMG-1, DNA-PK, and mTOR are targeted
Many biochemical actions of methylxanthines have been at low micromolar concentrations that also inhibit class I
identified, including antagonism of adenosine receptors, PI3Ks [41, 65, 82, 83]. The X-ray crystallographic structure
inhibition of cyclic nucleotide phosphodiesterases, and of p110γ bound to LY294002 shows that the morpholino
increased Ca2+ release from the sarcoplasmic reticulum. oxygen forms a hydrogen bond with the backbone amide of
These compounds also inhibit mTOR and related kinases, Val882 in the pocket that can be occupied by adenine in
J Chem Biol (2008) 1:27–36 33

ATP [58]. Substitution of this oxygen with nitrogen yields a inhibitor in combination with rapamycin, might be a viable
compound (LY303511) that is essentially inactive against therapeutic option for the treatment of certain cancers.
PI3K [79]. Treatment of cells with LY303511 inhibited Programs to develop potent and specific inhibitors of
agonist-induced S6K Thr389 phosphorylation and auto- DNA-PK have also yielded some compounds that inhibit
phosphorylation of mTOR at Ser2481, suggesting that mTOR [83, 87]. In general, these compounds inhibit DNA-
LY303511 might be an mTOR inhibitor. However, effects PK (IC50s in the sub-millimolar range) and p110α and
on mTOR activity were not directly tested using in vitro mTOR (low millimolar IC50s), but not ATM or ATR. One
assays, so whether or not LY303511 is a bona fide mTOR exception to this rule is 2-(morpholin-1-yl)pyrimido[2,1-α]
inhibitor remains an open question [84]. isoquinolin-4-one (compound 13 in Ref. [83]), which does
Since the advent of LY294002, there has been an not inhibit PI3K. We used this compound (named 401 in
explosion of interest in the synthesis of small molecule Ref. [88]) to examine the cellular effect of mTOR inhibition
PI3K inhibitors that show isoform selectivity and improved without complicating side effects on PI3K. The chemical
potency and specificity [1]. Unfortunately, so far there have structure of 401 is shown Fig. 2. First we performed in vitro
been no reports of an analogous effort to systematically kinase assays to confirm that 401 is selective for mTOR
produce mTOR inhibitors based on a structural or func- over p110α and p110β. The specificity profile was further
tional understanding of its catalytic site. However, some of widened by assaying 40 different protein kinases in the
the new compounds developed as PI3K inhibitors have also presence of 5 μM 401. As expected for an mTOR inhibitor,
been evaluated for activity against mTOR. Knight et al. treatment of cells with 401 blocked growth factor-induced
[85] determined the specificity profile of PI3K inhibitors phosphorylation of S6K Thr389 (an mTORC1 site) and Akt
from nine different chemical classes by measuring IC50 Ser473 (an mTORC2 site). By contrast, phosphorylation of
values in vitro against 55 purified kinases, including 15 Erk was not affected. These effects on cell signaling were
PI3K family members. Six compounds were reported to not due to inhibition of DNA-PK, as incubation of cells that
inhibit mTORC1 activity with IC50s≤10 μM. Interestingly, lack DNA-PK with 401 also decreased S6K and Akt
two of the compounds were less potent against mTORC2 phosphorylation. Finally, we examined the effect of 401 on
than mTORC1, suggesting that the catalytic site of the the growth and survival of mouse embryo fibroblasts
common mTOR subunit might be subtly altered by its (MEFs) deficient for TSC1. These cells have abnormally
association with the distinct binding partners in the two high mTORC1/S6K signaling and abnormally low Akt
complexes. The most potent mTOR inhibitor in this group activity due to feedback inhibition from the hyperactivated
was PI-103 (Fig. 2; IC50 =0.02 μM and 0.083 μM against mTORC1/S6K pathway. Long term treatment of these cells
mTORC1 and mTORC2, respectively). A model of the with rapamycin turns off the negative pathway and
three-dimensional structure of PI-103 bound to the active site upregulates Akt, which can provide a survival signal [23].
of p110γ showed that the inhibitor forms a hydrogen bond By contrast, we found that Akt phosphorylation remained
with Val882 in the “adenine pocket” and stretches deep into low in TSC1−/− MEFs cultured in the presence of 401.
an “affinity pocket” not accessed by ATP that contains the Furthermore, 401-treated cells exhibited strong growth
side chain of Ile879 [85]. The residues in mTOR at inhibition and increased apoptosis as compared with MEFs
equivalent positions are Val2240 and Ile2237, respectively, treated with rapamycin [88]. Treatment of TSC1−/− MEFs
suggesting that binding of PI-103 to the catalytic site of with an Akt inhibitor also increased apoptosis, suggesting
mTOR involves similar interactions in this region. that the cytotoxic effect of 401 might be due to suppression
Although PI-103 represents the first potent synthetic of mTORC2/Akt signaling. These results suggest that
inhibitor of mTOR, it is essentially equipotent against class inhibition of mTOR kinase activity by a small molecule
I PI3Ks (IC50 = 0.008–0.15 μM). Although this dual inhibitor such as 401 might be more effective than
specificity limits the use of PI-103 as a probe to study rapamycin at killing cancer cells that exhibit hyperactivated
mTOR function, PI-103 has proved to be useful as an mTOR signaling.
experimental compound in cancer research. PI-103 was
found to be the most active compound in a group of ten
isoform-selective PI3K inhibitors that were evaluated for Conclusions and outlook
the ability to block the proliferation of glioma cells in vitro
[86]. The cytostatic property of the compound was Upregulation of the PI3K/mTOR/Akt pathway is a common
attributed to its ability to inhibit both p110α and mTOR. feature of many proliferative disorders, including cancer.
PI-103 treatment of mice also reduced the size of Up to now, rapamycin and rapalogs are the most well
established tumor xenografts at doses that produced no studied mTOR inhibitors and they are now clinically used
observable toxicity [86]. These results suggest that dual- as cancer treatments. However, the possibility that inhibi-
specificity PI3K/mTOR inhibitors, or use of a PI3K tion of mTORC1 with these drugs might lead to Akt
34 J Chem Biol (2008) 1:27–36

upregulation and outgrowth of more aggressive lesions is a and raptor-independent pathway that regulates the cytoskeleton.
concern. Use of rapamycin plus an inhibitor of Akt or PI3K Curr Biol 14:1296–1302
7. Guertin DA, Sabatini DM (2005) An expanding role for mTOR in
is one way to circumvent this problem, and another is to use cancer. Trends Mol Med 11:353–361
a dual-specificity agent such as PI-103 that targets both 8. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005)
PI3K and mTOR. Another strategy is to develop drugs that Phosphorylation and regulation of Akt/PKB by the rictor-mTOR
selectively target the mTOR kinase domain, which should complex. Science 307:1098–1101
9. Wullschleger S, Loewith R, Hall MN (2006) TOR signaling in
inhibit both mTORC1 and mTORC2. A treatment combin- growth and metabolism. Cell 124:471–484
ing an mTORC2 inhibitor plus rapalogs would have a 10. Gangloff YG, Mueller M, Dann SG, Svoboda P, Sticker M, Spetz
similar effect. One caveat regarding these strategies is that JF, Um SH, Brown EJ, Cereghini S, Thomas G, Kozma SC (2004)
each drug class is expected to cause a distinct spectrum of Disruption of the mouse mTOR gene leads to early postimplanta-
tion lethality and prohibits embryonic stem cell development. Mol
toxicities. For example, mTORC2 inhibition might be less Cell Biol 24:9508–9516
toxic than PI3K or Akt inhibitors because it would affect 11. Murakami M, Ichisaka T, Maeda M, Oshiro N, Hara K,
mainly FOXO signaling and not other downstream effectors Edenhofer F, Kiyama H, Yonezawa K, Yamanaka S (2004)
of Akt. mTOR is essential for growth and proliferation in early mouse
embryos and embryonic stem cells. Mol Cell Biol 24:6710–
Considerable progress has been made in the last several 6718
years toward elucidating the structure and regulation of the 12. Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY,
two mTOR complexes, and it is now possible to assay each Moffat J, Brown M, Fitzgerald KJ, Sabatini DM (2006) Ablation
one in vitro. Determination of the three-dimensional struc- in mice of the mTORC components raptor, rictor, or mLST8
reveals that mTORC2 is required for signaling to Akt-FOXO and
ture of the mTOR kinase catalytic domain would be a major PKCalpha, but not S6K1. Dev Cell 11:859–871
breakthrough that would aid in the design and analysis of 13. Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, Huang
new inhibitors. Development of drugs specific for the mTOR Q, Qin J, Su B (2006) SIN1/MIP1 maintains rictor-mTOR
kinase domain or that disrupt the mTORC2 complex should complex integrity and regulates Akt phosphorylation and substrate
specificity. Cell 127:125–137
narrow the gaps in our knowledge about the importance of 14. Shiota C, Woo JT, Lindner J, Shelton KD, Magnuson MA (2006)
mTORC1 and mTORC2 in health and disease. Multiallelic disruption of the rictor gene in mice reveals that
mTOR complex 2 is essential for fetal growth and viability. Dev
Cell 11:583–589
Acknowledgments The authors’ research was supported by Carol 15. Li Y, Corradetti MN, Inoki K, Guan KL (2004) TSC2: filling the
M. Baldwin Breast Cancer Research Awards, and grants from the GAP in the mTOR signaling pathway. Trends Biochem Sci
Department of Veterans Affairs and the National Institutes of Health 29:32–38
DK62722. We greatly appreciate the expert graphical assistance of Jun 16. Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC,
Yong Choi. Blenis J (2002) Tuberous sclerosis complex-1 and -2 gene
products function together to inhibit mammalian target of
rapamycin (mTOR)-mediated downstream signaling. Proc Natl
Acad Sci USA 99:13571–13576
References 17. Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC (2002)
Identification of the tuberous sclerosis complex-2 tumor suppres-
sor gene product tuberin as a target of the phosphoinositide 3-
1. Marone R, Cmiljanovic V, Giese B, Wymann MP (2008) kinase/akt pathway. Mol Cell 10:151–162
Targeting phosphoinositide 3-kinase-Moving towards therapy. 18. Alessi DR, Sakamoto K, Bayascas JR (2006) LKB1-dependent
Biochim Biophys Acta 1784:159–185 signaling pathways. Annu Rev Biochem 75:137–163
2. Huang CH, Mandelker D, Schmidt-Kittler O, Samuels Y, 19. Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA,
Velculescu VE, Kinzler KW, Vogelstein B, Gabelli SB, Amzel DePinho RA, Cantley LC (2004) The tumor suppressor LKB1
LM (2007) The structure of a human p110alpha/p85alpha kinase directly activates AMP-activated kinase and regulates
complex elucidates the effects of oncogenic PI3Kalpha mutations. apoptosis in response to energy stress. Proc Natl Acad Sci USA
Science 318:1744–1748 101:3329–3335
3. Nobukuni T, Joaquin M, Roccio M, Dann SG, Kim SY, Gulati P, 20. Inoki K, Ouyang H, Zhu T, Lindvall C, Wang Y, Zhang X, Yang
Byfield MP, Backer JM, Natt F, Bos JL, Zwartkruis FJ, Thomas G Q, Bennett C, Harada Y, Stankunas K, Wang CY, He X,
(2005) Amino acids mediate mTOR/raptor signaling through MacDougald OA, You M, Williams BO, Guan KL (2006) TSC2
activation of class 3 phosphatidylinositol 3OH-kinase. Proc Natl integrates Wnt and energy signals via a coordinated phosphor-
Acad Sci USA 102:14238–14243 ylation by AMPK and GSK3 to regulate cell growth. Cell
4. Abraham RT (2004) PI 3-kinase related kinases: ‘big’ players in 126:955–968
stress-induced signaling pathways. DNA Repair (Amst) 3:883– 21. Byfield MP, Murray JT, Backer JM (2005) hVps34 is a nutrient-
887 regulated lipid kinase required for activation of p70 S6 kinase. J
5. Hara K, Maruki Y, Long X, Yoshino K, Oshiro N, Hidayat S, Biol Chem 280:33076–33082
Tokunaga C, Avruch J, Yonezawa K (2002) Raptor, a binding 22. Deyoung MP, Horak P, Sofer A, Sgroi D, Ellisen LW (2008)
partner of target of rapamycin (TOR), mediates TOR action. Cell Hypoxia regulates TSC1/2 mTOR signaling and tumor suppres-
110:177–189 sion through REDD1-mediated 14 3 3 shuttling. Genes Dev
6. Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, 22:239–251
Erdjument-Bromage H, Tempst P, Sabatini DM (2004) Rictor, a 23. Shah OJ, Wang Z, Hunter T (2004) Inappropriate activation of
novel binding partner of mTOR, defines a rapamycin-insensitive the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion,
J Chem Biol (2008) 1:27–36 35

insulin resistance, and cell survival deficiencies. Curr Biol tivity by the mammalian target of rapamycin. Mol Cell Biol
14:1650–1656 22:7428–7438
24. Yang Q, Inoki K, Kim E, Guan KL (2006) TSC1/TSC2 and Rheb 42. Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-
have different effects on TORC1 and TORC2 activity. Proc Natl Bromage H, Tempst P, Sabatini DM (2002) mTOR interacts with
Acad Sci USA 103:6811–6816 raptor to form a nutrient-sensitive complex that signals to the cell
25. Zhang H, Cicchetti G, Onda H, Koon HB, Asrican K, growth machinery. Cell 110:163–175
Bajraszewski N, Vazquez F, Carpenter CL, Kwiatkowski DJ 43. Elad G, Paz A, Haklai R, Marciano D, Cox A, Kloog Y (1999)
(2003) Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K- Targeting of K-Ras 4B by S-trans,trans-farnesyl thiosalicylic acid.
Akt signaling through downregulation of PDGFR. J Clin Invest Biochim. Biophys Acta 1452:228–242
112:1223–1233 44. McMahon LP, Yue W, Santen RJ, Lawrence JC Jr. (2005)
26. Tremblay F, Brule S, Hee Um S, Li Y, Masuda K, Roden M, Sun Farnesylthiosalicylic acid inhibits mammalian target of rapamycin
XJ, Krebs M, Polakiewicz RD, Thomas G, Marette A (2007) (mTOR) activity both in cells and in vitro by promoting
Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- dissociation of the mTOR-raptor complex. Mol Endocrinol
and obesity-induced insulin resistance. Proc Natl Acad Sci USA 19:175–183
104:14056–14061 45. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley
27. Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, AF, Markhard AL, Sabatini DM (2006) Prolonged rapamycin
Fumagalli S, Allegrini PR, Kozma SC, Auwerx J, Thomas G treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell
(2004) Absence of S6K1 protects against age- and diet-induced 22:159–168
obesity while enhancing insulin sensitivity. Nature 431:200–205 46. Bai X, Ma D, Liu A, Shen X, Wang QJ, Liu Y, Jiang Y (2007)
28. O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, Lane Rheb activates mTOR by antagonizing its endogenous inhibitor,
H, Hofmann F, Hicklin DJ, Ludwig DL, Baselga J, Rosen N FKBP38. Science 318:977–980
(2006) mTOR inhibition induces upstream receptor tyrosine 47. Kirchner GI, Meier-Wiedenbach I, Manns MP (2004) Clinical
kinase signaling and activates. Akt Cancer Res 66:1500–1508 pharmacokinetics of everolimus. Clin Pharmacokinet 43:83–95
29. Shi Y, Yan H, Frost P, Gera J, Lichtenstein A (2005) Mammalian 48. Terada N, Takase K, Papst P, Nairn AC, Gelfand EW (1995)
target of rapamycin inhibitors activate the AKT kinase in multiple Rapamycin inhibits ribosomal protein synthesis and induces G1
myeloma cells by up-regulating the insulin-like growth factor prolongation in mitogen-activated T lymphocytes. J Immunol
receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase 155:3418–3426
cascade. Mol Cancer Ther 4:1533–1540 49. Gutierrez-Dalmau A, Campistol JM (2007) Immunosuppressive
30. Tremblay F, Gagnon A, Veilleux A, Sorisky A, Marette A (2005) therapy and malignancy in organ transplant recipients: a system-
Activation of the mammalian target of rapamycin pathway acutely atic review. Drugs 67:1167–1198
inhibits insulin signaling to Akt and glucose transport in 3T3-L1 50. Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S,
and human adipocytes. Endocrinology 146:1328–1337 Hornung M, Bruns CJ, Zuelke C, Farkas S, Anthuber M, Jauch
31. Bjornsti MA, Houghton PJ (2004) The TOR pathway: a target for KW, Geissler EK (2002) Rapamycin inhibits primary and
cancer therapy. Nature reviews 4:335–348 metastatic tumor growth by antiangiogenesis: involvement of
32. Easton JB, Houghton PJ (2006) mTOR and cancer therapy. vascular endothelial growth factor. Nat Med 8:128–135
Oncogene 25:6436–6446 51. Boulay A, Zumstein-Mecker S, Stephan C, Beuvink I, Zilbermann
33. Dumont FJ, Su Q (1996) Mechanism of action of the immuno- F, Haller R, Tobler S, Heusser C, O'Reilly T, Stolz B, Marti A,
suppressant rapamycin. Life Sci 58:373–395 Thomas G, Lane HA (2004) Antitumor efficacy of intermittent
34. Kuo CJ, Chung J, Fiorentino DF, Flanagan WM, Blenis J, treatment schedules with the rapamycin derivative RAD001
Crabtree GR (1992) Rapamycin selectively inhibits interleukin-2 correlates with prolonged inactivation of ribosomal protein S6
activation of p70 S6 kinase. Nature 358:70–73 kinase 1 in peripheral blood mononuclear cells. Cancer Res
35. Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, Hall 64:252–261
MN (2004) Mammalian TOR complex 2 controls the actin 52. Moses JW, Leon MB, Popma JJ, Fitzgerald PJ, Holmes DR,
cytoskeleton and is rapamycin insensitive. Nature Cell Biology O'Shaughnessy C, Caputo RP, Kereiakes DJ, Williams DO,
6:1122–1128 Teirstein PS, Jaeger JL, Kuntz RE (2003) Sirolimus-eluting stents
36. Choi J, Chen J, Schreiber SL, Clardy J (1996) Structure of the versus standard stents in patients with stenosis in a native
FKBP12-rapamycin complex interacting with the binding domain coronary artery. N Engl J Med 349:1315–1323
of human FRAP. Science 273:239–242 53. Shillingford JM, Murcia NS, Larson CH, Low SH, Hedgepeth R,
37. Banaszynski LA, Liu CW, Wandless TJ (2005) Characterization Brown N, Flask CA, Novick AC, Goldfarb DA, Kramer-Zucker
of the FKBP.rapamycin.FRB ternary complex. J Am Chem Soc A, Walz G, Piontek KB, Germino GG, Weimbs T (2006) The
127:4715–4721 mTOR pathway is regulated by polycystin-1, and its inhibition
38. Leone M, Crowell KJ, Chen J, Jung D, Chiang GG, Sareth S, reverses renal cystogenesis in polycystic kidney disease. Proc Natl
Abraham RT, Pellecchia M (2006) The FRB domain of mTOR: Acad Sci USA 103:5466–5471
NMR solution structure and inhibitor design. Biochemistry 54. Brian PW, Curtis PJ, Hemming HG, Norris GLF (1957)
45:10294–10302 Wortmannin, an antibiotic produced by Penicillium wortmanni.
39. Veverka V, Crabbe T, Bird I, Lennie G, Muskett FW, Taylor RJ, Trans Br Mycol Soc 40:365–368
Carr MD (2008) Structural characterization of the interaction of 55. Nakanishi S, Kakita S, Takahashi I, Kawahara K, Tsukuda E,
mTOR with phosphatidic acid and a novel class of inhibitor: Sano T, Yamada K, Yoshida M, Kase H, Matsuda Y et al (1992)
compelling evidence for a central role of the FRB domain in Wortmannin, a microbial product inhibitor of myosin light chain
small molecule-mediated regulation of mTOR. Oncogene kinase. J Biol Chem 267:2157–2163
27:585–595 56. Arcaro A, Wymann MP (1993) Wortmannin is a potent phospha-
40. Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J (2001) tidylinositol 3-kinase inhibitor: the role of phosphatidylinositol
Phosphatidic acid-mediated mitogenic activation of mTOR sig- 3,4,5-trisphosphate in neutrophil responses. Biochem J 296( Pt
naling. Science 294:1942–1945 2):297–301
41. McMahon LP, Choi KM, Lin TA, Abraham RT, Lawrence JC Jr. 57. Yano H, Nakanishi S, Kimura K, Hanai N, Saitoh Y, Fukui Y,
(2002) The rapamycin-binding domain governs substrate selec- Nonomura Y, Matsuda Y (1993) Inhibition of histamine secretion
36 J Chem Biol (2008) 1:27–36

by wortmannin through the blockade of phosphatidylinositol 3- 74. Foukas LC, Daniele N, Ktori C, Anderson KE, Jensen J, Shepherd
kinase in RBL-2H3 cells. J Biol Chem 268:25846–25856 PR (2002) Direct effects of caffeine and theophylline on p110
58. Walker EH, Pacold ME, Perisic O, Stephens L, Hawkins PT, delta and other phosphoinositide 3-kinases. Differential effects on
Wymann MP, Williams RL (2000) Structural determinants of lipid kinase and protein kinase activities. J Biol Chem 277:37124–
phosphoinositide 3-kinase inhibition by wortmannin, LY294002, 37130
quercetin, myricetin, and staurosporine. Mol Cell 6:909–919 75. Blasina A, Price BD, Turenne GA, McGowan CH (1999) Caffeine
59. Wymann MP, Bulgarelli-Leva G, Zvelebil MJ, Pirola L, inhibits the checkpoint kinase ATM. Curr Biol 9:1135–1138
Vanhaesebroeck B, Waterfield MD, Panayotou G (1996) 76. Block WD, Merkle D, Meek K, Lees-Miller SP (2004) Selective
Wortmannin inactivates phosphoinositide 3-kinase by covalent inhibition of the DNA-dependent protein kinase (DNA-PK) by the
modification of Lys-802, a residue involved in the phosphate radiosensitizing agent caffeine. Nucleic Acids Res 32:1967–1972
transfer reaction. Mol Cell Biol 16:1722–1733 77. Sarkaria JN, Busby EC, Tibbetts RS, Roos P, Taya Y, Karnitz LM,
60. Balla A, Balla T (2006) Phosphatidylinositol 4-kinases: old Abraham RT (1999) Inhibition of ATM and ATR kinase activities
enzymes with emerging functions. Trends Cell Biol 16:351–361 by the radiosensitizing agent, caffeine. Cancer Res 59:4375–4382
61. Falasca M, Maffucci T (2007) Role of class II phosphoinositide 3- 78. Reinke A, Chen JC, Aronova S, Powers T (2006) Caffeine targets
kinase in cell signalling. Biochem Soc Trans 35:211–214 TOR complex I and provides evidence for a regulatory link
62. Liu Y, Jiang N, Wu J, Dai W, Rosenblum JS (2007) Polo-like between the FRB and kinase domains of Tor1p. J Biol Chem
kinases inhibited by wortmannin. Labeling site and downstream 281:31616–31626
effects. J Biol Chem 282:2505–2511 79. Vlahos CJ, Matter WF, Hui KY, Brown RF (1994) A specific
63. Liu Y, Shreder KR, Gai W, Corral S, Ferris DK, Rosenblum JS inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-
(2005) Wortmannin, a widely used phosphoinositide 3-kinase phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem
inhibitor, also potently inhibits mammalian polo-like kinase. 269:5241–5248
Chem Biol 12:99–107 80. Gharbi SI, Zvelebil MJ, Shuttleworth SJ, Hancox T, Saghir N,
64. Volinia S, Dhand R, Vanhaesebroeck B, MacDougall LK, Stein R, Timms JF, Waterfield MD (2007) Exploring the specificity of the
Zvelebil MJ, Domin J, Panaretou C, Waterfield MD (1995) A PI3K family inhibitor LY294002. Biochem J 404:15–21
human phosphatidylinositol 3-kinase complex related to the yeast 81. Bain J, Plater L, Elliott M, Shapiro N, Hastie J, McLauchlan H,
Vps34p-Vps15p protein sorting system. EMBO J 14:3339–3348 Klevernic I, Arthur S, Alessi D, Cohen P (2007) The selectivity of
65. Brunn GJ, Williams J, Sabers C, Wiederrecht G, Lawrence JC Jr., protein kinase inhibitors; a further update. Biochem J 408(3):297–
Abraham RT (1996) Direct inhibition of the signaling functions of 315
the mammalian target of rapamycin by the phosphoinositide 3- 82. Denning G, Jamieson L, Maquat LE, Thompson EA, Fields AP
kinase inhibitors, wortmannin and LY294002. EMBO J 15:5256– (2001) Cloning of a novel phosphatidylinositol kinase-related
5267 kinase: characterization of the human SMG-1 RNA surveillance
66. Chan DW, Son SC, Block W, Ye R, Khanna KK, Wold MS, protein. J Biol Chem 276:22709–22714
Douglas P, Goodarzi AA, Pelley J, Taya Y, Lavin MF, Lees-Miller 83. Griffin RJ, Fontana G, Golding BT, Guiard S, Hardcastle IR,
SP (2000) Purification and characterization of ATM from human Leahy JJ, Martin N, Richardson C, Rigoreau L, Stockley M,
placenta. A manganese-dependent, wortmannin-sensitive serine/ Smith GC (2005) Selective benzopyranone and pyrimido[2,1-a]
threonine protein kinase. J Biol Chem 275:7803–7810 isoquinolin-4-one inhibitors of DNA-dependent protein kinase:
67. Sarkaria JN, Tibbetts RS, Busby EC, Kennedy AP, Hill DE, synthesis, structure-activity studies, and radiosensitization of a
Abraham RT (1998) Inhibition of phosphoinositide 3-kinase human tumor cell line in vitro. J Med Chem 48:569–585
related kinases by the radiosensitizing agent wortmannin. Cancer 84. Kristof AS, Pacheco-Rodriguez G, Schremmer B, Moss J (2005)
Res 58:4375–4382 LY303511 (2-piperazinyl-8-phenyl-4H-1-benzopyran-4-one) acts
68. Yamashita A, Ohnishi T, Kashima I, Taya Y, Ohno S (2001) via phosphatidylinositol 3-kinase-independent pathways to inhibit
Human SMG-1, a novel phosphatidylinositol 3-kinase-related cell proliferation via mammalian target of rapamycin (mTOR)-
protein kinase, associates with components of the mRNA and non-mTOR-dependent mechanisms. J Pharmacol Exp Ther
surveillance complex and is involved in the regulation of 314:1134–1143
nonsense-mediated mRNA decay. Genes Dev 15:2215–2228 85. Knight ZA, Gonzalez B, Feldman ME, Zunder ER, Goldenberg
69. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, DD, Williams O, Loewith R, Stokoe D, Balla A, Toth B, Balla T,
Klevernic I, Arthur JS, Alessi DR, Cohen P (2007) The selectivity Weiss WA, Williams RL, Shokat KM (2006) A pharmacological
of protein kinase inhibitors: a further update. Biochem J 408:297– map of the PI3-K family defines a role for p110alpha in insulin
315 signaling. Cell 125:733–747
70. Ihle NT, Williams R, Chow S, Chew W, Berggren MI, Paine- 86. Fan QW, Knight ZA, Goldenberg DD, Yu W, Mostov KE, Stokoe
Murrieta G, Minion DJ, Halter RJ, Wipf P, Abraham R, D, Shokat KM, Weiss WA (2006) A dual PI3 kinase/mTOR
Kirkpatrick L, Powis G (2004) Molecular pharmacology and inhibitor reveals emergent efficacy in glioma. Cancer Cell 9:341–
antitumor activity of PX-866, a novel inhibitor of phosphoinositide- 349
3-kinase signaling. Mol Cancer Ther 3:763–772 87. Hardcastle IR, Cockcroft X, Curtin NJ, El-Murr MD, Leahy JJ,
71. Magkos F, Kavouras SA (2005) Caffeine use in sports, pharma- Stockley M, Golding BT, Rigoreau L, Richardson C, Smith GC,
cokinetics in man, and cellular mechanisms of action. Crit Rev Griffin RJ (2005) Discovery of potent chromen-4-one inhibitors of
Food Sci Nutr 45:535–562 the DNA-dependent protein kinase (DNA-PK) using a small-
72. Barnes PJ (2003) Theophylline: new perspectives for an old drug. molecule library approach. J Med Chem 48:7829–7846
Am J Respir Crit Care Med 167:813–818 88. Ballou LM, Selinger ES, Choi JY, Drueckhammer DG, Lin RZ
73. Scott PH, Lawrence JC Jr. (1998) Attenuation of mammalian (2007) Inhibition of mammalian target of rapamycin signaling by
target of rapamycin activity by increased cAMP in 3T3-L1 2-(morpholin-1-yl)pyrimido[2,1-alpha]isoquinolin-4-one. J Biol
adipocytes. J Biol Chem 273:34496–34501 Chem 282:24463–24470

You might also like