Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jan 1.
Published in final edited form as: Curr Top Microbiol Immunol. 2011;347:1–19. doi: 10.1007/82_2010_65

PI3K – From the Bench to the Clinic and Back

Bart Vanhaesebroeck a, Peter K Vogt b, Christian Rommel c
PMCID: PMC2954281  NIHMSID: NIHMS217237  PMID: 20549473

Abstract

From humble beginnings over 25 years ago as a lipid kinase activity associated with certain oncoproteins, PI3K (phosphoinositide 3-kinase) has been catapulted to the forefront of drug development in cancer, immunity and thrombosis, with the first clinical trials of PI3K pathway inhibitors now in progress. Here we give a brief overview of some key discoveries in the PI3K area and their impact, and include thoughts on the current state of the field, and where it could go from here.


PI3K has become a very intense area of research, with over 2000 publications on PI3K in PubMed for 2009 alone. The expectations for a therapeutic impact of intervention with PI3K activity are high, and progress in the clinical arena is being monitored by many. However, targeted therapies almost invariably encounter roadblocks, often exposing unresolved questions in the basic understanding of the target. PI3K will most likely be no exception. Below, we describe some of these early ‘surprises’ and how these inform and shape basic science investigations.

The discovery of the PI3K signalling pathway and its potential as a therapeutic target

Early work showed that a phosphatidylinositol kinase activity co-purified with various viral oncoproteins expressed in mammalian cells (Macara et al. 1984; Sugimoto et al. 1984) and that cellular transformation mediated by such oncoproteins was to some extent dependent on the association with this lipid kinase activity (Whitman et al. 1985). This oncoprotein-associated lipid kinase could phosphorylate phosphatidylinositol on the 3-OH position of the inositol ring, hereby generating PI3P, a novel type of phosphoinositide (Whitman et al. 1988). This finding was followed by the discovery of PI(3,4,5)P3 (phosphatidylinositol(3,4,5)trisphosphate; PIP3) in GPCR-stimulated neutrophils (Traynor-Kaplan et al. 1988; Traynor-Kaplan et al. 1989) and upon acute stimulation with tyrosine kinase agonists (Auger et al. 1989; Hawkins et al. 1992; Jackson et al. 1992). It was not known at the time that agonist-stimulated PI3K is a heterodimer made up of a p110 catalytic subunit and a regulatory subunit, namely p85 in the case of class IA PI3Ks and p101 in the case of the class IB p110γ. Early studies very much focused on a tyrosine-phosphorylated 85 kD protein found in PDGF-stimulated or polyoma middle T-transformed cells which associated with PI3K activity (Courtneidge and Heber 1987; Kaplan et al. 1987). This protein turned out to be the p85 regulatory subunit of PI3K, and its cDNA was cloned by several groups (Escobedo et al. 1991; Otsu et al. 1991; Skolnik et al. 1991). Several teams also purified the PI3K enzyme activity biochemically from various tissues (Carpenter et al. 1990; Fry et al. 1992; Morgan et al. 1990; Shibasaki et al. 1991; Stephens et al. 1994). Protein microsequencing allowed the design of oligonucleotide probes to isolate the first cDNA of a PI3K catalytic subunit, namely p110α (Hiles et al. 1992). This work revealed that the sequence of p110 was closely homologous to that of the product of vps34, a S. cerevisiae gene involved in endosomal sorting of proteins towards the vacuole, the yeast equivalent of the mammalian lysosome (Herman and Emr 1990). Follow-up work revealed that vps34 indeed had PI3K activity, but with a substrate specificity that was different from p110α, in that it can only phosphorylate PI (phosphatidylinositol) but not PI(4,5)P2 (phosphatidylinositol(4,5)bisphosphate) (Schu et al. 1993).

A concerted effort of many laboratories, using various techniques, including biochemical purification and degenerate PCR approaches, revealed the existence of multiple PI3K isoforms in mammals (Arcaro et al. 1998; Brown et al. 1997; Chantry et al. 1997; Domin et al. 1997; Hu et al. 1993; Misawa et al. 1998; Ono et al. 1998; Stephens et al. 1997; Stoyanov et al. 1995; Vanhaesebroeck et al. 1997b; Virbasius et al. 1996), but also in D. melanogaster (MacDougall et al. 1995), C. elegans (Morris et al. 1996), Dictyostelium (Zhou et al. 1995) and other species, even in plants. These findings led to the realisation that PI3Ks are an evolutionarily conserved family of enzymes which on the basis of structural and biochemical characteristics was divided into 3 classes (Vanhaesebroeck et al. 1997a; Zvelebil et al. 1996). Mammals have eight isoforms of PI3K (class IA: p110α, p110β, p110δ; class IB: p110γ; class II: PI3K-C2α, PI3K-C2β, PI3K-Cγ, and class III: vps34p). A single representative of each of the three PI3K classes is present in C. elegans and D. melanogaster. In yeast, only a class III PI3K is found (reviewed in Ref. Vanhaesebroeck et al. 2001).

The analysis of PI3K functions in the cell was greatly aided by two small molecule inhibitors, wortmannin and LY294002. Wortmannin was identified as a PI3K inhibitor in 1993 (Arcaro and Wymann 1993; Okada et al. 1994; Powis et al. 1994; Yano et al. 1993), and in 1994, Lilly laboratories published the LY294002 inhibitor (Vlahos et al. 1994). Interestingly, all these papers almost exclusively focused on probing the immunological aspects of PI3K function using these compounds. LY294002 and wortmannin have undoubtedly been instrumental in providing first insights into the cell biology of PI3Ks but may also have generated some false expectations due to lack of specificity (see below).

Concurrent with the isolation of the genes for the different PI3Ks was the realisation that the 3-phosphoinositides could selectively bind to defined target modules in proteins, thereby altering the localisation of such proteins and their conformation and activity. Among numerous protein domains that were defined during this time was the PH (pleckstrin homology) domain, a module that occurs in many proteins (Haslam et al. 1993; Mayer et al. 1993). A major discovery was that some PH domains could bind phosphoinositides (Harlan et al. 1994). The characterisation of other 3-phosphoinositide binding domains soon followed, including the FYVE (Fab 1, YOTB, Vac 1, EEA1) domain (Gaullier et al. 1998; Mu et al. 1995; Stenmark et al. 1996) and PX (Phox) domain (Cheever et al. 2001; Ellson et al. 2001; Kanai et al. 2001; Song et al. 2001; Xu et al. 2001) which both bind PI3P (phosphatidylinositol 3-phosphate).

One of the proteins that was reported (Haslam et al. 1993; Mayer et al. 1993) to have a PH domain was the Ser/Thr kinase Akt, which is the mammalian cellular homologue of the retroviral transforming gene v-Akt (Bellacosa et al. 1991). Akt was also independently cloned as a protein kinase related to PKA and PKC, hence its alternative names PKB (Coffer and Woodgett 1991) and Rac (related to A and C kinases) (Jones et al. 1991). Akt was subsequently confirmed as a PI3K target in cells stimulated with tyrosine kinase agonists, including PDGF and insulin (Burgering and Coffer 1995; Franke et al. 1995), and through its PH domain shown to bind PIP3 and PI(3,4)P2 with high specificity and affinity (Andjelkovic et al. 1997; Frech et al. 1997; Stokoe et al. 1997). An intact PH domain in Akt is crucial for its function (Stocker et al. 2002).

The regulation of Akt itself turned out to be rather complex. The PH domain recruits Akt to PIP3 and PI(3,4)P2 and the plasma membrane, where it becomes a substrate for the membrane-bound PDK1 kinase, which phosphorylates Akt on Thr308 (Alessi et al. 1997a; Alessi et al. 1997b; Stephens et al. 1998; Stokoe et al. 1997). Very early on, it was documented that Akt is also phosphorylated on Ser473 (Alessi et al. 1996), but it took more than a decade to identify the kinase that performs this phosphorylation. It turned out to be mTOR complexed with the Rictor protein, also referred to as mTORC2 (Sarbassov et al. 2005) (as opposed to mTORC1, the ‘classical’ mTOR in complex with Raptor).

A next step was to identify downstream substrates of Akt protein kinase activity. Akt was found to control other protein kinases either directly, such as GSKβ (Cross et al. 1995) or indirectly, such as p70 S6 kinase (Burgering and Coffer 1995). One of the Akt substrates turned out to be the pro-apoptotic protein BAD, which is inhibited in its apoptotic function upon phosphorylation by Akt (Datta et al. 1997; del Peso et al. 1997). Given that wortmannin and LY294002 had previously been shown to be able to induce cell death (Yao and Cooper 1995), these observations suggested the existence of a PI3K-Akt cell survival pathway.

It is often overlooked that studies in D. melanogaster and especially in C. elegans have been instrumental in delineating the generic layout of the PI3K pathway and key aspects of its biology. For example, studies in C. elegans uncovered the link between the insulin-receptor, PI3K and the FOXO transcription factors (Ogg et al. 1997) and between Akt and FOXO (Paradis and Ruvkun 1998). FOXO transcription factors were later shown to be a target for direct phosphorylation by Akt in mammalian cells (Brunet et al. 1999; Kops et al. 1999). Further seminal work in model organisms included the identification of AGE-1 as the C. elegans p110 paralog with a key function in the control of lifespan (Morris et al. 1996) and the identification of PI3K in Drosophila as an important determinant in the regulation of cell growth and size (Leevers et al. 1996).

Work from many groups further uncovered new elements of PI3K signaling, revealing the involvement of other PH domain-containing proteins, including regulators of small GTPases (GEFs and GAPs) (Klarlund et al. 1997; Krugmann et al. 2002; Welch et al. 2002) and various scaffolding and adaptor proteins (such as Gab1, Bam32, DAPP1) (Isakoff et al. 1998). These pathways have received much less attention over the years than Akt, and this may have had the effect of underestimating the importance of Akt-independent biology in PI3K action.

PI3K and human disease

Although the link between oncoproteins, growth factors and PI3K signaling, including the identification of PI3K as a Ras effector (Rodriguez-Viciana et al. 1994; Sjolander et al. 1991) and the demonstration that PI3K could act as a retroviral oncogene (Chang et al. 1997), provided some circumstantial evidence for a role of PI3K in cancer, genetic evidence from human cancer emerged only relatively late. An important breakthrough was the identification of the PTEN tumour suppressor as a PIP3-phosphatase (Maehama and Dixon 1998). The frequently occurring inactivation of PTEN in cancer leads to constitutive activation of the PI3K pathway. It was not until 2004, however, that cancer-specific activating mutations were reported in PIK3CA, which encodes the p110α isoform of PI3K (Campbell et al. 2004; Samuels et al. 2004). Surprisingly, no mutations in non-p110α isoforms have been detected thus far (Parsons et al. 2008; Samuels et al. 2004; TGCA 2008; Thomas et al. 2007; Wood et al. 2007). Mutations in the regulatory subunit, p85α, encoded by PIK3R1, have been also discovered, although they occur at low frequency (Jaiswal et al. 2009; Philp et al. 2001; TGCA 2008). Interestingly, these mutations can also activate p110β and p110δ, possibly providing a broader activation of the class IA PI3K pathway than PIK3CA mutations (Jaiswal et al. 2009). The sheer number of mutations directed to PI3K signaling in PTEN, PIK3CA, PIK3R1 and several upstream receptor tyrosine kinases makes this pathway one of the most deregulated and druggable biochemical activities in human cancer.

Since the mid nineties, evidence for non-redundant functions of the class IAPI3K isoforms began to emerge (Hill et al. 2000; Roche et al. 1998; Roche et al. 1994; Vanhaesebroeck et al. 1999). Isoform-specific functions were exemplified by mice with inactivated p110γ (Hirsch et al. 2000; Li et al. 2000; Sasaki et al. 2000) or p110δ (Clayton et al. 2002; Jou et al. 2002; Okkenhaug et al. 2002), PI3K isoforms that are preferentially expressed in leukocytes. These mice are viable and fertile but show largely non-overlapping immune phenotypes. The phenotypes of these genetically modified mice identified p110γ and p110δ as targets in immunity and inflammation (Rommel et al. 2007; Ruckle et al. 2006; Soond et al.).

Another area of isoform-specific function and possible therapeutic intervention is represented by the role of p110β in platelet biology and thrombosis (Jackson et al. 2005). The p110β isoform plays a key role in regulating the formation and stability of integrin/adhesion bonds, necessary for shear activation of platelets (Jackson et al. 2005). An isoform-selective p110β inhibitor eliminates occlusive thrombus formation but does not prolong bleeding time in vivo (Jackson et al. 2005). These studies defined p110β as a new target for antithrombotic therapy.

The development of PI3K inhibitors for human disease starts to inform basic science

In 2003, the first isoform-selective inhibitor, IC87114, which has high selectivity for p110δ, was published (Sadhu et al. 2003). Over the last decade, ever increasing efforts were made to create both isoform-selective and pan-PI3K inhibitors for therapeutic use, efforts aided by the first crystal structure of a PI3K, that of p110γ (Walker et al. 1999).

Isoform-selective inhibitors for p110δ (CAL101/hematologic malignancies) and p110β (AZD6482/thrombosis) have recently entered early clinical evaluation. Compounds that are effective against all class I PI3K isoforms, including sometimes mTOR, are currently being advanced into cancer patients with solid tumors. PI3K inhibitors have not yet been tested in allergy, inflammation and autoimmunity.

Several PI3K drug candidates have started to raise questions that impact on basic research, especially in the regulation of cell survival by PI3K. Indeed, inhibition of class I PI3K activity with pan-class I PI3K inhibitor compounds does not efficiently induce apoptosis, but rather lead to a G0/G1 cell cycle arrest (Dan et al. 2009; Fan et al. 2007; Guillard et al. 2009; Raynaud et al. 2007). In other words, inhibition of class I PI3K activity appears to be better at slowing down cell proliferation than at killing cells. This observation is reminiscent of what has been found in flies and worms, where inactivation of class I PI3K activity inhibits cell growth but does not induce cell death (Leevers et al. 1996; Morris et al. 1996). Mammalian cells have recently been shown to be able to survive and proliferate normally with extremely reduced levels of class I PI3K activity (Foukas et al. 2010).

Looking back, it is clear that the effect on cell survival has been most prominently associated with PI3K action. It is becoming increasingly clear that, while PI3K and Akt are effective modulators of anti-apoptotic signalling, in many systems, they are neither necessary nor sufficient to protect against cell death (reviewed in Ref. Vanhaesebroeck et al. 2001), These data suggest that the role of PI3K, and especially of Akt, in the control of cell survival and apoptosis may have been overestimated.

It is possible that the apoptosis-inducing activity of the pan-PI3K inhibitor LY294002, seen in some but not all cells, may be due to off-target effects. It is even more likely that cellular stress may have played a role in the outcome of some of the early studies on LY294002, for example when tested on explanted cells such as neurons which are undergoing tissue culture stress (Yao and Cooper 1995). An option for increasing therapeutic effectiveness of PI3K inhibitors in cancer could be to broaden the PI3K target spectrum to include class II and class III PI3Ks whose potential role in cancer is largely unexplored. It might also be of interest to target PI3K-C2α. Indeed, in a recent study, RNAi targeted to this isoform of PI3K led to cell death in half of the panel of cancer cells tested (Elis et al. 2008). PI3K-C2α is relatively resistant to LY294002 (Domin et al. 1997; Virbasius et al. 1996) and might not have been inhibited by the doses of LY294002 that allowed cells to survive in the presence of this compound. The class III PI3K, vps34, may also be an important cancer target, given that it has been implicated in autophagy, a response to which cells under stress can resort to overcome adverse conditions.

Evidence is slowly emerging that, in large panels of cancer cell lines tested in vitro, there is a lack of correlation between sensitivity to class I PI3K inhibitors and the mutational status of PIK3CA or PTEN (Edgar et al.). These data argue against the existence of ‘oncogene-addiction’ to the PI3K pathway in cultured cancer cells. One possible explanation for this finding could be that a gain-of-function in the PI3K pathway is important in cancer initiation, but that the cancer cells are no longer critically dependent on PI3K once the cancer is established. These observations suggest that patient selection on the basis of mutational status may not be as straightforward as originally hoped for. More work clearly needs to be done to define the molecular parameters that predict sensitivity of cancer cells to PI3K inhibition.

New evidence also shows that in cancer cell lines, there is no good correlation between the presence of PIK3CA mutations and the steady state or growth factor-stimulated activity of PI3K and Akt (Morrow et al. 2005; Stemke-Hale et al. 2008; Vasudevan et al. 2009). This is in contrast to engineered cell model systems where gain-of-function mutations in PIK3CA are linked to increased PI3K signalling. It is likely that in cancer cells, other signalling networks come into play and that regulatory feedback loops affect the status of the PI3K activities. Interestingly, some cells with mutant PIK3CA show a dependency on the PDK1 and SGK3 protein kinases (Vasudevan et al. 2009), and it will be important to determine the genes and signaling pathways that might modulate the sensitivity of PI3K mutant cells to PI3K inhibitors.

If (class I) PI3K inhibition alone does not induce cancer cell death, the question arises what are the cancer-cell intrinsic effects of such inhibition that could be exploited for therapy. A cancer-specific role of PI3K signalling in intracellular nutrient sensing and control of metabolic pathways needs to be considered (Coloff and Rathmell 2006; Foukas et al. 2006; Jones and Thompson 2009; Plas and Thompson 2005). Such a role is also supported by the phenotypes of PI3K inactivation in flies and worms (Leevers et al. 1996; Morris et al. 1996). Inhibition of PI3K in vivo has been documented to have a major impact on glucose uptake in tumor cells, as measured by 18fluoro deoxyglucose PET scans (Engelman et al. 2008). Other areas of cell-intrinsic impact of PI3K inhibition such as cell migration, invasion and metastasis also need to be examined.

It is most likely that class I PI3K inhibitors will be clinically effective only in combination with other interventions, such as targeted therapies against the EGF-R or MAPK pathways (Engelman et al. 2008; Faber et al. 2009; Sos et al. 2009), or more generic approaches such as chemo- and radiotherapy. One of the challenges for the future will be to delineate cancer types that might benefit from such combined therapies. An early example of such effective combination strategies is emerging in breast cancer where PI3K inhibitors can overcome resistance to EGF-R-directed therapy (Sergina et al. 2007).

It is important to keep in mind that most of the data on the impact of PI3K inhibition in cancer come from studies with cultured cell lines and xenografts. These conditions may affect the requirement for PI3K which may then differ significantly from the roles of PI3K in an autochthonous tumor growing in vivo. Indeed, the impact of PI3K inhibition on the stroma, including immune cells, fibroblasts, and endothelial cells, could be substantial but remains largely unexplored. A role of PI3K in developmental angiogenesis has recently been established (Graupera et al. 2008), but the functions of PI3K in tumor angiogenesis are not defined. An indirect role of PI3K blockade may also underpin the promising results of the phase I trials with the p110δ inhibitor CAL-101, which induced disease stabilisation in a substantial number of patients with B-cell lymphoma (Flinn et al. 2009). The direct impact of p110δ-centered inhibitors on the proliferation and survival of haematological cancer cells is modest, and it is possible that indirect actions of PI3K inhibitors come to play in this clinical setting.

Some outstanding questions in PI3K biology and signalling

While Akt has been the most studied target of PI3K, many questions on its regulation and function remain unanswered. Indeed, we still do not have a full understanding of its activation by PDK1 and mTORC2, of its inactivation and of the many feedback loops that control this kinase. We are largely ignorant of the mechanisms by which Akt regulates its cellular location and affects its many targets, notably those in the nucleus. We also have little definitive understanding of the specific, non-redundant functions of the three Akt isoforms. As aptly captured by Brian Hemmings when reviewing the field ten years after the molecular cloning of Akt, this is still ‘a hard Akt to follow’ (Brazil and Hemmings 2001). It will also be important to re-evaluate the pro-survival and growth-promoting role of Akt and to define the signalling context that would make it a potentially exploitable therapeutic target.

PI3K effectors other than Akt also deserve more attention and scrutiny. Indeed, other than Akt, PI3K regulates other tyrosine kinases (such as Btk) and affects adaptor proteins (such as Gab2) and a plethora of GEFs and GAPs for monomeric GTPases of the Rac, Ras and Arf families (Vanhaesebroeck et al. 2001). The regulation of these GEFs and GAPs is complex and difficult to track experimentally, but some of these proteins could play important roles in PI3K signalling pathways. This is illustrated by P-REX2a, which activates the small GTPase Rac and is regulated by both PIP3 and the Gβγ subunits of heterotrimeric G proteins, and which has recently been shown to interact with PTEN, inhibiting PTEN function (Fine et al. 2009).

The roles of the PI3K isoforms in human disease need to be further delineated. In a non-cancer context, class I PI3K isoforms have highly non-redundant functions, but it is not clear at this point how such specificity is achieved, as all PI3K isoforms activate Akt indiscriminately. It is possible that PI3K isoforms produce PIP3 in different cellular compartments, and they could also differentially regulate small GTPases such as RhoA (Papakonstanti et al. 2007; Papakonstanti et al. 2008). In cancer, some of this non-redundancy is lost, possibly because the pathways upstream of the PI3K isoforms have been deregulated (Vanhaesebroeck et al. 2010).

Powerful tools to address some of these questions now available. These include isoform-specific inhibitors for p110β, p110γ and p110δ as well as an array of mutant and transgenic mice. The differential roles of p110 isoforms in cancer remain an important topic. It is not clear why the gene encoding p110α is so selectively mutated in cancer. These mutations increase the activity of p110α by enhanced association with the plasma membrane (Gymnopoulos et al. 2007; Mandelker et al. 2009), or by release from a p85-mediated inhibition (Miled et al. 2007), but the detailed molecular mechanisms of increased downstream signalling remain to be determined. There is suggestive evidence that different mutations can have a differential biological output such as in breast cancer cells, where the E545K mutation of PIK3CA appears to be associated with an enhanced metastatic phenotype compared to the H1047R mutation (Pang et al. 2009).

Thus far, the focus of the field has been on class I PI3Ks and their action through the PH-domain-mediated binding of key effectors to PIP3 and PI(3,4)P2. Relatively little attention has been paid to class II and III PI3Ks, their physiological roles and possible involvement in disease. These PI3Ks operate through PI3P and its effector proteins which bind this lipid with their PX or FYVE domains. While PH domains are more abundant than PX and FYVE domains, only a very small subset of PH domains binds PIP3 or PI(3,4)P2 (Lemmon 2008). In contrast, all PX and FYVE domains bind to PI3P. Therefore PI3P has many more effectors than PIP3 and PI(3,4)P2. These effectors are very diverse and include p40 and p47 subunits of NADPH oxidase and proteins with sorting and scaffolding functions in membrane transport such as early endosome antigen-1 (EEA1), Hrs/vps27, ESCRT components, Alfy, kinesins and sorting nexin family members. PI3P-binding proteins also include the lipid kinase Fab1/PIKfyve (which converts PI3P to PI(3,5)P2), the protein kinase SGK3 and additional GAPs (such as RGS-PX1) (reviewed in Refs. Birkeland and Stenmark 2004; Di Paolo and De Camilli 2006; Hurley 2006; Lemmon 2008; Vanhaesebroeck et al. 2010).

A key question is whether PI3P is involved in acute signalling and to what extent it influences signalling by extracellular agonists. Class II PI3K isoforms have been reported to generate PI3P in an agonist-dependent manner (reviewed in Refs. Falasca and Maffucci 2009; Vanhaesebroeck et al. 2010) and vps34 has been shown to control amino acid-dependent activation of S6 kinase-1 through unknown intermediates (Byfield et al. 2005; Nobukuni et al. 2005). At present there are no small molecule inhibitors of class II and III PI3Ks in the public domain (Shuttleworth et al. 2009). The importance of PI3P in disease is underscored by the observation that germline inactivation of PI3P-phosphatases of the myotubularin family in humans can lead to neuropathies and myopathy (Nicot and Laporte 2008).

Last but not least, we know very little about the production of the PI3K lipids themselves, their levels in disease, their subcellular localisation and their dynamic interconversion to other phosphoinositides. The frequent loss of the tumor suppressor PTEN in cancer demonstrates the importance of 3-phosphoinositide turnover. More recent observations assign important roles to 5-phosphatases of PIP3, including IPP5E, whose inactivation is involved in ciliopathies (Bielas et al. 2009; Jacoby et al. 2009), and SHIP2, which has been implicated in insulin signalling and glucose homeostasis (Ooms et al. 2009). INPP4 is a 4-phosphatase of PI(3,4)P2; its INPP4B isoform is a tumor suppressor that inhibits PI3K signalling (Gewinner et al. 2009). PI3P turnover is regulated by myotubularin phosphatases, some of which have been implicated in myopathies and neuropathies (Nicot and Laporte 2008). These data show that it will be essential to monitor the levels and species of phosphoinositides in disease, in combination with proteomic and lipidomic profiling. Although it is now possible to monitor the subcellular distribution of 3-phosphoinositides with labelled lipid-binding domains, no progress has been made in the quantification of 3-phosphoinositides. Indeed, over the last decade, the entire field has almost exclusively relied on proxy readouts such as the phosphorylation of Akt. The disconnects between PI3K pathway activation and Akt phosphorylation that starts to surface (Vasudevan et al. 2009) make it imperative to develop new methods for monitoring 3-phosphoinositides in cells.

Concluding remarks

Remarkable progress has been made over the last two decades in our knowledge of PI3K biology and signalling. PI3Ks have been identified as powerful signaling enzymes that respond to diverse upstream inputs and feed into complex downstream networks. Class I PI3Ks generate the tightly regulated second messenger PIP3 signaling platform. At the level of cellular signalling, the four PI3K isoforms of class I, despite their identical lipid kinase activities, carry out largely non-redundant tasks, and recent evidence suggests that different isoforms can cooperate in achieving specific effects. The molecular basis for these distinctions and complementations is not understood. The extent to which different isoforms can substitute for each other is also not known.

High points in PI3K studies include genetically engineered mice, high resolution crystal structures, biochemical and cellular high throughput assays, cell-based and in vivo imaging assays, human genetics and isoform-selective inhibitors. There is an active debate in the field about selectively targeting single isoforms of PI3K versus a broader, pan-PI3K directed approach. First generation drugs against class I PI3K isoforms have entered clinical testing. Several other drugs targeting alternative components of the PI3K signaling network are at a similar stage of development. Despite many open questions, there is hope that an understanding of the genetic signatures that mark a role for PI3K in disease will translate into therapeutic benefits. First generation drugs are often “learning tools” that will be outperformed by better drugs and knowledge. Clinical experience, basic science and drug development are poised to interdigitate and to complement each other as the PI3K field evolves from a cellular signaling specialty to an area of broad medical significance and impact.

Acknowledgments

The authors apologize to the many authors whose primary research papers have not been cited due to space constraints. Research in the laboratory of BV is supported by Cancer Research UK, the MRC and BBSRC, EU, EMBO and the Ludwig Institute for Cancer Research. PKV is supported by grants from the National Cancer Institute and by The Stein Foundation. This is manuscript 20614 of The Scripps Research Institute. CR is supported by Intellikine Inc., a privately held biopharmaceutical company dedicated to the development of novel molecular targeted therapeutics.

References

  1. Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P, Hemmings BA. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 1996;15:6541–51. [PMC free article] [PubMed] [Google Scholar]
  2. Alessi DR, Deak M, Casamayor A, Caudwell FB, Morrice N, Norman DG, Gaffney P, Reese CB, MacDougall CN, Harbison D, Ashworth A, Bownes M. 3-Phosphoinositide-dependent protein kinase-1 (PDK1): structural and functional homology with the Drosophila DSTPK61 kinase. Curr Biol. 1997a;7:776–89. doi: 10.1016/s0960-9822(06)00336-8. [DOI] [PubMed] [Google Scholar]
  3. Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB, Cohen P. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 1997b;7:261–9. doi: 10.1016/s0960-9822(06)00122-9. [DOI] [PubMed] [Google Scholar]
  4. Andjelkovic M, Alessi DR, Meier R, Fernandez A, Lamb NJ, Frech M, Cron P, Cohen P, Lucocq JM, Hemmings BA. Role of translocation in the activation and function of protein kinase B. J Biol Chem. 1997;272:31515–24. doi: 10.1074/jbc.272.50.31515. [DOI] [PubMed] [Google Scholar]
  5. Arcaro A, Volinia S, Zvelebil MJ, Stein R, Watton SJ, Layton MJ, Gout I, Ahmadi K, Downward J, Waterfield MD. Human phosphoinositide 3-kinase C2beta, the role of calcium and the C2 domain in enzyme activity. J Biol Chem. 1998;273:33082–90. doi: 10.1074/jbc.273.49.33082. [DOI] [PubMed] [Google Scholar]
  6. Arcaro A, Wymann MP. Wortmannin is a potent phosphatidylinositol 3-kinase inhibitor: the role of phosphatidylinositol 3,4,5-trisphosphate in neutrophil responses. Biochem J. 1993;296(Pt 2):297–301. doi: 10.1042/bj2960297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Auger KR, Serunian LA, Soltoff SP, Libby P, Cantley LC. PDGF-dependent tyrosine phosphorylation stimulates production of novel polyphosphoinositides in intact cells. Cell. 1989;57:167–75. doi: 10.1016/0092-8674(89)90182-7. [DOI] [PubMed] [Google Scholar]
  8. Bellacosa A, Testa JR, Staal SP, Tsichlis PN. A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science. 1991;254:274–7. doi: 10.1126/science.254.5029.274. [DOI] [PubMed] [Google Scholar]
  9. Bielas SL, Silhavy JL, Brancati F, Kisseleva MV, Al-Gazali L, Sztriha L, Bayoumi RA, Zaki MS, Abdel-Aleem A, Rosti RO, Kayserili H, Swistun D, Scott LC, Bertini E, Boltshauser E, Fazzi E, Travaglini L, Field SJ, Gayral S, Jacoby M, Schurmans S, Dallapiccola B, Majerus PW, Valente EM, Gleeson JG. Mutations in INPP5E, encoding inositol polyphosphate-5-phosphatase E, link phosphatidyl inositol signaling to the ciliopathies. Nat Genet. 2009;41:1032–6. doi: 10.1038/ng.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Birkeland HC, Stenmark H. Protein targeting to endosomes and phagosomes via FYVE and PX domains. Curr Top Microbiol Immunol. 2004;282:89–115. doi: 10.1007/978-3-642-18805-3_4. [DOI] [PubMed] [Google Scholar]
  11. Brazil DP, Hemmings BA. Ten years of protein kinase B signalling: a hard Akt to follow. Trends Biochem Sci. 2001;26:657–64. doi: 10.1016/s0968-0004(01)01958-2. [DOI] [PubMed] [Google Scholar]
  12. Brown RA, Ho LK, Weber-Hall SJ, Shipley JM, Fry MJ. Identification and cDNA cloning of a novel mammalian C2 domain-containing phosphoinositide 3-kinase, HsC2-PI3K. Biochem Biophys Res Commun. 1997;233:537–44. doi: 10.1006/bbrc.1997.6495. [DOI] [PubMed] [Google Scholar]
  13. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, Blenis J, Greenberg ME. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96:857–68. doi: 10.1016/s0092-8674(00)80595-4. [DOI] [PubMed] [Google Scholar]
  14. Burgering BM, Coffer PJ. Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature. 1995;376:599–602. doi: 10.1038/376599a0. [DOI] [PubMed] [Google Scholar]
  15. Byfield MP, Murray JT, Backer JM. hVps34 is a nutrient-regulated lipid kinase required for activation of p70 S6 kinase. J Biol Chem. 2005;280:33076–82. doi: 10.1074/jbc.M507201200. [DOI] [PubMed] [Google Scholar]
  16. Campbell IG, Russell SE, Choong DY, Montgomery KG, Ciavarella ML, Hooi CS, Cristiano BE, Pearson RB, Phillips WA. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 2004;64:7678–81. doi: 10.1158/0008-5472.CAN-04-2933. [DOI] [PubMed] [Google Scholar]
  17. Carpenter CL, Duckworth BC, Auger KR, Cohen B, Schaffhausen BS, Cantley LC. Purification and characterization of phosphoinositide 3-kinase from rat liver. J Biol Chem. 1990;265:19704–11. [PubMed] [Google Scholar]
  18. Chang HW, Aoki M, Fruman D, Auger KR, Bellacosa A, Tsichlis PN, Cantley LC, Roberts TM, Vogt PK. Transformation of chicken cells by the gene encoding the catalytic subunit of PI 3-kinase. Science. 1997;276:1848–50. doi: 10.1126/science.276.5320.1848. [DOI] [PubMed] [Google Scholar]
  19. Chantry D, Vojtek A, Kashishian A, Holtzman DA, Wood C, Gray PW, Cooper JA, Hoekstra MF. p110delta, a novel phosphatidylinositol 3-kinase catalytic subunit that associates with p85 and is expressed predominantly in leukocytes. J Biol Chem. 1997;272:19236–41. doi: 10.1074/jbc.272.31.19236. [DOI] [PubMed] [Google Scholar]
  20. Cheever ML, Sato TK, de Beer T, Kutateladze TG, Emr SD, Overduin M. Phox domain interaction with PtdIns(3)P targets the Vam7 t-SNARE to vacuole membranes. Nat Cell Biol. 2001;3:613–8. doi: 10.1038/35083000. [DOI] [PubMed] [Google Scholar]
  21. Clayton E, Bardi G, Bell SE, Chantry D, Downes CP, Gray A, Humphries LA, Rawlings D, Reynolds H, Vigorito E, Turner M. A crucial role for the p110delta subunit of phosphatidylinositol 3-kinase in B cell development and activation. J Exp Med. 2002;196:753–63. doi: 10.1084/jem.20020805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Coffer PJ, Woodgett JR. Molecular cloning and characterisation of a novel putative protein-serine kinase related to the cAMP-dependent and protein kinase C families. Eur J Biochem. 1991;201:475–81. doi: 10.1111/j.1432-1033.1991.tb16305.x. [DOI] [PubMed] [Google Scholar]
  23. Coloff JL, Rathmell JC. Metabolic regulation of Akt: roles reversed. J Cell Biol. 2006;175:845–7. doi: 10.1083/jcb.200610119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Courtneidge SA, Heber A. An 81 kd protein complexed with middle T antigen and pp60c-src: a possible phosphatidylinositol kinase. Cell. 1987;50:1031–7. doi: 10.1016/0092-8674(87)90169-3. [DOI] [PubMed] [Google Scholar]
  25. Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378:785–9. doi: 10.1038/378785a0. [DOI] [PubMed] [Google Scholar]
  26. Dan S, Yoshimi H, Okamura M, Mukai Y, Yamori T. Inhibition of PI3K by ZSTK474 suppressed tumor growth not via apoptosis but G0/G1 arrest. Biochem Biophys Res Commun. 2009;379:104–9. doi: 10.1016/j.bbrc.2008.12.015. [DOI] [PubMed] [Google Scholar]
  27. Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 1997;91:231–41. doi: 10.1016/s0092-8674(00)80405-5. [DOI] [PubMed] [Google Scholar]
  28. del Peso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. Science. 1997;278:687–9. doi: 10.1126/science.278.5338.687. [DOI] [PubMed] [Google Scholar]
  29. Di Paolo G, De Camilli P. Phosphoinositides in cell regulation and membrane dynamics. Nature. 2006;443:651–7. doi: 10.1038/nature05185. [DOI] [PubMed] [Google Scholar]
  30. Domin J, Pages F, Volinia S, Rittenhouse SE, Zvelebil MJ, Stein RC, Waterfield MD. Cloning of a human phosphoinositide 3-kinase with a C2 domain that displays reduced sensitivity to the inhibitor wortmannin. Biochem J. 1997;326(Pt 1):139–47. doi: 10.1042/bj3260139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Edgar KA, Wallin JJ, Berry M, Lee LB, Prior WW, Sampath D, Friedman LS, Belvin M. Isoform-specific phosphoinositide 3-kinase inhibitors exert distinct effects in solid tumors. Cancer Res. 70:1164–72. doi: 10.1158/0008-5472.CAN-09-2525. [DOI] [PubMed] [Google Scholar]
  32. Elis W, Triantafellow E, Wolters NM, Sian KR, Caponigro G, Borawski J, Gaither LA, Murphy LO, Finan PM, Mackeigan JP. Down-regulation of class II phosphoinositide 3-kinase alpha expression below a critical threshold induces apoptotic cell death. Mol Cancer Res. 2008;6:614–23. doi: 10.1158/1541-7786.MCR-07-0262. [DOI] [PubMed] [Google Scholar]
  33. Ellson CD, Gobert-Gosse S, Anderson KE, Davidson K, Erdjument-Bromage H, Tempst P, Thuring JW, Cooper MA, Lim ZY, Holmes AB, Gaffney PR, Coadwell J, Chilvers ER, Hawkins PT, Stephens LR. PtdIns(3)P regulates the neutrophil oxidase complex by binding to the PX domain of p40(phox) Nat Cell Biol. 2001;3:679–82. doi: 10.1038/35083076. [DOI] [PubMed] [Google Scholar]
  34. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, Maira M, McNamara K, Perera SA, Song Y, Chirieac LR, Kaur R, Lightbown A, Simendinger J, Li T, Padera RF, Garcia-Echeverria C, Weissleder R, Mahmood U, Cantley LC, Wong KK. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14:1351–6. doi: 10.1038/nm.1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Escobedo JA, Navankasattusas S, Kavanaugh WM, Milfay D, Fried VA, Williams LT. cDNA cloning of a novel 85 kd protein that has SH2 domains and regulates binding of PI3-kinase to the PDGF beta-receptor. Cell. 1991;65:75–82. doi: 10.1016/0092-8674(91)90409-r. [DOI] [PubMed] [Google Scholar]
  36. Faber AC, Li D, Song Y, Liang MC, Yeap BY, Bronson RT, Lifshits E, Chen Z, Maira SM, Garcia-Echeverria C, Wong KK, Engelman JA. Differential induction of apoptosis in HER2 and EGFR addicted cancers following PI3K inhibition. Proc Natl Acad Sci U S A. 2009;106:19503–8. doi: 10.1073/pnas.0905056106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Falasca M, Maffucci T. Rethinking phosphatidylinositol 3-monophosphate. Biochim Biophys Acta. 2009 doi: 10.1016/j.bbamcr.2009.10.003. [DOI] [PubMed] [Google Scholar]
  38. Fan QW, Cheng CK, Nicolaides TP, Hackett CS, Knight ZA, Shokat KM, Weiss WA. A dual phosphoinositide-3-kinase alpha/mTOR inhibitor cooperates with blockade of epidermal growth factor receptor in PTEN-mutant glioma. Cancer Res. 2007;67:7960–5. doi: 10.1158/0008-5472.CAN-07-2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Fine B, Hodakoski C, Koujak S, Su T, Saal LH, Maurer M, Hopkins B, Keniry M, Sulis ML, Mense S, Hibshoosh H, Parsons R. Activation of the PI3K pathway in cancer through inhibition of PTEN by exchange factor P-REX2a. Science. 2009;325:1261–5. doi: 10.1126/science.1173569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Flinn IA, Byrd JC, Furman RR, Brown JR, Benson DM, Coutre SE, Kahl BS, Smith BD, Wagner-Johnston ND, Spurgeon SE, Giese NA, Yu AS. Evidence of Clinical Activity in a Phase 1 Study of CAL-101, An Oral P110δ Isoform-Selective Inhibitor of Phosphatidylinositol 3-Kinase, in Patients with Relapsed or Refractory B-Cell Malignancies ASH 2009 Abstract. 2009 [Google Scholar]
  41. Foukas LC, Berenjeno IM, Gray A, Khwaja A, Vanhaesebroeck B. Activity of any class IA PI 3-kinase isoform can sustain cell proliferation and survival. Proc Natl Acad Sci U S A. 2010 doi: 10.1073/pnas.0906461107. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Foukas LC, Claret M, Pearce W, Okkenhaug K, Meek S, Peskett E, Sancho S, Smith AJ, Withers DJ, Vanhaesebroeck B. Critical role for the p110alpha phosphoinositide-3-OH kinase in growth and metabolic regulation. Nature. 2006;441:366–70. doi: 10.1038/nature04694. [DOI] [PubMed] [Google Scholar]
  43. Franke TF, Yang SI, Chan TO, Datta K, Kazlauskas A, Morrison DK, Kaplan DR, Tsichlis PN. The protein kinase encoded by the Akt proto-oncogene is a target of the PDGF-activated phosphatidylinositol 3-kinase. Cell. 1995;81:727–36. doi: 10.1016/0092-8674(95)90534-0. [DOI] [PubMed] [Google Scholar]
  44. Frech M, Andjelkovic M, Ingley E, Reddy KK, Falck JR, Hemmings BA. High affinity binding of inositol phosphates and phosphoinositides to the pleckstrin homology domain of RAC/protein kinase B and their influence on kinase activity. J Biol Chem. 1997;272:8474–81. doi: 10.1074/jbc.272.13.8474. [DOI] [PubMed] [Google Scholar]
  45. Fry MJ, Panayotou G, Dhand R, Ruiz-Larrea F, Gout I, Nguyen O, Courtneidge SA, Waterfield MD. Purification and characterization of a phosphatidylinositol 3-kinase complex from bovine brain by using phosphopeptide affinity columns. Biochem J. 1992;288(Pt 2):383–93. doi: 10.1042/bj2880383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gaullier JM, Simonsen A, D’Arrigo A, Bremnes B, Stenmark H, Aasland R. FYVE fingers bind PtdIns(3)P. Nature. 1998;394:432–3. doi: 10.1038/28767. [DOI] [PubMed] [Google Scholar]
  47. Gewinner C, Wang ZC, Richardson A, Teruya-Feldstein J, Etemadmoghadam D, Bowtell D, Barretina J, Lin WM, Rameh L, Salmena L, Pandolfi PP, Cantley LC. Evidence that inositol polyphosphate 4-phosphatase type II is a tumor suppressor that inhibits PI3K signaling. Cancer Cell. 2009;16:115–25. doi: 10.1016/j.ccr.2009.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Graupera M, Guillermet-Guibert J, Foukas LC, Phng LK, Cain RJ, Salpekar A, Pearce W, Meek S, Millan J, Cutillas PR, Smith AJ, Ridley AJ, Ruhrberg C, Gerhardt H, Vanhaesebroeck B. Angiogenesis selectively requires the p110alpha isoform of PI3K to control endothelial cell migration. Nature. 2008;453:662–6. doi: 10.1038/nature06892. [DOI] [PubMed] [Google Scholar]
  49. Guillard S, Clarke PA, Te Poele R, Mohri Z, Bjerke L, Valenti M, Raynaud F, Eccles SA, Workman P. Molecular pharmacology of phosphatidylinositol 3-kinase inhibition in human glioma. Cell Cycle. 2009;8:443–53. doi: 10.4161/cc.8.3.7643. [DOI] [PubMed] [Google Scholar]
  50. Gymnopoulos M, Elsliger MA, Vogt PK. Rare cancer-specific mutations in PIK3CA show gain of function. Proc Natl Acad Sci U S A. 2007;104:5569–74. doi: 10.1073/pnas.0701005104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Harlan JE, Hajduk PJ, Yoon HS, Fesik SW. Pleckstrin homology domains bind to phosphatidylinositol-4,5-bisphosphate. Nature. 1994;371:168–70. doi: 10.1038/371168a0. [DOI] [PubMed] [Google Scholar]
  52. Haslam RJ, Koide HB, Hemmings BA. Pleckstrin domain homology. Nature. 1993;363:309–10. doi: 10.1038/363309b0. [DOI] [PubMed] [Google Scholar]
  53. Hawkins PT, Jackson TR, Stephens LR. Platelet-derived growth factor stimulates synthesis of PtdIns(3,4,5)P3 by activating a PtdIns(4,5)P2 3-OH kinase. Nature. 1992;358:157–9. doi: 10.1038/358157a0. [DOI] [PubMed] [Google Scholar]
  54. Herman PK, Emr SD. Characterization of VPS34, a gene required for vacuolar protein sorting and vacuole segregation in Saccharomyces cerevisiae. Mol Cell Biol. 1990;10:6742–54. doi: 10.1128/mcb.10.12.6742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hiles ID, Otsu M, Volinia S, Fry MJ, Gout I, Dhand R, Panayotou G, Ruiz-Larrea F, Thompson A, Totty NF, et al. Phosphatidylinositol 3-kinase: structure and expression of the 110 kd catalytic subunit. Cell. 1992;70:419–29. doi: 10.1016/0092-8674(92)90166-a. [DOI] [PubMed] [Google Scholar]
  56. Hill K, Welti S, Yu J, Murray JT, Yip SC, Condeelis JS, Segall JE, Backer JM. Specific requirement for the p85-p110alpha phosphatidylinositol 3-kinase during epidermal growth factor-stimulated actin nucleation in breast cancer cells. J Biol Chem. 2000;275:3741–4. doi: 10.1074/jbc.275.6.3741. [DOI] [PubMed] [Google Scholar]
  57. Hirsch E, Katanaev VL, Garlanda C, Azzolino O, Pirola L, Silengo L, Sozzani S, Mantovani A, Altruda F, Wymann MP. Central role for G protein-coupled phosphoinositide 3-kinase gamma in inflammation. Science. 2000;287:1049–53. doi: 10.1126/science.287.5455.1049. [DOI] [PubMed] [Google Scholar]
  58. Hu P, Mondino A, Skolnik EY, Schlessinger J. Cloning of a novel, ubiquitously expressed human phosphatidylinositol 3-kinase and identification of its binding site on p85. Mol Cell Biol. 1993;13:7677–88. doi: 10.1128/mcb.13.12.7677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hurley JH. Membrane binding domains. Biochim Biophys Acta. 2006;1761:805–11. doi: 10.1016/j.bbalip.2006.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Isakoff SJ, Cardozo T, Andreev J, Li Z, Ferguson KM, Abagyan R, Lemmon MA, Aronheim A, Skolnik EY. Identification and analysis of PH domain-containing targets of phosphatidylinositol 3-kinase using a novel in vivo assay in yeast. EMBO J. 1998;17:5374–87. doi: 10.1093/emboj/17.18.5374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Jackson SP, Schoenwaelder SM, Goncalves I, Nesbitt WS, Yap CL, Wright CE, Kenche V, Anderson KE, Dopheide SM, Yuan Y, Sturgeon SA, Prabaharan H, Thompson PE, Smith GD, Shepherd PR, Daniele N, Kulkarni S, Abbott B, Saylik D, Jones C, Lu L, Giuliano S, Hughan SC, Angus JA, Robertson AD, Salem HH. PI 3-kinase p110beta: a new target for antithrombotic therapy. Nat Med. 2005;11:507–14. doi: 10.1038/nm1232. [DOI] [PubMed] [Google Scholar]
  62. Jackson TR, Stephens LR, Hawkins PT. Receptor specificity of growth factor-stimulated synthesis of 3-phosphorylated inositol lipids in Swiss 3T3 cells. J Biol Chem. 1992;267:16627–36. [PubMed] [Google Scholar]
  63. Jacoby M, Cox JJ, Gayral S, Hampshire DJ, Ayub M, Blockmans M, Pernot E, Kisseleva MV, Compere P, Schiffmann SN, Gergely F, Riley JH, Perez-Morga D, Woods CG, Schurmans S. INPP5E mutations cause primary cilium signaling defects, ciliary instability and ciliopathies in human and mouse. Nat Genet. 2009;41:1027–31. doi: 10.1038/ng.427. [DOI] [PubMed] [Google Scholar]
  64. Jaiswal BS, Janakiraman V, Kljavin NM, Chaudhuri S, Stern HM, Wang W, Kan Z, Dbouk HA, Peters BA, Waring P, Dela Vega T, Kenski DM, Bowman KK, Lorenzo M, Li H, Wu J, Modrusan Z, Stinson J, Eby M, Yue P, Kaminker JS, de Sauvage FJ, Backer JM, Seshagiri S. Somatic mutations in p85alpha promote tumorigenesis through class IA PI3K activation. Cancer Cell. 2009;16:463–74. doi: 10.1016/j.ccr.2009.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Jones PF, Jakubowicz T, Pitossi FJ, Maurer F, Hemmings BA. Molecular cloning and identification of a serine/threonine protein kinase of the second-messenger subfamily. Proc Natl Acad Sci U S A. 1991;88:4171–5. doi: 10.1073/pnas.88.10.4171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009;23:537–48. doi: 10.1101/gad.1756509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Jou ST, Carpino N, Takahashi Y, Piekorz R, Chao JR, Wang D, Ihle JN. Essential, nonredundant role for the phosphoinositide 3-kinase p110delta in signaling by the B-cell receptor complex. Mol Cell Biol. 2002;22:8580–91. doi: 10.1128/MCB.22.24.8580-8591.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kanai F, Liu H, Field SJ, Akbary H, Matsuo T, Brown GE, Cantley LC, Yaffe MB. The PX domains of p47phox and p40phox bind to lipid products of PI(3)K. Nat Cell Biol. 2001;3:675–8. doi: 10.1038/35083070. [DOI] [PubMed] [Google Scholar]
  69. Kaplan DR, Whitman M, Schaffhausen B, Pallas DC, White M, Cantley L, Roberts TM. Common elements in growth factor stimulation and oncogenic transformation: 85 kd phosphoprotein and phosphatidylinositol kinase activity. Cell. 1987;50:1021–9. doi: 10.1016/0092-8674(87)90168-1. [DOI] [PubMed] [Google Scholar]
  70. Klarlund JK, Guilherme A, Holik JJ, Virbasius JV, Chawla A, Czech MP. Signaling by phosphoinositide-3,4,5-trisphosphate through proteins containing pleckstrin and Sec7 homology domains. Science. 1997;275:1927–30. doi: 10.1126/science.275.5308.1927. [DOI] [PubMed] [Google Scholar]
  71. Kops GJ, de Ruiter ND, De Vries-Smits AM, Powell DR, Bos JL, Burgering BM. Direct control of the Forkhead transcription factor AFX by protein kinase B. Nature. 1999;398:630–4. doi: 10.1038/19328. [DOI] [PubMed] [Google Scholar]
  72. Krugmann S, Anderson KE, Ridley SH, Risso N, McGregor A, Coadwell J, Davidson K, Eguinoa A, Ellson CD, Lipp P, Manifava M, Ktistakis N, Painter G, Thuring JW, Cooper MA, Lim ZY, Holmes AB, Dove SK, Michell RH, Grewal A, Nazarian A, Erdjument-Bromage H, Tempst P, Stephens LR, Hawkins PT. Identification of ARAP3, a novel PI3K effector regulating both Arf and Rho GTPases, by selective capture on phosphoinositide affinity matrices. Mol Cell. 2002;9:95–108. doi: 10.1016/s1097-2765(02)00434-3. [DOI] [PubMed] [Google Scholar]
  73. Leevers SJ, Weinkove D, MacDougall LK, Hafen E, Waterfield MD. The Drosophila phosphoinositide 3-kinase Dp110 promotes cell growth. EMBO J. 1996;15:6584–94. [PMC free article] [PubMed] [Google Scholar]
  74. Lemmon MA. Membrane recognition by phospholipid-binding domains. Nat Rev Mol Cell Biol. 2008;9:99–111. doi: 10.1038/nrm2328. [DOI] [PubMed] [Google Scholar]
  75. Li Z, Jiang H, Xie W, Zhang Z, Smrcka AV, Wu D. Roles of PLC-beta2 and -beta3 and PI3Kgamma in chemoattractant-mediated signal transduction. Science. 2000;287:1046–9. doi: 10.1126/science.287.5455.1046. [DOI] [PubMed] [Google Scholar]
  76. Macara IG, Marinetti GV, Balduzzi PC. Transforming protein of avian sarcoma virus UR2 is associated with phosphatidylinositol kinase activity: possible role in tumorigenesis. Proc Natl Acad Sci U S A. 1984;81:2728–32. doi: 10.1073/pnas.81.9.2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. MacDougall LK, Domin J, Waterfield MD. A family of phosphoinositide 3-kinases in Drosophila identifies a new mediator of signal transduction. Curr Biol. 1995;5:1404–15. doi: 10.1016/s0960-9822(95)00278-8. [DOI] [PubMed] [Google Scholar]
  78. Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem. 1998;273:13375–8. doi: 10.1074/jbc.273.22.13375. [DOI] [PubMed] [Google Scholar]
  79. Mandelker D, Gabelli SB, Schmidt-Kittler O, Zhu J, Cheong I, Huang CH, Kinzler KW, Vogelstein B, Amzel LM. A frequent kinase domain mutation that changes the interaction between PI3Kalpha and the membrane. Proc Natl Acad Sci U S A. 2009;106:16996–7001. doi: 10.1073/pnas.0908444106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Mayer BJ, Ren R, Clark KL, Baltimore D. A putative modular domain present in diverse signaling proteins. Cell. 1993;73:629–30. doi: 10.1016/0092-8674(93)90244-k. [DOI] [PubMed] [Google Scholar]
  81. Miled N, Yan Y, Hon WC, Perisic O, Zvelebil M, Inbar Y, Schneidman-Duhovny D, Wolfson HJ, Backer JM, Williams RL. Mechanism of two classes of cancer mutations in the phosphoinositide 3-kinase catalytic subunit. Science. 2007;317:239–42. doi: 10.1126/science.1135394. [DOI] [PubMed] [Google Scholar]
  82. Misawa H, Ohtsubo M, Copeland NG, Gilbert DJ, Jenkins NA, Yoshimura A. Cloning and characterization of a novel class II phosphoinositide 3-kinase containing C2 domain. Biochem Biophys Res Commun. 1998;244:531–9. doi: 10.1006/bbrc.1998.8294. [DOI] [PubMed] [Google Scholar]
  83. Morgan SJ, Smith AD, Parker PJ. Purification and characterization of bovine brain type I phosphatidylinositol kinase. Eur J Biochem. 1990;191:761–7. doi: 10.1111/j.1432-1033.1990.tb19185.x. [DOI] [PubMed] [Google Scholar]
  84. Morris JZ, Tissenbaum HA, Ruvkun G. A phosphatidylinositol-3-OH kinase family member regulating longevity and diapause in Caenorhabditis elegans. Nature. 1996;382:536–9. doi: 10.1038/382536a0. [DOI] [PubMed] [Google Scholar]
  85. Morrow CJ, Gray A, Dive C. Comparison of phosphatidylinositol-3-kinase signalling within a panel of human colorectal cancer cell lines with mutant or wild-type PIK3CA. FEBS Lett. 2005;579:5123–8. doi: 10.1016/j.febslet.2005.07.096. [DOI] [PubMed] [Google Scholar]
  86. Mu FT, Callaghan JM, Steele-Mortimer O, Stenmark H, Parton RG, Campbell PL, McCluskey J, Yeo JP, Tock EP, Toh BH. EEA1, an early endosome-associated protein. EEA1 is a conserved alpha-helical peripheral membrane protein flanked by cysteine “fingers” and contains a calmodulin-binding IQ motif. J Biol Chem. 1995;270:13503–11. doi: 10.1074/jbc.270.22.13503. [DOI] [PubMed] [Google Scholar]
  87. Nicot AS, Laporte J. Endosomal phosphoinositides and human diseases. Traffic. 2008;9:1240–9. doi: 10.1111/j.1600-0854.2008.00754.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Nobukuni T, Joaquin M, Roccio M, Dann SG, Kim SY, Gulati P, Byfield MP, Backer JM, Natt F, Bos JL, Zwartkruis FJ, Thomas G. Amino acids mediate mTOR/raptor signaling through activation of class 3 phosphatidylinositol 3OH-kinase. Proc Natl Acad Sci U S A. 2005;102:14238–43. doi: 10.1073/pnas.0506925102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, Tissenbaum HA, Ruvkun G. The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997;389:994–9. doi: 10.1038/40194. [DOI] [PubMed] [Google Scholar]
  90. Okada T, Sakuma L, Fukui Y, Hazeki O, Ui M. Blockage of chemotactic peptide-induced stimulation of neutrophils by wortmannin as a result of selective inhibition of phosphatidylinositol 3-kinase. J Biol Chem. 1994;269:3563–7. [PubMed] [Google Scholar]
  91. Okkenhaug K, Bilancio A, Farjot G, Priddle H, Sancho S, Peskett E, Pearce W, Meek SE, Salpekar A, Waterfield MD, Smith AJ, Vanhaesebroeck B. Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice. Science. 2002;297:1031–4. doi: 10.1126/science.1073560. [DOI] [PubMed] [Google Scholar]
  92. Ono F, Nakagawa T, Saito S, Owada Y, Sakagami H, Goto K, Suzuki M, Matsuno S, Kondo H. A novel class II phosphoinositide 3-kinase predominantly expressed in the liver and its enhanced expression during liver regeneration. J Biol Chem. 1998;273:7731–6. doi: 10.1074/jbc.273.13.7731. [DOI] [PubMed] [Google Scholar]
  93. Ooms LM, Horan KA, Rahman P, Seaton G, Gurung R, Kethesparan DS, Mitchell CA. The role of the inositol polyphosphate 5-phosphatases in cellular function and human disease. Biochem J. 2009;419:29–49. doi: 10.1042/BJ20081673. [DOI] [PubMed] [Google Scholar]
  94. Otsu M, Hiles I, Gout I, Fry MJ, Ruiz-Larrea F, Panayotou G, Thompson A, Dhand R, Hsuan J, Totty N, et al. Characterization of two 85 kd proteins that associate with receptor tyrosine kinases, middle-T/pp60c-src complexes, and PI3-kinase. Cell. 1991;65:91–104. doi: 10.1016/0092-8674(91)90411-q. [DOI] [PubMed] [Google Scholar]
  95. Pang H, Flinn R, Patsialou A, Wyckoff J, Roussos ET, Wu H, Pozzuto M, Goswami S, Condeelis JS, Bresnick AR, Segall JE, Backer JM. Differential enhancement of breast cancer cell motility and metastasis by helical and kinase domain mutations of class IA phosphoinositide 3-kinase. Cancer Res. 2009;69:8868–76. doi: 10.1158/0008-5472.CAN-09-1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Papakonstanti EA, Ridley AJ, Vanhaesebroeck B. The p110delta isoform of PI 3-kinase negatively controls RhoA and PTEN. EMBO J. 2007;26:3050–61. doi: 10.1038/sj.emboj.7601763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Papakonstanti EA, Zwaenepoel O, Bilancio A, Burns E, Nock GE, Houseman B, Shokat K, Ridley AJ, Vanhaesebroeck B. Distinct roles of class IA PI3K isoforms in primary and immortalised macrophages. J Cell Sci. 2008;121:4124–33. doi: 10.1242/jcs.032763. [DOI] [PubMed] [Google Scholar]
  98. Paradis S, Ruvkun G. Caenorhabditis elegans Akt/PKB transduces insulin receptor-like signals from AGE-1 PI3 kinase to the DAF-16 transcription factor. Genes Dev. 1998;12:2488–98. doi: 10.1101/gad.12.16.2488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA, Jr., Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321:1807–12. doi: 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Philp AJ, Campbell IG, Leet C, Vincan E, Rockman SP, Whitehead RH, Thomas RJ, Phillips WA. The phosphatidylinositol 3′-kinase p85alpha gene is an oncogene in human ovarian and colon tumors. Cancer Res. 2001;61:7426–9. [PubMed] [Google Scholar]
  101. Plas DR, Thompson CB. Akt-dependent transformation: there is more to growth than just surviving. Oncogene. 2005;24:7435–42. doi: 10.1038/sj.onc.1209097. [DOI] [PubMed] [Google Scholar]
  102. Powis G, Bonjouklian R, Berggren MM, Gallegos A, Abraham R, Ashendel C, Zalkow L, Matter WF, Dodge J, Grindey G, et al. Wortmannin, a potent and selective inhibitor of phosphatidylinositol-3-kinase. Cancer Res. 1994;54:2419–23. [PubMed] [Google Scholar]
  103. Raynaud FI, Eccles S, Clarke PA, Hayes A, Nutley B, Alix S, Henley A, Di-Stefano F, Ahmad Z, Guillard S, Bjerke LM, Kelland L, Valenti M, Patterson L, Gowan S, de Haven Brandon A, Hayakawa M, Kaizawa H, Koizumi T, Ohishi T, Patel S, Saghir N, Parker P, Waterfield M, Workman P. Pharmacologic characterization of a potent inhibitor of class I phosphatidylinositide 3-kinases. Cancer Res. 2007;67:5840–50. doi: 10.1158/0008-5472.CAN-06-4615. [DOI] [PubMed] [Google Scholar]
  104. Roche S, Downward J, Raynal P, Courtneidge SA. A function for phosphatidylinositol 3-kinase beta (p85alpha-p110beta) in fibroblasts during mitogenesis: requirement for insulin- and lysophosphatidic acid-mediated signal transduction. Mol Cell Biol. 1998;18:7119–29. doi: 10.1128/mcb.18.12.7119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Roche S, Koegl M, Courtneidge SA. The phosphatidylinositol 3-kinase alpha is required for DNA synthesis induced by some, but not all, growth factors. Proc Natl Acad Sci U S A. 1994;91:9185–9. doi: 10.1073/pnas.91.19.9185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Rodriguez-Viciana P, Warne PH, Dhand R, Vanhaesebroeck B, Gout I, Fry MJ, Waterfield MD, Downward J. Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 1994;370:527–32. doi: 10.1038/370527a0. [DOI] [PubMed] [Google Scholar]
  107. Rommel C, Camps M, Ji H. PI3K delta and PI3K gamma: partners in crime in inflammation in rheumatoid arthritis and beyond? Nat Rev Immunol. 2007;7:191–201. doi: 10.1038/nri2036. [DOI] [PubMed] [Google Scholar]
  108. Ruckle T, Schwarz MK, Rommel C. PI3Kgamma inhibition: towards an ‘aspirin of the 21st century’? Nat Rev Drug Discov. 2006;5:903–18. doi: 10.1038/nrd2145. [DOI] [PubMed] [Google Scholar]
  109. Sadhu C, Masinovsky B, Dick K, Sowell CG, Staunton DE. Essential role of phosphoinositide 3-kinase delta in neutrophil directional movement. J Immunol. 2003;170:2647–54. doi: 10.4049/jimmunol.170.5.2647. [DOI] [PubMed] [Google Scholar]
  110. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler KW, Vogelstein B, Velculescu VE. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
  111. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307:1098–101. doi: 10.1126/science.1106148. [DOI] [PubMed] [Google Scholar]
  112. Sasaki T, Irie-Sasaki J, Jones RG, Oliveira-dos-Santos AJ, Stanford WL, Bolon B, Wakeham A, Itie A, Bouchard D, Kozieradzki I, Joza N, Mak TW, Ohashi PS, Suzuki A, Penninger JM. Function of PI3Kgamma in thymocyte development, T cell activation, and neutrophil migration. Science. 2000;287:1040–6. doi: 10.1126/science.287.5455.1040. [DOI] [PubMed] [Google Scholar]
  113. Schu PV, Takegawa K, Fry MJ, Stack JH, Waterfield MD, Emr SD. Phosphatidylinositol 3-kinase encoded by yeast VPS34 gene essential for protein sorting. Science. 1993;260:88–91. doi: 10.1126/science.8385367. [DOI] [PubMed] [Google Scholar]
  114. Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, Moasser MM. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 2007;445:437–41. doi: 10.1038/nature05474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Shibasaki F, Homma Y, Takenawa T. Two types of phosphatidylinositol 3-kinase from bovine thymus. Monomer and heterodimer form. J Biol Chem. 1991;266:8108–14. [PubMed] [Google Scholar]
  116. Shuttleworth S, Silva F, Tomassi C, Cecil A, Hill T, Rogers H, Townsend P. Progress in the design and development of phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of chronic diseases. Progress in Medicinal Chemistry. 2009;48:81–131. doi: 10.1016/s0079-6468(09)04803-6. [DOI] [PubMed] [Google Scholar]
  117. Sjolander A, Yamamoto K, Huber BE, Lapetina EG. Association of p21ras with phosphatidylinositol 3-kinase. Proc Natl Acad Sci U S A. 1991;88:7908–12. doi: 10.1073/pnas.88.18.7908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Skolnik EY, Margolis B, Mohammadi M, Lowenstein E, Fischer R, Drepps A, Ullrich A, Schlessinger J. Cloning of PI3 kinase-associated p85 utilizing novel method for expression/cloning of target proteins for receptor tyrosine kinases. Cell. 1991;65:83–90. doi: 10.1016/0092-8674(91)90410-z. [DOI] [PubMed] [Google Scholar]
  119. Song X, Xu W, Zhang A, Huang G, Liang X, Virbasius JV, Czech MP, Zhou GW. Phox homology domains specifically bind phosphatidylinositol phosphates. Biochemistry. 2001;40:8940–4. doi: 10.1021/bi0155100. [DOI] [PubMed] [Google Scholar]
  120. Soond DR, Bjorgo E, Moltu K, Dale VQ, Patton DT, Torgersen KM, Galleway F, Twomey B, Clark J, Gaston JH, Tasken K, Bunyard P, Okkenhaug K. PI3K p110{delta} regulates T cell cytokine production during primary and secondary immune responses in mice and humans. Blood. 2010 doi: 10.1182/blood-2009-07-232330. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Sos ML, Fischer S, Ullrich R, Peifer M, Heuckmann JM, Koker M, Heynck S, Stuckrath I, Weiss J, Fischer F, Michel K, Goel A, Regales L, Politi KA, Perera S, Getlik M, Heukamp LC, Ansen S, Zander T, Beroukhim R, Kashkar H, Shokat KM, Sellers WR, Rauh D, Orr C, Hoeflich KP, Friedman L, Wong KK, Pao W, Thomas RK. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer. Proc Natl Acad Sci U S A. 2009;106:18351–6. doi: 10.1073/pnas.0907325106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL, Davies M, Carey M, Hu Z, Guan Y, Sahin A, Symmans WF, Pusztai L, Nolden LK, Horlings H, Berns K, Hung MC, van de Vijver MJ, Valero V, Gray JW, Bernards R, Mills GB, Hennessy BT. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008;68:6084–91. doi: 10.1158/0008-5472.CAN-07-6854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Stenmark H, Aasland R, Toh BH, D’Arrigo A. Endosomal localization of the autoantigen EEA1 is mediated by a zinc-binding FYVE finger. J Biol Chem. 1996;271:24048–54. doi: 10.1074/jbc.271.39.24048. [DOI] [PubMed] [Google Scholar]
  124. Stephens L, Anderson K, Stokoe D, Erdjument-Bromage H, Painter GF, Holmes AB, Gaffney PR, Reese CB, McCormick F, Tempst P, Coadwell J, Hawkins PT. Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B. Science. 1998;279:710–4. doi: 10.1126/science.279.5351.710. [DOI] [PubMed] [Google Scholar]
  125. Stephens L, Smrcka A, Cooke FT, Jackson TR, Sternweis PC, Hawkins PT. A novel phosphoinositide 3 kinase activity in myeloid-derived cells is activated by G protein beta gamma subunits. Cell. 1994;77:83–93. doi: 10.1016/0092-8674(94)90237-2. [DOI] [PubMed] [Google Scholar]
  126. Stephens LR, Eguinoa A, Erdjument-Bromage H, Lui M, Cooke F, Coadwell J, Smrcka AS, Thelen M, Cadwallader K, Tempst P, Hawkins PT. The G beta gamma sensitivity of a PI3K is dependent upon a tightly associated adaptor, p101. Cell. 1997;89:105–14. doi: 10.1016/s0092-8674(00)80187-7. [DOI] [PubMed] [Google Scholar]
  127. Stocker H, Andjelkovic M, Oldham S, Laffargue M, Wymann MP, Hemmings BA, Hafen E. Living with lethal PIP3 levels: viability of flies lacking PTEN restored by a PH domain mutation in Akt/PKB. Science. 2002;295:2088–91. doi: 10.1126/science.1068094. [DOI] [PubMed] [Google Scholar]
  128. Stokoe D, Stephens LR, Copeland T, Gaffney PR, Reese CB, Painter GF, Holmes AB, McCormick F, Hawkins PT. Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B. Science. 1997;277:567–70. doi: 10.1126/science.277.5325.567. [DOI] [PubMed] [Google Scholar]
  129. Stoyanov B, Volinia S, Hanck T, Rubio I, Loubtchenkov M, Malek D, Stoyanova S, Vanhaesebroeck B, Dhand R, Nurnberg B, et al. Cloning and characterization of a G protein-activated human phosphoinositide-3 kinase. Science. 1995;269:690–3. doi: 10.1126/science.7624799. [DOI] [PubMed] [Google Scholar]
  130. Sugimoto Y, Whitman M, Cantley LC, Erikson RL. Evidence that the Rous sarcoma virus transforming gene product phosphorylates phosphatidylinositol and diacylglycerol. Proc Natl Acad Sci U S A. 1984;81:2117–21. doi: 10.1073/pnas.81.7.2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. TGCA Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–8. doi: 10.1038/nature07385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Thomas RK, Baker AC, Debiasi RM, Winckler W, Laframboise T, Lin WM, Wang M, Feng W, Zander T, MacConaill L, Lee JC, Nicoletti R, Hatton C, Goyette M, Girard L, Majmudar K, Ziaugra L, Wong KK, Gabriel S, Beroukhim R, Peyton M, Barretina J, Dutt A, Emery C, Greulich H, Shah K, Sasaki H, Gazdar A, Minna J, Armstrong SA, Mellinghoff IK, Hodi FS, Dranoff G, Mischel PS, Cloughesy TF, Nelson SF, Liau LM, Mertz K, Rubin MA, Moch H, Loda M, Catalona W, Fletcher J, Signoretti S, Kaye F, Anderson KC, Demetri GD, Dummer R, Wagner S, Herlyn M, Sellers WR, Meyerson M, Garraway LA. High-throughput oncogene mutation profiling in human cancer. Nat Genet. 2007;39:347–51. doi: 10.1038/ng1975. [DOI] [PubMed] [Google Scholar]
  133. Traynor-Kaplan AE, Harris AL, Thompson BL, Taylor P, Sklar LA. An inositol tetrakisphosphate-containing phospholipid in activated neutrophils. Nature. 1988;334:353–6. doi: 10.1038/334353a0. [DOI] [PubMed] [Google Scholar]
  134. Traynor-Kaplan AE, Thompson BL, Harris AL, Taylor P, Omann GM, Sklar LA. Transient increase in phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol trisphosphate during activation of human neutrophils. J Biol Chem. 1989;264:15668–73. [PubMed] [Google Scholar]
  135. Vanhaesebroeck B, Guillermet-Guibert J, Graupera M, Bilanges B. The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol. 2010 doi: 10.1038/nrm2882. In press. [DOI] [PubMed] [Google Scholar]
  136. Vanhaesebroeck B, Jones GE, Allen WE, Zicha D, Hooshmand-Rad R, Sawyer C, Wells C, Waterfield MD, Ridley AJ. Distinct PI(3)Ks mediate mitogenic signalling and cell migration in macrophages. Nat Cell Biol. 1999;1:69–71. doi: 10.1038/9045. [DOI] [PubMed] [Google Scholar]
  137. Vanhaesebroeck B, Leevers SJ, Ahmadi K, Timms J, Katso R, Driscoll PC, Woscholski R, Parker PJ, Waterfield MD. Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem. 2001;70:535–602. doi: 10.1146/annurev.biochem.70.1.535. [DOI] [PubMed] [Google Scholar]
  138. Vanhaesebroeck B, Leevers SJ, Panayotou G, Waterfield MD. Phosphoinositide 3-kinases: a conserved family of signal transducers. Trends Biochem Sci. 1997a;22:267–72. doi: 10.1016/s0968-0004(97)01061-x. [DOI] [PubMed] [Google Scholar]
  139. Vanhaesebroeck B, Welham MJ, Kotani K, Stein R, Warne PH, Zvelebil MJ, Higashi K, Volinia S, Downward J, Waterfield MD. P110delta, a novel phosphoinositide 3-kinase in leukocytes. Proc Natl Acad Sci U S A. 1997b;94:4330–5. doi: 10.1073/pnas.94.9.4330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Vasudevan KM, Barbie DA, Davies MA, Rabinovsky R, McNear CJ, Kim JJ, Hennessy BT, Tseng H, Pochanard P, Kim SY, Dunn IF, Schinzel AC, Sandy P, Hoersch S, Sheng Q, Gupta PB, Boehm JS, Reiling JH, Silver S, Lu Y, Stemke-Hale K, Dutta B, Joy C, Sahin AA, Gonzalez-Angulo AM, Lluch A, Rameh LE, Jacks T, Root DE, Lander ES, Mills GB, Hahn WC, Sellers WR, Garraway LA. AKT-independent signaling downstream of oncogenic PIK3CA mutations in human cancer. Cancer Cell. 2009;16:21–32. doi: 10.1016/j.ccr.2009.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Virbasius JV, Guilherme A, Czech MP. Mouse p170 is a novel phosphatidylinositol 3-kinase containing a C2 domain. J Biol Chem. 1996;271:13304–7. doi: 10.1074/jbc.271.23.13304. [DOI] [PubMed] [Google Scholar]
  142. Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002) J Biol Chem. 1994;269:5241–8. [PubMed] [Google Scholar]
  143. Walker EH, Perisic O, Ried C, Stephens L, Williams RL. Structural insights into phosphoinositide 3-kinase catalysis and signalling. Nature. 1999;402:313–20. doi: 10.1038/46319. [DOI] [PubMed] [Google Scholar]
  144. Welch HC, Coadwell WJ, Ellson CD, Ferguson GJ, Andrews SR, Erdjument-Bromage H, Tempst P, Hawkins PT, Stephens LR. P-Rex1, a PtdIns(3,4,5)P3- and Gbetagamma-regulated guanine-nucleotide exchange factor for Rac. Cell. 2002;108:809–21. doi: 10.1016/s0092-8674(02)00663-3. [DOI] [PubMed] [Google Scholar]
  145. Whitman M, Downes CP, Keeler M, Keller T, Cantley L. Type I phosphatidylinositol kinase makes a novel inositol phospholipid, phosphatidylinositol-3-phosphate. Nature. 1988;332:644–6. doi: 10.1038/332644a0. [DOI] [PubMed] [Google Scholar]
  146. Whitman M, Kaplan DR, Schaffhausen B, Cantley L, Roberts TM. Association of phosphatidylinositol kinase activity with polyoma middle-T competent for transformation. Nature. 1985;315:239–42. doi: 10.1038/315239a0. [DOI] [PubMed] [Google Scholar]
  147. Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y, Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV, Ballinger DG, Sparks AB, Hartigan J, Smith DR, Suh E, Papadopoulos N, Buckhaults P, Markowitz SD, Parmigiani G, Kinzler KW, Velculescu VE, Vogelstein B. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108–13. doi: 10.1126/science.1145720. [DOI] [PubMed] [Google Scholar]
  148. Xu Y, Hortsman H, Seet L, Wong SH, Hong W. SNX3 regulates endosomal function through its PX-domain-mediated interaction with PtdIns(3)P. Nat Cell Biol. 2001;3:658–66. doi: 10.1038/35083051. [DOI] [PubMed] [Google Scholar]
  149. Yano H, Nakanishi S, Kimura K, Hanai N, Saitoh Y, Fukui Y, Nonomura Y, Matsuda Y. Inhibition of histamine secretion by wortmannin through the blockade of phosphatidylinositol 3-kinase in RBL-2H3 cells. J Biol Chem. 1993;268:25846–56. [PubMed] [Google Scholar]
  150. Yao R, Cooper GM. Requirement for phosphatidylinositol-3 kinase in the prevention of apoptosis by nerve growth factor. Science. 1995;267:2003–6. doi: 10.1126/science.7701324. [DOI] [PubMed] [Google Scholar]
  151. Zhou K, Takegawa K, Emr SD, Firtel RA. A phosphatidylinositol (PI) kinase gene family in Dictyostelium discoideum: biological roles of putative mammalian p110 and yeast Vps34p PI 3-kinase homologs during growth and development. Mol Cell Biol. 1995;15:5645–56. doi: 10.1128/mcb.15.10.5645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Zvelebil MJ, MacDougall L, Leevers S, Volinia S, Vanhaesebroeck B, Gout I, Panayotou G, Domin J, Stein R, Pages F, et al. Structural and functional diversity of phosphoinositide 3-kinases. Philos Trans R Soc Lond B Biol Sci. 1996;351:217–23. doi: 10.1098/rstb.1996.0019. [DOI] [PubMed] [Google Scholar]

RESOURCES