Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 1.
Published in final edited form as: Mol Cancer Res. 2021 Apr 30;19(8):1251–1257. doi: 10.1158/1541-7786.MCR-20-1066

Emerging Roles for AKT Isoform Preference in Cancer Progression Pathways

Seamus E Degan 1, Irwin H Gelman 1
PMCID: PMC8349788  NIHMSID: NIHMS1701740  PMID: 33931488

Abstract

The phosphoinositol-3 kinase (PI3K)-AKT pathway is one of the most mutated in human cancers, predominantly associated with the loss of the signaling antagonist, PTEN, and to lesser extents, with gain-of-function mutations in PIK3CA (encoding PI3K-p110α) and AKT1. In addition, most oncogenic driver pathways activate PI3K/AKT signaling. Nonetheless, drugs targeting PI3K or AKT have fared poorly against solid tumors in clinical trials as monotherapies, yet some have shown efficacy when combined with inhibitors of other oncogenic drivers, such as receptor tyrosine kinases or nuclear hormone receptors. There is growing evidence that AKT isoforms, AKT1, AKT2, and AKT3, have different, often distinct roles in either promoting or suppressing specific parameters of oncogenic progression, yet few if any isoform-preferred substrates have been characterized. This review will describe recent data showing that the differential activation of AKT isoforms is mediated by complex interplays between PTEN, PI3K isoforms and upstream tyrosine kinases, and that the efficacy of PI3K/AKT inhibitors will likely depend on the successful targeting of specific AKT isoforms and their preferred pathways.

Background

AKT (also known as protein kinase B (PKB)) is a serine/threonine kinase first identified in 1987 as the oncogene of the acute transforming retrovirus, AKT8, corresponding to AKT1 and AKT2 human proto-oncogene homologues (1). A third isoform, AKT3, was identified a decade later (2). Further research identified phosphoinositide 3-kinase (PI3K) as the upstream signaling activator of AKT (3) through its ability to generate PIP3 (4), and that the phosphatase and tensin homologue (PTEN) functions as a negative regulator of this PI3K/AKT pathway based on the ability of its lipid phosphatase activity to revert PIP3 to PIP2 (5,6) (Figure 1). The notion that AKT isoforms play non-redundant roles was made evident even at the initial discovery publication, which showed gene amplifications of AKT1, but not AKT2, in gastric adenocarcinomas (1). These non-redundant roles were further evidenced by the findings that Akt1−/− mice suffer embryonic lethality, Akt2−/− mice develop severe diabetic phenotypes, and Akt3−/− mice predominantly exhibit reduced brain sizes rather than cognitive deficiencies (711). Akt1−/− that do survive also exhibit impaired body growth (7). Only Akt2−/−;Akt3−/− intertypic crosses were viable, suggesting both compensatory and non-redundant roles in development (9).

Figure 1: AKT pathways and mediators.

Figure 1:

The activation of PI3K via receptor tyrosine kinases (RTK) or G-protein coupled receptors (GPCR) facilitates AKT-regulated cell growth, proliferation, survival, migration, and metabolisms mediated by the phosphorylation of substrates such as Palladin (PALLD), p21Waf1 (CDKN1A), S6K (RPS6KB1), FOXO family members or GSK3β. Actuation of maximal AKT serine/threonine kinase activity is mediated by a feedback cycle involving activation of mTORC2 followed by its phosphorylation of AKT on Ser473, and by PDK1 phosphorylation of AKT on Thr308.

AKT activation is regulated by a multilayer process, beginning with PI3K activation by receptors tyrosine kinases or G-protein coupled receptors. A major role for PI3K is as a lipid kinase that phosphorylates phosphoinositol 4,5-bisphosphate (PIP2) to produce PIP3. AKT is then recruited to plasma membrane sites through the interaction of its Pleckstrin Homology (PH) domain to PIP3 islands in the plasma membrane (12). Indeed, roughly 1% of all cancers (cBioPortal analysis of MSK-IMPACT pan-cancer cohort) have an E17K mutation in the AKT1-PH domain, resulting in increased association with PIP2 and oncogenicity in embryo fibroblasts (13). Following this, AKT is phosphorylated at two sites, T308 and S473, both of which are required for full activation: T308 is phosphorylated first by PDK1,which triggers a positive feedback mechanism mediated by the mTORC2 complex which phosphorylates AKT at S473, resulting in its maximal activation (1416). The finding that AKT-S473 phosphorylation can be carried out by other kinases, including DNA-PK and PKCγ (17,18) complicates the use of AKTpoS473 as a marker of PI3K activation1.

Shared and Distinct Roles of AKT Isoforms in Cellular Biology and Cancer

AKT is responsible for a variety of key parameters of normal or oncogenic cell behavior, including cell migration, proliferation, invasion, survival and metabolism (1922). Many of the roles for AKT isoforms are, in fact, redundant (Figure 2). These include negative regulation of i) Bcl-2 family proteins, ii) GSK3 isoforms responsible for cell survival and cellular metabolism, or iii) TSC2, a known tumor suppressor (23). There has been increased attention on defining both shared and isoform-specific AKT roles in these biologies. Noting that AKT activation regulates cellular energetics through the control of intracellular ATP levels (24,25), multiple studies identified key roles for AKT2 in mediating metabolic regulation induced in response to insulin or glucose stimulation, lipogenesis or GLUT4 trafficking (8,2628). However, there is also a role for AKT1 in metabolism including insulin production in pancreatic islet cells and glucose metabolism in breast tissue during lactation (29,30). Similarly, although AKT2 was shown to play a more dominant role in tumor migration and invasion, some overlapping roles for AKT1 are also identified (31,32). Indeed, a demonstrated E17K PH domain oncogenic mutation in AKT1 has been identified in 3–5% of all cancers in cBioPortal, and equivalent putative oncogenic mutations for AKT2 and AKT3, though less frequent, have also been identified, with the note that all these mutations cause increases in relative AKT kinase activity (33). This may lead to conditions such as Proteus syndrome with the E17K mutation (33). In addition, presence of the poS473 residue on AKT by immunohistochemistry (IHC) has been correlated with poor outcome in prostate cancer patients (34,35). This led to the notion that mutations that disrupt auto-inhibitory PH-kinase interactions can facilitate oncogenic progression by inducing the kinase activities of AKT isoforms (36). Many cancers with copy number losses or function-negating mutations in PTEN have increased AKT activation, as assessed by relative increases in poT308 and poS473 levels, or have gene amplifications of AKT isoform genes (37), which correlate with increased expression levels of AKT isoform proteins (38). Importantly, high AKT activation levels correlate with worse overall survival in hematological and solid cancers (39).

Figure 2: Roles for PI3K and AKT isoforms in specific parameters of oncogenesis.

Figure 2:

Specific and sometimes conflicting roles have been attributed to AKT1 or AKT2 in parameters of oncogenic growth, proliferation, survival motility or metabolism. Preferred upstream control of AKT1 or AKT2 is likely controlled PI3K containing p110α or p110β kinase domains, respectively. Though less clarified, several roles for AKT3 in oncogenic progression have been identified recently.

Even though all of the three AKT isoforms are considered to be oncogenic drivers, based on cases of AKT isoform gene amplification or kinase-activating mutation, recent data strongly suggest that they play overlapping and non-redundant roles depending on the specific oncogenic progression parameter studied. For example, in mouse transgenic models of Erbb2-driven mammary carcinoma, the expression of activated Akt1, in which the T308 and S473 residues are switched to phospho-mimetic Asp residues (Akt1T308D,S473D), increased primary tumor development but did not increase metastatic or invasive potential (31,40). In contrast, Akt2T309D,S474D had little effect on the latency of mammary tumor growth in transgenic mice induced by the expression of activated ErbB2 or polyomavirus middle T-ag, but it induced marked increases in pulmonary metastases and in tumor cell invasiveness (31,41). Further, in non-tumor biology, AKT1S473D, but not AKT2S474D, can induce lipogenesis in mature brown adipocytes (Martinez Calejman et al., 2020). These experiments are important in that they show that these phospho-mimetic mutations retain AKT1- or AKT2-specific signaling functions. In contrast, fusions of AKT isoforms to the N-terminal Src myristylation signal (“myr”- MGAG residues) induces constitutively-active AKT kinase activity presumably by bypassing the need for PH domain-mediated membrane association by AKT (42,43). Myr-AKT isoform variants can oncogenically transform embryo fibroblasts, and when expressed in transgenic mice via the mammary-specific MMTV promoter, increase susceptibility to carcinogen-induced mammary carcinomas (4345). In contrast, these same studies showed that overexpression of wt-AKT isoforms, whose intrinsic kinase activities were relatively much lower, were >100-fold poorer at transforming embryo fibroblasts. Lastly, the notion that forcing membrane association of AKT is sufficient to induce its oncogenic activity is underlined by the fact that the v-akt oncogene contains an N-terminally myristoylated GAG fusion that promotes membrane association (46,47). Taken together, these data strongly suggest that pro-oncogenic roles for AKT isoforms direct relate to both kinase activities and membrane association.

In contrast to the phospho-mimetic activated AKT isoform variants, however, there is no formal demonstration that the myr-AKT variants retain isoform-specific signaling, leading to the hypothesis that constitutive membrane association abrogates isoform-specific effects. Indeed, AKT isoforms may manifest their signaling specificities by acting in different cellular compartments. For example, Gonzalez and McGraw showed that although insulin induced equal activation of Akt1 and Akt2 in mouse adipocytes (based on relative poT308/9 levels), activation of the GLUT4 glucose transporter was facilitated by greater accumulation of Akt2 at the plasma membrane; expression of the Akt1-E17K variant, whose PH domain mutation facilitates constitutive plasma membrane association, overcame the Akt2 dependence for GLUT4 activation (48). Santi and Lee showed that in MDA-MB-231 human breast cancer cells, AKT1 was mainly found in the cytoplasm, AKT2 in the mitochondria and AKT3 in the nucleus and at the nuclear membrane, and that isoform-specific knockdown did not alter the localization of the other isoforms (49). They also showed whereas insulin induced the selectively accumulation of AKT2 to plasma membrane sites, swapping the AKT2-PH to AKT1 did not facilitate its movement to the plasma membrane. Indeed, swapping the AKT2-PH produced an AKT1 that was incapable of inducing cell proliferation and G1/S transition (50)Complicating this further are multiple, sometimes conflicting findings in breast cancer tissue samples that show enrichment of activated AKT1 and AKT2 in the nucleus (reviewed in (51)). These data suggest that AKT isoform-specific pathways could be facilitated by non-redundant interactions with signaling mediators enriched in particular cell compartments, and that some of these may be regulated by isoform-specific PH domains. An example of the latter is how the selective binding of TCL1b to AKT3 is dependent on its PH domain, a funciton that cannot be replacedby the AKT1 PH domain (52).

Although less is known about AKT3-specific roles in cancer progression, starting with the initial observation by Nakatani et al. in 1999 that AKT3 expression and phosphorylation are upregulated in ER+ breast cancers and androgen-independent prostate cancers, multiple studies have elucidated AKT3-specific oncogenic roles, as reviewed by Hinz and Jücker (2,51). These include mediating endocrine therapy resistance in ErbB2-driven mammary carcinomas and AKT inhibitor resistance in breast cancers, or suppressing chemotaxis and metastasis of triple-negative breast cancer cell lines (5355).

Non-redundant roles for AKT1 and AKT2 have also been shown in prostate cancer using genetically-engineered mouse models and tetracycline-regulated AKT isoform shRNA knockdowns: AKT1 promotes primary tumor growth, whereas AKT2 is responsible for development of distant metastasis and disease aggressiveness (56,57). More recent research has suggested that this isoform plasticity is dependent on other factors within the PI3K/AKT pathway. For example, PTEN-deficient tumors rely more on AKT2 than on AKT1 signaling for maintenance and survival under 3D growth conditions (56). A significant body of data indicate that the preferential activation of AKT isoforms can be influenced by which upstream PI3K isoforms are activated. For example, multiple studies show increased activation of PI3K-p110β in cancers with PTEN loss, corresponding to increased relative activation levels of AKT2 (56,58). In contrast, activation of Src-family kinases through the stimulation of specific growth factor receptor tyrosine kinases leads to the preferential activation of PI3K-p110α and the subsequent activation and dependence on AKT1 (59). Nonetheless, some plasticity exists within this axis: treatment of PTEN-null breast or prostate cancers with a PI3K-p110β inhibitor induces drug resistance by upregulating p110α and HER3/ERBB3 (5961). Thus, optimal cancer suppression requires therapeutically targeting multiple nodes upstream of AKT activation, such as multiple p110 isoforms and/or activated tyrosine kinases (61). Lastly, it is important to note the existence of AKT-independent PI3K cancer progression pathways (62,63), suggesting that AKT activation markers (e.g.- poT308 and poS473), though important, are insufficient to fully assess the role of activated PI3K in cancer progression.

AKT Isoform-Preferential Substrate Identification: Challenges and Successes

From a logical standpoint, it is reasonable to assume that non-overlapping or opposing roles for the AKT isoforms involve distinct sets of isoform-specific substrates, whereas the overlapping roles utilize shared substrates. As will be described below, limited attempts have been made to identify purely isoform-specific substrates, and even in these cases, it is unclear whether the isoform-specificity is only cell type- or context-specific. This has led to the likelihood that AKT isoform-dependent biologies utilize “preferential” substrates, yet to fully define these, cell-type and cancer-specific contexts will need to be considered. As an example, whereas AKT1 is pro-migratory and AKT2 anti-migratory in untransformed fibroblasts, opposite roles are manifest in breast cancer cell lines (64).

The quest to identify a consensus AKT substrate phosphorylation motif was heavily influenced by the initial substrates studied and subsequent experimental techniques that heavily focused on this canonical motif. The first AKT substrate validated was glycogen synthase kinase 3 beta (GSK3β), based on the ability of wortmannin- a PI3K inhibitor, to block insulin-induced GSK3β phosphorylation (65) and on the ability of immunoprecipitated PKB/AKT to phosphorylate GSK3β in vitro (66). The current list of >100 AKT substrates includes protein and lipid kinases, signaling proteins, transcription factors, ubiquitin ligases, metabolic enzymes and cell cycle regulators (66). The initial description of a possible consensus AKT substrate motif was developed by comparing the AKT/PKB phosphorylation sequence shared between GSK3β, MAPKAP kinase-1 (RPS6KA1/p90Rsk) and p70 S6Kinase (RPS6KB2), and then validated on a limited set of 7- to 10-mer peptides in in vitro kinase assays containing HA-tagged AKT1 immunopurified from HEK-293 cells, eventually defining the canonical substrate motif as RxRxxS/Tφ, with S/T representing the phosphorylated residues and φ representing hydrophobic amino acids (16,23,67). More recently, Balasuriya et al. (68) sought to assess how partial AKT1 phosphorylation (poS473 or poT308) affected substrate preference and maximal kinase activity when compared to fully activated AKT1 (containing both poS374 and poT308). They used three peptide libraries, one enriched for the AKT substrate motif found in 84 known substrates, plus two “oriented peptide array libraries” containing ~1011 peptides each, one of which (OPAL1) had Arg fixed at the −3 position, based on its known preference in canonical AKT substrate motif. They showed less dependence on Arg at the −5 position for AKT1poS473/T308, consistent with a Proline at the −5 position in the AKT phosphorylation sites on AMPK and ATP-citrate lyase (ACLY) (69). However, this study did not attempt to identify potentially non-canonical AKT1 substrate motifs using the more random OPAL2 peptide library. Indeed, one drawback to substrate site/motif identification using in vitro assays is that they lack the context of cellular compartmentalization, and thus, “substrates” that might never interact with AKT isoforms might be identified artefactually. In contrast, a problem for substrate identification in cells is that other kinases, such as RSK, PIM2 or protein kinase A (PKA), can phosphorylate versions of the AKT substrate motif (67,7072). PIM2 and AKT can phosphorylate overlapping sites on the anti-apoptotic protein, BAD, and on the cell cycle regulator, p21, and the AKT phosphorylation site on ACLY (S455) can be also phosphorylated by PKA, mTOR or BCKDK (7378). This points to the complication that substrates identified by phosphoproteomics analyses in cells cannot readily distinguish between direct and indirect substrates. Thus, even when using a canonical AKT substrate motif, confidence in the growing list of shared and preferential AKT isoform substrates requires combining data from in vitro kinase assays and from cellular proteomics data using either isoform-specific knockouts, knockdowns or overexpressions, as described below, or AKT inhibitory drugs (79)

The importance of alternative AKT substrate motifs increases with the discussion of how specific pathways or biologies are controlled by specific AKT isoforms (51). Thus, in addition to shared substrates that encode the RxRxxS/Tφ motif, phosphorylation of AKT isoform-preferred substrates is likely facilitated via either non-redundant, non-canonical motifs or substrate conformations. For example, Palladin, was identified because the phosphorylation motif surrounding S507 conformed to the canonical AKT motif (80). Newer data show that Palladin is a preferred substrate of AKT1 but not AKT2, whereas AKT2, but not AKT1, regulates its expression (32,81). Girardi et al. suggest that the ability of casein kinase 2 to phosphorylate AKT1, but not AKT2, helps drive the kinase specificity of AKT1 for palladin (82).

There have been a small number of phosphoproteome analyses to identify AKT isoform-preferential substrates. For example, (83) performed LC-MS/MS on phospho-serine or -threonine peptides from Akt1/2/3 knockout (TKO) lung fibroblasts (from transgenic mice) vs. TKO cells re-expressing Akt1, Akt2 and/or Akt3. Sixteen new substrates were identified, including IWS1, an RNA processing regulator, which appeared as an Akt1- and Akt3-preferred substrate. Although this analysis minimized the possibility of isoform compensation- a phenomenon described previously (9), it is restricted to basal AKT isoform activities in a single cell-type context (fibroblasts), and moreover, there was no analysis of non-canonical substrate motifs. Cenni et al. identified Ankrd2/ARPP as a novel AKT substrate based on combining proteomics analysis of peptides captured using beads coated with antibodies that recognize the canonical phosphorylated RxxS/T motif (84). Using AKT isoform-specific siRNAs as well as in vitro kinase assays with immunoprecipitated HA-tagged AKT isoforms, they showed that Ankrd2 is an AKT2-preferred substrate. Likewise, the insulin-induced phosphorylation of MYO5A could be abrogated by siRNA to AKT2 but not to AKT1 (85). Nonetheless, the vast majority of the 167 AKT substrates listed on the Cell Signaling site (https://www.cellsignal.com/contents/resources-reference-tables/pi3k-akt-substrates-table/science-tables-akt-substrate) are listed as AKT1-specific, with only seven listed as AKT2-preferred (CD34, EZRIN, GAPDH, MYO5A, QIK/SIK2, SFRS5 and STXBP4) and none as preferred by AKT3 alone. Indeed, the true AKT isoform specificity of this list needs reevaluation: many of the AKT1- or AKT2-specific substrate designations may be due to the fact that these were the only kinases tested, or because “total” AKT activity was attributed to AKT1, or because in a given cell context, only one isoform is predominantly expressed. An example of the latter is EZRIN, which was identified as an AKT2-specific substrate in Caco-2 cells, even though these cells expressed little AKT1 or AKT3 (86). This suggests that some previous identifications of isoforms-specific substrates might actually reflect cell type-preferential AKT isoform expression.

A continuing confounding issue is the contribution of non-AKT kinases (e.g., PKA or RSK) in the phosphorylation of a putative AKT isoform-specific substrate under specific biological conditions. For example, based on the assumed role for AKT2 in insulin-mediated inhibition of gluconeogenesis (87,88), Dentin et al. showed that purified AKT2 could phosphorylate SIK2/QIK in vitro at a canonical AKT substrate site, Ser358, that was shown to be induced by insulin (87,88). However, subsequent work showed that whereas insulin induced Akt2 activation in mouse liver cells, it could not induce Sik2poS358. In contrast, Sik2poS358 could be induced by PKA activated by glucagon or a cell-permeable cAMP analogue (Bt2-cAMP), even in Akt2−/− hepatocytes or Akt2+/+ hepatocytes treated with the pan-AKT inhibitor, MK-2206 (89). Thus, although it is listed as such, there remains no direct evidence that SIK2 is a bona fide AKT2 substrate in cells.

It is important to note at this point that while the focus is on canonical AKT phosphorylation as the mechanism for determining substrate preference, there are other factors that may contribute to this phenomenon. These factors, highlighted in the review by Hinz and Jücker, include post-translational modification, miRNA regulation, extracellular activation, and others (51). While these other factors may indeed contribute to some of the specificity, there is limited research into what roles these different cellular processes have on AKT isoform preferentiality.

Translating AKT Isoforms and Substrate Preferentiality to the Clinic

Given the growing evidence of AKT isoform-specific roles in cancer progression, the lack of clarity in reference to isoform-preferred substrates and pathways adds to the lack of understanding of how to therapeutically target the AKT axis. This is especially evident in PTEN-deficient cancers, which are assumed to be driven by increased AKT signaling. Consistent with our lack of full understanding of the oncogenic roles for PI3K and AKT, drugs targeting PI3K or AKT have performed poorly as monotherapies in solid tumors even though this axis is one of the most mutated in cancers (90,91). One pan-AKT inhibitor, Ipatasertib/RG7440, has been used in combination with endocrine and CDK4/6 targeted therapy in metastatic breast cancer (92). Indeed, this combination arose from evidence that resistance to chronic treatment of estrogen receptor-positive breast cancers with the CDK4/6 inhibitor, Palbociclib (Ibrance), caused a dependence on upregulated PI3K/AKT signaling as a driver of cyclin E/CDK2-mediated proliferation (93). In theory, targeting all three AKT isoforms with a pan-inhibitor such as Ipatasertib should override both isoform-specific dependence and compensatory isoform activity. However, even within the class of so-called pan-AKT inhibitors, there can be >10-fold selective inhibition of one AKT isoform over others (Table 1). As examples, using in vitro assays containing purified AKT1, AKT2 or AKT3 plus a commonly recognized GSKα 15-mer peptide substrate, Dumble et al. showed that GSK2142795 inhibits AKT2 8.6-fold or 4.7-fold more potently than AKT3 or AKT1, respectively; for GSK2110183, inhibition of AKT1 is 25-fold or 32.5-fold more potent than for AKT2 or AKT3, respectively (94). Moreover, even though cancer cell lines with either mutant PIK3CA or PTEN loss were relatively more sensitive to these two drugs than those expressing WT genes, the drugs levels to needed to establish 50% growth inhibition (GI50) in 2D cultures were typically >100-fold higher than those needed to inhibit AKT isoform kinase activities. Even in cancer lines where mutant PIK3CA or PTEN loss was the only identified oncogenic driver, GI50 values varied >50-fold. Thus, in addition to not understanding how this class of drugs is targeting AKT isoforms, there is a disconnect between an assumed dependency on oncogenic AKT signaling and drug potency. As an example of this disconnect, Li et al., show that whereas AKT1 knockdown was sufficient to inhibit the invasiveness of MCF-7 and MDA-MB-231 human breast cancer cells, treatment with the “pan-AKT” inhibitor MK2206 (not a clinical candidate inhibitor) induced increased experimental lung metastasis formation in vivo (95).

Table1:

Specificity of pan-AKT inhibitors for AKT isoforms

Drug IC50 (nM) reference
AKT1 AKT2 AKT3
uprosertib/GSK2142795 180 328 38 Dumble et al., 2014
afuresertib/GSK2110183 0.08 2 2.6 Dumble et al., 2014
MK2206 5 12 65 Hirai et al., 2010
capivasertib/AZD5363 3 7 7 Davies et al., 2012
Ipatasertib/GDC-0068 5 18 8 Lin et al., 2011

The only AKT1- or AKT2-specific inhibitors described to date are A-674563 and CCT128930, respectively, both of which are ATP-competitive inhibitors, yet neither of which is in clinical trials. No AKT3-specific small drug inhibitors have been described. A-674563 (IC50=11nM against AKT1) is also a potent PKA and CDK2 inhibitor (IC50’s of 16nM and 46nM, respectively), whereas CCT128930 (IC50=6nM against AKT2) requires 28-fold more drug (IC50=168nM) to inhibit PKA (96). However, no data exist comparing the selectivity of these two drugs against purified AKT isoforms. Moreover, the mechanism-of-action of A-674563 in tumor cells remains unclear in that treatment of A427 lung carcinoma cells decreased the phosphorylation of some substrates assumed to be shared by all AKT isoforms, such as FOXO1, yet paradoxically increased (PRAS40) or had no effect (GSK3β) on the phosphorylation of other common substrates (97). A recent study describes AKT1-, 2- or 3-specific nanobodies, however, these have not been tested for their ability to block AKT isoform-specific activation in cells (98).

The needed development of improved AKT isoform-specific inhibitors and the elucidation of isoform-preferred substrates will undoubtedly allow for the ability to dissect the molecular pathways controlled by each AKT isoform during normal development or cancer progression. These data will also identify new, potentially targetable cancer biomarkers involved in specific progression parameters. Lastly, they will also help explain how possible compensatory effects by other AKT isoforms, upstream PI3K-p110 isoforms and other oncogenic mediators such as receptor and non-receptor tyrosine kinases regulate response to drugs targeting of the PI3K-AKT axis (5961,99). Clarification of these important issues will help improve the therapeutic targeting of this critical signaling pathway in solid tumors.

ACKNOWLEDGMENTS

We thank Henry Withers for critical review of the manuscript, and Asoke Mal and Eugene Kandel for critical discussions on AKT. This work was supported by the Roswell Park Alliance Foundation and by the National Cancer Institute grants R21-CA235092 (IHG) and P30-CA016056 (Roswell Park Comprehensive Cancer Center Support Grant).

Footnotes

Conflict of Interest: The authors declare no potential conflicts of interest.

1

Per convention for protein modifications adopted by EURESCO conference on Functional Protein Modules (Seefeld, Austria, 6–11 October 2001) described in Aasland et al. (FEBS Lett., 2002, 513(1):141–144): “po” represents protein phosphorylation.

References

  • 1.Staal SP. Molecular cloning of the akt oncogene and its human homologues AKT1 and AKT2: Amplification of AKT1 in a primary human gastric adenocarcinoma. Proc Natl Acad Sci 1987;84:5034–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nakatani K, Sakaue H, Thompson DA, Weigel RJ, Roth RA. Identification of a human Akt3 (Protein kinase B γ) which contains the regulatory serine phosphorylation site. Biochem Biophys Res Commun 1999;257:906–10. [DOI] [PubMed] [Google Scholar]
  • 3.Franke TF, Yang S-I, Chan TO, Datta K, Kazlauskas A, Morrison DK, et al. 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] [PubMed] [Google Scholar]
  • 4.Franke TF, Kaplan DR, Cantley LC, Toker A. Direct regulation of the Akt proto-oncogene product by phosphatidylinositol-3,4-bisphosphate. Science 1997;275:665–8. [DOI] [PubMed] [Google Scholar]
  • 5.Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275:1943–7. [DOI] [PubMed] [Google Scholar]
  • 6.Steck PA, Pershouse MA, Jasser SA, Yung WKA, Lin H, Ligon AH, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 1997;15:356–62. [DOI] [PubMed] [Google Scholar]
  • 7.Chen WS, Xu PZ, Gottlob K, Chen ML, Sokol K, Shiyanova T, et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the akt1 gene. Genes Develop 2001;15:2203–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Garofalo RS, Orena SJ, Rafidi K, Torchia AJ, Stock JL, Hildebrandt AL, et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKBβ. J Clin Invest 2003;112:197–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dummler B, Tschopp O, Hynx D, Yang Z-Z, Dirnhofer S, Hemmings BA. Life with a Single Isoform of Akt: Mice Lacking Akt2 and Akt3 Are Viable but Display Impaired Glucose Homeostasis and Growth Deficiencies. Molec Cell Biol 2006;26:8042–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tschopp O, Yang ZZ, Brodbeck D, Dummler BA, Hemmings-Mieszczak M, Watanabe T, et al. Essential role of protein kinase Bγ (PKBγ/Akt3) in postnatal brain developmental but not in glucose homeostasis. Development 2005;132:2943–54. [DOI] [PubMed] [Google Scholar]
  • 11.Easton RM, Cho H, Roovers K, Shineman DW, Mizrahi M, Forman MS, et al. Role for Akt3/Protein Kinase Bγ in Attainment of Normal Brain Size. Molec Cell Biol 2005;25:1869–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.James SR, Downes CP, Gigg R, Grove SJA, Holmes AB, Alessi DR. Specific binding of the Akt-1 protein kinase to phosphatidylinositol 3,4,5-trisphosphate without subsequent activation. Biochem J 1996;315:709–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dannemann N, Hart JR, Ueno L, Vogt PK. Phosphatidylinositol 4,5-bisphosphate-specific AKT1 is oncogenic. Int J Cancer 2010;127:239–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PRJ, Reese CB, et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Bα. Curr Biol 1997;7:261–9. [DOI] [PubMed] [Google Scholar]
  • 15.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] [PubMed] [Google Scholar]
  • 16.Alessi DR, Caudwell FB, Andjelkovic M, Hemmings BA, Cohen P. Molecular basis for the substrate specificity of protein kinase B; comparison with MAPKAP kinase-1 and p70 S6 kinase. FEBS Lett 1996;399:333–8. [DOI] [PubMed] [Google Scholar]
  • 17.Bozulic L, Surucu B, Hynx D, Hemmings BA. PKBα/Akt1 Acts Downstream of DNA-PK in the DNA Double-Strand Break Response and Promotes Survival. Mol Cell 2008;30:203–13. [DOI] [PubMed] [Google Scholar]
  • 18.Kawakami Y, Nishimoto H, Kitaura J, Maeda-Yamamoto M, Kato RM, Littman DR, et al. Protein kinase C βII regulates Akt phosphorylation on Ser-473 in a cell type- and stimulus-specific fashion. J Biol Chem 2004;279:47720–5. [DOI] [PubMed] [Google Scholar]
  • 19.Dillon RL, White DE, Muller WJ. The phosphatidyl inositol 3-kinase signaling network: Implications for human breast cancer. Oncogene. 2007. page 1338–45. [DOI] [PubMed] [Google Scholar]
  • 20.Jia S, Liu Z, Zhang S, Liu P, Zhang L, Lee SH, et al. Essential roles of PI(3)K–p110β in cell growth, metabolism and tumorigenesis. Nature 2008;454:776–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hers I, Vincent EE, Tavaré JM. Akt signalling in health and disease. Cell Signal 2011;23:1515–27. [DOI] [PubMed] [Google Scholar]
  • 22.Park BK, Zeng X, Glazer RI. Akt1 induces extracellular matrix invasion and matrix metalloproteinase-2 activity in mouse mammary epithelial cells. Cancer Res 2001;61:7647–53. [PubMed] [Google Scholar]
  • 23.Manning BD, Cantley LC. AKT/PKB Signaling: Navigating Downstream. Cell 2007;129:1261–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hahn-Windgassen A, Nogueira V, Chen CC, Skeen JE, Sonenberg N, Hay N. Akt activates the mammalian target of rapamycin by regulating cellular ATP level and AMPK activity. Journal of Biological Chemistry. J Biol Chem 2005;280:32081–9. [DOI] [PubMed] [Google Scholar]
  • 25.Gottlob K, Majewski N, Kennedy S, Kandel E, Robey RB, Hay N. Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and mitochondrial hexokinase. Genes Develop 2001;15:1406–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sanchez-Gurmaches J, Tang Y, Jespersen NZ, Wallace M, Martinez Calejman C, Gujja S, et al. Brown Fat AKT2 Is a Cold-Induced Kinase that Stimulates ChREBP-Mediated De Novo Lipogenesis to Optimize Fuel Storage and Thermogenesis. Cell Metab 2018;27:195–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Calera MR, Martinez C, Liu H, El Jack AK, Birnbaum MJ, Pilch PF. Insulin increases the association of Akt-2 with Glut4-containing vesicles. Journal of Biological Chemistry. J Biol Chem 1998;273:7201–4. [DOI] [PubMed] [Google Scholar]
  • 28.Martinez Calejman C, Trefely S, Entwisle SW, Luciano A, Jung SM, Hsiao W, et al. mTORC2-AKT signaling to ATP-citrate lyase drives brown adipogenesis and de novo lipogenesis. Nat Commun 2020;11:575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Boxer RB, Stairs DB, Dugan KD, Notarfrancesco KL, Portocarrero CPP, Keister BA, et al. Isoform-specific requirement for Akt1 in the developmental regulation of cellular metabolism during lactation. Cell Metab 2006;4:475–90. [DOI] [PubMed] [Google Scholar]
  • 30.Buzzi F, Xu L, Zuellig RA, Boller SB, Spinas GA, Hynx D, et al. Differential Effects of Protein Kinase B/Akt Isoforms on Glucose Homeostasis and Islet Mass. Molec Cell Biol 2010;30:601–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Maroulakou IG, Oemler W, Naber SP, Tsichlis PN. Akt1 ablation inhibits, whereas Akt2 ablation accelerates, the development of mammary adenocarcinomas in mouse mammary tumor virus (MMTV)-ErbB2/Neu and MMTV-polyoma middle T transgenic mice. Cancer Res 2007;67:167–77. [DOI] [PubMed] [Google Scholar]
  • 32.Chin YR, Toker A. The Actin-Bundling Protein Palladin Is an Akt1-Specific Substrate that Regulates Breast Cancer Cell Migration. Mol Cell 2010;38:333–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, Robbins CM, et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 2007;448:439–44. [DOI] [PubMed] [Google Scholar]
  • 34.Kreisberg JI, Malik SN, Prihoda TJ, Bedolla RG, Troyer DA, Kreisberg S, et al. Phosphorylation of Akt (Ser473) is an excellent predictor of poor clinical outcome in prostate cancer. Cancer Res 2004;64:5232–6. [DOI] [PubMed] [Google Scholar]
  • 35.Sircar K, Yoshimoto M, Monzon FA, Koumakpayi IH, Katz RL, Khanna A, et al. PTEN genomic deletion is associated with p-Akt and AR signalling in poorer outcome, hormone refractory prostate cancer. J Pathol 2009;218:505–13. [DOI] [PubMed] [Google Scholar]
  • 36.Parikh C, Janakiraman V, Wu WI, Foo CK, Kljavin NM, Chaudhuri S, et al. Disruption of PH-kinase domain interactions leads to oncogenic activation of AKT in human cancers. Proc Natl Acad Sci 2012;109:19368–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Altomare DA, Testa JR. Perturbations of the AKT signaling pathway in human cancer. Oncogene 2005;24:7455–64. [DOI] [PubMed] [Google Scholar]
  • 38.Shao X, Lv N, Liao J, Long J, Xue R, Ai N, et al. Copy number variation is highly correlated with differential gene expression: A pan-cancer study. BMC Med Genet 2019;20:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yao Z, Gao G, Yang J, Long Y, Wang Z, Hu W, et al. Prognostic Role of the Activated p-AKT Molecule in Various Hematologic Malignancies and Solid Tumors: A Meta-Analysis. Front Oncol 2020;10:588200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hutchinson JN, Jin J, Cardiff RD, Woodgett JR, Muller WJ. Activation of Akt-1 (PKB-α) Can Accelerate ErbB-2-Mediated Mammary Tumorigenesis but Suppresses Tumor Invasion. Cancer Res 2004;64:3171–8. [DOI] [PubMed] [Google Scholar]
  • 41.Dillon RL, Marcotte R, Hennessy BT, Woodgett JR, Mills GB, Muller WJ. Akt1 and Akt2 play distinct roles in the initiation and metastatic phases of mammary tumor progression. Cancer Res 2009;69:5057–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kohn AD, Takeuchi F, Roth RA. Akt, a pleckstrin homology domain containing kinase, is activated primarily by phosphorylation. J Biol Chem 1996;271:21920–6. [DOI] [PubMed] [Google Scholar]
  • 43.Mende I, Malstrom S, Tsichlis PN, Vogt PK, Aoki M. Oncogenic transformation induced by membrane-targeted Akt2 and Akt3. Oncogene 2001;20:4419–23. [DOI] [PubMed] [Google Scholar]
  • 44.Liu X, Powlas J, Shi Y, Oleksijew AX, Shoemaker AR, De Jong R, et al. Rapamycin inhibits Akt-mediated oncogenic transformation and tumor growth. Anticancer Res; 2004;24:2697–704. [PubMed] [Google Scholar]
  • 45.Wu Y, Kim J, Elshimali Y, Sarkissyan M, Vadgama J V. Activation of Akt1 accelerates carcinogen-induced tumorigenesis in mammary gland of virgin and post-lactating transgenic mice. BMC Cancer 2014;14:266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ahmed NN, Franke TF, Bellacosa A, Datta K, Gonzalez-Portal ME, Taguchi T, et al. The proteins encoded by c-akt and v-akt differ in post-translational modification, subcellular localization and oncogenic potential. Oncogene 1993;8:1957–63. [PubMed] [Google Scholar]
  • 47.Bellacosa A, Franke TF, Gonzalez-Portal ME, Datta K, Taguchi T, Gardner J, et al. Structure, expression and chromosomal mapping of c-akt: Relationship to v-akt and its implications. Oncogene 1993;8:745–54. [PubMed] [Google Scholar]
  • 48.Gonzalez E, McGraw TE. Insulin-modulated Akt subcellular localization determines Akt isoform-specific signaling. Proc Natl Acad Sci U S A 2009;106:7004–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Santi SA, Lee H. The Akt isoforms are present at distinct subcellular locations. Am J Physiol Cell Physiol; 2010;298:C580–91. [DOI] [PubMed] [Google Scholar]
  • 50.Yun SJ, Tucker DF, Kim EK, Kim MS, Do KH, Ha JM, et al. Differential regulation of Akt/protein kinase B isoforms during cell cycle progression. FEBS Lett 2009;583:685–90. [DOI] [PubMed] [Google Scholar]
  • 51.Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Comm Signal 2019;17:154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Laine J, Künstle G, Obata T, Noguchi M. Differential regulation of Akt kinase isoforms by the members of the TCL1 oncogene family. J Biol Chem 2002;277:3743–51. [DOI] [PubMed] [Google Scholar]
  • 53.Grottke A, Ewald F, Lange T, Nörz D, Herzberger C, Bach J, et al. Downregulation of AKT3 Increases Migration and Metastasis in Triple Negative Breast Cancer Cells by Upregulating S100A4. Dittmar T, editor. PLOS ONE 2016;11:e0146370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Stottrup C, Tsang T, Chin YR. Upregulation of AKT3 confers resistance to the AKT Inhibitor MK2206 in breast cancer. Mol Cancer Ther 2016;15:1964–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Grabinski N, Möllmann K, Milde-Langosch K, Müller V, Schumacher U, Brandt B, et al. AKT3 regulates ErbB2, ErbB3 and estrogen receptor α expression and contributes to endocrine therapy resistance of ErbB2+ breast tumor cells from Balb-neuT mice. Cell Signal 2014;26:1021–9. [DOI] [PubMed] [Google Scholar]
  • 56.Chin YR, Yuan X, Balk SP, Toker A. PTEN-deficient tumors depend on AKT2 for maintenance and survival. Cancer Discov 2014;4:945–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chen ML, Xu PZ, Peng XD, Chen WS, Guzman G, Yang X, et al. The deficiency of Akt1 is sufficient to suppress tumor development in Pten+/− mice. Genes Dev 2006;20:1569–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Xie S, Ni J, McFaline-Figueroa JR, Wang Y, Bronson RT, Ligon KL, et al. Divergent Roles of PI3K Isoforms in PTEN-Deficient Glioblastomas. Cell Reports 2020;32:108196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell 2011;19:58–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bosch A, Li Z, Bergamaschi A, Ellis H, Toska E, Prat A, et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer. Science Transl Med 2015;7:283ra51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-Outmezguine V, et al. Feedback Suppression of PI3Kα Signaling in PTEN-Mutated Tumors Is Relieved by Selective Inhibition of PI3Kβ. Cancer Cell 2015;27:109–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Reina-Campos M, Linares JF, Duran A, Cordes T, L’Hermitte A, Badur MG, et al. Increased Serine and One-Carbon Pathway Metabolism by PKCλ/ι Deficiency Promotes Neuroendocrine Prostate Cancer. Cancer Cell 2019;35:385–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hsu AH, Lum MA, Shim K-S, Frederick PJ, Morrison CD, Chen B, et al. Crosstalk between PKCα and PI3K/AKT Signaling Is Tumor Suppressive in the Endometrium. Cell Reports 2018;24:655–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhou GL, Tucker DF, Sun SB, Bhatheja K, Birnbaum MJ, Field J. Opposing roles for Akt1 and Akt2 in Rac/Pak signaling and cell migration. J Biol Chem 2006;281:36443–53. [DOI] [PubMed] [Google Scholar]
  • 65.Cross DAE, 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] [PubMed] [Google Scholar]
  • 66.Manning BD, Toker A. Leading Edge Review AKT/PKB Signaling: Navigating the Network. Cell 2017;169:381–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hutti JE, Jarrell ET, Chang JD, Abbott DW, Storz P, Toker A, et al. A rapid method for determining protein kinase phosphorylation specificity. Nature Meth 2004;1:27–9. [DOI] [PubMed] [Google Scholar]
  • 68.Balasuriya N, Davey NE, Johnson JL, Liu H, Biggar KK, Cantley LC, et al. Phosphorylation-dependent substrate selectivity of protein kinase B (AKT1). J Biol Chem 2020;295:8120–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Berwick DC, Hers I, Heesom KJ, Kelly Moule S, Tavaré JM. The identification of ATP-citrate lyase as a protein kinase B (Akt) substrate in primary adipocytes. J Biol Chem 2002;277:33895–900. [DOI] [PubMed] [Google Scholar]
  • 70.Dumaz N, Marais R. Protein kinase A blocks Raf-1 activity by stimulating 14–3-3 binding and blocking Raf-1 interaction with Ras. J Biol Chem 2003;278:29819–23. [DOI] [PubMed] [Google Scholar]
  • 71.Zimmermann S, Moelling K. Phosphorylation and regulation of Raf by Akt (protein kinase B). Science 1999;286:1741–4. [DOI] [PubMed] [Google Scholar]
  • 72.Romeo Y, Zhang X, Roux PP. Regulation and function of the RSK family of protein kinases. Biochem J 2012;441:553–69. [DOI] [PubMed] [Google Scholar]
  • 73.Fox CJ, Hammerman PS, Cinalli RM, Master SR, Chodosh LA, Thompson CB. The serine/threonine kinase Pim-2 is a transcriptionally regulated apoptotic inhibitor. Genes Dev 2003;17:1841–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Potapova IA, El-Maghrabi MR, Doronin S V., Benjamin WB. Phosphorylation of recombinant human ATP:citrate lyase by cAMP-dependent protein kinase abolishes homotropic allosteric regulation of the enzyme by citrate and increases the enzyme activity. Allosteric activation of atp:citrate lyase by phosphorylated sugars. Biochem 2000;39:1169–79. [DOI] [PubMed] [Google Scholar]
  • 75.White PJ, McGarrah RW, Grimsrud PA, Tso SC, Yang WH, Haldeman JM, et al. The BCKDH Kinase and Phosphatase Integrate BCAA and Lipid Metabolism via Regulation of ATP-Citrate Lyase. Cell Metab 2018;27:1281–1293.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen Y, Qian J, He Q, Zhao H, Toral-Barza L, Shi C, et al. mTOR complex-2 stimulates acetyl-CoA and de novo lipogenesis through ATP citrate lyase in HER2/PIK3CA-hyperactive breast cancer. Oncotarget 2016;7:25224–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yan B, Zemskova M, Holder S, Chin V, Kraft A, Koskinen PJ, et al. The PIM-2 Kinase Phosphorylates BAD on Serine 112 and Reverses BAD-induced Cell Death. J Biol Chem 2003;278:45358–67. [DOI] [PubMed] [Google Scholar]
  • 78.Ju X, Katiyar S, Wang C, Liu M, Jiao X, Li S, et al. Akt1 governs breast cancer progression in vivo. Proc Natl Acad Sci 2007;104:7438–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wiechmann S, Ruprecht B, Siekmann T, Zheng R, Frejno M, Kunold E, et al. Chemical Phosphoproteomics Sheds New Light on the Targets and Modes of Action of AKT Inhibitors. ACS Chem Biol 2021;16:631–641. [DOI] [PubMed] [Google Scholar]
  • 80.Obata T, Yaffe MB, Leparc GG, Piro ET, Maegawa H, Kashiwagi A, et al. Peptide and protein library screening defines optimal substrate motifs for AKT/PKB. J Biol Chem 2000;275:36108–15. [DOI] [PubMed] [Google Scholar]
  • 81.Chin YR, Toker A. Akt2 regulates expression of the actin-bundling protein palladin. FEBS Lett 2010;584:4769–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Girardi C, James P, Zanin S, Pinna LA, Ruzzene M. Differential phosphorylation of Akt1 and Akt2 by protein kinase CK2 may account for isoform specific functions. Biochim Biophys Acta - Mol Cell Res 2014;1843:1865–74. [DOI] [PubMed] [Google Scholar]
  • 83.Sanidas I, Polytarchou C, Hatziapostolou M, Ezell SA, Kottakis F, Hu L, et al. Phosphoproteomics Screen Reveals Akt Isoform-Specific Signals Linking RNA Processing to Lung Cancer. Molec Cell 2014;53:577–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cenni V, Bavelloni A, Beretti F, Tagliavini F, Manzoli L, Lattanzi G, et al. Ankrd2/ARPP is a novel Akt2 specific substrate and regulates myogenic differentiation upon cellular exposure to H 2O 2. Molec Biol Cell 2011;22:2946–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yoshizaki T, Imamura T, Babendure JL, Lu J-C, Sonoda N, Olefsky JM. Myosin 5a Is an Insulin-Stimulated Akt2 (Protein Kinase Bβ) Substrate Modulating GLUT4 Vesicle Translocation. Molec Cell Biol 2007;27:5172–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shiue H, Musch MW, Wang Y, Chang EB, Turner JR. Akt2 phosphorylates ezrin to trigger NHE3 translocation and activation. J Biol Chem 2005;280:1688–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dentin R, Liu Y, Koo SH, Hedrick S, Vargas T, Heredia J, et al. Insulin modulates gluconeogenesis by inhibition of the coactivator TORC2. Nature 2007;449:366–9. [DOI] [PubMed] [Google Scholar]
  • 88.Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB, et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKBβ). Science 2001;292:1728–31. [DOI] [PubMed] [Google Scholar]
  • 89.Patel K, Foretz M, Marion A, Campbell DG, Gourlay R, Boudaba N, et al. The LKB1-salt-inducible kinase pathway functions as a key gluconeogenic suppressor in the liver. Nature Commun 2014;5:4535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hanker AB, Kaklamani V, Arteaga CL. Challenges for the Clinical Development of PI3K Inhibitors: strategies to Improve Their Impact in solid Tumors. 2019; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Rodon J, Dienstmann R, Serra V, Tabernero J. Development of PI3K inhibitors: lessons learned from early clinical trials. Nat Rev Clini Oncol 2013;10:143–53. [DOI] [PubMed] [Google Scholar]
  • 92.Vicier C, Isambert N, Dalenc F, Campone M, Levy C, Rezai K, et al. TAKTIC: A prospective, multicenter, uncontrolled, phase IB/II study of LY2780301 (LY) in combination with weekly paclitaxel (wP) in HER2-negative locally advanced (LA) or metastatic breast cancer (MBC) patients. J Clin Oncol 2019;37:1091–1091. [Google Scholar]
  • 93.Herrera-Abreu MT, Palafox M, Asghar U, Rivas MA, Cutts RJ, Garcia-Murillas I, et al. Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer. Cancer Res 2016;76:2301–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dumble M, Crouthamel MC, Zhang SY, Schaber M, Levy D, Robell K, et al. Discovery of novel AKT inhibitors with enhanced anti-tumor effects in combination with the MEK inhibitor. PLoS ONE 2014;9:e100880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Li W, Hou JZ, Niu J, Xi ZQ, Ma C, Sun H, et al. Akt1 inhibition promotes breast cancer metastasis through EGFR-mediated β-catenin nuclear accumulation. Cell Communication and Signaling. BioMed Central Ltd; 2018;16:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Luo Y, Shoemaker AR, Liu X, Woods KW, Thomas SA, de Jong R, et al. Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo. Mol Cancer Ther 2005;4:977–86. [DOI] [PubMed] [Google Scholar]
  • 97.Chorner PM, Moorehead RA. A-674563, a putative AKT1 inhibitor that also suppresses CDK2 activity, inhibits human NSCLC cell growth more effectively than the pan-AKT inhibitor, MK-2206. PLoS ONE 2018;13:e0193344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Merckaert T, Zwaenepoel O, Gevaert K, Gettemans J. Development and characterization of protein kinase B/AKT isoform-specific nanobodies. Ramos JW, editor. PLOS ONE 2020;15:e0240554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, et al. Reciprocal Feedback Regulation of PI3K and Androgen Receptor Signaling in PTEN-Deficient Prostate Cancer. Cancer Cell 2011;19:575–86. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES