throbber
Oncogene (2005) 24, 7482–7492
`& 2005 Nature Publishing Group All rights reserved 0950-9232/05 $30.00
`
`www.nature.com/onc
`
`The Akt/PKB pathway: molecular target for cancer drug discovery
`
`Jin Q Cheng*,1, Craig W Lindsley2, George Z Cheng3, Hua Yang1 and Santo V Nicosia1
`
`1Departments of Pathology and Interdisciplinary Oncology, H Lee Moffitt Cancer Center and Research Institute, University of South
`Florida College of Medicine, 12902 Magnolia Drive, SRB3, Tampa, FL 33612, USA; 2Department of Medicinal Chemistry, Merck &
`Co., West Point, PA 19486, USA; 3Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA
`
`The serine/threonine kinase Akt/PKB pathway presents
`an exciting new target for molecular therapeutics, as it
`functions as a cardinal nodal point
`for transducing
`extracellular (growth factor and insulin) and intracellular
`(receptor
`tyrosine kinases, Ras and Src) oncogenic
`signals. In addition, alterations of the Akt pathway have
`been detected in a number of human malignancies. Ectopic
`expression of Akt, especially constitutively activated Akt,
`is sufficient to induce oncogenic transformation of cells
`and tumor formation in transgenic mice as well as
`chemoresistance. Akt has a wide range of downstream
`targets that regulate tumor-associated cell processes such
`as cell growth, cell cycle progression, survival, migration,
`epithelial–mesenchymal
`transition
`and
`angiogenesis.
`Blockage of Akt signaling results in apoptosis and growth
`inhibition of tumor cells with elevated Akt. The observed
`dependence of certain tumors on Akt signaling for survival
`and growth has wide implications for cancer therapy,
`offering the potential for preferential tumor cell killing. In
`the last several years, through combinatorial chemistry,
`high-throughput and virtual screening, and traditional
`medicinal chemistry, a number of inhibitors of the Akt
`pathway have been identified. This review focuses on
`ongoing translational efforts to therapeutically target the
`Akt pathway.
`Oncogene (2005) 24, 7482–7492. doi:10.1038/sj.onc.1209088
`
`Keywords: Akt; cancer; therapeutics; inhibitor
`
`Akt proteins: overview and rationale as antitumor targets
`
`Akt was originally discovered as an oncogene trans-
`duced by the acute transforming retrovirus (Akt-8),
`which was isolated from an AKR thymoma (Staal et al.,
`1977; Staal, 1987), and subsequently found to encode a
`serine/threonine protein kinase (Bellacosa et al., 1991).
`Akt is also known as protein kinase B (Coffer and
`Woodgett, 1991) and RAC-PK (Jones et al., 1991). Viral
`akt highly activated and oncogenic due to the fact that
`v-akt is associated with the cell membrane through a
`myristylated Gag protein sequence fused to the N-
`terminus of Akt (Bellacosa et al., 1991). The important
`
`*Correspondence: JQ Cheng; E-mail: chengjq@moffitt.usf.edu
`
`role of Akt in transformation and cancer was shortly
`thereafter strengthened by the cloning of the AKT2 gene
`(Cheng et al., 1992) and the discovery that AKT2 is
`frequently amplified and overexpressed in human
`cancers (Cheng et al., 1992, 1996; Bellacosa et al.,
`1995). To date, three Akt family members have been
`identified in mammals, designated Akt1/PKBa, Akt2/
`PKBb and Akt3/PKBg (Testa and Bellacosa, 2001). The
`members of Akt family share similar domain structure
`and are activated by various stimuli in a phosphatidy-
`linositol 3-kinase (PI3K)-dependent manner (Burgering
`and Coffer, 1995; Franke et al., 1995; Liu et al., 1998;
`Shaw et al., 1998). Activation of Akt depends on the
`integrity of
`the pleckstrin homology (PH) domain,
`which mediates its membrane translocation, and on
`the phosphorylation of Thr308 in the activation loop and
`Ser473 (Chan et al., 1999; Datta et al., 1999; Testa and
`Bellacosa, 2001; Brazil et al., 2002). Phosphoinositides,
`PtdIns-3,4-P2 and PtdIns-3,4,5-P3, produced by PI3 K
`bind directly to the PH domain of Akt, driving a
`conformational change in the molecule, which enables
`the activation loop of Akt to be phosphorylated by
`PDK1 at Thr308 (Alessi et al., 1997). Full activation of
`AKT1 is also associated with phosphorylation of Ser473
`(Alessi et al., 1996) within a C-terminal hydrophobic
`motif characteristic of kinases in the AGC kinase family.
`Although the role of PDK1 in Thr308 phosphorylation is
`well established, the mechanism of Ser473 phosphoryla-
`tion is controversial. A number of candidate enzymes
`responsible for this modification have been put forward,
`including integrin-linked kinase (Persad et al., 2001),
`PDK1 when in a complex with the kinase PRK2
`(Balendran et al., 1999), Akt itself, through autopho-
`sphorylation (Toker and Newton, 2000), PKCa (Parto-
`vian and Simons, 2004), PKCbII (Kawakami et al.,
`2004), DNA-dependent kinase (Feng et al., 2004), and
`the rictor-mTOR complex (Sarbassov et al., 2005). The
`activity of Akt
`is negatively regulated by tumor
`suppressor PTEN, which is frequently mutated in
`human malignancy (Li et al., 1997; Steck et al., 1997;
`Parsons, 2004). PTEN encodes a dual-specificity protein
`and lipid phosphatase that reduces intracellular levels of
`PtdIns-3,4,5-P3 by converting them to PtdIns-4,5-P2,
`thereby inhibiting the PI3K/Akt pathway (Stambolic
`et al., 1998). Akt phosphorylates and/or interacts with a
`number of molecules
`to exert
`its normal cellular
`functions, which include roles in cell proliferation,
`survival and differentiation (Chan et al., 1999; Datta
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 1 of 11
`
`

`

`7483
`
`et al., 1999; Testa and Bellacosa, 2001; Brazil et al.,
`2002). Gene knockout studies have defined the biologi-
`cal
`importance of Akt members in normal cells. In
`particular, Akt2-null mice develop typical
`type II
`diabetes (Cho et al., 2001a, b), while Akt1- and Akt3-
`deficient mice do not display a diabetic phenotype but
`exhibit a decrease in the sizes of all organs and a
`selective impairment of brain development, respectively
`(Chen et al., 2001; Cho et al., 2001a, b; Easton et al.,
`2005; Tschopp et al., 2005). Moreover, although Akt1-
`and Akt3-deficient brains are reduced in size to
`approximately the same degree, the absence of Akt1
`reduces neuronal cell number, whereas the lack of Akt3
`results in smaller and fewer cells in which mTOR
`signaling is attenuated (Easton et al., 2005).
`Several lines of evidence suggest that Akt is a critical
`target for anticancer drug discovery. First, Akt sits at
`the crossroads of multiple oncogenic and tumor
`suppressor signaling networks (see related Reviews in
`this issue). Almost all known oncogenic growth factors,
`angiogenic factors and cytokines activate Akt by
`binding to cognate receptors on cell surface. Further,
`Akt is also activated by steroid hormones, such as
`estrogen and androgen through a mechanism indepen-
`dent of their nuclear receptors (Sun et al., 2001a; Sun
`et al., 2003). In addition, Akt is shown to be activated by
`constitutively active Ras and Src (Datta et al., 1996; Liu
`et al., 1998). Second, frequent deregulations of many
`components of the Akt signaling pathway have been
`observed in human cancer (see review by Altomare and
`Testa in this issue). Of the Akt family, only the AKT2
`gene is frequently amplified in human cancer. Further,
`overexpression of AKT2 RNA and/or protein is also
`more commonly observed in human cancer than are
`AKT1 and AKT3 (Testa and Bellacosa, 2001; Kim
`et al., 2005). However, recurrent activation of the three
`Akt family members has been detected in a variety of
`types of human malignancy (Yuan et al., 2000; Sun
`et al., 2001a, b, 2003; Altomare et al., 2003, 2004, 2005;
`Balsara et al., 2004). Activation of Akt is primarily the
`result of aberrant upstream molecules of Akt, which
`include overproduction of growth factors, upregulation
`and/or mutation of receptor tyrosine kinases, Ras and
`Src as well as PIK3CA and PTEN. While a point
`mutation of AKT2 has been reported in familial diabetes
`(George et al., 2004), a dominant mutation of Akt has
`not been identified in human tumor. Third, ectopic
`expression of constitutively active Akt and even wild-
`type Akt2 results in oncogenic transformation in vitro
`and in vivo (Cheng et al., 1997; Hutchinson et al., 2001;
`Malstrom et al., 2001; Mende et al., 2001; Sun et al.,
`2001a, b; Majumder et al., 2003). Furthermore, a
`number of studies have shown that overexpression
`and/or activation of Akt render tumor cells resistant
`to chemotherapeutic drugs and signal molecule inhibi-
`tors such as Gleevec, Iressa, Herceptin and retinoid acid
`(Cheng et al., 2002; Arlt et al., 2003; Knuefermann et al.,
`2003; Yuan et al., 2003; Nagata et al., 2004). In
`addition, Akt targets many signal molecules to regulate
`tumor development-associated cell processes such as
`apoptosis, cell proliferation, differentiation, migration
`
`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`and angiogenesis. Finally, knockdown of Akt by
`antisense or siRNA significantly reduces tumor growth
`and invasiveness and induces apoptosis and cell growth
`arrest only in tumor cells overexperssing Akt (Cheng
`et al., 1996; Chen et al., 2001; Asnaghi et al., 2004; Remy
`et al., 2004; Tabellini et al., 2005).
`These observations make Akt an attractive target for
`anticancer drug discovery, and it has been postulated
`that inhibition of Akt alone or in combination with
`standard cancer chemotherapeutics will reduce the
`apoptotic threshold and preferentially kill cancer cells.
`The development of specific and potent inhibitors will
`allow this hypothesis to be tested in animals. The
`majority of small molecule inhibitors in this nascent field
`are classic ATP-competitive inhibitors, which provide
`little specificity. Phosphatidylinositol (PI) analogs have
`been reported to inhibit Akt, but these inhibitors may
`also have specificity problems with respect to other PH
`domain containing proteins and may have poor
`bioavailability. Recently, small chemical compounds
`triciribine/Akt/protein kinase B inhibitor-2 (API-2) and
`allosteric inhibitors have been reported which are PH
`domain dependent, and the latter also exhibit Akt
`isozyme selectivity.
`In addition,
`inhibitors
`toward
`upstream regulators and downstream targets of Akt
`have also been tested for their capability of reversing the
`phenotype of cancer cells expressing altered Akt. This
`review focuses on the ongoing efforts to therapeutically
`target individual components of Akt pathway including
`Akt
`itself as well as its upstream regulators and
`downstream effectors (Figure 1). Some of these efforts
`involve AKT-specific inhibition based on structure-
`based analyses (see also the Review by Kumar and
`Madison in this issue).
`
`Therapeutic targeting of upstream regulators of Akt
`
`PDK1 inhibitors
`
`PDK1 is a serine/threonine protein kinase that can
`phosphorylate and activate a number of kinases in the
`AGC kinase superfamily (Mora et al., 2004). The first
`identified and best characterized PDK1 substrates are
`the three members of the Akt family (Mitsiades et al.,
`2004). PDK1 phosphorylates the activation loop of Akt
`(also called the T-loop) on residue Thr308, which
`primarily regulates Akt activation (Alessi et al., 1997).
`Therefore, a PDK1 inhibitor should significantly block
`activation of Akt.
`Three potent PDK1 inhibitors, BX-795, BX-912 and
`BX-320 (Figure 2a), recently identified by screening of
`compound libraries, have IC50 between 11 and 30 nM
`(Feldman et al., 2005). The inhibitors blocked PDK1/
`Akt signaling in tumor cells resulting in the inhibition of
`anchorage-independent growth and the induction of
`apoptosis in a variety of tumor cell lines. A number of
`cancer cell lines with elevated Akt activity were >30-
`fold more sensitive to growth inhibition by PDK1
`inhibitors in soft agar than on tissue culture plastic,
`consistent with the cell survival function of the PDK1/
`
`Oncogene
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 2 of 11
`
`

`

`7484
`
`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`PIP3
`competitor
`
`Akt
`inhibitor
`
`PI-3,4,
`
`5-P3
`
`PH
`
`Kinase
`domain
`
`Akt
`-T308-p
`
`RD
`
`-S473-p
`
`PTEN
`
`PI 3-kinase
`
`p110
`
`Src
`
`p85
`
`PI-4,
`5-P2
`
`TRK
`inhibitor
`
`RAS
`
`PDK1
`
`FTI/GGTI
`
`PI3K
`inhibitor
`
`PDK1
`inhibitor
`
`mTOR
`
`mTOR
`inhibitor
`
`p70S6K
`
`4E-BP1
`
`5’top mRNA translation
`
`Cap dependent translation
`
`Figure 1 Therapeutic targeting of the Akt pathway including Akt itself as well as its upstream regulators and downstream effectors
`
`Figure 2 Compound structures for inhibitors of PDK1 (a) and PI3K (b)
`
`Akt signaling pathway, which is particularly important
`for unattached cells. BX-320 inhibited the growth of
`LOX melanoma tumors in the lungs of nude mice after
`injection of tumor cells into the tail vein. The effect of
`BX-320 on cancer cell growth in vitro and in vivo
`indicates that PDK1 inhibitors may have clinical utility
`as anticancer agents.
`The staurosporine derivative UCN-01 (7-hydroxys-
`taurosporine), a drug now in clinical trials, has been
`shown to potently inhibit PDK1 (IC50¼ 33 nM) in vitro.
`UCN-01-induced PDK1 inhibition was also observed in
`
`human tumor xenografts (Sato et al., 2002). Over-
`expression of constitutively active Akt diminished the
`cytotoxic effects of UCN-01, suggesting that UCN-01
`may in part exert its cytotoxicity by inhibiting PDK1/
`Akt
`survival pathway. Crystal
`structure analyses
`showed that staurosporine and UCN-01 form a complex
`with the kinase domain of PDK1 (Komander et al.,
`2003). Although staurosporine and UCN-01 interact
`with the PDK1 active site in an overall similar manner,
`the UCN-01 7-hydroxy group (Figure 2a), which is not
`present in staurosporine, generates direct and water-
`
`Oncogene
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 3 of 11
`
`

`

`7485
`
`mediated hydrogen bonds with active-site residues. This
`moiety is hydrogen-bonded directly to Thr222 and
`indirectly via an ordered water molecule to Gln220 of
`PDK1 (Zhao et al., 2002; Johnson and Pinto, 2002;
`Komander et al., 2003). A different water-mediated
`hydrogen-bonding network is also observed in other
`UCN-01 complexes and might serve as a starting point
`for further structure-based optimization. In addition,
`recent studies show that UCN-01 inhibits other kinases
`such as PKC and Chk1 and transcriptionally upregu-
`lates the cyclin-dependent kinase inhibitor p21waf1/cip1
`(Senderowicz, 2003a, b).
`
`PI3K inhibitors
`
`Dissection of PI3K/Akt signaling pathway has been
`aided greatly by two pharmacological PI3K inhibitors,
`wortmannin and LY294002 (Figure 2b). Wortmannin is
`a fungal metabolite and a potent inhibitor of type I
`PI3K, with an IC50 range for inhibition of PI3K from 2
`to 4 nM (Arcaro and Wymann, 1993; Vlahos et al.,
`1994). Wortmannin inhibits PI3K activity by binding
`covalently to a conserved lysine residues in the ATP-
`binding site of the enzyme (Wymann et al., 1996).
`Wortmannin has antitumor activity in vitro and in vivo,
`suggesting that it might offer a valuable approach to
`treat cancer. However, a major disadvantage of the use
`of wortmannin is its stability in an aqueous environ-
`ment. Wortmannin is soluble in organic solvents, which
`may limit its use in clinical trials. Currently, water-
`soluble wortmannin conjugates are being developed to
`circumvent this issue. LY294002 is a flavonoid deriva-
`tive and a reversible, ATP-competitive inhibitor with
`IC50 for recombinant PI3K in the low micromolar
`range. A number of in vitro studies have shown that
`LY294002 alone has antiproliferative and proapoptotic
`activities (Wetzker and Rommel, 2004). Relatively, few
`in vivo studies have been conducted to demonstrate the
`efficacy of LY294002 on the inhibition of tumor growth,
`but
`these
`studies
`showed that administration of
`LY294002 in human cancer xenografts inhibited tumor
`growth and induced apoptosis (Semba et al., 2002; Fan
`et al., 2003). Although inhibition of the PI3K/Akt
`pathway by wortmannin or LY294002 alone may inhibit
`cell proliferation, promote apoptosis and/or inhibit
`tumor growth,
`the combination of wortmannin or
`LY294002 with traditional cytotoxic drugs or radiation
`enhances the effectiveness of these treatments (Wetzker
`and Rommel, 2004).
`It
`is noteworthy that neither wortmannin nor
`LY294002 displays selectivity for different members of
`the class I PI3K (Finan and Thomas, 2004). Wortman-
`nin also inhibits class III PI3K to reduce autophagy, a
`class II programmed cell death (Shintani and Klionsky,
`2004). LY294002 also inhibits casein kinase 2 with
`similar potency to PI3K. At higher concentrations,
`wortmannin inhibits PI3K-related enzymes, such as
`mTOR, ATM and PI4-kinase b (Finan and Thomas,
`2004). Moreover, methylxanthines, such as caffeine and
`theophylline, inhibit p110d, even though their activity is
`rather weak (Arcaro and Wymann, 1993; Abraham, 2004).
`
`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`Recently, several new compounds have been de-
`scribed to have some selectivity for individual members
`of PI3K (Figure 2b). PIramed have described several
`imidazopyridine derivatives that exhibit excellent PI3K
`inhibitory activities, especially against p110a. A series of
`morpholino-substituted compounds related LY294002
`have shown isoform selectivity. Quinolone and pyrido-
`pyrimidine (Kinacia) are approximately 100-fold more
`potent against p110a/b as compared to p110g (Finan
`and Thomas, 2004). ICOS Corporation has recently
`claimed a new PI3K inhibitor, IC87114, which selec-
`tively inhibits p110d with IC50¼ 0.5 mM and >50-fold
`selectivity over the other class PI3K isoforms (Sadhu
`et al., 2003). In addition, Novartis has described 5-
`phenylthiazole derivatives as PI3K inhibitors (Finan
`and Thomas, 2004). However, antitumor activity of
`these compounds needs further investigation both in
`vitro and in vivo. It is noteworthy that use of PI3K
`inhibitors may be associated with undesirable side
`effects because of the many important cellular targets
`of this lipid kinase.
`
`Inhibitors of the prenylation
`
`Protein prenylation, including farnesylation and geranyl-
`geranylation,
`is a lipid post-translational modification
`required for the cancer-causing activity of proteins such as
`the GTPase Ras. Farnesyltransferase and geranylgeranyl-
`transferase I inhibitors (FTIs/GGTIs) represent a new
`class of anticancer drugs that exhibit a remarkable ability
`to inhibit malignant transformation without significant
`toxicity to normal cells, especially FTIs are currently in
`clinical trials. However, the mechanism of FTI and GGTI
`antitumor activity remains elusive. It has been shown that
`FTIs inhibit PI3K/Akt-mediated growth factor- and
`adhesion-dependent survival pathways and induce apop-
`tosis in human cancer cells that overexpress Akt (Jiang
`et al., 2000; Prendergast, 2000; Sebti and Der, 2003).
`Furthermore, overexpression of Akt, but not oncogenic H-
`Ras, sensitizes NIH3T3 cells to FTI-277, and a high serum
`level prevents FTI-277-induced apoptosis in H-Ras- but
`not Akt-transformed NIH3T3 cells. A constitutively active
`form of Akt rescues human cancer cells from FTI-277-
`induced apoptosis. Integrin-dependent activation of Akt is
`also blocked by FTI-277. In addition, GGTI-298 and
`GGTI-2166 have also been shown to inhibit PI3K/Akt
`pathway, resulting in apoptosis in human cancer cells
`(Dan et al., 2004). Thus, a mechanism for FTIs and
`GGTIs inhibition of human tumor growth is by inducing
`apoptosis through inhibition of the PI3K/Akt pathway.
`However, neither FTIs nor GGTIs directly inhibits PI3K/
`Akt, suggesting that the unidentified prenylated proteins
`that activate PI3K/Akt are the targets of FTIs and/or
`GGTIs. Recent studies suggest that RhoB could be a
`candidate to mediate this action (Liu and Prendergast,
`2000; Adini et al., 2003; Jiang et al., 2004).
`
`RTK inhibitors
`
`Among RTKs, EGFR and Her2/Neu/ErbB2 are fre-
`quently altered in human cancer and primarily activate
`
`Oncogene
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 4 of 11
`
`

`

`7486
`
`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`the PI3K/Akt pathway. Two major approaches have
`been used to target the ErbB family, that is, small-
`molecule tyrosine kinase inhibitors and humanized
`antibodies against the receptor extracellular domains
`(Yu and Hung, 2000; Chen et al., 2003). In general,
`antibodies bind to the extracellular domain of
`the
`receptors,
`inhibiting their activation by ligand, and
`promoting receptor internalization and downregulation,
`whereas small molecules competitively inhibit ATP
`binding to the receptor, thereby hindering autopho-
`sphorylation and kinase activation. At present, the most
`advanced of the newer therapies in clinical development
`are anti-EGFR monoclonal antibody IMC-C225 (ce-
`tuximab, Erbitux; Imclone), anti-ErbB2 and the rever-
`sible small-molecule inhibitors of EGFR, ZD-1839
`(gefitinib, Iressa; AstraZeneca) and OSI-774 (erlotinib,
`Tarceva; OSI Pharmaceuticals). Both ZD-1839 and
`OSI-774 have been through phase I and phase II trials.
`Promising single-agent clinical antitumor activity has
`been reported in advanced NSCLC, head and neck
`cancer and prostate carcinoma. Furthermore, huma-
`nized monoclonal antibodies, IMC-C225 against EGFR
`and trastuzumab (Herceptin) targeted ErbB2, are also in
`phase II and phase III trials. These agents potently
`inhibit EGFR and ErbB2 resulting in the reduction of
`Akt kinase activity (Yakes et al., 2002; Mitsiades et al.,
`2004). A recent study has shown that patients who
`become resistance to Herceptin have elevated levels of
`Akt due to loss of PTEN, suggesting that elevated Akt
`activation is responsible for Herceptin resistance (Na-
`gata et al., 2004).
`
`Akt inhibitors
`
`Akt antibody
`
`Antibodies and antibody-based reagents have been used
`for the treatment of cancer. As described above, the
`humanized IgG1 trastuzamab (Herceptin) is an effective
`treatment for breast cancers that overexpress ErbB2 (Yu
`and Hung, 2000). Genetic engineering of antibodies can
`be used to modify and enhance antibody efficacy. For
`example, mouse monoclonal antibodies can be chimer-
`ized by such approaches to prevent the production of
`human antimurine antibodies when administered to
`immune-competent patients (Clark, 2000). An alterna-
`tive strategy is to replace the antibody gene present in
`mouse B cells with human antibody genes. These
`modified B cells can then be used to produce hybridoma
`cell
`lines that express fully humanized monoclonal
`antibodies that avoid cross-species immune response
`(Fishwild et al., 1996). Over a decade ago, McCafferty
`et al. (1990) demonstrated that recombinant antibody
`fragments could be displayed on the tip of M13
`bacteriophage. Some of
`the advantages of phage-
`displayed recombinant antibodies over the conventional
`polyclonal or monoclonal antibodies are quick genera-
`tion time, cheap production cost, and,
`importantly,
`accessibility to the antibody DNA for further genetic
`manipulations. Recently, Shin et al. (2005) developed
`
`novel recombinant anti-Akt single-chain antibodies by
`panning a mouse phage-displayed scFv recombinant
`antibody library using GST-Akt1 fusion protein. To
`generate a membrane-permeable version of the anti-
`Akt1-scFv, the scFv gene was subcloned into a GST
`expression vector carrying a membrane-translocating
`sequence (MTS) from Kaposi fibroblast growth factor.
`A purified GST-anti-Akt1-MTS fusion protein accumu-
`lates intracellularly and inhibits activation of all three
`Akt family members. Interestingly, in vitro kinase assay
`shows that GST-anti-Akt1-MTS also inhibits constitu-
`tively active forms of Myr-Akt1, -Akt2 and -Akt3 as
`well as phosphomimetic mutant of Akt-DD, where
`Thr308 and Ser473 are replaced with aspartic acid.
`Furthermore, GST-anti-Akt1-MTS induces apoptosis in
`cancer cell lines that express constitutively active Akt. In
`addition, anti-Akt scFv exhibits antitumor activity in
`PyVmT-expressing transgenic tumors
`implanted in
`mouse dorsal window chambers (Shin et al., 2005).
`These data indicate that GST-anti-Akt1-MTS is a cell-
`permeable inhibitor of Akt and that this approach can
`be used to generate compounds that target tumor cells
`dependent on aberrant Akt for their growth.
`
`PI analog inhibitors
`
`As PtdIns(3,4,5)P3 directly binds to the PH domain of
`Akt and PDK1 and is required for activation of Akt, the
`development of a PtdIns(3,4,5)P3 analog would be a
`reasonable approach to develop an Akt inhibitor. This
`mode of inhibition would prevent Akt translocation to
`the plasma membrane and activation. The feasibility of
`this approach was suggested by the demonstration that
`D-3-deoxy-myo-inositols inhibited the growth of trans-
`formed cells (Powis et al., 1991). It was subsequently
`found that the inositol derivative DPI had an IC50 of
`35 mM against H-29 colon cancer cell growth (Kozi-
`kowski et al., 1995). A recent study examined 24
`modified phosphatidylinositol ether
`lipid analogues
`(PIAs) and found that five of them, PIA5, 6, 23, 24,
`and 25 (Figure 3), with modifications at two sites on the
`inositol ring, inhibited Akt with IC50o5 mM (Castillo
`et al., 2004a, b). PIAs decreased phosphorylation of
`many downstream targets of Akt without affecting
`upstream kinases, such as PI3K or PDK1. Importantly,
`PIAs selectively induced apoptosis in cancer cell lines
`with high levels of endogenous Akt activity. These
`findings identify PIAs as effective Akt inhibitors, and
`provide proof of principle for targeting the PH domain
`of Akt. However, whether PIAs are effective in vivo and
`whether PIAs affect other PH-domain containing
`proteins are currently unknown.
`Perifosine is a novel orally bioavailable alkylpho-
`spholipid and structurally resembles naturally occurring
`phospholipids (Figure 3). Perifosine is known to be a
`CDK inhibitor and has displayed significant antiproli-
`ferative activity in vitro and in vivo in several human
`tumor model systems (Senderowicz, 2003a, b; Vink
`et al., 2005). It has been shown that perifosine can
`cause cell cycle arrest with induction of p21WAF1/CIP1 in a
`p53-independent fashion. By searching for the under-
`
`Oncogene
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 5 of 11
`
`

`

`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`7487
`
`Figure 3 Compound structures for representive Akt inhibitors
`
`lying mechanism, Kondapaka et al. (2003) has demon-
`strated that perifosine inhibits Akt phosphoylation by
`decreasing the plasma membrane localization of Akt,
`and this is substantially relieved by Myr-Akt along with
`relief of downstream drug effect on induction of
`p21WAF1/CIP1. While perifosine does not directly affect
`PI3K, PDK1 or Akt activity, its antitumor activity is, at
`least in part, mediated by inhibition of the Akt pathway
`(Kondapaka et al., 2003). In a recent phase I clinical
`trial, 1 patient had a partial response to perifosine and 2
`patients had stable disease among 42 patients with
`incurable solid malignancies, thus justifying additional
`investigation of this agent in a phase II trial (Van
`Ummersen et al., 2004).
`
`ATP-competitive inhibitors
`
`Through the elucidation of the sequence and structural
`composition of kinase active sites, coupled with the
`solution of numerous ATP-competitive ligand complex
`structures, significant advances have been made in
`developing inhibitors that are highly selective. This has
`been the case not only for kinases that are divergent in
`primary structure, but also for isoforms with highly
`conserved structure and ATP-binding sites. However,
`due to the fact that there is a high degree of homology in
`the ATP-binding pocket between Akt, PKA and PKC
`(Yang et al., 2002), many typical PKA and PKC
`inhibitors have been identified as inhibitors of Akt
`(Reuveni et al., 2002). So far, no ATP-competitive
`inhibitors specific against Akt have been identified.
`While a recent report described the design and synthesis
`of three Akt-selective inhibitors, their ability to inhibit
`Akt has not been tested in either cell culture or animal
`models (Breitenlechner et al., 2005).
`
`Pseudosubstrate inhibitors
`
`In contrast to ATP-competitive inhibitors, pseudosub-
`strate peptide inhibitors bind to the peptide/protein
`substrate sites of
`the catalytic domain and have
`been proven to be selective and potent for many kinases
`(Luo et al., 2004). This selectivity derives from the
`much larger peptide-kinase contact region that has
`evolved to discriminate between various protein sub-
`strates. A 14-mer peptide (AKTide-2T) was identified
`that binds to the substrate binding site of Akt1 and
`inhibits Akt1 activity (Luo et al., 2004). A hybrid
`peptide was recently constructed between this sequence
`and a sequence (amino acids 16–24) of the forkhead
`transcription factor FOXO3. The resulting 20-mer
`peptide construct inhibits Akt1 with a Ki of 1.1 mM.
`Replacement of a putative phosphorylation site serine
`(Ser) in the sequence with an alanine (Ala) moiety
`resulted in a further 10-fold improvement of potency.
`Efforts to truncate the peptide sequence showed that the
`20-mer could be truncated to a 17-mer with only slight
`loss of potency but further chain shortening resulted in
`larger losses of potency. These peptides are highly
`selective versus p70S6K, p90S6K PKA, Cdc2, Src, and
`PKC. Interestingly, the original 20-mer was equally
`active against SGK but the subsequent Ala for Ser
`replacement resulted in a highly selective inhibitor.
`Fusion peptides were also constructed to allow cell
`uptake and demonstrated dose-dependent inhibition of
`GSK3b phosphorylation. While these peptides are
`potent and selective inhibitors of Akt, their size makes
`them poor leads for small molecule inhibitor develop-
`ment. Significant
`truncation of
`the peptides and
`incorporation of peptidomimetic functionality would
`likely be needed to lead to cell-permeable molecules with
`good pharmacokinetics.
`
`Oncogene
`
`NOVARTIS EXHIBIT 2051
`Par v. Novartis, IPR 2016-01479
`Page 6 of 11
`
`

`

`7488
`
`API-2/triciribine
`
`Akt as target for anticancer drug discovery
`JQ Cheng et al
`
`By screening of the National Cancer Institute Diversity
`Set 2000-compound library, which are derived from
`approximate 140 000 compounds, a small molecule Akt
`pathway inhibitor, API-2, has recently been identified
`(Yang et al., 2004). API-2 suppressed the kinase activity
`and phosphorylation level of all
`three Akt
`family
`members. The inhibition of Akt kinase resulted in
`suppression of cell growth and induction of apoptosis in
`human cancer cells that harbor constitutively activated
`Akt due to overexpression of Akt or other genetic
`alterations such as PTEN mutation. API-2 is highly
`selective for Akt and does not inhibit PI3K, PDK1,
`PKC, SGK, PKA, Stat3, Erk-1/2 or JNK. Furthermore,
`API-2 potently inhibited tumor growth in nude mice of
`human cancer cells in which Akt is aberrantly expressed/
`activated but not of those cancer cells in which it is not.
`Recent data suggest
`that API-2 inhibition of Akt
`depends on the interaction with the PH domain of Akt.
`API-2 is a synthetic small molecule compound
`identified previously and named triciribine (TCN,
`NSC-154020; Figure 3) or tricyclic nucleoside (Chung
`et al., 1980; Wotring et al., 1986). Its 5’phosphate ester,
`triciribine monophosphate (TCN-P, NSC-280594) is the
`chemical entity advanced into clinical trials because it is
`more soluble than the parent drug (Wotring et al., 1986).
`TCN and TCN-P have antiviral and antineoplastic
`activity at low micromolar or submicromolar concen-
`trations (Migawa et al., 2005). By the early 1980s, TCN-
`P had been identified as an inhibitor of DNA and
`protein synthesis, and shown preclinical activity against
`leukemias and carcinomas. A number of phase I and II
`clinical trials of TCN/API-2 have been conducted in
`patients with advanced tumors, including carcinomas of
`the breast, colon, bladder, ovary, pancreas and lung
`(Cobb et al., 1983; Mittelman et al., 1983; Feun et al.,
`1984, 1993; Powis et al., 1986; Schilcher et al., 1986;
`Hoffman et al., 1996). Owing to the fact that TCN-p
`was used as a cytotoxic drug, the majority of clinical
`trials were carried out with high doses of the drug in
`order to achieve maximal clinical efficacy. While it
`exhibited antitumor activity in some patients, TCN/
`API-2 had significant
`side effects at high doses,
`including hepatotoxicity, hypertriglyceridemia, throm-
`bocytopenia, and hyperglycemia, which hampers its
`application in the clinic.
`As TCN/API-2 specifically targets Akt, it is reason-
`able to speculate that a given tumor with high levels of
`Akt activity will be more sensitive/responsive to TCN/
`API-2 treatment and that lower doses of TCN/API-2
`should achieve clinical efficacy without significant side
`effects in patients whose tumors exhibit elevated Akt
`levels. In fact, a recent study has shown that TCN/API-2
`effectively induces apoptosis and cell growth arrest in
`tumor cells with activation of Akt at a concentration of
`10 mM. In xenograft experiments, no detectable side
`effects were observed in 50 mice treated with TCN/API-
`2 at a concentration of l mg/kg/day, which significantly
`inhibited tumor growth in cancer cells overexpressing

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket