PKI-402 is a selective, reversible, ATP-competitive, equipotent inhibitor of class I phosphatidylinositol 3-kinases (PI3K), including PI3K-α mutants, and mammalian target of rapamycin (mTOR; IC50 versus PI3K-α = 2 nmol/L). PKI-402 inhibited growth of human tumor cell lines derived from breast, brain (glioma), pancreas, and non–small cell lung cancer tissue and suppressed phosphorylation of PI3K and mTOR effector proteins (e.g., Akt at T308) at concentrations that matched those that inhibited cell growth. In MDA-MB-361 [breast: Her2+ and PIK3CA mutant (E545K)], 30 nmol/L PKI-402 induced cleaved poly(ADP-ribose) polymerase (PARP), a marker for apoptosis. In vivo, PKI-402 inhibited tumor growth in MDA-MB-361, glioma (U87MG), and lung (A549) xenograft models. In MDA-MB-361, PKI-402 at 100 mg/kg (daily for 5 days, one round) reduced initial tumor volume of 260 mm3 to 129 mm3 and prevented tumor regrowth for 70 days. In MDA-MB-361 tumors, PKI-402 (100 mg/kg, single dose) suppressed Akt phosphorylation (at T308) and induced cleaved PARP. Suppression of phosphorylated Akt (p-Akt) was complete at 8 hours and still evident at 24 hours. Cleaved PARP was evident at 8 and 24 hours. In normal tissue (heart and lung), PKI-402 (100 mg/kg) had minimal effect on p-Akt, with no detectable cleaved PARP. Preferential accumulation of PKI-402 in tumor tissue was observed. Complete, sustained suppression of Akt phosphorylation may cause tumor regression in MDA-MB-361 and other xenograft models. We are testing whether dual PI3K/mTOR inhibitors can durably suppress p-Akt, induce cleaved PARP, and cause tumor regression in a diverse set of human tumor xenograft models. Mol Cancer Ther; 9(4); 976–84. ©2010 AACR.

Phosphatidylinositol 3-kinase (PI3K)-α plays a key role in the biology of human cancer. PI3K-α is a lipid kinase that is a central component in the PI3K/Akt/mammalian target of rapamycin (mTOR) signaling pathway. This pathway regulates cell proliferation, growth, survival, and apoptosis (1, 2). The deregulated activation of PI3K-α and its downstream effectors, including Akt and mTOR, has been linked to tumor initiation and maintenance. PI3K/Akt/mTOR pathway activation can be caused by loss of PTEN (the phosphatase that regulates PI3K signaling), overexpression or activation of some receptor tyrosine kinases [e.g., epidermal growth factor receptor (EGFR) and HER-2], interaction with activated Ras, overexpression of the PI3K-α gene (PIKC3A), or mutations in PIKC3A that cause elevated PI3K-α kinase activity (13). Aberrantly elevated PI3K/Akt/mTOR pathway signaling has been implicated in poor prognosis and survival in patients with various lymphatic tumors, as well as breast, prostate, lung, glioblastoma, melanoma, colon, and ovarian cancers (14). Additionally, PI3K/Akt/mTOR pathway activation contributes to resistance of cancer cells to both targeted anticancer therapies and conventional cytotoxic agents (5, 6). An effective inhibitor of the PI3K/Akt/mTOR pathway could both prevent cancer cell proliferation and induce programmed cell death (apoptosis; refs. 1, 2, 5). Therefore, the quest of the Wyeth PI3K inhibitor discovery project was to identify potent small-molecule inhibitors of the PI3K/Akt/mTOR signaling pathway, which we hypothesized would exert antitumor activity in a broad array of preclinical tumor models.

PKI-402 is an example of small molecules we have identified as potent inhibitors of the PI3K/mTOR signaling pathway. PKI-402 is a reversible, ATP-competitive, and equipotent inhibitor of class I PI3Ks, including the E545K and H1047R PI3K-α mutants, and mTOR (IC50 versus PI3K-α = 2 nmol/L). Selectivity of PKI-402 was established in a screen against 236 diverse human kinases. PKI-402 caused in vitro growth inhibition of human tumor cell lines derived from a diverse set of human tumor tissues, including breast, brain (glioma), pancreas, and non–small cell lung cancer (NSCLC) tissues. In vitro, PKI-402 suppressed phosphorylation of PI3K and mTOR effector proteins, particularly phosphorylated Akt (p-Akt) at T308, at concentrations that closely matched those that inhibited tumor cell growth. In MDA-MB-361, a breast tumor line with mutant PI3K-α (E545K) and elevated levels of Her2 receptor, PKI-402 induced cleaved poly(ADP-ribose) polymerase (PARP), a marker for apoptosis. In vivo, PKI-402 displayed antitumor activity (i.v. route) in breast [MDA-MB-361: Her2+ and PIK3CA (E545K)], glioma (U87MG and PTEN), and NSCLC (A549; K-Ras and STK11) xenograft models.

A key question about PI3K/mTOR signaling inhibitors is: can they cause tumor regression in preclinical models? Data we present here show that PKI-402 caused regression in the MDA-MB-361 xenograft model. PKI-402 effect was most pronounced at 100 mg/kg (daily for 5 days, one round), which reduced initial tumor volume and prevented tumor regrowth for 70 days. In MDA-MB-361 tumor tissue, PKI-402 at 100 mg/kg (single dose) fully suppressed p-Akt at both the T308 and the S473 sites at 8 hours and induced cleaved PARP. At 24 hours, p-Akt suppression was still evident, as was cleaved PARP. Biomarker analysis of heart and lung tissue showed minimal effect on p-Akt and no induction of cleaved PARP by PKI-402 at 100 mg/kg. Preferential accumulation of PKI-402 in tumor tissue was observed. Data presented here indicate that sustained suppression of p-Akt correlates with tumor regression in the MDA-MB-361 xenograft model.

Enzyme assays

Enzyme assays were done in fluorescent polarization (FP) format, adapted from the Echelon K-1100 PI3K FP assay kit protocol (7). Human class I PI3Ks and PI3K-α mutants (E545K and H1047R) were produced in Sf9 or purchased from Upstate Biotech. GST-GRP1 (murine) was produced in Escherichia coli and isolated by GST-Sepharose. Assay buffers were reaction buffer [20 mmol/L HEPES (pH 7.1), 2 mmol/L MgCl2, 0.05% CHAPS, and 0.01% β-mercaptoethanol] and stop/detection buffer [100 mmol/L HEPES (pH 7.5), 4 mmol/L EDTA, 0.05% CHAPS]. FP reaction was run for 30 min at room temperature in 20 μL of reaction buffer containing 20 μmol/L phosphatidylinositol 4,5-bisphosphate (PIP2), 25 μmol/L ATP, and <4% DMSO (compound solvent). FP reaction was stopped with 20 μL of stop/detection buffer (10 nmol/L probe and 40 nmol/L GST-GRP), and after 2 h, data were collected using an Envision plate reader (Perkin-Elmer).

Selectivity of PKI-402 was evaluated in the Invitrogen 236 human kinase panel at [ATP] = Km for each enzyme.

Cell culture, growth inhibition, and translocation assays

Cell lines obtained from the American Type Culture Collection were MDA-MB-361, MDA-MB-468, T47D, MCF7, BT474, HT29, HCT116, DLD1, U87MG, H157, NCI-H460, A549, NCI-H1975, NCI-H1650, NCI-H2170, KB, 786-0, A498, MIA-PaCa-2, and PC3. Mutational status (http://www.sanger.ac.uk/genetics/CGP/cosmic/) of various oncogenes is listed in Table 1. U2OS cells engineered to monitor FOXO1–green fluorescent protein (GFP) cellular translocation were from Thermo Scientific. All cell lines were propagated at 37°C in 5% CO2 incubators in supplier-recommended growth medium.

Table 1.

PKI-402 IC50 values in human tumor cell line growth inhibition assays

Cell lineMutationsIC50 (μmol/L)SD (n ≥ 3)
Breast 
    MDA-MB-361 PIK3CA 0.006 0.001 
    MDA-MB-468 PTEN, RB, p53 0.009 0.001 
    T47D p53, PIK3CA 0.013 0.002 
    BT474 PIK3CA, p53 0.026 0.006 
    MCF7 PIK3CA 0.101 0.014 
Colon 
    HCT116 K-Ras, PIK3CA 0.033 0.002 
    HT29 B-Raf, PIK3CA, p53 0.136 0.041 
    DLD1 K-Ras, PIK3CA 0.227 0.009 
Lung (NSCLC) 
    NCI-H157 K-Ras, STK11, p53 0.036 0.004 
    NCI-H460 K-Ras, PIK3CA, STK11 0.089 0.003 
    A549 K-Ras, STK11 0.116 0.031 
    NCI-H2170 p53 0.127 0.027 
    NCI-H1975 EGFR, PIK3CA, p53 0.210 0.015 
    NCI-H1650 EGFR, p53 0.349 0.061 
Pancreas 
    MIA-PaCa-2 K-Ras, p53 0.051 0.003 
Brain (glioma) 
    U87MG PTEN 0.077 0.017 
Prostate 
    PC3 PTEN, p53 0.016 0.002 
Epidermoid 
    KB  0.027 0.003 
Renal 
    A498 VHL 0.268 0.002 
    786-0 PTEN, VHL, p53 0.334 0.060 
Cell lineMutationsIC50 (μmol/L)SD (n ≥ 3)
Breast 
    MDA-MB-361 PIK3CA 0.006 0.001 
    MDA-MB-468 PTEN, RB, p53 0.009 0.001 
    T47D p53, PIK3CA 0.013 0.002 
    BT474 PIK3CA, p53 0.026 0.006 
    MCF7 PIK3CA 0.101 0.014 
Colon 
    HCT116 K-Ras, PIK3CA 0.033 0.002 
    HT29 B-Raf, PIK3CA, p53 0.136 0.041 
    DLD1 K-Ras, PIK3CA 0.227 0.009 
Lung (NSCLC) 
    NCI-H157 K-Ras, STK11, p53 0.036 0.004 
    NCI-H460 K-Ras, PIK3CA, STK11 0.089 0.003 
    A549 K-Ras, STK11 0.116 0.031 
    NCI-H2170 p53 0.127 0.027 
    NCI-H1975 EGFR, PIK3CA, p53 0.210 0.015 
    NCI-H1650 EGFR, p53 0.349 0.061 
Pancreas 
    MIA-PaCa-2 K-Ras, p53 0.051 0.003 
Brain (glioma) 
    U87MG PTEN 0.077 0.017 
Prostate 
    PC3 PTEN, p53 0.016 0.002 
Epidermoid 
    KB  0.027 0.003 
Renal 
    A498 VHL 0.268 0.002 
    786-0 PTEN, VHL, p53 0.334 0.060 

Cell growth inhibition was determined using the CellTiter 96 AQueous proliferation assay from Promega. Manufacturer's protocol was used, with adjustments for cell line growth characteristics. Data were collected after 72 h using a Wallac Victor2 V (Perkin-Elmer) 1420 multilabel HTS counter. FOXO-GFP translocation in U2OS cells was quantified after 60-min PKI-402 exposure using a Cellomics ArrayScan VTI Reader.

Cell lysis and western blotting

Briefly, 3 × 105 cells were seeded onto six-well microtiter plates (Nunc) 24 h before being exposed to PKI-402 in complete growth medium. Cells were exposed to compound for 4 h (unless indicated otherwise in figure legends). Subsequently, cell growth medium was removed and cells were washed twice with cold (4°C) PBS. Cell lysis buffer (0.2 mL) was added with mixing to insure cell lysis. Cell lysis buffer was 20 mmol/L Tris-HCl (pH 7.5), 150 mmol/L NaCl, 1 mmol/L Na2EDTA, 1 mmol/L EGTA, 1% NP40, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L β-glycerophosphate, 1 mmol/L Na3VO4, and 1 μg/mL leupeptin. Cell lysates were spun at 14,000 rpm for 30 s. Supernatant (60 μL) was combined with 30 μL of 3× protein gel loading buffer [187.5 mmol/L Tris-HCl (pH 6.8), 6% (w/v) SDS, 15% glycerol, 0.03% (w/v) bromphenol blue, and 125 mmol/L DTT]. Samples were boiled (5 min) and subjected to SDS-PAGE, transferred to nitrocellulose, and probed with various antibodies. Antibodies from Cell Signaling Technology were anti-Akt, anti–p-Akt at T308, anti–p-Akt at S473, anti-4EBP1, anti–phospho-4EBP1 (p-4EBP1) at T37/46, anti–p70 S6 kinase (p70S6K), anti–phospho-p70S6K (p-p70S6K) at T389, anti-PARP, anti–cleaved PARP at D214, anti–phospho-endothelial nitric oxide synthase (ENOS) at S1177, anti–phospho-glycogen synthase kinase 3α/β (GSK3α/β) at S21/9, anti–phospho-PRAS40 at T246, and anti–phospho-p44/42 mitogen-activated protein kinase (MAPK; extracellular signal-regulated kinase 1/2; T202/Y204). Conjugate (horseradish peroxidase) secondary antibody enabled chemiluminescent detection. Inhibition of protein phosphorylation was quantified from Western blots analyzed on the Bio-Rad Fluor-S MultiImager using Quantity One Analysis software.

Caspase activation assay

The Caspase-Glo 3/7(Promega) luminescent assay was used to measure caspase-3/7 activity in MDA-MB-361 and HCT116 cell lines. Cells were exposed to PKI-402 for 4 or 24 h. Assays were done in 96-well format (∼4,000 cells per well). Caspase-3/7 activity was quantitated using a Wallac Victor2 V 1420 multilabel HTS counter.

Establishment of xenograft tumors, efficacy studies, and biomarker analysis

Establishment of tumors, group randomization, tumor, and body weight recording during efficacy studies were described elsewhere (8). PKI-402 or vehicle was administered by i.v. route. All in vivo studies using nude mice were conducted under an approved Institutional Animal Care and Use Committee protocol. Tumor weight was calculated by the formula tumor weight (mg) = (d2 × D/2), where d and D are the shortest and longest diameters of the tumor, respectively, measured in millimeters. Significant (statistically, Student's t test) reduction in the tumor growth of treated groups compared with controls (vehicle) was defined as P < 0.05.

Pharmacodynamic (biomarker) measurements were done on tumor-bearing female nude mice administered PKI-402. Tumor or normal tissue samples were collected from euthanized animals, homogenized, washed twice with cold (4°C) PBS, and then treated with cell lysis buffer (described above). Cell lysates were processed and probed with the various antibodies as described above.

Enzyme assays

PKI-402 (Fig. 1) is a pan class I PI3K inhibitor with a 2 nmol/L IC50 against PI3K-α. IC50 determinations for PKI-402 against class I PI3Ks (α, β, δ, and γ), the two most common mutant forms of PI3K-α (E545K and H1047R; ref. 9), and the PI3K-related kinase family member mTOR are shown in Fig. 1.

Figure 1.

Structure of PKI-402 and IC50 (μmol/L) inhibition data for class I PI3Ks, the most frequently occurring mutant forms (E545K and H1047R) of PI3K-α, and mTOR.

Figure 1.

Structure of PKI-402 and IC50 (μmol/L) inhibition data for class I PI3Ks, the most frequently occurring mutant forms (E545K and H1047R) of PI3K-α, and mTOR.

Close modal

PKI-402 is an ATP-competitive triazolopyrimidine scaffold compound (Supplementary Data). Mutant forms of PI3K-α reported to have elevated lipid kinase activity (10) were inhibited by PKI-402 at concentrations equivalent to the IC50 for wild-type PI3K-α. PI3K-β, PI3K-δ, and PI3K-γ isoforms were inhibited at concentrations only 3- to 8-fold higher than that of PI3K-α. mTOR kinase inhibition (IC50, 3 nmol/L) indicated that PKI-402 was an equipotent class I PI3K/mTOR inhibitor.

Characterization of PKI-402 against a panel of 236 human protein kinases (Invitrogen) confirmed a highly selective profile (Supplementary Data). In this kinase panel, only C-Raf and B-Raf (wild-type and V600E mutant) were inhibited by PKI-402. Subsequent IC50 values of 7 μmol/L were determined for all Raf enzymes. The IC50s for PKI-402 against all other kinases in the Invitrogen panel were >10 μmol/L.

Cell growth inhibition assay

Table 1 lists IC50 data for human tumor cell growth inhibition by PKI-402. Cell lines from an array of tumor tissues, including breast, colon, lung, epidermoid, renal, pancreas, brain, and prostate, were tested. PKI-402 was a potent cell growth inhibitor in most human tumor cell lines, with IC50 values of <250 nmol/L (17 of 20). In all cases where examined, PKI-402 was found to suppress phosphorylation of downstream effectors of PI3K signaling (e.g., Akt) at concentrations that closely matched growth inhibition IC50.

Higher IC50 values against A498, 786-0, and NCI-H1650 (IC50 range, 268–349 nmol/L) were observed, but when we evaluated phosphorylation of the PI3K effector Akt (T308) in A498 and 785-0, we found that it occurred at concentrations that closely matched growth inhibition IC50s (Supplementary Data).

PKI-402 in vitro profile in biomarker, caspase activation, and FOXO1-GFP translocation assays

PKI-402 effect on phosphorylation of PI3K and mTOR effector proteins and activation of caspase-3/7 in MDA-MB-361 [Her2+ and PIK3CA (E545K)]

The effect of PKI-402 on a representative group of PI3K/mTOR effector proteins in MDA-MB-361 is shown in Fig. 2A. This was done to link PKI-402 enzyme inhibition to cellular antiproliferative effects. PKI-402 lipid kinase activity in cells was not directly measured. Class I PI3Ks convert PIP2 to PIP3 (phosphatidylinositol 3,4,5-trisphosphate) at the inner cell membrane (1, 2). PIP3 is bound by the pleckstrin homology domains of both PDK1 and Akt serine/threonine kinases, resulting in their close proximity at the inner cell membrane where PDK1 phosphorylates (at T308) and activates Akt. Suppression of cellular PIP3 by PKI-402 was indirectly shown by potent (IC50, <10 nmol/L) suppression of Akt phosphorylation at T308 (Fig. 2A). Full activation of Akt kinase occurs when the mTOR-containing TORC2 protein complex phosphorylates Akt at S473. Figure 2A shows that potent (IC50, <30 nmol/L) suppression of Akt phosphorylation at S473 was also caused by PKI-402. PKI-402 did not affect the overall Akt content in MDA-MB-361 cells at any concentration tested, indicating that p-Akt suppression was not an artifact of compound cytotoxicity.

Figure 2.

In vitro profile of PKI-402. MDA-MB-361 [Her2+/PIK3CA (E545K)] cells were exposed to PKI-402 (0.003–0.3 μmol/L) for 4 h. A, PKI-402 suppression of p-Akt (both T308 and S473) and suppression of phosphorylation of Akt effectors [GSK3 (at S9/21), PRAS40 (at T246), and ENOS (at S1177)] and mTOR effectors [p70S6K (at T389) and p-4EBP1 (at T37/46)]. PKI-402 induction of cleaved PARP (cPARP) at 0.1 and 0.3 μmol/L. B, caspase-3/7 activity in MDA-MB-361 after a 4-h exposure to PKI-402, HKI-272 (Her2 inhibitor), or PD0325901 (MEK inhibitor) at 0.03 to 3.0 μmol/L. (◊), maximal caspase-3/7 activity was caused by 1.0 to 3.0 μmol/L PKI-402. Neither HKI-272 (□) nor PD0325901 (Δ) induced caspase-3/7 activity. C and D, PKI-402 combined with 0.1 μmol/L of either HKI-272 (×) or PD0325901 (+) did not increase caspase-3/7 activity. U2OS (FOXO1-GFP) cells were exposed to PKI-402 (0.003–10.0 μmol/L) for 1 h. C, nuclear to cytoplasmic ratio of FOXO1-GFP in U2OS cells after 60-min incubation with PKI-402 (IC50, 21 nmol/L). D, translocation of FOXO1-GFP (green) in U2OS from the cytoplasm (left) to the nucleus (right) after 60-min exposure to PKI-402. Nuclei (blue) were stained with Hoechst 33342.

Figure 2.

In vitro profile of PKI-402. MDA-MB-361 [Her2+/PIK3CA (E545K)] cells were exposed to PKI-402 (0.003–0.3 μmol/L) for 4 h. A, PKI-402 suppression of p-Akt (both T308 and S473) and suppression of phosphorylation of Akt effectors [GSK3 (at S9/21), PRAS40 (at T246), and ENOS (at S1177)] and mTOR effectors [p70S6K (at T389) and p-4EBP1 (at T37/46)]. PKI-402 induction of cleaved PARP (cPARP) at 0.1 and 0.3 μmol/L. B, caspase-3/7 activity in MDA-MB-361 after a 4-h exposure to PKI-402, HKI-272 (Her2 inhibitor), or PD0325901 (MEK inhibitor) at 0.03 to 3.0 μmol/L. (◊), maximal caspase-3/7 activity was caused by 1.0 to 3.0 μmol/L PKI-402. Neither HKI-272 (□) nor PD0325901 (Δ) induced caspase-3/7 activity. C and D, PKI-402 combined with 0.1 μmol/L of either HKI-272 (×) or PD0325901 (+) did not increase caspase-3/7 activity. U2OS (FOXO1-GFP) cells were exposed to PKI-402 (0.003–10.0 μmol/L) for 1 h. C, nuclear to cytoplasmic ratio of FOXO1-GFP in U2OS cells after 60-min incubation with PKI-402 (IC50, 21 nmol/L). D, translocation of FOXO1-GFP (green) in U2OS from the cytoplasm (left) to the nucleus (right) after 60-min exposure to PKI-402. Nuclei (blue) were stained with Hoechst 33342.

Close modal

TORC1 as well as TORC2 mTOR complexes were inhibited by PKI-402. Examples of PKI-402 effect on TORC1 were suppression of 4EBP1 and p70S6 kinase phosphorylation. Both p70S6K and 4EBP1 phosphorylation was inhibited at IC50s of <10 nmol/L (Fig. 2A).

PKI-402 suppression of Akt phosphorylation caused consequent effects on Akt effectors such as PRAS40, ENOS, and GSK3. PRAS40 regulates mTOR activity, ENOS generates nitrous oxide in blood vessels and is involved in regulation of vascular function and angiogenesis, and GSK3 is a serine/threonine protein kinase that regulates cell cycle progression and glucose metabolism (2, 11, 12). Akt phosphorylation of PRAS40 at T246 was suppressed at an IC50 of <30 nmol/L (Fig. 2A). Akt phosphorylation of ENOS at S1177 and GSK3α/GSK3β at S9/S21 was suppressed by PKI-402 at IC50s of <10 nmol/L (Fig. 2A).

An effect of PKI-402 particularly evident in MDA-MB-361 cells was induction of cleaved PARP, an indicator of cells undergoing apoptosis (13, 14). Complete PKI-402 suppression of Akt phosphorylation in MDA-MB-361 correlated with detectable cleaved PARP at 0.1 μmol/L PKI-402 (Fig. 2A). Cleaved PARP was detected in MDA-MB-361 within 4 hours after exposure to PKI-402.

We also measured PKI-402 activation of caspase-3 (Caspase-3/7 assay, Promega) in MDA-MB-361 because caspase-3 is a critical mediator of apoptosis, being either partially or totally responsible for the proteolytic cleavage of many key proteins including the nuclear enzyme PARP (14). Figure 2B shows that PKI-402 caused a dose-dependent increase in caspase-3/7 activity at 4 hours. MDA-MB-361 cells overexpress the Her2 receptor; therefore, we tested the ability of HKI-272 (irreversible Her2 kinase inhibitor; ref. 15) or PD0325901 [MAP/extracellular signal-regulated kinase kinase (MEK) inhibitor; ref. 16] to either induce caspase-3/7 activity alone or enhance PKI-402–induced caspase activity. Neither PD0325901 nor HKI-272 caused caspase-3/7 activation at concentrations tested (Fig. 2B). When PKI-402 was combined with 0.1 μmol/L (shown) or 1.0 μmol/L (data not shown) of either PD0325901 or HKI-272, caspase-3/7 activation in MDA-MB-361 cells was unaffected. These data indicate the significant addiction of MDA-MB-361 to PI3K/mTOR signaling to maintain cell viability. Less than 10% of MDA-MB-361 cells exposed to PKI-402 at 0.3 μmol/L (or higher) for 24 hours remained viable (Supplementary Data).

PKI-402 effect on FOXO1(FKHR)-GFP translocation in U2OS

Forkhead proteins (e.g., FOXO1) comprise a highly conserved family of transcription factors that control genes encoding proteins regulating insulin production, apoptosis, and cell cycle (17). FOXO1 activity is regulated by Akt-mediated phosphorylation. Akt-phosphorylated FOXO1 is sequestered in the cytosol by 14-3-3 protein, and unphosphorylated FOXO1 locates to the cell nucleus. Figure 2B shows that PKI-402, which suppressed Akt phosphorylation (T308 and S473; Fig. 2A), caused translocation of FOXO1(FKHR)-GFP to cell nuclei in U2OS cells engineered (Thermo Scientific) to monitor compound-mediated FOXO1-GFP nuclear translocation. The IC50 value for PKI-402 effect on FOXO1-GFP nuclear translocation was 21 nmol/L (Fig. 2C). Figure 2D shows FOXO1-GFP (green) cytoplasmic distribution in untreated U2OS cells (left), with nuclear translocation evident (at right) after 1-hour exposure to PKI-402.

PKI-402 induction of cleaved PARP and activation of caspase-3/7 in HCT116 cells

To determine whether the PKI-402 effect on cell survival observed in MDA-MB-361 breast tumor cells was unique, or also evident in other tumor cells, we evaluated the sensitivity of the colon tumor line HCT116 (K-Ras and PIK3CA) to this compound. In vitro growth inhibition IC50 of HCT116 by PKI-402 was 0.033 μmol/L. Suppression of p-Akt-T308 was observed at ≥0.1 μmol/L after 4-hour exposure to PKI-402. However, no cleaved PARP was evident after 4-hour exposure to PKI-402 up to 3 μmol/L. Longer exposure (24 hours) of HCT116 to PKI-402 again showed suppression of p-Akt-T308 (IC50, <0.1 μmol/L; Fig. 3A, top), with only a trace cleaved PARP band at 3.0 μmol/L.

Figure 3.

PKI-402 suppression of p-Akt-T308, induction of cleaved PARP, and activation of caspase-3/7 alone, and with 0.1 μmol/L PD0325901 (MEK) in HCT116 (K-Ras and PIK3CA) at 24 h. A, top, cleaved PARP, p-Akt-T308, p-MAPK (T202/Y204), and actin after PKI-402 (0.03–3.0 μmol/L) alone for 24 h [IC50s, <0.03 μmol/L (p-Akt-T308) and ≥3.0 μmol/L (cleaved PARP)]. No PKI-402 effect was evident on p-MAPK (T202/Y204) or actin. Bottom, effect of combined PKI-402 and 0.1 μmol/L PD0325901. The p-Akt-T308 IC50 was <0.03 μmol/L, cleaved PARP was now evident at 0.1 μmol/L, and p-MAPK (T202/Y204) was fully suppressed. B, induction of caspase-3/7 activity by PKI-402 (□), PD0325901 (Δ), and combined (◊) PKI-402 and PD0325901 (at 0.1 μmol/L) in HCT116 exposed to compounds for 24 h.

Figure 3.

PKI-402 suppression of p-Akt-T308, induction of cleaved PARP, and activation of caspase-3/7 alone, and with 0.1 μmol/L PD0325901 (MEK) in HCT116 (K-Ras and PIK3CA) at 24 h. A, top, cleaved PARP, p-Akt-T308, p-MAPK (T202/Y204), and actin after PKI-402 (0.03–3.0 μmol/L) alone for 24 h [IC50s, <0.03 μmol/L (p-Akt-T308) and ≥3.0 μmol/L (cleaved PARP)]. No PKI-402 effect was evident on p-MAPK (T202/Y204) or actin. Bottom, effect of combined PKI-402 and 0.1 μmol/L PD0325901. The p-Akt-T308 IC50 was <0.03 μmol/L, cleaved PARP was now evident at 0.1 μmol/L, and p-MAPK (T202/Y204) was fully suppressed. B, induction of caspase-3/7 activity by PKI-402 (□), PD0325901 (Δ), and combined (◊) PKI-402 and PD0325901 (at 0.1 μmol/L) in HCT116 exposed to compounds for 24 h.

Close modal

Because HCT116 cells have both PIK3CA and K-Ras mutations, we tested whether a MEK inhibitor, PD0325901, could influence PKI-402 effect on cleaved PARP induction. PD0325901 IC50 for growth inhibition of HCT116 was 0.230 μmol/L, and IC50 for phosphorylated MAPK (p-MAPK) (T202/Y204) suppression was ∼0.050 μmol/L (24 hours). Cleaved PARP was not detected in HCT116 after PD0325901 exposure at 24 hours (up to 3.0 μmol/L; data not shown). When HCT116 cells were exposed to 0.1 μmol/L PD0325901 and PKI-402 (from 0.03 to 3.0 μmol/L) for 24 hours, a substantial increase in cleaved PARP was observed. Cleaved PARP signal was detectable at 0.1 μmol/L, with a dose-dependent increase up to 3.0 μmol/L (Fig. 3A, bottom).

The effect of PKI-402 alone and in combination with PD0325901 on caspase-3/7 activity was measured at 24 hours (Fig. 3B). PKI-402, or PD0325901, alone weakly induced caspase-3/7 activity at the highest compound concentration (3.0 μmol/L). In contrast, PD0325901 (0.1 μmol/L) combined with PKI-402 increased caspase-3/7 activity at PKI-402 concentrations from 0.03 to 3.0 μmol/L (5-fold increase at 3.0 μmol/L). This effect will be investigated in future in vivo efficacy studies with HCT116.

In vivo efficacy and biomarker profile of PKI-402 in MDA-MB-361 (breast) tumor xenografts

PKI-402 at 25, 50, and 100 mg/kg caused regression of MDA-MB-361 tumors (Fig. 4A). PKI-402 effect was most pronounced at 100 mg/kg (daily for 5 days, one round), which reduced an initial tumor volume of 260 mm3 to 129 mm3 and prevented tumor regrowth for 70 days. PKI-402 given at 100 mg/kg daily for 5 days was well tolerated; however, continuous exposure at this level for longer than 7 days did cause weight loss. Tumor regrowth occurred between days 16 and 20 when PKI-402 was administered at 25 and 50 mg/kg (daily for 5 days, two rounds; both regimens well tolerated). However, when PKI-402 was readministered at 100 mg/kg, large tumors were significantly (P < 0.02) reduced in volume. In the 25 mg/kg treatment group, PKI-402 readministered at 100 mg/kg (daily for 5 days, one round) on day 37 caused ∼650 mm3 tumors to shrink to ∼200 mm3, a ∼69% reduction in tumor volume. In the 50 mg/kg treatment group, PKI-402 readministered on day 45 (100 mg/kg, daily for 3 days, two rounds) caused ∼600 mm3 tumors to ultimately shrink to ∼200 mm3, a ∼58% reduction in tumor volume. In Fig. 4B, photographic evidence of PKI-402 effect on MDA-MB-361 tumors treated at 100 mg/kg (daily for 5 days, one round) is shown.

Figure 4.

In vivo efficacy against MDA-MB-361 xenografts and in vivo biomarkers in tumor and heart tissue. A, PKI-402 given at 100 mg/kg (Δ) daily for 5 d for one round or at 25 (▴) or 50 (▪) mg/kg daily for 5 d for two rounds. The 100 mg/kg treatment caused tumor regression with no regrowth until day ∼70. Initial tumor regression in both 50 and 25 mg/kg groups was followed by tumor regrowth at days 16 to 20. Readministration of PKI-402 (↓, at 100 mg/kg, daily for 3 d) either once (25 mg/kg group, on day 37) or twice (50 mg/kg group, on day 45 and then again on day 53) caused shrinkage (>50%) of large tumors. Points, mean tumor volume from 10 mice; bars, SEM. B, a control tumor and a tumor from the 100 mg/kg treatment group at day 14. C, cleaved PARP, p-Akt (T308 and S473), p-p70S6K (T389), p-S6 (S235/236), and actin status in tumor xenograft tissue. PKI-402 was given in single-dose format at 100, 50, and 25 mg/kg. Tumor tissue was harvested at 8 h after compound administration. Suppression of protein phosphorylation and cleaved PARP induction was greatest at the 100 mg/kg dose. D, comparison PKI-402 effect on MDA-MB-361 tumor and heart tissues in MDA-MB-361 tumor-bearing nude mice. PKI-402 was given at 100 mg/kg (single dose) and tissue was harvested at 24 h. Note the lack of cleaved PARP in the heart tissue.

Figure 4.

In vivo efficacy against MDA-MB-361 xenografts and in vivo biomarkers in tumor and heart tissue. A, PKI-402 given at 100 mg/kg (Δ) daily for 5 d for one round or at 25 (▴) or 50 (▪) mg/kg daily for 5 d for two rounds. The 100 mg/kg treatment caused tumor regression with no regrowth until day ∼70. Initial tumor regression in both 50 and 25 mg/kg groups was followed by tumor regrowth at days 16 to 20. Readministration of PKI-402 (↓, at 100 mg/kg, daily for 3 d) either once (25 mg/kg group, on day 37) or twice (50 mg/kg group, on day 45 and then again on day 53) caused shrinkage (>50%) of large tumors. Points, mean tumor volume from 10 mice; bars, SEM. B, a control tumor and a tumor from the 100 mg/kg treatment group at day 14. C, cleaved PARP, p-Akt (T308 and S473), p-p70S6K (T389), p-S6 (S235/236), and actin status in tumor xenograft tissue. PKI-402 was given in single-dose format at 100, 50, and 25 mg/kg. Tumor tissue was harvested at 8 h after compound administration. Suppression of protein phosphorylation and cleaved PARP induction was greatest at the 100 mg/kg dose. D, comparison PKI-402 effect on MDA-MB-361 tumor and heart tissues in MDA-MB-361 tumor-bearing nude mice. PKI-402 was given at 100 mg/kg (single dose) and tissue was harvested at 24 h. Note the lack of cleaved PARP in the heart tissue.

Close modal

Biomarker evaluation was done on MDA-MB-361 tumor tissue from mice treated with 25, 50, and 100 mg/kg (single dose) of PKI-402. Figure 4C shows the complete suppression of p-Akt (at T308 and S473) and p-p70S6K at 8 hours after administration of PKI-402 at 100 mg/kg. Distinct PKI-402 effect on this set of PI3K/mTOR effector proteins was also evident at the 50 and 25 mg/kg dosing levels. Cleaved PARP was evident when PKI-402 was given at 50 and 100 mg/kg. Table 2 lists serum and tumor tissue concentrations after 50 or 100 mg/kg single-dose administration of PKI-402. At 50 mg/kg PKI-402, preferential accumulation in tumor tissue relative to serum was evident at 4 hours and persisted up to 8 hours. Tumor/serum ratio (6.2) peaked at 6 hours. Tumor/serum ratio for PKI-402 at 100 mg/kg was 8.9 at 24 hours.

Table 2.

Concentration (ng/mL) of PKI-402 in serum and tumor following a single dose

Pharmacokinetic summaryng/mL
CompoundRouteDoseFormulation1 h4 h6 h8 h24 h
PKI-402 i.v. 50 mg/kg Saline/lactic acid Serum 6,228 1,215 527 549  
Tumor 8,319 6,576 3,265 2,219  
PKI-402 i.v. 100 mg/kg Saline/lactic acid Serum  814 132 
Tumor    2,813 1,184 
Pharmacokinetic summaryng/mL
CompoundRouteDoseFormulation1 h4 h6 h8 h24 h
PKI-402 i.v. 50 mg/kg Saline/lactic acid Serum 6,228 1,215 527 549  
Tumor 8,319 6,576 3,265 2,219  
PKI-402 i.v. 100 mg/kg Saline/lactic acid Serum  814 132 
Tumor    2,813 1,184 

NOTE: PKI-402 accumulation in tumor tissue was observed at 4 to 8 h (50 mg/kg) and at 8 to 24 h (100 mg/kg).

The effect of PKI-402 on PI3K/mTOR effector protein phosphorylation and cleaved PARP in both tumor and normal (e.g., heart and lung) tissue was assessed. In MDA-MB-361 tumor tissue, PKI-402 (100 mg/kg, single dose) effect on p-Akt (both T308 and S473), p-p70S6K, and p-pS6 was reduced (compared with 8 hours) but still evident at 24 hours (Fig. 4D, left). Cleaved PARP signal, however, was equal if not greater than that observed at 8 hours (Fig. 4C). In both heart (Fig. 4D, right) and lung tissue (data not shown) from nude mice bearing MDA-MB-361 tumors, PKI-402 (single dose, 100 mg/kg) reduced p-Akt (T308 and S473) at 24 hours, but there was no detectable cleaved PARP.

In vivo efficacy of PKI-402 in A549 (NSCLC) and U87MG (glioma) tumor xenografts

PKI-402 was tested for antitumor efficacy in the A549 (NSCLC) and U87MG (glioma) xenograft models. A549 cells contain mutant K-Ras and STK11. Because these mutations influence both PI3K and mTOR activity, we tested PKI-402 in vivo growth-inhibitory effects on A549 tumors. Figure 5A shows that PKI-402 significantly (P < 0.02) inhibited the growth of A549 tumors in nude mice at 25 and 50 mg/kg (daily for 5 days, for three rounds).

Figure 5.

PKI-402 in vivo efficacy against A549 (NSCLC) and U87MG (glioma) xenografts. A, PKI-402 antitumor activity against A549 xenografts at 10 mg/kg (

graphic
), 25 mg/kg (
graphic
), and 50 (▪) mg/kg (21-d study). B, PKI-402 antitumor activity against U87MG xenografts at 100 mg/kg (▴) in a 7-d study. Points, mean tumor volume from 10 mice; bars, SEM.

Figure 5.

PKI-402 in vivo efficacy against A549 (NSCLC) and U87MG (glioma) xenografts. A, PKI-402 antitumor activity against A549 xenografts at 10 mg/kg (

graphic
), 25 mg/kg (
graphic
), and 50 (▪) mg/kg (21-d study). B, PKI-402 antitumor activity against U87MG xenografts at 100 mg/kg (▴) in a 7-d study. Points, mean tumor volume from 10 mice; bars, SEM.

Close modal

PKI-402 inhibition of U87MG tumor growth showed efficacy in a PTEN-negative glioblastoma multiforme tumor line. PKI-402 at 100 mg/kg (daily for 5 days, one round) caused significant (P < 0.01) reduction in tumor growth relative to untreated controls (Fig. 5B). Rapid growth of U87MG tumors in this model was evident by the ∼5-fold increase in control tumor size over the 7-day study period. PKI-402 clearly attenuated this growth, permitting only a ∼2.3-fold increase in tumor size.

We identified PKI-402 in enzyme assays that measured activity of class I PI3Ks and mTOR. PKI-402 was a potent inhibitor of these enzymes, including the most frequently occurring mutant forms of PI3K-α (8). Analysis of PKI-402 against a panel of 236 human kinases showed a highly selective profile. In vitro, PKI-402 inhibition of cell growth and suppression of Akt phosphorylation correlated with induction of apoptosis in MDA-MB-361 (breast) human tumor cells. The proapoptotic effects of PKI-402 alone or in combination (i.e., PD0325901) on other cell lines (HCT116; Fig. 3) suggest that PKI-402 could have a broad range of activity and possibly more than a cytostatic effect on xenograft tumor models.

A key question about PI3K/mTOR signaling inhibitors is: can they cause tumor regression in preclinical models? Based on current data disclosed about other PI3K/mTOR inhibitors (e.g., BEZ235, PI-103, and SF1126; refs. 18, 19), PKI-402 shows a distinctive single-agent profile in that it can potently induce cell death in vitro and cause tumor regression in vivo (MDA-MB-361 model).

Indeed, at 25, 50, and 100 mg/kg, PKI-402 caused regression of MDA-MB-361 tumors (Fig. 4A). PKI-402 effect was most pronounced at 100 mg/kg (daily for 5 days, one round), which reduced an initial tumor volume (260 mm3 to 129 mm3) and prevented tumor regrowth for 70 days. Tumor regrowth occurred between days 16 and 20 when PKI-402 was administered at 25 and 50 mg/kg (daily for 5 days, two rounds). However, these enlarged tumors remained sensitive to PKI-402. For example, when PKI-402 was readministered at 100 mg/kg (daily for 3 days, one round) at day 37 (25 mg/kg group), large tumors (∼600 mm3) were reduced in volume by 69%.

Biomarker analysis done with MDA-MB-361 tumor tissue from mice given PKI-402 at 100 mg/kg (single dose) showed sustained suppression of Akt phosphorylation at both the T308 and the S473 sites and induction of cleaved PARP. Suppression of p-Akt was complete at 8 hours and still evident at 24 hours, whereas cleaved PARP was evident at both 8 and 24 hours. Biomarker analysis of heart and lung tissue from MDA-MB-361 tumor-bearing mice given PKI-402 at 100 mg/kg showed minimal effect on p-Akt and no induction of cleaved PARP (Fig. 4D). Preferential accumulation of PKI-402 in tumor tissue was observed (Table 2). It may be that complete and sustained suppression of p-Akt is needed to cause tumor regression in the MDA-MB-361 and other tumor xenograft models. We are doing detailed pharmacokinetic, pharmacodynamic, and antitumor efficacy studies to confirm this hypothesis. If this link is established, it could become a key criterion for advancing compounds to clinical development.

Finally, we established PKI-402 antitumor efficacy in additional in vivo models. These were A549 (NSCLC; K-Ras/STK11) and U87MG (glioma, PTEN). Clinical outcome for NSCLC is especially bleak (20). PKI-402 efficacy against A549 tumors (Fig. 5) supports the concept that inhibition of the K-Ras effector PI3K is an effective antitumor strategy (1, 5). In cells lacking functional STK11, such as A549, defective AMPK regulation of mTOR, and in turn deregulation of mTOR-controlled cell metabolism, suggests possible sensitivity to PKI-402 mTOR-inhibitory effects. PKI-402 significantly inhibited the growth of A549 tumors in nude mice at both 25 and 50 mg/kg (daily for 5 days, three rounds).

As with NSCLC, patients with PTEN-negative glioblastoma multiforme tumors generally have a poor prognosis (21). In clinical settings, such tumors are resistant to EGFR inhibitors, radiotherapy, and most alkylating agents (22). PKI-402 (100 mg/kg, daily for 5 days) inhibited U87MG tumor growth, with significant reduction in tumor growth relative to untreated controls. Despite the high dosing level needed for efficacy, these data suggest that compounds such as PKI-402 may be useful either as a single agent or in combination with cytostatic or cytotoxic (e.g., temozolomide; ref. 23) drugs in treatment of glioblastoma multiforme.

All authors are employees of Wyeth Research. No other potential conflicts of interest were disclosed.

We thank Ker Yu and Lourdes Toral-Barza for evaluating PKI-402 in their mTOR kinase assay.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Engelman
JA
,
Luo
J
,
Cantley
LC
. 
The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism
.
Nat Rev Genet
2006
;
7
:
606
19
.
2
Shaw
RJ
,
Cantley
LC
. 
Ras, PI(3)K and mTOR signaling controls tumor cell growth
.
Nature
2006
;
441
:
424
30
.
3
Kok
K
,
Geering
B
,
Vanhaesebroeck
B
. 
Regulation of phosphoinositide 3-kinase expression in health and disease
.
Trends Biochem Sci
2009
;
34
:
115
27
.
4
Vanhaesebroeck
B
,
Leevers
SJ
,
Ahmadi
K
, et al
. 
Synthesis and function of 3-phosphorylated inositol lipids
.
Annu Rev Biochem
2001
;
70
:
535
602
.
5
Yuan
TL
,
Cantley
LC
. 
PI3K pathway alterations in cancer: variations on a theme
.
Oncogene
2008
;
27
:
5497
510
.
6
Keniry
M
,
Parsons
R
. 
The role of PTEN signaling perturbations in cancer and in targeted therapy
.
Oncogene
2008
;
27
:
5477
85
.
7
Yang
X
,
Li
P
,
Feldberg
L
, et al
. 
A directly labeled TR-FRET assay for monitoring phosphoinositide-3-kinase activity
.
Comb Chem High Throughput Screen
2006
;
9
:
565
70
.
8
Golas
JM
,
Arndt
K
,
Etienne
C
, et al
. 
SKI-606, a 4-anilino-3-quinolinecarbonitrile dual inhibitor of Src and Abl kinases, is a potent antiproliferative agent against chronic myelogenous leukemia cells in culture and causes regression of K562 xenografts in nude mice
.
Cancer Res
2003
;
63
:
375
81
.
9
Bader
AG
,
Kang
S
,
Vogt
PK
. 
Cancer-specific mutations in PIK3CA are oncogenic in vivo
.
Proc Natl Acad Sci U S A
2006
;
103
:
1475
9
.
10
Carson
JD
,
Van Aller
G
,
Lehr
R
, et al
. 
Effects of oncogenic p110α subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase
.
Biochem J
2008
;
409
:
519
24
.
11
Fulton
D
,
Gratton
JP
,
McCabe
TJ
, et al
. 
Regulation of endothelium-derived nitric oxide production by the protein kinase Akt
.
Nature
1999
;
399
:
597
601
.
12
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
.
13
Tewari
M
,
Quan
LT
,
O'Rourke
K
, et al
. 
Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase
.
Cell
1995
;
81
:
801
9
.
14
Fernandes-Alnemri
T
,
Litwack
G
,
Alnemri
ES
. 
CPP32, a novel human apoptotic protein with homology to Caenorhabditis elegans cell death protein Ced-3 and mammalian interleukin-1β-converting enzyme
.
J Biol Chem
1994
;
269
:
30761
4
.
15
Kwak
EL
,
Sordella
R
,
Bell
DW
, et al
. 
Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib
.
Proc Natl Acad Sci U S A
2005
;
102
:
7665
70
.
16
Sosman
JA
,
Puzanov
I
. 
Molecular targets in melanoma from angiogenesis to apoptosis
.
Clin Cancer Res
2006
;
12
:
2376
83s
.
17
Arden
KC
. 
FoxO: linking new signaling pathways
.
Mol Cell
2004
;
14
:
416
8
.
18
Engelman
JA
. 
Targeting PI3K signalling in cancer: opportunities, challenges and limitations
.
Nat Rev Cancer
2009
;
9
:
550
62
.
19
Liu
P
,
Cheng
H
,
Roberts
TM
,
Zhao
JJ
. 
Targeting the phosphoinositide 3-kinase pathway in cancer
.
Nat Rev Drug Discov
2009
;
8
:
627
44
.
20
Sharma
SV
,
Bell
DW
,
Settleman
J
,
Haber
DA
. 
Epidermal growth factor receptor mutations in lung cancer
.
Nat Rev Cancer
2007
;
7
:
169
81
.
21
Mellinghoff
IK
,
Wang
MY
,
Vivanco
I
, et al
. 
Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors
.
N Engl J Med
2005
;
353
:
2012
24
.
22
Norden
AD
,
Drappatz
J
,
Wen
PY
. 
Novel anti-angiogenic therapies for malignant gliomas
.
Lancet Neurol
2008
;
7
:
1152
60
.
23
Maira
SM
,
Stauffer
F
,
Brueggen
J
, et al
. 
Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity
.
Mol Cancer Ther
2008
;
7
:
1851
63
.

Supplementary data