The introduction of MAPK pathway inhibitors paved the road for significant advancements in the treatment of BRAF-mutant (BRAFMUT) melanoma. However, even BRAF/MEK inhibitor combination therapy has failed to offer a curative treatment option, most likely because these pathways constitute a codependent signaling network. Concomitant PTEN loss of function (PTENLOF) occurs in approximately 40% of BRAFMUT melanomas. In this study, we sought to identify the nodes of the PTEN/PI3K pathway that would be amenable to combined therapy with MAPK pathway inhibitors for the treatment of PTENLOF/BRAFMUT melanoma. Large-scale compound sensitivity profiling revealed that PTENLOF melanoma cell lines were sensitive to PI3Kβ inhibitors, albeit only partially. An unbiased shRNA screen (7,500 genes and 20 shRNAs/genes) across 11 cell lines in the presence of a PI3Kβ inhibitor identified an adaptive response involving the IGF1R–PI3Kα axis. Combined inhibition of the MAPK pathway, PI3Kβ, and PI3Kα or insulin-like growth factor receptor 1 (IGF1R) synergistically sustained pathway blockade, induced apoptosis, and inhibited tumor growth in PTENLOF/BRAFMUT melanoma models. Notably, combined treatment with the IGF1R inhibitor, but not the PI3Kα inhibitor, failed to elevate glucose or insulin signaling. Taken together, our findings provide a strong rationale for testing combinations of panPI3K, PI3Kβ + IGF1R, and MAPK pathway inhibitors in PTENLOF/BRAFMUT melanoma patients to achieve maximal response. Cancer Res; 76(2); 390–402. ©2015 AACR.

The observation that approximately 50% of malignant melanomas harbor mutations in the BRAF gene has triggered intensive efforts to develop inhibitors of mutant BRAF, including vemurafenib and dabrafenib. Although the introduction of these drugs has revolutionized the treatment of BRAF-mutant (BRAFMUT) melanoma patients, sustained benefit is limited by intrinsic and acquired resistance. The major mechanism of acquired resistance appears to involve reactivation of MAPK signaling, and indeed dual BRAF/MEK inhibition has improved clinical responses compared with single-agent treatment (1). Nevertheless, BRAF/MEK inhibitor combination therapy fails to provide a curative treatment option for patients with BRAF-mutant melanoma most likely due to concurrent genetic alterations conferring codependencies. In accordance with a complementary role of PTEN, PTENLOF alterations were identified in approximately 40% of BRAFMUT melanoma and were shown to promote Braf-driven metastatic melanoma in a genetically engineered mouse model (2). Furthermore, PTENLOF alterations are associated with BRAF inhibitor resistance in cellular models and in patients as well as decreased progression-free survival upon dabrafenib treatment (3–5). Similarly, decreased PTEN protein levels were found to predict for vemurafenib insensitivity, highlighting the urgent need for the optimization of targeted therapies for this genetic context (6).

PTEN encodes a lipid phosphatase that antagonizes PI3K activity by dephosphorylating phosphatidylinositol-3,4,5-trisphosphate. Class IA PI3Ks are heterodimeric lipid kinases composed of a p85 regulatory subunit and one of three catalytic subunits, namely p110α, p110β, or p110δ (PI3Kα, PI3Kβ, or PI3Kδ). Whereas PI3Kα and PI3Kβ are ubiquitously expressed, expression of PI3Kδ has been reported to be largely restricted to the hematopoietic system. In contrast with this, interrogation of the Cancer Cell Line Encyclopedia (CCLE) and associated mRNA expression profiles revealed significant expression levels of all three isoforms in the majority of cell lines derived from diverse lineages (7). Nevertheless, PTENLOF cancers and cell lines of different lineages have been shown to specifically depend on PI3Kβ (8–10). In this study, we demonstrate for the first time the exquisite dependence of PTENLOF melanoma on the PI3Kβ isoform. Although PI3Kβ inhibitors were only partially efficacious, using an unbiased RNAi approach, we identified a homogeneous pattern of chemosensitization to PI3Kβ inhibition with PI3Kα, insulin-like growth factor receptor 1 (IGF1R), and components of the MAPK pathway as top hits in PTENLOF/BRAFMUT melanoma. We subsequently confirmed that dual inhibition of PI3Kα/β was required to permanently block PI3K signaling and that addition of PI3Kα/β inhibitors strongly synergized with MAPK pathway inhibitors. In comparison with direct targeting of PI3Kα, anti-IGF1R antibodies, which presumably block receptor tyrosine kinase (RTK)–mediated PI3Kα activation in the tumor tissue only, were as efficacious, but did not lead to the induction of glucose abnormalities. These data provide a strong rationale for the first-line combination of MAPK pathway inhibitors with panPI3K or PI3Kβ/IGF1R inhibitors to overcome intrinsic and acquired resistance to BRAF inhibitors in PTENLOF/BRAFMUT melanoma.

Cell lines and reagents

Cell lines were obtained from and cultured as recommended by the ATCC, Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, and Health Science Research Resources Bank. Authenticity was confirmed by single-nucleotide polymorphism fingerprinting, and cells were cultivated less than 20 passages before use.

High-throughput cell line profiling and manual cell proliferation assays

The sensitivity profiles of melanoma lines used for this study were extracted from a high-throughput profiling approach described previously (7). Manual determination of the antiproliferative effect of compounds or compound combinations was performed as described previously using methylene blue or resazurin staining as readout (11, 12). Detailed information is summarized in Supplementary Materials and Methods.

Synthetic lethality pooled shRNA screen

Lentiviral supernatants of barcoded custom library (Cellecta) of approximately 170,000 shRNAs targeting approximately 7,500 genes (divided into three sublibraries) were produced according to the manufacturer's recommended protocols. After infection and puromycin selection, cells were cultivated ±1 μmol/L rac-KIN-193 at a minimal representation of 1,000 cells/shRNA for five population doublings. The representation of each shRNA barcode was measured by next-generation sequencing on an Illumina HiSeq platform. Hits for each panel of cell lines were identified as described in Supplementary Materials and Methods.

Immunoblotting and coimmunoprecipitation assays

Immunoblotting and coimmunoprecipitation experiments were done according to standard methods. Detailed information and a comprehensive list of antibodies are provided in Supplementary Materials and Methods.

Reverse-phase protein array

Reverse-phase protein array (RPPA) was performed as described previously (13). In brief, cells were seeded in duplicate in 96-well plates at an appropriate density, and the following day, they were treated with serial compound dilutions for 1 hour, washed with ice-cold PBS, and lysed in CLB96 lysis buffer (Bayer Technology). Pulverized tumor samples were processed accordingly.

Proteomics

Immunoprecipitates were separated by SDS-PAGE and in-gel digested using a Trypsin/Lys-C Mix (Promega). Peptides were analyzed by LC-MS/MS (LTQ OrbiTrap Elite; Thermo) as described previously (14), using spectral counting as a proxy for the protein quantity.

Cell death and apoptosis assay

Induction of cell death was determined in triplicates on an Array Scan VTI platform (Cellomics) using 2 μg/mL propidium iodide (PI) (Life Technologies) and 1 μg/mL Hoechst33342 (Sigma). Induction of apoptosis was determined in triplicates by Annexin V/PI staining (Life Technologies) according to the manufacturer's flow cytometry protocol.

Tumor growth inhibition in vivo

WM-266-4 cells (2 × 106) were injected subcutaneously into the flanks of nude mice. At a tumor volume of 200 to 300 cm3, mice were treated with the indicated compounds. Detailed information, including analysis of glucose/insulin levels, is provided in Supplementary Materials and Methods.

PTENLOF melanomas specifically depend on PI3Kβ

To determine whether specific compound mechanisms selectively impair the growth of PTENLOF melanoma cells, we compared the activity of 1397 compounds for their effect on the viability of 14 PTENLOF and 27 PTENWT melanoma lines from the CCLE based on their activity area (combined measure of compound potency and efficacy) as described elsewhere (7). This analysis revealed that PI3Kβ inhibition best discriminates among 390 different modes of action tested between PTENLOF and PTENWT melanoma lines (Fig. 1A; Supplementary Table S1). In contrast, PI3Kα, PI3Kδ, PI3Kγ, or panPI3K inhibitors did not score significantly as selectively active compounds toward PTENLOF melanoma lines. Interestingly, sensitivity to PI3Kα-selective inhibitors was detected primarily in PTENWT lines, but these compounds did not pass the significance cutoff (P < 0.01; data not shown).

Figure 1.

PTENLOF melanomas specifically depend on PI3Kβ. A, analysis showing drugs of distinct modes of action that exhibit superior activity area in growth inhibition in 14 PTENLOF over 27 PTENWT melanoma lines. The Wilcoxon test was conducted to determine the statistical significance between PTENLOF and PTENWT panels and compounds were ranked accordingly. The dashed line represents a cutoff of P < 0.01; P values included in illustration refer to PI3Kβi. B, immunoblot analysis documenting the effect of serial dilutions (0.01–20 μmol/L) of PI3Kαi and PI3Kβi on phosphorylated and total Akt levels in RVH-421. C, RVH-421 cells were treated with serial dilutions of the indicated drugs, and their effect on phosphorylated and total Akt was evaluated by RPPA. D, scatter plots showing Amax (expressed as percentage of DMSO) versus GI50 of the indicated compounds in proliferation assays of a panel of BRAFMUT melanoma lines. Each data point reflects the mean of at least two independent experiments. P values indicate the statistical significance of differentiation between PTENLOF/BRAFMUT and PTENWT/BRAFMUT lines (two-tailed t test based on GI50). PI3Kβi1/2, rac-KIN-193/TGX221; panPI3Ki, GDC0941; PI3Kαi, BYL719.

Figure 1.

PTENLOF melanomas specifically depend on PI3Kβ. A, analysis showing drugs of distinct modes of action that exhibit superior activity area in growth inhibition in 14 PTENLOF over 27 PTENWT melanoma lines. The Wilcoxon test was conducted to determine the statistical significance between PTENLOF and PTENWT panels and compounds were ranked accordingly. The dashed line represents a cutoff of P < 0.01; P values included in illustration refer to PI3Kβi. B, immunoblot analysis documenting the effect of serial dilutions (0.01–20 μmol/L) of PI3Kαi and PI3Kβi on phosphorylated and total Akt levels in RVH-421. C, RVH-421 cells were treated with serial dilutions of the indicated drugs, and their effect on phosphorylated and total Akt was evaluated by RPPA. D, scatter plots showing Amax (expressed as percentage of DMSO) versus GI50 of the indicated compounds in proliferation assays of a panel of BRAFMUT melanoma lines. Each data point reflects the mean of at least two independent experiments. P values indicate the statistical significance of differentiation between PTENLOF/BRAFMUT and PTENWT/BRAFMUT lines (two-tailed t test based on GI50). PI3Kβi1/2, rac-KIN-193/TGX221; panPI3Ki, GDC0941; PI3Kαi, BYL719.

Close modal

Accordingly, treatment of the PTENLOF/BRAFMUT melanoma cell line RVH-421 with the PI3Kβi rac-KIN-193, but not with the selective PI3Kαi BYL719, led to a reduction in Akt phosphorylation within 1 hour in a dose-dependent manner (Fig. 1B). Quantitative analysis using RPPA validated this finding in three different cell lines (Fig. 1C; Supplementary Fig. S1A). Short-term treatment with two different PI3Kβi's (rac-KIN-193/TGX-221) reduced Akt phosphorylation similarly to the panPI3Ki (GDC-0941), whereas a PI3Kα inhibitor reduced Akt phosphorylation only at high concentrations where it is also active against other PI3K isoforms.

To test whether inhibition of Akt phosphorylation led to growth inhibition, the antiproliferative effect of these compounds was determined in a panel of 9 PTENLOF/BRAFMUT and 11 PTENWT/BRAFMUT melanoma lines (Fig. 1D). Concordance of PTENLOF calls according to the CCLE and absence of PTEN protein was confirmed by immunoblotting the cell lines (Supplementary Fig. S1B). Analysis of the maximum efficacy (Amax) and GI50 values confirmed that PI3Kβ inhibition, but not PI3Kα or panPI3K inhibition, selectively impaired the growth of PTENLOF/BRAFMUT compared with PTENWT/BRAFMUT melanoma lines. However, both PI3Kβi's only partially blocked proliferation (Amax ≥ 25%) in most melanoma lines. Interestingly, the panPI3Ki was as efficient as the PI3Kβi's in reducing pAkt levels, but superior in inhibiting proliferation. This observation suggests either that short-term pAkt inhibition might not be indicative of complete or prolonged pathway inhibition or that an alternative non-Akt substrate of PI3K plays a critical role in the regulation of proliferation.

Taken together, our unbiased approach enabled us to identify PI3Kβ as the primary PI3K isoform driving Akt signaling in this genetic context.

Deep-coverage pooled shRNA screen identifies PIK3CA, IGF1R, and MAPK signaling as the most synergistic partners to PI3Kβ inhibition in PTENLOF/BRAFMUT melanoma

As the treatment with PI3Kβi's yielded only partial responses in PTENLOF/BRAFMUT melanoma lines, we sought to determine whether additional signaling nodes might contribute to mitigating the overall response. To discover such nodes, a deep-coverage pooled shRNA screen was performed wherein a lentiviral shRNA library targeting approximately 7,500 genes with approximately 20 shRNAs/gene was introduced into 5 PTENLOF/BRAFMUT and 6 PTENWT/BRAFMUT melanoma lines. After five population doublings (±PI3Kβi), the relative abundance of each individual shRNA was determined by deep sequencing (Fig. 2A). Redundant siRNA activity (RSA) scores of drug-treated and untreated control samples were compared using Limma's linear model within each genotype to identify gene products that were selectively required in the presence of PI3Kβi in the PTENLOF/BRAFMUT cells only (15). PIK3CA, IGF1R, BRAF, MAP2K1, and MAPK1 were identified as top sensitizers to PI3Kβ inhibition in a genotype-selective manner with a marked distinction from any other hit in this unbiased approach (Fig. 2B).

Figure 2.

Deep-coverage pooled shRNA screen identifies PIK3CA, IGF1R, and MAPK signaling as the most synergistic partners to PI3Kβ inhibition in PTENLOF/BRAFMUT melanoma. A, schematic screening workflow. B, volcano plot showing statistical significance compared with changes in shRNA representation upon PI3Kβi relative to vehicle treatment for each gene. Each data point represents −log10 (P value) by the fold change obtained by the drug-treated versus untreated control comparison within 5 PTENLOF/BRAFMUT or 6 PTENWT/BRAFMUT lines. The shRNA library was split into three sublibraries, with well-known oncogenic drivers being represented in two to three sublibraries. C, synergistic inhibition of proliferation upon PI3Kβ/PI3Kα inhibitor combination in RVH-421 and WM-266-4. Dose matrices highlight percentages of proliferation inhibition relative to DMSO, with 100 indicating complete block of proliferation and >100 indicative of cell death. PI3Kβi1/2, rac-KIN-193/GSK-2636771; PI3Kαi, BYL719.

Figure 2.

Deep-coverage pooled shRNA screen identifies PIK3CA, IGF1R, and MAPK signaling as the most synergistic partners to PI3Kβ inhibition in PTENLOF/BRAFMUT melanoma. A, schematic screening workflow. B, volcano plot showing statistical significance compared with changes in shRNA representation upon PI3Kβi relative to vehicle treatment for each gene. Each data point represents −log10 (P value) by the fold change obtained by the drug-treated versus untreated control comparison within 5 PTENLOF/BRAFMUT or 6 PTENWT/BRAFMUT lines. The shRNA library was split into three sublibraries, with well-known oncogenic drivers being represented in two to three sublibraries. C, synergistic inhibition of proliferation upon PI3Kβ/PI3Kα inhibitor combination in RVH-421 and WM-266-4. Dose matrices highlight percentages of proliferation inhibition relative to DMSO, with 100 indicating complete block of proliferation and >100 indicative of cell death. PI3Kβi1/2, rac-KIN-193/GSK-2636771; PI3Kαi, BYL719.

Close modal

The above data suggested that PI3Kα and IGF1R signaling were attenuating the response to PI3Kβ inhibitors. To further test this notion, and to ask whether a PI3Kαi would hence act synergistically with a PI3Kβi in PTENLOF/BRAFMUT melanoma lines, two PTENLOF/BRAFMUT lines were treated with a PI3Kβi and a PI3Kαi in a matrix combination setting (Fig. 2C). Although single-agent treatment with PI3Kαi did not affect proliferation, combination with PI3Kβi1 led to an enhanced antiproliferative effect at all concentrations tested (Fig. 2C). Synergy scores (SS > 2) demonstrated that the combination of both drugs acted in a robust synergistic manner. The structurally diverse PI3Kβi2 GSK-2636771 yielded similar results, indicating that these synergistic effects are likely on-target. In conclusion, the combination of specific small-molecule inhibitors recapitulated the synthetic lethal interaction identified by RNAi in the chemosensitization screen.

Small-molecule inhibition of PI3Kβ triggers the recruitment of p85 to IRS2 in PTENLOF/BRAFMUT melanoma

The fact that PI3Kαi activity was only observed in combination with a PI3Kβi raised the possibility that PI3Kβ inhibition leads to the activation of PI3Kα. Activation of PI3Kα is regulated by membrane recruitment of the p110α/p85 complex to tyrosine-phosphorylated RTKs or their adaptor proteins via the interaction of p85 SH2 domains with phosphorylated YXXM motifs (16–18). In order to examine PI3Kα recruitment to RTKs or adaptor proteins, we next studied the interaction of p85 with phosphotyrosine-containing proteins upon PI3Kβi treatment. Immunoblot analysis of p85 coimmunoprecipitates using a pTyr antibody revealed one specific signal upon PI3Kβi treatment in RVH-421 cells (Fig. 3A). Label-free mass spectrometric analysis of these immunocomplexes identified 20 proteins that were enriched upon PI3Kβi treatment (Supplementary Table S2). Among these, only the adaptor protein insulin receptor substrate 2 (IRS2) that mediates the activation of signaling pathways in response to ligand stimulation of cell surface receptors is reported to be Tyr-phosphorylated and harbors 6 YXXM motifs, therefore being a potent activator of PI3K (16, 19). Subsequent, direct coimmunoprecipitation assays confirmed that PI3Kβ inhibition induced a robust p85/IRS2 interaction (Fig. 3B). These studies also demonstrated PI3Kβi-induced tyrosine phosphorylation of IRS2 with concomitant reduction of pIRS2 S731 levels and the consequential change in IRS2 protein migration in the SDS gel (20). Time-course experiments revealed that the interaction was detectable within 30 minutes of treatment (Supplementary Fig. S2A).

Figure 3.

Small-molecule inhibition of PI3Kβ triggers the recruitment of p85 to IRS2 in PTENLOF/BRAFMUT melanoma. A, immunoprecipitation of p85 from lysates derived from RVH-421 treated ±PI3Kβi for 2 hours. Precipitates were separated by SDS-PAGE and immunoblotted with p85- and pTyr antibodies. B–D, coimmunoprecipitation experiments were carried out using cell lysates of RVH-421 (B and C) and A-375 (D) treated with the indicated compounds for 2 hours and blotted for p85, IRS2, and pTyr (IRS2). A total of 2.5% of the amount of lysate used for each IP was immunoblotted with the indicated antibodies as input control. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; and PI3Kαi, BYL719.

Figure 3.

Small-molecule inhibition of PI3Kβ triggers the recruitment of p85 to IRS2 in PTENLOF/BRAFMUT melanoma. A, immunoprecipitation of p85 from lysates derived from RVH-421 treated ±PI3Kβi for 2 hours. Precipitates were separated by SDS-PAGE and immunoblotted with p85- and pTyr antibodies. B–D, coimmunoprecipitation experiments were carried out using cell lysates of RVH-421 (B and C) and A-375 (D) treated with the indicated compounds for 2 hours and blotted for p85, IRS2, and pTyr (IRS2). A total of 2.5% of the amount of lysate used for each IP was immunoblotted with the indicated antibodies as input control. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; and PI3Kαi, BYL719.

Close modal

To explore the correlation of PI3K pathway inhibition and p85/IRS2 interaction in more detail, RVH-421 cells were treated with a panel of PI3K inhibitors of different isoform specificities (Fig. 3C). Both the PI3Kβi and the panPI3Ki reduced phosphorylation of Akt, S6, NDRG1, a known substrate of the mTORC2-regulated kinase SGK, and IRS2 (Fig. 3C, bottom; ref. 21), whereas the PI3Kαi did not affect PI3K signaling in this cell line. In keeping with this, treatment with both the PI3Kβi and the panPI3Ki triggered the p85/IRS2 interaction, whereas the PI3Kαi did not (Fig. 3C, top). In contrast, in the PTENWT/BRAFMUT cell line A-375, the PI3KαI, but not the PI3KβI, caused the same consequences as described before for the PI3Kβi in the PTENLOF/BRAFMUT setting (Fig. 3D).

In conclusion, our data support the notion that PI3K signaling is reactivated upon PI3Kβ inhibition in PTENLOF/BRAFMUT melanoma by recruitment of PI3K to an activated RTK via the adaptor molecule IRS2.

Small-molecule inhibition of mTOR, PDK1, and Akt relieves feedback inhibition on IRS2 in PTENLOF/BRAFMUT melanoma

In addition to the role of PI3Kβ in activating the classical PI3K pathway, PI3Kβ exhibits Akt/mTOR-independent functions, including the regulation of autophagy (22). To investigate whether classical PI3Kβ downstream signaling accounts for the induction of p85/IRS2 interaction, we asked whether inhibition of mTOR, PDK1, and Akt with selective small molecules would also trigger the p85/IRS2 interaction. The catalytic mTORi AZD8055, the allosteric mTORC1i RAD001 (Fig. 4A; Supplementary Fig. S2B), the catalytic PDK1i GSK-2334470 (Fig. 4B; Supplementary Fig. S2C), and MK2206, which inhibits Akt membrane localization (Fig. 4C; Supplementary Fig. S2D), all induced p85/IRS2 complex formation in a dose-dependent manner.

Figure 4.

Small-molecule inhibition of mTOR, PDK1, and Akt relieves feedback inhibition on IRS2 in PTENLOF/BRAFMUT melanoma. A–C, RVH-421 cells were treated with the indicated compounds at various concentrations for 2 hours. The p85 or control IgG antibody precipitates were immunoblotted with the indicated antibodies. PI3Kβi, rac-KIN-193; mTORi, AZD8055, mTORC1i, RAD001; PDK1i, GSK-2334470; and AKTi, MK2206.

Figure 4.

Small-molecule inhibition of mTOR, PDK1, and Akt relieves feedback inhibition on IRS2 in PTENLOF/BRAFMUT melanoma. A–C, RVH-421 cells were treated with the indicated compounds at various concentrations for 2 hours. The p85 or control IgG antibody precipitates were immunoblotted with the indicated antibodies. PI3Kβi, rac-KIN-193; mTORi, AZD8055, mTORC1i, RAD001; PDK1i, GSK-2334470; and AKTi, MK2206.

Close modal

These data strongly suggest that PI3Kβ inhibition leads to the loss of feedback inhibition mediated by Ser/Thr kinases, including Akt, mTORC1, and S6K and, subsequently, to the IRS2-dependent recruitment of PI3Kα through activated RTKs.

Small-molecule inhibition of IGF1R signaling potentiates PI3Kβ inhibitor activity in PTENLOF/BRAFMUT melanoma

As IGF1R was found among the highest scoring sensitizers to PI3Kβ inhibition (Fig. 2B), we speculated that IGF1R was the sole RTK mediating the reactivation of PI3K signaling upon PI3Kβi treatment. In agreement with this, AEW541 treatment (IGF1Ri) blocked PI3Kβi-induced p85/IRS2 interaction in a dose-dependent manner (Fig. 5A). In keeping with previous reports showing an increase in RTK expression levels by PI3K inhibition (23), long-term PI3Kβi treatment led to an increase in IGF1R levels in the four PTENLOF/BRAFMUT models tested (Supplementary Fig. S3). To further validate IGF1R inhibition as a sensitizer to PI3Kβ inhibition, cell viability was determined upon treatment with this combination. Whereas the IGF1Ri had only minor antiproliferative effects, a fixed concentration of the compound enhanced PI3Kβi efficacy at all concentrations tested (Fig. 5B; Supplementary Fig. S4A). Furthermore, matrix combination assays confirmed the synergistic activity of IGF1Ri with two structurally distinct PI3Kβi's, but not the PI3Kαi, in two PTENLOF/BRAFMUT lines (Fig. 5C; Supplementary Fig. S4B). PanPI3Ki treatment also led to synergistic effects, albeit less significant due to its stronger efficacy as single agent. Importantly, the selectivity of this synergy with PI3Kβi's in the genetic context of PTENLOF was underscored by the lack of combination activity in the PTENWT/BRAFMUT melanoma cell line A-375 (Supplementary Fig. S4C).

Figure 5.

Small-molecule inhibition of IGF1R signaling potentiates PI3Kβ inhibitor activity in PTENLOF/BRAFMUT melanoma. A, RVH-421 cells were treated with the indicated compounds for 72 hours. The p85, IRS2, and control antibody immunoprecipitates and input lysates were immunoblotted with the indicated phospho-specific or total target protein antibodies. B and C, synergistic proliferation inhibition upon PI3K/IGF1R inhibitor combination in RVH-421 cells. RVH-421 cells were treated with PI3Ki or IGF1Ri as single agents or in combination, with increasing concentrations of PI3Ki and a fixed concentration of 1.1 μmol/L IGF1Ri (B) or in a matrix setting (C). The data shown in B represent the mean (±SEM) of two replicates. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; PI3Kαi, BYL719; and IGF1Ri, AEW541.

Figure 5.

Small-molecule inhibition of IGF1R signaling potentiates PI3Kβ inhibitor activity in PTENLOF/BRAFMUT melanoma. A, RVH-421 cells were treated with the indicated compounds for 72 hours. The p85, IRS2, and control antibody immunoprecipitates and input lysates were immunoblotted with the indicated phospho-specific or total target protein antibodies. B and C, synergistic proliferation inhibition upon PI3K/IGF1R inhibitor combination in RVH-421 cells. RVH-421 cells were treated with PI3Ki or IGF1Ri as single agents or in combination, with increasing concentrations of PI3Ki and a fixed concentration of 1.1 μmol/L IGF1Ri (B) or in a matrix setting (C). The data shown in B represent the mean (±SEM) of two replicates. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; PI3Kαi, BYL719; and IGF1Ri, AEW541.

Close modal

These data indicate that IGF1R inhibition acts in combination specifically with PI3Kβ inhibitors to block proliferation of PTENLOF/BRAFMUT melanoma by interfering with the reactivation of PI3K signaling through PI3Kα.

Concomitant inhibition of PI3Kβ, IGF1R, and MAPK signaling leads to a complete and sustained pathway blockade and induction of cell death in PTENLOF/BRAFMUT melanoma

In keeping with the known dependence of BRAFMUT melanoma on the MAPK pathway, the pooled shRNA screen identified shRNAs targeting key nodes of MAPK signaling (BRAF, MAP2K1, and MAPK1) among the strongest sensitizers to PI3Kβ inhibition (Fig. 2B). Indeed, the two PI3Kβi's and the panPI3Ki showed strong synergy upon combination treatment with the BRAFi (LGX818) or the MEKi (MEK162) while the PI3Kαi did not (Fig. 6A and B; Supplementary Fig. S5A and S5B). As expected, synergy was not observed in the PTENWT background (Supplementary Fig. S5C and S5D). Thus, our data support the notion that adding PI3Kβ inhibitors to MAPK pathway inhibitors in PTENLOF/BRAFMUT melanoma elicits a robust and specific combinatorial activity.

Figure 6.

Concomitant inhibition of PI3Kβ, IGF1R, and MAPK signaling leads to complete and sustained pathway blockade and induction of cell death in PTENLOF/BRAFMUT melanoma. A and B, determination of PI3K/BRAF (A) and PI3K/MEK (B) inhibitor synergy in RVH-421 using proliferation assays. Dose matrices highlight percentages of proliferation inhibition relative to DMSO, with 100 indicating complete block of proliferation and >100 indicative of cell death. C, effects of treatment with the indicated inhibitors as single agents or in combination for 2 or 72 hours on RVH-421 were evaluated by immunoblotting using the indicated antibodies. D, fractions of living (A, PI), necrotic (A, PI+), early (A+, PI), and late (A+, PI+) apoptotic RVH-421 and WM-266-4 cells were measured upon treatment with the indicated compounds for 72 hours. Data are represented as mean (±SD) of triplicates. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; PI3Kαi, BYL719; BRAFi, LGX818; and MEKi, MEK162.

Figure 6.

Concomitant inhibition of PI3Kβ, IGF1R, and MAPK signaling leads to complete and sustained pathway blockade and induction of cell death in PTENLOF/BRAFMUT melanoma. A and B, determination of PI3K/BRAF (A) and PI3K/MEK (B) inhibitor synergy in RVH-421 using proliferation assays. Dose matrices highlight percentages of proliferation inhibition relative to DMSO, with 100 indicating complete block of proliferation and >100 indicative of cell death. C, effects of treatment with the indicated inhibitors as single agents or in combination for 2 or 72 hours on RVH-421 were evaluated by immunoblotting using the indicated antibodies. D, fractions of living (A, PI), necrotic (A, PI+), early (A+, PI), and late (A+, PI+) apoptotic RVH-421 and WM-266-4 cells were measured upon treatment with the indicated compounds for 72 hours. Data are represented as mean (±SD) of triplicates. PI3Kβi1/2, rac-KIN-193/GSK-2636771; panPI3Ki, GDC0941; PI3Kαi, BYL719; BRAFi, LGX818; and MEKi, MEK162.

Close modal

In a next step, the impact of IGF1R-mediated activation of PI3Kα on the ability of a PI3Kβi to permanently block the PI3K/mTOR pathway was assessed in two cell lines (Fig. 6C; Supplementary Fig. S6A and S6B). Although treatment with the PI3Kβi for 2 hours efficiently blocked PI3K/mTOR signaling, a rebound after 72 hours of treatment was detected (lanes 1/2 and 11/12). Loss of activity due to compound instability was excluded as the compound had been refreshed appropriately. Dual PI3Kα/β or panPI3K inhibition prevented this PI3K/mTOR signaling rebound (lane 16; Supplementary Fig. S7A). Consistent with an exclusive role for IGF1R in PI3Kα activation, the IGF1Ri (or an anti-IGF1R antibody; Supplementary Fig. S6B) substituted for the PI3Kαi in preventing this rebound (lane 17). The elevated MAPK signaling in this BRAFMUT setting might interfere with PI3K/mTOR signaling by regulation of the activity of the TSC1/2 complex and phosphorylation of S6 at Ser235/236 by its effector RSK (24–26). Indeed, concomitant inhibition of MEK/PI3Kβ/PI3Kα led to a significant decrease not only in Erk but also in S6 phosphorylation when treatment was continued for 72 hours. This sustained MAPK/PI3K/mTOR blockade led to the induction of PARP cleavage (lanes 9 and 19). Also, combined inhibition of MEK/PI3Kβ/IGF1R restored complete pathway inhibition in long-term assays and triggered PARP cleavage (lanes 10 and 20).

Furthermore, the induction of apoptosis triggered by the MEK and BRAF inhibitors was more pronounced when the MAPK pathway inhibitors were combined with a PI3Kβi as measured by Annexin V/PI staining in four PTENLOF/BRAFMUT melanoma lines (Fig. 6D; Supplementary Fig. S7B). Most importantly, however, further addition of either the PI3Kαi or the IGF1Ri was associated with an even more pronounced increase in the fraction of early and/or late apoptotic cells underscoring the need for sustained concomitant blockade of MAPK and PI3K signaling in order to maximize impact on cell viability in this genetic context (Fig. 6D; Supplementary Fig. S7B). In addition, matrix combination assays revealed that PI3K pathway inhibition using PI3Kβi/PI3Kαi or PI3Kβi/IGF1Ri combinations also enhanced the antiproliferative effect of the current BRAF/MEK inhibitor regimen in the setting of PTENLOF/BRAFMUT and shifted the response toward cell death (Supplementary Fig. S8A and S8B).

Taken together, these results show that, in PTENLOF/BRAFMUT melanoma, the benefit of adding PI3Kβ to MAPK pathway inhibition is limited due to reactivation of PI3Kα by relieving a feedback inhibition on IGF1R signaling (Supplementary Fig. S9). Thus, sustained and concomitant inhibition of PI3Kβ together with PI3Kα or IGF1R and MAPK signaling is necessary to achieve complete long-term pathway inhibition and maximize induction of cell death.

Concomitant inhibition of PI3Kβ, IGF1R, and MAPK signaling blocks tumor growth in vivo

Due to the dominant role of PI3Kα in glucose homeostasis, the substitution of PI3Kα with IGF1R inhibitors provides the unique opportunity to circumvent dose-limiting on-target side effects of PI3Kα inhibition in insulin-sensitive tissues. Therefore, the efficacy and effects on glucose homeostasis of PI3Kαi or IGF1Ri treatments in combination with PI3Kβi/BRAFi were determined in the PTENLOF/BRAFMUT xenograft model WM-266-4. Contrary to the in vitro findings, the PI3Kαi and PI3Kβi were almost equally efficient in inhibiting tumor growth, most likely due to the reported antiangiogenic effect of PI3Kα inhibition (27). The inhibition of both isoforms individually boosted the effect of the BRAFi, but dual PI3Kα/β inhibition increased the efficacy of BRAFi treatment even further. Interestingly, although IGF1R inhibition (anti-IGF1R antibody) substituted for PI3Kα inhibition in the blockade of tumor growth (Fig. 7A), there were profound differences in their effects on glucose homeostasis. As expected, PI3Kαi treatment resulted in hyperglycemia that was particularly pronounced in the triple PI3Kαi/PI3Kβi/BRAFi combination. Strikingly, no effects on insulin signaling and body weight were observed in any of the anti-IGF1R antibody treatment arms (Fig. 7B, data not shown). Pharmacokinetics analysis confirmed the expected exposures excluding possible drug–drug interactions (data not shown).

Figure 7.

Concomitant inhibition of PI3Kβ/PI3Kα or PI3Kβ/IGF1R maximizes the efficacy of BRAF inhibitors in PTENLOF/BRAFMUT melanoma in vivo. A, in vivo efficacy on tumor growth. Mice bearing WM-266-4 xenografts were treated with the indicated compounds at the indicated doses and schedule for 15 days. Tumor volume was measured by caliper. Data are represented as mean ± SEM; sample size, 6 mice per group. *, P < 0.05 vs. vehicle controls; §, P < 0.05 vs. best dual combination (ANOVA on ranks and post hoc Tukey test). B, in vivo modulation of glucose and insulin. Mice from efficacy study were sacrificed after 4 hours after last dose. Blood glucose and plasma insulin levels were assessed and are represented as mean ± SEM (n = 5–6 per time point). *, P < 0.05 vs. vehicle controls; #, P < 0.05 vs. triple combination (ANOVA on ranks and post hoc Dunn or Tukey test). PI3Kβi, GSK2636771; PI3Kαi, BYL719; BRAFi, LGX818; and IGF1Ri, figitumumab-like antibody.

Figure 7.

Concomitant inhibition of PI3Kβ/PI3Kα or PI3Kβ/IGF1R maximizes the efficacy of BRAF inhibitors in PTENLOF/BRAFMUT melanoma in vivo. A, in vivo efficacy on tumor growth. Mice bearing WM-266-4 xenografts were treated with the indicated compounds at the indicated doses and schedule for 15 days. Tumor volume was measured by caliper. Data are represented as mean ± SEM; sample size, 6 mice per group. *, P < 0.05 vs. vehicle controls; §, P < 0.05 vs. best dual combination (ANOVA on ranks and post hoc Tukey test). B, in vivo modulation of glucose and insulin. Mice from efficacy study were sacrificed after 4 hours after last dose. Blood glucose and plasma insulin levels were assessed and are represented as mean ± SEM (n = 5–6 per time point). *, P < 0.05 vs. vehicle controls; #, P < 0.05 vs. triple combination (ANOVA on ranks and post hoc Dunn or Tukey test). PI3Kβi, GSK2636771; PI3Kαi, BYL719; BRAFi, LGX818; and IGF1Ri, figitumumab-like antibody.

Close modal

In summary, these data demonstrate a clear improvement of efficacy of sole BRAF inhibition in the PTENLOF/BRAFMUT melanoma background by concomitant dual PI3Kα/β blockade using either direct PI3Kα or IGF1R inhibitors and provide a strong rationale to test such combinations in humans.

Here, we have demonstrated for the first time that PTENLOF in the setting of BRAFMUT melanoma causes selective sensitivity to PI3Kβ inhibitors. However, the major limitation of PI3Kβ as a drug target in this genetic setting is the immediate rebound of PI3Kα signaling. Therefore, concomitant inhibition of PI3Kα is required to achieve sustained PI3K pathway blockade and enhance the apoptotic response upon treatment with the current BRAF/MEK inhibitor regimen.

In parallel with our findings, recent studies revealed that the contribution of individual isoforms to PI3K signaling upon PTENLOF is highly context dependent. Although PTENLOF tumors and cancer derived from prostate and breast lineage specifically depend on PI3Kβ signaling (8–10), in PTENLOF cell lines and tumors derived from the endometrial lineage, dual inhibition of PI3Kα/β is required to reduce cell viability or tumor growth, especially in a Kras-mutant background (28, 29). Similarly, both PI3Kα/β contribute to the PTEN hamartoma tumor syndrome (30), whereas T-cell acute lymphoblastic leukemia driven by PTENLOF relies on the activity of PI3Kγ/δ in line with the predominant role of these isoforms in the hematopoietic lineage (31). These observations underscore the need for the determination of isoform dependency in different indications as well as in-depth analysis of coexisting genetic alterations. Indeed, recent studies revealed a mechanism of actively induced compensation by the various PI3K isoforms (32, 33). Similarly, while we observed a primary sensitivity to PI3Kβ inhibitors in our melanoma panel and a trend toward PI3Kδ sensitivity (Fig. 1A), combination of PI3Kβi with PI3Kαi was necessary to inhibit PI3K signaling upon long-term treatment. In contrast, long-term treatment with PI3Kδi did not synergize with PI3Kβi to robustly inhibit PI3K signaling, but dual PI3Kδ/α inhibition caused mild reduction of pAkt (Supplementary Fig. S3). In contrast with our findings, a recent study using genetically engineered murine models and three cell lines distinct to ours excluded a role of the PI3Kβ isoform in PTENLOF/BRAFMUT melanoma but demonstrated the sole involvement of PI3Kδ/α instead (34). Taken together, these findings indicate that the isoforms' interplay and the corresponding tumor dependency is diverse and highly context specific. Thus, depending on the adaptive input from the upstream RTKs and G protein coupled receptors, PTEN loss will amplify the signal of different class IA PI3K isoforms. This suggests clinical strategies that would either require the combined use of isoform-specific inhibitors in specifically selected subsets of PTENLOF/BRAFMUT melanoma patients or the use of panPI3K inhibitors across the whole PTENLOF/BRAFMUT melanoma population. However, systemic PI3Kα inhibition will ultimately lead to the induction of on-target dose-limiting toxicities that are even exacerbated when combined with MEK inhibitors. Alternative scheduling regimens or targeting the mechanism of PI3Kα reactivation in the tumor tissue specifically might circumvent this problem. Our data provide evidence that in PTENLOF/BRAFMUT melanoma PI3Kβ inhibition triggers PI3Kα activation via IGF1R and that IGF1R inhibition indeed substitutes for PI3Kα inhibition to block tumor growth, providing a unique opportunity to minimize the induction of glucose abnormalities.

In contrast with several reports pointing to highly heterogeneous resistance mechanisms to inhibitors of PI3K/mTOR involving a diverse set of RTKs (35, 36), we identified exclusively and reproducibly shRNAs targeting IGF1R as sensitizers of PI3Kβ inhibition across a panel of PTENLOF/BRAFMUT melanoma lines by blocking the reactivation of PI3Kα signaling.

Activation of PI3K signaling via the IGF1R has been shown to contribute to the emergence of resistance to targeted therapies in a variety of human cancers, including BRAFMUT melanoma treated with a BRAF inhibitor (37–40). To achieve increased efficacy of therapy and prevention of resistance, it might therefore be beneficial to improve patient stratification beyond BRAFMUT status and extend the standard treatment of MAPK inhibition upfront with PI3K pathway inhibitors, especially in a setting where PTEN is already lost. Based on the results presented here, however, we speculate that in patients with BRAFMUT and concomitant PTENLOF, a combination of MAPK pathway inhibition with dual PI3Kα/β inhibitors or with panPI3K inhibitors may be more effective than sole combination of MAPK pathway with PI3Kβ inhibitors. One of the major concern with such strategies is, however, the induction of changes in glucose homeostasis due to blockade of the PI3Kα isoform in insulin-sensitive tissues (41), leading to dose-limiting hyperglycemia. Therefore, the observation that PI3Kα was exclusively activated by IGF1R in PTENLOF/BRAFMUT melanoma provided the unique opportunity to successfully replace the PI3Kα inhibition by a combination of IGF1R and PI3Kβ inhibitors, thereby minimizing on-target side effects on insulin-sensitive tissues (42).

The pharmaceutical industry has heavily pursued the development of various PI3K and MAPK pathway inhibitors with distinct modes of action over the last decade. The availability of these compounds together with the data presented in this work strongly argues for the initiation of preclinical and clinical studies to evaluate the potential long-term benefit for patients with PTENLOF/BRAFMUT melanoma treated with a combination of MAPK pathway inhibitors together with pan PI3K or PI3Kβ and IGF1R monoclonal antibodies.

W.R. Sellers is VP/Global Head of Oncology at and has ownership interest (including patents) in Novartis. F. Hofmann has ownership interest (including patents) in Novartis stocks. No potential conflicts of interest were disclosed by the other authors.

Conception and design: B. Herkert, C. Schnell, H. Voshol, W.R. Sellers, F. Hofmann, S.M. Brachmann

Development of methodology: B. Herkert, A. Kauffmann, H. Erasimus, E. Billy, D. Ruddy, D. Guthy

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Herkert, S. Mollé, C. Schnell, T. Ferrat, H. Voshol, J. Juengert, H. Erasimus, G. Marszalek, M. Kazic-Legueux, D. Ruddy, M. Stump, D. Guthy, M. Ristov, K. Calkins

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Herkert, A. Kauffmann, S. Mollé, C. Schnell, H. Voshol, M. Kazic-Legueux, E. Billy, M. Stump, S.-M. Maira, W.R. Sellers

Writing, review, and/or revision of the manuscript: B. Herkert, A. Kauffmann, C. Schnell, H. Voshol, M. Stump, K. Calkins, S.-M. Maira, W.R. Sellers, F. Hofmann, S.M. Brachmann

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D. Ruddy

Study supervision: B. Herkert, F. Hofmann, M.N. Hall, S.M. Brachmann

The authors thank Kristine Yu, Kalyani Gampa, Iris Kao, Philippe Megel, and Odile Weber for technical assistance and Christine Fritsch, Tobias Schmelzle, Michael Schlabach, and Karen Yu for discussions.

All research costs were covered by Novartis AG. No additional external funding was received for this study.

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.
Flaherty
KT
,
Infante
JR
,
Daud
A
,
Gonzalez
R
,
Kefford
RF
,
Sosman
J
, et al
Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations
.
N Engl J Med
2012
;
367
:
1694
703
.
2.
Dankort
D
,
Curley
DP
,
Cartlidge
RA
,
Nelson
B
,
Karnezis
AN
,
Damsky
WE
, et al
Braf(V600E) cooperates with Pten loss to induce metastatic melanoma
.
Nat Genet
2009
;
41
:
544
52
.
3.
Xing
F
,
Persaud
Y
,
Pratilas
CA
,
Taylor
BS
,
Janakiraman
M
,
She
Q-B
, et al
Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring V600EBRAF
.
Oncogene
2012
;
31
:
446
57
.
4.
Paraiso
KHT
,
Xiang
Y
,
Rebecca
VW
,
Abel
E V
,
Chen
YA
,
Munko
AC
, et al
PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression
.
Cancer Res
2011
;
71
:
2750
60
.
5.
Nathanson
KL
,
Martin
A-M
,
Wubbenhorst
B
,
Greshock
J
,
Letrero
R
,
D'Andrea
K
, et al
Tumor genetic analyses of patients with metastatic melanoma treated with the BRAF inhibitor dabrafenib (GSK2118436)
.
Clin Cancer Res
2013
;
19
:
4868
78
.
6.
Trunzer
K
,
Pavlick
AC
,
Schuchter
L
,
Gonzalez
R
,
McArthur
GA
,
Hutson
TE
, et al
Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma
.
J Clin Oncol
2013
;
31
:
1767
74
.
7.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
2012
;
483
:
603
7
.
8.
Wee
S
,
Wiederschain
D
,
Maira
S-M
,
Loo
A
,
Miller
C
,
deBeaumont
R
, et al
PTEN-deficient cancers depend on PIK3CB
.
Proc Natl Acad Sci U S A
2008
;
105
:
13057
62
.
9.
Ni
J
,
Liu
Q
,
Xie
S
,
Carlson
C
,
Von
T
,
Vogel
K
, et al
Functional characterization of an isoform-selective inhibitor of PI3K-p110 as a potential anticancer agent
.
Cancer Discov
2012
;
2
:
425
33
.
10.
Torbett
NE
,
Luna-Moran
A
,
Knight
ZA
,
Houk
A
,
Moasser
M
,
Weiss
W
, et al
A chemical screen in diverse breast cancer cell lines reveals genetic enhancers and suppressors of sensitivity to PI3K isoform-selective inhibition
.
Biochem J
2008
;
415
:
97
110
.
11.
Guagnano
V
,
Kauffmann
A
,
Wöhrle
S
,
Stamm
C
,
Ito
M
,
Barys
L
, et al
FGFR genetic alterations predict for sensitivity to NVP-BGJ398, a selective Pan-FGFR inhibitor
.
Cancer Discov
2012
;
2
:
1118
33
.
12.
Lehár
J
,
Krueger
AS
,
Avery
W
,
Heilbut
AM
,
Johansen
LM
,
Price
ER
, et al
Synergistic drug combinations tend to improve therapeutically relevant selectivity
.
Nat Biotechnol
2009
;
27
:
659
66
.
13.
Martiny-baron
G
,
Haasen
D
,
Dorazio
DD
,
Voshol
J
,
Fabbro
D
. 
Protein Microarrays
.
Korf
U
,
editor
.
Totowa, NJ
:
Humana Press
; 
2011
.
14.
Sausgruber
N
,
Coissieux
M-M
,
Britschgi
A
,
Wyckoff
J
,
Aceto
N
,
Leroy
C
, et al
Tyrosine phosphatase SHP2 increases cell motility in triple-negative breast cancer through the activation of SRC-family kinases
.
Oncogene
2015
;
34
:
2272
8
.
15.
König
R
,
Chiang
C
,
Tu
BP
,
Yan
SF
,
DeJesus
PD
,
Romero
A
, et al
A probability-based approach for the analysis of large-scale RNAi screens
.
Nat Methods
2007
;
4
:
847
9
.
16.
Songyang
Z
,
Shoelson
SE
,
Chaudhuri
M
,
Gish
G
,
Pawson
T
,
Haser
WG
, et al
SH2 domains recognize specific phosphopeptide sequences
.
Cell
1993
;
72
:
767
78
.
17.
Gillham
H
,
Golding
MC
,
Pepperkok
R
,
Gullick
WJ
. 
Intracellular movement of green fluorescent protein-tagged phosphatidylinositol 3-kinase in response to growth factor receptor signaling
.
J Cell Biol
1999
;
146
:
869
80
.
18.
Yu
J
,
Zhang
Y
,
McIlroy
J
,
Rordorf-Nikolic
T
,
Orr
GA
,
Backer
JM
. 
Regulation of the p85/p110 phosphatidylinositol 3′-kinase: stabilization and inhibition of the p110alpha catalytic subunit by the p85 regulatory subunit
.
Mol Cell Biol
1998
;
18
:
1379
87
.
19.
Landis
J
,
Shaw
LM
. 
Insulin receptor substrate-2 mediated phosphatidylinositol-3-kinase signaling selectively inhibits glycogen synthase kinase-3β to regulate aerobic glycolysis
.
J Biol Chem
2014
;
289
:
18603
13
.
20.
Haruta
T
,
Uno
T
,
Kawahara
J
,
Takano
A
,
Egawa
K
,
Sharma
PM
, et al
A rapamycin-sensitive pathway down-regulates insulin signaling via phosphorylation and proteasomal degradation of insulin receptor substrate-1
.
Mol Endocrinol
2000
;
14
:
783
94
.
21.
García-Martínez
JM
,
Alessi
DR
. 
mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1)
.
Biochem J
2008
;
416
:
375
85
.
22.
Dou
Z
,
Pan
JA
,
Dbouk
HA
,
Ballou
LM
,
DeLeon
JL
,
Fan
Y
, et al
Class IA PI3K p110?? subunit promotes autophagy through Rab5 small GTPase in response to growth factor limitation
.
Mol Cell
2013
;
50
:
29
42
.
23.
Chakrabarty
A
,
Sanchez
V
,
Kuba
MG
,
Rinehart
C
,
Arteaga
CL
. 
Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors
.
Proc Natl Acad Sci
2012
;
109
:
2718
23
.
24.
Ma
L
,
Chen
Z
,
Erdjument-Bromage
H
,
Tempst
P
,
Pandolfi
PP
. 
Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosis and cancer pathogenesis
.
Cell
2005
;
121
:
179
93
.
25.
Magnuson
B
,
Ekim
B
,
Fingar
DC
. 
Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks
.
Biochem J
2012
;
441
:
1
21
.
26.
Roux
PP
,
Shahbazian
D
,
Vu
H
,
Holz
MK
,
Cohen
MS
,
Taunton
J
, et al
RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation
.
J Biol Chem
2007
;
282
:
14056
64
.
27.
Schnell
CR
,
Barbe
S
,
Caravatti
G
,
Brachmann
S
,
Guthy
D
,
Maira
SM
, et al
NVP-BYL719, a selective inhibitor of the class Ia PI3K isoform alpha impairs angiogenesis and microvascular permeability
.
Cancer Res
2012
;
72
:
Suppl; abstr 3743
.
28.
Weigelt
B
,
Warne
PH
,
Lambros
MB
,
Reis-Filho
JS
,
Downward
J
. 
PI3K pathway dependencies in endometrioid endometrial cancer cell lines
.
Clin Cancer Res
2013
;
19
:
3533
44
.
29.
Schmit
F
,
Utermark
T
,
Zhang
S
,
Wang
Q
,
Von
T
,
Roberts
TM
, et al
PI3K isoform dependence of PTEN-deficient tumors can be altered by the genetic context
.
Proc Natl Acad Sci U S A
2014
;
111
:
6395
400
.
30.
Wang
Q
,
Von
T
,
Bronson
R
,
Ruan
M
,
Mu
W
,
Huang
A
, et al
Spatially distinct roles of class Ia PI3K isoforms in the development and maintenance of PTEN hamartoma tumor syndrome
.
Genes Dev
2013
;
27
:
1568
80
.
31.
Subramaniam
PS
,
Whye
DW
,
Efimenko
E
,
Chen
J
,
Tosello
V
,
De Keersmaecker
K
, et al
Targeting nonclassical oncogenes for therapy in T-ALL
.
Cancer Cell
2012
;
21
:
459
72
.
32.
Costa
C
,
Ebi
H
,
Martini
M
,
Beausoleil
SA
,
Faber
AC
,
Jakubik
CT
, et al
Measurement of PIP3 levels reveals an unexpected role for p110β in early adaptive responses to p110α-specific inhibitors in luminal breast cancer
.
Cancer Cell
2015
;
27
:
97
108
.
33.
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
.
34.
Deuker
M
,
Marsh Durban
V
,
Phillips
W
,
McMahon
M
. 
PI3′-kinase inhibition forestalls the onset of MEK1/2 inhibitor resistance in BRAF-mutated melanoma
.
Cancer Discov
2015
;
5
:
143
53
.
35.
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
.
36.
Liu
P
,
Cheng
H
,
Santiago
S
,
Raeder
M
,
Zhang
F
,
Isabella
A
, et al
Oncogenic PIK3CA-driven mammary tumors frequently recur via PI3K pathway-dependent and PI3K pathway-independent mechanisms
.
Nat Med
2011
;
17
:
1116
20
.
37.
Shi
Y
,
Yan
H
,
Frost
P
,
Gera
J
,
Lichtenstein
A
. 
Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade
.
Mol Cancer Ther
2005
;
4
:
1533
40
.
38.
Jones
HE
,
Goddard
L
,
Gee
JMW
,
Hiscox
S
,
Rubini
M
,
Barrow
D
, et al
Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells
.
Endocr Relat Cancer
2004
;
11
:
793
814
.
39.
Tamburini
J
,
Chapuis
N
,
Bardet
V
,
Park
S
,
Sujobert
P
,
Willems
L
, et al
Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: Rationale for therapeutic inhibition of both pathways
.
Blood
2008
;
111
:
379
82
.
40.
Villanueva
J
,
Vultur
A
,
Lee
JT
,
Somasundaram
R
,
Fukunaga-Kalabis
M
,
Cipolla
AK
, et al
Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K
.
Cancer Cell
2010
;
18
:
683
95
.
41.
Bendell
JC
,
Rodon
J
,
Burris
HA
,
De Jonge
M
,
Verweij
J
,
Birle
D
, et al
Phase I, dose-escalation study of BKM120, an oral pan-class I PI3K inhibitor, in patients with advanced solid tumors
.
J Clin Oncol
2012
;
30
:
282
90
.
42.
Jia
S
,
Roberts
TM
,
Zhao
JJ
. 
Should individual PI3 kinase isoforms be targeted in cancer?
Curr Opin Cell Biol
2009
;
21
:
199
208
.

Supplementary data