New treatments for triple-negative breast cancer (TNBC) are urgently needed. Despite there being little evidence of clinical activity as single-agent therapies, we show that dual blockade of PI3Kα and CDK4/6 is synergistically effective against multiple RB1-wild-type TNBC models. Combined PI3Kα and CDK4/6 inhibition significantly increased apoptosis, cell-cycle arrest, and tumor immunogenicity and generated immunogenic cell death in human TNBC cell lines. Combination treatment also significantly improved disease control in human xenograft models compared with either monotherapy. Combined PI3Kα and CDK4/6 inhibition significantly increased tumor-infiltrating T-cell activation and cytotoxicity and decreased the frequency of immunosuppressive myeloid-derived suppressor cells in a syngeneic TNBC mouse model. Notably, combined PI3Kα and CDK4/6 inhibition, along with inhibition of immune checkpoints PD-1 and CTLA-4, induced complete and durable regressions (>1 year) of established TNBC tumors in vivo. Overall, our results illustrate convergent mechanisms of PI3Kα and CDK4/6 blockade on cell-cycle progression, DNA damage response, and immune-modulation and may provide a novel therapeutic approach for TNBC. Cancer Res; 77(22); 6340–52. ©2017 AACR.

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is clinically characterized by its lack of expression of estrogen- and progesterone receptors (ER/PR) and HER2 (1). TNBC has the worst outcome of all breast cancer subtypes (1). Cytotoxic chemotherapy is the mainstay of current treatment for TNBC patients. However, although these regimens can prove effective for a subgroup of TNBC patients, in general, once relapsed, remissions are brief and are frequently followed by rapid disease progression and death. In contrast, the existence of a tumor immune infiltrate has been reported to be a robust prognostic factor in TNBC (2). However, the causal mechanisms underlying this immune response are unclear. It has been proposed that DNA damage with resultant activation of stimulator of interferon genes (STING) and/or mutational load and neoantigen production may be responsible (3, 4). Regardless, this immune response seems not to be sufficient to induce primary tumor clearance in humans.

Cell-cycle control is frequently dysregulated in breast cancer (5). The transition from G1 to S phase of the cell cycle is controlled by interactions between cyclin-dependent kinases 4 and 6 (CDK4/6), cyclin D1, and retinoblastoma protein (RB). CDK4/6 inhibition has been shown to be an effective therapeutic strategy for ER-positive breast cancers in clinical trials (6–10). However, TNBC is a molecularly heterogeneous disease characterized by genomic instability along with high expression of cell-cycle genes including cyclin E1 (11) and has shown resistance to single-agent CDK4/6 inhibition (6, 12).

The complex biology of TNBC suggests that combination treatments will be required to achieve effective and durable disease control. Combined treatment with PI3K and CDK4/6 inhibitors has been shown to be effective in mitigating early adaption responses to single-agent PI3K and CDK4/6 inhibitors for overcoming single-agent inhibitor resistance in ER-positive breast cancers (13, 14). We sought to determine whether combined PI3K and CDK4/6 inhibition would be a similarly useful strategy for TNBC. We also hypothesized that induction of cell death could promote an immune response, given that TNBC may be amendable to immune approaches (2, 15–17).

Here, we show that synergistic interactions of PI3Kα and CDK4/6 inhibitors resulted in more effective disease control of TNBC both in vitro and in vivo. Combination treatment resulted in increased cell-cycle arrest, apoptosis, calreticulin cell-surface expression, and tumor immunogenicity. Finally, we show that combination of PI3Kα and CDK4/6 inhibitors with immune-checkpoint blockade resulted in long-lasting tumor regression in a syngeneic immunocompetent mouse model of TNBC.

Cell lines and culture

All culture media were supplemented with 10% heat-inactivated FBS. HCC70, HCC1806, and MDA-MB-468 were kindly provided by Professor Roger Daly from Monash University, Australia. All other human cell lines were obtained from the ATCC. The human cell lines have been authenticated by short tandem repeat analysis and were maintained in DMEM or RPMI1640 (Gibco). AT3OVA (18) mouse TNBC cell line was maintained in SAFC DMEM. We have previously characterized AT3OVA and found that it was wild-type for Rb1, Pik3ca, and Trp53 (19). All human and mouse cell lines have been verified to be negative for mycoplasma contamination and were maintained at 37°C in a 5% CO2 incubator. Characteristics of the human cell lines are further detailed in Supplementary Table S1.

Drug compounds

BYL719 (Alpelisib) and LEE011 (Ribociclib) were kindly supplied by Novartis. PD991 (Palbociclib; PD-0332991) was purchased from SelleckChem. All drugs for in vitro use were reconstituted in DMSO at 10 mmol/L. BYL719, LEE011, and PD991 were reconstituted in 0.5% methylcellulose (Sigma Aldrich) for in vivo experiments. Immune-checkpoint antibodies and the isotype control used in in vivo experiments were obtained from BioXCell: anti-mouse PD-1 mAb (RMP1-14), anti-mouse CTLA-4 mAb (9H10), and Rat IgG2a isotype control mAb (2A3). Doses used are detailed in figure legends.

Combination treatment synergy quantitation

Drug combination studies were performed according to the Chou–Talalay method of synergy quantitation (20). MDA-MB-468, HCC1143, HCC70, HCC1806, MDA-MB-231, HS578T, and MDA-MB-453 cells were treated in vitro for 72 hours with the combination of BYL719 and LEE011 over a range of concentrations held at a fixed ratio based on the GI50 (drug concentration required for 50% cell growth inhibition) of each drug specific for each cell line. CalcuSyn 2.0 (Biosoft) performs multiple drug dose-effect calculations using the Median Effects methods described by Chou and Talalay (21) and was used to determine the combination index (CI), which offers quantitative definition for additive effect (CI = 1), synergism (CI < 1), and antagonism (CI > 1) of drug combinations.

Flow cytometry analysis

Cells were plated in 24-well plates and treated the following day with the indicated agents. Cells and tissues were lysed and processed as described in Supplementary Methods. FACS analysis was performed on an LSR II flow cytometer (BD Biosciences).

Western blot analysis

Cell lines were plated in 6-well plates and treated the following day as indicated. Cells and tissues were lysed and processed as described in Supplementary Methods. Signal intensities for each protein of interest were quantitated using Image J (https://imagej.net) and presented in Supplementary Fig. S1.

3′RNA sequencing

MDA-MB-231 cells were plated in 6-well plates and treated the following day as indicated. Two replicates were included. After 24 hours, total cell RNA was extracted using a PureLink RNA Mini kit (ThermoFisher Scientific) following the manufacturer's instructions. The quantity and integrity of the total RNA were determined using TapeStation 2200 system (Agilent Technologies) and Qubit RNA High Sensitivity assay kit (ThermoFisher Scientific). Note that 500 ng total RNA was used for library preparation according to the manufacturer's instructions (QuantSeq 3′ mRNA-Seq FWD; Lexogen). Indexed libraries were pooled and sequenced on a NextSeq500 (Illumina). Briefly, the library was generated with an oligo-dT containing the Illumina Read2 linker and a random forward primer containing the Illumina Read1 linker. The library was then amplified with PCR primers containing sample indices and the Illumina clustering sequences. Five to 15 million single-end 75 bp reads were generated per sample.

Gene set enrichment analysis analysis

Gene set enrichment analysis (GSEA) was performed using the GSEA tool (22) on the entire normalized RNA expression count matrix without limiting the input to only differentially expressed genes. The Hallmark collection of 50 predefined gene sets and C5 Gene Ontology (GO) collection of 5,917 predefined gene sets from the Molecular Signatures Database (MSigDB, Broad Institute) were used for analysis. The gene sets (3,576 C5 and 49 Hallmark gene sets) included in the analysis were limited to those that contained between 15 and 500 genes. Permutation (by gene sets) was conducted 1,000 times according to default weighted enrichment statistics and using difference of class metrics to calculate and rank genes according to their differential expression levels between two treatment groups. Significant gene sets were those defined with a nominal P < 0.05. Calculation of the false discovery rate (FDR) was used to correct for multiple comparisons and gene set sizes. Significantly enriched gene sets with FDR < 0.25 were selected for hypothesis generation.

In vivo studies

Six- to 8-week-old female Nod SCID γ (NSG) mice were used for MDA-MB-231, HCC1806, and patient-derived xenograft models. For MDA-MB-231 and HCC1806 xenografts, 2 × 106 cells (suspended in PBS:Matrigel, 1:1) were injected into the fourth mammary fat pad of the NSG mice. For experiments with the patient-derived xenograft (PDX) model 12006, a single tumor from a previously established NSG mouse (passage 5) was excised and small fragments were implanted directly into the fourth mammary fat pads of 6- to 8-week-old female NSG mice. AT3OVA (5 × 105) cells were suspended in PBS and injected into the fourth mammary fat pads of 6- to 8-week-old female immune-competent C57BL/6 mice.

For all models, tumor volume (length × width2 × 0.5) was assessed by caliper measurements every 3 to 4 days. Once tumors reached an average of 100 to 150 mm3 (human xenograft models) or 50 to 100 mm3 (mouse syngeneic model), mice were randomized to commence drug treatment (day 1). BYL719, LEE011, and PD991 were administered via oral gavage. BYL719 was given daily throughout the duration of the experiment. LEE011 or PD991 were given daily on days 1 to 21 of each 28-day cycle (3 weeks on, 1 week off). Anti–PD-1 and/or anti–CTLA-4 were administered via intraperitoneal injections on days 1, 5, 9, and 13 only. For 7-day immune response analysis, BYL719 and LEE011 were administered daily with no breaks with dosing. For these experiments, anti–PD-1 and/or anti–CTLA-4 were only administered on days 1 and 5.

Mice were euthanized if the tumors reached ethical limit of 1,400 mm3 or if the animals displayed health indicators that met the institutional criteria for sacrifice. All animal experiments were approved by the Peter MacCallum Cancer Centre Animal Experimentation Ethics Committee (E539 and E556) and conducted in accordance with the National Health and Medical Research Council Australian Code of Practice for the Care and Use of Animals for Scientific Purposes.

Statistical analysis

One-way ANOVA with Tukey multiple comparison test was used to compare between treatment groups. Tumor growth curves were analyzed using two-way repeated measures ANOVA with Tukey multiple comparison test. Survival differences between treatment groups were determined using the Kaplan–Meier log rank analysis. Statistical analyses (not including differential gene expression or GSEA analyses) were performed using Prism 7 (GraphPad). A two-tailed P < 0.05 was considered statistically significant. All data are expressed as mean ± SEM.

Combined PI3Kα and CDK4/6 inhibition is synergistic against RB1-wild-type TNBC cell lines in vitro

We quantitated the level of synergy between BYL719 (PI3Kα inhibitor) and LEE011 (CDK4/6 inhibitor) in seven human TNBC cell lines including six RB1-wild-type and one RB1-mutant cell line. Of the six RB1-wild-type cell lines, three were classified as basal-like, two mesenchymal-like, and one was of the luminal androgen receptor (LAR) subtype (23). We demonstrated that the interaction between BYL719 and LEE011 was synergistic in all six RB1-wild-type TNBC cell lines but was antagonistic in the RB1-mutant cell line, MDA-MB-468 (Fig. 1A).

Figure 1.

Combined PI3Kα and CDK4/6 inhibitor treatment is synergistic in TNBC cell lines. A, Synergy quantification as indicated by CI values of BYL719 and LEE011 treatment in TNBC cell lines. Gray bar, CI values indicating additive effect. *, MDA-MB-468 carries RB1 mutation; BL, basal-like subtype; MSL, mesenchymal-like subtype; LAR, luminal androgen receptor subtype. Data depicted are the mean ± SEM. B, Percent cell growth of TNBC cells relative to vehicle treatment treated with media containing vehicle, BYL719, LEE011, or the combination at their GI50 doses for 72 hours in vitro. Data depicted are the mean ± SEM. C, Representative Western blots depicting changes in protein expression in HCC1806 and MDA-MB-231 cells following 72-hour treatment with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination. Lysates were made and probed with the indicated antibodies. D, Tumor growth of HCC1806, MDA-MB-231, and PDX12006 xenografts in vivo. HCC1806 and PDX12006 xenografts were treated with vehicle, BYL719 (15 mg/kg), LEE011 (60 mg/kg), or the combination. MDA-MB-231 xenografts were treated with vehicle, BYL719 (25 mg/kg), PD991 (120 mg/kg), or the combination. N = 8 mice per treatment group. Data depicted are the mean fold change in tumor volume ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA (A and B), or two-way ANOVA (D).

Figure 1.

Combined PI3Kα and CDK4/6 inhibitor treatment is synergistic in TNBC cell lines. A, Synergy quantification as indicated by CI values of BYL719 and LEE011 treatment in TNBC cell lines. Gray bar, CI values indicating additive effect. *, MDA-MB-468 carries RB1 mutation; BL, basal-like subtype; MSL, mesenchymal-like subtype; LAR, luminal androgen receptor subtype. Data depicted are the mean ± SEM. B, Percent cell growth of TNBC cells relative to vehicle treatment treated with media containing vehicle, BYL719, LEE011, or the combination at their GI50 doses for 72 hours in vitro. Data depicted are the mean ± SEM. C, Representative Western blots depicting changes in protein expression in HCC1806 and MDA-MB-231 cells following 72-hour treatment with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination. Lysates were made and probed with the indicated antibodies. D, Tumor growth of HCC1806, MDA-MB-231, and PDX12006 xenografts in vivo. HCC1806 and PDX12006 xenografts were treated with vehicle, BYL719 (15 mg/kg), LEE011 (60 mg/kg), or the combination. MDA-MB-231 xenografts were treated with vehicle, BYL719 (25 mg/kg), PD991 (120 mg/kg), or the combination. N = 8 mice per treatment group. Data depicted are the mean fold change in tumor volume ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA (A and B), or two-way ANOVA (D).

Close modal

As expected from the above results, combination treatment with BYL719 and LEE011 significantly inhibited cell growth compared with single-agent and vehicle treatments in the synergistic lines (Fig. 1B). Consistent with this, we show in HCC1806 and MDA-MB-231 cell lines that BYL719 and LEE011 single-agent treatments resulted in reduction of AKT and RB phosphorylation, respectively, compared with vehicle treatment, and both remained suppressed with combination treatment (Fig. 1C).

Combination treatment is efficacious in TNBC xenografts in vivo

The efficacy of combined PI3Kα and CDK4/6 inhibition was next examined in vivo. HCC1806 and MDA-MB-231 xenograft tumor models were treated with BYL719, CDK4/6 inhibitor (LEE011 or PD991), the combination, or vehicle control. In both models, strong synergism was observed with significant reduction of tumor growth of mice treated with combination therapy compared with single-agent and vehicle treatments (Fig. 1D).

We next treated a patient-derived TNBC xenograft (PDX12006) with the combination of BYL719 and LEE011. This tumor sample had oncogenic TP53 p.S241C (COSM10709) and ARID1A p.R2232W (COSM3724474) mutations and was classified as mesenchymal subtype (P < 0.001). Similar to what we observed in HCC1806 and MDA-MB-231 xenograft models, combination treatment in the PDX model exhibited significant tumor growth inhibition (Fig. 1D).

BYL719 and LEE011 treatment significantly increases apoptosis

We next examined combined BYL719 and LEE011 treatment on cell cycle and apoptosis in vitro. Combined treatment significantly increased the effect on G1 cell-cycle arrest compared with single-agent BYL719 and vehicle treatments in both basal-like and mesenchymal-like TNBC cell lines (Fig. 2A). In HCC70 and MDA-MB-231 lines, G1 cell-cycle arrest was significantly greater following combination treatment compared with BYL719 and LEE011 single-agent treatments as well as vehicle treatment.

Figure 2.

Combined PI3Kα and CDK4/6 inhibitor treatment induces cell-cycle arrest and pronounced apoptosis in TNBC cell lines. A, Cell-cycle analysis in TNBC lines treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination for 72 hours. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA comparing percentage of cells in G0–G1 between treatment groups. B, TNBC cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination for 72 hours and analyzed using flow cytometry for Annexin V–positive and propidium iodide (PI)–negative cells. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA. C, Representative Western blots depicting changes in protein expression in HCC1806 and MDA-MB-231 cells following 72-hour treatment with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination. Lysates were made and probed with the indicated antibodies. Quantitation of relative signal intensities of protein expression presented in Supplementary Fig. S1.

Figure 2.

Combined PI3Kα and CDK4/6 inhibitor treatment induces cell-cycle arrest and pronounced apoptosis in TNBC cell lines. A, Cell-cycle analysis in TNBC lines treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination for 72 hours. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA comparing percentage of cells in G0–G1 between treatment groups. B, TNBC cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination for 72 hours and analyzed using flow cytometry for Annexin V–positive and propidium iodide (PI)–negative cells. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA. C, Representative Western blots depicting changes in protein expression in HCC1806 and MDA-MB-231 cells following 72-hour treatment with vehicle, 1 μmol/L of BYL719, 1 μmol/L of LEE011, or the combination. Lysates were made and probed with the indicated antibodies. Quantitation of relative signal intensities of protein expression presented in Supplementary Fig. S1.

Close modal

Notably, combined treatment with BYL719 and LEE011 significantly increased apoptosis as indicated by an increase in percentage of Annexin V+/propidium iodide cells compared with single-agent and vehicle treatments (Fig. 2B).

We next analyzed protein components of the PI3K and CDK4/6 pathways by Western blot analysis in HCC1806 and MDA-MB-231 cells treated with vehicle, BYL719, LEE011, or the combination for 72 hours in vitro (Fig. 2C). Treatment with LEE011 was associated with increased expression of cyclin D1, consistent with previous reports (13, 14). Phosphorylation levels of eukaryotic translation initiation factor 4E-binding protein-1 (4EBP1 Thr37/40), known to be a direct substrate of mTOR and one of the regulators of cyclin D1 expression (24, 25), were maintained across treatment groups in MDA-MB-231 and only slightly decreased with BYL719 and LEE011 combined treatment in HCC1806 compared with vehicle treatment, suggesting incomplete inhibition of mTOR activity. Nonetheless, we observed that combination treatment removed AKT inhibition on GSK3β and p21Cip1/WAF1 (p21) tumor suppressors. p-GSK3β Ser9 and p-p21 Thr145 expression levels were suppressed with combined BYL719 and LEE011 treatment compared with single-agent and vehicle treatments in MDA-MB-231 cells, whereas in HCC1806 cells, suppression was observed with single-agent BYL719 and combination therapy compared with vehicle treatment.

Next, we assessed the CDK2/cyclin E complex, which associates with CDK2 and initiates CDK2 activation shortly before entry into S phase (26). We demonstrate that p-CDK2 Thr160 levels were reduced with LEE011 treatment and further decreased with combination treatment in both cell lines. Furthermore, cyclin E2 levels were markedly decreased with LEE011 and combination treatments compared with vehicle control; however, this was also accompanied by an increase in cyclin E1 expression with LEE011 and combination treatments.

These results suggest that decreased cell survival and proliferation due to combined PI3Kα and CDK4/6 blockade is likely mediated through multiple cell-cycle–related pathways in TNBC.

Synergistic effects of combination treatment on cell division processes and immune responses

To further understand the mechanism by which BYL719 and LEE011 were effective in combination in an unbiased manner, we performed next-generation RNA sequencing and GSEA using MDA-MB-231 cells treated with vehicle BYL719, LEE011, or the combination. BYL719 and LEE011 single-agent and the combination treatment groups were each compared with vehicle treatment.

Not surprisingly, we observed that the “E2F targets” hallmark gene set was the top enriched (P < 0.05) downregulated gene set across all comparisons (Fig. 3A). Compared with single-agent and vehicle treatment groups, the combined therapy was associated with significant downregulation of E2F target genes involved in cell-cycle progression, consistent with our results on cell-cycle inhibition above.

Figure 3.

Combined BYL719 and LEE011 treatment downregulates cell-cycle processes and induces DNA damage in TNBC. A–C, MDA-MB-231 cells were treated in vitro for 24 hours with vehicle, 1 μmol/L BYL719, 1 μmol/L LEE011, or the combination, after which, total RNA was extracted and analyzed via 3'RNAseq. A, GSEA enrichment plot and heatmap of differentially expressed genes in the Hallmark E2F targets gene set. V, Vehicle; B, BYL719; L, LEE011; BL, BYL719 and LEE011. B, Top five most enriched GSEA GO processes with the largest absolute normalized enrichment score (NES) with smallest FDR P value among significantly enriched gene sets with FDR < 0.25. ES, enrichment score; NP, nominal P value. C, Heatmap of core-enriched genes in top enriched downregulated Hallmark and GO process cell-cycle–related gene sets. Genes that were significantly differentially expressed (P < 0.05) in combination treatment compared with vehicle treatment are shown. Gene names that are highlighted in yellow encode proteins involved with DNA damage and repair processes. D, Combined BYL719 and LEE011 treatment induces DNA damage. HCC1806 and MDA-MB-231 cell lines were treated with 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry. Phosphorylated γH2AX Ser139-positive cells were quantitated as a percentage of live cells. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; and ****, P < 0.0001 by one-way ANOVA.

Figure 3.

Combined BYL719 and LEE011 treatment downregulates cell-cycle processes and induces DNA damage in TNBC. A–C, MDA-MB-231 cells were treated in vitro for 24 hours with vehicle, 1 μmol/L BYL719, 1 μmol/L LEE011, or the combination, after which, total RNA was extracted and analyzed via 3'RNAseq. A, GSEA enrichment plot and heatmap of differentially expressed genes in the Hallmark E2F targets gene set. V, Vehicle; B, BYL719; L, LEE011; BL, BYL719 and LEE011. B, Top five most enriched GSEA GO processes with the largest absolute normalized enrichment score (NES) with smallest FDR P value among significantly enriched gene sets with FDR < 0.25. ES, enrichment score; NP, nominal P value. C, Heatmap of core-enriched genes in top enriched downregulated Hallmark and GO process cell-cycle–related gene sets. Genes that were significantly differentially expressed (P < 0.05) in combination treatment compared with vehicle treatment are shown. Gene names that are highlighted in yellow encode proteins involved with DNA damage and repair processes. D, Combined BYL719 and LEE011 treatment induces DNA damage. HCC1806 and MDA-MB-231 cell lines were treated with 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry. Phosphorylated γH2AX Ser139-positive cells were quantitated as a percentage of live cells. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; and ****, P < 0.0001 by one-way ANOVA.

Close modal

The top enriched (P < 0.05) downregulated GO processes for each comparison were all related to cell division processes, with particular enrichment for downregulation of genes in chromosome/chromatid segregation processes as shown in Fig. 3B.

A heatmap of the “core-enriched” genes in the top enriched (P < 0.05) hallmark and GO process gene sets shows striking downregulation of these genes (log fold change, –0.39 to –3.74; median log fold change = –1.66) with the combination treatment compared with vehicle treatment (Fig. 3C). Interestingly, we noted that genes involved in DNA damage and repair processes (highlighted in yellow in Fig. 3C) made up a sizeable proportion among those downregulated.

Given these findings, we next investigated if there was objective evidence of DNA damage. Validating our observations above, we found that both LEE011 single-agent and combination treatments were associated with increased levels of γH2AX phosphorylation on Ser139 (γH2AX, a marker for DNA damage; Fig. 3D).

Combination BYL719 and LEE011 treatment increases tumor immunogenicity in vitro

In our GSEA analysis, we observed that immune-related pathways were also among the top enriched (P < 0.05) upregulated hallmarks. These included upregulation of IFNα, IFNγ, TNFα, and innate immune (complement) responses (Fig. 4A and B). We also observed significant upregulation of genes (log fold change, 0.24 to 1.51; median log fold change = 0.55) involved in antigen presentation that included HLAs HLA-A (human major histocompatibility complex class I; MHCI), HLA-DMA (human MHCII), CTSD, ICAM, RELB, PSME1, and TAPBP, in response to combination therapy compared with vehicle treatment (highlighted in yellow in Fig. 4B).

Figure 4.

Combined BYL719 and LEE011 treatment upregulates immune response gene expression and increases tumor immunogenicity in TNBC. A and B, MDA-MB-231 cells were treated in vitro for 24 hours with vehicle, 1 μmol/L BYL719, 1 μmol/L LEE011, or the combination, after which, total RNA was extracted and analyzed via 3'RNAseq. A, Top five upregulated enriched GSEA Hallmarks with highest positive normalized enrichment score (NES) with smallest FDR P value among significantly enriched gene sets with FDR < 0.25. ES, enrichment score; NP, nominal P value. B, Heatmap of core-enriched genes in the top enriched upregulated Hallmark immune-related gene sets. Genes that were significantly differentially expressed (P < 0.05) in combination compared with vehicle treatments are shown. Gene names that are highlighted in yellow encode proteins involved with antigen presentation. C, HCC1806 and MDA-MB-231 cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry for tumor cell-surface expression of human leukocyte antigens (HLA-ABC and HLA-DR), CD86, and PD-L1. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA. D, HCC1806 and MDA-MB-231 cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry for cell-surface calreticulin (CRT) expression. Data depicted are the mean ± SEM. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA.

Figure 4.

Combined BYL719 and LEE011 treatment upregulates immune response gene expression and increases tumor immunogenicity in TNBC. A and B, MDA-MB-231 cells were treated in vitro for 24 hours with vehicle, 1 μmol/L BYL719, 1 μmol/L LEE011, or the combination, after which, total RNA was extracted and analyzed via 3'RNAseq. A, Top five upregulated enriched GSEA Hallmarks with highest positive normalized enrichment score (NES) with smallest FDR P value among significantly enriched gene sets with FDR < 0.25. ES, enrichment score; NP, nominal P value. B, Heatmap of core-enriched genes in the top enriched upregulated Hallmark immune-related gene sets. Genes that were significantly differentially expressed (P < 0.05) in combination compared with vehicle treatments are shown. Gene names that are highlighted in yellow encode proteins involved with antigen presentation. C, HCC1806 and MDA-MB-231 cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry for tumor cell-surface expression of human leukocyte antigens (HLA-ABC and HLA-DR), CD86, and PD-L1. Data depicted are the mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by one-way ANOVA. D, HCC1806 and MDA-MB-231 cell lines were treated with vehicle, 1 μmol/L of BYL719, 1 μmol/L LEE011, or the combination for 72 hours and analyzed using flow cytometry for cell-surface calreticulin (CRT) expression. Data depicted are the mean ± SEM. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA.

Close modal

We next investigated whether combined BYL719 and LEE011 treatment could modulate tumor immunogenicity given the transcriptional findings above. We examined tumor cell-surface expression of HLA antigens as well as the immune-checkpoint ligands, programmed death ligand 1 (PD-L1), and CD86 in HCC1806 and MDA-MB-231 cell lines (Fig. 4C). We found that LEE011 single-agent as well as the combination treatments significantly increased expression of HLA antigens (expression was further increased in the presence of IFNγ), as well as CD86, the ligand for immune-checkpoint cytotoxic T-lymphocyte–associated protein 4 (CTLA-4), in both cell lines. BYL719 seemed to have an inhibitory effect on the expression of PD-L1, consistent with the literature that this ligand is regulated through the PI3K/AKT signaling pathway (27–29).

Given the above results, along with observations of increased DNA damage and tumor immunogenicity, we next assessed whether combined therapy could induce an increase in “immunogenic” cell death. We examined levels of tumor cell-surface calreticulin expression, given that this protein, usually present on the endoplasmic reticulum, is translocated to the plasma membrane surface of cells undergoing immunogenic cell death in the early, preapoptotic phase, facilitating engulfment of dying cells by dendritic cells (30). Consistent with our hypothesis, we observed pronounced increased expression of cell-surface calreticulin following combination therapy on both HCC1806 and MDA-MB-231 cells compared with single-agent and vehicle treatments (Fig. 4D).

Collectively, these findings suggest that the combined BYL719 and LEE011 treatment enhanced immunogenic cell death and could promote tumor immunogenicity. These findings provided the rationale to evaluate whether combination therapy could enhance immune responses in vivo.

Combined BYL719 and LEE011 inhibition increases T-cell activation and reduces immunosuppressive cell populations in vivo

AT3OVA is a well-characterized syngeneic mouse model of TNBC and was used to examine whether immune responses were important for the therapeutic effect following combined PI3Kα and CDK4/6 inhibition in vivo. AT3OVA tumor–bearing immune-competent mice were treated with BYL719, LEE011, the combination, or vehicle control. We observed that combined treatment exerted greater control of AT3OVA tumor growth compared with single-agent or vehicle treatments (Fig. 5A).

Figure 5.

Combined PI3Kα and CDK4/6 inhibition increases tumor-infiltrating CD8+ and CD4+ T-cell activation and cytotoxic potential as well as decreases frequency of MDSCs in AT3OVA tumors in mice. A, Tumor growth of AT3OVA. Tumor-bearing mice were treated with vehicle, BYL719 (10 mg/kg), LEE011 (40 mg/kg), or the combination. N = 8 mice per treatment group. Data depicted are the mean tumor volume ± SEM. B–D, AT3OVA tumor–bearing mice were treated with vehicle, BYL719 (10 mg/kg), LEE011 (40 mg/kg), or the combination for 7 days. Tumors were then harvested, and the tumor infiltrating CD8+ T cells (B), CD4+ T cells (C), and mMDSCs (D) were analyzed using flow cytometry. N = 5 to 11 mice per treatment group. Data depicted are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by two-way ANOVA (A) or one-way ANOVA (B–D).

Figure 5.

Combined PI3Kα and CDK4/6 inhibition increases tumor-infiltrating CD8+ and CD4+ T-cell activation and cytotoxic potential as well as decreases frequency of MDSCs in AT3OVA tumors in mice. A, Tumor growth of AT3OVA. Tumor-bearing mice were treated with vehicle, BYL719 (10 mg/kg), LEE011 (40 mg/kg), or the combination. N = 8 mice per treatment group. Data depicted are the mean tumor volume ± SEM. B–D, AT3OVA tumor–bearing mice were treated with vehicle, BYL719 (10 mg/kg), LEE011 (40 mg/kg), or the combination for 7 days. Tumors were then harvested, and the tumor infiltrating CD8+ T cells (B), CD4+ T cells (C), and mMDSCs (D) were analyzed using flow cytometry. N = 5 to 11 mice per treatment group. Data depicted are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by two-way ANOVA (A) or one-way ANOVA (B–D).

Close modal

We next characterized the effect of treatment on immune cell populations in the tumor microenvironment by flow cytometry. Compared with single-agent and vehicle treatment cohorts, the combined treatment markedly enhanced infiltrating CD8+ and CD4+ T-cell activity as indicated by an increase in CD69, granzyme B, and IFNγ expression (Fig. 5B and C). Further evidence of enhanced T-cell activation following combined therapy was indicated by a significant increase in coexpression of programmed cell death protein 1 (PD-1) and CTLA-4 immune-checkpoint proteins in CD8+ and CD4+ T cells compared with single-agent and vehicle treatment groups (Fig. 5B and C). Single-agent or combination treatments were not observed to affect proliferation or frequency of tumor-infiltrating CD8+ and CD4+ T cells (Supplementary Fig. S2A and S2B).

In addition to activating adaptive immune cells, combined BYL719 and LEE011 treatment significantly increased the frequency of tumor-infiltrating natural killer T (NKT) cells, and there was a trend for increased mature NK cells (NK1.1+/CD11bhigh/CD27low) compared with single-agent LEE011 and vehicle treatments (Supplementary Fig. S2C). Granzyme B expression by mature NK cells was also observed to be significantly greater with combination treatment compared with BYL719 and LEE011 single-agent and vehicle treatment groups, indicating an increase in their cytotoxic potential (Supplementary Fig. S2C).

Interestingly, combined BYL719 and LEE011 treatment was effective in significantly reducing the frequency of immunosuppressive monocytic myeloid-derived suppressor cells (mMDSC; Fig. 5D). We also observed a significant decrease in proliferation of CD4+ FOXP3+ regulatory T cells (Treg) following combination therapy as indicated by the significant decrease in expression of the Ki67 proliferative marker (Supplementary Fig. S2B).

In summary, combined PI3Kα and CDK4/6 inhibition resulted in enhanced innate and adaptive antitumor immune responses. The increase in expression of both PD-1 and CTLA-4 on T cells suggested that the addition of immune-checkpoint blockade could further augment the antitumor therapeutic effect of PI3Kα and CDK4/6 inhibition.

Immune-checkpoint blockade in combination with PI3Kα and CDK4/6 inhibition results in long-lasting tumor regression in vivo

We next examined the efficacy of combining BYL719 and LEE011 with immune-checkpoint blockade in the AT3OVA model. We tested anti–PD-1 and anti–CTLA-4 mAbs given that these immune checkpoints were found to be upregulated on T cells following PI3Kα and CDK4/6 inhibition and that they are currently being evaluated in clinical trials. Anti–PD-1 and anti–CTLA-4 treatments were administered on day 1 along with BYL719 and LEE011 treatments in tumor-bearing mice. All treatments ceased by day 50.

We observed that tumors treated with anti–PD-1 and anti–CTLA-4 as single agents performed worse in terms of tumor growth control and survival compared with BYL719 and LEE011 combination treatment (Fig. 6A). However, we found that the quadruple combination treatment group (comprising BYL719, LEE011, anti–PD-1, and anti–CTLA-4) performed the best among all treatment groups (Fig. 6A). All tumors (10/10) treated with the quadruple combination remained regressed during the 50 days of treatment; whereas tumor growth continued in all other treatment groups despite being under constant drug pressure (Supplementary Fig. S3). Of note, 50% (5/10) of tumors treated with the quadruple combination remained regressed for more than a year, even after treatment cessation, with resultant significant improvement in survival compared with BYL719 and LEE011 as well as anti–PD-1 and anti–CTLA-4 treatment groups (Fig. 6A). Notably, there was no toxicity observed in mice following the full treatment schedule.

Figure 6.

Immune-checkpoint blockade in combination with BYL719 and LEE011 treatment induces effective, long-lasting tumor regression in vivo. A, Tumor growth and survival of AT3OVA. Tumor-bearing mice were treated with indicated drug combinations with the following doses: BYL719 (10 mg/kg), LEE011 (40 mg/kg), anti–PD-1 (200 μg), and anti–CTLA-4 (150 μg). There were 6 mice in BYL719 + LEE011 + anti–PD-1 and BYL719 + LEE011 + anti–CTLA-4 groups. There were 10 to 14 mice in all other groups. All treatments ceased by day 50, and tumor growth data are depicted in Fig. 5A. Data depicted are the mean tumor volume ± SEM. B and C, AT3OVA tumor–bearing mice were treated with the indicated drug combinations, with doses as in A. Treatment was for 7 days in vivo. Tumors were then harvested. B, Expression of tumor immunogenicity markers, major histocompatibility complex class I (MHCI; H2KB), MHCII and PDL-1 (C), and granzyme B expression by tumor-infiltrating CD8+ T and CD4+ T cells was analyzed using flow cytometry. Combination: treatment with BYL719, LEE011, anti–PD-1, and anti–CTLA-4. N = 5 to 11 mice per treatment group. Data depicted are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by two-way ANOVA (A) or Kaplan–Meier log rank test, or one-way ANOVA (B and C).

Figure 6.

Immune-checkpoint blockade in combination with BYL719 and LEE011 treatment induces effective, long-lasting tumor regression in vivo. A, Tumor growth and survival of AT3OVA. Tumor-bearing mice were treated with indicated drug combinations with the following doses: BYL719 (10 mg/kg), LEE011 (40 mg/kg), anti–PD-1 (200 μg), and anti–CTLA-4 (150 μg). There were 6 mice in BYL719 + LEE011 + anti–PD-1 and BYL719 + LEE011 + anti–CTLA-4 groups. There were 10 to 14 mice in all other groups. All treatments ceased by day 50, and tumor growth data are depicted in Fig. 5A. Data depicted are the mean tumor volume ± SEM. B and C, AT3OVA tumor–bearing mice were treated with the indicated drug combinations, with doses as in A. Treatment was for 7 days in vivo. Tumors were then harvested. B, Expression of tumor immunogenicity markers, major histocompatibility complex class I (MHCI; H2KB), MHCII and PDL-1 (C), and granzyme B expression by tumor-infiltrating CD8+ T and CD4+ T cells was analyzed using flow cytometry. Combination: treatment with BYL719, LEE011, anti–PD-1, and anti–CTLA-4. N = 5 to 11 mice per treatment group. Data depicted are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001 by two-way ANOVA (A) or Kaplan–Meier log rank test, or one-way ANOVA (B and C).

Close modal

In terms of mechanism, we observed a significant increase in MHCI (H2KB) expression on AT3OVA tumor cells following combined BYL719 and LEE011 treatment in vivo (Fig. 6B), validating our results that BYL719 and LEE011 treatment increases tumor immunogenicity in vitro (Fig. 4C). Although changes in MHCII expression were less impressive with combined BYL719 and LEE011 treatment, similar to results obtained in vitro, we show that addition of immune-checkpoint blockade in the quadruple combination treatment group resulted in a pronounced and significant increase in MHCII as well as PDL-1 expression on AT3OVA tumor cells compared with other treatment groups (Fig. 6B). Moreover, we found that granzyme B expression in CD8+ and CD4+ T cells was significantly increased following quadruple combination treatment compared with all other treatment groups (Fig. 6C). These results support that the addition of immune-checkpoint blockade further augments tumor immunogenicity and subsequent T-cell cytotoxicity responses following PI3Kα and CDK4/6 inhibition in TNBC.

TNBC remains the most challenging breast cancer subtype to treat with the poorest prognosis and median survival after relapse. There are currently no targeted therapies approved for the treatment of TNBC, although Olaparib in BRCA1 germline-deficient breast cancers may soon see this change (31). The majority of TNBC, however, are BRCA1/2 wild-type, and their prognosis remains poor particularly after relapse. New therapies or drug combinations that can achieve durable disease control are needed. Immunotherapy, in particular immune-checkpoint blockade, has shown remarkable success in other solid cancer types; however, response rates in metastatic TNBC patients remain low compared with those observed for immunogenic diseases such as melanoma (32, 33).

In this study, we have shown synergistic antitumor effects for the combination of PI3Kα (BYL719) with CDK4/6 (LEE011, PD991) inhibitors in a variety of TNBC preclinical models including basal-like, mesenchymal, mesenchymal-like, and LAR TNBC subtypes. Cooperative increase in cell-cycle arrest, DNA damage, replicative stress, and apoptosis all contributed to the antitumor effects of combined PI3Kα and CDK4/6 inhibition in TNBC.

A recent report suggested synergism for the CDK4/6 inhibitor (PD991) with two PI3K inhibitors (Pictilisib, GDC-0941 and Taselisib, GDC-0032) for only TNBC cell lines that are of the luminal-like subtype or those that harbor activating mutations in PIK3CA (encoding PI3 kinase catalytic α subunit, PI3Kα; ref. 34). However, our data suggest wide-ranging synergism in a variety of TNBC models in vivo and in vitro, regardless of PIK3CA mutation status, with PI3Kα inhibitor (BYL719) together with either CDK4/6 inhibitors (LEE011 or PD991). The PI3K inhibitors vary markedly in their potency across the PI3K catalytic isoforms (PI3Kα/β/γ/δ), and we hypothesize that this may explain the differences in findings. GDC-0941 is a pan-isoform PI3K inhibitor, whereas GDC-0032 is a β isoform–sparing PI3K inhibitor (IC50 against PI3Kβ = 9.1 nmol/L) and has potent inhibitory effects on PI3Kα (IC50 = 0.29 nmol/L), PI3Kγ (IC50 = 0.97 nmol/L), and PI3Kδ (IC50 = 0.12 nmol/L) isoforms (35, 36). Whereas BYL719 is a selective PI3Kα inhibitor, exhibiting the highest potency against PI3Kα activation (IC50 against PI3Kα = 19 nmol/L) compared with other isoforms such as PI3Kβ (IC50 = 1,156 nmol/L), PI3Kγ (IC50 = 320 nmol/L), and PI3Kδ (IC50 = 290 nmol/L; ref. 37). Furthermore, we differed in our approaches in compound synergy assessment and have additionally evaluated synergistic effects on apoptosis and cell-cycle arrest.

The LAR subtype of TNBC has been previously shown to be susceptible to combined PI3K and CDK4/6 pathway inhibition due to the presence of PIK3CA mutations (14, 34). Moreover, tumors of the LAR subtype have been shown to be heavily enriched in hormonally regulated pathways that include androgen/estrogen metabolism as well as display luminal gene expression patterns, similar to ER-positive luminal breast cancers (23). RB has been shown to have a highly specific and indispensable role in mediating antitumor responses to CDK4/6 inhibition (6, 38, 39). Consistent with other reports (34), the antagonistic interaction of the two compounds in the RB1-mutant cell line MDA-MB-468 further suggests that RB remains essential for synergistic interactions following PI3K and CDK4/6 pathway inhibitions in TNBC. In The Cancer Genome Atlas study, the percentage of TNBC tumors where RB1 was mutated or lost was reported to be only 20% (11), indicating that combined PI3Kα and CDK4/6 inhibitor treatment could be beneficial to the majority of TNBC patients.

Gene expression analyses revealed that combined PI3Kα and CDK4/6 inhibition in TNBC significantly downregulates genes involved in multiple key prosurvival cellular processes including DNA damage and repair responses. We have shown that this corresponds with an increase in DNA damage. The observed increased cyclin E1 expression in addition to reduced expression of DNA repair genes with combined PI3Kα and CDK4/6 inhibitor treatment could have contributed to further catastrophic genomic instability in our models (40, 41).

The clinical importance of tumor-infiltrating lymphocytes (TIL) has been an emerging area of research in breast cancer, particularly in TNBC where higher levels of TILs are associated with both increase in response to chemotherapy and improved survival (2, 15). However, many patients with advanced TNBC have few or no TILs present at diagnosis. Therefore, our novel observation that combined PI3Kα and CDK4/6 inhibition could effectively promote antitumor immunity in TNBC was of high interest. To our knowledge, we have shown for the first time that combined PI3Kα and CDK4/6 inhibition increases tumor antigen presentation as well as immunogenic cell death, which would potentially facilitate the eradication of tumor cells by the immune system (30).

Further evaluation of immune responses to PI3Kα and CDK4/6 inhibition in mouse immunocompetent environment revealed that combination treatment in vivo results in increased activation and cytotoxicity of both adaptive and innate immune cell populations as well as decreased frequency of immune-suppressive MDSCs within the tumor environment. The observation of increased antitumor immunity as a result of suppressing PI3K and CDK4/6 pathways was intriguing as both these pathways have been long thought to be important for immune cell function and proliferation (42, 43). Nonetheless, recent studies have shown that adaptive and innate effector cell function and survival were not affected with PI3Kα inhibition (44, 45). Moreover, our work, along with other reports, has highlighted that various PI3K isoforms as well as some CDKs could be essential for modulation of the immune-suppressive tumor microenvironment (46–49). These studies have demonstrated that blockade of PI3Kδ or γ isoforms reduces the function of Tregs and MDSCs to a greater extent compared with effector T cells, thereby maintaining better control of tumor growth (48, 49). Interestingly, in the clinical setting, neutropenia is reported as an on-target side effect of CDK4/6 inhibition (50, 51). This effect could conceivably contribute to our novel observation of decreased MDSC frequency with PI3Kα and CDK4/6 inhibition. The functional effect of combined therapy on regulatory cell types such MDSCs and Tregs warrants further investigation in future studies.

Taken together, our work suggests that dual inhibition of PI3Kα and CDK4/6 acts in concert to promote synergistic disease control, apoptosis, and antitumor immunity. Importantly, further combination with immune-checkpoint blockade significantly increased tumor immunogenicity and T-cell cytotoxicity, resulting in effective and durable (>1 year) tumor regression with significantly better overall survival compared with all other treatment groups.

In summary, our findings suggest that combined PI3Kα and CDK4/6 inhibition is synergistic and immunogenic against a diverse range of PIK3CA- and RB1-wild-type preclinical TNBC models. We also conclude that antitumor efficacy can be further enhanced with the addition of immune-checkpoint blockade. Our results have identified a novel approach that warrants evaluation in clinical trials for patients in critical need of new effective therapies.

S. Loi has received funding to her institution from Novartis and Pfizer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: Z.L. Teo, W.A. Phillips, P.K. Darcy, S. Loi

Development of methodology: Z.L. Teo, B. Virassamy, S. Loi

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Z.L. Teo, S. Versaci, S. Dushyanthen, C.P. Mintoff, B. Virassamy, S. Loi

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Z.L. Teo, S. Dushyanthen, F. Caramia, P. Savas, M. Zethoven, B. Virassamy, G.A. McArthur, P.K. Darcy, S. Loi

Writing, review, and/or revision of the manuscript: Z.L. Teo, P. Savas, M. Zethoven, S.J. Luen, G.A. McArthur, W.A. Phillips, P.K. Darcy, S. Loi

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Z.L. Teo, F. Caramia, B. Virassamy, S. Loi

Study supervision: P.K. Darcy, S. Loi

The authors wish to acknowledge staff from the Peter MacCallum Cancer Centre Animal Facility, FACS Facility, and Molecular Genomics Core Facility for their assistance.

This work was supported by a National Health and Medical Research Council (NHMRC) of Australia Project Grant (1123208; S. Loi, P.K. Darcy, W.A. Phillips). Z.L. Teo was supported by a NHMRC Early Career Fellowship (1106967). P.K. Darcy was supported by a NHMRC Senior Research Fellowship (1041828). S. Loi was supported by the Cancer Council Victoria Australia John Colebatch Fellowship as well as the Breast Cancer Research Foundation NY. S. Loi has received funding to her institution from Novartis and Pfizer.

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.
Metzger-Filho
O
,
Tutt
A
,
de Azambuja
E
,
Saini
KS
,
Viale
G
,
Loi
S
, et al
Dissecting the heterogeneity of triple-negative breast cancer
.
J Clin Oncol
2012
;
30
:
1879
87
.
2.
Loi
S
,
Sirtaine
N
,
Piette
F
,
Salgado
R
,
Viale
G
,
Van Eenoo
F
, et al
Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98
.
J Clin Oncol
2013
;
31
:
860
7
.
3.
Parkes
EE
,
Walker
SM
,
Taggart
LE
,
McCabe
N
,
Knight
LA
,
Wilkinson
R
, et al
Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer
.
J Nat Cancer Inst
2017
;
109
:
djw199
.
4.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
5.
Musgrove
EA
,
Caldon
CE
,
Barraclough
J
,
Stone
A
,
Sutherland
RL
. 
Cyclin D as a therapeutic target in cancer
.
Nat Rev Cancer
2011
;
11
:
558
72
.
6.
DeMichele
A
,
Clark
AS
,
Tan
KS
,
Heitjan
DF
,
Gramlich
K
,
Gallagher
M
, et al
CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment
.
Clin Cancer Res
2015
;
21
:
995
1001
.
7.
Finn
RS
,
Crown
JP
,
Lang
I
,
Boer
K
,
Bondarenko
IM
,
Kulyk
SO
, et al
The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study
.
Lancet Oncol
2015
;
16
:
25
35
.
8.
Turner
NC
,
Ro
J
,
André
F
,
Loi
S
,
Verma
S
,
Iwata
H
, et al
Palbociclib in hormone-receptor–positive advanced breast cancer
.
N Engl J Med
2015
;
373
:
209
19
.
9.
Hortobagyi
GN
,
Stemmer
SM
,
Burris
HA
,
Yap
YS
,
Sonke
GS
,
Paluch-Shimon
S
, et al
Ribociclib as first-line therapy for HR-positive, advanced breast cancer
.
N Engl J Med
2016
;
375
:
1738
48
.
10.
Finn
RS
,
Martin
M
,
Rugo
HS
,
Jones
S
,
Im
SA
,
Gelmon
K
, et al
Palbociclib and letrozole in advanced breast cancer
.
N Engl J Med
2016
;
375
:
1925
36
.
11.
Koboldt
DC
,
Fulton
RS
,
McLellan
MD
,
Schmidt
H
,
Kalicki-Veizer
J
,
McMichael
JF
, et al
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
12.
Finn
RS
,
Dering
J
,
Conklin
D
,
Kalous
O
,
Cohen
DJ
,
Desai
AJ
, et al
PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro
.
Breast Cancer Res
2009
;
11
:
R77
.
13.
Herrera-Abreu
MT
,
Palafox
M
,
Asghar
U
,
Rivas
MA
,
Cutts
RJ
,
Garcia-Murillas
I
, et al
Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer
.
Cancer Res
2016
;
76
:
2301
13
.
14.
Vora
SR
,
Juric
D
,
Kim
N
,
Mino-Kenudson
M
,
Huynh
T
,
Costa
C
, et al
CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors
.
Cancer Cell
2014
;
26
:
136
49
.
15.
Denkert
C
,
von Minckwitz
G
,
Brase
JC
,
Sinn
BV
,
Gade
S
,
Kronenwett
R
, et al
Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2–positive and triple-negative primary breast cancers
.
J Clin Oncol
2015
;
33
(
9
):
983
91
.
16.
Loi
S
. 
Host antitumor immunity plays a role in the survival of patients with newly diagnosed triple-negative breast cancer
.
J Clin Oncol
2014
;
32
:
2935
7
.
17.
Emens
L
,
Adams
S
,
Loi
S
,
Schmid
P
,
Schneeweiss
A
,
Rugo
H
, et al
A phase III randomized trial of atezolizumab in combination with nab-paclitaxel as first line therapy for patients with metastatic triple-negative breast cancer (mTNBC)
.
Cancer Res
2016
;
76
:
OT1-01-6
.
18.
Mattarollo
SR
,
Loi
S
,
Duret
H
,
Ma
Y
,
Zitvogel
L
,
Smyth
MJ
. 
Pivotal role of innate and adaptive immunity in anthracycline chemotherapy of established tumors
.
Cancer Res
2011
;
71
:
4809
20
.
19.
Dushyanthen
S
,
Teo
ZL
,
Caramia
F
,
Savas
P
,
Mintoff
C
,
Virassamy
B
, et al
Agonist immunotherapy restores T cell function following MEK inhibition improving efficacy in breast cancer
.
Nat Commun
2017
;
8
:
606
.
20.
Chou
TC
. 
Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies
.
Pharmacol Rev
2006
;
58
:
621
81
.
21.
Chou
T-C
,
Talalay
P
. 
Analysis of combined drug effects: a new look at a very old problem
.
Trends Pharmacol Sci
1983
;
4
:
450
4
.
22.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Nat Acad Sci U S A
2005
;
102
:
15545
50
.
23.
Lehmann
BD
,
Bauer
JA
,
Chen
X
,
Sanders
ME
,
Chakravarthy
AB
,
Shyr
Y
, et al
Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies
.
J Clin Invest
2011
;
121
:
2750
67
.
24.
Averous
J
,
Fonseca
BD
,
Proud
CG
. 
Regulation of cyclin D1 expression by mTORC1 signaling requires eukaryotic initiation factor 4E-binding protein 1
.
Oncogene
2008
;
27
:
1106
13
.
25.
Hara
K
,
Yonezawa
K
,
Kozlowski
MT
,
Sugimoto
T
,
Andrabi
K
,
Weng
Q-P
, et al
Regulation of eIF-4E BP1 Phosphorylation by mTOR
.
J Biol Chem
1997
;
272
:
26457
63
.
26.
Ohtsubo
M
,
Theodoras
AM
,
Schumacher
J
,
Roberts
JM
,
Pagano
M
. 
Human cyclin E, a nuclear protein essential for the G1-to-S phase transition
.
Mol Cell Biol
1995
;
15
:
2612
24
.
27.
Atefi
M
,
Avramis
E
,
Lassen
A
,
Wong
DJ
,
Robert
L
,
Foulad
D
, et al
Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma
.
Clin Cancer Res
2014
;
20
:
3446
57
.
28.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
8
.
29.
Crane
CA
,
Panner
A
,
Murray
JC
,
Wilson
SP
,
Xu
H
,
Chen
L
, et al
PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer
.
Oncogene
2009
;
28
:
306
12
.
30.
Obeid
M
,
Tesniere
A
,
Ghiringhelli
F
,
Fimia
GM
,
Apetoh
L
,
Perfettini
JL
, et al
Calreticulin exposure dictates the immunogenicity of cancer cell death
.
Nat Med
2007
;
13
:
54
61
.
31.
Robson
M
,
Im
S-A
,
Senkus
E
,
Xu
B
,
Domchek
SM
,
Masuda
N
, et al
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Eng J Med
2017
:
1
11
.
32.
Adams
S
,
Diamond
JR
,
Hamilton
EP
,
Pohlmann
PR
,
Tolaney
SM
,
Molinero
L
, et al
Phase Ib trial of atezolizumab in combination with nab-paclitaxel in patients with metastatic triple-negative breast cancer (mTNBC)
.
J Clin Oncol
2016
;
34
:
1009
.
33.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
34.
Turner
NC
,
Asghar
U
,
Barr
AR
,
Cutts
R
,
Beaney
M
,
Babina
IS
, et al
Single-cell dynamics determines response to CDK4/6 inhibition in triple negative breast cancer
.
Clin Cancer Res
2017
;
23
:
5561
72
.
35.
Ndubaku
CO
,
Heffron
TP
,
Staben
ST
,
Baumgardner
M
,
Blaquiere
N
,
Bradley
E
, et al
Discovery of 2-{3-[2-(1-isopropyl-3-methyl-1H-1,2-4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (GDC-0032): a beta-sparing phosphoinositide 3-kinase inhibitor with high unbound exposure and robust in vivo antitumor activity
.
J Med Chem
2013
;
56
:
4597
610
.
36.
Folkes
AJ
,
Ahmadi
K
,
Alderton
WK
,
Alix
S
,
Baker
SJ
,
Box
G
, et al
The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-t hieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer
.
J Med Chem
2008
;
51
:
5522
32
.
37.
Furet
P
,
Guagnano
V
,
Fairhurst
RA
,
Imbach-Weese
P
,
Bruce
I
,
Knapp
M
, et al
Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation
.
Bioorg Med Chem Lett
2013
;
23
:
3741
8
.
38.
Dean
JL
,
Thangavel
C
,
McClendon
AK
,
Reed
CA
,
Knudsen
ES
. 
Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure
.
Oncogene
2010
;
29
:
4018
32
.
39.
Fry
DW
,
Harvey
PJ
,
Keller
PR
,
Elliott
WL
,
Meade
M
,
Trachet
E
, et al
Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts
.
Mol Cancer Ther
2004
;
3
:
1427
38
.
40.
Spruck
CH
,
Won
KA
,
Reed
SI
. 
Deregulated cyclin E induces chromosome instability
.
Nature
1999
;
401
:
297
300
.
41.
Negrini
S
,
Gorgoulis
VG
,
Halazonetis
TD
. 
Genomic instability–an evolving hallmark of cancer
.
Nature Rev Mol Cell Biol
2010
;
11
:
220
8
.
42.
Okkenhaug
K
,
Vanhaesebroeck
B
. 
PI3K in lymphocyte development, differentiation and activation
.
Nat Rev Immunol
2003
;
3
:
317
30
.
43.
Wells
AD
,
Morawski
PA
. 
New roles for cyclin-dependent kinases in T cell biology: linking cell division and differentiation
.
Nat Rev Immunol
2014
;
14
:
261
70
.
44.
So
L
,
Yea
SS
,
Oak
JS
,
Lu
M
,
Manmadhan
A
,
Ke
QH
, et al
Selective inhibition of phosphoinositide 3-kinase p110alpha preserves lymphocyte function
.
J Biol Chem
2013
;
288
:
5718
31
.
45.
Yea
SS
,
So
L
,
Mallya
S
,
Lee
J
,
Rajasekaran
K
,
Malarkannan
S
, et al
Effects of novel isoform-selective phosphoinositide 3-kinase inhibitors on natural killer cell function
.
PLoS One
2014
;
9
:
e99486
.
46.
Matsuura
I
,
Denissova
NG
,
Wang
G
,
He
D
,
Long
J
,
Liu
F
. 
Cyclin-dependent kinases regulate the antiproliferative function of Smads
.
Nature
2004
;
430
:
226
31
.
47.
Tone
Y
,
Furuuchi
K
,
Kojima
Y
,
Tykocinski
ML
,
Greene
MI
,
Tone
M
. 
Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer
.
Nat Immunol
2008
;
9
:
194
202
.
48.
Kaneda
MM
,
Messer
KS
,
Ralainirina
N
,
Li
H
,
Leem
CJ
,
Gorjestani
S
, et al
PI3Kgamma is a molecular switch that controls immune suppression
.
Nature
2016
;
539
:
437
42
.
49.
Ali
K
,
Soond
DR
,
Pineiro
R
,
Hagemann
T
,
Pearce
W
,
Lim
EL
, et al
Inactivation of PI(3)K p110delta breaks regulatory T-cell-mediated immune tolerance to cancer
.
Nature
2014
;
510
:
407
11
.
50.
Flaherty
KT
,
Lorusso
PM
,
Demichele
A
,
Abramson
VG
,
Courtney
R
,
Randolph
SS
, et al
Phase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancer
.
Clin Cancer Res
2012
;
18
:
568
76
.
51.
Schwartz
GK
,
LoRusso
PM
,
Dickson
MA
,
Randolph
SS
,
Shaik
MN
,
Wilner
KD
, et al
Phase I study of PD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (Schedule 2/1)
.
Br J Cancer
2011
;
104
:
1862
8
.