1 Introduction

The PI3K/AKT/mTOR pathway plays a central role in cell physiology by transmitting signal transduction events in response to extracellular stimuli. This pathway controls many cellular functions such as proliferation, growth, survival, motility, and metabolism [1]. Mutations in this signalling route are frequently found in cancer, being particularly common in breast cancer, where about 60% of tumors harbor genetic alterations that hyperactivate the PI3K/AKT/mTOR pathway [2]. Preclinical studies have proven these alterations to be oncogenic drivers and, as such, therapeutic targets. Several drugs against PI3K, mTOR, and AKT are in clinical development. However, so far, the results have been modest with only one drug approved, everolimus, for the treatment of metastatic estrogen receptor (ER)-positive breast cancer. Recent advances in preclinical and clinical research are shedding some light on how to best approach the therapeutic inhibition of PIK3/AKT/mTOR signaling in patients. Drugs targeting this pathway face many challenges to succeed such as the identification of a biomarker predictive of tumor response, development of rational combinations with other compounds to overcome mechanisms of adaptive resistance, and optimization of approaches to minimize toxicity while still inhibiting their molecular target(s). In this review, we will discuss the current knowledge on drug development against the pathway, its limitations, and suggestions for an effective drug development.

2 Function and genetic alterations of PI3K/AKT pathway in breast cancer

Phosphoinositide 3-kinases (PI3Ks) are a family of three different classes of lipid kinases. Class I PI3K is the most studied and clearly implicated in oncogenic transformation and tumor growth [13]. Class I PI3Ks are heterodimers consisting of a p85 regulatory subunit and a p110 catalytic subunit (p110α, p110β, p110γ, or p110δ). PI3K receives signals from growth factor receptor tyrosine kinases, such as ERBB receptors, FGFR and IGF-1R, and G protein-coupled receptors. Activated receptors phosphorylate adaptor proteins which, in turn, bind the amino-terminal SH2 domain of p85. This binding frees p110 from the inhibitory effect of p85 which then catalyzes the conversion of phosphatidylinositol bisphosphate, PI(4,5)P2, to phosphatidylinositol triphosphate, PI(3,4,5)P3. PIP3 recruits PDK1 and AKT, through their pleckstrin homology (PH) domain, to the plasma membrane. PDK1 phosphorylates AKT at Thr308. The mTOR/Rictor (TORC2) complex phosphorylates AKT at Ser473, resulting in full activation of this enzyme. PTEN and INPP4B dephosphorylate PIP3 in positions 3 and 5 of the inositol ring, respectively, thereby negatively regulating PI3K signaling output [4, 5]. Activated AKT phosphorylates and inhibits tuberous sclerosis complex 1 and 2 (TSC1/2) resulting in accumulation of Ras homolog enriched in brain (RHEB) which activates the complex mTOR/Raptor (TORC1). TORC1 phosphorylates ribosomal protein S6 kinase (S6K1) and eIF4E binding protein 1 (4E-BP1) promoting messenger RNA (mRNA) translation, protein synthesis, and autophagy. AKT also phosphorylates GSK3α, GSK3β, FoxO transcription factors, MDM2, BAD, and p27KIP1 to facilitate survival and cell cycle entry (Fig. 1).

Fig. 1
figure 1

The PI3K/AKT/mTOR pathway signaling as a therapeutic target. Activated growth factor receptors phosphorylate adaptor proteins like IRS1 which recruit p85/p110 (PI3K) dimers to the plasma membrane. PI3K is a heterodimer composed of a p85 regulatory subunit and a p110 catalytic subunit. p85 binding to IRS1 relieves its inhibitory effect on p110. Activated p110 catalyzes the conversion of PIP2 to PIP3. PTEN and INPP4B dephosphorylate PIP3, thus negatively regulating PI3K. PIP3 recruits PDK1 and AKT to the membrane. Full activation of AKT requires its phosphorylation by PDK1 and mTORC2. Activated AKT inhibits the complex TSC1/2, resulting in RHEB-GTP accumulation which, in turn, activates TORC1. Activated TORC1 phosphorylates ribosomal protein S6 kinase (S6K1) and eIF4E binding protein 1 (4E-BP1) promoting mRNA translation, protein synthesis, and autophagy. Small-molecule inhibitors discussed in the text are included in the figure. Dashed lines represent the inhibitory feedback loop relieved upon inhibition of the pathway

Mutations of the PIK3CA gene, which encodes p110α, are the most common genetic alteration in breast cancer occurring at frequency of 45% in luminal A, 30% in luminal B, 39% in HER2-enriched, and 9% in basal-like breast cancer subtypes. More than 80% of the mutations cluster within the helical (E542K and E545K) or the kinase (H1047R) domains of p110α [6]. Helical domain mutations increase catalytic activity by reducing the repression of p110α by p85 [7] or facilitating the interaction of p110α with IRS1 [8], whereas kinase domain mutations mainly increase the retention of p110α at the plasma membrane [9]. Preclinical data from cell-based studies and genetically engineered mice (GEMs) have clearly shown that these mutations activate PI3K/AKT/mTOR signaling and are oncogenic drivers by promoting cell transformation, tumor initiation, progression, and resistance to apoptosis [1012]. However, data from knock-in GEMs, where the PIK3CA mutant protein is expressed at physiologic levels in the mammary gland, do not show pathway hyperactivation. In this knock-in models, mammary tumors develop after a long latency [13], suggesting that additional genetic alterations are needed to recapitulate a PI3K-induced transformed phenotype [14, 15]. This is consistent with data from primary breast cancers showing a disconnection between PIK3CA mutation and PI3K pathway activation. For example, luminal tumors, despite having the highest incidence of PIK3CA mutations, do not exhibit high levels of (activated) p-AKT, p-S6, and p-4EBP1 [16]. Further, PIK3CA mutations have been associated in early ER-positive breast cancer with good prognosis and are not a negative predictive factor for response to adjuvant endocrine therapy [1719]. In triple-negative breast cancer, the PI3K/AKT/mTOR activation appears to be driven mainly by loss of PTEN (30%) or INPP4B (40%). Genomic loss of these tumor suppressors is associated with increased levels of p-AKT, p-S6, and p-4EBP1 [16, 20]. Around 3% of luminal tumors harbor AKT mutations in the PH domain (E17K), which result in constitutive localization at the plasma membrane and resulting activation of AKT [21]. Mutations in PIK3R1, the gene encoding the p85 regulatory subunit of PI3K, have also been reported although with a lower frequency (≈2%). Interestingly, PIK3R1 mutations cluster in the region of the protein that contacts p110, thus reducing the inhibitory effect of p85 on the isozyme [22, 23].

3 Inhibition of mTOR in cancer

mTOR is a serine/threonine kinase composed by the following two distinct protein complexes: a rapamycin- and nutrient-sensitive multiprotein complex (TORC1) and a growth factor-sensitive but nutrient- and rapamycin-insensitive complex (TORC2). TORC1 responds to amino acids, stress, oxygen, energy requirements and growth factors, and promotes cell growth and cell cycle progression. TORC2 responds to growth factors and regulates cell survival and metabolism, as well as the cytoskeleton [24]. Inhibitors of mTOR such as rapamycin (sirolimus) and the rapalogs temsirolimus (CCI-779), everolimus (RAD001), and deforolimus (AP23573) have been extensively evaluated in hematological malignancies, renal cancer, and as treatment for transplant rejection.

4 mTOR inhibitors in ER-positive breast cancer

Preclinical studies show that PI3K/AKT/mTOR activation is a mechanism of acquired resistance to long-term estrogen deprivation [2527]. Results from two randomized trials, BOLERO-2 and TAMRAD [28, 29], suggested that the addition of the TORC1 inhibitor everolimus to anti-estrogen therapy can reverse endocrine resistance. In both studies, all patients had been previously exposed to aromatase inhibitors (AIs) and most of them developed progression after an initial response (acquired resistance). Patients were randomized to receive anti-estrogen therapy (exemestane in BOLERO-2, tamoxifen in TAMRAD) plus everolimus vs the anti-estrogen alone. The addition of everolimus increased the median progression-free survival (PFS) in both studies (BOLERO-2 PFS 7.8 vs 3.2 months, p < 0.0001; TAMRAD PFS 8.6 vs 4.5 months, p < 0.01). This led in 2012 to the approval of everolimus in combination with endocrine therapy after progression on AIs from the FDA and the EMA. The phase III trial HORIZON [30] evaluated the addition of temsirolimus to letrozole vs letrozole alone in AI-naive patients. There was no difference in outcome between the two arms of the study probably because HORIZON, different to BOLERO-2 and TAMRAD, tested the impact of TORC1 inhibition on primary endocrine resistance. In both BOLERO-2 and TAMRAD, the addition of everolimus to endocrine therapy produced higher rate of stomatitis, rash, fatigue, anorexia, anemia, and thrombocytopenia than endocrine therapy alone, which produced frequent treatment discontinuation and dose modifications. A recent study showed that stomatitis, the most common adverse event of everolimus, can be considerably reduced with the prophylactic use of dexamethasone oral solution [31]. This strategy could help to maintain the dose level and probably the drug efficacy. All this toxicity highlights one limitation of inhibitors of the PI3K/AKT/mTOR pathway and the importance of knowledge and practice on the management of adverse events associated with drugs like everolimus.

Predictive biomarkers of response to everolimus in breast cancer are not yet established. A biomarker analysis in ≈300 patients from BOLERO-2 using targeted exome sequencing, mostly of DNA from primary tumors, failed to identify any correlation between PIK3CA-activating mutations or other gene alterations in the PI3K/AKT/mTOR pathway and benefit from everolimus [32]. Also, selected PIK3CA mutations in plasma tumor at study entry did not predict for benefit from everolimus [33]. Interestingly, 10 patients with somatic mTOR kinase mutations derived clinical benefit from everolimus, which is in agreement with reports in renal and bladder cancers [34, 35].

5 mTOR inhibitors in HER2-overexpressing breast cancer

Laboratory studies have suggested that inhibition of PI3K/AKT/mTOR pathway is required for the anti-tumor action of HER2-targeted therapies and that it can also mediate resistance to anti-HER2 therapies [3638]. Based on this background, everolimus has been evaluated as treatment of de novo and acquired resistance to trastuzumab in two phase III trials, BOLERO-1 and BOLERO-3. In BOLERO-1 [39], addition of everolimus to trastuzumab and paclitaxel as first-line treatment of HER2-positive advanced breast cancer did not prolong PFS (15 vs 14 months). BOLERO-3 [40] only included patients progressing on trastuzumab-paclitaxel; in this trial, the addition of everolimus to trastuzumab-vinorelbine prolonged PFS compared to trastuzumab-vinorelbine (7.0 vs 5.8 months, p = 0.0067). Both studies showed that patients with ER-negative/HER2-positive tumors derived more benefit from everolimus.

A genomic and immunohistochemical analysis of tumors from 377 patients enrolled in BOLERO-1 and BOLERO-3 showed that the benefit of adding everolimus was restricted to patients with PIK3CA mutations, PTEN loss, or “hyperactive” PI3K pathway, defined as low PTEN expression/mutation and/or known PIK3CA and/or AKT1 E17K mutation [41]. These results are in agreement with preclinical studies that suggest that PI3K/AKT/mTOR activation is causal to resistance to anti-HER2 therapies. For example, GEMs with transgenic mammary tumors HER2 and PIK3CAH1047R are highly resistant to anti-HER2 combinations (trastuzumab/pertuzumab and trastuzumab/lapatinib) with the addition of the pan-PI3K inhibitor buparlisib restoring drug sensitivity [42]. A biomarker analysis in CLEOPATRA (phase III study evaluating trastuzumab/taxane ± pertuzumab in HER2-positive metastatic breast cancer) showed that patients with PTEN loss and PIK3CA mutations derived lower benefit of the anti-HER2 therapy [43]. In the setting of neoadjuvant treatment, a recent meta-analysis including almost 1000 patients [44] suggested that patients with HER2+/PIK3CA mutant tumors had lower rates of pathologic complete response to trastuzumab, lapatinib, or the combination compared to HER2+ breast cancers with wild-type PIK3CA. This detrimental effect was restricted to ER-positive tumors but had no impact in overall survival. These results of the interaction of PI3K activation with prognosis and intrinsic resistance to anti-HER2 therapies in the neoadjuvant and metastatic setting are hard to reconcile with data from the adjuvant setting. Neither PIK3CA hot spot-activating mutation or PTEN loss predicted for lack of benefit from adjuvant trastuzumab in two randomized clinical trials including more than 3000 patients [45, 46].

6 New TORC1/TORC2 inhibitors

Everolimus is an allosteric inhibitor of TORC1 but does not affect TORC2. Blockage of TORC1 relieves a negative-feedback loop between S6K and IRS1, leading to an increase in phosphorylation of AKT on Ser473 by uninhibited TORC2. This has been seen in tumor biopsies of patients on treatment with rapalogs [47, 48], underscoring an intrinsic limitation of the anti-tumor activity of TORC1 inhibitors. Catalytic TORC1/TORC2 inhibitors, such as AZD2014 or MLN0128, are in clinical development with the aim of a more profound and complete blockade of mTOR complexes and avoidance of the compensatory activation of AKT [49, 50]. Laboratory studies have already shown that TORC1/TORC2 inhibitors induce a better blockade of PI3K/AKT/mTOR signaling as measured by inhibition of p-4EBP1, p-S6, and p-AKT compared to rapalogs [51]. These drugs are also active against everolimus-resistant acquired mutations in the rapamycin-binding domain of mTOR [52, 53].

7 PI3K inhibitors in breast cancer

A plethora of compounds have been developed to inhibit PI3K in breast cancer (Table 1). They are classified according to the specificity for each PI3K isoform in (1) pan-PI3K, targeting all class I isoforms, isoform-specific PI3K inhibitors, and dual-PI3K/mTOR inhibitors. As single agents, these drugs are far from the response rates obtained with the inhibition of other oncogenic kinases in other cancer types (such as those targeting mutant EGFR and mutant ALK in lung cancer and mutant BRAF in melanoma). Co-existing genetic alterations in tumors, compensatory feedback loops, and associated toxicity that precludes an adequate dose intensity are possible explanations for this lack of efficacy. Early results with pan-PI3K inhibitors in breast cancer are disappointing, with several studies showing increase toxicity and not a substantial improvement over endocrine therapy alone. Several phase III registration trials with PI3Kα-specific inhibitors are ongoing at the time of this writing (Table 1).

Table 1 Selected ongoing phase II and III clinical trial PI3K/AKT/mTOR inhibitors in breast cancer

8 Pan-PI3K inhibitors in combination with endocrine therapy

Preclinical studies and retrospective analysis of some clinical trials have suggested that ER+/PIK3CA mutant tumors exhibit a lower response to anti-estrogens compared to ER+/PIK3CA wild-type tumors [54, 55]. In addition, some patients with ER+ breast cancer that progress on anti-estrogen therapy respond clinically to PI3K inhibitors [56]. ER+ human breast cancer cell lines that adapt to estrogen deprivation exhibit amplification of PI3K/AKT/mTOR signaling, and PI3K pathway inhibitors prevent acquired hormone independence [26]. Low levels of estradiol can rescue ER+/PIK3CA mutant cells from the lethal effect of PI3K inhibitors [57]. Further, inhibition of PI3K/AKT results in upregulation of ERα mRNA and protein and ER transcriptional activity [5860], also suggesting co-regulation of ER and PI3K pathways. Finally, combined inhibition of ER and PI3K is synergistic against ER+/PIK3CA mutant xenografts [60, 61]. Taken together, these data strongly suggest that combined inhibition of ER and PI3K is a robust therapeutic approach to target these cancers.

Pan-PI3K inhibitors such as pictilisib (GDC-0941) or buparlisib (BKM120) target all class I p110 isoforms. Although they provide a potential advantage by broadly targeting multiple oncogenic PI3K isozymes, this approach narrows their therapeutic window by increasing adverse events [62].

Pictilisib (GDC-0941) is an oral ATP-competitive reversible inhibitor of all four class I PI3K isoforms [63, 64]. In a presurgical study by Schmid et al. [65], the combination of picitisilib and anastrozole was superior to anastrozole alone at inhibiting breast cancer cell proliferation as measured by Ki67 IHC. PIK3CA mutations were not predictive of response in this study. The phase II FERGI trial evaluated the addition of pictisilib to fulvestrant in post-menopausal patients progressing on an AI [66]. Pictisilib and fulvestrant did not increase PFS compared to fulvestrant and placebo (hazard ratio of 0·74, p = 0.096). Of note, no difference in outcome was seen in patients with PIK3CA mutant tumors. Patients treated with pictisilib showed a high rate of serious adverse event, mostly rash, pneumonitis, diarrhea, stomatitis, and transaminitis. This toxicity led to dose modifications in 45% of cases and treatment discontinuation in 24%. Of note, only 18% of patients developed hyperglycemia, which is an on-target effect of pictilisib as a result of inhibition of p110α, suggesting that adequate inhibition of PI3K was not achieved in FERGI, thus limiting the evaluability of this study.

Two early-phase clinical trials tested the pan-PI3K inhibitor buparlisib in combination with endocrine therapy, reporting good activity and a toxicity profile characterized by transaminitis, hyperglycemia, diarrhea, and mood disorders (anxiety, depression, irritability) [56, 67]. Both studies showed that the intermittent dose of buparlisib (100 mg 5 days on/2 days off) had lower rates of adverse events than daily dosing (100 mg/day). Buparlisib in combination with fulvestrant was studied in the phase III BELLE-2 trial for post-menopausal patients with metastatic breast cancer progressing on an AI [68]. Patients were randomized to receive fulvestrant plus buparlisib or placebo. Randomization was stratified by PI3K/AKT/mTOR pathway activation (defined by PIK3CA mutation and/or PTEN loss) status. Buparlisib modestly increased the median PFS by 1.9 months (6.9 vs 5.0 months, p < 0.001). For patients with pathway activation, there was no difference in the benefit of buparlisib. In a subset of patients (n = 581) where PIK3CA mutation was assessed in plasma tumor, cell-free DNA at trial entry, buparlisib plus fulvestrant showed a significant increase in PFS when compared to fulvestrant alone (7 vs 3.2 months, HR 0.56, p < 0.001) for PIK3CA mutants. Serious adverse events were reported in 23% of patients, dose reductions in 46%, and dose interruptions in 55%. The toxicity profile was characterized by transaminitis, hyperglycemia, mood disorders, and rash (Table 2).

Table 2 PI3K/AKT/mTOR pathway inhibitor potencies and common toxicities

At this time, the low benefit/toxicity ratio of buparlisib is not encouraging for the further development of this drug. Results of a phase III trial of fulvestrant ± buparlisib in patients with ER+ breast cancer progressing on everolimus (BELLE-3; NCT01633060) will be reported at the end of 2016.

9 PI3K alfa-specific inhibitors in combination with endocrine therapy

PI3Kα is the isoform predominantly mutated in cancer. Its selective inhibition has been shown to block PI3K/AKT signaling in response to different growth factor stimuli [6971]. The rationale for the development of PIK3α-specific inhibitors is to maximize the inhibition of p110α while sparing the patient from the side effects associated with inhibition of all p110 isozymes. Two PI3Kα inhibitors are in clinical development for breast cancer in combination with endocrine therapy, alpelisib (BYL719) and taselisib (GDC-0032).

Alpelisib has been studied as single agent [72], showing preferential activity against those tumors with PIK3CA mutations and a toxicity profile overall lower than that seen with pan-PI3K inhibitors and consisting of hyperglycemia, nausea, diarrhea, decreased appetite, fatigue, vomiting, and rash. Alpelisib plus fulvestrant is being studied in a phase III trial for patients with metastatic ER-positive breast cancer progressing on an AI (SOLAR1; NCT02437318). Two other phase I trials of alpelisib in combination with AIs have reported also showing preferential activity in patients with PIK3CA mutant cancers [73, 74]. In the study conducted by our group, the clinical benefit rate in patients with PIK3CA mutant vs PIK3CA wild tumors was 41 vs 20%, respectively.

Taselisib is a potent inhibitor of p110α, p110δ, and p110γ, with a 30-fold less potency against p110β active to p110α. It has also a greater selectivity against PIK3CA mutant isoforms than wild type and it is superior in terms of pathway inhibition and induction of apoptosis to other PI3K inhibitors [75, 76]. A phase I trial of taselisib + letrozole [77] reported the following grade ≥3 adverse events: diarrhea 14%, hyperglycemia 7%, stomatitis 4%, and fatigue 4%; the recommended phase II dose was 6 mg/day. A phase Ib trial of taselisib in combination with tamoxifen was presented in ASCO 2016 [78], with a similar toxicity profile. A phase II trial of the combination taselisib + fulvestrant [79] in 60 patients reported colitis (13.3%) and diarrhea (11.7%) as the most common grade ≥3 adverse events and an 18% rate of treatment discontinuation. Clinical response rate was higher in patients with PIK3CA mutant compared to PIK3CA wild-type cancers (41 vs 14%, respectively). Taselisib is currently being evaluated with fulvestrant in a randomized phase III study in patients with metastatic ER+ tumors with and without PIK3CA mutations and with previous exposure to an AI (SANDPIPER; NCT02340221).

10 AKT inhibitors

AKT is a serine/threonine kinase with three isoforms (AKT1, AKT2, and AKT3). It is a downstream target of the PI3K and plays an important role in cancer cell survival, cell cycle entry, and glucose metabolism [2]. E17K-AKT1 is the most common somatic alteration in AKT, occurring in about 2% of all breast cancers. E17K increases lipid affinity to the AKT PH domain, resulting in constitutive membrane localization and activation of AKT [21]. There are several phase I and II clinical trials with allosteric and catalytic AKT inhibitors. In breast cancer, clinical results have been communicated with MK-2206 and AZD5363. A phase I trial of the combination of the allosteric pan-AKT inhibitor MK-2206 with fulvestrant reported grade 3 rash as the dose-limiting toxicity and a clinical benefit rate of 42%. Response was not associated with PI3K mutation status [80]. A recent report by Hyman and colleagues suggests that the E17K-AKT1 mutation could be a predictive marker for the efficacy of the pan-AKT catalytic inhibitor AZD5363. AZD5363 was evaluated in a phase I trial in patients with solid tumors harboring E17K-AKT1; 33 of 41 assessable patients (including 21 with ER-positive breast cancer) had regression of tumors and persistent AKT1-E17K allele decay in plasma tumor DNA [81].

11 Conclusions

The PI3K/AKT/mTOR pathway is the most frequently mutated network in breast cancer, providing multiple molecular targets that can be exploited with a therapeutic intent. However, biomarkers which identify PI3K-dependent cancers, that as a result of that dependence would be more likely to respond to these drugs, are not yet established. Following the approval of TORC1 inhibitors, which disable one of the key hubs downstream PI3K, PI3Kα inhibitors are poised to be the next ones to be approved for use in breast cancer. Large ongoing randomized registration trials with PI3Kα inhibitors include an adequate number of patients with cancers with activating “hot spot” PIK3CA mutations in order to determine if the benefit of these drugs, if present, will be limited to against cancers of that genotype. Although manageable, on-target toxicities induced by these drugs are not insignificant. Despite early signals of clinical activity, significant challenges for the therapeutic targeting of PI3K/AKT remain. These include mechanisms of compensation in tumor and host tissues upon treatment with these drugs that limit their optimal biological dosing and anti-tumor effect but also the incomplete knowledge about targeted drugs that should be used in combination with PI3K pathway inhibitors. It is clear that the benefit of these drugs will require development of rational combinations before they become a significant component of the anti-cancer portfolio in breast cancer.