Next Article in Journal
HSF1 Can Prevent Inflammation following Heat Shock by Inhibiting the Excessive Activation of the ATF3 and JUN&FOS Genes
Next Article in Special Issue
Augmented Antitumor Effect of Unripe Rubus coreanus Miquel Combined with Oxaliplatin in a Humanized PD-1/PD-L1 Knock-In Colorectal Cancer Mouse Model
Previous Article in Journal
Simple Detection of Unstained Live Senescent Cells with Imaging Flow Cytometry
Previous Article in Special Issue
The AGEs/RAGE Transduction Signaling Prompts IL-8/CXCR1/2-Mediated Interaction between Cancer-Associated Fibroblasts (CAFs) and Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer

1
Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
2
Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(16), 2508; https://doi.org/10.3390/cells11162508
Submission received: 9 July 2022 / Revised: 6 August 2022 / Accepted: 9 August 2022 / Published: 12 August 2022
(This article belongs to the Special Issue Recent Advances in Cancer Therapy)

Abstract

:
Phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB/AKT) and mechanistic target of rapamycin (mTOR) (PAM) pathways play important roles in breast tumorigenesis and confer worse prognosis in breast cancer patients. The inhibitors targeting three key nodes of these pathways, PI3K, AKT and mTOR, are continuously developed. For breast cancer patients to truly benefit from PAM pathway inhibitors, it is necessary to clarify the frequency and mechanism of abnormal alterations in the PAM pathway in different breast cancer subtypes, and further explore reliable biomarkers to identify the appropriate population for precision therapy. Some PI3K and mTOR inhibitors have been approved by regulatory authorities for the treatment of specific breast cancer patient populations, and many new-generation PI3K/mTOR inhibitors and AKT isoform inhibitors have also been shown to have good prospects for cancer therapy. This review summarizes the changes in the PAM signaling pathway in different subtypes of breast cancer, and the latest research progress about the biomarkers and clinical application of PAM-targeted inhibitors.

1. Introduction

Phosphatidylinositol 3-kinases (PI3Ks) are lipid kinases that can fall into three classes (I, II and III) in mammals [1]. It has been extensively studied for its important functions in physiology and diseases. In particular, class I PI3K is a well-studied subtype and has been confirmed to be associated with the occurrence and development of cancer [2]. Class I PI3Ks consist of a catalytic subunit p110 (p110α, p110β, p110γ or p110δ) encoded by PIK3CA, PIK3CB and PIK3CD, respectively, and a regulatory subunit p85 (p85α, p85β and p85γ) encoded by PIK3R1, PIK3R2 and PIK3R3, respectively (Figure 1). The class I PI3K, protein kinase B (PKB/AKT) and mechanistic target of rapamycin (mTOR) pathway (PAM pathway) display abnormal activation frequently in human cancer and plays a vital part in cell survival, proliferation, motility and metabolism (Figure 2) [3,4,5].
Different from class I PI3K(PIK3C1), class II/III PI3K (PIK3C2/3) mainly phosphorylates phosphatidylinositol to phosphatidylinositol 3-phosphate (PI3P), thereby regulating autophagy and vesicular trafficking [9,10]. Class II PI3K is also involved in angiogenesis and cell migration. In breast cancer, low expression of PIK3C2 seems to increase sensitivity to chemotherapeutic drugs [11]. However, PIK3C2 is less sensitive to classical PI3K inhibitors. It warrants further studies to develop selective PIK3C2 inhibitors [12,13]. In addition, PIK3C3 has been confirmed to regulate tumor cell proliferation by inducing autophagy and regulating iron metabolism [14]. Preclinical studies have shown that targeting PIK3C3 has anti-tumor activity in breast, colorectal and prostate cancer [15,16]. This review focuses on the well-studied class I PI3K and the related PAM pathway in breast cancer.
Abnormal enhancement of the PI3K/AKT/mTOR pathway often promotes excessive cell multiplication and resistance to apoptosis, and participates in the development and progression of various tumors [17]. Disturbance of the PAM pathway is particularly usual in breast cancer, with approximately 70% of breast cancer patients having alterations in this pathway [18,19]. A huge number of experiments in vitro and in vivo have shown that inhibiting key components of the PAM pathway can inhibit cancer cell proliferation and survival, and affect tumor microenvironment, angiogenesis, cancer metastasis and metabolism, thereby exerting anti-tumor effects and overcoming endocrine therapy resistance [20,21,22,23,24]. Many of small-molecule inhibitors targeting the PAM pathway have been tested in preclinical and clinical studies. Thus far, only a few PI3K and mTOR inhibitors have been approved for the treatment of breast cancer [25].

2. Changes of the PAM Pathway in Different Breast Cancer Subtypes

The mechanism of abnormal activation of the PAM pathways in breast cancer includes variations in key molecules, such as amplification or overexpression of RTKs (e.g., HER2/ERBB2) and KRAS (kirsten rat sarcoma viral oncogene) mutation [26]. PIK3CA, PIK3CB and PIK3R1 mutations are frequently detected in breast cancer [22,27,28,29]. There are common p110α (PIK3CA) variants in the acidic cluster of the helical domain (E542, E545, and Q546) and the histidine residue (H1047) in the kinase domain (Figure 1). In addition, AKT1 mutations and inactivation of tumor suppressor gene PTEN, TSC1/2 (Tuberous Sclerosis Complex 1/2) or INPP4B [18,30,31,32] are involved in the abnormal activation of the PAM pathways. Some of these changes can be prognostic factors or biomarkers for targeted therapy (Table 1).
The frequency of changes in the above-mentioned genes may vary among different subtypes of breast cancer (Table 1) [47,48,49,50,51,52,53]. Breast cancer can be roughly divided into four subtypes: luminal A (60–70%), luminal B (10–20%), HER2-enriched (13–15%) and triple-negative (10–15%) [52]. In estrogen receptor (ER)+ and HER2+ breast cancer, the most common mechanism of abnormal activation of the PAM pathway is PIK3CA mutation, accounting for 47% of cases of the ER+/HER2− (luminal A) subtype, 33% of the ER+/HER2+ (luminal B) subtype, 23%–39% of the ER-/HER2+ subtype and 8–25% of the triple-negative breast cancer (TNBC) subtype [33,34,36]. In advanced and metastatic breast cancer, PI3KCA mutations may lead to chemotherapy resistance and a poor prognosis. For HER2 positive breast cancer, PIK3CA mutations are associated with worse prognosis [37,54]. Among patients receiving neoadjuvant regimens containing docetaxel, carboplatin, trastuzumab and lapatinib, those with tumors with PIK3CA/ERBB family mutations seem to develop pathologic complete response (pCR) more than those with wild-type tumors [55]. For TNBC, the most common abnormal mechanism of PAM is PTEN inactivation or downregulation, accounting for 67% of cases [56,57]. In addition, mTOR hyperphosphorylation is associated with poor outcomes of patients with stage I/II TNBC [46]. Metaplastic breast cancer is a type of TNBC. Several studies found strong enrichment in mutations of PIK3CA/PIK3R1, P53 and PTEN, and aberrations of RAS-MAPK pathways in metaplastic breast cancer [38]. In addition, PDK1 amplification is present in 20–38% of all breast cancer subtypes and is involved in aberrant activation of the PAM pathway [35,45]. AKT1 activating mutations (E17K) are also observed in 7% of ER+ metastatic breast cancer patients [42]. Therefore, it is necessary to select corresponding biomarkers according to different breast cancer subtypes to guide targeted therapy and evaluate prognosis.
Inhibiting the key nodes in PAM pathway can exert anti-tumor effects [43,44]. In HR+ breast cancer, the activation of PI3K pathway by PIK3CA mutation promotes ligand-independent ER activation, which is one of the important mechanisms of endocrine therapy resistance [53,58,59,60]. Inhibition of PI3K can delay or reverse endocrine therapy resistance and improve patient prognosis. In addition, AKT activation may confer resistance to anticancer agents such as the ER antagonists tamoxifen and fulvestrant. Combination of AKT inhibitors with tamoxifen and fulvestrant may improve their effectiveness [61,62]. Pan-mTORC1/2 inhibitors can also reverse endocrine resistance, chemoresistance and radiation resistance [63]. For patients with HER2+ breast cancer, inhibition of PI3K or mTOR can become a new therapeutic regimen after secondary resistance to anti-HER2 therapy [64,65,66]. For TNBC, PAM inhibitors may have clinical benefits in PTEN-deficient population [67,68]. Owing to the complexity and interactions among the diverse components in PAM pathway, inhibition of a single target may result in compensatory changes in other targets. After inhibition of PI3K, the mTOR pathway can be abnormally activated to counteract this effect [69,70,71]. The mTOR inhibitor may promote the expression of insulin receptor substrates, which may upregulate the AKT pathway [72].
Multiple changes in PAM pathways may co-exist in breast cancer. For example, the co-existence of PIK3CA mutation, PTEN deletion and HER2 amplification is detected in breast cancer [2,73]. Hence, inhibition of a single target may not achieve anti-tumor effects in these circumstances. This not only suggests the molecular mechanism of drug resistance in breast cancer, but also the feasibility of combined therapy. Yang et al. have shown that temsirolimus (mTORC1 inhibitor) in combination with dactolisib (dual PI3K-mTOR inhibitor) or ZSTK474 (pan-PI3K inhibitor) can collectively inhibit cancer cell growth and overcome cellular resistance to temsirolimus [74]. Tang et al. also proved that using different PAM inhibitors at the same time can reach better anti-cancer effects [75].

3. Preclinical and Clinical Development of PAM Pathway Inhibitors in Different Subtypes of Breast Cancer

3.1. PAM Inhibitors for Treating ER+/HER2− Breast Cancer

About 70% of breast cancer is ER+ and HER2− [37]. Endocrine therapy is the standard regimen for these patients [25]. Abnormal activation of the PAM pathway is one of the important reasons of endocrine resistance [59], which can be overcome or reversed by targeting pathways’ components which activated during acquired drug resistance [76,77]. Therefore, PAM pathway inhibitors have been extensively studied in this population (Table 2). PAM pathway inhibitors are classified into PI3K, AKT, mTOR, and dual PI3K-mTOR inhibitors. Alpelisib (PI3K inhibitor) and everolimus (mTOR inhibitor) have been approved by the U.S. Food and Drug Administration (FDA) for the clinical treatment of breast cancer [25]. Many AKT and mTOR inhibitors have initially shown preclinical activity or are currently undergoing clinical trials.

3.1.1. PI3K Inhibitors for Treating ER+/HER2− Breast Cancer

PI3K inhibitors can fall into three categories. The first category includes pan-PI3K inhibitors, which non-selectively act on the ATP-binding pockets of all class I PI3K isoforms (p110α, p110β, p110γ, and p110δ) [89]. Both buparisib and pictilisib are pan-PI3K inhibitors. Phase III clinical trials show that, as the second-line treatment of ER+ and HER2− advanced or metastatic breast cancer patients, buparisib combined with fulvestrant can significantly improve PFS, while the most frequent grade 3–4 adverse events are elevated alanine aminotransferase, hyperglycemia, hypertension and fatigue [78,79]. These severe adverse reactions may result in discontinuation of the drug. Due to the high blood–brain barrier-penetrating properties of buparlisib, depression and anxiety are also common psychiatric side-effects. Pictilisib, another pan-PI3K inhibitor, did not result in a significant survival benefit [80,81]. However, these studies consider that combining PI3K inhibition with endocrine therapy is reasonable in patients with ER +/HER2− breast cancer [36,90] (Table 2).
The second class of specific PI3K inhibitors are represented by the drugs alpelisib (BYL719) and taselisib (GDC-0032). The ATP binding sites of type I PI3K are highly homologous. The different residues next to the ATP binding sites can be divided into the adjacent hinge region (four residues) and the variable region located at the p-loop. Non-conserved residues in these two regions are key to the subtype selectivity of pan-PI3K inhibitors. Alpelisib can form a dihydrogen bond with Q859 in the hinge region of PI3Kα, while other subunits at this site are too short to form the same structure, so it can selectively inhibit p110α [91]. So far, alpelisib is the only PI3K inhibitor approved by the FDA for breast cancer treatment. In the phase III SOLAR-1 trial, ER+/HER2− advanced breast cancer patients with relapsed or progressed disease after endocrine therapy received alpelisib and fulvestrant treatment. This trial showed that alpelisib plus fulvestrant demonstrated better overall efficacy compared with placebo (26.6% vs. 12.8%). Furthermore, in patients with PIK3CA mutation, combination of alpelisib and fulvestrant greatly prolonged the mPFS (p = 0.001, Table 2). In contrast, fulvestrant plus alpelisib had no PFS benefit (7.4 months vs. 5.6 months, HR: 0.85; 95% CI: 0.58–1.25) in the PIK3CA wild-type group [81,82]. The positive results of SOLAR-1 trial led to the approvement of alpelisib plus fulvestrant for treating advanced or metastatic breast cancer with ER expression and PIK3CA mutation [25]. The recently updated American Society of Clinical Oncology (ASCO) guideline recommended that patients with locally recurrent unresectable or metastatic hormone receptor positive and HER2-negative breast cancer should be subject to testing of PIK3CA mutations to determine their eligibility for treatment with the alpelisib plus fulvestrant [92]. The BYLieve phase II trial assessment of the effectiveness and security of alpelisib plus fulvestrant for patients treated with CDK4/6 inhibitors [83]. Other studies have attempted to use alpelisib for neoadjuvant treatment of breast cancer, but the results of the NEO-ORB phase II study showed that alpelisib combined with letrozole for ER+/HER2− and early-stage breast cancer had no additional clinical benefit [84]. Taselisib inhibits p110α, δ and γ, but is 30-fold less potent against p110β. The SANDPIPER phase III clinical trial evaluated the safety of taselisib plus fulvestrant for postmenopausal breast cancer with disease recurrence/progression during or after an aromatase inhibitor. This trial indicated that taselisib increased the frequency of grade 3–5 adverse events (16.4% in placebo arm vs. 49.5% in taselisib arm). Based on this trail, taselisib combined with fulvestrant was discontinued, suggesting that follow-up PI3K inhibitors should improve selectivity for key isoforms, not just selectivity [85]. Other compounds, such as GDC0077 [93], eganelisib and samotolisib are also in preclinical studies.
Dual PI3K-mTOR inhibitors can target the catalytic pockets of mTOR and PI3K enzymes based on structural similarity. Since mTOR inhibitors may enhance the PI3K/PDK1 axis, an inhibitor targeting both PI3K and mTOR may have better anti-cancer activity [94]. Gedatolisib (PF-05212384) is the represent drug of dual PI3K-mTOR inhibitors. Preclinical studies have demonstrated that gedatolisib combined with letrozole/palbociclib or fulvestrant/palbociclib has antitumor activity with manageable toxicity [86,95]. In addition, a phase I trial of samotolisib (dual PI3K-mTOR inhibitors) in combination with cyclin dependent kinase (CDK) inhibitors in ER+ breast cancer patients is ongoing [88]. Recent studies indicate that PI3K/mTOR inhibitors combined with paclitaxel can enhance tumor response to immunosuppressants and may provide a viable treatment for metastatic breast cancer. Therefore, dual target inhibitors in combination with immunotherapy will also be the focus of future research [87,88,96].

3.1.2. AKT Inhibitors for Treating ER+/HER2− Breast Cancer

There are different kinds of AKT inhibitors. Pan-AKT inhibitors bind to the ATP pocket of AKT1/2/3 and suppress their activity [97] (Table 3). Another kind of AKT inhibitor is allosteric inhibitor [98]. Allosteric Akt inhibitor such as MK-2206 is a kind of PH-domain dependent inhibitor [99]. A phase II trial (NCT01776008) showed that MK-2206 did not increase the efficacy of anastrozole monotherapy in patients with PIK3CA-mutated ER+ breast cancer [100].
Capivasertib (AZD5363) is another inhibitor of all three AKT isoforms [101]. A randomized study assessed the effects of capivasertib plus fulvestrant in ER+, HER2− advanced breast cancer patients resistant to endocrine therapy (FAKTION). This trial and its updated analysis showed that the addition of capivasertib to fulvestrant resulted in a significant improvement of progression-free survival, objective response rate (ORR) [102] and overall survival [103] in participants with aromatase inhibitor-resistant ER-positive, HER2−negative advanced breast cancer. Additionally, the expanded biomarker testing suggested that capivasertib was predominantly effective in patients with PI3K/AKT/PTEN pathway-altered tumors (38.9 vs. 20.0 months, p = 0.0047) [103]. The grade 3–4 adverse events were hypertension (capivasertib arm vs. placebo: 32% vs. 24%), diarrhea (14% vs. 4%) and rash (20% vs. 0) [102]. Further study and stricter monitoring and management of adverse reactions are needed. Moreover, a basket trial of capivasertib treatment of patients with AKT1 (E17K)-mutated tumors demonstrated an objective response rate of 33%, with clinical benefit in ER+/HER2− breast cancer [111]. Multiple preclinical studies have confirmed that the combination of PARP and PI3K/AKT pathway inhibitors has synergistic antitumor activity in breast cancer susceptibility gene (BRCA)-deficient cancer models [112]. Phase I trials of olaparib, a PARP inhibitor, and capivasertib in BRCA1/2 and non-BRCA1/2 mutated breast cancer patients are ongoing [113].

3.1.3. mTOR Inhibitors for Treating ER+/HER2− Breast Cancer

It is known that mTOR can phosphorylate ERα at ser118, making it insensitive to tamoxifen [71,114]. The BOLERO-2 study showed that the median progression-free survival in postmenopausal ER+/HER2− breast cancer patients resistant to aromatase inhibitor was improved by everolimus in combination with exemestane compared to placebo and exemestane [104]. These findings prompted the FDA to approve everolimus for the patients with ER+, HER2− advanced disease or its combination with exemestane for the treatment of relapse or progression after the use of nonsteroidal aromatase inhibitors in postmenopausal ER+/HER2− advanced breast cancer patients without visceral disease [25,105,106,108]. The European Medicines Agency (EMA) also approved everolimus for ER+/HER2− advanced breast cancer patients after failure of non-steroidal aromatase inhibitors treatment. Exploration of everolimus in the neoadjuvant offsetting for breast cancer has yielded initial results. A study of everolimus plus letrozole for preoperative neoadjuvant treatment of breast cancer was conducted. Everolimus combined with letrozole resulted in a superior response and inhibition of tumor proliferation than letrozole alone [110,115]. However, further studies are required to confirm the effectiveness and safety of the drug.
Sapanisertib (MLN0128) has dual specificity for the mTOR complex (mTORC1 and mTORC2), and is a new generation of ATP-competitive mTOR kinase inhibitors. A phase II study of sapanisertib in combination with exemestane or fulvestrant in postmenopausal women with previously treated everolimus-sensitive or-resistant breast cancer was conducted. It was well tolerated and showed significant clinical benefit [109]. However, follow-up research is necessary. Some studies have found that compensatory IGF signaling could reduce the effectiveness of mTOR inhibitor in combination with endocrine therapy [108]. Further studies may focus on the effects of combining IGF axis inhibitors and mTOR inhibitors.

3.2. PAM Inhibitors for Treating HER2+ Breast Cancer

HER2 is overexpressed in about 20–25% of breast cancers [52]. HER2−targeted therapy is the standard of care for these patients [26]. PAM pathway is one of the major signaling pathway downstream of HER2. Its abnormal activation, such as PIK3CA mutation and constitutively active AKT, is involved in the development of primary and secondary resistance to HER2− targeted therapy [116,117]. PAM pathway inhibitors may help restore tumor sensitivity to anti-HER2 therapy (Table 4), but the efficacy and safety need to be further explored.
Buparlisib is a pan-PI3K inhibitor [118,119]. A phase II trial of buparlisib plus trastuzumab in trastuzumab-resistant, HER2−positive advanced breast cancer patients was conducted. Similar to many pan-PI3K inhibitors, the common adverse reactions of buparlisib were diarrhea (54%) and nausea (48%). Unfortunately, this trial failed to demonstrate a benefit of addling buparlisib to trastuzumab [119]. Copanlisib (BAY80-6946), a PI3Kα/δ inhibitor, was shown to be synergistic with anti-HER2 therapy in trastuzumab-resistant breast cancer cells [125]. A phase Ib trial indicated that copanlisib in combination with trastuzumab was well-tolerated in HER2+ metastatic breast cancer patients [126]. In a phase I trial (BYL-719) for alpelisib in combination with T-DM1(Ado-trastuzumab emtansine) in trastuzumab-resistant and/or T-DM1-resistant, HER2+ and metastatic breast cancer patients, the CBR (CR + PR) in the entire patient population and patients with prior T-DM1 treatment was 71% and 60%, respectively [120]. To further improve the efficacy and safety, specific PI3K inhibitors are still in development. Of note, GDC0941 and XL-147(pan-PI3K inhibitors) or BEZ235 (dual PI3K/mTOR inhibitor) can increase HER2/3 expression thereby promoting RAS/MAPK (mitogen-activated protein kinases) signaling [127,128,129]. Hence, PAM inhibitors combined with MEK inhibitors may be useful to overcome drug resistance [130].
The combination of MK-2206 with standard neoadjuvant therapy given rise to pCR rates in breast cancer patients with HER2 over expression [121]. However, MK-2206 has not yet been developed further. AKT/mTOR is highly activated in lapatinib-resistant HER2+ breast cancer cells [131]. The mTOR inhibitor INK-128 can restore the sensitivity of lapatinib-resistant HER2+ breast cancer cells to TKIs [127]. Recent study indicates that everolimus combined with T-DM1 has a strong in vivo and in vitro antitumor effect on HER2 positive breast cancer [132]. In a phase II clinical trial, the mTOR inhibitor sirolimus combined with trastuzumab was well tolerated in patients with trastuzumab-resistant, HER2-positive and advanced breast cancer [124]. The phase III BOLERO-3 study of everolimus combined with vinorelbine and trastuzumab in HER2-positive, trastuzumab-resistant advanced breast cancer was conducted. Compared with the placebo arm, everolimus combined with vinorelbine and trastuzumab significantly prolonged the mPFS (5.78 vs. 7.0 month; p = 0.0067) [123]. The exploratory analysis found that patients with PIK3CA mutations, PTEN deletions or tumors with an overactive PI3K pathway could gain PFS benefit from everolimus [122]. However, further studies are still in progress [124].

3.3. PAM Inhibitors for Treating Triple-Negative Breast Cancer (TNBC)

TNBC has a high degree of malignancy and rapid metastasis [52]. Abnormal activation of the PAM pathway is also common in TNBC, especially the type of luminal androgen receptor (LAR) in the Fudan classification [133]. This population may benefit from PI3K-targeted therapy (Table 5). A trial of the pan-PI3K inhibitor buparlisib in metastatic TNBC patients was conducted. Although the downregulation of vital components in the PI3K pathway was observed in patients with stable disease, the trial failed to observe a clear objective response. Inhibition of PI3K alone may not be sufficient for treating TNBC [134]. A phase II study of the sequential treatment of metastatic TNBC with PI3K-α inhibitor serabelisib, cisplatin and nab-paclitaxel is underway. In addition, a phase I study of eganelisib (dual PI3K-mTOR inhibitor) for treating advanced or metastatic TNBC is under recruitment.
AKT inhibitors combined with paclitaxel have shown remarkable antitumor activity as first-line drugs in the treatment of metastatic breast cancer. In the phase II PAKT study of the AKT1-3 isoform inhibitor capivasertib combined with paclitaxel as a first-line treatment of TNBC, combination of capivasertib and paclitaxel significantly prolonged PFS and OS compared with paclitaxel alone (p = 0.04, Table 5). The difference in survival benefit was more obvious in patients with PIK3CA/AKT1/PTEN-altered tumors (9.3 vs. 3.7 months; p = 0.01) [135]. In the LOTU phase II study for first-line treatment of TNBC, the pan-AKT inhibitor GDC-0068 (Ipatasertib) also showed an advantage in prolonging PFS compared with paclitaxel alone (p = 0.037) [136]. Another ongoing phase II trial, FAIRLANE, is also exploring the efficacy and safety of GDC-0068 in combination with paclitaxel in patients with grade IA-IIIA TNBC [137]. Some studies have confirmed that suppressing AKT and/or p70S6K (p70 ribosomal protein S6 kinase) activation might synergize with paclitaxel [141]. LY2780301 is a dual inhibitor of p70S6K and AKT. The phase Ib/II TAKTIC trial aims to evaluate LY2780301 in combination with weekly paclitaxel for treating HER2-negative advanced breast cancer patients. The combination of LY2780301 and paclitaxel demonstrated ORR benefit, while the main grade 3–4 drug-related adverse events included neuropathy (8%) and ungual toxicity (25%) [138].
The mTOR inhibitor everolimus was also used in the neoadjuvant treatment of TNBC. However, compared with the placebo group, combination of everolimus with cisplatin and paclitaxel did not demonstrate CR benefit (pCR: 36% vs. 49%) [139]. A study of tamirolimus or everolimus in combination with doxorubicin and bevacizumab for the treatment of metastatic TNBC showed initial results. mTOR inhibitors prolonged the ORR to 21%, and improved the 6-month clinical benefit rate to 40% [140]. The objective response benefit was associated with PI3K pathway aberration (p = 0.04) [140]. The status of PTEN may be a biomarker of PAM inhibitors for TNBC [142].

4. Conclusions and Perspectives

The PI3K/AKT/mTOR pathway adjusts cell proliferation and metabolism. Abnormalities in key targets lead to over-activation of this pathway, which is involved in tumor development, progression and drug resistance. PAM pathway inhibitors may be reliable antitumor agents (Table 6). At present, the PI3K inhibitor alpelisib and mTOR inhibitor everolimus have been approved by the FDA and EMA for the treatment of advanced ER+ breast cancer. The new generation of AKT inhibitors capivasertib and the highly selective ATP-competitive mTOR kinase inhibitors sapanisertib and sirolimus remain to be extensively evaluated. Most PAM inhibitors have limited effectiveness due to weak isoform selection inhibition, feedback regulation of downstream pathways, and tandem interference with other signaling pathways. Perhaps stronger PI3K subtype-specific inhibitors such as GDC-0077(p110α inhibitor) may show a better antitumor efficacy. In addition, the novel AKT inhibitor INY-03-041, which is composed of Ipatasertib-NH2, a ten-hydrocarbon linker and a cereblon ligand lenalidomide, can target all three AKT protein for proteasomal degradation [143]. More and more AKT degraders have been developed [144]. Preclinical studies demonstrate that these AKT degraders can effectively suppress tumor growth [144]. It remains to know whether these compounds are tolerable and have superior efficacy in clinical setting.
In conclusion, different combination treatments may be considered for different breast cancer subtypes. In ER+ breast cancer patients, PAM inhibitors combined with CDK inhibitors or other anti-estrogen therapies are expected to overcome drug resistance and prolong survival. For breast cancer with HER2-overexpressed breast cancer, the combination of mTOR inhibitor with anti-HER2 drugs (trastuzumab, TDM-1) may be an alternative after the progression of second-line therapy. For TNBC, concurrent targeting PAM and other key pathway nodes (EGFR, MEK) may be of benefit. Moreover, it remains to evaluate the efficacy of combined treatment of breast cancer patients with immune checkpoint inhibitors and PAM inhibitors. Currently, the combination of PARP and AKT inhibitors has also shown prospective results in breast cancer patients with BRCA1/2 mutation [145,146]. Future studies should focus on improving subtype selectivity and reducing toxic side effects. The combination of different PAM pathway inhibitors, or a combination with immunotherapy drugs, may also be a strategy to further improve efficacy [147]. Screening for reliable biomarkers that predict the effectiveness of combination regiments is also critical.

Author Contributions

Conceptualization, T.L. and X.Z.; Supervision, H.Z. and P.H.; writing—original draft preparation, K.Z. and Y.W.; visualization, Y.F.; Writing—review and editing, T.L. and H.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Key Research and Development Project of Science and Technology Department of Sichuan Province, grant number 00402053A2231 and the 135 project for disciplines of excellence, West China Hospital, Sichuan University, grant number: ZYGD18012.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thanks to all authors for their contributions to this manuscript. Figure 2 was created with BioRender, thanks to BioRender (https://app.biorender.com/ (accessed on 26 June 2022).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Bilanges, B.; Posor, Y.; Vanhaesebroeck, B. PI3K isoforms in cell signalling and vesicle trafficking. Nat. Rev. Mol. Cell Biol. 2019, 20, 515–534. [Google Scholar] [CrossRef] [PubMed]
  2. Vanhaesebroeck, B.; Perry, M.W.D.; Brown, J.R.; André, F.; Okkenhaug, K. PI3K inhibitors are finally coming of age. Nat. Rev. Drug Discov. 2021, 20, 741–769. [Google Scholar] [CrossRef] [PubMed]
  3. Hoxhaj, G.; Manning, B.D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef] [PubMed]
  4. Arafeh, R.; Samuels, Y. PIK3CA in cancer: The past 30 years. Semin. Cancer Biol. 2019, 59, 36–49. [Google Scholar] [CrossRef] [PubMed]
  5. Mangé, A.; Coyaud, E.; Desmetz, C.; Laurent, E.; Béganton, B.; Coopman, P.; Raught, B.; Solassol, J. FKBP4 connects mTORC2 and PI3K to activate the PDK1/Akt-dependent cell proliferation signaling in breast cancer. Theranostics 2019, 9, 7003–7015. [Google Scholar] [CrossRef]
  6. Hua, H.; Kong, Q.; Yin, J.; Zhang, J.; Jiang, Y. Insulin-like growth factor receptor signaling in tumorigenesis and drug resistance: A challenge for cancer therapy. J. Hematol. Oncol. 2020, 13, 64. [Google Scholar] [CrossRef] [PubMed]
  7. Mayer, I.A.; Arteaga, C.L. The PI3K/AKT pathway as a target for cancer treatment. Annu. Rev. Med. 2016, 67, 11–28. [Google Scholar] [CrossRef]
  8. Yin, Y.; Hua, H.; Li, M.; Liu, S.; Kong, Q.; Shao, T.; Wang, J.; Luo, Y.; Wang, Q.; Luo, T.; et al. mTORC2 promotes type I insulin-like growth factor receptor and insulin receptor activation through the tyrosine kinase activity of mTOR. Cell Res. 2016, 26, 46–65. [Google Scholar] [CrossRef] [PubMed]
  9. Thoresen, S.B.; Pedersen, N.M.; Liestøl, K.; Stenmark, H. A phosphatidylinositol 3-kinase class III sub-complex containing VPS15, VPS34, Beclin 1, UVRAG and BIF-1 regulates cytokinesis and degradative endocytic traffic. Exp. Cell Res. 2010, 316, 3368–3378. [Google Scholar] [CrossRef] [PubMed]
  10. Posor, Y.; Eichhorn-Gruenig, M.; Puchkov, D.; Schöneberg, J.; Ullrich, A.; Lampe, A.; Müller, R.; Zarbakhsh, S.; Gulluni, F.; Hirsch, E.; et al. Spatiotemporal control of endocytosis by phosphatidylinositol -3, 4-bisphosphate. Nature 2013, 499, 233–237. [Google Scholar] [CrossRef] [PubMed]
  11. Gulluni, F.; Martini, M.; De Santis, M.C.; Campa, C.C.; Ghigo, A.; Margaria, J.P.; Ciraolo, E.; Franco, I.; Ala, U.; Annaratone, L.; et al. Mitotic spindle assembly and genomic stability in breast cancer require PI3K-C2α scaffolding function. Cancer Cell. 2017, 32, 444–459.e7. [Google Scholar] [CrossRef] [PubMed]
  12. Kroesbergen, J.; Roozen, A.M.; Gelsema, W.J.; de Ligny, C.L. 99mTc bone scanning agents--IV. Chemical characterization of 99mTc(Sn)-pyrophosphate complexes. Int. J. Rad. Appl. Instrum. B 1988, 15, 209–214. [Google Scholar] [CrossRef]
  13. Gulluni, F.; De Santis, M.C.; Margaria, J.P.; Martini, M.; Hirsch, E. Class II PI3K functions in cell biology and disease. Trends Cell Biol. 2019, 29, 339–359. [Google Scholar] [CrossRef] [PubMed]
  14. Jiang, X.; Bao, Y.; Liu, H.; Kou, X.; Zhang, Z.; Sun, F.; Qian, Z.; Lin, Z.; Li, X.; Liu, X.; et al. VPS34 stimulation of p62 phosphorylation for cancer progression. Oncogene 2017, 36, 6850–6862. [Google Scholar] [CrossRef] [PubMed]
  15. Young, C.D.; Arteaga, C.L.; Cook, R.S. Dual inhibition of type I and type III PI3 kinases increases tumor cell apoptosis in HER2+ breast cancers. Breast Cancer Res. 2015, 17, 148. [Google Scholar] [CrossRef] [PubMed]
  16. Chu, C.A.; Wang, Y.W.; Chen, Y.L.; Chen, H.W.; Chuang, J.J.; Chang, H.Y.; Ho, C.L.; Chang, C.; Chow, N.H.; Lee, C.T. The role of phosphatidylinositol 3-kinase catalytic subunit type 3 in the pathogenesis of human cancer. Int. J. Mol. Sci. 2021, 22, 10964. [Google Scholar] [CrossRef]
  17. Verret, B.; Cortes, J.; Bachelot, T.; Andre, F.; Arnedos, M. Efficacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol. 2019, 30 (Suppl. 10), x12–x20. [Google Scholar] [CrossRef]
  18. Hinz, N.; Jücker, M. Distinct functions of AKT isoforms in breast cancer: A comprehensive review. Cell Commun. Signal. 2019, 17, 154. [Google Scholar] [CrossRef]
  19. López-Knowles, E.; O’Toole, S.A.; McNeil, C.M.; Millar, E.K.; Qiu, M.R.; Crea, P.; Daly, R.J.; Musgrove, E.A.; Sutherland, R.L. PI3K pathway activation in breast cancer is associated with the basal-like phenotype and cancer-specific mortality. Int. J. Cancer 2010, 126, 1121–1131. [Google Scholar] [CrossRef]
  20. Zhang, J.; Liu, J.; Zhang, H.; Wang, J.; Hua, H.; Jiang, Y. The role of network-forming collagens in cancer progression. Int. J. Cancer 2022, 151, 833–842. [Google Scholar] [CrossRef]
  21. Nakanishi, Y.; Walter, K.; Spoerke, J.M.; O’Brien, C.; Huw, L.Y.; Hampton, G.M.; Lackner, M.R. Activating mutations in PIK3CB confer resistance to PI3K inhibition and define a novel oncogenic role for p110β. Cancer Res. 2016, 76, 1193–1203. [Google Scholar] [CrossRef] [PubMed]
  22. Jiang, Y.; Zhang, H.; Wang, J.; Liu, Y.; Luo, T.; Hua, H. Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J. Hematol. Oncol. 2022, 15, 34. [Google Scholar] [CrossRef] [PubMed]
  23. Manning, B.D.; Toker, A. AKT/PKB signaling: Navigating the network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [PubMed]
  24. Courtney, K.D.; Corcoran, R.B.; Engelman, J.A. The PI3K pathway as drug target in human cancer. J. Clin. Oncol. 2010, 28, 1075–1083. [Google Scholar] [CrossRef]
  25. Gradishar, W.J.; Moran, M.S.; Abraham, J.; Aft, R.; Agnese, D.; Allison, K.H.; Blair, S.L.; Burstein, H.J.; Dang, C.; Elias, A.D.; et al. Breast Cancer, Version 4.2021 Featured Updates to the NCCN Guidelines. JNCCN J. Natl. Compr. Cancer Netw. 2021, 19, 485–494. [Google Scholar] [CrossRef]
  26. Butti, R.; Das, S.; Gunasekaran, V.P.; Yadav, A.S.; Kumar, D.; Kundu, G.C. Receptor tyrosine kinases (RTKs) in breast cancer: Signaling, therapeutic implications and challenges. Mol. Cancer 2018, 17, 34. [Google Scholar] [CrossRef]
  27. Bachman, K.E.; Argani, P.; Samuels, Y.; Silliman, N.; Ptak, J.; Szabo, S.; Konishi, H.; Karakas, B.; Blair, B.G.; Lin, C.; et al. The PIK3CA gene is mutated with high frequency in human breast cancers. Cancer Biol. Ther. 2004, 3, 772–775. [Google Scholar] [CrossRef]
  28. Zhang, H.; Liu, G.; Dziubinski, M.; Yang, Z.; Ethier, S.P.; Wu, G. Comprehensive analysis of oncogenic effects of PIK3CA mutations in human mammary epithelial cells. Breast Cancer Res. Treat. 2008, 112, 217–227. [Google Scholar] [CrossRef]
  29. Kan, Z.; Jaiswal, B.S.; Stinson, J.; Janakiraman, V.; Bhatt, D.; Stern, H.M.; Yue, P.; Haverty, P.M.; Bourgon, R.; Zheng, J.; et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 2010, 466, 869–873. [Google Scholar] [CrossRef]
  30. Fortier, A.M.; Asselin, E.; Cadrin, M. Functional specificity of Akt isoforms in cancer progression. Biomol. Concepts 2011, 2, 1–11. [Google Scholar] [CrossRef]
  31. Rodon, J.; Dienstmann, R.; Serra, V.; Tabernero, J. Development of PI3K inhibitors: Lessons learned from early clinical trials. Nat. Rev. Clin. Oncol. 2013, 10, 143–153. [Google Scholar] [CrossRef] [PubMed]
  32. Costa, C.; Wang, Y.; Ly, A.; Hosono, Y.; Murchie, E.; Walmsley, C.S.; Huynh, T.; Healy, C.; Peterson, R.; Yanase, S.; et al. PTEN loss mediates clinical cross-resistance to CDK4/6 and PI3Kα inhibitors in breast cancer. Cancer Discov. 2020, 10, 72–85. [Google Scholar] [CrossRef] [PubMed]
  33. Stemke-Hale, K.; Gonzalez-Angulo, A.M.; Lluch, A.; Neve, R.M.; Kuo, W.L.; Davies, M.; Carey, M.; Hu, Z.; Guan, Y.; Sahin, A.; et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008, 68, 6084–6091. [Google Scholar] [CrossRef] [PubMed]
  34. Pérez-Tenorio, G.; Alkhori, L.; Olsson, B.; Waltersson, M.A.; Nordenskjöld, B.; Rutqvist, L.E.; Skoog, L.; Stål, O. PIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancer. Clin. Cancer Res. 2007, 13, 3577–3584. [Google Scholar] [CrossRef] [PubMed]
  35. Deng, L.; Chen, J.; Zhong, X.R.; Luo, T.; Wang, Y.P.; Huang, H.F.; Yin, L.J.; Qiu, Y.; Bu, H.; Lv, Q.; et al. Correlation between activation of PI3K/AKT/mTOR pathway and prognosis of breast cancer in Chinese women. PLoS ONE 2015, 10, e0120511. [Google Scholar] [CrossRef] [PubMed]
  36. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [PubMed]
  37. Miller, T.W.; Rexer, B.N.; Garrett, J.T.; Arteaga, C.L. Mutations in the phosphatidylinositol 3-kinase pathway: Role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res. 2011, 13, 224. [Google Scholar] [CrossRef]
  38. Coussy, F.; El Botty, R.; Lavigne, M.; Gu, C.; Fuhrmann, L.; Briaux, A.; de Koning, L.; Dahmani, A.; Montaudon, E.; Morisset, L.; et al. Combination of PI3K and MEK inhibitors yields durable remission in PDX models of PIK3CA-mutated metaplastic breast cancers. J. Hematol. Oncol. 2020, 13, 13. [Google Scholar] [CrossRef]
  39. Crowder, R.J.; Phommaly, C.; Tao, Y.; Hoog, J.; Luo, J.; Perou, C.M.; Parker, J.S.; Miller, M.A.; Huntsman, D.G.; Lin, L.; et al. PIK3CA and PIK3CB inhibition produce synthetic lethality when combined with estrogen deprivation in estrogen receptor-positive breast cancer. Cancer Res. 2009, 69, 3955–3962. [Google Scholar] [CrossRef]
  40. Chen, L.; Yang, L.; Yao, L.; Kuang, X.Y.; Zuo, W.J.; Li, S.; Qiao, F.; Liu, Y.R.; Cao, Z.G.; Zhou, S.L.; et al. Characterization of PIK3CA and PIK3R1 somatic mutations in Chinese breast cancer patients. Nat. Commun. 2018, 9, 1357. [Google Scholar] [CrossRef]
  41. Ellis, M.J.; Lin, L.; Crowder, R.; Tao, Y.; Hoog, J.; Snider, J.; Davies, S.; DeSchryver, K.; Evans, D.B.; Steinseifer, J.; et al. Phosphatidyl-inositol-3-kinase alpha catalytic subunit mutation and response to neoadjuvant endocrine therapy for estrogen receptor positive breast cancer. Breast Cancer Res. Treat. 2010, 119, 379–390. [Google Scholar] [CrossRef] [PubMed]
  42. Smyth, L.M.; Tamura, K.; Oliveira, M.; Ciruelos, E.M.; Mayer, I.A.; Sablin, M.P.; Biganzoli, L.; Ambrose, H.J.; Ashton, J.; Barnicle, A.; et al. Capivasertib, an AKT Kinase Inhibitor, as Monotherapy or in Combination with Fulvestrant in Patients with AKT1E17K-Mutant, ER-Positive Metastatic Breast Cancer. Clin. Cancer Res. 2020, 26, 3947–3957. [Google Scholar] [CrossRef] [PubMed]
  43. Loi, S.; Haibe-Kains, B.; Majjaj, S.; Lallemand, F.; Durbecq, V.; Larsimont, D.; Gonzalez-Angulo, A.M.; Pusztai, L.; Symmans, W.F.; Bardelli, A.; et al. PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 10208–10213. [Google Scholar] [CrossRef]
  44. Kalinsky, K.; Jacks, L.M.; Heguy, A.; Patil, S.; Drobnjak, M.; Bhanot, U.K.; Hedvat, C.V.; Traina, T.A.; Solit, D.; Gerald, W.; et al. PIK3CA mutation associates with improved outcome in breast cancer. Clin. Cancer Res. 2009, 15, 5049–5059. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, N.; Fu, J.; Li, Z.; Jiang, N.; Chen, Y.; Peng, J. The landscape of PDK1 in breast cancer. Cancers 2022, 14, 811. [Google Scholar] [CrossRef] [PubMed]
  46. Dai, M.; Yan, G.; Wang, N.; Daliah, G.; Edick, A.M.; Poulet, S.; Boudreault, J.; Ali, S.; Burgos, S.A.; Lebrun, J.J. In vivo genome-wide CRISPR screen reveals breast cancer vulnerabilities and synergistic mTOR/Hippo targeted combination therapy. Nat. Commun. 2021, 12, 3055. [Google Scholar] [CrossRef]
  47. Dong, C.; Wu, J.; Chen, Y.; Nie, J.; Chen, C. Activation of PI3K/AKT/mTOR pathway causes drug resistance in breast cancer. Front. Pharmacol. 2021, 12, 628690. [Google Scholar] [CrossRef]
  48. Gonzalez-Angulo, A.M.; Ferrer-Lozano, J.; Stemke-Hale, K.; Sahin, A.; Liu, S.; Barrera, J.A.; Burgues, O.; Lluch, A.M.; Chen, H.; Hortobagyi, G.N.; et al. PI3K pathway mutations and PTEN levels in primary and metastatic breast cancer. Mol. Cancer Ther. 2011, 10, 1093–1101. [Google Scholar] [CrossRef]
  49. Graudenzi, A.; Cava, C.; Bertoli, G.; Fromm, B.; Flatmark, K.; Mauri, G.; Castiglioni, I. Pathway-based classification of breast cancer subtypes. Front. Biosci. (Landmark Ed.) 2017, 22, 1697–1712. [Google Scholar] [CrossRef]
  50. Banerji, S.; Cibulskis, K.; Rangel-Escareno, C.; Brown, K.K.; Carter, S.L.; Frederick, A.M.; Lawrence, M.S.; Sivachenko, A.Y.; Sougnez, C.; Zou, L.; et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 2012, 486, 405–409. [Google Scholar] [CrossRef]
  51. Fujimoto, Y.; Morita, T.Y.; Ohashi, A.; Haeno, H.; Hakozaki, Y.; Fujii, M.; Kashima, Y.; Kobayashi, S.S.; Mukohara, T. Combination treatment with a PI3K/Akt/mTOR pathway inhibitor overcomes resistance to anti-HER2 therapy in PIK3CA-mutant HER2-positive breast cancer cells. Sci. Rep. 2020, 10, 21762. [Google Scholar] [CrossRef] [PubMed]
  52. Harbeck, N.; Penault-Llorca, F.; Cortes, J. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  53. Ng, C.K.; Piscuoglio, S.; Geyer, F.C.; Burke, K.A.; Pareja, F.; Eberle, C.A.; Lim, R.S.; Natrajan, R.; Riaz, N.; Mariani, O.; et al. The Landscape of somatic genetic alterations in metaplastic breast carcinomas. Clin. Cancer Res. 2017, 23, 3859–3870. [Google Scholar] [CrossRef] [PubMed]
  54. Shen, L.S.; Jin, X.Y.; Wang, X.M.; Tou, L.Z.; Huang, J. Advances in endocrine and targeted therapy for hormone-receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer. Chin. Med. J. 2020, 133, 1099–1108. [Google Scholar] [CrossRef]
  55. Toomey, S.; Eustace, A.J.; Fay, J.; Sheehan, K.M.; Carr, A.; Milewska, M.; Madden, S.F.; Teiserskiene, A.; Kay, E.W.; O’Donovan, N.; et al. Impact of somatic PI3K pathway and ERBB family mutations on pathological complete response (pCR) in HER2-positive breast cancer patients who received neoadjuvant HER2-targeted therapies. Breast Cancer Res. 2017, 19, 87. [Google Scholar] [CrossRef]
  56. Ertay, A.; Liu, H.; Liu, D.; Peng, P.; Hill, C.; Xiong, H.; Hancock, D.; Yuan, X.; Przewloka, M.R.; Coldwell, M.; et al. WDHD1 is essential for the survival of PTEN-inactive triple-negative breast cancer. Cell Death Dis. 2020, 11, 1001. [Google Scholar] [CrossRef]
  57. Shoman, N.; Klassen, S.; McFadden, A.; Bickis, M.G.; Torlakovic, E.; Chibbar, R. Reduced PTEN expression predicts relapse in patients with breast carcinoma treated by tamoxifen. Mod. Pathol. 2005, 18, 250–259. [Google Scholar] [CrossRef]
  58. Bosch, A.; Li, Z.; Bergamaschi, A.; Ellis, H.; Toska, E.; Prat, A.; Tao, J.J.; Spratt, D.E.; Viola-Villegas, N.T.; Castel, P.; et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer. Sci. Transl. Med. 2015, 7, 283ra51. [Google Scholar] [CrossRef]
  59. Hanker, A.B.; Sudhan, D.R.; Arteaga, C.L. Overcoming endocrine resistance in breast cancer. Cancer Cell 2020, 37, 496–513. [Google Scholar] [CrossRef]
  60. Li, D.; Ji, H.; Niu, X.; Yin, L.; Wang, Y.; Gu, Y.; Wang, J.; Zhou, X.; Zhang, H.; Zhang, Q. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 2020, 111, 47–58. [Google Scholar] [CrossRef] [PubMed]
  61. Hua, H.; Zhang, H.; Chen, J.; Wang, J.; Liu, J.; Jiang, Y. Targeting Akt in cancer for precision therapy. J. Hematol. Oncol. 2021, 14, 128. [Google Scholar] [CrossRef] [PubMed]
  62. Hua, H.; Kong, Q.; Zhang, H.; Wang, J.; Luo, T.; Jiang, Y. Targeting mTOR for cancer therapy. J. Hematol. Oncol. 2019, 12, 71. [Google Scholar] [CrossRef] [PubMed]
  63. Dey, N.; De, P.; Leyland-Jones, B. PI3K-AKT-mTOR inhibitors in breast cancers: From tumor cell signaling to clinical trials. Pharmacol. Ther. 2017, 175, 91–106. [Google Scholar] [CrossRef] [PubMed]
  64. Howlader, N.; Altekruse, S.F.; Li, C.I.; Chen, V.W.; Clarke, C.A.; Ries, L.A.; Cronin, K.A. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J. Natl. Cancer Inst. 2014, 106, dju055. [Google Scholar] [CrossRef] [PubMed]
  65. García-García, C.; Ibrahim, Y.H.; Serra, V.; Calvo, M.T.; Guzmán, M.; Grueso, J.; Aura, C.; Pérez, J.; Jessen, K.; Liu, Y.; et al. Dual mTORC1/2 and HER2 blockade results in antitumor activity in preclinical models of breast cancer resistant to anti-HER2 therapy. Clin. Cancer Res. 2012, 18, 2603–2612. [Google Scholar] [CrossRef] [PubMed]
  66. Khan, M.A.; Jain, V.K.; Rizwanullah, M.; Ahmad, J.; Jain, K. PI3K/AKT/mTOR pathway inhibitors in triple-negative breast cancer: A review on drug discovery and future challenges. Drug Discov. Today 2019, 24, 2181–2191. [Google Scholar] [CrossRef]
  67. Pascual, J.; Turner, N.C. Targeting the PI3-kinase pathway in triple-negative breast cancer. Ann. Oncol. 2019, 30, 1051–1060. [Google Scholar] [CrossRef]
  68. Denkert, C.; Liedtke, C.; Tutt, A.; von Minckwitz, G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet 2017, 389, 2430–2442. [Google Scholar] [CrossRef]
  69. Elkabets, M.; Vora, S.; Juric, D.; Morse, N.; Mino-Kenudson, M.; Muranen, T.; Tao, J.; Campos, A.B.; Rodon, J.; Ibrahim, Y.H.; et al. mTORC1 inhibition is required for sensitivity to PI3K p110α inhibitors in PIK3CA-mutant breast cancer. [published correction appears in Sci Transl Med. 2018 Nov 14;10,]. Sci. Transl. Med. 2013, 5, 196ra99. [Google Scholar] [CrossRef]
  70. Petrossian, K.; Nguyen, D.; Lo, C.; Kanaya, N.; Somlo, G.; Cui, Y.X.; Huang, C.S.; Chen, S. Use of dual mTOR inhibitor MLN0128 against everolimus-resistant breast cancer. Breast Cancer Res. Treat. 2018, 170, 499–506. [Google Scholar] [CrossRef]
  71. Yang, H.; Rudge, D.G.; Koos, J.D.; Vaidialingam, B.; Yang, H.J.; Pavletich, N.P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497, 217–223. [Google Scholar] [CrossRef] [PubMed]
  72. O’Reilly, K.E.; Rojo, F.; She, Q.B.; Solit, D.; Mills, G.B.; Smith, D.; Lane, H.; Hofmann, F.; Hicklin, D.J.; Ludwig, D.L.; et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006, 66, 1500–1508. [Google Scholar] [CrossRef] [PubMed]
  73. Endicott, S.J.; Ziemba, Z.J.; Beckmann, L.J.; Boynton, D.N.; Miller, R.A. Inhibition of class I PI3K enhances chaperone-mediated autophagy. J. Cell Biol. 2020, 219, e202001031. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, S.; Xiao, X.; Meng, X.; Leslie, K.K. A mechanism for synergy with combined mTOR and PI3 kinase inhibitors. PLoS ONE 2011, 6, e26343. [Google Scholar] [CrossRef] [PubMed]
  75. Tang, K.D.; Ling, M.T. Targeting drug-resistant prostate cancer with dual PI3K/mTOR inhibition. Curr. Med. Chem. 2014, 21, 3048–3056. [Google Scholar] [CrossRef]
  76. O’Brien, N.A.; McDermott, M.S.; Conklin, D.; Luo, T.; Ayala, R.; Salgar, S.; Chau, K.; DiTomaso, E.; Babbar, N.; Su, F.; et al. Targeting activated PI3K/mTOR signaling overcomes acquired resistance to CDK4/6-based therapies in preclinical models of hormone receptor-positive breast cancer. Breast Cancer Res. 2020, 22, 89. [Google Scholar] [CrossRef]
  77. Boulay, A.; Rudloff, J.; Ye, J.; Zumstein-Mecker, S.; O’Reilly, T.; Evans, D.B.; Chen, S.; Lane, H.A. Dual inhibition of mTOR and estrogen receptor signaling in vitro induces cell death in models of breast cancer. Clin. Cancer Res. 2005, 11, 5319–5328. [Google Scholar] [CrossRef]
  78. Baselga, J.; Im, S.A.; Iwata, H.; Cortés, J.; De Laurentiis, M.; Jiang, Z.; Arteaga, C.L.; Jonat, W.; Clemons, M.; Ito, Y.; et al. Buparlisib plus fulvestrant versus placebo plus fulvestrant in postmenopausal, hormone receptor-positive, HER2-negative, advanced breast cancer (BELLE-2): A randomised, double-blind, placebo-controlled, phase 3 trial. [published correction appears in Lancet Oncol. 2019 Feb;20, e71–e72]. Lancet Oncol. 2017, 18, 904–916. [Google Scholar] [CrossRef]
  79. Di Leo, A.; Johnston, S.; Lee, K.S.; Ciruelos, E.; Lønning, P.E.; Janni, W.; O’Regan, R.; Mouret-Reynier, M.A.; Kalev, D.; Egle, D.; et al. Buparlisib plus fulvestrant in postmenopausal women with hormone- receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018, 19, 87–100. [Google Scholar] [CrossRef]
  80. Krop, I.E.; Mayer, I.A.; Ganju, V.; Dickler, M.; Johnston, S.; Morales, S.; Yardley, D.A.; Melichar, B.; Forero-Torres, A.; Lee, S.C.; et al. Pictilisib for oestrogen receptor-positive, aromatase inhibitor-resistant, advanced or metastatic breast cancer (FERGI): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2016, 17, 811–821. [Google Scholar] [CrossRef]
  81. Vuylsteke, P.; Huizing, M.; Petrakova, K.; Roylance, R.; Laing, R.; Chan, S.; Abell, F.; Gendreau, S.; Rooney, I.; Apt, D.; et al. Pictilisib PI3Kinase inhibitor (a phosphatidylinositol 3-kinase [PI3K] inhibitor) plus paclitaxel for the treatment of hormone receptor-positive, HER2-negative, locally recurrent, or metastatic breast cancer: Interim analysis of the multicentre, placebo-controlled, phase II randomised PEGGY study. Ann. Oncol. 2016, 27, 2059–2066. [Google Scholar] [CrossRef] [PubMed]
  82. André, F.; Ciruelos, E.M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I.A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.S.; et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from SOLAR-1. Ann. Oncol. 2021, 32, 208–217. [Google Scholar] [CrossRef]
  83. Rugo, H.S.; Lerebours, F.; Ciruelos, E.; Drullinsky, P.; Ruiz-Borrego, M.; Neven, P.; Park, Y.H.; Prat, A.; Bachelot, T.; Juric, D.; et al. Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): One cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol. 2021, 22, 489–498. [Google Scholar] [CrossRef]
  84. Mayer, I.A.; Prat, A.; Egle, D.; Blau, S.; Fidalgo, J.; Gnant, M.; Fasching, P.A.; Colleoni, M.; Wolff, A.C.; Winer, E.P.; et al. A Phase II Randomized study of neoadjuvant letrozole plus alpelisib for hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer (NEO-ORB). Clin. Cancer Res. 2019, 25, 2975–2987. [Google Scholar] [CrossRef] [PubMed]
  85. Dent, S.; Cortés, J.; Im, Y.H.; Diéras, V.; Harbeck, N.; Krop, I.E.; Wilson, T.R.; Cui, N.; Schimmoller, F.; Hsu, J.Y.; et al. Phase III randomized study of taselisib or placebo with fulvestrant in estrogen receptor-positive, PIK3CA-mutant, HER2-negative, advanced breast cancer: The SANDPIPER trial. Ann. Oncol. 2021, 32, 197–207. [Google Scholar] [CrossRef] [PubMed]
  86. Shor, R.E.; Dai, J.; Lee, S.Y.; Pisarsky, L.; Matei, I.; Lucotti, S.; Lyden, D.; Bissell, M.J.; Ghajar, C.M. The PI3K/mTOR inhibitor Gedatolisib eliminates dormant breast cancer cells in organotypic culture, but fails to prevent metastasis in preclinical settings. Mol. Oncol. 2022, 16, 130–147. [Google Scholar] [CrossRef]
  87. Clinical Trails.gov. Study of the Safety and Pharmacology of GDC-0980 in Combination with Paclitaxel with or without Bevacizumab in Patients with Locally Recurrent or Metastatic Breast Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT01254526 (accessed on 26 March 2021).
  88. Clinical Trails.gov. A Study of LY2835219 (Abemaciclib) in Combination with Therapies for Breast Cancer That Has Spread. Available online: https://clinicaltrials.gov/ct2/show/NCT02057133 (accessed on 26 March 2021).
  89. Qin, H.; Liu, L.; Sun, S.; Zhang, D.; Sheng, J.; Li, B.; Yang, W. The impact of PI3K inhibitors on breast cancer cell and its tumor microenvironment. PeerJ 2018, 6, e5092. [Google Scholar] [CrossRef]
  90. Markham, A. Alpelisib: First global approval. Drugs 2019, 79, 1249–1253. [Google Scholar] [CrossRef]
  91. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef]
  92. Henry, N.L.; Somerfield, M.R.; Dayao, Z.; Elias, A.; Kalinsky, K.; McShane, L.M.; Moy, B.; Park, B.H.; Shanahan, K.M.; Sharma, P.; et al. Biomarkers for systemic therapy in metastatic breast cancer: ASCO guideline update. J. Clin. Oncol. 2022, JCO-22. [Google Scholar] [CrossRef]
  93. Song, K.W.; Edgar, K.A.; Hanan, E.J.; Hafner, M.; Oeh, J.; Merchant, M.; Sampath, D.; Nannini, M.A.; Hong, R.; Phu, L.; et al. RTK-dependent inducible degradation of mutant PI3Kα drives GDC-0077 (Inavolisib) efficacy. Cancer Discov. 2022, 12, 204–219. [Google Scholar] [CrossRef] [PubMed]
  94. Kumar, S.; Bhattacharyya, S.; Das, A.; Singh, G.; Bal, A. In vitro effect of PIK3CA/mTOR inhibition in triple-negative breast cancer subtype cell lines. Breast Dis. 2022, 41, 241–247. [Google Scholar] [CrossRef] [PubMed]
  95. Shah, N.; Mohammad, A.S.; Saralkar, P.; Sprowls, S.A.; Vickers, S.D.; John, D.; Tallman, R.M.; Lucke-Wold, B.P.; Jarrell, K.E.; Pinti, M.; et al. Investigational chemotherapy and novel pharmacokinetic mechanisms for the treatment of breast cancer brain metastases. Pharmacol. Res. 2018, 132, 47–68. [Google Scholar] [CrossRef]
  96. Yan, C.; Yang, J.; Saleh, N.; Chen, S.C.; Ayers, G.D.; Abramson, V.G.; Mayer, I.A.; Richmond, A. Inhibition of the PI3K/mTOR pathway in breast cancer to enhance response to immune checkpoint inhibitors in breast cancer. Int. J. Mol. Sci. 2021, 22, 5207. [Google Scholar] [CrossRef] [PubMed]
  97. Halder, A.K.; Cordeiro, M.N.D.S. AKT Inhibitors: The road ahead to computational modeling-guided discovery. Int. J. Mol. Sci. 2021, 22, 3944. [Google Scholar] [CrossRef]
  98. Xiang, R.F.; Wang, Y.; Zhang, N.; Xu, W.B.; Cao, Y.; Tong, J.; Li, J.M.; Wu, Y.L.; Yan, H. MK2206 enhances the cytocidal effects of bufalin in multiple myeloma by inhibiting the AKT/mTOR pathway. Cell Death Dis. 2017, 8, e2776. [Google Scholar] [CrossRef]
  99. Akcakanat, A.; Meric-Bernstam, F. MK-2206 window of opportunity study in breast cancer. Ann. Transl. Med. 2018, 6 (Suppl. S1), S57. [Google Scholar] [CrossRef]
  100. Ma, C.X.; Suman, V.; Goetz, M.P.; Northfelt, D.; Burkard, M.E.; Ademuyiwa, F.; Naughton, M.; Margenthaler, J.; Aft, R.; Gray, R.; et al. A Phase II trial of neoadjuvant MK-2206, an AKT inhibitor, with Anastrozole in clinical stage II or III PIK3CA-mutant ER-positive and HER2-negative breast cancer. Clin. Cancer Res. 2017, 23, 6823–6832. [Google Scholar] [CrossRef]
  101. Turner, N.C.; Alarcón, E.; Armstrong, A.C.; Philco, M.; López Chuken, Y.A.; Sablin, M.P.; Tamura, K.; Gómez Villanueva, A.; Pérez-Fidalgo, J.A.; Cheung, S.; et al. BEECH: A dose-finding run-in followed by a randomised phase II study assessing the efficacy of AKT inhibitor capivasertib (AZD5363) combined with paclitaxel in patients with estrogen receptor-positive advanced or metastatic breast cancer, and in a PIK3CA mutant sub-population. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 774–780. [Google Scholar] [CrossRef]
  102. Jones, R.H.; Casbard, A.; Carucci, M.; Cox, C.; Butler, R.; Alchami, F.; Madden, T.A.; Bale, C.; Bezecny, P.; Joffe, J.; et al. Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive breast cancer (FAKTION): A multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2020, 21, 345–357. [Google Scholar] [CrossRef]
  103. Howell, S.J.; Casbard, A.; Carucci, M.; Ingarfield, K.; Butler, R.; Morgan, S.; Meissner, M.; Bale, C.; Bezecny, P.; Moon, S.; et al. Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive, HER2-negative breast cancer (FAKTION): Overall survival, updated progression-free survival, and expanded biomarker analysis from a randomised, phase 2 trial. Lancet Oncol. 2022, 23, 851–864. [Google Scholar] [CrossRef] [PubMed]
  104. Prat, A.; Brase, J.C.; Cheng, Y.; Nuciforo, P.; Paré, L.; Pascual, T.; Martínez, D.; Galván, P.; Vidal, M.; Adamo, B.; et al. Everolimus plus exemestane for hormone receptor-positive advanced breast cancer: A PAM50 intrinsic subtype analysis of BOLERO-2. Oncologist 2019, 24, 893–900. [Google Scholar] [CrossRef] [PubMed]
  105. Schmid, P.; Zaiss, M.; Harper-Wynne, C.; Ferreira, M.; Dubey, S.; Chan, S.; Makris, A.; Nemsadze, G.; Brunt, A.M.; Kuemmel, S.; et al. Fulvestrant plus vistusertib vs fulvestrant plus everolimus vs fulvestrant alone for women with hormone receptor-positive metastatic breast cancer: The MANTA phase 2 randomized clinical trial. JAMA Oncol. 2019, 5, 1556–1564. [Google Scholar] [CrossRef] [PubMed]
  106. Kornblum, N.; Zhao, F.; Manola, J.; Klein, P.; Ramaswamy, B.; Brufsky, A.; Stella, P.J.; Burnette, B.; Telli, M.; Makower, D.F.; et al. Randomized phase II trial of fulvestrant plus everolimus or placebo in postmenopausal women with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer resistant to aromatase inhibitor therapy: Results of PrE0102. J. Clin. Oncol. 2018, 36, 1556–1563. [Google Scholar] [CrossRef] [PubMed]
  107. Jerusalem, G.; de Boer, R.H.; Hurvitz, S.; Yardley, D.A.; Kovalenko, E.; Ejlertsen, B.; Blau, S.; Özgüroğlu, M.; Landherr, L.; Ewertz, M.; et al. Everolimus plus exemestane vs everolimus or capecitabine monotherapy for estrogen receptor-positive, HER2-negative advanced breast cancer: The BOLERO-6 Randomized Clinical Trial. JAMA Oncol. 2018, 4, 1367–1374. [Google Scholar] [CrossRef]
  108. Schmid, P.; Sablin, M.P.; Bergh, J.; Im, S.A.; Lu, Y.S.; Martínez, N.; Neven, P.; Lee, K.S.; Morales, S.; Pérez-Fidalgo, J.A.; et al. A phase Ib/II study of xentuzumab, an IGF-neutralising antibody, combined with exemestane and everolimus in hormone receptor-positive, HER2-negative locally advanced/metastatic breast cancer. Breast Cancer Res. 2021, 23, 8. [Google Scholar] [CrossRef] [PubMed]
  109. Lim, B.; Potter, D.A.; Salkeni, M.A.; Silverman, P.; Haddad, T.C.; Forget, F.; Awada, A.; Canon, J.L.; Danso, M.; Lortholary, A.; et al. Sapanisertib plus exemestane or fulvestrant in women with hormone receptor-positive/HER2-negative advanced or metastatic breast cancer. Clin. Cancer Res. 2021, 27, 3329–3338. [Google Scholar] [CrossRef]
  110. Wolff, A.C.; Lazar, A.A.; Bondarenko, I.; Garin, A.M.; Brincat, S.; Chow, L.; Sun, Y.; Neskovic-Konstantinovic, Z.; Guimaraes, R.C.; Fumoleau, P.; et al. Randomized phase III placebo-controlled trial of letrozole plus oral temsirolimus as first-line endocrine therapy in postmenopausal women with locally advanced or metastatic breast cancer. J. Clin. Oncol. 2013, 31, 195–202. [Google Scholar] [CrossRef]
  111. Kalinsky, K.; Hong, F.; McCourt, C.K.; Sachdev, J.C.; Mitchell, E.P.; Zwiebel, J.A.; Doyle, L.A.; McShane, L.M.; Li, S.; Gray, R.J.; et al. Effect of capivasertib in patients with an AKT1 E17K-mutated tumor: NCI-MATCH subprotocol EAY131-Y nonrandomized trial. JAMA Oncol. 2021, 7, 271–278. [Google Scholar] [CrossRef]
  112. Hosford, S.R.; Miller, T.W. Clinical potential of novel therapeutic targets in breast cancer: CDK4/6, Src, JAK/STAT, PARP, HDAC, and PI3K/AKT/mTOR pathways. Pharmgenom. Pers. Med. 2014, 7, 203–215. [Google Scholar] [CrossRef]
  113. Yap, T.A.; Kristeleit, R.; Michalarea, V.; Pettitt, S.J.; Lim, J.S.; Carreira, S.; Roda, D.; Miller, R.; Riisnaes, R.; Miranda, S.; et al. Phase I trial of the PARP inhibitor olaparib and AKT inhibitor capivasertib in patients with BRCA1/2- and non-BRCA1/2-mutant cancers. Cancer Discov. 2020, 10, 1528–1543. [Google Scholar] [CrossRef] [PubMed]
  114. Oshiro, N.; Yoshino, K.I.; Hidayat, S.; Tokunaga, C.; Hara, K.; Eguchi, S.; Avruch, J.; Yonezawa, K. Dissociation of raptor from mTOR is a mechanism of rapamycin-induced inhibition of mTOR function. Genes Cells 2004, 9, 359–366. [Google Scholar] [CrossRef] [PubMed]
  115. Ballhausen, A.; Wheler, J.J.; Karp, D.D.; Piha-Paul, S.A.; Fu, S.; Pant, S.; Tsimberidou, A.M.; Hong, D.S.; Subbiah, V.; Holley, V.R.; et al. Phase I study of everolimus, letrozole, and trastuzumab in patients with hormone receptor-positive metastatic breast cancer or other solid tumors. Clin. Cancer Res. 2021, 27, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  116. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef] [PubMed]
  117. Chandarlapaty, S.; Sakr, R.A.; Giri, D.; Patil, S.; Heguy, A.; Morrow, M.; Modi, S.; Norton, L.; Rosen, N.; Hudis, C.; et al. Frequent mutational activation of the PI3K-AKT pathway in trastuzumab-resistant breast cancer. Clin. Cancer Res. 2012, 18, 6784–6791. [Google Scholar] [CrossRef]
  118. Pistilli, B.; Pluard, T.; Urruticoechea, A.; Farci, D.; Kong, A.; Bachelot, T.; Chan, S.; Han, H.S.; Jerusalem, G.; Urban, P.; et al. Phase II study of buparlisib (BKM120) and trastuzumab in patients with HER2+ locally advanced or metastatic breast cancer resistant to trastuzumab-based therapy. Breast Cancer Res. Treat. 2018, 168, 357–364. [Google Scholar] [CrossRef]
  119. Guerin, M.; Rezai, K.; Isambert, N.; Campone, M.; Autret, A.; Pakradouni, J.; Provansal, M.; Camerlo, J.; Sabatier, R.; Bertucci, F.; et al. PIKHER2: A phase IB study evaluating buparlisib in combination with lapatinib in trastuzumab-resistant HER2-positive advanced breast cancer. Eur. J. Cancer 2017, 86, 28–36. [Google Scholar] [CrossRef]
  120. Jain, S.; Shah, A.N.; Santa-Maria, C.A.; Siziopikou, K.; Rademaker, A.; Helenowski, I.; Cristofanilli, M.; Gradishar, W.J. Phase I study of alpelisib (BYL-719) and trastuzumab emtansine (T-DM1) in HER2-positive metastatic breast cancer (MBC) after trastuzumab and taxane therapy. Breast Cancer Res. Treat. 2018, 171, 371–381. [Google Scholar] [CrossRef] [PubMed]
  121. Chien, A.J.; Tripathy, D.; Albain, K.S.; Symmans, W.F.; Rugo, H.S.; Melisko, M.E.; Wallace, A.M.; Schwab, R.; Helsten, T.; Forero-Torres, A.; et al. MK-2206 and standard neoadjuvant chemotherapy improves response in patients with human epidermal growth factor receptor 2-positive and/or hormone receptor-negative breast bancers in the I-SPY 2 trial. J. Clin. Oncol. 2020, 38, 1059–1069. [Google Scholar] [CrossRef]
  122. André, F.; Hurvitz, S.; Fasolo, A.; Tseng, L.M.; Jerusalem, G.; Wilks, S.; O’Regan, R.; Isaacs, C.; Toi, M.; Burris, H.A.; et al. Molecular alterations and everolimus efficacy in human epidermal growth factor receptor 2-overexpressing metastatic breast cancers: Combined exploratory biomarker analysis from BOLERO-1 and BOLERO-3. J. Clin. Oncol. 2016, 34, 2115–2124. [Google Scholar] [CrossRef]
  123. André, F.; O’Regan, R.; Ozguroglu, M.; Toi, M.; Xu, B.; Jerusalem, G.; Masuda, N.; Wilks, S.; Arena, F.; Isaacs, C.; et al. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014, 15, 580–591. [Google Scholar] [CrossRef]
  124. Acevedo-Gadea, C.; Hatzis, C.; Chung, G.; Fishbach, N.; Lezon-Geyda, K.; Zelterman, D.; DiGiovanna, M.P.; Harris, L.; Abu-Khalaf, M.M. Sirolimus and trastuzumab combination therapy for HER2-positive metastatic breast cancer after progression on prior trastuzumab therapy. Breast Cancer Res. Treat. 2015, 150, 157–167. [Google Scholar] [CrossRef]
  125. Liu, N.; Rowley, B.R.; Bull, C.O.; Schneider, C.; Haegebarth, A.; Schatz, C.A.; Fracasso, P.R.; Wilkie, D.P.; Hentemann, M.; Wilhelm, S.M.; et al. BAY 80-6946 is a highly selective intravenous PI3K inhibitor with potent p110α and p110δ activities in tumor cell lines and xenograft models. Mol. Cancer Ther. 2013, 12, 2319–2330. [Google Scholar] [CrossRef]
  126. Keegan, N.M.; Furney, S.J.; Walshe, J.M.; Gullo, G.; Kennedy, M.J.; Smith, D.; McCaffrey, J.; Kelly, C.M.; Egan, K.; Kerr, J.; et al. Phase Ib trial of copanlisib, a phosphoinositide-3 kinase (PI3K) inhibitor, with trastuzumab in advanced pre-treated HER2-positive breast cancer “PantHER”. Cancers 2021, 13, 1225. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, Y.; Li, J.; Chen, J.J.; Gao, X.; Huang, Z.; Shen, Q. Multifunctional nanoparticles loading with docetaxel and GDC0941 for reversing multidrug resistance mediated by PI3K/Akt signal pathway. Mol. Pharm. 2017, 14, 1120–1132. [Google Scholar] [CrossRef] [PubMed]
  128. Fourneaux, B.; Chaire, V.; Lucchesi, C.; Karanian, M.; Pineau, R.; Laroche-Clary, A.; Italiano, A. Dual inhibition of the PI3K/AKT/mTOR pathway suppresses the growth of leiomyosarcomas but leads to ERK activation through mTORC2: Biological and clinical implications. Oncotarget 2017, 8, 7878–7890. [Google Scholar] [CrossRef] [PubMed]
  129. Holbro, T.; Beerli, R.R.; Maurer, F.; Koziczak, M.; Barbas, C.F., 3rd; Hynes, N.E. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2003, 100, 8933–8938. [Google Scholar] [CrossRef]
  130. Greger, J.G.; Eastman, S.D.; Zhang, V.; Bleam, M.R.; Hughes, A.M.; Smitheman, K.N.; Dickerson, S.H.; Laquerre, S.G.; Liu, L.; Gilmer, T.M. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol. Cancer Ther. 2012, 11, 909–920. [Google Scholar] [CrossRef]
  131. Gayle, S.S.; Arnold, S.L.; O’Regan, R.M.; Nahta, R. Pharmacologic inhibition of mTOR improves lapatinib sensitivity in HER2-overexpressing breast cancer cells with primary trastuzumab resistance. Anti-Cancer Agents Med. Chem. 2012, 12, 151–162. [Google Scholar] [CrossRef]
  132. Casadevall, D.; Hernández-Prat, A.; García-Alonso, S.; Arpí-Llucià, O.; Menéndez, S.; Qin, M.; Guardia, C.; Morancho, B.; Sánchez-Martín, F.J.; Zazo, S.; et al. mTOR inhibition and trastuzumab-emtansine (T-DM1) in HER2-positive breast cancer. Mol. Cancer Res. 2022, 6, 1108–1121. [Google Scholar] [CrossRef]
  133. Lawrence, M.S.; Stojanov, P.; Mermel, C.H.; Robinson, J.T.; Garraway, L.A.; Golub, T.R.; Meyerson, M.; Gabriel, S.B.; Lander, E.S.; Getz, G. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014, 505, 495–501. [Google Scholar] [CrossRef] [PubMed]
  134. Garrido-Castro, A.C.; Saura, C.; Barroso-Sousa, R.; Guo, H.; Ciruelos, E.; Bermejo, B.; Gavilá, J.; Serra, V.; Prat, A.; Paré, L.; et al. Phase 2 study of buparlisib (BKM120), a pan-class I PI3K inhibitor, in patients with metastatic triple-negative breast cancer. Breast Cancer Res. 2020, 22, 120. [Google Scholar] [CrossRef] [PubMed]
  135. Schmid, P.; Abraham, J.; Chan, S.; Wheatley, D.; Brunt, A.M.; Nemsadze, G.; Baird, R.D.; Park, Y.H.; Hall, P.S.; Perren, T.; et al. Capivasertib plus paclitaxel versus placebo plus paclitaxel as first-line therapy for metastatic triple-negative breast cancer: The PAKT trial. J. Clin. Oncol. 2020, 38, 423–433. [Google Scholar] [CrossRef]
  136. Ippen, F.M.; Grosch, J.K.; Subramanian, M.; Kuter, B.M.; Liederer, B.M.; Plise, E.G.; Mora, J.L.; Nayyar, N.; Schmidt, S.P.; Giobbie-Hurder, A.; et al. Targeting the PI3K/Akt/mTOR pathway with the pan-Akt inhibitor GDC-0068 in PIK3CA-mutant breast cancer brain metastases. Neuro. Oncol. 2019, 21, 1401–1411. [Google Scholar] [CrossRef]
  137. Oliveira, M.; Saura, C.; Nuciforo, P.; Calvo, I.; Andersen, J.; Passos-Coelho, J.L.; Gil, M.G.; Bermejo, B.; Patt, D.A.; Ciruelos, E.; et al. FAIRLANE, a double-blind placebo-controlled randomized phase II trial of neoadjuvant ipatasertib plus paclitaxel for early triple-negative breast cancer. Ann. Oncol. 2019, 30, 1289–1297. [Google Scholar] [CrossRef]
  138. Vicier, C.; Sfumato, P.; Isambert, N.; Dalenc, F.; Robert, M.; Levy, C.; Rezai, K.; Provansal, M.; Adélaïde, J.; Garnier, S.; et al. TAKTIC: A prospective, multicentre, uncontrolled, phase IB/II study of LY2780301, a p70S6K/AKT inhibitor, in combination with weekly paclitaxel in HER2-negative advanced breast cancer patients. Eur. J. Cancer 2021, 159, 205–214. [Google Scholar] [CrossRef] [PubMed]
  139. Jovanović, B.; Mayer, I.A.; Mayer, E.L.; Abramson, V.G.; Bardia, A.; Sanders, M.E.; Kuba, M.G.; Estrada, M.V.; Beeler, J.S.; Shaver, T.M.; et al. A randomized phase II neoadjuvant study of cisplatin, paclitaxel with or without everolimus in patients with stage II/III triple-negative breast cancer (TNBC): Responses and long-term outcome correlated with increased frequency of DNA damage response gene mutations, TNBC subtype, AR status, and Ki67. Clin. Cancer Res. 2017, 23, 4035–4045. [Google Scholar] [CrossRef] [PubMed]
  140. Basho, R.K.; Gilcrease, M.; Murthy, R.K.; Helgason, T.; Karp, D.D.; Meric-Bernstam, F.; Hess, K.R.; Herbrich, S.M.; Valero, V.; Albarracin, C.; et al. Targeting the PI3K/AKT/mTOR pathway for the treatment of mesenchymal triple-negative breast cancer: Evidence from a phase 1 trial of mTOR inhibition in combination with liposomal doxorubicin and bevacizumab. JAMA Oncol. 2017, 3, 509–515. [Google Scholar] [CrossRef] [PubMed]
  141. Zhang, Q.; Wang, X.; Cao, S.; Sun, Y.; He, X.; Jiang, B.; Yu, Y.; Duan, J.; Qiu, F.; Kang, N. Berberine represses human gastric cancer cell growth in vitro and in vivo by inducing cytostatic autophagy via inhibition of MAPK/mTOR/p70S6K and Akt signaling pathways. Biomed. Pharmacother. 2020, 128, 110245. [Google Scholar] [CrossRef]
  142. Zecchin, D.; Moore, C.; Michailidis, F.; Horswell, S.; Rana, S.; Howell, M.; Downward, J. Combined targeting of G protein-coupled receptor and EGF receptor signaling overcomes resistance to PI3K pathway inhibitors in PTEN-null triple negative breast cancer. EMBO Mol. Med. 2020, 12, e11987. [Google Scholar] [CrossRef]
  143. You, I.; Erickson, E.C.; Donovan, K.A.; Eleuteri, N.A.; Fischer, E.S.; Gray, N.S.; Toker, A. Discovery of an AKT Degrader with Prolonged Inhibition of Downstream Signaling. Cell Chem. Biol. 2020, 27, 66–73.e7. [Google Scholar] [CrossRef] [PubMed]
  144. Yu, X.; Xu, J.; Shen, Y.; Cahuzac, K.M.; Park, K.S.; Dale, B.; Liu, J.; Parsons, R.E.; Jin, J. Discovery of Potent, Selective, and In Vivo Efficacious AKT Kinase Protein Degraders via Structure-Activity Relationship Studies. J. Med. Chem. 2022, 65, 3644–3666. [Google Scholar] [CrossRef] [PubMed]
  145. Li, Y.; Wang, Y.; Zhang, W.; Wang, X.; Chen, L.; Wang, S. BKM120 sensitizes BRCA-proficient triple negative breast cancer cells to olaparib through regulating FOXM1 and Exo1 expression. Sci. Rep. 2021, 11, 4774. [Google Scholar] [CrossRef] [PubMed]
  146. Gallyas FJr Sumegi, B.; Szabo, C. Role of Akt Activation in PARP Inhibitor Resistance in Cancer. Cancers 2020, 12, 532. [Google Scholar] [CrossRef] [PubMed]
  147. El Guerrab, A.; Bamdad, M.; Bignon, Y.J.; Penault-Llorca, F.; Aubel, C. Co-targeting EGFR and mTOR with gefitinib and everolimus in triple-negative breast cancer cells. Sci. Rep. 2020, 10, 6367. [Google Scholar] [CrossRef]
Figure 1. The genes and proteins of class I PI3K. The catalytic subunits p110α/p110β, the regulatory subunits p85α/p85β and the genes that encoded these subunits (PIK3CA, PIK3CB, PIK3R1 and PIK3R2) are shown. ABD, albumin binding domain; RBD, RAS-binding domain; C2, protein kinase C conserved region 2; Rho-GAP, Rho GTPase-activating protein; SH2, Src homology 2 domain; iSH2, inter-SH2; cSH2, C-terminal SH2.
Figure 1. The genes and proteins of class I PI3K. The catalytic subunits p110α/p110β, the regulatory subunits p85α/p85β and the genes that encoded these subunits (PIK3CA, PIK3CB, PIK3R1 and PIK3R2) are shown. ABD, albumin binding domain; RBD, RAS-binding domain; C2, protein kinase C conserved region 2; Rho-GAP, Rho GTPase-activating protein; SH2, Src homology 2 domain; iSH2, inter-SH2; cSH2, C-terminal SH2.
Cells 11 02508 g001
Figure 2. The PAM signaling pathways. PAM pathway can be activated by G protein-coupled receptor (GPCR) and receptor tyrosine kinases (RTKs), including human epidermal growth factor receptor 2 (HER2/ERBB2), fibroblast growth factor receptor (FGFR), insulin and insulin-like growth factor-1 receptor (InsR/IGF-1R), which PtdIns (4,5) P2 (PIP2) to generate the second messenger PtdIns (3,4,5) P3 (PIP3) [6]. PTEN (phosphatase and tensin homolog) dephosphorylates PIP3 to generate PIP2, while INPP4B (inositol polyphosphate-4-phosphatase type II B) dephosphorylates PtdIns(3,4)P2 to generate PtdIns(3)P (PI3P). Proteins, containing a pleckstrin homology (PH) domain, are recruited to the cytomembrane by PIP3, including AKT, 3-phosphoinositide-dependent kinase 1 (PDK1), and serum and glucocorticoid-induced kinase (SGK). The main downstream target of PI3K is AKT. It is activated by PDK1 and mTOR complex 2 (mTORC2), and phosphorylates a large number of downstream effector proteins, including mTOR complex 1 (mTORC1), forkhead box protein O1 (FoxO1), glycogen synthesis kinase (GSK3) and murine double minute 2 (MDM2). The AKT-mediated phosphorylation of GSK3β, FOXO1 and MDM2 directly or indirectly controls cellular growth and survival. The activated mTORC1 ultimately regulates cellular processes, such as the initiation of mRNA transcription, cell growth, autophagy and protein synthesis, via phosphorylation of 4EBP1 and S6K1 [1,7]. In addition, mTORC2 can phosphorylate both IGF-1R and AKT [8]. S6K-mediated phosphorylation of PDK1 negatively feed back to inhibit PDK1 [7]. PKC and SGK are also involved in PI3K signaling independent of AKT.
Figure 2. The PAM signaling pathways. PAM pathway can be activated by G protein-coupled receptor (GPCR) and receptor tyrosine kinases (RTKs), including human epidermal growth factor receptor 2 (HER2/ERBB2), fibroblast growth factor receptor (FGFR), insulin and insulin-like growth factor-1 receptor (InsR/IGF-1R), which PtdIns (4,5) P2 (PIP2) to generate the second messenger PtdIns (3,4,5) P3 (PIP3) [6]. PTEN (phosphatase and tensin homolog) dephosphorylates PIP3 to generate PIP2, while INPP4B (inositol polyphosphate-4-phosphatase type II B) dephosphorylates PtdIns(3,4)P2 to generate PtdIns(3)P (PI3P). Proteins, containing a pleckstrin homology (PH) domain, are recruited to the cytomembrane by PIP3, including AKT, 3-phosphoinositide-dependent kinase 1 (PDK1), and serum and glucocorticoid-induced kinase (SGK). The main downstream target of PI3K is AKT. It is activated by PDK1 and mTOR complex 2 (mTORC2), and phosphorylates a large number of downstream effector proteins, including mTOR complex 1 (mTORC1), forkhead box protein O1 (FoxO1), glycogen synthesis kinase (GSK3) and murine double minute 2 (MDM2). The AKT-mediated phosphorylation of GSK3β, FOXO1 and MDM2 directly or indirectly controls cellular growth and survival. The activated mTORC1 ultimately regulates cellular processes, such as the initiation of mRNA transcription, cell growth, autophagy and protein synthesis, via phosphorylation of 4EBP1 and S6K1 [1,7]. In addition, mTORC2 can phosphorylate both IGF-1R and AKT [8]. S6K-mediated phosphorylation of PDK1 negatively feed back to inhibit PDK1 [7]. PKC and SGK are also involved in PI3K signaling independent of AKT.
Cells 11 02508 g002
Table 1. Frequencies of changes of PAM pathway in different molecular subtypes of breast cancer.
Table 1. Frequencies of changes of PAM pathway in different molecular subtypes of breast cancer.
Gene
(Protein)
AlterationEffect on SignalingCorrelation with PrognosisFrequencyReference
Luminal (ER+)HER2+TNBC (ER−, PR−, HER2−)
AB
PTENInactivation and mutation/reduced expressionover activation of PI3K signalingNegative in TNBC29–44%22%67%[33,34,35]
PIK3CA
(p110α/PI3K)
Activating mutationHyperactivation of PI3K signaling* Positive in luminal,
negative in metastatic/HER2+ breast cancer
47%33%23–39%8–25%[33,36,37,38]
PIK3CB
(p110β/PI3K)
Amplification
/Mutation
PIP3 accumulates and activates AKTIrrelevant5%[29,39]
PIK3R1
(p85α/PI3K)
Inactivating mutationDerepression of catalytic activity of p110α-2% of Early breast cancer
11% of Metastatic breast cancer
[40,41]
AKT1Activating mutationHyperactivation of AKTIrrelevant2.6–7.4%[33,42,43,44]
AKT2AmplificationIrrelevant2.8–4%[18]
AKT3AmplificationPositive in luminal A breast cancer15%[40]
PDK1AmplificationHyperactivation of AKT-20–38%[45]
(mTOR)p-mTOR expressionHyperactivation of mTORNegative in TNBC39%37.5–72.1%[46]
* Positive: Associated with a better prognosis; Negative: Associated with a worse prognosis; Irrelevant: No significant correlation with prognosis; p-mTOR: phosphorylated mTOR.
Table 2. Clinical trials of PI3K inhibitors in ER+/HER2− breast cancer.
Table 2. Clinical trials of PI3K inhibitors in ER+/HER2− breast cancer.
TargetDrugStudy (Phase)Patient PopulationRegimen and OutcomeFDA/EMA ApprovalReference
Pan-PI3KBuparisib
(BKMI20)
BELLE-2
(III)
HR (+), HER2 (−), ABC/MBC
(second line)
buparlisib + fulvestrant vs. placebo + fulvestrant
(mPFS: 6.9 vs. 5.0 months;
HR: 0.78; p = 0.00021)
N[78]
BELLE-3
(II)
HR (+), HER2 (−), ABC/MBC relapsed on or after endocrine therapy and mTOR inhibitorsbuparlisib vs. Placebo
(mPFS: 3.9 vs. 1.8 months; HR: 0.67; p = 0.0003)
[79]
Pictilisib
(GDC-0941)
FERGI
(II)
HR (+), HER2 (−), ABC/MBC
Al-resistant
pictilisib + fulvestrant vs. placebo + fulvestrant (mPFS:6.6 vs. 5.1 months; HR: 0.74;
p = 0.096)
N[80]
PEGGY
(II)
HR (+), HER2 (−)
metastatic breast cancer
Pictilisib + paclitaxel vs. placebo + paclitaxel (mPFS:8.2 vs. 7.8 months; HR: 0.95) [81]
PI3K
(p110α)
Alpelisib
(BYL719)
SOLAR-1
(III)
HR (+), HER2 (−), ABC
Received endocrine therapy previously
PIK3CA-mutated: alpelisib vs. placebo
(mPFS 11.0 vs. 5.7 months; HR: 0.65; p < 0.001);
(mOS: 39.3 vs. 31.4 months; HR: 0.86; p = 0.15)
Y[82]
BYLieve
(II)
HR (+), HER2 (−),
PIK3CA-mutant ABC
progressed on/after prior therapy, including CDK inhibitors
proportion of without disease progression at 6 month was 50.4% (95% CI: 41.2–59.6). [83]
NEO-ORB
(II)
HR (+), HER2 (−)
Postmenopausal women
Tlc-T3 breast cancer
Alpelisib + letrozole vs. placebo + letrozde,
ORR: 43% vs. 45%,
PIK3CA-wild-type vs. mutant ORR: 63% vs. 61%
[84]
Taselisib
(GDC0032)
SANDPIPER
(III)
Postmenopausal women, disease
recurrence/progression during/after AI
Taselisib vs. placebo
(PFS: 7.4 vs. 5.4 months;
HR: 0.70: p = 0.0037)
N[85]
PI3K-mTORGedatolisibNCT02684032
(I)
metastatic breast cancerNAN[86]
ApitolisibNCT01254526
(Ib)
locally recurrent breast cancer
or metastatic breast cancer
NAN[87]
SamotolisibNCT02057133
(I)
In combination with: letrozole, anastrozole, tamoxifen, exemestaneNAN[88]
Al, aromatase inhibitor; mPFS, median progression-free survival; HR, hazard ratio: MBC, metastatic breast cancer. ABC, advanced breast cancer; ORR, objective response rate; NA, not applicable or discontinued owing to drug toxicity; EMA, European Medicines Agency; N, not yet approved; Y, approved. FDA, Food and Drug Administration.
Table 3. Clinical trials of AKT and mTOR inhibitors in ER+/HER2− breast cancer.
Table 3. Clinical trials of AKT and mTOR inhibitors in ER+/HER2− breast cancer.
TargetDrugStudy (Phase)Patient PopulationRegimen and OutcomeFDA/EMA ApprovedReference
AKT1-3Capivasertib (AZD5363)BEECH
(II)
ER (+), HER2 (−) ABC/MBC (first-line)capivasertib + paclitaxel vs. placebo + paclitaxel (mPFS: 10.9 vs. 8.4 months; HR: 0.80; p = 0.308)
PIK3CA+ sub-population (mPFS: 10.9 vs. 10.8 months; HR: 1.11; p = 0.760)
N[101]
FAKTION
(II)
ER (+)/HER (−)
ABC/MBC;
Postmenopausal relapsed or progressed on AI
capivasertib + fulvestrant vs. placebo + fulvestrant
(mPFS: 10.3 vs. 4.8 months; HR: 0.58; p = 0.0044; mOS: 29.3 vs. 23.4 months; HR: 0.66; p = 0.035)
[102,103]
Pan-AKTMK-2206NCT01776008
(II)
Endocrine resistant, ER+ breast cancer0% pCRN-
mTOR
(mTORC1)
Everolimus (RAD001)BOLERO-2
(III)
ER (+)/HER2−, AI-resistant and postmenopausal
ABC
Everolimus + exemestrane vs. placebo + exemestrane (final PFS:11.0 vs. 4.1 months; HR: 0.38; p < 0.0001) [104]
MANTA
(II)
HR+, postmenopausal and AI-resistant locally ABC or MBCEverolimus +fulvestrant vs. fulvestrant (mPFS: 12.3 vs. 5.4 months; HR: 0.63; p = 0.01)A[105]
PrE0102
(II)
ER (+)/HER2−, AI-resistant and postmenopausal MBCEverolimus + fulvestrant vs. placebo + fulvestrant (mPFS:10.3 vs.5.1 months; HR: 0.61; p = 0.02)
ORR: 18.2 vs. 12.3%; p = 0.47
[106]
BOLERO-6
(III)
ER (+)/HER2−ABCEverolimus + exemestrane vs. Exemestrane vs. capecitabine mOS: 23.1 vs. 29.3 months vs. 25.6 months [107]
NCT02123823
(I-II)
ER (+)/HER2−
ABC and MBC
xentuzumab + everolimus + exemestane, vs. exemestane + everolimus (mPFS: 7.3 vs. 5.6 months; p = 0.9057) [108]
mTOR
(mTORC1/2)
MLN0128NCT02049957
(I-II)
HR+/HER2− and
AI-resistant MBC
everolimus-sensitive vs. everolimus-resistant cohorts, 1 CBR-16: 45% vs. 23%,
2 ORR: 8% vs. 2%
N[109]
ApanisertibNCT02756364
(II)
HR+/HER2− and
AI-resistant MBC
NAN-
Pan-mTORTemsirolimusHORIZON
(III)
HR+, postmenopausal and AI-naïve ABCTemsirolimus vs. placebo + letrozole
(mPFS: 9.0 vs. 5.6 months; HR: 0.7, p < 0.009)
N[110]
1 CBR-16, clinical benefit rate at 16 weeks; 2 ORR, overall response rate.
Table 4. Clinical trials of PAM inhibitors in HER2+ breast cancer.
Table 4. Clinical trials of PAM inhibitors in HER2+ breast cancer.
TargetDrugStudy (Phase)Patient PopulationRegimen and OutcomeReference
Pan-PI3KBuparlisib
(BKM120)
BKM120 (II)Trastuzumab-resistant
HER2+ breast cancer
buparlisib + trastuzumab: ORR:10%
(ORR ≥ 25%)
[118]
PIKHER
(II)
Trastuzumab-resistant
HER2+ ABC
DCR: 79%; 95% CI: 57–92%,
CBR: 29%; 95% CI: 12–51%.
[119]
AlpelisibBYL-719
(I)
Trastuzumab- and taxane-resistant
HER2+ MBC
Alpelisib + T-DM1: ORR: 43%.
T-DM1-resistant (n = 10): ORR 30%.
mPFS 8.1 months
[120]
Pan-AktMK-2206SPY2
(II)
High-risk, early-stage
Breast cancer with neoadjuvant therapy
MK-2206 vs. control: pCR 61.8% vs. 35% (control: standard taxane- and anthracycline-based neoadjuvant therapy)[121]
AKT-1Ipatasertib
(IPAT)
SOLTI-1507
(Ib)
HER2+ ABC or MBC with PIK3CA mutationNA-
mTOREverolimusBOLERO-1
(III)
HER2+, HR-
primary ABC
Everolimus vs. placebo + trastuzumab
mPFS: 20.3 vs.13.1 months; HR: 0.66; p = 0.0049
[122]
BOLERO-3
(III)
Taxane-pretreated and trastuzumab-resistant HER2+ ABCEverolimus vs. Placebo
+ trastuzumab, vinorelbine
mPFS: 7.0 vs.5.8 months; HR: 0.78; p = 0.0067
[123]
SirolimusM124188
(II)
HER2(+) MBCTrastuzumab + sirolimus
ORR: 1/9 (11%) CBR: 4/9 (44%)
[124]
Table 5. Clinical trials of PAM inhibitors in triple negative breast cancer (TNBC).
Table 5. Clinical trials of PAM inhibitors in triple negative breast cancer (TNBC).
TargetDrugStudy (Phase)Patient PopulationRegimen and OutcomeReference
Pan-PI3KBuparlisib
(BKM120)
NCT01790932
(II)
Metastatic TNBCCBR:12% (6 patients, all SD ≥ 4 months) mPFS: 1.8 months (95% CI: 1.6–2.3)
mOS: 11.2 months (95% CI: 6.2–25)
[134]
AKT1-3Capivasertib(II)Metastatic TNBCCapivasertib vs. Placebo + paclitaxel
(mPFS: 5.9 vs. 4.2 months; p = 0.06)
(mOS 19.1 vs. 12.6 months; p = 0.04)
[135]
Pan-AKTGDC-0068
(Ipatasertib)
LOTU
(II)
Metastatic TNBCIpatasertib vs. placebo + paclitaxel
(mPFS 6.2 vs. 4.9 months; HR: 0.60; p = 0.037)
[136]
FAIRLANE
(II)
Early TNBCIpatasertib + paclitaxel vs. placebo + paclitaxel
pCR rates: 17% vs. 13%
[137]
LY2780301TAKTIC
(Ib/II)
HER2-ABC6-month ORR:63.9% [48.8–76.8][138]
mTOREverolimus
(DAE)
NCT00930930
(II)
II/III TNBC
(Neoadjuvant therapy)
Everolimus vs. placebo
(5pCR: 36% vs. 49%)
[139]
Temsirolimus
(DAT)
NCT00761644
(II)
Metaplastic TNBCDoxorubicin + bevacizumab + DAT or DAE
ORR: 21%; CBR: 40%
PI3K pathway alteration: ORR: (31% vs. 0%; p = 0.04); CBR (44% vs. 45%; p > 0.99).
[140]
PI3K-mTOREganelisibNCT03719326
(I/Ib)
Advanced or metastatic
TNBC
In combination with pegylated liposomal doxorubicin (PLD) or A2aR/A2bR antagonist-1(AB928)
NA
-
CI, confident interval; SD, stable disease; mOS, median overall survival; mPFS, median Progression-Free Survival; pCR, pathological complete response; NA: not available.
Table 6. Lists of clinical trials of some PAM inhibitors in different subtypes of breast cancer.
Table 6. Lists of clinical trials of some PAM inhibitors in different subtypes of breast cancer.
TargetDrugHR+, HER2−HER2+Triple Negative Breast Cancer
Class I PI3KBuparisibNCT01633060-NCT01790932
NCT01339442
NCT01610284
PictilisibNCT01437566NCT00928330NCT01918306
NCT01740336
AlpelisibNCT02379247NCT05230810NCT02038010
NCT03386162
NCT04208178
TaselisibNCT02340221NCT02390427NCT02457910
NCT02273973
PI3K-mTORGedatolisibNCT02684032NCT03698383NCT03243331
NCT01920061
ApitolisibNCT01254526--
SamotolisibNCT02057133--
Eganelisib--NCT03961698
AKTCapivasertibNCT01277757-NCT03997123
NCT01992952 NCT03742102
MK-2206NCT01776008--
Ipatasertib-NCT03840200NCT02301988
NCT03800836
LY2780301--NCT01980277
mTOREverolimus NCT02216786NCT00912340NCT01931163
NCT01797120NCT00876395
NCT01783444
NCT02123823
MLN0128NCT02049957-NCT02719691
NCT02988986
ApanisertibNCT02756364--
TemsirolimusNCT02152943NCT00411788NCT02723877
NCT01248494
SirolimusNCT00411788NCT01783444-
Ridaforolimus-NCT00736970-
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhu, K.; Wu, Y.; He, P.; Fan, Y.; Zhong, X.; Zheng, H.; Luo, T. PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer. Cells 2022, 11, 2508. https://doi.org/10.3390/cells11162508

AMA Style

Zhu K, Wu Y, He P, Fan Y, Zhong X, Zheng H, Luo T. PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer. Cells. 2022; 11(16):2508. https://doi.org/10.3390/cells11162508

Chicago/Turabian Style

Zhu, Kunrui, Yanqi Wu, Ping He, Yu Fan, Xiaorong Zhong, Hong Zheng, and Ting Luo. 2022. "PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer" Cells 11, no. 16: 2508. https://doi.org/10.3390/cells11162508

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop