You're using an outdated browser. Please upgrade to a modern browser for the best experience.
PI3K Inhibitors in Breast Cancer: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Paola Fuso.

Breast cancer is the leading cause of death in the female population and despite significant efforts made in diagnostic approaches and treatment strategies adopted for advanced breast cancer, the disease still remains incurable. Therefore, development of more effective systemic treatments constitutes a crucial need. Recently, several clinical trials were performed to find innovative predictive biomarkers and to improve the outcome of metastatic breast cancer through innovative therapeutic algorithms. In the pathogenesis of breast cancer, the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB/AKT)-mammalian target of rapamycin (mTOR) axis is a key regulator of cell proliferation, growth, survival, metabolism, and motility, making it an interest and therapeutic target. Nevertheless, the PI3K/AKT/mTOR cascade includes a complex network of biological events, needing more sophisticated approaches for their use in cancer treatment.

  • PI3K inhibitors
  • subtype breast cancer
  • biomarkers

1. Introduction

Breast cancer (BC) has been divided into four subgroups according to the different expressions of hormone and human epidermal growth factor (HER2) receptors: luminal A, luminal B, HER2 enriched, and basal-like. Each subtype is characterized by different clinical manifestations and prognosis [1,2,3,4][1][2][3][4]. The most advanced genomic assays have expanded BC molecular sequencing, shedding light on the detection of further genetic alterations and new potentially predictive and prognostic biomarkers [5].
Genetic dysregulation in the phosphoinositide 3-kinase (PI3K) pathway is frequently observed in BC. Alterations in the PI3K/AKT/mTOR pathway can either increase or abolish PI3K activity. Moreover, mutations in the tumor suppressing system, such as in the genes encoding for phosphatase and tensin homolog (PTEN) and inositol polyphosphate 4-phosphatase (INPP4B), can determine not only cancer growth, progression, survival, metabolism, protein synthesis and angiogenesis but also the risk of resistance to endocrine therapy and chemotherapy [6]. Therefore, a better understanding of the PI3K axis in BC in the metastatic settings is crucial, in order to better target the different molecular subtypes.
PI3Ks represents a group of heterodimeric lipid kinases at plasma and intracellular membranes characterized by catalytic (p110) and regulatory (p85) subunits. PI3K is divided into three classes with different biochemical properties and specificity. Class I PI3Ks, the most involved in BC, are divided by PI3Ks class IA, class IB, and class IC. The activation of the specific receptor—by the extracellular ligand—stimulates PI3K to catalyze phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-triphosphate (PIP3), recruiting two central mediators of the PI3K cascade, the protein kinase B (AKT) and the phosphoinositide-dependent kinase 1 (PDK1). Then, the phosphorylation of AKT by PDK1 determines a series of different downstream signals mediated by the activation of mammalian target of rapamycin (mTOR), promoting the metabolic and proliferative setting of the cell. The tumor suppressors PTEN and INPP4B are negative regulators of AKT as they catalyze the dephosphorylation of PIP3 to PIP2, limiting the activation of the PI3K signaling axis [7,8,9][7][8][9].

2. Pan-PI3K Inhibitors

Activating mutations in PIK3CA, the gene encoding for the p110α catalytic subunits of PI3K, are associated with the growth and survival of cancer cells that play a role in cell survival, proliferation, differentiation, and glucose transport [4,14,15,16][4][10][11][12]. Dysregulation of the PI3K-pathway may also contribute to resistance to a variety of anticancer agents [17,18][13][14]. First-generation PI3K inhibitors (PI3Ki), also known as pan-inhibitors of PI3K, target all four catalytic isoforms of class I PI3Ks (α, β, γ, and δ) [19,20][15][16]. These inhibitors encompass a broad spectrum of activities, and this broader inhibition leads to a higher risk of adverse events (AEs) and off-target toxicity, which have limited their use at therapeutic doses and caused treatment discontinuation [10,21,22][17][18][19].

2.1. Pan-PI3K Inhibitors in HR+/HER2− Breast Cancer Subtypes

In luminal BC subtype PIK3CA activating mutations occur in roughly 40% of cases and, in this setting, the pan-PI3Ki mostly include buparlisib (BKM120) and pictilisib (GDC-094).

2.1.1. Buparlisib

Buparlisib (BKM120) is an orally selective pan-PI3Ki that inhibits all four class I PI3K isoforms (p110α, β, γ, and δ), as well as somatically mutated p110α, with no activity against the class III PI3K, or mammalian target of rapamycin (mTOR). In cellular assays and in vivo models of human cancers, buparlisib showed significant dose-dependent tumor growth inhibition, potent antiproliferative and proapoptotic activities [26,27][20][21]. Miller et al. reported that the hyperactivation of the PI3K pathway, or PTEN loss expression, promoted antioestrogen resistance in HR+/HER2− MBC, inducing ER-transcriptional independent activity and growth of BC cells [24,25][22][23]. Therefore, buparlisib plus fulvestrant has been successfully evaluated in a phase I study in postmenopausal women with HR+ MBC, previously treated with endocrine therapy [28][24]. PIK3CA mutations proved to be a biomarker of poor prognosis, such as PgR negativity, TP53 mutations, and loss of PTEN expression. Conversely, mutations in AKT1 and ESR1 did not prevent tumor response. Treatment-related adverse events (AEs) were generally mild and included hepatic toxicity, rash and hyperglycemia [29,30][25][26]. The phase III BELLE-2 clinical trial investigated the efficacy and safety of buparlisib plus fulvestrant in HR+/HER2− post-menopausal MBC patients, who were progressive during/after treatment with aromatase inhibitors (AI) and had received up to one prior line of chemotherapy [23][27]. Patients were randomized to receive fulvestrant plus buparlisib or placebo and stratified according to PI3K status in tumor tissue by Sanger sequencing, and visceral disease status. The trial met its primary endpoint demonstrating a relatively modest benefit in progression-free survival (PFS) (6.9 versus 5.0 months compared to placebo; HR 0.78 CI 0.67–0.89; p < 0.001). Many patients, in the buparlisib arm, discontinued the study early (1.9 months) due to adverse events (higher transaminase levels, hyperglycemia, rash, and mood disturbances) probably influencing the potential benefits of combination therapy. In the exploratory analysis on PIK3CA mutations identified in circulating tumor DNA (ctDNA), the combination treatment was associated with an improvement in PFS compared with fulvestrant alone only in patients with PIK3CA mutations. In contrast, prespecified analyses of PIK3CA mutation and/or loss of PTEN expression by Sanger sequencing in archival tissue samples did not show increased PFS with buparlisib, potentially suggesting a tumor evolution from initial diagnosis and treatment beginning. Likewise, BELLE-3 trial explored the ability of fulvestrant plus buparlisib to restore endocrine sensitivity, compared to fulvestrant alone, in patients with luminal MBC subtype previously treated with ET and mTOR inhibitors [31][28]. The trial showed that the combination therapy was associated with better PFS (median 3.9 months versus 1.8 months; HR 0.67 CI 0.53–0.84; p < 0.001), particularly in patients with real-time polymerase chain reaction (RT-PCR) or ctDNA PIK3CA mutations. In ctDNA-detected PI3K alterations the median PFS was 4.2 months versus 1.6 months (HR 0.46 CI 0.29–0.73; p = 0.00031) while it was 4.7 months versus 1.4 months (HR 0.39 CI 0.23–0.65; p < 0.001) in the group tested by RT-PCR. On the contrary, in the BELLE-2 trial, patients with PIK3CA mutations tested in tumor tissue, PFS was not different between treatments. The discrepancy between the two studies could be explained by the different sensitivity of the test used for PIK3CA assessment, or by the absence of stratification, based on ctDNA at randomization and subsequently added as a secondary endpoint. Additionally, Baselga et al., in the combined analysis from BELLE-2 and BELLE-3 trials, observed that assessing PIK3CA-mutational status in ctDNA resulted in larger clinical benefits compared with tissue samples PCR [32,33][29][30]. The safety profile of buparlisib was consistent in BELLE-3 and BELLE-2. Psychiatric complications, related to the ability of buparlisib to penetrate the blood-brain barrier [34][31], such as depression, anxiety, and suicidal ideation, were reported in 2% of patients enrolled in the BELLE-3 trial, in contrast with the absence of suicidal ideation in BELLE-2 study. In phase II/III clinical trial BELLE-4 [35][32], the combination of paclitaxel plus buparlisib in MBC as first-line treatment was tested, showing that the addition of buparlisib to paclitaxel did not improve median PFS when compared to paclitaxel alone (8.0 months versus 9.2 months in buparlisib and placebo group, respectively; HR 1.18 CI 0.82–1.68). Furthermore, the HRs, in the group of patients with PI3K mutations, PTEN loss expression and PI3K wild type, were similar to the overall enrolled population. Buparlisib treatment was associated with a higher frequency of serious AEs (30.2% in the buparlisib arm versus 20.9% in the placebo group) and more than 40% of patients, in the combination arm, experienced diarrhea (55%), rash (43%), hyperglycemia, and nausea (41%).

2.1.2. Pictilisib

Pictilisib (GDC-094) is an oral class I PI3K pan-inhibitor. In vitro experiments showed preliminary antitumor activity (equipotent inhibition of the p110α and -δPI3K isoforms and less potent inhibition of p110β and -γ isoforms) and, in human tumor xenograft murine models, pictilisib demonstrated a strong inhibitory effect on the growth of human U87MG glioblastoma and IGROV1 ovarian cancer [36][33]. In the dose-escalation phase I clinical trial, Sarker et al. [37][34] evaluated the preliminary clinical activity of pictilisib in sixty unselected and heavily pretreated patients with advanced solid tumors. A low response rate, consisting of one partial response (PR) and one stable disease (SD), was observed. The toxicity profile of pictilisib was similar to buparlisib and the most frequent grade 3/4 AEs were rash, hyperglycemia, and pneumonitis. In thPis study pictilisib-related hyperglycemia was limited to grade 1–2; grade > 3 hyperglycemia occurred only in one patient. Mood alterations were not significant due to the low central nervous system (CNS) penetration of pictilisib compared to Buparlisib. According to previous results by Schmid et al., in a phase II, open-label, randomized trial [38][35], the addiction of pictilisib to anastrozole determined a synergic effect and was associated with increased inhibition of tumor cell proliferation over anastrozole alone in postmenopausal women with luminal-B early BC. In the two-part, randomized, double-blind, placebo-controlled phase II clinical trial FERGI, [39][36] investigating the benefit of adding pictilisib to fulvestrant in postmenopausal patients with HR+/HER2− MBC resistant to AI, no difference in median PFS was found in the combination treatment arm, regardless of PI3KCA mutation status. The median PFS was 6.6 months and 5.1 months in the experimental arm and control arm, respectively (HR 0.74 CI 0.52–1.06; p = 0.096) in ITT-population and 6.5 months and 5.1 months, respectively (HR 0.73 95% CI 0.42–1.28; p = 0.268) in women with PIK3CA-mutations. Pictilisib treatment was affected by drug toxicity, potentially limiting its efficacy. Rash, diarrhea, increase transaminase level, hyperglycemia, pneumonitis, bronchopneumonia, pleural effusions, and fatigue were the most common AEs in the pictilisib arm and in approximately 60% of cases grade 3 or worse adverse events were observed, with serious AEs occurring in up to 16% of participants. Another phase II randomized placebo-controlled clinical trial, PEGGY [40][37], failed to meet the primary endpoint. This study, similar to BELLE 4, was conducted to evaluate the benefit of adding PI3K-inhibitors to paclitaxel vs. paclitaxel alone in pre-and post-menopausal HR-positive/HER2-negative advanced BC patients. Genetic alterations in the PIK3CA gene were assessed in about 35% and 32% of patients in both arms, respectively. At the interim analyses, no significant differences in terms of PFS and overall response (ORR) were observed, irrespective of PIK3CA mutation status. Indeed, the addition of pictilisib to paclitaxel did not improve median PFS in the entire (8.2 months vs. 7.8 months; HR 0.95 CI 0.62–1.46; p = 0.83) and in the PI3K pathway-activated study population (7.3 months vs. 5.8 months; HR 1.06 CI 0.52–2.12; p = 0.88). Toxicity due to pictilisib administration included maculopapular rash, hypertension, and hyperglycemia, and experimental treatment was also associated with higher grade ≥ 3 AEs, dose reduction, and discontinuation.

2.2. Pan-PI3K Inhibitors in HER2+ Breast Cancer Subtypes

Although the anti-HER2 agents demonstrated strong efficacy in patients with HER2 + BC, de novo or acquired resistance to HER2-target therapies represents a wide field of research. About 25% of HER2 + BC harbor PIK3CA mutations confer resistance to anti-HER2 therapy and poorer prognosis [41,42,43,44,45,46][38][39][40][41][42][43]. Preclinical studies showed that HER2-signalling is extremely deregulated and is largely mediated by p110α rather than one of the other class-I PI3K isoforms, providing a robust rationale to target the PI3K pathway [47,48,49][44][45][46]. The tolerability and activity of buparlisib in combination with trastuzumab were investigated in a phase Ib/II dose-escalation trial in PIK3CA unselected HER2-positive BC, progressive to trastuzumab. The recommended dose of buparlisib was reached at 100 mg/day plus weekly intravenous trastuzumab but, even if evidence of clinical activity was observed, the study did not meet the pre-established primary endpoint such as objective response > 25% [50,51][47][48]. Furthermore, Guerin et al. studied buparlisib in combination with lapatinib, the orally dual anti-HER2/HER1 tyrosine kinase inhibitor, in the phase Ib/II trial PIKHER2, in HER2-positive trastuzumab-resistant MBC patients, independently to PIK3CA mutational status [52][49]. Twenty-four patients were treated at five different dose regimens; the selected dose was 80 mg/daily for buparlisib and 1000 mg/daily for lapatinib. Main drug-related adverse events leading to discontinuation of treatment were gastrointestinal disorders, skin rash, depression and anxiety. The disease control rate was 79% (95% CI 57–92%) including 4% of CR and a 29% of clinical benefit rate (CBR) (95% CI 12–51%).

2.3. Pan-PI3K Inhibitors in Triple Negative Breast Cancer Subtypes

TNBC represents the most aggressive BC with an incidence of 15–20% and a high heterogeneity in the mutational profile. Due to limited possibility of target therapies, standard chemotherapy still represents the milestone of treatment [53][50]. Approximately 10% of TNBC harbors a germline mutation in BRCA1 or BRCA2 genes and recent evidence has shown that platinum-based therapy offers promising activity both in early and metastatic settings [54,55,56,57,58,59,60][51][52][53][54][55][56][57]. In TNBC, activating PIK3CA mutations, the majority located in the p110α subunit, are the second most frequent molecular aberrations after TP53 mutations and occur in 7–9% of primary TNBC, with a likely higher rate in advanced TNBC [1]. PIK3CA mutations, with additional inactivating alterations in PTEN [61][58] and activating mutations in AKT1, globally occur in about 25% of TNBC [62][59]. In this subtype, dysregulation of the PI3K pathway has been correlated with chemotherapy resistance [63][60] and loss of PTEN function confers resistance to PDL1-blockade and leads to increased PI3Kβ signaling. Indeed, based on preclinical models, combined therapy with an anti-PDL1 agent and a PI3Kβ inhibitor showed an improvement in cancer growth inhibition [62,64][59][61]. However, the role of the PI3K pathway-targeted therapy in TNBC is also unclear. The activation of the PI3K pathway is more linked with the androgen receptor-positive subtype of TNBC and less correlated to TNBC compared to HR-positive and HER2-positive BC [62,65][59][62]. In the BELLE-4 trial TNBC patients (about 25% of total) tended to have a worse prognosis with buparlisib versus placebo arm (5.5 versus 9.3 months; HR 1.86 CI 0.91–3.79) and versus HR+ population (9.2 vs. 9.2 months; HR 1.00 CI 0.66–1.529). In this trial, the poorer prognosis in the TNBC subgroup, may be explained by a lower duration of paclitaxel exposure in the buparlisib group suggesting that the toxicity of buparlisib may have compromised the adequate administration of chemotherapy; hence, threse authoarchers failed to confirm the PIK3CA mutation’s predictive role in TNBC subtype [35][32]. In preclinical models, the pan-PI3Ki BKM120 sensitized BRCA-proficient TNBC to PARP inhibitor olaparib: threse authoarchers demonstrated that the dual PI3K and PARP inhibition significantly downregulated BRCA1/2 expression and reduced tumor cell growth [66][63]. Likewise, a second study showed synergic activity of buparlisib combined with olaparib in an MMTV-CreBRCA1f/fp53 +/− mouse model of BC [67][64]. Additionally, Matulonis UA et al., in a phase I dose-escalation trial, demonstrated anticancer activity in BC (54% of which TNBC) and ovarian cancer and in both germline BRCA (gBRCA)-mutated and gBRCA-wild-type patients [68][65].

References

  1. Bray, F.; Ferlay, J. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424.
  2. Heer, E.; Harper, A. Global burden and trends in premenopausal and postmenopausal breast cancer: A population-based study. Lancet Glob. Health 2020, 8, e1027–e1037.
  3. Perou, C.M.; Sørli, T. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752.
  4. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumors. Nature 2012, 490, 61–70.
  5. Kawaji, H.; Kubo, M. Comprehensive molecular profiling broadens treatment options for breast cancer patients. Cancer Med. 2021, 10, 529–539.
  6. Guerrero-Zotano, A.; Mayer, I.A. PI3K/AKT/mTOR: Role in breast cancer progression, drug resistance, and treatment. Cancer Metastasis Rev. 2016, 35, 515–524.
  7. Fruman, D.; Chiu, H. The PI3K pathway in human disease. Cell 2017, 170, 605–635.
  8. Engelman, J.; Luo, J. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006, 7, 606–619.
  9. Thorpe, L.; Yuzugullu, H. PI3K in cancer: Divergent roles of isoforms, modes of activation, and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24.
  10. Fabi, A.; Metro, G.; Di Benedetto, A.; Nisticò, C.; Vici, P.; Melucci, E.; Antoniani, B. Clinical significance of PTEN and p-Akt co-expression in HER2-positive metastatic breast cancer patients treated with trastuzumab-based therapies. Oncology 2010, 78, 141–149.
  11. Bader, A.G.; Kang, S. Oncogenic PI3K deregulates transcription and translation. Nat. Rev. Cancer 2005, 5, 921–929.
  12. Vivanco, I.; Sawyers, C.L. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2002, 2, 489–501.
  13. Fekete, M.; Santiskulvong, C. Effect of PI3K/Akt pathway inhibition-mediated G1 arrest on chemosensitization in ovarian cancer cells. Anticancer Res. 2012, 32, 445–452.
  14. Carden, C.P.; Stewart, A. The association of PI3 kinase signaling and chemoresistance in advanced ovarian cancer. Mol. Cancer Ther. 2012, 11, 1609–1617.
  15. Garces, A.E.; Stocks, M.J. PI3K Clinical Candidates and Recent Inhibitor Design Strategies: A Medicinal Chemistry Perspective. Med. Chem. 2019, 62, 4815–4850.
  16. Yang, J.; Nie, J. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26.
  17. Ellis, H.; Ma, C.X. PI3K Inhibitors in Breast Cancer Therapy. Curr. Oncol. Rep. 2019, 21, 110.
  18. Verret, B.; Cortes, J. Efficacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol. 2019, 30, 12–20.
  19. Janku, F.; Yap, T.A. Targeting the PI3K pathway in cancer: Are we making headway? Nat. Rev. Clin. Oncol. 2018, 15, 273–291.
  20. Maira, S.M.; Pecchi, S. Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol. Cancer Ther. 2012, 11, 317–328.
  21. Sanchez, C.G.; Ma, C.X. Preclinical modeling of combined phosphatidylinositol-3-kinase inhibition with endocrine therapy for estrogen receptor-positive breast cancer. Breast Cancer Res. 2011, 13, R21.
  22. Miller, T.; Hennessy, W. Hyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancer. J. Clin. Investig. 2010, 120, 2406–2413.
  23. Miller, T.; Rexer, W. Mutations in the phosphatidylinositol 3-kinase pathway: Role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res. 2011, 13, 224.
  24. Ma, C.X.; Luo, J. A phase I trial of BKM120 (Buparlisib) in combination with fulvestrant in postmenopausal women with estrogen receptor-positive metastatic breast cancer. Clin. Cancer Res. 2016, 22, 1583–1591.
  25. Bendell, J.C.; Rodon, J. Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 2012, 30, 282–290.
  26. Mayer, I.A.; Abramson, V.G. Stand up to cancer phase Ib study of pan-phosphoinositide-3-kinase inhibitor buparlisib with letrozole in estrogen receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer. J. Clin. Oncol. 2014, 32, 1202–1209.
  27. Baselga, J.; Im, S.A.; Iwata, H.; Cortés, J.; De Laurentiis, M.; Jiang, Z.; Campone, M. 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. Lancet Oncol. 2017, 18, 904–916.
  28. Di Leo, A.; Johnston, S. Buparlisib plus fulvestrant in postme nopausal 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.
  29. Baselga, J.; Sellami, D. Abstract A050: PIK3CA mutation status in tumor tissue and ctDNA as a biomarker for PFS in patients with HRþ, HER2- ABC treated with buparlisib or placebo plus fulvestrant: Results from the BELLE-2 and BELLE-3 randomized studies. Mol. Cancer Ther. 2018, 17 (Suppl. 1), A050.
  30. Chang, D.Y.; Ma, W.L. Role of Alpelisib in the Treatment of PIK3CA-Mutated Breast Cancer: Patient Selection and Clinical Perspectives (Review). Ther. Clin. Risk Manag. 2021, 17, 193–207.
  31. Nanni, P.; Nicoletti, G. Multiorgan metastasis of human HER-2+ breast cancer in Rag2−/−;Il2rg−/− mice and treatment with PI3K inhibitor. PLoS ONE 2012, 7, e39626.
  32. Martín, M.; Chan, A.; Dirix, L.; O’Shaughnessy, J.; Hegg, R.; Manikhas, A.; Delaloge, S. A randomized adaptive phase II/III study of buparlisib, a pan-class I PI3K inhibitor, combined with paclitaxel for the treatment of HER2- advanced breast cancer (BELLE-4). Ann. Oncol. 2017, 28, 313–320.
  33. Folkes, A.; Ahmadi., K. The identification of 2-(1Hindazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thienopyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J. Med. Chem. 2008, 51, 5522–5532.
  34. Sarker, D.; Ang, J.E. First-in-human phase I study of pictilisib (GDC-0941), a potent pan-class I phosphatidylinositol-3-kinase (PI3K) inhibitor, in patients with advanced solid tumors. Clin. Cancer Res. 2015, 21, 77–86.
  35. Schmid, P.; Pinder, S.E. Phase II randomized preoperative window-of-opportunity study of the PI3K inhibitor pictilisib plus anastrozole compared with anastrozole alone in patients with estrogen receptor-positive breast cancer. J. Clin. Oncol. 2016, 34, 1987–1994.
  36. Krop, I.E.; Mayer, I.A. 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.
  37. Vuylsteke, P.; Huizing, M. Pictilisib PI3Kinase inhibitor (a phosphatidylinositol 3-kinase 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.
  38. Bahrami, A.; Khazaei, M. The therapeutic potential of PI3K/AKT/ mTOR inhibitors in breast cancer: Rationale and progress. J. Cell Biochem. 2018, 119, 213–222.
  39. Berns, K.; Horlings, H.M. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 2007, 12, 395–402.
  40. Pernas, S.; Tolaney, S.M. HER2-positive breast cancer: New therapeutic frontiers and overcoming resistance. Ther. Adv. Med. Oncol. 2019, 11, 1758835919833519.
  41. Baselga, J.; Cortes, J. Biomarker analyses in CLEOPATRA: A phase III, placebo-controlled study of pertuzumab in human epidermal growth factor receptor 2-positive, firstline metastatic breast cancer. J. Clin. Oncol. 2014, 32, 3753–3761.
  42. Baselga, J.; Lewis, G. Relationship between tumor biomarkers and efficacy in EMILIA, a phase III study of trastuzumab emtansine in HER2-positive metastatic breast cancer. Clin. Cancer Res. 2016, 22, 3755–3763.
  43. Chandarlapaty, S.; Sakr, R.A. Frequent mutational activation of the PI3K-AKT pathway in trastuzumab-resistant breast cancer. Clin. Cancer Res. 2012, 18, 6784–6791.
  44. Wilks, S. Potential of overcoming resistance to HER2-targeted therapies through the PI3K/Akt/mTOR pathway. Breast 2015, 24, 548–555.
  45. Li, H.; Prever, L. Targeting PI3K/AKT/mTOR Signaling Pathway in Breast Cancer (Review). Cancers 2021, 13, 3517.
  46. Hanker, A.B.; Pfefferl, A.D. Mutant PIK3CA accelerates HER2-driven transgenic mammary tumors and induces resistance to combinations of anti-HER2 therapies. Proc. Natl. Acad. Sci. USA 2013, 110, 14372–14377.
  47. Saura, C.; Bendell, J. Phase Ib study of buparlisib plus trastuzumab in patients with HER2-positive advanced or metastatic breast cancer that has progressed on Trastuzumab-based therapy. Clin. Cancer Res. 2014, 20, 1935–1945.
  48. Pistilli, B.; Pluard, T. 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.
  49. Guerin, M.; Rezai, K. PIKHER2: A phase IB study evaluating buparlisib in combination with lapatinib in trastuzumabresistant HER2-positive advanced breast cancer. Eur. J. Cancer 2017, 86, 28–36.
  50. Zhang, Z.; Richmond, A. The Role of PI3K Inhibition in the Treatment of Breast Cancer, Alone or Combined with Immune Checkpoint Inhibitors. Front. Mol. Biosci. 2021, 8, 648663.
  51. Poggio, F.; Bruzzone, M. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: A systematic review and meta-analysis. Ann. Oncol. 2018, 29, 1497–1508.
  52. Byrski, T.; Dent, R. Results of a phase II open-label, non-randomized trial of cisplatin chemotherapy in patients with BRCA1-positive metastatic breast cancer. Breast Cancer Res. 2012, 14, R110.
  53. Tutt, A.; Tovey, H. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: The TNT Trial. Nat. Med. 2018, 24, 628–637.
  54. Robson, M.; Im, S.A. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 2017, 377, 523–533.
  55. Litton, J.K.; Rugo, H.S. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N. Engl. J. Med. 2018, 379, 753–763.
  56. Turner, N.C.; Telli, M.L. Final results of a phase 2 study of talazoparib (TALA) following platinum or multiple cytotoxic regimens in advanced breast cancer patients (pts) with germline BRCA1/2 mutations (ABRAZO). J. Clin. Oncol. 2017, 35 (Suppl. 15), 1007.
  57. Andrè, F.; Zielinski, C.C. Optimal strategies for the treatment of metastatic triple-negative breast cancer with currently approved agents. Ann. Oncol. 2012, 23, vi46–vi51.
  58. Shah, S.P.; Roth, A. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 2012, 486, 395–399.
  59. Pascual, J.; Turner, N.C. Targeting the PI3-kinase pathway in triple negative breast cancer. Ann. Oncol. 2019, 30, 1051–1060.
  60. Drullinsky, P.R.; Hurvitz, S.A. Mechanistic basis for PI3K inhibitor antitumor activity and adverse reactions in advanced breast cancer. Breast Cancer Res. Treat. 2020, 181, 233–248.
  61. Peng, W.; Chen, J.Q. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 2016, 6, 202–216.
  62. LoRusso, P. Inhibition of the PI3K/AKT/mTOR pathway in solid tumors. J. Clin. Oncol. 2016, 34, 3803–3815.
  63. Ibrahim, Y.H.; Garcia, G. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2012, 2, 1036–1047.
  64. Juvekar, A.; Burga, L.N. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2012, 2, 1048–1063.
  65. Matulonis, U.A.; Wulf, G.M. Phase I dose escalation study of the PI3kinase pathway inhibitor BKM120 and the oral poly (ADP ribose) polymerase (PARP) inhibitor olaparib for the treatment of highgrade serous ovarian and breast cancer. Ann. Oncol. 2017, 28, 512–518.
More
Academic Video Service