Next Article in Journal
Neuronal Repressor REST Controls Ewing Sarcoma Growth and Metastasis by Affecting Vascular Pericyte Coverage and Vessel Perfusion
Next Article in Special Issue
Insights into the Role of Estrogen Receptor β in Triple-Negative Breast Cancer
Previous Article in Journal
Spectrum-Wide Exploration of Human Adenoviruses for Breast Cancer Therapy
Previous Article in Special Issue
The Landscape of Targeted Therapies in TNBC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges

Department of Medical Oncology & Molecular Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA 91010, USA
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(6), 1404; https://doi.org/10.3390/cancers12061404
Submission received: 3 May 2020 / Revised: 25 May 2020 / Accepted: 26 May 2020 / Published: 29 May 2020

Abstract

:
Triple negative breast cancer (TNBC) is an aggressive breast cancer with historically poor outcomes, primarily due to the lack of effective targeted therapies. The tumor molecular heterogeneity of TNBC has been well recognized, yet molecular subtype driven therapy remains lacking. While neoadjuvant anthracycline and taxane-based chemotherapy remains the standard of care for early stage TNBC, the optimal chemotherapy regimen is debatable. The addition of carboplatin to anthracycline, cyclophosphamide, and taxane (ACT) regimen is associated with improved complete pathologic response (pCR). Immune checkpoint inhibitor (ICI) combinations significantly increase pCR in TNBC. Increased tumor infiltrating lymphocyte (TILs) or the presence of DNA repair deficiency (DRD) mutation is associated with increased pCR. Other targets, such as poly-ADP-ribosyl polymerase inhibitors (PARPi) and Phosphatidylinositol-3-kinase/Protein Kinase B/mammalian target of rapamycin (PI3K-AKT-mTOR) pathway inhibitors, are being evaluated in the neoadjuvant setting. This review examines recent progress in neoadjuvant therapy of TNBC, including platinum, ICI, PARPi, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) pathway targeted therapies, and novel tumor microenvironment (TME) targeted therapy, in addition to biomarkers for the prediction of pCR.

1. Introduction

Triple negative breast cancer (TNBC) accounts for 15% of all breast cancer and it is characterized by the lack of expression of estrogen receptor (ER)/progesterone receptor (PR)/human epidermal growth factor receptor-2 (HER-2), earlier recurrence, tendency of visceral metastasis, and worse overall survival [1,2,3]. The mainstay of treatment for early stage TNBC is neoadjuvant chemotherapy, followed by definitive surgery. Response to initial chemotherapy predicts clinical outcomes in breast cancer [4,5,6]. Neoadjuvant therapy has become increasingly used for the treatment of tumor ≥2 cm in standard-of-care clinical practice, and pathological response is routinely assessed for the evaluation of overall prognosis. Pathological complete response (pCR) was associated with better prognosis in neoadjuvant TNBC trials and has become a surrogate marker of survival [7,8]. The prognosis of TNBC is poor, particularly when pCR was not achieved [9].
Conventional neoadjuvant chemotherapy regimen composed of adriamycin, cyclophosphamide, and paclitaxel (ACT) results in a pCR rate of 35–45% [8,10,11]. Tumor heterogeneity of TNBC is well recognized [12,13,14], yet molecular subtype driven therapy has not become a routine clinical practice, largely due to the lack of effective targeted therapies. Recent clinical trials incorporating immune checkpoint inhibitors (ICI) or targeted therapy, such as poly-ADP-ribosyl polymerase inhibitors (PARPi), may have the potential to personalize neoadjuvant therapy in early stage TNBC [15,16,17]. Other agents, including the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) pathway inhibitors and androgen receptor (AR) targeted therapies, may be applicable for certain molecular subtypes of TNBCs. Novel approaches in conducting neoadjuvant clinical trials, such as I-SPY 2 and ARTEMIS, may accelerate the progress to bring effective targeted therapies to the neoadjuvant setting [18,19,20,21]. In this review, we will summarize recent neoadjuvant trials focusing on the following perspectives: (a) chemotherapy optimization with the addition of carboplatin; (b) the addition of ICI to chemotherapy backbones; (c) clinical trial design by the evaluation of novel targeted agents such as I-SPY 2; (d) biomarker driven identification of clinically relevant patient subgroups to enable a more precise treatment approach (ARTEMIS). Many promising targeted therapies and approaches that are discussed in this review may lead to a paradigm shift of neoadjuvant therapies for early stage TNBC.

2. Molecular Heterogeneity of Triple Negative Breast Cancer

A subset of TNBC is chemosensitive and 35–45% of patients achieve pCR despite the poor prognosis and aggressive nature. This might be explained by the tumor molecular heterogeneity of TNBC. Several molecular classifiers that are based on mRNA profiling of TNBC have been identified. Lehmann et al. reported TNBC-7 subtypes [12]: basal-like 1 (BL1), basal-like 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL), luminal androgen receptor (LAR), and unstable (UNS). The BL1 subtype expresses genes that are related to cell cycle and proliferation, which is consistent with the elevated DNA damage response pathway observed in this subtype. This molecular property explains the robust response to neoadjuvant chemotherapy of the BL1 subtype [14], specifically sensitivity to platinum agents [22]. The BL2 subtype expresses genes that are associated with growth factor signaling. The IM subtype highly expresses immune cell signaling genes. The M and MSL subtypes express genes that are involved in cell motility, and the MSL subtype is also associated with cell differentiation. The LAR subtype demonstrates distinctive gene expression that is enriched in hormone-regulated pathways, such as AR signaling, and it is the least proliferative subtype resulting in enhanced chemotherapy resistance. The clinical relevancy of the TNBC 7-subtype was further investigated by determining pCR rates after neoadjuvant chemotherapy. Of 146 patients with TNBC, molecular subtype and pCR status were significantly associated (p = 0.04379) and TNBC subtype was an independent predictor of pCR status (p = 0.022) by a likelihood ratio test [23]. The BL1 subtype had the highest pCR rate (52%); BL2 and LAR had the lowest (0% and 10%, respectively). Similarly, in a study conducted by Santonja et al., 125 TNBC patients treated with neoadjuvant anthracyclines and/or taxanes +/− carboplatin showed BL1 tumors had the highest pCR to carboplatin containing regimens (80% vs. 23%, p = 0.027) and LAR tumors had the lowest pCR to all treatments (14.3% vs. 42.7%, p = 0.045 when excluding MSL samples) [22]. Later, these seven subtypes were refined into four types (TNBC type-4): BL1, BL2, M, and LAR with evidence of IM and MSL subtypes representing tumors with substantial infiltrating lymphocytes and mesenchymal cells, respectively. The BL1 subtype demonstrated the highest pCR rate of 40–50% [14].
Burstein et al. subdivided TNBCs into TNBC-4 subtypes: LAR, mesenchymal (MES), basal-like immunosuppressed (BLIS), and basal-like immune-activated (BLIA) [13]. The LAR subtype demonstrated molecular evidence of ER activation suggesting response to anti-estrogen or anti-androgen therapies, as described in Lehmann’s subtypes [12]. MES subtype was characterized by pathways of cell cycle, mismatch repair, and DNA damage repair. The BLIS subtype exhibited a downregulation of immune and cytokine pathways that are associated with the worst clinical outcomes. Contrary to BLIS, the BLIA subtype showed the best clinical outcomes with upregulated immunoregulation pathways.
Despite the fact that TNBC subtyping provides an in-depth understanding of the tumor heterogeneity of TNBC [24,25,26], its clinical application has been limited due to the complexity of gene signatures. Table 1 summarizes molecular subtypes of TNBC and potential targets for therapies.

3. Platinum in TNBC

Platinum salts are DNA damaging agents that show an increased efficacy in the tumors with a defected DNA repair system. The platinum salts react with DNA inside cells and distort the double helix of DNA inducing single-strand breaks (SSB) and double-strand breaks (DSB). When these damages cannot be efficiently repaired, it results in cell death [28]. These agents have shown activity in cancers with germline BRCA mutation, as BRCA 1/2 proteins have an essential role in repairing DNA damage [29,30,31].
A high proportion of TNBC exhibits BRCA-like status (BRCAness), which indicates that these tumors are highly sensitive to platinum salts [32,33]. Platinum-based chemotherapy has been investigated in the neoadjuvant setting, with the goal to increase pCR and improve clinical outcome. Carboplatin-containing regimens demonstrated superior pCR rates when compared with standard regimen in two large randomized trials. The CALGB 40603/Alliance trial studied the clinical benefit of adding carboplatin +/− bevacizumab to neoadjuvant chemotherapy in stage II/III TNBC [34]. A total of 443 patients with stage II/ III TNBC received paclitaxel 80 mg/m2 weekly × 12, followed by dose dense doxorubicin and cyclophosphamide (ddAC) × 4, and were randomly assigned to concurrent carboplatin (AUC 6) once every three weeks × 4 ± bevacizumab 10 mg/kg every two weeks × 9. With carboplatin, the percentage of patients who achieved pCR increased significantly from 41% to 54% (odds ratio (OR), 1.71; p = 0.0029). The trial was not powered to detect long term overall survival (OS) and the addition of carboplatin to standard chemotherapy did not improve long term OS [35]. In the GeparSixto trial, 595 patients with stage II and III TNBC were randomized to receive either carboplatin or no carboplatin on a backbone regimen with paclitaxel, liposomal doxorubicin, and bevacizumab [36]. The pCR rates were significantly improved in the carboplatin group: 53.2% vs. 36.9 (p = 0.005). In both the CALGB 40603 and GeparSixto trials, hematological toxicities, including neutropenia and thrombocytopenia, were increased in the carboplatin group. The result from a meta-analysis of nine randomized controlled trials (RCTs) (N =  2109) showed that platinum-based neoadjuvant chemotherapy significantly increased pCR rate from 37.0% to 52.1% (OR 1.96, 95% confidential interval (CI) 1.46–2.62, p < 0.001) [37]. In addition, an increased pCR rate persisted after restricting the analysis to the three RCTs (N = 611) that used the same standard regimen in both groups of weekly paclitaxel (with or without carboplatin), followed by doxorubicin and cyclophosphamide (AC) (OR 2.53, 95% CI 1.37–4.66, p = 0.003). In two of the RCTs (N = 748) with survival data reported, no significant difference in event free survival (EFS) (hazard ratio (HR) 0.72, 95% CI 0.49–1.06, p = 0.094) and OS (HR 0.86, 95% CI 0.46–1.63, p = 0.651) were observed. Significantly increased grade 3/4 hematological adverse events (AEs) were observed with platinum-based neoadjuvant chemotherapy.
Our single center phase II trial of carboplatin plus nab-paclitaxel (carboplatin AUC6 every four weeks × 4 and weekly nab-paclitaxel at 100 mg/m2 × 16 week) in stage II-III TNBC (N = 67) demonstrated a pCR rate of 48% with reasonable tolerability [38]. Sharma et al. reported a pCR rate of 55% with the combination of carboplatin and docetaxel (carboplatin AUC6, docetaxel 75 mg/m2 every three weeks × 6, N = 190) [39]. Table 2 shows the pCR rates from clinical trials that explored the efficacy of carboplatin.
In addition to studies with carboplatin, cisplatin 75 mg/m2, every three weeks × 4 was evaluated in a randomized phase II study of neoadjuvant cisplatin vs. AC in germline BRCA carriers with HER2-negative (TBCRC 031) [46]. The pCR rate was 18% with cisplatin and 26% with AC, yielding a risk ratio (RR) of 0.70 (90% CI, 0.39 to 1.20).
Other chemotherapy agents were evaluated to improve clinical outcomes in TNBC without compelling results. Nab-paclitaxel, an albumin-bound particle form of paclitaxel, has shown preferential tumor uptake and favorable safety profiles when compared to paclitaxel [47,48,49]. The clinical benefit from nab-paclitaxel in TNBC is still controversial [50,51]. When nab-paclitaxel was used instead of paclitaxel in phase III trial, the improvement of pCR was not statistically significant, showing pCR 41.3% with nab-paclitaxel vs. 37.7% with paclitaxel in the TNBC group (OR 0.85; 90% CI, 0.49–1.45) [52]. The GeparSepto trial that included about 20% of TNBC demonstrated significantly higher pCR in nab-paclitaxel subgroup than in the paclitaxel subgroup (48% vs. 26%, p = 0.00027) [53]. Additionally, nab-paclitaxel showed superior pCR when given with carboplatin as compared with gemcitabine [44].

4. Immune Check Point Inhibitors

Introducing immunotherapy in the oncology field has changed the landscape of cancer treatment. Programmed death-1 (PD-1) is a T-cell inhibitory receptor that regulates the immune system by downregulating T-cell response upon binding with its ligand, programmed death ligand-1 (PD-L1) expressed on cancer cells. While the activation of this pathway prevents cancer cells from immune mediated cell death, the inhibition of PD-1 or PD-L1 can restore anti-tumor effects of T-cells. Among all of the breast cancer subtypes, TNBC is especially immunogenic, exhibiting increased expression of PD-L1 [54,55]. This immunogenicity observed in TNBC attracted ICI as a treatment option.
PD-L1 inhibitor atezolizumab showed progression free survival (PFS) benefit in metastatic TNBC (mTNBC) and a complete response (CR) of 10%, which lead to the first ICI approval in TNBC [56]. ICIs have been investigated in several neoadjuvant trials in TNBC. The primary objective of these trials is to test pCR from adding ICI to chemotherapy, mainly taxane and anthracycline. Notably, carboplatin has been utilized when considering its efficacy in TNBC based on previous trials. KEYNOTE-173 is a phase Ib study that showed improved pCR rate from programmed cell-death 1 (PD-1) inhibitor pembrolizumab combined with neoadjuvant chemotherapy [16]. KEYNOTE-522 is a phase III study of neoadjuvant chemotherapy (paclitaxel and carboplatin, then doxorubicin or epirubicin and cyclophosphamide) combined with pembrolizumab or placebo, followed by adjuvant pembrolizumab or placebo in patients with TNBC [15]. An interim analysis reported significantly higher pCR rate in the pembrolizumab combined group (64.8% vs. 51.2%) than in the chemotherapy alone group, regardless of PD-L1 status. Event-free survival (EFS) was significantly higher in the pembrolizumab group during median follow up of 15.5 months. Grade 3 or higher AEs were 76.8% and 72.2%, respectively, with neutropenia as the most common serious AEs in both groups. This is the first phase III trial supporting the role of ICI in neoadjuvant and adjuvant treatment, and a long-term survival result is expected. In I-SPY 2, the overall pCR rate reached 60% when pembrolizumab was given with paclitaxel followed by AC [57].
Durvalumab has also been evaluated for neoadjuvant treatment. In a phase I/II trial, durvalumab with nab-paclitaxel and sequential ddAC carried 55% of pCR in the PD-L1 positive group [58]. GeparNuevo trial randomized patients with TNBC to nab-paclitaxel with or without durvalumab [59]. All of the patients then received epirubicin and cyclophosphamide (EC) as neoadjuvant chemotherapy. Among 174 patients, the pCR rate with durvalumab was 53.4% vs. placebo 44.2%. Interestingly, this increased pCR rate was seen exclusively in patients that were treated with durvalumab alone before the initiation of chemotherapy (pCR 61%). In the NeoTRIP study (NCT002620280), 280 patients with TNBC were randomized to receive neoadjuvant carboplatin AUC 2 and nab-paclitaxel at 125 mg/m2 intravenously (IV) on days 1 and 8 with or without atezolizumab at 1200 mg intravenously on day 1 [60]. The pCR rate was 43.5% (95% CI, 35.1%–52.2%) with atezolizumab and 40.8% (95% CI, 32.7%–49.4%) without atezolizumab in the intent-to-treat population, which led to an odds ratio of 1.11 (95% CI, 0.69–1.79; p = 0.066). In this study, 49% of patients had cT2 disease, 59% had cN1 nodal status, and 56% were PD-L1 positive. The role of atezolizumab in the neoadjuvant setting is currently being investigated in the GeparDouze/NSABP B-59 (NCT03281954) trial. TNBC patients will receive neoadjuvant atezolizumab combined with chemotherapy (carboplatin plus paclitaxel and AC or EC), followed by adjuvant atezolizumab [61]. Clinical trials evaluating ICIs in early stage TNBC are described in Table 3.
Based on these encouraging pCR rates, ICIs may play an important role in neoadjuvant therapy for TNBC and eventually become standard-of-care for a subset of TNBCs. However, well defined biomarkers for the better identification of appropriate patients remain lacking. Long-term survival benefits from adding ICIs need to be evaluated in order to adopt ICIs as neoadjuvant treatment in clinical practice.

5. TME Targeting for Neoadjuvant Treatment

The tumor microenvironment (TME) is associated with immune suppression, escape from immune detection and development of drug resistance, and is being increasingly recognized as a potential target for treatment of TNBC [63]. Tumor associated macrophages (TAMs) promote the progression and metastasis of TNBC by releasing inhibitory cytokines, reducing functions of tumor infiltrating lymphocytes (TILs), promoting TREG (regulatory T-cells), and modulating PD-1/PD-L1 expression in TME [64]. TME targeted therapies are undergoing active clinical trial investigation. The combination of cabiralizumab, an antibody that inhibits the colony stimulating factor-1 receptor (CSF1R) and it blocks the activation and survival of macrophages, and ICI with neoadjuvant chemotherapy might improve efficacy by decreasing TAMs and increasing TILs in early stage TNBC [65,66]. Currently, cabiralizumab is being used in combination with nivolumab and neoadjuvant chemotherapy in patients with localized TNBC (NCT04331067).

6. PARP Inhibitors for Neoadjuvant Treatment

DNA repair deficiency and PARP inhibitors: BRCA 1/2 mutation is one of the greatest genetic risk factors of developing breast cancer. BRCA 1 and BRCA2 are tumor suppressor genes that play a major role in the DNA repair system, specifically in homologous recombination, which repairs double-stranded breaks (DSBs). When homologous recombination does not function (homologous recombination deficiency, HRD), commonly seen in cases of BRCA 1/2 mutations, DSBs result in genomic instability. Poly ADP-ribose polymerase (PARP) 1 is a protein that binds to single stranded breaks (SSBs) during the DNA repair process. PARPi traps PARP1 and induces cell death by preventing SSB repair, followed by DSBs without functional homologous recombination in patients with BRCA mutations. PARPi showed efficacy in patients with BRCA mutations. The OlympiAD trial is a randomized phase III trial that compared olaparib with standard chemotherapy in patients with metastatic breast cancer and germline BRCA mutation [67]. The significantly longer PFS shown in the olaparib group was more prominent in the TNBC (HR 0.43 in TNBC vs. 0.82 in non-TNBC) and made up of 50% of this study. Talazoparib is aPARPi that is approved for advanced breast cancer with BRCA mutation through the EMBRCA trial [68]. In this trial, patients with germline BRCA mutation were randomized in a 2:1 ratio to receive talazoparib or single-agent therapy of the physician’s choice. Among a total of 287 patients who received talazoparib, 45% patients were TNBC. The median PFS was significantly longer in the talazoparib group.
The efficacy of PARPi was further investigated in the neoadjuvant setting. I-SPY 2 trial studied the neoadjuvant PARPi veliparib and carboplatin followed by AC as compared with standard neoadjuvant chemotherapy (paclitaxel followed by AC) [69]. The estimated pCR rate in TNBC was 51% in the veliparib-carboplatin group vs. 26% in the control group. This study demonstrates that patients with TNBC can benefit from PARPi and carboplatin as neoadjuvant treatment. However, the grade 3 or 4 hematologic toxicity was much higher in the veliparib-carboplatin group than in control group. BrighTNess trial is a phase III randomized trial to confirm clinical benefit of adding veliparib and carboplatin in TNBC [45]. A total of 634 patients with stage II-III TNBC were randomized (2:1:1) to receive veliparib/carboplatin/paclitaxel, carboplatin/paclitaxel, or paclitaxel, followed by AC after the randomized portion. The pCR rate in veliparib/carboplatin/paclitaxel group (53%) was significantly higher than the paclitaxel group (31%), but adding veliparib to carboplatin/paclitaxel did not improve pCR (58%). Grade 3–4 hematology toxicity was significantly increased from adding carboplatin, regardless of using veliparib.
Several studies have been conducted to evaluate the role of PARPi as neoadjuvant treatment in early stage BRCA mutated or HRD breast cancer. Most patients enrolled in these studies are TNBC. MD Anderson reported a pilot study of neoadjuvant talazoparib in patients with germline BRCA mutations [70]. Fifteen of 20 enrolled patients were TNBC, and 50% achieved pCR after six months of single agent talazoparib. Hematologic toxicity was the most common AE with 40% Grade 3 anemia and 15% Grade 3 neutropenia. This enhanced pCR from single agent talazoparib offers a different approach for patients with early stage BRCA mutated TNBC. A confirmatory trial is currently ongoing in order to verify the benefit of single agent talazoparib (NCT02401347). GeparOLA trial (NCT02789332) is a phase II randomized trial to evaluate neoadjuvant paclitaxel and olaparib in patients with HRD [71]. The study randomized 107 patients, including 77 TNBC, to either weekly paclitaxel and daily olaparib or weekly paclitaxel and carboplatin for 12 weeks, and then followed with EC. Interestingly, an improved pCR rate in the olaparib group was achieved in the hormone receptor-positive group (29 patients). In TNBC, the olaparib group showed a pCR rate of 56% and the carboplatin group showed 59.3%. Carboplatin is a DNA damaging agent, and works in a similar way to PARPi by inducing DNA damage through DSBs in HDR resulting in a comparable pCR. Table 4 summarizes clinical trials utilizing PAPRi.
The benefit of adding PARPi to neoadjuvant chemotherapy is still controversial for all TNBCs, despite its correlation with genomic instability. The improved pCR from adding carboplatin (also a DNA breaking agent) makes the use of PARPi more controversial. Large randomized trials are needed to determine whether adding carboplatin, PARPi, or both can achieve better pCR. Importantly, the toxicities from adding carboplatin or PARPi should be considered as increased hematologic AEs were observed in previous trials.

7. PI3K/AKT/mTOR Targeted Therapies

Phosphatidylinositol-3-kinase (PI3K)/AKT/ mammalian target of rapamycin (mTOR) signaling is the most commonly activated cancer driver pathway, leading to cell proliferation and survival (Figure 1). The mutation of PIK3CA, the gene encoding the subunit p110α of PI3K, or deactivation of phosphatase and tensin homolog (PTEN), negative regulator of PI3K, can contribute to the progression of cancer [72,73,74]. PIK3CA mutation is found in 20–40% of breast cancer and it is associated with increased resistance to chemotherapy [75,76,77]. The inhibition of this pathway has been actively investigated [78], and the mTOR inhibitor everolimus and the PI3K-α inhibitor alpelisib were FDA approved for hormone receptor-positive metastatic breast cancer [79,80]. The alterations of the PI3K/PTEN/AKT pathway (including PIK3CA mutations, PTEN inactivating mutations, and AKT1 activating mutations) occur in 25% of primary TNBC and possibly at a modestly higher frequency in mTNBC [81,82,83]. In the phase II LOTUS trial, the patients were randomly assigned (1:1) to receive intravenous paclitaxel 80 mg/m2 (days 1, 8, 15) with either ipatasertib, a pan-AKT inhibitor at 400 mg or placebo once per day (days 1–21) every 28 days. Median PFS in the intention-to-treat (ITT) population was 6.2 months (95% CI 3.8–9.0) with ipatasertib versus 4.9 months (3.6–5.4) with placebo (HR 0.60, 95% CI 0.37–0.98; p = 0.037). In the 48 patients with PTEN-low tumors, the median PFS was 6.2 months (95% CI 3.6–9.1) with ipatasertib vs. 3.7 months (range 1.9–7.3 months) with placebo (HR 0.59, 95% CI 0.26–1.32, p = 0.18) [84]. The Phase III IPATUNITY130 trial (NCT03337724), where patient receive either paclitaxel and ipatasertib or paclitaxel and placebo, will confirm survival benefit [85]. The AKT inhibitor Capivasertib showed significantly longer PFS and OS in mTNBC when added to paclitaxel as first line treatment of mTNBC [86].
Ipatasertib has been studied in neoadjuvant TNBC in a phase II neoadjuvant FAIRLANE study of weekly paclitaxel plus ipatasertib or placebo with the following endpoints: pCR rate, PTEN-low population assessed via IHC and PIK3CA/AKT1/PTEN-altered tumors using next generation sequencing (NGS) [87]. The addition of ipatasertib showed a small increase in pCR rate of 17% vs. 13% in ITT. The clinical response rate by breast MRI of ipatasertib was numerically improved, but not statistically significant in the biomarker-selected patients: PTEN-low tumors (32% vs. 6%) and PIK3CA/AKT1/PTEN-altered tumors (39% vs. 9%). In the adaptive neoadjuvant phase II I-SPY 2 trial, the AKT inhibitor MK-2206 plus standard neoadjuvant chemotherapy of weekly paclitaxel followed by AC achieved an estimated pCR rate of 40% when compared with 22% from chemotherapy alone in the TNBC subgroup [88]. These results support further evaluation of AKT inhibition + paclitaxel and AC neoadjuvant chemotherapy in patients with PIK3CA/AKT1/PTEN-altered tumors.

8. Androgen Receptor Targeting in TNBC

AR is a potential therapeutic target considering 10–40% of TNBC express AR of 1 to 10% of stained tumor cells [89,90,91]. The efficacy of AR inhibitors has been studied in AR-positive mTNBC. The AR inhibitor enzalutamide has demonstrated clinical benefit rate at 16 weeks of 33% in mTNBC with AR ≥ 10% [92]. Abiraterone, an inhibitor of 17α-Hydroxylase/C17,20-lyase (CYP17) required enzyme for androgen biosynthesis, had modest objective responsive rate (ORR) of 6.7%, PFS of 2.8 months and six month clinical benefit rate (CBR) of 20% in AR-positive (≥10% IHC) mTNBC [93]. Single agent AR targeted therapy appears to be modest, and combination therapy with other targeted agents are currently under investigation. Enzalutamide plus paclitaxel neoadjuvant therapy is currently ongoing (NCT02689427). Enzalutamide in combination with taselisib (NCT02457910) or alpelisib (03207529) trials in mTNBC are actively accruing patients.

9. Biomarkers Predicting pCR in TNBC

9.1. Tumor Infiltrating Lymphocytes

While immunotherapy is successful across a variety of tumor types, biomarkers precisely predicting response to therapy remain to be identified. Understanding the tumor immune microenvironment holds promise for optimal cancer therapy. TILs and PD-L1 and are the most commonly used biomarkers to evaluate the response to ICI. The presence of stromal tumor infiltrating lymphocytes (sTILs) is widely recognized as a good prognostic factor in both adjuvant and neoadjuvant chemotherapy [94,95,96]. Loi et al. reported that higher levels of TILs were associated with decreased distant recurrent in TNBC, and improved disease free survival (DFS) and OS [97]. Two pooled analyses with a large number of patients demonstrated that increased TILs predict pCR and improved survival in TNBC. The German Breast Cancer Group analyzed pretreatment core biopsies from 3771 patients for sTILs following the guidelines of the International TIL working group [98]. TILs were predefined in three groups: low (0–10%), intermediate (11–59%), and high TILs (≥60%). Increased TIL percentile predicted response to neoadjuvant chemotherapy in TNBC: pCR was achieved in 80/260 (31%) of patients with low TILs, 117/373 (31%) of patients with intermediate TILs, and 136/273 (50%) of patients with high TILs (p < 0.0001). A 10% increase in TILs was associated with longer DFS in TNBC (HR 0.93 (95% CI 0·87–0·98), p = 0.011) and longer overall survival in TNBC (HR 0·92 (95% CI 0·86–0·99), p = 0.032). These findings were reproduced in a different pooled analysis with 2148 patients from nine studies for adjuvant chemotherapy [99]. Mean sTILs was 23% and increased sTILs were significantly associated with improved survival: HR for a 10% increase in sTILs was 0.83 (95% CI, 0.78–0.87) for distant DFS and 0.83 (95% CI, 0.79–0.88) for OS. sTILs significantly decreased in metastatic TNBCs as compared with matched primary [100,101]. Higher TIL PD1 expression was associated with better prognosis in early stage TNBCs [102]. These results further support the approach of introducing ICIs early in the neoadjuvant or adjuvant setting, since primary tumors are more immunogenic.

9.2. PD-L1

PD-L1 expression on tumor cells or immune cells has been evaluated as a biomarker of treatment response to anti-PD-1 or anti-PD-L1 therapies [103,104]. Measuring PD-L1 expression remains controversial due to different methods and antibodies. The expression of PL-L1 in TNBC was estimated to be 40–65% on immune cells [105,106]. Mittendorf et al. reported 19% (20 /105 TNBC) of tumor cells were PD-L1 positive, defined by >5% of membranous staining by IHC [54]. In IMpassion 130 trial, intratumoral CD8 correlated with PD-L1 immune cell expression, and was therefore predictive of prolonged PFS (HR, 0.74; 95% CI, 0.61–0.91) and OS (HR, 0.66; 95% CI, 0.50–0.88) with atezolizumab and nab-paclitaxel vs. placebo and nab-paclitaxel [107]. sTILs were not well correlated with PD-L1 immune cell expression, and only predicted prolonged PFS with atezolizumab when compared with placebo (HR, 0.66; 95% CI, 0.50–0.86). There is a lack of quantitative association between PD-L1 expression and response. Indeed, the response to ICI is not linearly associated with increasing levels of expression, and the methods and antibodies used for PD-L1 assessment remain controversial [108]. It has been observed that PD-L1 negative patients may still derive benefit from ICIs. The knowledge gap in PD-L1 testing across different trials needs to be mitigated in order to best characterize patients who might benefit from ICIs.

9.3. Immune Gene Signatures

In addition to TILs and PD-L1, multi-gene signatures have been studied as a more comprehensive tool capturing the immunogenicity of TNBC. The GeparSixto trial was analyzed for mRNA markers from pretherapeutic formalin-fixed paraffine embedded core biopsied samples [109]. A GeparSixto immune signature (GSIS) composed of seven immune-activating genes (CXCL9, CCL5, CD8A, CD80, CXCL13, IGKC, CD21) and five immunosuppressive (IDO1, PD-1, PD-L1, CTLA4, FOXP3) genes was validated as a marker for immune reaction. GSIS revealed that the increased mRNA expression level of these genes, including immunosuppressive genes, was associated with pCR. In our neoadjuvant carboplatin and nab-paclitaxel trial, GSIS was significantly associated with pCR and residual cancer burden (RCB) in a multivariate model (submitted) [38].

9.4. Combined Modality of Gene Signatures and IHCs

Using laser capture microdissection gene expression profiles, the tumor immune microenvironment (TIME) was captured and subclassified from therapy-naïve TNBC tumors. An “immune hot” TIME exhibited tumor infiltration of granzyme B+CD8+ T cells (GzmB+CD8+ T cells), a type 1 IFN signature, and elevated expression of multiple immune inhibitory molecules, including indoleamine 2,3-dioxygenase (IDO) and PD-L1, was associated with good outcomes. An “immune-cold” TIME with an absence of tumoral CD8+ T cells was defined by elevated expression of the immunosuppressive marker B7-H4, signatures of fibrotic stroma, and was associated with poor outcomes [110]. This laboratory approach appears to be labor-intensive and might not be easily adapted in the clinic.

9.5. BRCAness or DNA Repair Defect

Recent advanced technology can capture functional HRD beyond BRCA 1/2 mutations (BRCAness) that shares molecular features of BRCA alteration with a scoring system. myChoice HRD® by Myriad Genetics (Salt Lake City, UT, USA) is a commercially available test for assessing HRD. This is a NGS-based in vitro test that determines genomic instability that is based on an algorithmic scoring system of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST) [111]. TNBC is strongly related with BRCA mutation and HRD. Among all of the patients with TNBC, 10–15% of patients have germline BRCA 1/2 mutations [112,113] and 40–60% of patients are positive for HRD [114,115,116]. Other genes that are involved in the HR process, such as PALB2 and RAD51, have been discovered to play an important role in TNBC [117]. TNBC with BRCA mutation or HRD is more sensitive to chemotherapy or PARPi [111,118,119,120]. However, evaluating HRD has not been standardized in clinical practice. Currently there are few commercially available methods to evaluate HRD, and this needs to be further studied before being used in clinical practice.

10. Novel Neoadjuvant Clinical Trial Approach

Several ongoing trials have been evaluating the addition of novel targeted therapy agents to standard chemotherapy in the neoadjuvant setting, including I-SPY 2 and ARTEMIS trial. The I-SPY 2 trial utilizes an adaptive design for evaluating the addition of novel agents to paclitaxel, followed by AC (P-AC) in high-risk early stage breast cancer [121]. The addition of veliparib to P-AC had an estimated pCR of 51% [69] and adding pembrolizumab to P-AC had an estimated pCR rate of 60% [57]. Although improved pCR is encouraging, the addition of veliparib or pembrolizumab has not been conclusively shown to improve long-term outcome. This might be attributed to the small sample size.
Precision medicine based on the genomic tests has been adopted in clinical trials. In the metastatic setting, the utility of genomic mutation driven therapies has been tested in basket trials, such as NCI-MATCH (Molecular Analysis for Therapy Choice), which contains a multi-arm design with each arm testing a single drug on a histology-agnostic fashion [122,123]. Despite the appealing concept of precision medicine for management of metastatic breast cancer, the implementation of such approaches in the neoadjuvant setting remains challenging. While TNBC patients with pCR/RCB-0 or RCB-1 have better survival, those with extensive residual disease (RCB-II or RCB-III) after neoadjuvant chemotherapy (NACT) have poor prognoses [124,125,126]. The ARTEMIS (NCT 02276443) is a randomized phase II trial to determine whether precision neoadjuvant therapy (P-NAT) impacts the rates of pathologic response (RCB 0–I) while using a CLIA-certified chemosensitivity mRNA gene signature (GES) and subtyping of TNBC by IHC to select targeted therapy trials for chemotherapy-insensitive tumors [19,20,21]. The initial study plan was to randomize 360 patients with TNBC as 2:1 ratio to “know” vs. “not know” P-NAT. Chemotherapy-sensitive tumors received chemotherapy, and chemotherapy-insensitive tumors were enrolled in a clinical trial. The first interim analysis (N = 133 patients with RCB status) revealed a RCB 0–1 rate of 56% (“know” P-NAT) vs. 62% (“not know” P-NAT); p = 1.0; thus, randomization was discontinued for futility [19]. A total of 232 patients were enrolled, including 168 evaluable for RCB. In the ultrasound-resistant cohort (N = 43), RCB 0–I rates were higher in patients treated with targeted therapy (N = 30) vs. AC-T (N = 13); (30% vs. 8%; odds ratio = 5.1 with 95% CI, 0.6–45.7; p = 0.11). GES failed to improve the rates of RCB 0–I in TNBC; however, in patients with resistant disease identified by ultrasound after AC, RCB 0–I rates were higher in patients that were treated with targeted therapy as compared to chemotherapy alone. This trial again demonstrated a persistent gap between tumor biology and the clinical application of precision medicine in the neoadjuvant setting.

11. De-Escalation vs. Escalation of NAC Regimen in TNBC

The optimization of a neoadjuvant chemotherapy regimen in early stage TNBC continues to evolve. The key question remains to be the appropriate selection of a neoadjuvant regimen based on patient and disease characteristics. Recently, the promising pCR rate that was reported from KEYNOTE 522 using pembrolizumab plus carboplatin/paclitaxel followed by AC potentially shifts the standard-of-care regimen for early stage TNBC neoadjuvant therapy toward more intensive chemotherapy backbone, such as weekly carbo/taxol followed by AC, although significant Grade 3–4 AEs raised the question of whether every patient requires such an intensive regimen.
There is existing evidence showing the carboplatin/taxane regimen remains highly active and it could serve as a chemotherapy backbone for immunotherapy or targeted therapy combinations (de-escalated chemotherapy). Recent trials demonstrated a promising pCR rate of “anthracycline-free regimen”. Our single center phase II trial of carboplatin plus nab-paclitaxel (carboplatin AUC6 every four weeks × 4 and weekly nab-paclitaxel at 100 mg/m2 × 16 week) in stage II-III TNBC (N = 67) demonstrated a pCR rate of 48% with reasonable tolerability [38]. Sharma et al. reported a pCR rate of 55% with the combination of carboplatin and docetaxel (carboplatin AUC6, docetaxel 75 mg/m2 every three weeks × 6, N = 190) [39]. WSG-ADAPT-TN trial reported by Gluz et al. also reflected a de-escalation concept with a 12 week neoadjuvant regimen [44]. When patients were treated with carboplatin AUC2 with nab-paclitaxel 125 mg/m2 on day 1 and 8 for four three-week cycles, the pCR rate was 44.9 (N = 154). The expression of immunological genes (CD8, PD-L1), basal-like mRNA expression profile, and high Ki-67 were associated with pCR in a multi-variate model (p < 0.05) [127]. All three trials are consistent with a favorable toxicity profile and high efficacy using carboplatin and taxane based anthracyclin-free regimen. These data support further research while using de-escalated chemotherapy backbone for combination therapy with ICI or targeted therapy. Currently, the ongoing NeoPACT trial combining carboplatin AUC6, docetaxel 75 mg, and pembrolizumab 200 mg every three weeks is actively enrolling patients, and the results of the trial result are eagerly awaited (NCT03639948). Confirmatory analysis of biomarkers predicting patients who can achieve pCR without the use of anthracycline and/or ICIs is critical for patient selection.

12. Conclusions and Future Direction

Recent progress has been made in neoadjuvant therapy for early stage TNBC. ICI and PARPi may become standard-of-care for appropriate subtypes of TNBC. Carboplatin remains an important treatment in BRCA-associated tumors or HRD tumors. Novel clinical trial design, such as I-SPY 2 or ARTEMIS, might vastly facilitate testing novel targeted therapy in the neoadjuvant setting. The many promising targeted therapies and approaches that are discussed in this review may lead to a paradigm shift of neoadjuvant therapies for early stage TNBC.

Author Contributions

Conceptualization, J.S.L. and Y.Y.; writing—original draft preparation, J.S.L.; writing—review and editing, S.E.Y. and Y.Y.; visualization, S.E.Y.; supervision, Y.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Y.Y. has contracted clinical trials and research projects sponsored by Novartis, Eisai, Pfizer, Merck, Genentech, and Puma. Y.Y. is an advisory board of Immunomedics, Pfizer, Genentech, and Novartis and a speaker bureau of Eisai, Novartis, AstraZeneca, Genentech, and Daiichi. The other authors declare no conflict of interest.

References

  1. Billar, J.A.Y.; Dueck, A.C.; Stucky, C.-C.H.; Gray, R.J.; Wasif, N.; Northfelt, D.W.; McCullough, A.E.; Pockaj, B.A. Triple-negative breast cancers: Unique clinical presentations and outcomes. Ann. Surg. Oncol. 2010, 17, 384–390. [Google Scholar] [CrossRef] [PubMed]
  2. Malorni, L.; Shetty, P.B.; De Angelis, C.; Hilsenbeck, S.; Rimawi, M.F.; Elledge, R.; Osborne, C.K.; De Placido, S.; Arpino, G. Clinical and biologic features of triple-negative breast cancers in a large cohort of patients with long-term follow-up. Breast Cancer Res. Treat. 2012, 136, 795–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Li, X.; Yang, J.; Peng, L.; Sahin, A.A.; Huo, L.; Ward, K.C.; O’Regan, R.; Torres, M.A.; Meisel, J.L. Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res. Treat. 2017, 161, 279–287. [Google Scholar] [CrossRef] [PubMed]
  4. Broglio, K.R.; Quintana, M.; Foster, M.; Olinger, M.; McGlothlin, A.; Berry, S.M.; Boileau, J.-F.; Brezden-Masley, C.; Chia, S.; Dent, S.; et al. Association of Pathologic Complete Response to Neoadjuvant Therapy in HER2-Positive Breast Cancer With Long-Term Outcomes: A Meta-Analysis. JAMA Oncol. 2016, 2, 751–760. [Google Scholar] [CrossRef] [Green Version]
  5. Kozak, M.M.; Jacobson, C.E.; Von Eyben, R.; Pollom, E.L.; Telli, M.; Horst, K.C. Outcomes Following Neoadjuvant Chemotherapy for Breast Cancer in Women Aged 40 Years and Younger: Impact of Pathologic Nodal Response. J. Natl. Compr. Cancer Netw. 2018, 16, 845–850. [Google Scholar] [CrossRef] [Green Version]
  6. Cortazar, P.; Zhang, L.; Untch, M.; Mehta, K.; Costantino, J.P.; Wolmark, N.; Bonnefoi, H.; Cameron, D.; Gianni, L.; Valagussa, P.; et al. Pathological complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014, 384, 164–172. [Google Scholar] [CrossRef] [Green Version]
  7. Liedtke, C.; Mazouni, C.; Hess, K.R.; André, F.; Tordai, A.; Mejia, J.A.; Symmans, W.F.; Gonzalez-Angulo, A.M.; Hennessy, B.; Green, M.; et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2008, 26, 1275–1281. [Google Scholar] [CrossRef]
  8. Biswas, T.; Efird, J.T.; Prasad, S.; Jindal, C.; Walker, P.R. The survival benefit of neoadjuvant chemotherapy and pCR among patients with advanced stage triple negative breast cancer. Oncotarget 2017, 8, 112712–112719. [Google Scholar] [CrossRef] [Green Version]
  9. Von Minckwitz, G.; Untch, M.; Blohmer, J.-U.; Costa, S.D.; Eidtmann, H.; Fasching, P.A.; Gerber, B.; Eiermann, W.; Hilfrich, J.; Huober, J.; et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J. Clin. Oncol. 2012, 30, 1796–1804. [Google Scholar] [CrossRef] [Green Version]
  10. Chen, V.E.; Gillespie, E.F.; Zakeri, K.; Murphy, J.D.; Yashar, C.M.; Lu, S.; Einck, J.P. Pathologic response after neoadjuvant chemotherapy predicts locoregional control in patients with triple negative breast cancer. Adv. Radiat. Oncol. 2017, 2, 105–109. [Google Scholar] [CrossRef] [Green Version]
  11. Gamucci, T.; Pizzuti, L.; Sperduti, I.; Mentuccia, L.; Vaccaro, A.; Moscetti, L.; Marchetti, P.; Carbognin, L.; Michelotti, A.; Iezzi, L.; et al. Neoadjuvant chemotherapy in triple-negative breast cancer: A multicentric retrospective observational study in real-life setting. J. Cell. Physiol. 2018, 233, 2313–2323. [Google Scholar] [CrossRef] [PubMed]
  12. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Burstein, M.D.; Tsimelzon, A.; Poage, G.M.; Covington, K.R.; Contreras, A.; Fuqua, S.A.W.; Savage, M.I.; Osborne, C.K.; Hilsenbeck, S.G.; Chang, J.C.; et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin. Cancer Res. 2015, 21, 1688–1698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lehmann, B.D.; Jovanović, B.; Chen, X.; Estrada, M.V.; Johnson, K.N.; Shyr, Y.; Moses, H.L.; Sanders, M.E.; Pietenpol, J.A. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368. [Google Scholar] [CrossRef]
  15. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kümmel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef]
  16. Schmid, P.; Salgado, R.; Park, Y.H.; Muñoz-Couselo, E.; Kim, S.B.; Sohn, J.; Im, S.-A.; Foukakis, T.; Kuemmel, S.; Dent, R.; et al. Pembrolizumab plus chemotherapy as neoadjuvant treatment of high-risk, early-stage triple-negative breast cancer: Results from the phase 1b open-label, multicohort KEYNOTE-173 study. Ann. Oncol. 2020. [Google Scholar] [CrossRef]
  17. Litton, J.; Symmans, F.; Gogineni, K.; Saltzman, M.; Telli, M.; Usha, L.; Chakrabarti, J.; Tudor, I.; Quek, R.; Czibere, A. Abstract OT3-03-02: A phase 2, open-label, single-arm, multi-center study of talazoparib for neoadjuvant treatment of germline BRCA1/2 mutation patients with early-stage triple-negative breast cancer (TNBC). Cancer Res. 2019, 79. [Google Scholar] [CrossRef]
  18. Wolf, D.M.; Yau, C.; Sanil, A.; Brown-Swigart, L.; Hirst, G.; Buxton, M.; Paoloni, M.; Olopade, O.; Rugo, H.; De Michele, A.; et al. Abstract 858: Combining sensitivity markers to identify triple-negative breast cancer patients most responsive to veliparib/carboplatin: Results from the I-SPY 2 TRIAL. Cancer Res. 2016, 76, 858. [Google Scholar] [CrossRef]
  19. Seth, S.; Huo, L.; Rauch, G.M.; Adrada, B.E.; Piwnica-Worms, H.; Thompson, A.M.; Mittendorf, E.A.; Litton, J.K.; Symmans, W.F.; Draetta, G.F.; et al. Delineating longitudinal patterns of response to neoadjuvant systemic therapy (NAST) in triple-negative breast cancer (TNBC): Profiling results from a randomized, TNBC enrolling trial to confirm molecular profiling improves survival (ARTEMIS; NCT02276443). JCO 2019, 37, 586. [Google Scholar] [CrossRef]
  20. Moulder, S.L.; Hess, K.R.; Candelaria, R.P.; Rauch, G.M.; Santiago, L.; Adrada, B.; Yang, W.T.; Gilcrease, M.Z.; Huo, L.; Stauder, M.C.; et al. Precision neoadjuvant therapy (P-NAT): A planned interim analysis of a randomized, TNBC enrolling trial to confirm molecular profiling improves survival (ARTEMIS). JCO 2018, 36, 518. [Google Scholar] [CrossRef]
  21. Garber, H.; Rauch, G.; Adrada, B.; Candelaria, R.; Mittendorf, E.; Thompson, A.; Litton, J.; Damodaran, S.; Lim, B.; Arun, B.; et al. Abstract P2-16-09: Residual cancer burden in patients with early stage triple negative breast cancer who progress on anthracycline-based neoadjuvant chemotherapy in an ongoing clinical trial (ARTEMIS). Cancer Res. 2020, 80. [Google Scholar] [CrossRef]
  22. Santonja, A.; Sánchez-Muñoz, A.; Lluch, A.; Chica-Parrado, M.R.; Albanell, J.; Chacón, J.I.; Antolín, S.; Jerez, J.M.; De la Haba, J.; De Luque, V.; et al. Triple negative breast cancer subtypes and pathologic complete response rate to neoadjuvant chemotherapy. Oncotarget 2018, 9, 26406–26416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Masuda, H.; Baggerly, K.A.; Wang, Y.; Zhang, Y.; Gonzalez-Angulo, A.M.; Meric-Bernstam, F.; Valero, V.; Lehmann, B.D.; Pietenpol, J.A.; Hortobagyi, G.N.; et al. Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes. Clin. Cancer Res. 2013, 19, 5533–5540. [Google Scholar] [CrossRef] [Green Version]
  24. Jézéquel, P.; Kerdraon, O.; Hondermarck, H.; Guérin-Charbonnel, C.; Lasla, H.; Gouraud, W.; Canon, J.-L.; Gombos, A.; Dalenc, F.; Delaloge, S.; et al. Identification of three subtypes of triple-negative breast cancer with potential therapeutic implications. Breast Cancer Res. 2019, 21, 65. [Google Scholar] [CrossRef] [PubMed]
  25. Jiang, Y.-Z.; Ma, D.; Suo, C.; Shi, J.; Xue, M.; Hu, X.; Xiao, Y.; Yu, K.-D.; Liu, Y.-R.; Yu, Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 2019, 35, 428–440.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Prado-Vázquez, G.; Gámez-Pozo, A.; Trilla-Fuertes, L.; Arevalillo, J.M.; Zapater-Moros, A.; Ferrer-Gómez, M.; Díaz-Almirón, M.; López-Vacas, R.; Navarro, H.; Maín, P.; et al. A novel approach to triple-negative breast cancer molecular classification reveals a luminal immune-positive subgroup with good prognoses. Sci. Rep. 2019, 9, 1538. [Google Scholar] [CrossRef] [PubMed]
  27. Collignon, J.; Lousberg, L.; Schroeder, H.; Jerusalem, G. Triple-negative breast cancer: Treatment challenges and solutions. Breast Cancer 2016, 8, 93–107. [Google Scholar] [CrossRef] [Green Version]
  28. Garutti, M.; Pelizzari, G.; Bartoletti, M.; Malfatti, M.C.; Gerratana, L.; Tell, G.; Puglisi, F. Platinum Salts in Patients with Breast Cancer: A Focus on Predictive Factors. Int. J. Mol. Sci. 2019, 20, 3390. [Google Scholar] [CrossRef] [Green Version]
  29. Byrski, T.; Gronwald, J.; Huzarski, T.; Grzybowska, E.; Budryk, M.; Stawicka, M.; Mierzwa, T.; Szwiec, M.; Wisniowski, R.; Siolek, M.; et al. Pathologic complete response rates in young women with BRCA1-positive breast cancers after neoadjuvant chemotherapy. J. Clin. Oncol. 2010, 28, 375–379. [Google Scholar] [CrossRef]
  30. Byrski, T.; Dent, R.; Blecharz, P.; Foszczynska-Kloda, M.; Gronwald, J.; Huzarski, T.; Cybulski, C.; Marczyk, E.; Chrzan, R.; Eisen, A.; et al. 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. [Google Scholar] [CrossRef] [Green Version]
  31. Vencken, P.M.L.H.; Kriege, M.; Hoogwerf, D.; Beugelink, S.; Van der Burg, M.E.L.; Hooning, M.J.; Berns, E.M.; Jager, A.; Collée, M.; Burger, C.W.; et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann. Oncol. 2011, 22, 1346–1352. [Google Scholar] [CrossRef] [PubMed]
  32. Isakoff, S.J.; Mayer, E.L.; He, L.; Traina, T.A.; Carey, L.A.; Krag, K.J.; Rugo, H.S.; Liu, M.C.; Stearns, V.; Come, S.E.; et al. TBCRC009: A Multicenter Phase II Clinical Trial of Platinum Monotherapy With Biomarker Assessment in Metastatic Triple-Negative Breast Cancer. J. Clin. Oncol. 2015, 33, 1902–1909. [Google Scholar] [CrossRef] [PubMed]
  33. Vollebergh, M.A.; Lips, E.H.; Nederlof, P.M.; Wessels, L.F.A.; Wesseling, J.; Vd Vijver, M.J.; De Vries, E.G.E.; Van Tinteren, H.; Jonkers, J.; Hauptmann, M.; et al. Genomic patterns resembling BRCA1- and BRCA2-mutated breast cancers predict benefit of intensified carboplatin-based chemotherapy. Breast Cancer Res. 2014, 16, R47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sikov, W.M.; Berry, D.A.; Perou, C.M.; Singh, B.; Cirrincione, C.T.; Tolaney, S.M.; Kuzma, C.S.; Pluard, T.J.; Somlo, G.; Port, E.R.; et al. Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance). J. Clin. Oncol. 2015, 33, 13–21. [Google Scholar] [CrossRef] [Green Version]
  35. Sikov, W.M.; Polley, M.-Y.; Twohy, E.; Perou, C.M.; Singh, B.; Berry, D.A.; Tolaney, S.M.; Somlo, G.; Port, E.R.; Ma, C.X.; et al. CALGB (Alliance) 40603: Long-term outcomes (LTOs) after neoadjuvant chemotherapy (NACT) +/- carboplatin (Cb) and bevacizumab (Bev) in triple-negative breast cancer (TNBC). JCO 2019, 37, 591. [Google Scholar] [CrossRef]
  36. Von Minckwitz, G.; Schneeweiss, A.; Loibl, S.; Salat, C.; Denkert, C.; Rezai, M.; Blohmer, J.U.; Jackisch, C.; Paepke, S.; Gerber, B.; et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): A randomised phase 2 trial. Lancet Oncol. 2014, 15, 747–756. [Google Scholar] [CrossRef]
  37. Poggio, F.; Bruzzone, M.; Ceppi, M.; Pondé, N.F.; La Valle, G.; Del Mastro, L.; De Azambuja, E.; Lambertini, M. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: A systematic review and meta-analysis. Ann. Oncol. 2018, 29, 1497–1508. [Google Scholar] [CrossRef]
  38. Yuan, Y.; Frankel, P.; Li, M.; Kruper, L.; Jones, V.; Treece, T.; Waisman, J.; Yim, J.; Tumyan, L.; Schmolze, D.; et al. Abstract P1-15-07: Phase II trial of neoadjuvant carboplatin and nab-paclitaxel in patients with locally advanced triple negative breast cancer. Cancer Res. 2019, 79. [Google Scholar] [CrossRef]
  39. Sharma, P.; López-Tarruella, S.; García-Saenz, J.A.; Ward, C.; Connor, C.S.; Gómez, H.L.; Prat, A.; Moreno, F.; Jerez-Gilarranz, Y.; Barnadas, A.; et al. Efficacy of Neoadjuvant Carboplatin plus Docetaxel in Triple-Negative Breast Cancer: Combined Analysis of Two Cohorts. Clin. Cancer Res. 2017, 23, 649–657. [Google Scholar] [CrossRef] [Green Version]
  40. Alba, E.; Chacon, J.I.; Lluch, A.; Anton, A.; Estevez, L.; Cirauqui, B.; Carrasco, E.; Calvo, L.; Segui, M.A.; Ribelles, N.; et al. A randomized phase II trial of platinum salts in basal-like breast cancer patients in the neoadjuvant setting. Results from the GEICAM/2006-03, multicenter study. Breast Cancer Res. Treat. 2012, 136, 487–493. [Google Scholar] [CrossRef]
  41. Ando, M.; Yamauchi, H.; Aogi, K.; Shimizu, S.; Iwata, H.; Masuda, N.; Yamamoto, N.; Inoue, K.; Ohono, S.; Kuroi, K.; et al. Randomized phase II study of weekly paclitaxel with and without carboplatin followed by cyclophosphamide/epirubicin/5-fluorouracil as neoadjuvant chemotherapy for stage II/IIIA breast cancer without HER2 overexpression. Breast Cancer Res. Treat. 2014, 145, 401–409. [Google Scholar] [CrossRef]
  42. Zhang, P.; Yin, Y.; Mo, H.; Zhang, B.; Wang, X.; Li, Q.; Yuan, P.; Wang, J.; Zheng, S.; Cai, R.; et al. Better pathologic complete response and relapse-free survival after carboplatin plus paclitaxel compared with epirubicin plus paclitaxel as neoadjuvant chemotherapy for locally advanced triple-negative breast cancer: A randomized phase 2 trial. Oncotarget 2016, 7, 60647–60656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Schneeweiss, A.; Möbus, V.; Tesch, H.; Hanusch, C.; Denkert, C.; Lübbe, K.; Huober, J.; Klare, P.; Kümmel, S.; Untch, M.; et al. Intense dose-dense epirubicin, paclitaxel, cyclophosphamide versus weekly paclitaxel, liposomal doxorubicin (plus carboplatin in triple-negative breast cancer) for neoadjuvant treatment of high-risk early breast cancer (GeparOcto-GBG 84): A randomised phase III trial. Eur. J. Cancer 2019, 106, 181–192. [Google Scholar] [CrossRef] [PubMed]
  44. Gluz, O.; Nitz, U.; Liedtke, C.; Christgen, M.; Grischke, E.-M.; Forstbauer, H.; Braun, M.; Warm, M.; Hackmann, J.; Uleer, C.; et al. Comparison of Neoadjuvant Nab-Paclitaxel+Carboplatin vs Nab-Paclitaxel+Gemcitabine in Triple-Negative Breast Cancer: Randomized WSG-ADAPT-TN Trial Results. J. Natl. Cancer Inst. 2018, 110, 628–637. [Google Scholar] [CrossRef]
  45. Loibl, S.; O’Shaughnessy, J.; Untch, M.; Sikov, W.M.; Rugo, H.S.; McKee, M.D.; Huober, J.; Golshan, M.; Von Minckwitz, G.; Maag, D.; et al. Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): A randomised, phase 3 trial. Lancet Oncol. 2018, 19, 497–509. [Google Scholar] [CrossRef]
  46. Tung, N.; Arun, B.; Hacker, M.R.; Hofstatter, E.; Toppmeyer, D.L.; Isakoff, S.J.; Borges, V.; Legare, R.D.; Isaacs, C.; Wolff, A.C.; et al. TBCRC 031: Randomized Phase II Study of Neoadjuvant Cisplatin Versus Doxorubicin-Cyclophosphamide in Germline BRCA Carriers With HER2-Negative Breast Cancer (the INFORM trial). J. Clin. Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
  47. Nyman, D.W.; Campbell, K.J.; Hersh, E.; Long, K.; Richardson, K.; Trieu, V.; Desai, N.; Hawkins, M.J.; Von Hoff, D.D. Phase I and pharmacokinetics trial of ABI-007, a novel nanoparticle formulation of paclitaxel in patients with advanced nonhematologic malignancies. J. Clin. Oncol. 2005, 23, 7785–7793. [Google Scholar] [CrossRef]
  48. Gradishar, W.J.; Tjulandin, S.; Davidson, N.; Shaw, H.; Desai, N.; Bhar, P.; Hawkins, M.; O’Shaughnessy, J. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J. Clin. Oncol. 2005, 23, 7794–7803. [Google Scholar] [CrossRef]
  49. Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.; Beals, B.; Dykes, D.; et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res. 2006, 12, 1317–1324. [Google Scholar] [CrossRef] [Green Version]
  50. Futamura, M.; Nagao, Y.; Ishihara, K.; Takeuchi, M.; Nakada, T.; Kawaguchi, Y.; Asano, M.; Kumazawa, I.; Shiroko, T.; Morimitsu, K.; et al. Preoperative neoadjuvant chemotherapy using nanoparticle albumin-bound paclitaxel followed by epirubicin and cyclophosphamide for operable breast cancer: A multicenter phase II trial. Breast Cancer 2017, 24, 615–623. [Google Scholar] [CrossRef] [Green Version]
  51. Kuwayama, T.; Nakamura, S.; Hayashi, N.; Takano, T.; Tsugawa, K.; Sato, T.; Kitani, A.; Okuyama, H.; Yamauchi, H. Randomized Multicenter Phase II Trial of Neoadjuvant Therapy Comparing Weekly Nab-paclitaxel Followed by FEC With Docetaxel Followed by FEC in HER2- Early-stage Breast Cancer. Clin. Breast Cancer 2018, 18, 474–480. [Google Scholar] [CrossRef] [PubMed]
  52. Gianni, L.; Mansutti, M.; Anton, A.; Calvo, L.; Bisagni, G.; Bermejo, B.; Semiglazov, V.; Thill, M.; Chacon, J.I.; Chan, A.; et al. Comparing Neoadjuvant Nab-paclitaxel vs Paclitaxel Both Followed by Anthracycline Regimens in Women With ERBB2/HER2-Negative Breast Cancer-The Evaluating Treatment With Neoadjuvant Abraxane (ETNA) Trial: A Randomized Phase 3 Clinical Trial. JAMA Oncol. 2018, 4, 302–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Untch, M.; Jackisch, C.; Schneeweiss, A.; Schmatloch, S.; Aktas, B.; Denkert, C.; Schem, C.; Wiebringhaus, H.; Kümmel, S.; Warm, M.; et al. NAB-Paclitaxel Improves Disease-Free Survival in Early Breast Cancer: GBG 69–GeparSepto. JCO 2019, 37, 2226–2234. [Google Scholar] [CrossRef]
  54. Mittendorf, E.A.; Philips, A.V.; Meric-Bernstam, F.; Qiao, N.; Wu, Y.; Harrington, S.; Su, X.; Wang, Y.; Gonzalez-Angulo, A.M.; Akcakanat, A.; et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol. Res. 2014, 2, 361–370. [Google Scholar] [CrossRef] [Green Version]
  55. Soliman, H.; Khalil, F.; Antonia, S. PD-L1 expression is increased in a subset of basal type breast cancer cells. PLoS ONE 2014, 9, e88557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.-A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef] [PubMed]
  57. Nanda, R.; Liu, M.C.; Yau, C.; Shatsky, R.; Pusztai, L.; Wallace, A.; Chien, A.J.; Forero-Torres, A.; Ellis, E.; Han, H.; et al. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women With Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncol. 2020. [Google Scholar] [CrossRef]
  58. Pusztai, L.; Reisenbichler, E.; Bai, Y.; Fischbach, N.; Persico, J.; Adelson, K.; Katoch, A.; Horowitz, N.; Lannin, D.; Killelea, B.; et al. Abstract PD1-01: Durvalumab (MEDI4736) concurrent with nab-paclitaxel and dose dense doxorubicin cyclophosphamide (ddAC) as neoadjuvant therapy for triple negative breast cancer (TNBC). Cancer Res. 2020, 80. [Google Scholar] [CrossRef]
  59. Loibl, S.; Untch, M.; Burchardi, N.; Huober, J.; Sinn, B.V.; Blohmer, J.-U.; Grischke, E.-M.; Furlanetto, J.; Tesch, H.; Hanusch, C.; et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: Clinical results and biomarker analysis of GeparNuevo study. Ann. Oncol. 2019, 30, 1279–1288. [Google Scholar] [CrossRef] [Green Version]
  60. Gianni, L.; Huang, C.-S.; Egle, D.; Bermejo, B.; Zamagni, C.; Thill, M.; Anton, A.; Zambelli, S.; Bianchini, G.; Russo, S.; et al. Abstract GS3-04: Pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple negative, early high-risk and locally advanced breast cancer. NeoTRIPaPDL1 Michelangelo randomized study. Cancer Res. 2020, 80. [Google Scholar] [CrossRef]
  61. Geyer, C.E.; Loibl, S.; Rastogi, P.; Seiler, S.; Costantino, J.P.; Vijayvergia, N.; Cortazar, P.; Lucas, P.C.; Denkert, C.; Mamounas, E.P.; et al. NSABP B-59/GBG 96-GeparDouze: A randomized double-blind phase III clinical trial of neoadjuvant chemotherapy (NAC) with atezolizumab or placebo in Patients (pts) with triple negative breast cancer (TNBC) followed by adjuvant atezolizumab or placebo. JCO 2018, 36. [Google Scholar] [CrossRef]
  62. Mittendorf, E.; Barrios, C.; Harbeck, N.; Miles, D.; Saji, S.; Zhang, H.; Duc, A.-N.; Rafii, S.; Lai, C. Abstract OT2-07-03: IMpassion031: A phase III study comparing neoadjuvant atezolizumab vs placebo in combination with nab-paclitaxel–based chemotherapy in early triple-negative breast cancer (TNBC). Cancer Res. 2018, 78. [Google Scholar] [CrossRef]
  63. Deepak, K.G.K.; Vempati, R.; Nagaraju, G.P.; Dasari, V.R.; Nagini, S.; Rao, D.N.; Malla, R.R. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020, 153, 104683. [Google Scholar] [CrossRef] [PubMed]
  64. Santoni, M.; Romagnoli, E.; Saladino, T.; Foghini, L.; Guarino, S.; Capponi, M.; Giannini, M.; Cognigni, P.D.; Ferrara, G.; Battelli, N. Triple negative breast cancer: Key role of Tumor-Associated Macrophages in regulating the activity of anti-PD-1/PD-L1 agents. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 78–84. [Google Scholar] [CrossRef]
  65. Ngambenjawong, C.; Gustafson, H.H.; Pun, S.H. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv. Drug Deliv. Rev. 2017, 114, 206–221. [Google Scholar] [CrossRef] [Green Version]
  66. Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.-M.; Ries, C.H.; Rüttinger, D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef]
  67. Robson, M.; Im, S.-A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N. Engl. J. Med. 2017, 377, 523–533. [Google Scholar] [CrossRef]
  68. Hurvitz, S.A.; Gonçalves, A.; Rugo, H.S.; Lee, K.-H.; Fehrenbacher, L.; Mina, L.A.; Diab, S.; Blum, J.L.; Chakrabarti, J.; Elmeliegy, M.; et al. Talazoparib in Patients with a Germline BRCA-Mutated Advanced Breast Cancer: Detailed Safety Analyses from the Phase III EMBRACA Trial. Oncologist 2019. [Google Scholar] [CrossRef] [Green Version]
  69. Rugo, H.S.; Olopade, O.I.; DeMichele, A.; Yau, C.; Van ’t Veer, L.J.; Buxton, M.B.; Hogarth, M.; Hylton, N.M.; Paoloni, M.; Perlmutter, J.; et al. Adaptive Randomization of Veliparib-Carboplatin Treatment in Breast Cancer. N. Engl. J. Med. 2016, 375, 23–34. [Google Scholar] [CrossRef]
  70. Litton, J.K.; Scoggins, M.E.; Hess, K.R.; Adrada, B.E.; Murthy, R.K.; Damodaran, S.; DeSnyder, S.M.; Brewster, A.M.; Barcenas, C.H.; Valero, V.; et al. Neoadjuvant Talazoparib for Patients With Operable Breast Cancer With a Germline BRCA Pathogenic Variant. JCO 2019, 38, 388–394. [Google Scholar] [CrossRef]
  71. Fasching, P.A.; Jackisch, C.; Rhiem, K.; Schneeweiss, A.; Klare, P.; Hanusch, C.; Huober, J.B.; Link, T.; Untch, M.; Schmatloch, S.; et al. GeparOLA: A randomized phase II trial to assess the efficacy of paclitaxel and olaparib in comparison to paclitaxel/carboplatin followed by epirubicin/cyclophosphamide as neoadjuvant chemotherapy in patients (pts) with HER2-negative early breast cancer (BC) and homologous recombination deficiency (HRD). JCO 2019, 37, 506. [Google Scholar] [CrossRef]
  72. Li, G.; Robinson, G.W.; Lesche, R.; Martinez-Diaz, H.; Jiang, Z.; Rozengurt, N.; Wagner, K.-U.; Wu, D.-C.; Lane, T.F.; Liu, X.; et al. Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland. Development 2002, 129, 4159–4170. [Google Scholar] [PubMed]
  73. Campbell, I.G.; Russell, S.E.; Choong, D.Y.H.; Montgomery, K.G.; Ciavarella, M.L.; Hooi, C.S.F.; Cristiano, B.E.; Pearson, R.B.; Phillips, W.A. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 2004, 64, 7678–7681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Levine, D.A.; Bogomolniy, F.; Yee, C.J.; Lash, A.; Barakat, R.R.; Borgen, P.I.; Boyd, J. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clin. Cancer Res. 2005, 11, 2875–2878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Knuefermann, C.; Lu, Y.; Liu, B.; Jin, W.; Liang, K.; Wu, L.; Schmidt, M.; Mills, G.B.; Mendelsohn, J.; Fan, Z. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene 2003, 22, 3205–3212. [Google Scholar] [CrossRef] [Green Version]
  76. 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] [Green Version]
  77. Isakoff, S.J.; Engelman, J.A.; Irie, H.Y.; Luo, J.; Brachmann, S.M.; Pearline, R.V.; Cantley, L.C.; Brugge, J.S. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005, 65, 10992–11000. [Google Scholar] [CrossRef] [Green Version]
  78. LoRusso, P.M. Inhibition of the PI3K/AKT/mTOR Pathway in Solid Tumors. J. Clin. Oncol. 2016, 34, 3803–3815. [Google Scholar] [CrossRef]
  79. Baselga, J.; Campone, M.; Piccart, M.; Burris, H.A.; Rugo, H.S.; Sahmoud, T.; Noguchi, S.; Gnant, M.; Pritchard, K.I.; Lebrun, F.; et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N. Engl. J. Med. 2012, 366, 520–529. [Google Scholar] [CrossRef] [Green Version]
  80. 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]
  81. Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [CrossRef] [PubMed] [Green Version]
  82. Shah, S.P.; Roth, A.; Goya, R.; Oloumi, A.; Ha, G.; Zhao, Y.; Turashvili, G.; Ding, J.; Tse, K.; Haffari, G.; et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 2012, 486, 395–399. [Google Scholar] [CrossRef]
  83. Pascual, J.; Turner, N.C. Targeting the PI3-kinase pathway in triple-negative breast cancer. Ann. Oncol. 2019, 30, 1051–1060. [Google Scholar] [CrossRef] [Green Version]
  84. Kim, S.-B.; Dent, R.; Im, S.-A.; Espié, M.; Blau, S.; Tan, A.R.; Isakoff, S.J.; Oliveira, M.; Saura, C.; Wongchenko, M.J.; et al. Ipatasertib plus paclitaxel versus placebo plus paclitaxel as first-line therapy for metastatic triple-negative breast cancer (LOTUS): A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2017, 18, 1360–1372. [Google Scholar] [CrossRef]
  85. Dent, R.; Kim, S.-B.; Oliveira, M.; Isakoff, S.J.; Barrios, C.H.; O’Shaughnessy, J.; Lu, X.; Wongchenko, M.; Bradley, D.; Mani, A.; et al. IPATunity130: A pivotal randomized phase III trial evaluating ipatasertib (IPAT) + paclitaxel (PAC) for PIK3CA/AKT1/PTEN-altered advanced triple-negative (TN) or hormone receptor-positive HER2-negative (HR+/HER2–) breast cancer (BC). JCO 2018, 36. [Google Scholar] [CrossRef]
  86. 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]
  87. Oliveira, M.; Saura, C.; Nuciforo, P.; Calvo, I.; Andersen, J.; Passos-Coelho, J.L.; Gil Gil, M.; 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]
  88. 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 Cancers in the I-SPY 2 Trial. J. Clin. Oncol. 2020, 38, 1059–1069. [Google Scholar] [CrossRef]
  89. Niemeier, L.A.; Dabbs, D.J.; Beriwal, S.; Striebel, J.M.; Bhargava, R. Androgen receptor in breast cancer: Expression in estrogen receptor-positive tumors and in estrogen receptor-negative tumors with apocrine differentiation. Mod. Pathol. 2010, 23, 205–212. [Google Scholar] [CrossRef]
  90. Park, S.; Koo, J.; Park, H.S.; Kim, J.-H.; Choi, S.-Y.; Lee, J.H.; Park, B.-W.; Lee, K.S. Expression of androgen receptors in primary breast cancer. Ann. Oncol. 2010, 21, 488–492. [Google Scholar] [CrossRef]
  91. Astvatsaturyan, K.; Yue, Y.; Walts, A.E.; Bose, S. Androgen receptor positive triple negative breast cancer: Clinicopathologic, prognostic, and predictive features. PLoS ONE 2018, 13, e0197827. [Google Scholar] [CrossRef] [Green Version]
  92. Traina, T.A.; Miller, K.; Yardley, D.A.; Eakle, J.; Schwartzberg, L.S.; O’Shaughnessy, J.; Gradishar, W.; Schmid, P.; Winer, E.; Kelly, C.; et al. Enzalutamide for the Treatment of Androgen Receptor-Expressing Triple-Negative Breast Cancer. J. Clin. Oncol. 2018, 36, 884–890. [Google Scholar] [CrossRef] [PubMed]
  93. Bonnefoi, H.; Grellety, T.; Tredan, O.; Saghatchian, M.; Dalenc, F.; Mailliez, A.; L’Haridon, T.; Cottu, P.; Abadie-Lacourtoisie, S.; You, B.; et al. A phase II trial of abiraterone acetate plus prednisone in patients with triple-negative androgen receptor positive locally advanced or metastatic breast cancer (UCBG 12-1). Ann. Oncol. 2016, 27, 812–818. [Google Scholar] [CrossRef]
  94. Loi, S.; Sirtaine, N.; Piette, F.; Salgado, R.; Viale, G.; Van Eenoo, F.; Rouas, G.; Francis, P.; Crown, J.P.A.; Hitre, E.; et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J. Clin. Oncol. 2013, 31, 860–867. [Google Scholar] [CrossRef] [PubMed]
  95. Dieci, M.V.; Criscitiello, C.; Goubar, A.; Viale, G.; Conte, P.; Guarneri, V.; Ficarra, G.; Mathieu, M.C.; Delaloge, S.; Curigliano, G.; et al. Prognostic value of tumor-infiltrating lymphocytes on residual disease after primary chemotherapy for triple-negative breast cancer: A retrospective multicenter study. Ann. Oncol. 2014, 25, 611–618. [Google Scholar] [CrossRef]
  96. Criscitiello, C.; Esposito, A.; Trapani, D.; Curigliano, G. Prognostic and predictive value of tumor infiltrating lymphocytes in early breast cancer. Cancer Treat. Rev. 2016, 50, 205–207. [Google Scholar] [CrossRef]
  97. Loi, S.; Michiels, S.; Salgado, R.; Sirtaine, N.; Jose, V.; Fumagalli, D.; Kellokumpu-Lehtinen, P.-L.; Bono, P.; Kataja, V.; Desmedt, C.; et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: Results from the FinHER trial. Ann. Oncol. 2014, 25, 1544–1550. [Google Scholar] [CrossRef]
  98. Denkert, C.; Von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; Budczies, J.; Huober, J.; Klauschen, F.; Furlanetto, J.; et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: A pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018, 19, 40–50. [Google Scholar] [CrossRef]
  99. Loi, S.; Drubay, D.; Adams, S.; Pruneri, G.; Francis, P.A.; Lacroix-Triki, M.; Joensuu, H.; Dieci, M.V.; Badve, S.; Demaria, S.; et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J. Clin. Oncol. 2019, 37, 559–569. [Google Scholar] [CrossRef]
  100. He, T.-F.; Yost, S.E.; Frankel, P.H.; Dagis, A.; Cao, Y.; Wang, R.; Rosario, A.; Tu, T.Y.; Solomon, S.; Schmolze, D.; et al. Multi-panel immunofluorescence analysis of tumor infiltrating lymphocytes in triple negative breast cancer: Evolution of tumor immune profiles and patient prognosis. PLoS ONE 2020, 15, e0229955. [Google Scholar] [CrossRef] [Green Version]
  101. Hutchinson, K.E.; Yost, S.E.; Chang, C.-W.; Johnson, R.M.; Carr, A.R.; McAdam, P.R.; Halligan, D.L.; Chang, C.-C.; Schmolze, D.; Liang, J.; et al. Comprehensive Profiling of Poor-Risk Paired Primary and Recurrent Triple-Negative Breast Cancers Reveals Immune Phenotype Shifts. Clin. Cancer Res. 2020, 26, 657–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Ren, X.; Wu, H.; Lu, J.; Zhang, Y.; Luo, Y.; Xu, Q.; Shen, S.; Liang, Z. PD1 protein expression in tumor infiltrated lymphocytes rather than PDL1 in tumor cells predicts survival in triple-negative breast cancer. Cancer Biol. Ther. 2018, 19, 373–380. [Google Scholar] [CrossRef] [PubMed]
  103. Patel, S.P.; Kurzrock, R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol. Cancer Ther. 2015, 14, 847. [Google Scholar] [CrossRef]
  104. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
  105. Beckers, R.K.; Selinger, C.I.; Vilain, R.; Madore, J.; Wilmott, J.S.; Harvey, K.; Holliday, A.; Cooper, C.L.; Robbins, E.; Gillett, D.; et al. Programmed death ligand 1 expression in triple-negative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome. Histopathology 2016, 69, 25–34. [Google Scholar] [CrossRef] [PubMed]
  106. Cimino-Mathews, A.; Thompson, E.; Taube, J.M.; Ye, X.; Lu, Y.; Meeker, A.; Xu, H.; Sharma, R.; Lecksell, K.; Cornish, T.C.; et al. PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum. Pathol. 2016, 47, 52–63. [Google Scholar] [CrossRef] [Green Version]
  107. Emens, L.; Loi, S.; Rugo, H.; Schneeweiss, A.; Diéras, V.; Iwata, H.; Barrios, C.; Nechaeva, M.; Molinero, L.; Nguyen Duc, A.; et al. Abstract GS1-04: IMpassion130: Efficacy in immune biomarker subgroups from the global, randomized, double-blind, placebo-controlled, phase III study of atezolizumab + nab -paclitaxel in patients with treatment-naïve, locally advanced or metastatic triple-negative breast cancer. In Proceedings of the San Antonio Breast Cancer Symposium, San Antonio, TX, USA, 4–8 December 2018. [Google Scholar]
  108. Sun, W.Y.; Lee, Y.K.; Koo, J.S. Expression of PD-L1 in triple-negative breast cancer based on different immunohistochemical antibodies. J. Transl. Med. 2016, 14, 173. [Google Scholar] [CrossRef] [Green Version]
  109. Denkert, C.; Von Minckwitz, G.; Brase, J.C.; Sinn, B.V.; Gade, S.; Kronenwett, R.; Pfitzner, B.M.; Salat, C.; Loi, S.; Schmitt, W.D.; et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J. Clin. Oncol. 2015, 33, 983–991. [Google Scholar] [CrossRef]
  110. Gruosso, T.; Gigoux, M.; Manem, V.S.K.; Bertos, N.; Zuo, D.; Perlitch, I.; Saleh, S.M.I.; Zhao, H.; Souleimanova, M.; Johnson, R.M.; et al. Spatially distinct tumor immune microenvironments stratify triple-negative breast cancers. J. Clin. Investig. 2019, 129, 1785–1800. [Google Scholar] [CrossRef] [Green Version]
  111. Telli, M.L.; Timms, K.M.; Reid, J.; Hennessy, B.; Mills, G.B.; Jensen, K.C.; Szallasi, Z.; Barry, W.T.; Winer, E.P.; Tung, N.M.; et al. Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer. Clin. Cancer Res. 2016, 22, 3764–3773. [Google Scholar] [CrossRef] [Green Version]
  112. Sharma, P.; Klemp, J.R.; Kimler, B.F.; Mahnken, J.D.; Geier, L.J.; Khan, Q.J.; Elia, M.; Connor, C.S.; McGinness, M.K.; Mammen, J.M.W.; et al. Germline BRCA mutation evaluation in a prospective triple-negative breast cancer registry: Implications for hereditary breast and/or ovarian cancer syndrome testing. Breast Cancer Res. Treat. 2014, 145, 707–714. [Google Scholar] [CrossRef]
  113. Shimelis, H.; LaDuca, H.; Hu, C.; Hart, S.N.; Na, J.; Thomas, A.; Akinhanmi, M.; Moore, R.M.; Brauch, H.; Cox, A.; et al. Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J. Natl. Cancer Inst. 2018, 110, 855–862. [Google Scholar] [CrossRef]
  114. Akashi-Tanaka, S.; Watanabe, C.; Takamaru, T.; Kuwayama, T.; Ikeda, M.; Ohyama, H.; Mori, M.; Yoshida, R.; Hashimoto, R.; Terumasa, S.; et al. BRCAness predicts resistance to taxane-containing regimens in triple negative breast cancer during neoadjuvant chemotherapy. Clin. Breast Cancer 2015, 15, 80–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Manié, E.; Popova, T.; Battistella, A.; Tarabeux, J.; Caux-Moncoutier, V.; Golmard, L.; Smith, N.K.; Mueller, C.R.; Mariani, O.; Sigal-Zafrani, B.; et al. Genomic hallmarks of homologous recombination deficiency in invasive breast carcinomas. Int. J. Cancer 2016, 138, 891–900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sharma, P.; Barlow, W.E.; Godwin, A.K.; Pathak, H.; Isakova, K.; Williams, D.; Timms, K.M.; Hartman, A.R.; Wenstrup, R.J.; Linden, H.M.; et al. Impact of homologous recombination deficiency biomarkers on outcomes in patients with triple-negative breast cancer treated with adjuvant doxorubicin and cyclophosphamide (SWOG S9313). Ann. Oncol. 2018, 29, 654–660. [Google Scholar] [CrossRef] [PubMed]
  117. Couch, F.J.; Hart, S.N.; Sharma, P.; Toland, A.E.; Wang, X.; Miron, P.; Olson, J.E.; Godwin, A.K.; Pankratz, V.S.; Olswold, C.; et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J. Clin. Oncol. 2015, 33, 304–311. [Google Scholar] [CrossRef]
  118. Telli, M.L.; Hellyer, J.; Audeh, W.; Jensen, K.C.; Bose, S.; Timms, K.M.; Gutin, A.; Abkevich, V.; Peterson, R.N.; Neff, C.; et al. Homologous recombination deficiency (HRD) status predicts response to standard neoadjuvant chemotherapy in patients with triple-negative or BRCA1/2 mutation-associated breast cancer. Breast Cancer Res. Treat. 2018, 168, 625–630. [Google Scholar] [CrossRef]
  119. Loibl, S.; Weber, K.E.; Timms, K.M.; Elkin, E.P.; Hahnen, E.; Fasching, P.A.; Lederer, B.; Denkert, C.; Schneeweiss, A.; Braun, S.; et al. Survival analysis of carboplatin added to an anthracycline/taxane-based neoadjuvant chemotherapy and HRD score as predictor of response-final results from GeparSixto. Ann. Oncol. 2018, 29, 2341–2347. [Google Scholar] [CrossRef]
  120. Gruber, J.J.; Afghahi, A.; Hatton, A.; Scott, D.; McMillan, A.; Ford, J.M.; Telli, M.L. Talazoparib beyond BRCA: A phase II trial of talazoparib monotherapy in BRCA1 and BRCA2 wild-type patients with advanced HER2-negative breast cancer or other solid tumors with a mutation in homologous recombination (HR) pathway genes. JCO 2019, 37, 3006. [Google Scholar] [CrossRef]
  121. Barker, A.D.; Sigman, C.C.; Kelloff, G.J.; Hylton, N.M.; Berry, D.A.; Esserman, L.J. I-SPY 2: An adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin. Pharmacol. Ther. 2009, 86, 97–100. [Google Scholar] [CrossRef]
  122. McNeil, C. NCI-MATCH launch highlights new trial design in precision-medicine era. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef]
  123. Flaherty, K.T.; Gray, R.; Chen, A.; Li, S.; Patton, D.; Hamilton, S.R.; Williams, P.M.; Mitchell, E.P.; Iafrate, A.J.; Sklar, J.; et al. The Molecular Analysis for Therapy Choice (NCI-MATCH) Trial: Lessons for Genomic Trial Design. J. Natl. Cancer Inst. 2020. [Google Scholar] [CrossRef] [Green Version]
  124. Symmans, W.F.; Peintinger, F.; Hatzis, C.; Rajan, R.; Kuerer, H.; Valero, V.; Assad, L.; Poniecka, A.; Hennessy, B.; Green, M.; et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J. Clin. Oncol. 2007, 25, 4414–4422. [Google Scholar] [CrossRef] [PubMed]
  125. Symmans, W.F.; Wei, C.; Gould, R.; Yu, X.; Zhang, Y.; Liu, M.; Walls, A.; Bousamra, A.; Ramineni, M.; Sinn, B.; et al. Long-Term Prognostic Risk After Neoadjuvant Chemotherapy Associated With Residual Cancer Burden and Breast Cancer Subtype. J. Clin. Oncol. 2017, 35, 1049–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Yau, C.; Van der Noordaa, M.; Wei, J.; Osdoit, M.; Reyal, F.; Hamy, A.-S.; Lae, M.; Martin, M.; Del Monte, M.; Boughey, J.C.; et al. Abstract GS5-01: Residual cancer burden after neoadjuvant therapy and long-term survival outcomes in breast cancer: A multi-center pooled analysis. Cancer Res. 2020, 80. [Google Scholar] [CrossRef]
  127. Gluz, O.; Kolberg-Liedtke, C.; Prat, A.; Christgen, M.; Gebauer, D.; Kates, R.; Paré, L.; Grischke, E.-M.; Forstbauer, H.; Braun, M.; et al. Efficacy of deescalated chemotherapy according to PAM50 subtypes, immune and proliferation genes in triple-negative early breast cancer: Primary translational analysis of the WSG-ADAPT-TN trial. Int. J. Cancer 2020, 146, 262–271. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of PI3K/AKT/mTOR pathway activation and targeted therapies. Activating mutations in the α catalytic domain of PI3K and/or PTEN mutation lead to pathway activation. PI3K signaling pathway linking RTK signaling leads to downstream activation of PI3K/AKT/mTOR, promoting cell proliferation and survival. RTK receptor tyrosine kinase, PI3K phosphatidylinositol-3-kinase, PTEN phosphatase and tensin homolog, AKT Protein Kinase B, mTORC mechanistic target of rapamycin complex.
Figure 1. Mechanisms of PI3K/AKT/mTOR pathway activation and targeted therapies. Activating mutations in the α catalytic domain of PI3K and/or PTEN mutation lead to pathway activation. PI3K signaling pathway linking RTK signaling leads to downstream activation of PI3K/AKT/mTOR, promoting cell proliferation and survival. RTK receptor tyrosine kinase, PI3K phosphatidylinositol-3-kinase, PTEN phosphatase and tensin homolog, AKT Protein Kinase B, mTORC mechanistic target of rapamycin complex.
Cancers 12 01404 g001
Table 1. Triple negative breast cancer (TNBC) molecular subtype and potential targets for therapy.
Table 1. Triple negative breast cancer (TNBC) molecular subtype and potential targets for therapy.
Molecular SubtypesGenomic AlterationsPotential Therapeutic Targets
Basal-like 1 (BL1)Cell cycle
DNA repair (ATR–BRCA pathway)
Proliferation
PARP inhibitors
Carboplatin, Cisplatin
Other chemotherapy
Basal-like 2 (BL2)Growth factor signaling pathways (EGFR, MET, NGF, Wnt/β-catenin, IGF-1R)
Glycolysis, gluconeogenesis
Expression of myoepithelial markers
mTOR inhibitors
Growth-factor inhibitors
Immunomodulatory (IM)Immune cell processes (CTLA4, IL12, IL7 pathways, antigen processing/presentation)
Gene signature for medullary BC (rare TNBC with a favorable prognosis)
PD1/PD-L1 inhibitors
Other immune checkpoint inhibitors
Mesenchymal-like (M)Cell motility
Cell differentiation
Growth factor signaling (Notch, PDGFR, FGFR, TGFβ)
EMT
mTOR inhibitors
EMT-targeted therapy
CSC-targeted therapy
AXL inhibitor
Mesenchymal stem-like (MSL)Low proliferation
Angiogenesis genes
Similar to Mesenchymal-like (M)
PI3K inhibitors
Antiangiogenic therapy
Src antagonist
Luminal androgen receptor (LAR)Androgen receptor
Luminal gene expression pattern
Molecular apocrine subtype
Antiandrogen blockade
CDK4/6 inhibitors
Immune checkpoint inhibitors
Modified from Lehmann et al. [12,14] and Collignon et al. [27];AXL, tyrosine-protein kinase receptor UFO; CSC, cancer stem cells; EGFR, epidermal growth factor receptor; EMT, epithelial-mesenchymal-transition; FGFR, fibroblast growth factor receptors; IGF-1R, insulin-like growth factor receptor; IL, interleukin; MET, hepatocyte growth factor; mTOR,, mammalian target of rapamycin; NGF, nerve growth factor; PARP, poly ADP ribose polymerase; PDGFR, platelet-derived growth factor receptors; PD1, programmed cell death 1; PD-L1, programmed death-ligand 1; PI3K, phosphatidylinositol 3-kinase; TGFβ, transforming growth factor beta.
Table 2. Pathological complete response (pCR) rate in neoadjuvant trials with carboplatin in early stage TNBC.
Table 2. Pathological complete response (pCR) rate in neoadjuvant trials with carboplatin in early stage TNBC.
TrialsTreatmentNumber of Patients with TNBC*pCR Ratep-Value
GEICAM/2006–03 [40]EC followed by T + carboplatin
vs. without carboplatin
48 vs. 4630% in both armsN/A
GeparSixto
GBG66 [36]
P and NPLD with Bev + carboplatin vs. without carboplatin158 vs. 15753.2% vs. 36.9%0.005
GALGB 40603
Alliance [34]
(weekly) P + carboplatin, followed by ddAC (with or without Bev)
vs. without carboplatin
221 vs. 21254% vs. 41%0.0029
Ando et al. [41](weekly) P + carboplatin followed by EC/5-FU vs. without carboplatin37 vs. 3861.2% vs. 26.3%0.003
Zhang et al. [42]P + carboplatin vs. P + E47 vs. 4438.6% vs. 14.0%0.014
GeparOcto
GBG84 [43]
(weekly) P and NPLD + carboplatin vs. E followed by P followed by C203 vs. 20051.7% vs. 48.50.518
WSG-ADAPT-TN [44]Nab-P + carboplatin vs. Nab-P + G154 vs. 18245.9% vs. 28.7%0.002
BrighTNess [45]P + carboplatin followed by AC vs. without carboplatin160 vs. 15858% vs. 31%0.0001
Sharma et al. [39]T + carboplatin19055%N/A
Yuan et al. [38]Nab-P +carboplatin6748%N/A
E: epirubicin; C: cyclophosphamide; T: docetaxel; P: paclitaxel; NPLD: non-pegylated liposomal doxorubicin; Bev: bevacizumab; A: doxorubicin; dd: dose-dense; G: gemcitabine; N/A: not applicable *pCR in the both breast and axilla (ypT0/is ypN0).
Table 3. Immune checkpoint inhibitor trials in early stage TNBC.
Table 3. Immune checkpoint inhibitor trials in early stage TNBC.
TrialsTreatmentNumber of Patients with TNBC*pCR Ratep-Value
GeparNuevo [59]Nab P + durvalumab followed by EC + durvalumab vs. without durvalumab88 vs. 8653.4% vs. 44.2%0.287
KEYNOTE 173 [16]Nab P with or without Cb + pembrolizumab followed by AC6060%N/A
ISPY2 [57](weekly) P + pembrolizumab followed by AC vs. without pembrolizumab29 vs.60% vs. 22%N/A
KEYNOTE 522 [15]Cb/P + pembrolizumab followed by AC or EC + pembrolizumab vs. without pembrolizumab401 vs. 20164.8% vs. 51.2%<0.001
NeoTRIP (2019, abstract) [60]Cb and Nab P + atezolizumab vs. without atezolizumab138 vs. 14243.5% vs. 40.8%0.66
NeoPACT (NCT03639948)Phase II trial of Cb and T + pembrolizumabRecruiting with accrual goal of 100N/AN/A
Impassion 031 (NCT 03197935) [62]Phase III trial of Nab P + atezolizumab followed by ddAC + atezolizumab (continue atezolizumab as adjuvant after surgery)Completed accrual with 204 patientsN/AN/A
GeparDouze (NCT03281954) [61]Phase III trial of (weekly) P and Cb + atezolizumab followed by AC or EC + atezolizumabRecruiting with accrual goal of 1520N/AN/A
P: paclitaxel; Cb: carboplatin; A: doxorubicin; C: cyclophosphamide; E: epirubicin; T: docetaxel; dd: dose-dense; N/A: not applicable. *pCR in the both breast and axilla (ypT0/is ypN0).
Table 4. Poly-ADP-ribosyl polymerase (PARP) inhibitors in neoadjuvant TNBC trials.
Table 4. Poly-ADP-ribosyl polymerase (PARP) inhibitors in neoadjuvant TNBC trials.
TrialsTreatmentNumber of Patients with TNBC*pCR Ratep-Value
I-SPY 2 [69]P and Cb + veliparib followed by AC vs. P followed by AC39 vs. 2151% vs. 26%Not reported (95% PI, 33–66% vs. 9–43%)
BrighTNess [45]Arm 1: P and Cb + veliparib
Arm 2: P and Cb
Arm 3: P
All arms followed by AC
316 vs. 169 vs. 5853% vs. 58% vs. 31%Arm 1 vs 2: 0.357
Arm 1 vs. 3: <0.0001
GeparOLA [71]P+ olaparib vs. P + Cb, followed by EC50 vs. 2756.0% vs. 59.3%Not reported
NCT02401347Phase II of talazoparibRecruiting with accrual goal of 40N/AN/A
P: paclitaxel; Cb: carboplatin; AC: doxorubicin and cyclophosphamide; EC: epirubicin and cyclophosphamide; N/A: not applicable. *pCR in the both breast and axilla (ypT0/is ypN0).

Share and Cite

MDPI and ACS Style

Lee, J.S.; Yost, S.E.; Yuan, Y. Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges. Cancers 2020, 12, 1404. https://doi.org/10.3390/cancers12061404

AMA Style

Lee JS, Yost SE, Yuan Y. Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges. Cancers. 2020; 12(6):1404. https://doi.org/10.3390/cancers12061404

Chicago/Turabian Style

Lee, Jin Sun, Susan E. Yost, and Yuan Yuan. 2020. "Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges" Cancers 12, no. 6: 1404. https://doi.org/10.3390/cancers12061404

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