• Open Access
    Review

    Early-stage triple negative breast cancer: the therapeutic role of immunotherapy and the prognostic value of pathological complete response

    Pierluigi De Santis 1
    Martina Perrone 1
    Chiara Guarini 1
    Anna Natalizia Santoro 1
    Carmelo Laface 1
    Daniela Carrozzo 1
    Gaia Rachele Oliva 2
    Palma Fedele 1*

    Explor Target Antitumor Ther. 2024;5:232–250 DOI: https://doi.org/10.37349/etat.2024.00215

    Received: May 26, 2023 Accepted: December 26, 2023 Published: February 28, 2024

    Academic Editor: Laura Cerchia, Institute of Experimental Endocrinology and Oncology “G. Salvatore”-National Research Council (IEOS-CNR), Italy; Simona Camorani, Institute of Experimental Endocrinology and Oncology “G. Salvatore”-National Research Council (IEOS-CNR), Italy

    This article belongs to the special issue Innovative Strategies to Target Triple-negative Breast Cancer

    Abstract

    Triple negative breast cancer (TNBC) represents an aggressive disease associated with a high risk of recurrence after curative treatment and a poor prognosis in the metastatic setting. Chemotherapy was for years the only treatment available in the early and metastatic setting, due to the lack of actionable targets. Clinical practice has changed following the results obtained with the addition of immunotherapy to standard chemotherapy, the development of novel drugs [i.e. antibody-drug conjugates (ADCs)], and the use of targeted treatments for patients carrying germline pathogenic breast cancer susceptibility genes (BRCA) 1 or BRCA 2 variants. The treatment of early-stage disease has had a shift in clinical practice since July 2021, after the Food and Drug Administration (FDA) approval of pembrolizumab in association with chemotherapy as neoadjuvant treatment for TNBC and as a single agent in the subsequent adjuvant setting. This intensive treatment based on the combination of a poly-chemotherapy and an immune checkpoint inhibitor (ICI) led to the improvement of short- and long-term outcomes, but it has highlighted some new unmet clinical needs in the treatment of early-stage TNBC: the selection of the most effective adjuvant therapy and the integration of pembrolizumab with other therapeutic strategies [capecitabine, poly(ADP-ribose) polymerase (PARP) inhibitors] based on the achievement of pathologic complete response (pCR); the identification of predictive biomarkers to select patients who could most benefit from the addition of ICI, to minimize toxicities and to maximize outcomes; the possibility of de-escalating chemotherapy in favor of immune-combo or novel agents, such as ADCs; the role of immunotherapy in estrogen receptor (ER)-low patients. The advent of immunotherapy not only addresses current challenges in TNBC treatment but also holds the promise of a radical transformation in its therapeutic paradigm, enhancing significantly clinical outcomes and offering new perspectives for patients grappling with this aggressive form of breast cancer.

    Keywords

    Triple negative breast cancer, immunotherapy, pathological complete response, neoadjuvant combination treatment, adjuvant treatment

    Introduction

    Triple negative Breast Cancer (TNBC) is a histological subtype of breast cancer (BC) characterized by the immunohistochemical lack of expression (< 1%) of estrogen receptor (ER), progesterone receptor (PgR), and human epidermal growth factor receptor 2 (HER2). It accounts for approximately 10–20% of all BC, affecting mainly young, premenopausal women, and individuals with inherited gene alterations, such as BC susceptibility genes 1/2 (BRCA 1/2) mutations [13]. It notably presents an aggressive biological behavior with a trend to have a higher grade and an often lymph node involvement at diagnosis, an inclination to metastasize after curative treatment, and a poorer prognosis in metastatic setting when compared with other BC subtypes [4, 5].

    For decades, treatment for early TNBC has been based on surgery and subsequent adjuvant chemotherapy (CHT) for the reduction of disease recurrence [6]. Therefore, conventional cytotoxic CHT has represented the backbone of systemic treatment in the early TNBC, including neoadjuvant treatment, which used to reduce tumor size in larger tumors increasing the chances of a breast-conserving surgery [7, 8]. In recent years the development of novel therapeutic approaches has been difficult, due to the heterogeneity of TNBC and lack of therapeutic targets [9, 10]. Nevertheless, immunotherapy and poly(ADP-ribose) polymerase (PARP) inhibitors have shown survival benefits in recent studies.

    Specifically, combinations of immune checkpoint inhibitors (ICIs) with CHT or other alternative therapeutic compounds could emerge as a successful therapeutic approach in the management of TNBC patients. Despite the progress in ICIs representing a notable milestone in TNBC treatment, additional investigations are necessary to tackle this issue comprehensively. A profound comprehension of tumor subtypes, alongside tumor microenvironment (TME) and in terms of molecular, genetic, and immune aspects, would amplify the potential for developing targeted immunotherapy to achieve superior therapeutic effectiveness, especially in TNBC [11].

    Therefore, in this review, we aimed to investigate the role of immunotherapy in early-stage TNBC, the prognostic value of pathologic complete response (pCR) with its therapeutic implications, and the future perspectives regarding the systemic treatment of early TNBC, including the discovery of new biomarkers.

    The landscape of immunotherapy in TNBC

    The immune system plays a crucial role in TNBC compared to the other molecular subtypes of BC. Although originally BC was considered non-immunogenic, TNBC has a high immunogenic potential, making it a promising candidate for immunotherapy, especially with ICIs [12, 13]. TNBC immunogenicity is related to intrinsic tumor cell signatures and tumoral surrounding microenvironment features.

    Over the last decades thanks to emerging technologies such as next-generation sequencing (NGS), the knowledge of the molecular and genetic background of TNBC improved, bringing to light its intertumoral and intratumoral heterogeneity.

    A first classification divided TNBC into six subtypes: basal-like 1 (BL1), basal-like 2 (BL2), mesenchymal (M), M stem-like (MSL), immunomodulatory (IM), and luminal androgen receptor (LAR) [14].

    Subsequently, analyzing RNA and DNA-based profiles of 198 TNBC tumors, a four-type classification of TNBC was shaped: basal-like immunosuppressed (BLIS), basal-like immune-activated (BLIA), M and LAR [15]. This classification was further revised with the identification of four specific TNBC subtypes: BL1, BL2, M, and LAR, omitting IM and MSL because of the dependence of these two subtypes on the TME features [14].

    In addition, TNBC could be classified into three microenvironment phenotypes or clusters:

    • Cluster 1: “immune-desert” with poor immune cell permeation, due to a high presence of MYC amplifications and, consequently, a lower recruitment of innate immune cells.

    • Cluster 2: “innate immune-inactivated” characterized by a hyper-activation of phosphatidylinositide 3-kinase/protein kinase B (PI3K-AKT) pathway in tumor cells, low tumor antigen burden and infiltration of deactivated innate immune cells, fibroblasts, and endothelial cells. Clusters 1 and 2 are therefore referred to as “cold tumors”.

    • Cluster 3: “immune-inflamed”, the so-called “hot tumor” that represents about 30% of TNBCs and is characterized by an abundant adaptive and innate immune cells infiltration and with a high expression of immune checkpoint molecules [16].

    The potential “hot” conversion of “cold” tumors could improve the efficacy of cancer immunotherapy. For example, local IM therapies can express a synergistic effect with immunotherapy by acting on components of the TME and immune system function, such as elevating the expression of tumor antigens and increasing the recruitment of activated immune cells in the TME [17].

    TNBC cancer cell immunological features include genomic instability and high tumor mutational burden (TMB), resulting in more somatic mutations and neoantigens [18].

    Moreover, approximately 10–20% of TNBC harbor BRCA 1 or BRCA 2 germinal mutations, with a consequent hereditary deficit in the DNA repair mechanism and strong genomic instability. Several studies have demonstrated that TNBC-carrying BRCA mutations are more sensitive to DNA-damaging drugs such as anthracyclines, but also platinum agents and PARP inhibitors [1921]. Sensitivity to these drugs was also observed in tumors with alterations in other genes, sharing BRCA-mutant phenotype in the absence of a BRCA 1/2 mutation, namely “BRCAness” [22, 23].

    Tumors with BRCA 1/2 mutations or BRCAness TNBC are more immunogenic than TNBC without these genetic alterations [2426].

    Compared to the other BC subtypes the immunogenic TME features in TNBC consist of higher levels of vascular endothelial growth factor (VEGF), that promote tumor cell growth and migration such as mitogen-activated protein kinases (MAPKs), tumor-associated macrophages (TAMs), and tumor-infiltrating lymphocytes (TILs), white blood cells that migrate towards the tumor, leading to an important immunogenic effect and consequently that are involved in killing cancer cells [2729].

    TAMs regulate the interaction between the immune system and cancer cells. CD163+ M2 macrophages, which are associated with tumors characterized by higher proliferation and poorer differentiation [30], are more present in TNBC and basal-like BC [31]. A prosperous infiltration of TILs is found in TNBC tumors and the stroma surrounding them, with a recognized predictive and prognostic role, specifically for CD4+ CD8+ T cells [32]. Several studies have reported better response to neoadjuvant CHT (NACT) [33] and better clinical outcomes in BC with high TIL infiltrate [3439]. Based on this evidence, the international TILs working group started standardizing the evaluation of BC TILs to use it in clinical practice identifying those patients that may benefit from emerging immunotherapies with ICIs or combination therapies [40].

    All these TME elements contribute to TNBC immunogenicity which also appears to be closely related to the concept of TMB, depending on the ineffective DNA repair system with the consequent generation of high rates of neoantigens. The upregulated antigen presentation system leads to an increasing number of innate and adaptative immune cells and many cytokines interplaying with cancer cells. However, the exact relationship between TMB, neoantigens, and immune infiltration is not yet completely understood, and some studies have reported an inverse association between immune cells in TME and the rate of somatic copy number alterations [41, 42].

    Moreover, although TMB is comparable across the three clusters of TNBC, the “immune-inflated” phenotype is characterized by a higher degree of immune cells in the TME, but also a high expression of immune checkpoints by cancer cells [16]. The rate of TILs, indeed, has been positively related to programmed death ligand 1 (PD-L1) expression [43]. PD-L1 is an immune checkpoint that mediates local immune escape in many tumors inducing saturation of activated T cells. Even if PD-L1 prognostic role is yet controversial [44], however it results more overexpressed in TNBC compared with other BC and it can predict responsiveness to immunotherapy [16].

    Therefore, TNBC represents an aggressive BC subtype, associated with high mutational load, high tumor immunogenicity and TME diversity.

    New paradigms in early TNBC: from CHT to immunotherapy

    CHT in adjuvant treatment for TNBC

    In early TNBC patients, CHT represents the mainstay of adjuvant and neoadjuvant treatments. Adjuvant CHT is recommended for tumor sizes greater than 1.0 cm and patients with nodal involvement, regardless of tumor size. Therefore, it can be considered for tumor sizes between 0.6–1.0 cm [45]. A recent large meta-analysis demonstrated that adjuvant CHT with anthracyclines-containing regimens plus taxanes, compared with no CHT, can reduce BC mortality rates by about 40% during the first decade after diagnosis. Moreover, regimens with higher cumulative and dose-dense schedules of anthracycline (with granulocyte colony-stimulating factor support) have shown better survival benefits and more reductions in recurrence [46]. Three-weekly docetaxel and paclitaxel can be considered in adjuvant setting, but weekly paclitaxel, in a subgroup analysis, has shown improved outcomes and is preferred for TNBC [47]. In TNBC in frail patients with a known history of heart disease, to minimize the cardiotoxicity of adjuvant treatments, docetaxel combined with cyclophosphamide (TC) has proven to be a viable alternative to doxorubicin and cyclophosphamide (AC), demonstrating a favorable disease-free survival (DFS) [48]. Therefore, there is a broad spectrum of chemotherapeutic treatments for early TNBC that should be customised according to the patient and expected toxicities.

    The role of platinum in adjuvant setting for TNBC

    TNBC patients commonly harbor BRCA 1/2 or BRCAness mutations with a homologous recombination deficiency (HRD) that makes them particularly susceptible to platinum agents due to their ability to hit cancer cells that have deficient DNA repair mechanisms [4951]. Several retrospective single-center studies have explored the role of adjuvant platinum combined with standard anthracycline and taxane-based regimens, with controversial results not showing clear clinical benefits [52, 53]. Nevertheless, a recent phase III trials have demonstrated a longer 5-year DFS (86.5% vs. 80.3%) with similar results in distant DFS and relapse-free survival (RFS) of platinum-containing adjuvant regimens (paclitaxel-carboplatin) compared to a standard anthracyclines-containing regimen followed by taxane, however with no benefit in overall survival (OS) [54].

    Another important factor is platinum resistance. Platinum sensitivity may be affected by changes in the hazard ratio (HR) pathway or, in the case of patients with BRCA 1/2 mutations, by the secondary appearance of new BRCA 1 or 2 mutations that make cancer cells less sensitive to platinum [55, 56].

    Other mechanisms of resistance to platinum compounds are:

    • Modification of drug transport within the tumor cell, by determining decreased influx or increased efflux.

    • Increase of detoxification systems.

    • Decrease of cell apoptosis [57].

    Therefore, the benefit of adjuvant platinum-based regimens remains controversial and needs validation by prospective adjuvant ongoing trials.

    Neoadjuvant treatments for TNBC

    NACT

    Several treatment guidelines recommend NACT as the preferred option for stage II or III TNBC and for stage I with a tumor size greater than 1 cm. It can be considered in stage I TNBC with a tumor size from 0.6 cm to 1 cm and/or in the case of tumors with nodal micrometastases. [6, 45]. There is no significant difference in survival benefits between patients receiving neoadjuvant or adjuvant CHT after surgical resection. However, neoadjuvant treatments can be useful for inoperable tumors rendering them operable and they can also downstage patients with operable BC promoting breast-conservation [58, 59]. The use of neoadjuvant treatments provides important prognostic information based on response to therapy. Achieving a pCR, defined as the lack of cancer cells in tissue samples of breast and axillary lymph nodes, after a neoadjuvant treatment, is associated with favorable disease-free and OS in early TNBC, as demonstrated in Collaborative Trials in Neoadjuvant Breast Cancer (CTNeoBC) pooled analysis. In this study, patients with early BC treated with NACT and followed by surgery who obtained pCR (ypT0 ypN0, ypT0/is ypN0) were associated with improved event-free survival (EFS) and OS, especially in TNBC (HR = 0.24 and HR = 0.16, respectively) [33]. Like adjuvant treatment, traditional NACT is based on anthracyclines and taxanes, and a dose-dense regimen is preferred in neoadjuvant settings based on proven improved DFS and OS in a large meta-analysis [60].

    In recent years, the use of platinum-based combination regimens has been the focus of neoadjuvant treatment to increase the rate of pCR in TNBC. Three recent studies demonstrated that combining platinum with taxane and anthracycline led to an improvement in the pCR rate in TNBC, with a similar survival benefit [6163]. In Brightness Trial patients with II–III stage TNBC were randomly assigned to receive paclitaxel alone, paclitaxel and carboplatin and this combination with a PARP inhibitor, veliparib followed by AC. Although the addition of veliparib and carboplatin was associated with an increase of patients who achieved a pCR compared to paclitaxel alone (53% vs. 31%, P < 0.0001), but not to paclitaxel and carboplatin, this benefit could be related to the addition of the carboplatin [63]. The initial rationale for using the combination of platinum in NACT was that sporadic TNBC can show BRCAness with a major response to platinum regimens [50, 51]. However, the greatest benefit was seen in patients who were germline BRCA wild type, and only a marginal benefit was observed in the germline BRCA mutant subgroup, as was shown in the recent GeparOLA trial. In this trial, patients were randomized to neoadjuvant therapy with paclitaxel and carboplatin vs. neoadjuvant therapy with paclitaxel and olaparib (PARP inhibitor). In both arms, the combination of epirubicin and cyclophosphamide was administered next. This study, although limited by a small number of patients enrolled, showed an advantage for the carboplatin arm in patients without BRCA mutation (germ or somatic) and high HRD. The 4-year invasive DFS (iDFS) rate with olaparib-paclitaxel was 81.2% vs. 93.4% with carboplatin-paclitaxel (CP) [HR = 3.03; 95% confidence interval (CI) = 0.67–13.67; log-rank P = 0.1290]. The 4-year OS rate was 89.2% with the olaparib combination vs. 96.6% with carboplatin (HR = 3.27; 95% CI = 0.39–27.20; log-rank P = 0.2444). The trend of the iDFS curves was similar in the two treatment arms and independent of germline or somatic BRCA mutation [64].

    Platinum combinations are currently recommended for selected patients with TNBC who require adequate local control before surgical resection [45]. A more recent phase III trial presented at the San Antonio Breast Cancer Symposium evaluating the efficacy and safety of adding carboplatin to standard sequential taxane-anthracycline NACT in patients with TNBC who had no evidence of metastatic disease, has observed improvements in terms of DFS (5-year DFS were 70.6% and 64.5% respectively with a HR = 0.79, 95% CI = 0.61–1.02, P = 0.073) and OS (5-year OS were 74.0% and 66.7% respectively with a HR = 0.75, 95% CI = 0.57–0.98, P = 0.034) with the addition of carboplatin, but these benefits were limited to patients who were 50 years of age or younger. Therefore, the pCR in the intention-to-treat population was 54.5% in the carboplatin arm and 40.3% in the control arm (P < 0.001) [65].

    The inclusion of platinum agents as NACT for TNBC remains controversial. Long-term outcomes and new prospective studies are needed to clarify the role of platinum agents in this setting.

    Neoadjuvant immunotherapy

    The success of ICIs in metastatic TNBC led to expand their role in neoadjuvant settings. Pembrolizumab and atezolizumab have shown progression-free survival (PFS) benefits in phase III trials in advanced setting [66, 67]. In contrast to atezolizumab that showed conflicting results [68, 69], pembrolizumab consistently showed OS benefits in advanced TNBC [66, 70]. In early-stage TNBC two studies evaluated atezolizumab in neoadjuvant setting. In the NeoTRIPaPDL1 trial, no improvement in pCR was shown with the addition of atezolizumab to a non-anthracycline-containing CHT regimen [71]. More recent Impassion031 phase III study evaluating the association of atezolizumab to a standard NACT (nab-palclitaxel weekly for 12 weeks followed by 4 cycles of AC), has demonstrated a significant improvement of pCR rates in intention to treat (ITT) population (58% in atezolizumab arm vs. 41% in placebo arm, P = 0.0044), regardless of PD-L1 status, meeting the primary endpoint of the study [72]. Therefore, in early BC the combination of pembrolizumab with paclitaxel-carboplatin followed by anthracycline increased pCR rate and EFS rate in the KEYNOTE-522 trial, representing a turning point for the role of immunotherapy in neoadjuvant therapy of TNBC and establishing pembrolizumab as a standard treatment during neoadjuvant treatment for stage II and III TNBC. The trial evaluated the combination of pembrolizumab (18 cycles, 200 mg every 3 weeks) combined with four cycles of paclitaxel (weekly or 3-weekly) and carboplatin (3-weekly), followed by 3-weekly AC for 4 cycles, compared to placebo with CHT. Pembrolizumab arm showed a 13.6% improvement in pCR [64.8% (95% CI = 59.9–69.5%) vs. 51.2% (95% CI = 44.1–58.3%)] and in EFS rate [84.5% (95% CI = 81.7–86.9%) vs. 76.8% (95% CI = 72.2–80.7%)], meeting the primary endpoint of the study, regardless nodal involvement and PD-L1 status. The average duration of follow-up is still immature, but a trend of superiority in terms of OS in the pembrolizumab arm was nevertheless detected [73]. Limits of this study are the lack of biomarkers that predict what patient may benefit from the addition of pembrolizumab and the non-utilization of dose-dense schedule of AC which showed superior OS benefit in the neoadjuvant setting in TNBC [74].

    Moreover, the recent GeparNuevo trial showed that durvalumab (1,500 mg every 4 weeks) added to NACT consisting of nab-paclitaxel 125 mg/m2 weekly for 12 weeks, followed by epirubicin/cyclophosphamide every 2 weeks, in early TNBC significantly improved iDFS (85.6% with durvalumab vs. 77.2% with placebo HR = 0.48, 95% CI = 0.24–0.97, stratified log-rank P = 0.036) and OS (95.2% vs. 83.5% with a HR = 0.24, 95% CI = 0.08–0.72, P = 0.006), despite a modest pCR increase and no adjuvant component of durvalumab [75]. Future studies should aim to define the role of immunotherapy in the treatment of early TNBC, to define the ideal duration of these treatments, and should research new biomarkers to personalize treatments.

    Pathological complete response: prognostic role and therapeutic implications

    In clinical practice, the achievement of pCR after neoadjuvant treatment is correlated to the improvement of long-term benefits concerning EFS and OS. Its prognostic value is greatest in aggressive tumor subtypes, like in TNBC (EFS: HR = 0.24; OS: HR = 0.16) [33]. Patients who have residual invasive BC after the receipt of NACT have a high risk of relapse. Patients with TNBC who do not experience pCR have an estimated 5-year EFS of 57% and OS of 47% (compared with 90% EFS and 84% OS, respectively, for patients with early-stage TNBC who demonstrate pCR) [76, 77].

    After pre-operative CHT and surgical treatment, patients can receive postoperative radiation therapy (RT). Patients with hormone receptor-positive BC [hormone receptor-positive (HR+) BC] are candidates for adjuvant endocrine treatment. However, until recently, no adjuvant CHT was expected as standard in patients with TNBC. Only follow-up was recommended in those who have pCR or in those with residual invasive BC after the receipt of neoadjuvant regimens [78]. To address the unmet clinical need for optimal adjuvant treatment in the subgroup of patients with TNBC at high risk of recurrence (those who have not achieved the pCR after NACT containing anthracycline, taxane, or both), the Capecitabine for Residual Cancer as Adjuvant Therapy (CREATE-X) was designed. The trial did not include only patients with TNBC but also patients with HR+ HER2 negative BC [79]. The results of this phase III trial showed that the addition of adjuvant capecitabine (1,250 mg per square meter of body-surface area, twice per day, on days 1 to 14, every 3 weeks for six or eight cycles) was safe and effective in prolonging DFS and OS among the ITT population. The study showed a superior DFS in the capecitabine group than in the control group (74.1% vs. 67.6% at 5 years; HR = 0.70; P = 0.01). Therefore, OS was longer in the experimental group: 89.2% vs. 83.6% of the patients were alive at 5 years (HR = 0.59; P = 0.01). Thirty percent of the patients had triple negative (TN) disease, and they represent the subgroup with poor prognosis (approximately half the patients with TNBC who had a pCR did not have the recurrence of the disease) [33]. The benefit of capecitabine vs. control in DFS and OS was notable among this subgroup of patients (HR = 0.58 and HR = 0.52, respectively) [79].

    The reflection in the treatment algorithm due to these results was significant.

    Some limits of this study are the exclusion of patients who had reached the pCR, for whom only follow-up was indicated, and the lack of efficacy results selected for residual cancer burden (RCB). The RCB quantifies the extent of residual disease after neoadjuvant treatment at the time of surgery. This score uses the diameter of residual disease, percentage of vital tumor cells, and diameter of the largest involved lymph node to calculate the amount of residual disease. It has been validated with distinct prognostic RCB classes in all BC subtypes, with the most significant discriminatory power in TN and Her-2 positive BC. It is categorized as RCB-0 (equivalent to a pCR), RCB-1, RCB-2, and RCB-3, reflecting increasingly larger residual cancer and respective poor prognoses (in terms of EFS) [80]. Finally, the CREATE-X trial did not examine capecitabine efficacy in patients with germline BRCA 1 or BRCA 2 pathogenic variants (less than 15% of those enrolled) [79].

    OlympiA is a phase III study designed to investigate how the PARP inhibitor olaparib might improve DFS and OS in patients with resected HR+ BC and TNBC with germline BRCA 1 or BRCA 2 mutation. It enrolled patients treated with CHT (containing anthracyclines, taxanes or the combination of both) in neoadjuvant or adjuvant setting and randomized them to receive olaparib (orally administered at the dose of 300 mg twice daily) vs. placebo for 1 year after surgical resection (and radiotherapy when indicated). Also in this trial, patients with TNBC who underwent NACT followed by surgery were required to have residual invasive BC in the breast and/or resected lymph nodes (non-pCR) [81]. Postneoadjuvant capecitabine was not foreseen in this trial. iDFS, the primary endpoint of the study, was significantly longer among patients assigned to receive olaparib than among those assigned placebo (HR = 0.58; P < 0.001). The percentage of patients alive and free of invasive disease at 3 years was 85.9% in the olaparib group and 77.1% in the placebo group. The benefit of adjuvant olaparib was observed irrespective of the germline BRCA mutation (BRCA 1 vs. BRCA 2), the hormone-receptor status, or the timing of previous CHT (neoadjuvant vs. adjuvant) [81] 4-year iDFS for the olaparib group was 82.7% (vs. 75.4% in placebo group) and 4-year distant DFS (DDFS) was 86.5% (vs. 79.1%). Adjuvant olaparib improves OS, with an HR of 0.68 and a P value of 0.009 at 3.5 years of median follow-up, meeting the significance threshold for OS at the second planned interim analysis. The OS benefit at 4 years in the olaparib arm compared with the placebo arm was reported (89.8% vs. 86.4%, respectively) [82].

    Both studies have defined the standard of adjuvant therapy post-NACT for patients with BRCA wild type (CREATE-X) and BRCA mutated (OlympiA) TNBC, that did not reach the pCR.

    The low percentage of BRCA mutated patients enrolled in the CREATE-X, the absence of pre-planned subgroup analyzes for this population do not allow for a description of the efficacy of capecitabine in this subgroup of patients.

    Moreover, there are no prospective randomized trials between capecitabine and olaparib to guide the clinical decision in this population, nor combination or sequence data between these two drugs.

    It would also be important to consider the potentially severe toxicity profile of such a combination, given their overlapping side effects (in particular, cytopenias).

    The treatment paradigm of early TNBC has had a real evolution since July 2021, with the introduction of immunotherapy following the Food and Drug Administration (FDA) approval of pembrolizumab for high-risk TNBC (tumor size > 1 cm but ≤ 2 cm in diameter with nodal involvement or tumor size > 2 cm in diameter regardless of nodal involvement), regardless of tumor PD-L1 expression, in combination with CHT as neoadjuvant treatment, and then continued as a single agent as adjuvant treatment after surgery for a total duration of approximately 1 year [83].

    Results from the KEYNOTE-522 study were the basis for this approval, demonstrating a significantly higher rate of pCR at the time of definitive surgery among patients who received pembrolizumab plus NACT than among those who received placebo plus NACT and an improvement in long-term benefits [73, 84]. The aim of the trial was not to identify the contributions of the neoadjuvant and adjuvant treatment phases, so it is difficult to define if these long-term results are related to exposure to adjuvant pembrolizumab or a lesser RCB at the end of the neoadjuvant phase in the pembrolizumab–CHT group.

    An exploratory analysis of the study then provided data to further describe the prognosis related to the RCB after neoadjuvant experimental treatment (Figure 1) [85].

    The unmet need for the optimal adjuvant treatment according to RCB [85]

    The HR for recurrence event in subgroups RBO-0, RCB-1, RCB-2, and RCB-3 are respectively 0.70 (rates: 5.2% vs. 7.3% in the pembrolizumab + CHT vs. placebo + CHT), 0.92 (rates: 17.4% vs. 20%), 0.52 (rates: 25.5% vs. 44.3%), 1.24 (72.5% vs. 69.2%).

    The rate of recurrence was numerically lower in all RCB groups with pembrolizumab + CHT, except in the small RCB-3 subset (that is represented by 5% and 7% of the population in the study, respectively in the experimental and control group). Pembrolizumab shifted RCB to lower categories in most patients (RCB-0: 63% vs. 56% of patients in the experimental vs. the control arm; RCB-1: 9% vs. 11%; RCB 18% vs. 20%).

    No patients in this trial received adjuvant capecitabine, and there are no randomized efficacy and safety data showing that multiagent therapy with pembrolizumab and capecitabine is superior to single-agent therapy in high-risk patients (stage II–III) who did not reach pCR.

    At the time, only results from phase II studies in metastatic TNBC demonstrated no new safety signals with this combination [86, 87].

    Pembrolizumab has also not been studied in combination with olaparib in the adjuvant setting, for the treatment of patients with BRCA mutations. No efficacy data are reported in the literature, even if some safety data are reported in the metastatic setting, in some early-phase studies that have evaluated the combination of PARPis and ICIs, not reporting unexpected toxicities [88, 89].

    Prospective trials would be needed to define what is the optimal adjuvant strategy according to RCB (single-agent CHT or poly-CHT), how the clinician should decide between olaparib, immunotherapy or capecitabine in the treatment of the population with BRCA mutations and whether these therapies can be administered in combination or sequence, with data in terms of efficacy and safety.

    Additional treatment strategies with new drugs are being studied as adjuvant treatment after NACT, with antibody-drug conjugates (ADCs) such as datopotamab deruxtecan (with or without durvalumab in TROPICS-Breast 03, ClinicalTrials.gov identifier: NCT05629585), and patritumab deruxtecan (HER3-DXd) which showed promising clinical response and biological changes in early TNBC [SOLTI TOT-HER3 window of opportunity trial part B, presented at European Society for Medical Oncology (ESMO) Breast 2023], or with ICIs (A-BRAVE trial, NCT02926196 and SWOG S1418/BR006 trial, NCT02954874).

    New biomarkers and frontiers in TNBC

    Recent progress in integrating ICIs and novel agents has revolutionized the therapeutic approach for early TNBC. Treatment strategies now emphasize escalating chemotherapeutic agents based on standard neoadjuvant regimens. An example is a phase II trial (ACTRN12617000651381) presented at the San Antonio Breast Cancer Symposium 2022 evaluating in high-risk TNBC, the addition of ipilimumab and nivolumab to neoadjuvant paclitaxel following a suboptimal response to anthracycline-based CHT (< 50% tumor reduction) and resulting in promising objective response rate (ORR) (43.7%) and pCR (18.8%) rates, regardless of PD-L1 status.

    However, it is also crucial to identify subgroups of patients with favorable prognoses, where NACT could potentially be de-escalated. Therefore, discovering novel biomarkers to categorize patients with good prognoses and safely de-escalate NACT is essential.

    TILs show promise as a biomarker for selecting patients who may have favorable outcomes with treatment de-escalation. In recent trials, higher TILs levels were associated with a higher pCR rate [71, 75, 90] and with a better response [75, 91]. Liquid biopsies, such as circulating tumor DNA (ctDNA), could serve as promising markers for identifying patients who might benefit from de-escalating or escalating neoadjuvant or adjuvant treatment. Rapid ctDNA clearance during NACT in early TNBC is linked to a high likelihood of achieving pCR [92]. Conversely, detecting ctDNA after completing NACT and surgery is associated with higher recurrence rates and poorer prognoses [93]. The use of dynamic biomarkers, such as ctDNA, to guide the choice of treatments in high-risk patients appears increasingly to be an important resource to be exploited in future studies.

    Furthermore, ADCs are emerging. Particularly, sacituzumab govitecan (SG) an ADC targeting Trop-2 was approved in metastatic TNBC patients who received ≥ 2 prior systemic therapies in the light of the results of the phase III ASCENT study. In this trial patients were randomized (1:1) to receive sacituzumab govitecan 10 mg/kg via intravenous infusion on day 1 and day 8 of a 21-day treatment cycle or a treatment of physician’s choice (TPC) achieving the primary endpoint (PFS 4.8 vs. 1.7 months) and also demonstrating an advantage in terms of OS (11.8 months vs. 6.9 months) [94]. Another single-arm phase II trial is evaluating SG and atezolizumab in combination as adjuvant treatment for patients with TNBC who have residual invasive disease after neoadjuvant therapy and detectable ctDNA (ClinicalTrials.gov identifier NCT04434040).

    Finally, it is essential to redefine, with new dedicated trials, the role of ER-low (1–9%) BC which, biologically and prognostically very similar to TNBC, could potentially benefit from the addition of immunotherapy to CHT and the role of HER-2 low [score 1+ or 2+ not amplified in fluorescence in situ hybridization (FISH)] BC in the light of recent results of efficacy of trastuzumab deruxtecan in advanced BC HER-2 low. Therefore, future studies are likely to expand the armamentarium at our disposal in this setting.

    New frontiers in early TNBC are summarized in Figure 2.

    New frontiers in early TNBC

    Interpretation and clinical implications

    TNBC has long been a challenging disease to treat due to its aggressive behavior and the lack of target therapies [95].

    Thanks to recent developments on TNBC, a series of therapeutic targets have been identified for the treatment of metastatic and early setting diseases. Especially for the radically operable disease, the chances of cure are increased with treatments aimed at reducing the odds of recurrence after tumor removal.

    Anthracycline and taxane-based poly-CHT remains the standard of treatment, most often administered preoperatively to assess tumor sensitivity. It aims to increase the rate of local control, making it useful to guide breast-conserving surgery and to ensure survival benefits by reaching the pCR.

    The introduction of immunotherapy in association with poly-CHT in the neoadjuvant setting has increased the rate of pCR, guaranteeing better results in terms of long-term benefits in the KEYNOTE-522, the pivotal trial that led to the approval in clinical practice of the use of the anti-PD1, pembrolizumab, in the early setting disease (neoadjuvant and adjuvant setting). These clinical findings were based on preclinical investigations that overturned the previous belief that BC was not an immunogenic disease [12].

    The actual need is to define the optimal adjuvant strategy after neoadjuvant chemo-immunotherapy, which must be affected by the patient’s risk of recurrence based on the histological prognostic and evidence after radical surgery, the individual’s tolerance of therapy-induced side effects (Figure 3).

    Current treatment algorithm for stage II–III TNBC

    In patients with low RCB and a low overall risk of recurrence, pembrolizumab alone should be continued. In patients with poor prognostic features of high RCB, this strategy may not be the best choice. Patients with high RCB, BRCA wild type, could benefit from capecitabine alone, although it would be reasonable to use a combination of capecitabine and pembrolizumab. Patients with high RCB, germline BRCA mutations, could benefit from olaparib (according to the inclusion criteria of the OlympiA trial), although it would be reasonable to use olaparib and pembrolizumab in combination or sequentially.

    However, none of these strategies, in monotherapy and/or in combination, have evidence from specific randomized trials after the neoadjuvant immunotherapy. There are no data on efficacy and safety in this setting. Currently, the best schedule is not known, and new data are awaited on new adjuvant strategies.

    Extensive efforts will also be required to investigate and expand access to immunotherapy to ER-low populations (ER 1–9%), not included in KEYNOTE-522. It represents a subgroup that does not formally meet the definition of TNBC, but shares biology, with nearly 90% of these tumors harboring a basal-like intrinsic subtype, and prognosis with TNBC and could share the same benefit from the addition of immunotherapy [96, 97].

    Furthermore, novel active agents are emerging for the treatment of TNBC and could provide an opportunity for a de-escalation of traditional CHT, the anti-trophoblast cell-surface antigen 2 (Trop2) sacituzumab govitecan that is currently being investigated in the early setting, including in combination with immunotherapy in the ASPRIA trial (ClinicalTrials.gov identifier NCT04434040).

    Conclusions

    This review highlights the multitude of advances in the treatment of early-stage TNBC and the important issues raised.

    The management of triple-negative breast cancer (TNBC) has seen notable advancements with the identification of therapeutic targets and successful integration of immunotherapy in neoadjuvant treatment. However, the current challenge lies in determining the optimal adjuvant strategy post-chemo-immunotherapy, tailoring decisions to individual patient characteristics and prognostic factors. The uncertainty surrounding the efficacy and safety of these strategies necessitates further randomized studies, while ongoing research explores novel approaches, such as the potential use of innovative agents like sacituzumab govitecan in the context of de-escalating traditional CHT. The imperative to extend access to immunotherapy to subgroups, such as those with low ER expression, holds crucial promise, paving the way for a more personalized and targeted future direction in TNBC treatment.

    In the next few years, it will be necessary to design new prospective clinical trials and wait for the results of those in progress, for a better knowledge of the efficacy of combination therapies, therapeutic sequences and new target drugs for the treatment of a disease which up to a few years ago was considered “untargetable”. This should be accompanied by a commitment to biomarker discovery, which could help the oncologist make the best decision for patient care.

    Abbreviations

    AC:

    doxorubicin and cyclophosphamide

    ADCs:

    antibody-drug conjugates

    BC:

    breast cancer

    BRCA:

    breast cancer susceptibility genes

    CHT:

    chemotherapy

    CI:

    confidence interval

    CREATE-X:

    Capecitabine for Residual Cancer as Adjuvant Therapy

    ctDNA:

    circulating tumor DNA

    DFS:

    disease-free survival

    EFS:

    event-free survival

    ER:

    estrogen receptor

    FDA:

    Food and Drug Administration

    HER2:

    human epidermal growth factor receptor 2

    HR:

    hazard ratio

    HR+:

    hormone receptor-positive

    ICI:

    immune checkpoint inhibitor

    iDFS:

    invasive disease-free survival

    IM:

    immunomodulatory

    LAR:

    luminal androgen receptor

    M:

    mesenchymal

    NACT:

    neoadjuvant chemotherapy

    OS:

    overall survival

    PARP:

    Poly(ADP-ribose) polymerase

    pCR:

    pathologic complete response

    PD-L1:

    programmed death ligand 1

    RCB:

    residual cancer burden

    TILs:

    tumor-infiltrating lymphocytes

    TMB:

    tumor mutational burden

    TME:

    tumor microenvironment

    TNBC:

    triple negative breast cancer

    Declarations

    Author contributions

    PDS: Conceptualization, Investigation, Writing—original draft, Writing—review & editing, Validation, Supervision. MP, CG, and GRO: Conceptualization, Investigation, Writing—original draft, Writing—review & editing. ANS, PF, and CL: Validation, Writing—review & editing, Supervision. DC: Investigation. All authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Sporikova Z, Koudelakova V, Trojanec R, Hajduch M. Genetic markers in triple-negative breast cancer. Clin Breast Cancer. 2018;18:e84150. [DOI] [PubMed]
    Howard FM, Olopade OI. Epidemiology of triple-negative breast cancer: a review. Cancer J. 2021;27:816. [DOI] [PubMed]
    Almansour NM. Triple-negative breast cancer: a brief review about epidemiology, risk factors, signaling pathways, treatment and role of artificial intelligence. Front Mol Biosci. 2022;9:836417. [DOI] [PubMed] [PMC]
    Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:34652. [DOI] [PubMed] [PMC]
    Azim HA, Ghosn M, Oualla K, Kassem L. Personalized treatment in metastatic triple-negative breast cancer: the outlook in 2020. Breast J. 2020;26:6980. [DOI] [PubMed]
    Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al.; ESMO Guidelines Committee. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2019;30:1194220. Erratum in: Ann Oncol. 2019;30:1674. Erratum in: Ann Oncol. 2021;32:284. [DOI] [PubMed]
    Bear HD, Anderson S, Brown A, Smith R, Mamounas EP, Fisher B, et al.; National Surgical Adjuvant Breast and Bowel Project Protocol B-27. The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: preliminary results from National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol. 2003;21:416574. [DOI] [PubMed]
    Golshan M, Loibl S, Wong SM, Houber JB, O’Shaughnessy J, Rugo HS, et al. Breast conservation after neoadjuvant chemotherapy for triple-negative breast cancer: surgical results from the BrighTNess randomized clinical trial. JAMA Surg. 2020;155:e195410. Erratum in: JAMA Surg. 2021;156:503. [DOI] [PubMed] [PMC]
    Turner N, Lambros MB, Horlings HM, Pearson A, Sharpe R, Natrajan R, et al. Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene. 2010;29:201323. [DOI] [PubMed] [PMC]
    Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer. 2020;6:54. [DOI] [PubMed] [PMC]
    Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, et al. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer. 2023;22:105. [DOI] [PubMed] [PMC]
    Liu Z, Li M, Jiang Z, Wang X. A comprehensive immunologic portrait of triple-negative breast cancer. Transl Oncol. 2018;11:31129. [DOI] [PubMed] [PMC]
    Farshbafnadi M, Pastaki Khoshbin A, Rezaei N. Immune checkpoint inhibitors for triple-negative breast cancer: from immunological mechanisms to clinical evidence. Int Immunopharmacol. 2021;98:107876. [DOI] [PubMed]
    Lehmann BD, Jovanović B, Chen X, Estrada MV, Johnson KN, Shyr Y, et al. Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS One. 2016;11:e0157368. [DOI] [PubMed] [PMC]
    Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SAW, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21:168898. [DOI] [PubMed] [PMC]
    Xiao Y, Ma D, Zhao S, Suo C, Shi J, Xue MZ, et al.; AME Breast Cancer Collaborative Group. Multi-omics profiling reveals distinct microenvironment characterization and suggests immune escape mechanisms of triple-negative breast cancer. Clin Cancer Res. 2019;25:500214. [DOI] [PubMed]
    Pelly VS, Moeini A, Roelofsen LM, Bonavita E, Bell CR, Hutton C, et al. Anti-inflammatory drugs remodel the tumor immune environment to enhance immune checkpoint blockade efficacy. Cancer Discov. 2021;11:260219. [DOI] [PubMed] [PMC]
    Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2014;2:36170. [DOI] [PubMed] [PMC]
    Wang C, Zhang J, Wang Y, Ouyang T, Li J, Wang T, et al. Prevalence of BRCA1 mutations and responses to neoadjuvant chemotherapy among BRCA1 carriers and non-carriers with triple-negative breast cancer. Ann Oncol. 2015;26:5238. [DOI] [PubMed]
    Hahnen E, Lederer B, Hauke J, Loibl S, Kröber S, Schneeweiss A, et al. Germline mutation status, pathological complete response, and disease-free survival in triple-negative breast cancer: secondary analysis of the GeparSixto randomized clinical trial. JAMA Oncol. 2017;3:137885. [DOI] [PubMed] [PMC]
    Pohl-Rescigno E, Hauke J, Loibl S, Möbus V, Denkert C, Fasching PA, et al. Association of germline variant status with therapy response in high-risk early-stage breast cancer: a secondary analysis of the GeparOcto randomized clinical trial. JAMA Oncol. 2020;6:7448. [DOI] [PubMed] [PMC]
    Vollebergh MA, Lips EH, Nederlof PM, Wessels LF, Wesseling J, Vd Vijver MJ, et al. Genomic patterns resembling BRCA1- and BRCA2-mutated breast cancers predict benefit of intensified carboplatin-based chemotherapy. Breast Cancer Res. 2014;16:R47. [DOI] [PubMed] [PMC]
    Belli C, Duso BA, Ferraro E, Curigliano G. Homologous recombination deficiency in triple negative breast cancer. Breast. 2019;45:1521. [DOI] [PubMed]
    van Verschuer VMT, Hooning MJ, van Baare-Georgieva RD, Hollestelle A, Timmermans AM, Koppert LB, et al. Tumor-associated inflammation as a potential prognostic tool in BRCA1/2-associated breast cancer. Hum Pathol. 2015;46:18290. [DOI] [PubMed]
    Nolan E, Savas P, Policheni AN, Darcy PK, Vaillant F, Mintoff CP, et al.; Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer (kConFab); Perou CM, Visvader JE, Gray DHD, Loi S, Lindeman GJ. Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer. Sci Transl Med. 2017;9:eaal4922. [DOI] [PubMed] [PMC]
    Parkes EE, Walker SM, Taggart LE, McCabe N, Knight LA, Wilkinson R, et al. Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. J Natl Cancer Inst. 2016;109:djw199. [DOI] [PubMed] [PMC]
    Castaneda CA, Mittendorf E, Casavilca S, Wu Y, Castillo M, Arboleda P, et al. Tumor infiltrating lymphocytes in triple negative breast cancer receiving neoadjuvant chemotherapy. World J Clin Oncol. 2016;7:38794. [DOI] [PubMed] [PMC]
    García-Teijido P, Cabal ML, Fernández IP, Pérez YF. Tumor-infiltrating lymphocytes in triple negative breast cancer: the future of immune targeting. Clin Med Insights Oncol. 2016;10:319. [DOI] [PubMed] [PMC]
    Gomez-Macias GS, Molinar-Flores G, Lopez-Garcia CA, Santuario-Facio S, Decanini-Arcaute H, Valero-Elizondo J, et al. Immunotyping of tumor-infiltrating lymphocytes in triple-negative breast cancer and genetic characterization. Oncol Lett. 2020;20:140. [DOI] [PubMed] [PMC]
    Sousa S, Brion R, Lintunen M, Kronqvist P, Sandholm J, Mönkkönen J, et al. Human breast cancer cells educate macrophages toward the M2 activation status. Breast Cancer Res. 2015;17:101. [DOI] [PubMed] [PMC]
    Medrek C, Pontén F, Jirström K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306. [DOI] [PubMed] [PMC]
    Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 2016;13:22841. [DOI] [PubMed]
    Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:16472. [DOI] [PubMed]
    Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, 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;19:4050. [DOI] [PubMed]
    Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. 2014;32:295966. [DOI] [PubMed] [PMC]
    Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol. 2013;31:8607. [DOI] [PubMed]
    Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, 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;25:154450. [DOI] [PubMed]
    Hida AI, Watanabe T, Sagara Y, Kashiwaba M, Sagara Y, Aogi K, et al. Diffuse distribution of tumor-infiltrating lymphocytes is a marker for better prognosis and chemotherapeutic effect in triple-negative breast cancer. Breast Cancer Res Treat. 2019;178:28394. [DOI] [PubMed]
    Ibrahim EM, Al-Foheidi ME, Al-Mansour MM, Kazkaz GA. The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: a meta-analysis. Breast Cancer Res Treat. 2014;148:46776. [DOI] [PubMed]
    Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al.; International TILs Working Group 2014. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 2015;26:25971. [DOI] [PubMed] [PMC]
    Karn T, Jiang T, Hatzis C, Sänger N, El-Balat A, Rody A, et al. Association between genomic metrics and immune infiltration in triple-negative breast cancer. JAMA Oncol. 2017;3:170711. [DOI] [PubMed] [PMC]
    Safonov A, Jiang T, Bianchini G, Győrffy B, Karn T, Hatzis C, et al. Immune gene expression is associated with genomic aberrations in breast cancer. Cancer Res. 2017;77:331724. [DOI] [PubMed]
    Lotfinejad P, Asghari Jafarabadi M, Abdoli Shadbad M, Kazemi T, Pashazadeh F, Sandoghchian Shotorbani S, et al. Prognostic role and clinical significance of tumor-infiltrating lymphocyte (TIL) and programmed death ligand 1 (PD-L1) expression in triple-negative breast cancer (TNBC): a systematic review and meta-analysis study. Diagnostics (Basel). 2020;10:704. [DOI] [PubMed] [PMC]
    Zhu X, Zhang Q, Wang D, Liu C, Han B, Yang JM. Expression of PD-L1 attenuates the positive impacts of high-level tumor-infiltrating lymphocytes on prognosis of triple-negative breast cancer. Cancer Biol Ther. 2019;20:110512. [DOI] [PubMed] [PMC]
    NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Breast Cancer. National Comprehensive Cancer Network® (NCCN®); c2020 [cited 2023 May 21]. Available from: https://www2.tri-kobe.org/nccn/guideline/breast/english/breast.pdf
    Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Anthracycline-containing and taxane-containing chemotherapy for early-stage operable breast cancer: a patient-level meta-analysis of 100 000 women from 86 randomised trials. Lancet. 2023;401:1277-1292. [DOI] [PubMed]
    Sparano JA, Zhao F, Martino S, Ligibel JA, Perez EA, Saphner T, et al. Long-term follow-up of the E1199 phase III trial evaluating the role of taxane and schedule in operable breast cancer. J Clin Oncol. 2015;33:235360. [DOI] [PubMed] [PMC]
    Jones SE, Savin MA, Holmes FA, O’Shaughnessy JA, Blum JL, Vukelja S, et al. Phase III trial comparing doxorubicin plus cyclophosphamide with docetaxel plus cyclophosphamide as adjuvant therapy for operable breast cancer. J Clin Oncol. 2006;24:53817. Erratum in: J Clin Oncol. 2007;25:1819. [DOI] [PubMed]
    Wheate NJ, Collins JG. Multi-nuclear platinum drugs: a new paradigm in chemotherapy. Curr Med Chem Anticancer Agents. 2005;5:26779. [DOI] [PubMed]
    Garutti M, Pelizzari G, Bartoletti M, Malfatti MC, Gerratana L, Tell G, et al. Platinum salts in patients with breast cancer: a focus on predictive factors. Int J Mol Sci. 2019;20:3390. [DOI] [PubMed] [PMC]
    Chalasani P, Livingston R. Differential chemotherapeutic sensitivity for breast tumors with “BRCAness”: a review. Oncologist. 2013;18:90916. [DOI] [PubMed] [PMC]
    Vetter M, Fokas S, Biskup E, Schmid T, Schwab F, Schoetzau A, et al. Efficacy of adjuvant chemotherapy with carboplatin for early triple negative breast cancer: a single center experience. Oncotarget. 2017;8:7561726. [DOI] [PubMed] [PMC]
    Su YW, Hung CY, Lam HB, Chang YC, Yang PS. A single institution experience of incorporation of cisplatin into adjuvant chemotherapy for patients with triple-negative breast cancer of unknown BRCA mutation status. Clin Med Insights Oncol. 2018;12:1179554918794672. [DOI] [PubMed] [PMC]
    Yu KD, Ye FG, He M, Fan L, Ma D, Mo M, et al. Effect of adjuvant paclitaxel and carboplatin on survival in women with triple-negative breast cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2020;6:13906. [DOI] [PubMed] [PMC]
    Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, et al. Secondary Somatic Mutations Restoring BRCA1/2 Predict Chemotherapy Resistance in Hereditary Ovarian Carcinomas. J Clin Oncol. 2011;29:300815. [DOI] [PubMed] [PMC]
    Guillemette S, Serra RW, Peng M, Hayes JA, Konstantinopoulos PA, Green MR, et al. Resistance to therapy in BRCA2 mutant cells due to loss of the nucleosome remodeling factor CHD4. Genes Dev. 2015;29:48994. [DOI] [PubMed] [PMC]
    Zhou J, Kang Y, Chen L, Wang H, Liu J, Zeng S, et al. The drug-resistance mechanisms of five platinum-based antitumor agents. Front Pharmacol. 2020;11:343. [DOI] [PubMed] [PMC]
    Rastogi P, Anderson SJ, Bear HD, Geyer CE, Kahlenberg MS, Robidoux A, et al. Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol. 2008;26:77885. Erratum in: J Clin Oncol. 2008;26:2793. [DOI] [PubMed]
    Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018;19:2739. [DOI] [PubMed] [PMC]
    Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Increasing the dose intensity of chemotherapy by more frequent administration or sequential scheduling: a patient-level meta-analysis of 37 298 women with early breast cancer in 26 randomised trials. Lancet. 2019;393:144052. [DOI] [PubMed] [PMC]
    von Minckwitz G, Schneeweiss A, Loibl S, Salat C, Denkert C, Rezai M, 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;15:74756. [DOI] [PubMed]
    Sikov WM, Berry DA, Perou CM, Singh B, Cirrincione CT, Tolaney SM, 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;33:1321. [DOI] [PubMed] [PMC]
    Loibl S, O’Shaughnessy J, Untch M, Sikov WM, Rugo HS, McKee MD, 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;19:497509. [DOI] [PubMed]
    Fasching PA, Link T, Hauke J, Seither F, Jackisch C, Klare P, et al.; German Breast Group and Arbeitsgemeinschaft Gynäkologische Onkologie Breast. Neoadjuvant paclitaxel/olaparib in comparison to paclitaxel/carboplatinum in patients with HER2-negative breast cancer and homologous recombination deficiency (GeparOLA study). Ann Oncol. 2021;32:4957. [DOI] [PubMed]
    Gupta S, Nair NS, Hawaldar R, Vanmali V, Parmar V, Gulia S, et al. Abstract GS5-01: Addition of platinum to sequential taxane-anthracycline neoadjuvant chemotherapy in patients with triple-negative breast cancer: a phase III randomized controlled trial. Cancer Res. 2023;83:GS5-01. [DOI]
    Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, et al.; KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:181728. [DOI] [PubMed]
    Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al.; IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:210821. [DOI] [PubMed]
    Miles D, Gligorov J, André F, Cameron D, Schneeweiss A, Barrios C, et al.; IMpassion131 investigators. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol. 2021;32:9941004. [DOI] [PubMed]
    Emens LA, Adams S, Barrios CH, Diéras V, Iwata H, Loi S, et al. First-line atezolizumab plus nab-paclitaxel for unresectable, locally advanced, or metastatic triple-negative breast cancer: IMpassion130 final overall survival analysis. Ann Oncol. 2021;32:98393. Erratum in: Ann Oncol. 2021;32:1650. [DOI] [PubMed]
    Cortes J, Rugo HS, Cescon DW, Im SA, Yusof MM, Gallardo C, et al.; KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022;387:21726. [DOI] [PubMed]
    Gianni L, Huang CS, Egle D, Bermejo B, Zamagni C, Thill M, et al. Pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple-negative, early high-risk and locally advanced breast cancer: NeoTRIP Michelangelo randomized study. Ann Oncol. 2022;33:53443. [DOI] [PubMed]
    Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg R, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020;396:1090100. [DOI] [PubMed]
    Schmid P, Cortes J, Dent R, Pusztai L, McArthur H, Kümmel S, et al.; KEYNOTE-522 Investigators. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386:55667. [DOI] [PubMed]
    Del Mastro L, Poggio F, Blondeaux E, De Placido S, Giuliano M, Forestieri V, et al.; Gruppo Italiano Mammella Investigators. Fluorouracil and dose-dense adjuvant chemotherapy in patients with early-stage breast cancer (GIM2): end-of-study results from a randomised, phase 3 trial. Lancet Oncol. 2022;23:157182. [DOI] [PubMed]
    Loibl S, Schneeweiss A, Huober J, Braun M, Rey J, Blohmer JU, et al.; GBG and AGO-B. Neoadjuvant durvalumab improves survival in early triple-negative breast cancer independent of pathological complete response. Ann Oncol. 2022;33:114958. [DOI] [PubMed]
    Kuroi K, Toi M, Ohno S, Nakamura S, Iwata H, Masuda N, et al. Prognostic significance of subtype and pathologic response in operable breast cancer; a pooled analysis of prospective neoadjuvant studies of JBCRG. Breast Cancer. 2015;22:48695. [DOI] [PubMed]
    Spring LM, Fell G, Arfe A, Sharma C, Greenup R, Reynolds KL, et al. Pathologic complete response after neoadjuvant chemotherapy and impact on breast cancer recurrence and survival: a comprehensive meta-analysis. Clin Cancer Res. 2020;26:283848. [DOI] [PubMed] [PMC]
    Coates AS, Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart-Gebhart MJ, et al.; Panel Members. Tailoring therapies—improving the management of early breast cancer: St Gallen international expert consensus on the primary therapy of early breast cancer 2015. Ann Oncol. 2015;26:153346. [DOI] [PubMed] [PMC]
    Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017;376:214759. [DOI] [PubMed]
    Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007;25:441422. [DOI] [PubMed]
    Tutt ANJ, Garber JE, Kaufman B, Viale G, Fumagalli D, Rastogi P, et al.; OlympiA Clinical Trial Steering Committee and Investigators. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med. 2021;384:2394405. [DOI] [PubMed] [PMC]
    Geyer CE Jr, Garber JE, Gelber RD, Yothers G, Taboada M, Ross L, et al.; OlympiA Clinical Trial Steering Committee and Investigators. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann Oncol. 2022;33:125068. [DOI] [PubMed] [PMC]
    FDA D. I.S.C.D. burst edition: FDA approval of Keytruda (pembrolizumab) for high-risk early-stage triple-negative breast cancer [Internet]. [cited 2023 May 21]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-keytruda-pembrolizumab-high-risk-early-stage-triple-negative
    Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al.; KEYNOTE-522 Investigators. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382:81021. [DOI] [PubMed]
    Pusztai L, Denkert C, O’Shaughnessy J, Cortes J, Dent RA, McArthur HL, et al. Event-free survival by residual cancer burden after neoadjuvant pembrolizumab + chemotherapy versus placebo + chemotherapy for early TNBC: exploratory analysis from KEYNOTE-522. J Clin Oncol. 2022;40:503. [DOI]
    Shah AN, Flaum L, Helenowski I, Santa-Maria CA, Jain S, Rademaker A, et al. Phase II study of pembrolizumab and capecitabine for triple negative and hormone receptor-positive, HER2–negative endocrine-refractory metastatic breast cancer. J Immunother Cancer. 2020;8:e000173. [DOI] [PubMed] [PMC]
    Page D, Pucilowska J, Bennetts L, Kim I, Sanchez K, Martel M, et al. Updated efficacy of first or second-line pembrolizumab plus in metastatic triple negative breast cancer and correlations with baseline lymphocyte and naïve CD4+ T-cell count. 2018 San Antonio Breast Cancer Symposium. San Antonio: Books, Presentations, Posters, Etc. 2018.
    Vinayak S, Tolaney SM, Schwartzberg L, Mita M, McCann G, Tan AR, et al. Open-label clinical trial of niraparib combined with pembrolizumab for treatment of advanced or metastatic triple-negative breast cancer. JAMA Oncol. 2019;5:113240. [DOI] [PubMed] [PMC]
    Domchek SM, Postel-Vinay S, Im SA, Park YH, Delord JP, Italiano A, et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): an open-label, multicentre, phase 1/2, basket study. Lancet Oncol. 2020;21:115564. [DOI] [PubMed]
    Nederlof I, De Bortoli D, Bareche Y, Hooijer GKJ, Sotiriou C, Van De Vijver MJ, et al. Relationship between tumor infiltrating lymphocytes (TILs) and response to pembrolizumab (pembro)+chemotherapy (CT) as neoadjuvant treatment (NAT) for triple-negative breast cancer (TNBC): phase Ib KEYNOTE-173 trial. Ann Oncol. 2019;30:III2. [DOI]
    Doroshow DB, Bhalla S, Beasley MB, Sholl LM, Kerr KM, Gnjatic S, et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2021;18:34562. [DOI] [PubMed]
    Magbanua MJM, Swigart LB, Wu HT, Hirst GL, Yau C, Wolf DM, et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol. 2021;32:22939. [DOI] [PubMed] [PMC]
    Radovich M, Jiang G, Hancock BA, Chitambar C, Nanda R, Falkson C, et al. Association of circulating tumor DNA and circulating tumor cells after neoadjuvant chemotherapy with disease recurrence in patients with triple-negative breast cancer: preplanned secondary analysis of the BRE12-158 randomized clinical trial. JAMA Oncol. 2020;6:14105. [DOI] [PubMed] [PMC]
    Bardia A, Hurvitz SA, Tolaney SM, Loirat D, Punie K, Oliveira M, et al.; ASCENT Clinical Trial Investigators. Sacituzumab govitecan in metastatic triple-negative breast cancer. N Engl J Med. 2021;384:152941. [DOI] [PubMed]
    Loibl S, Poortmans P, Morrow M, Denkert C, Curigliano G. Breast cancer. Lancet. 2021;397:175069. [DOI] [PubMed]
    Schrodi S, Braun M, Andrulat A, Harbeck N, Mahner S, Kiechle M, et al. Outcome of breast cancer patients with low hormone receptor positivity: analysis of a 15-year population-based cohort. Ann Oncol. 2021;32:141024. [DOI] [PubMed]
    Villegas SL, Nekljudova V, Pfarr N, Engel J, Untch M, Schrodi S, et al. Therapy response and prognosis of patients with early breast cancer with low positivity for hormone receptors – An analysis of 2765 patients from neoadjuvant clinical trials. Eur J Cancer. 2021;148:15970. [DOI] [PubMed]