Next Article in Journal
Does Molecular Profiling of KRAS-Mutant Non-Squamous Non-Small Cell Lung Cancer (NSCLC) Help in Treatment Strategy Planning?
Next Article in Special Issue
Tailoring the Evidence for Exercise Oncology within Breast Cancer Care
Previous Article in Journal
Current and New Novel Combination Treatments for Metastatic Triple-Negative Breast Cancer
Previous Article in Special Issue
Locoregional Management of Breast Cancer: A Chronological Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Triple-Negative Breast Cancer: A Review of Current Curative Intent Therapies

1
Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
2
Department of Oncology, University of Calgary, Calgary, AB T2N 4N2, Canada
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(7), 4768-4778; https://doi.org/10.3390/curroncol29070378
Submission received: 10 May 2022 / Revised: 27 June 2022 / Accepted: 28 June 2022 / Published: 7 July 2022
(This article belongs to the Special Issue Evolving Paradigm of Curative Intent Breast Cancer Management)

Abstract

:
Breast cancer is the most commonly diagnosed malignancy in women, with triple-negative breast cancer (TNBC) accounting for 10–20% of cases. Historically, fewer treatment options have existed for this subtype of breast cancer, with cytotoxic chemotherapy playing a predominant role. This article aims to review the current treatment paradigm for curative-intent TNBC, while also reviewing potential future developments in this landscape. In addition to chemotherapy, recent advances in the understanding of the molecular biology of TNBC have led to promising new studies of targeted and immune checkpoint inhibitor therapies in the curative-intent setting. The appropriate selection of TNBC patient subgroups with a higher likelihood of benefit from treatment is critical to identify the best treatment approach.

1. Introduction

In Canada, 28 900 new cases of breast cancer are projected in 2022, representing over 25% of all new cancer diagnoses in women [1]. Triple-negative breast cancer (TNBC) accounts for approximately 10–20% of all cases [2]. TNBC is associated with aggressive tumor phenotypes and a higher risk of recurrence (particularly within the first three to five years after diagnosis) despite higher initial response rates to curative-intent treatment [3]. In the metastatic setting, survival outcomes are much worse than hormone-receptor and/or HER-2-positive breast cancers [2]. Historically, treatment options for TNBC were limited to cytotoxic chemotherapy. However, TNBC represents a highly heterogenous group of tumors, and understanding the molecular biology of a specific tumor allows for personalized treatment (in turn, minimizing unnecessary treatment toxicity while improving cancer outcomes) [4]. This article reviews the current landscape of curative-intent TNBC treatment strategies, in addition to looking ahead toward future directions and possible therapeutic approaches. Treatment options are summarized for adjuvant (Table 1) and neoadjuvant (Table 2) TNBC, and drug classes and mechanisms of action are detailed in Table 3.

2. Cytotoxic Chemotherapy

TNBC shows initial sensitivity to cytotoxic chemotherapy, with higher response rates observed compared to other breast cancer subtypes [3]. Approximately 30–40% of TNBC cases will achieve a pathologic complete response (pCR) after treatment with a third-generation sequential anthracycline and taxane-based chemotherapy regimen [27], with pCR appearing to provide a valid surrogacy for recurrence-free and overall survival after treatment with chemotherapy in the TNBC subtype [28,29]. Superiority of the treatment with an anthracycline and taxane (over an anthracycline-sparing regimen) was established in the ABC group of trials (combining the USOR 06-090, NSABP B-46/USOR 07,132, and NSABP-B49 studies), which failed to demonstrate noninferiority of the anthracycline-sparing regimen [5]. Subgroup analysis of this study showed absolute four-year invasive disease-free survival benefits of 2.5% in node-negative TNBC patients, 10.9% in TNBC patients with N1 disease (1–3 lymph nodes involved), and 11.0% in TNBC patients with four or more lymph nodes involved. However, wide confidence intervals were noted in each of these subgroups, with the hazard ratio crossing 1.0.

2.1. Capecitabine

The value of pCR as a surrogate endpoint for survival in TNBC patients treated with chemotherapy led to studies assessing the impact of treatment escalation in patients where a pCR was not achieved. In the CREATE-X clinical trial, 910 patients with HER-2-negative breast cancer and without a pCR after neoadjuvant chemotherapy were randomized to standard care with or without capecitabine for between six and eight cycles of treatment [6]. While the overall study population benefited from capecitabine, the improvement in outcomes was exclusively driven by the TNBC subgroup, for both disease-free survival (69.8% versus 56.1%, HR 0.58, 95% CI 0.39–0.87) and overall survival (78.8% versus 70.3%, HR 0.52, 95% CI 0.30–0.90). Conversely, the GEICAM/2003-11_CIBOMA/2004-01 trial (which also compared capecitabine in the adjuvant setting to observation) failed to show an improvement in disease-free survival (79.6% versus 76.8%, HR 0.82, 95% CI 0.63–1.06) or overall survival (86.2% versus 85.9%, HR 0.92, 95% CI 0.66–1.28) [7]. However, this study also included patients with a pCR after neoadjuvant therapy (a lower-risk group), potentially accounting for the difference in efficacy. A systematic review and meta-analysis of capecitabine usage in the neoadjuvant and adjuvant settings showed significant improvements in disease-free survival (HR 0.75, 95% CI 0.65–0.86, p < 0.001) and overall survival (HR 0.63, 95% CI 0.53–0.77, p < 0.001) [30].
The SYSUCC-001 clinical trial also assessed the role of adjuvant capecitabine therapy, but at a lower dose (650 mg/m2 twice daily) given continuously for one year as maintenance therapy [8]. This trial randomized 424 patients with TNBC to capecitabine versus observation, showing an improvement in five-year disease-free survival (82.8% versus 73.0%, HR 0.64, 95% CI 0.42–0.95, p = 0.03), but without a statistically significant difference in five-year overall survival (85.5% versus 81.3%, HR 0.75, 95% CI 0.47–1.19, p = 0.22).

2.2. Platinum Agents

Platinum cytotoxic agents (such as carboplatin or cisplatin) cause DNA strand breaks via cross-linkage of DNA strands [31], increasing their effectiveness in tumors with impaired DNA repair pathways (a finding commonly seen in basal-like-1 TNBC [32,33], as well as in patients with hereditary mutations such as BRCA or other homologous recombination pathway genes [34]). Therefore, multiple studies have focused on the role of platinum-based chemotherapy for TNBC, with mixed results. In the GeparSixto clinical trial, the addition of carboplatin to neoadjuvant therapy resulted in a pCR in 53.2% of TNBC patients (84/158), compared to 36.9% (58/157) without carboplatin (p = 0.005) [13]. Similarly, in the CALGB 40,603 and BrighTNess studies, early-stage TNBC patients had higher pCR rates with the addition of carboplatin [14,15]. A post hoc analysis of BrighTNess also showed improvements in event-free survival rates with the addition of carboplatin to neoadjuvant chemotherapy (HR 0.57, p = 0.02), although no difference in overall survival outcomes was seen [16]. One meta-analysis of nine randomized controlled trials confirmed a significant increase in pCR rate with platinum-based neoadjuvant therapy (52.1% versus 37.0%, p < 0.001) [35].
In the adjuvant setting (particularly after neoadjuvant treatment with an anthracycline and taxane), the role of platinum agents is less clear. The EA1131 study [9] randomized patients with residual disease after neoadjuvant therapy to receive either capecitabine or platinum-based chemotherapy, and failed to demonstrate noninferiority of the platinum agent. The three-year event-free survival in both arms of this study was much worse than anticipated (42% in the platinum chemotherapy arm, versus 49% in the capecitabine arm). This is considered to relate to a higher proportion of basal-like tumors (78%) in the EA1131 study, or a higher burden of residual disease compared to the CREATE-X trial. The ongoing NRG BR-003 clinical trial (NCT02488967) [36] is randomizing higher-risk patients (with either node-positive disease or a tumor size of at least 3.0 cm) not treated in the neoadjuvant setting to receive standard adjuvant anthracycline/taxane combination chemotherapy with or without carboplatin. This study is estimated to have a primary completion date in November 2023 and will provide important insight into the role of platinum therapy in the adjuvant setting. A recent meta-analysis (looking at 2425 patients across eight clinical trials in the neoadjuvant and adjuvant settings) showed improvements in disease-free survival and overall survival with the addition of carboplatin to an anthracycline/taxane backbone, both with trial-level and individual patient-level analyses [37]. Overall, at this time, outside of clinical trials, the role of platinum chemotherapy in TNBC is primarily restricted to the neoadjuvant setting.

3. Targeted Therapies

PARP Inhibitors

Poly-ADP-ribose polymerases (PARP) are a group of diverse enzymes involved in DNA damage repair of single-stranded breaks, amongst other cellular functions [38]. Double-stranded DNA breaks are repaired through multiple mechanisms, including homologous recombination, but in the setting of an impairment in these pathways (such as that seen with mutations in the BRCA1 or BRCA2 genes, for example), PARP plays a more critical role. In patients with mutations in BRCA1/2 (and, to a lesser extent, other homologous recombination pathway genes as well), the inhibition of PARP leads to the inhibition of an effective DNA damage response, and subsequently cell death through a process known as synthetic lethality [39].
The role of PARP inhibitors in the neoadjuvant setting currently remains limited. The I-SPY-2 adaptive randomized phase II trial estimated an 88% predicted probability of success for the addition of veliparib plus carboplatin for neoadjuvant therapy in TNBC, with a pCR rate of 51% versus 26% in the control group (anthracycline/taxane chemotherapy) [17,18]. This led to the development of the phase III BrighTNess trial, which randomized patients to three arms—standard treatment (paclitaxel followed by doxorubicin/cyclophosphamide), standard treatment plus carboplatin, or standard treatment plus carboplatin and veliparib. As noted above, while the addition of carboplatin improved pCR [15] and event-free survival [16] rates compared to standard therapy, the veliparib arm did not provide any additional benefit, and increased toxicity. However, this study was not restricted to patients with BRCA mutations, potentially accounting for the negative result. A single-arm, nonrandomized phase II study of talazoparib (NEOTALA) assessed patients with germline BRCA1/2 mutations and locally advanced HER-2-negative breast cancer (tumor size at least 1.5 cm) [19]. Patients were treated with talazoparib 1 mg daily for 24 weeks prior to surgery, followed by adjuvant therapy based on investigator choice. The results of this study were presented at the 2021 American Society of Clinical Oncology Annual Meeting, with a reported pCR rate of 49.2% in the intention-to-treat population based on independent central review (a pCR rate comparable to those expected from anthracycline/taxane chemotherapy combinations, despite these patients not having received neoadjuvant cytotoxic therapy). Larger studies in the BRCA1/2-mutated population are needed to clarify the role of PARP inhibition and cytotoxic chemotherapy in neoadjuvant TNBC.
Conversely, in the adjuvant setting, PARP inhibition plays a critical role. The international phase III, double-blinded, multi-center OlympiA clinical trial [10] assessed the role of adjuvant olaparib therapy in patients with germline BRCA1/2 mutations and high-risk HER-2-negative breast cancer (defined as having lymph node involvement or tumor size greater than 2.0 cm if not treated with neoadjuvant therapy, or the presence of residual disease if treated with neoadjuvant therapy). Patients were assigned to one year of olaparib (300 mg twice daily) or placebo therapy, with treatment started within 12 weeks of completion of locoregional therapy. Although hormone-receptor-positive tumors were also allowed on this study, over 80% of the patients included had TNBC. The results of the OlympiA study demonstrated a significant improvement in invasive disease-free survival at three years with olaparib compared to placebo (85.9% versus 77.1%, HR 0.58, 99.5% CI 0.41–0.82, p < 0.001), as well as distant disease-free survival (87.5% versus 80.4%, HR 0.57, 99.5% CI 0.39–0.83, p < 0.001). While fewer deaths were reported in the olaparib group compared to placebo, the between-group difference did not cross the boundary for significance at the first interim analysis. However, an update presented at the European Society of Medical Oncology Virtual Plenary in March 2022 confirmed an improvement in overall survival, with a 3-year survival of 92.0% with olaparib compared to 89.1% with placebo (HR 0.68, 98.5% CI 0.47–0.97, p = 0.009) [11]. Of note, the OlympiA study did not allow TNBC patients with residual disease after neoadjuvant therapy to receive adjuvant capecitabine therapy. As a result, there is a lack of clarity as to the utility of capecitabine in these patients, and further studies are needed to address this.

4. Immunotherapy

The role of immunotherapy in treating curative-intent TNBC is rapidly evolving. Given the potential toxicities associated with checkpoint inhibitor therapy (some of which can be persistent, debilitating, or even fatal), risks and benefits must be carefully considered, particularly in the adjuvant setting [40].

4.1. Neoadjuvant Therapy

The only current US Federal Drug Administration (FDA) approval for immunotherapy in early-stage TNBC is for pembrolizumab [41], given in the neoadjuvant and adjuvant setting, based on the results of the KEYNOTE-522 clinical trial. In this trial, patients with stage II/III TNBC were randomized to receive either pembrolizumab or placebo for eight cycles in the neoadjuvant setting (given concurrently with paclitaxel and carboplatin, followed by anthracycline and cyclophosphamide chemotherapy) [20]. After surgery, pembrolizumab or placebo was continued every three weeks for up to nine additional cycles. This trial had two primary endpoints, including pathologic complete response rate (pCR) and event-free survival. At the first interim analysis (consisting of the first 602 patients randomized), an absolute improvement of 13.6% (p < 0.001) was seen in pCR rate with pembrolizumab-chemotherapy (64.8%) versus placebo-chemotherapy (51.2%). More recent data from 1174 randomized patients showed an absolute improvement in event-free survival (disease progression precluding surgery, local/distant recurrence, second primary cancer, or death from any cause) of 7.7% (HR 0.63, p < 0.001) with pembrolizumab-chemotherapy (84.5%) vs. placebo-chemotherapy (76.8%) [42].
Atezolizumab was studied in the neoadjuvant setting for patients with stage II/III TNBC in the Impassion031 study [21]. In this study, patients received nab-paclitaxel followed by dose-dense doxorubicin and cyclophosphamide, either with atezolizumab or placebo. This was followed by adjuvant atezolizumab to complete 12 months of treatment. The pCR rate with atezolizumab was 58%, compared to 41% in the placebo arm (in patients with PD-L1-positive tumors, the pCR rate was 69% for atezolizumab, compared to 49% for placebo).
In the phase II GeparNUEVO study [43], patients with TNBC with a tumor size of 2 cm or greater were randomized to receive durvalumab versus placebo as a single dose two weeks prior starting neoadjuvant chemotherapy (window-of-opportunity phase). This was followed by durvalumab versus placebo in combination with neoadjuvant chemotherapy (nab-paclitaxel for 12 weeks, followed by dose-dense epirubicin and cyclophosphamide for four cycles). In total, 174 patients were randomized, of which 36.4% had stage I tumors. However, after 117 patients were recruited onto the study, the Independent Data Monitoring Committee recommended eliminating the window-of-opportunity portion of the study, due to delays in receiving systemic chemotherapy. In the overall population, the pCR rate with durvalumab-chemotherapy was 53.4%, compared to 44.2% with placebo-chemotherapy (OR 1.45, p = 0.224). However, in patients treated in the window-of-opportunity phase, the pCR rate was 61.0% with durvalumab, compared to 41.4% with placebo. Additional data from GeparNUEVO presented at ASCO 2021 showed significant improvements in the 3-year invasive disease-free survival (84.9% vs. 76.9%, HR 0.54, p = 0.0559) and 3-year overall survival with durvalumab versus placebo (95.1% vs. 83.1%, HR 0.26, p = 0.076) [22]. The findings of this study raise several important questions regarding the role of immunotherapy in curative-intent TNBC, including regarding the necessity of adjuvant immunotherapy, as well as the utility of pCR as a surrogate endpoint for checkpoint inhibitors (given a modest improvement in pCR but a substantial improvement in overall survival). More information regarding the appropriateness of pCR as a surrogate endpoint in neoadjuvant immunotherapy TNBC trials may come from the NeoTRIP Michelangelo study [44]. This study is assessing carboplatin and nab-paclitaxel chemotherapy with or without atezolizumab in the neoadjuvant setting (all patients received anthracycline chemotherapy in the adjuvant setting). Preliminary analysis of this study showed no improvement in pCR rates with the addition of atezolizumab, but the primary outcome for this trial is event-free survival, and these results are still pending. Additionally, the sequencing of immunotherapy and chemotherapy requires further research, as the benefit of durvalumab in achieving pCR was seen primarily in patients treated in the window-of-opportunity phase.

4.2. Adjuvant Therapy

Data supporting the use of immune checkpoint inhibitors in the adjuvant setting are sparse, but multiple trials are currently underway. The SWOG S1418/BR-006 study is comparing one year of pembrolizumab vs. observation after completion of all other standard adjuvant therapies for TNBC with residual malignancy (either tumor size greater than 1 cm or positive lymph nodes) after neoadjuvant chemotherapy [12]. The Impassion-030 (ALEXANDRA) study is comparing adjuvant chemotherapy with or without atezolizumab in patients with stage I-II TNBC [45]. Overall, the use of adjuvant immunotherapy for TNBC should be restricted to the clinical trial setting at this time.

5. Future Directions

5.1. Circulating Tumor DNA (ctDNA)

Circulating tumor DNA is released following cancer cell death and presents an enticing target to assess disease response and activity [46]. Recent meta-analysis data showed that ctDNA detection after treatment for early breast cancer is associated with a significant reduction in disease-free survival (HR 8.32, 95% CI 3.01–22.99, p < 0.01) [47]. Additionally, another study in TNBC assessed 33 patients with residual disease after neoadjuvant therapy [48]. ctDNA was detected in four of these patients, all of whom recurred within nine months of surgery. However, an additional 10 patients ultimately had disease recurrence without detectable ctDNA, leading to a calculated specificity of 100% but a sensitivity of only 33% in this small population. A large phase III randomized trial (ZEST, NCT04915755) [49] is currently recruiting and will provide critical insights into the role of ctDNA. This trial is enrolling patients with TNBC or BRCA1/2 mutations after completion of standard neoadjuvant and/or adjuvant therapies. Patients are screened using a personalized ctDNA assay, and patients with detectable ctDNA are randomized to receive either niraparib (a PARP inhibitor) versus placebo. If positive, this trial would confirm not only the prognostic value of ctDNA assays, but also their predictive value in the personalization of treatment escalation—a truly practice-changing finding.

5.2. Other Targeted Pathways

Studies are ongoing to assess the role of other targeted treatments in TNBC, including in the curative-intent setting. The phase II randomized FAIRLANE study assessed neoadjuvant ipatasertib versus placebo in combination with paclitaxel chemotherapy for 12 weeks in patients with TNBC with a tumor size of at least 1.5 cm. In the overall population, there was no improvement in pCR rate with the addition of ipatasertib [23]. However, in the subgroup of tumors with mutations in the PIK3CA/AKT1/PTEN pathway, a pCR rate of 39% with ipatasertib was seen, compared to just 9% in the placebo arm. Another potential treatment strategy is the androgen receptor (AR), which is expressed in many TNBC patients, although the data supporting the use of treatments such as enzalutamide or abiraterone are extremely limited and primarily only in the metastatic setting [24]. Therefore, these treatments should not be used in the curative-intent setting outside of a clinical trial at this time.

5.3. Antibody–Drug Conjugates (ADCs)

While there are no currently approved antibody–drug conjugates in the curative-intent setting for TNBC, several molecules have demonstrated activity in the metastatic setting. These include sacituzumab govitecan (an ADC consisting of an antibody targeting the human trophoblast cell-surface antigen 2 (Trop-2) linked to SN-38, a potent topoisomerase inhibitor) [25], as well as trastuzumab deruxtecan (an ADC consisting of an antibody targeting the HER-2 receptor linked to another topoisomerase inhibitor, deruxtecan). Recent data showed an impressive efficacy of trastuzumab deruxtecan in previously treated metastatic TNBC with a low expression of HER-2 (defined as 1+ on immunohistochemistry (IHC) or 2+ on IHC with negative in situ hybridization testing) [26]. A phase III clinical trial of sacituzumab govitecan in patients with high relapse risk after standard neoadjuvant therapy (NCT04595565) is currently recruiting. At this time, ADCs should not be used for curative-intent TNBC treatment outside of a clinical trial.

6. Conclusions

In summary, patients with TNBC have more curative-intent treatment options than ever before. While cytotoxic chemotherapy continues to play a critical role in TNBC management, new targeted and immunotherapy options are creating seismic shifts in the approach to managing patients. Additional research is needed to better understand appropriate sequencing, and escalation and de-escalation of therapies.

Funding

This research received no external funding.

Conflicts of Interest

N.A.N. receives research support from AstraZeneca, Pfizer, Merck, Seagen and Novartis. She has received speaker honoraria from AstraZeneca, Pfizer, Merck, Seagen, Roche, Exact science, Eli Lily, Gilead and Novartis. O.F.K. has participated in advisory boards and received speaker honoraria from Pfizer and AstraZeneca.

References

  1. Brenner, D.R.; Poirier, A.; Woods, R.R.; Ellison, L.F.; Billette, J.M.; Demers, A.A.; Zhang, S.X.; Yao, C.; Finley, C.; Fitzgerald, N.; et al. Projected estimates of cancer in Canada in 2022. CMAJ 2022, 194, E601–E607. [Google Scholar] [CrossRef] [PubMed]
  2. Waks, A.G.; Winer, E.P. Breast cancer treatment: A review. JAMA 2019, 321, 288–300. [Google Scholar] [CrossRef] [PubMed]
  3. Carey, L.A.; Dees, E.C.; Sawyer, L.; Gatti, L.; Moore, D.T.; Collichio, F.; Ollila, D.W.; Sartor, C.I.; Graham, M.L.; Perou, C.M. The triple negative paradox: Primary tumor chemosensitivity of breast cancer subtypes. Clin. Cancer Res. 2007, 13, 2329–2334. [Google Scholar] [CrossRef] [Green Version]
  4. Abuhadra, N.; Stecklein, S.; Sharma, P.; Moulder, S. Early-stage triple-negative breast cancer: Time to optimize personalized strategies. Oncologist 2022, 27, 30–39. [Google Scholar] [CrossRef]
  5. Blum, J.L.; Flynn, P.J.; Yothers, G.; Asmar, L.; Geyer, C.E.; Jacobs, S.A.; Robert, N.J.; Hopkins, J.O.; O’Shaughnessy, J.A.; Dang, C.T.; et al. Anthracyclines in early breast cancer: The ABC trials—USOR 06-090, NSABP B-46-I/USOR 07132, and NSABP B-49 (NRG oncology). J. Clin. Oncol. 2017, 35, 2647–2655. [Google Scholar] [CrossRef] [PubMed]
  6. Masuda, N.; Lee, S.-J.; Ohtani, S.; Im, Y.-H.; Lee, E.-S.; Yokota, I.; Kuroi, K.; Im, S.-A.; Park, B.-W.; Kim, S.-B.; et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N. Engl. J. Med. 2017, 376, 2147–2159. [Google Scholar] [CrossRef]
  7. Lluch, A.; Barrios, C.H.; Torrecillas, L.; Ruiz-Borrego, M.; Bines, J.; Segalla, J.; Guerrero-Zotano, Á.; García-Sáenz, J.A.; Torres, R.; de la Haba, J.; et al. Phase III trial of adjuvant capecitabine after standard neo-/adjuvant chemotherapy in patients with early triple-negative breast cancer (GEICAM/2003-11_CIBOMA/2004-01). J. Clin. Oncol. 2020, 38, 203–213. [Google Scholar] [CrossRef]
  8. Wang, X.; Wang, S.S.; Huang, H.; Cai, L.; Zhao, L.; Peng, R.J.; Lin, Y.; Tang, J.; Zeng, J.; Zhang, L.H.; et al. Effect of capecitabine maintenance therapy using lower dosage and higher frequency vs observation on disease-free survival among patients with early-stage triple-negative breast cancer who had received standard treatment: The SYSUCC-001 randomized clinical trial. JAMA 2021, 325, 50–58. [Google Scholar] [CrossRef]
  9. Mayer, I.A.; Zhao, F.; Arteaga, C.L.; Symmans, W.F.; Park, B.H.; Burnette, B.L.; Tevaarwerk, A.J.; Garcia, S.F.; Smith, K.L.; Makower, D.F.; et al. Randomized phase III postoperative trial of platinum-based chemotherapy versus capecitabine in patients with residual triple-negative breast cancer following neoadjuvant chemotherapy: ECOG-ACRIN EA1131. J. Clin. Oncol. 2021, 39, 2539–2551. [Google Scholar] [CrossRef]
  10. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; de Azambuja, E.; Fielding, A.; Balmaña, J.; et al. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef]
  11. Tutt, A.N.J.; Garber, J.E.; Gelber, R.D. OlympiA Phase III: OS Results of Olaparib in gBRCAm Breast Cancer. Available online: https://oncologypro.esmo.org/meeting-resources/esmo-virtual-plenary-resources/olympia-phase-iii-pre-specified-event-driven-analysis-of-overall-survival-of-olaparib-in-gbrcam-breast-cancer (accessed on 9 May 2022).
  12. Pusztai, L.; Barlow, W.; Ganz, P.; Henry, N.; White, J.; Jagsi, R.; Mammen, J.; Lew, D.; Mejia, J.; Karantza, V.; et al. Abstract OT1-02-04: SWOG S1418/NRG -BR006: A randomized, phase III trial to evaluate the efficacy and safety of MK-3475 as adjuvant therapy for triple receptor-negative breast cancer with >1 cm residual invasive cancer or positive lymph nodes (>pN1mic) after neoadjuvant chemotherapy. Cancer Res. 2018, 78 (Suppl. 4), OT1-02-04. [Google Scholar] [CrossRef]
  13. 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]
  14. Shepherd, J.H.; Ballman, K.; Polley, M.Y.C.; Campbell, J.D.; Fan, C.; Selitsky, S.; Fernandez-Martinez, A.; Parker, J.S.; Hoadley, K.A.; Hu, Z.; et al. CALGB 40603 (alliance): Long-term outcomes and genomic correlates of response and survival after neoadjuvant chemotherapy with or without carboplatin and bevacizumab in triple-negative breast cancer. J. Clin. Oncol. 2022, 40, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Geyer, C.E.; Sikov, W.M.; Huober, J.; Rugo, H.S.; Wolmark, N.; O’Shaughnessy, J.; Maag, D.; Untch, M.; Golshan, M.; Lorenzo, J.P.; et al. Long-term efficacy and safety of addition of carboplatin with or without veliparib to standard neoadjuvant chemotherapy in triple-negative breast cancer: 4-year follow-up data from BrighTNess, a randomized phase III trial. Ann. Oncol. 2022, 33, 384–394. [Google Scholar] [CrossRef]
  17. Wang, H.; Yee, D. I-SPY 2: A neoadjuvant adaptive clinical trial designed to improve outcomes in high-risk breast cancer. Curr. Breast Cancer Rep. 2019, 11, 303–310. [Google Scholar] [CrossRef]
  18. 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]
  19. Litton, J.K.; Beck, J.T.; Jones, J.M.; Andersen, J.; Blum, J.L.; Mina, L.A.; Brig, R.; Danso, M.A.; Yuan, Y.; Abbattista, A.; et al. Neoadjuvant talazoparib in patients with germline BRCA1/2 (gBRCA1/2) mutation-positive, early HER2-negative breast cancer (BC): Results of a phase 2 study. J. Clin. Oncol. 2021, 39 (Suppl. 15), 505. [Google Scholar] [CrossRef]
  20. 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]
  21. Mittendorf, E.A.; Zhang, H.; Barrios, C.H.; Saji, S.; Jung, K.H.; Hegg, R.; Koehler, A.; Sohn, J.; Iwata, H.; Telli, M.L.; 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, 1090–1100. [Google Scholar] [CrossRef]
  22. Loibl, S.; Schneeweiss, A.; Huober, J.B.; Braun, M.; Rey, J.; Blohmer, J.U.; Furlanetto, J.; Zahm, D.M.; Hanusch, C.; Thomalla, J.; et al. Durvalumab improves long-term outcome in TNBC: Results from the phase II randomized GeparNUEVO study investigating neodjuvant durvalumab in addition to an anthracycline/taxane based neoadjuvant chemotherapy in early triple-negative breast cancer (TNBC). J. Clin. Oncol. 2021, 39 (Suppl. 15), 506. [Google Scholar] [CrossRef]
  23. 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] [PubMed]
  24. Ge, J.; Zuo, W.; Chen, Y.; Shao, Z.; Yu, K. The advance of adjuvant treatment for triple-negative breast cancer. Cancer Biol. Med. 2022, 19, 187. [Google Scholar] [CrossRef] [PubMed]
  25. Bardia, A.; Mayer, I.A.; Vahdat, L.T.; Tolaney, S.M.; Isakoff, S.J.; Diamond, J.R.; O’Shaughnessy, J.; Moroose, R.L.; Santin, A.D.; Abramson, V.G.; et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N. Engl. J. Med. 2019, 380, 741–751. [Google Scholar] [CrossRef]
  26. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N. Engl. J. Med. 2022. Online publication ahead of print. [Google Scholar] [CrossRef]
  27. 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]
  28. 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]
  29. 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]
  30. Huo, X.; Li, J.; Zhao, F.; Ren, D.; Ahmad, R.; Yuan, X.; Du, F.; Zhao, J. The role of capecitabine-based neoadjuvant and adjuvant chemotherapy in early-stage triple-negative breast cancer: A systematic review and meta-analysis. BMC Cancer 2021, 21, 78. [Google Scholar] [CrossRef]
  31. Kelland, L. The resurgence of platinum-based cancer chemotherapy. Nat. Rev. Cancer 2007, 7, 573–584. [Google Scholar] [CrossRef]
  32. Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef] [PubMed]
  33. 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. [Google Scholar] [CrossRef]
  34. Ali, R.M.M.; McIntosh, S.A.; Savage, K.I. Homologous recombination deficiency in breast cancer: Implications for risk, cancer development, and therapy. Genes Chromosomes Cancer 2021, 60, 358–372. [Google Scholar] [CrossRef]
  35. 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] [PubMed]
  36. Doxorubicin Hydrochloride and Cyclophosphamide Followed by Paclitaxel with or without Carboplatin in Treating Patients with Triple-Negative Breast Cancer—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02488967 (accessed on 9 May 2022).
  37. Pathak, N.; Sharma, A.; Elavarasi, A.; Sankar, J.; Deo, S.V.S.; Sharma, D.N.; Mathur, S.; Kumar, S.; Prasad, C.P.; Kumar, A.; et al. Moment of truth-adding carboplatin to neoadjuvant/adjuvant chemotherapy in triple negative breast cancer improves overall survival: An individual participant data and trial-level meta-analysis. Breast 2022, 64, 7–18. [Google Scholar] [CrossRef] [PubMed]
  38. Thompson, L.H.; Schild, D. Homologous recombinational repair of DNA ensures mammalian chromosome stability. Mutat Res. 2001, 477, 131–153. [Google Scholar] [CrossRef]
  39. Nijman, S.M. Synthetic lethality: General principles, utility and detection using genetic screens in human cells. FEBS Lett. 2011, 585, 1–6. [Google Scholar] [CrossRef] [Green Version]
  40. Emens, L.A.; Adams, S.; Cimino-Mathews, A.; Disis, M.L.; Gatti-Mays, M.E.; Ho, A.Y.; Kalinsky, K.; McArthur, H.L.; Mittendorf, E.A.; Nanda, R.; et al. Society for immunotherapy of cancer (SITC) clinical practice guideline on immunotherapy for the treatment of breast cancer. J. Immunother. Cancer 2021, 9, 2597. [Google Scholar] [CrossRef]
  41. US Food and Drug Administration (FDA). Highlights of Prescribing Information—Pembrolizumab. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125514s110lbl.pdf (accessed on 9 May 2022).
  42. Schmid, P.; Cortes, J.; Dent, R.; Pusztai, L.; McArthur, H.; Kümmel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; et al. Event-free survival with pembrolizumab in early triple-negative breast cancer. N. Engl. J. Med. 2022, 386, 556–567. [Google Scholar] [CrossRef]
  43. 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]
  44. Gianni, L.; Huang, C.S.; Egle, D.; Bermejo, B.; Zamagni, C.; Thill, M.; Anton, A.; Zambelli, S.; Bianchini, G.; Russo, S.; 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, 534–543. [Google Scholar] [CrossRef] [PubMed]
  45. McArthur, H.L.; Ignatiadis, M.; Guillaume, S.; Bailey, A.; Martinez, J.L.; Brandao, M.; Metzger, O.; Lai, C.; Fumagalli, D.; Daly, F.; et al. ALEXANDRA/IMpassion030: A phase III study of standard adjuvant chemotherapy with or without atezolizumab in early-stage triple-negative breast cancer. J. Clin. Oncol. 2019, 37 (Suppl. 15), TPS598. [Google Scholar] [CrossRef]
  46. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef] [PubMed]
  47. Cullinane, C.; Fleming, C.; O’Leary, D.P.; Hassan, F.; Kelly, L.; O’Sullivan, M.J.; Corrigan, M.A.; Redmond, H.P. Association of circulating tumor DNA with disease-free survival in breast cancer: A systematic review and meta-analysis. JAMA Netw. Open 2020, 3, e2026921. [Google Scholar] [CrossRef]
  48. Chen, Y.-H.; Hancock, B.A.; Solzak, J.P.; Brinza, D.; Scafe, C.; Miller, K.D.; Radovich, M. Next-generation sequencing of circulating tumor DNA to predict recurrence in triple-negative breast cancer patients with residual disease after neoadjuvant chemotherapy. NPJ Breast Cancer 2017, 3, 24. [Google Scholar] [CrossRef]
  49. Efficacy and Safety Comparison of Niraparib to Placebo in Participants with Human Epidermal Growth Factor 2 Negative (HER2-) Breast Cancer Susceptibility Gene Mutation (BRCAmut) or Triple-Negative Breast Cancer (TNBC) with Molecular Disease—Full Text View—ClinicalTrials.gov. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04915755 (accessed on 9 May 2022).
Table 1. Adjuvant Triple Negative Breast Cancer (TNBC) curative-intent treatment.
Table 1. Adjuvant Triple Negative Breast Cancer (TNBC) curative-intent treatment.
Trial
(NCT Number)
PhaseStageTreatmentPatients in AnalysisOutcomesRefs
ChemotherapyABC Trials
(NCT00493870, NCT00887536, NCT01547741)
IIIEarly
II– III
Docetaxel and cyclophosphamide versus doxorubicin and cyclophosphamide1288DFS: HR 1.42, 95% CI 1.04–1.94
OS: not significant
(favoring AC)
[5]
CREATE-X
(NCT00130533)
IIIEarly
I–IIIb
Patients without pCR, standard of care with or without capecitabine286DFS: HR 0.58, 95% CI 0.39–0.87
OS: HR 0.52, 95% CI 0.30–0.90
[6]
GEICAM/2003-11_CIBOMA/2004-01
(NCT00130533)
IIIEarly
I–III
Capecitabine versus observation869DFS: HR 0.82, 95% CI 0.63–1.0
OS: HR 0.92, 95% CI 0.66–1.28
[7]
SYSUCC-001
(NCT01112826)
IIIEarly
I–III
Low-dose capecitabine vs. observation424DFS: HR 0.64, 95% CI 0.42–0.95
OS: HR 0.75, 95% CI 0.47–1.19
[8]
EA1131
(NCT02445391)
IIIEarly
II–III
Neoadjuvant chemotherapy followed by adjuvant capecitabine versus platinum308DFS: 49.4%, 95% CI 39.0 to 59.0 versus 42.0%, 95% CI 30.5–53.1
DFS: HR 1 (Ref) versus 1.06, 95% CI 0.62–1.81
[9]
Targeted therapyOlympiaA
(NCT02032823)
IIIEarly
II–III
Olaparib versus placebo in patients with BRCA1/2 mutations1509DFS, distant: HR 0.57, 99.5% CI 0.39–0.83
OS: HR 0.68, 98.5% CI 0.47–0.97
[10,11]
ImmunotherapySWOG S1418/BR-006
(NCT02954874)
IIIEarly
II–III
Patients without pCR received pembrolizumab versus observationRecruitment on goingNot yet reported[12]
NCT, national clinical trial; DFS, disease-free survival; OS, overall survival; HR, hazard ratio; CI confidence interval; pCR, pathological complete response; BRCA, BReast CAncer gene.
Table 2. Neoadjuvant Triple Negative Breast Cancer TNBC curative-intent treatment.
Table 2. Neoadjuvant Triple Negative Breast Cancer TNBC curative-intent treatment.
Trial
(NCT Number)
PhaseStageTreatmentPatients in AnalysisOutcomesRefs
ChemotherapyGeparSixto
(NCT01426880)
IIEarly
II–III
Neoadjuvant chemotherapy with versus without carboplatin315Achieving a pCR: OR 1.94, 95% CI 1.24–3.04[13]
CALGB 40603
(NCT00861705)
IIEarly
II–III
Weekly paclitaxel with versus without carboplatin, followed by AC113The addition of carboplatin increased pCR from 41% to 61%[14]
BrighTNess
(NCT02032277)
IIIEarly
II–III
Paclitaxel with versus without carboplatin318EFS: HR 0.57, 95% CI 0.36–0.91[15,16]
Targeted therapyI-SPY-2
(NCT02032277)
IIEarly
II–III
Paclitaxel with versus without veliparib–carboplatin116Rate of pCR: 51%, 95% CI 36–66 versus 26%, 95% CI 9–43[17,18]
BrighTNess
(NCT02032277)
IIIEarly
II–III
Paclitaxel and carboplatin with versus without veliparib476Rate of pCR:
53% versus 58% p = 0.36
[15,16]
NEOTALA
(NCT03499353)
IIEarly
I–III
Single-agent talazoparib in patients with BRCA1/2 mutations61Rate of pCR:
49.2%, 95% CI 34.0–64.5
[19]
ImmunotherapyKEYNOTE-522
(NCT03036488)
IIIEarly
II–III
Paclitaxel and carboplatin with versus without pembrolizumab, followed by AC1174 -EFS
602 -pCR
EFS: HR 0.63, 95% CI 0.48–0.82
Rate of pCR: 64.8%, increase of 13.6%, 95% CI 5.4–21.8
[20]
IMpassion031
(NCT03498716)
IIIEarly
II–III
nab-paclitaxel followed by ddAC with versus without atezolizumab333Rate of pCR: 58%, increase of 17%, 95% CI 6–27[21]
GeparNUEVO
(NCT02685059)
IIEarly
II–III
nab-paclitaxel followed by ddAC with versus without durvalumab174Rate of pCR: 53.4%, increase of 9.2%, NS
Achieving a pCR OR 1.45, 95% CI 0.80–2.63
DFS: HR 0.54
OS: HR 0.26
[22]
NCT, national clinical trial; DFS, disease-free survival; OS, overall survival; EFS, event-free survival; NS, not significant; HR, hazard ratio; OR, odds ratio; CI confidence interval; pCR, pathological complete response; BRCA, BReast CAncer gene; AC, anthracycline + cyclophosphamide; ddAC, dose dense anthracycline + cyclophosphamide.
Table 3. Details of agents used in Triple Negative Breast Cancer (TNBC).
Table 3. Details of agents used in Triple Negative Breast Cancer (TNBC).
Agent TypeAgent ClassAgentMechanismRelevant TrialsRef
ChemotherapyAnti-metabolitesCapecitabineProdrug that is converted to 5-FU, and subsequent metabolites inhibit formation of thymidylate, necessary for DNA synthesisCREATE-X[6]
Alkylating agentsPlatinum
(carboplatin)
Reactive platinum complexes inhibit DNA synthesis by forming interstrand and intrastrand cross-linking of DNA moleculesGeparSixto[13]
CALGB 40603[14]
BrighTNess[15,16]
CyclophosphamideProdrug that is metabolized to its active form phosphoramide mustard that forms cross-links between strands of DNAABC Trials[5]
CALGB 40603[14]
GeparNUEVO[22]
Anti-microtubuleTaxanes
(paclitaxel, docetaxel)
Prevents effective microtubules by binding and promoting stabilization and growth ABC Trials
(See Table 1 and Table 2)
[5]
Cytotoxic antibioticsDoxorubicinIntercalates with DNA leading to topoisomerase II inhibition and subsequent apoptosisABC Trials
(See Table 1 and Table 2)
[5]
Targeted therapyPARPiVeliparib
Talazoparib
Olaparib
Inhibition of PARP leads to ineffective repair of DNA SSBs, leading to DSBs, and apoptosisBrighTNess[15,16]
I-SPY-II[17]
NEOTALA[19]
OlympiaA[10]
AKTiIpatasertibInhibition of AKT slows down upregulated cell division pathwaysFAIRLANE[23]
ImmunotherapyPD-L1 inhibitorAtezolizumab
Durvalumab
MAB checkpoint inhibitor blocks PD-L1 interrupting the interaction with PD-1 on T-cells, enhancing antitumor immune response and leading to increased T-cell activation against tumorsIMPassion031[21]
GeparNUEVO[22]
PD-1 inhibitorPembrolizumabMAB checkpoint inhibitor that blocks PD-1 as opposed to PD-L1, enhancing antitumor immune response and leading to increased T-cell activation against tumorsKEYNOTE522[20]
Androgen DeprivationAndrogen receptor signaling inhibitor (ARSI)EnzalutamidePrevents the androgen receptor from translocating through the cell, preventing DNA transcriptionMDV3100-11[24]
Pregnenolone AnalogueAbirateroneSuppresses CPY17A1-mediated androgen synthesis and direct AR-inhibitory propertiesUCBG 12-1[24]
Antibody–Drug Conjugates Sacituzumab govitecan-hziyAntibody targeting Trop-2 linked to SN-38 (topoisomerase inhibitor), with chemotherapy released after cell internalization of the antibody.NCT01631552[25]
Trastuzumab deruxtecanAntibody targeting HER-2 linked to a topoisomerase inhibitor (deruxtecan). Used in patients with low expression of HER-2.DESTINY-Breast04[26]
5-FU, 5-fluorouracil; PARP, Poly-adenosine diphosphate-ribose polymerase; SSB, single-strand break; DSB, double-strand break; AKT, also called Protein kinase B; PD-L1, Programmed death-ligand 1; PD-1, Programmed death 1; MAB, monoclonal antibody; AR, androgen receptor; Trop-2, Human trophoblast cell-surface antigen 2; HER-2, Human epidermal growth factor receptor 2.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

MacDonald, I.; Nixon, N.A.; Khan, O.F. Triple-Negative Breast Cancer: A Review of Current Curative Intent Therapies. Curr. Oncol. 2022, 29, 4768-4778. https://doi.org/10.3390/curroncol29070378

AMA Style

MacDonald I, Nixon NA, Khan OF. Triple-Negative Breast Cancer: A Review of Current Curative Intent Therapies. Current Oncology. 2022; 29(7):4768-4778. https://doi.org/10.3390/curroncol29070378

Chicago/Turabian Style

MacDonald, Isaiah, Nancy A. Nixon, and Omar F. Khan. 2022. "Triple-Negative Breast Cancer: A Review of Current Curative Intent Therapies" Current Oncology 29, no. 7: 4768-4778. https://doi.org/10.3390/curroncol29070378

Article Metrics

Back to TopTop