Next Article in Journal
High Neuroticism Is Related to More Overall Functional Problems and Lower Function Scores in Men Who Had Surgery for Non-Relapsing Prostate Cancer
Previous Article in Journal
Changes in Inflammatory Markers Predict the Prognosis of Resected Hepatocellular Carcinoma with Child–Pugh A
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Treatment Strategies for Residual Disease following Neoadjuvant Chemotherapy in Patients with Early-Stage Breast Cancer

1
Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan
2
School of Medicine, University of Jordan, Amman 11941, Jordan
3
Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut 1107, Lebanon
4
School of Medicine, University of Balamand, Koura 3843, Lebanon
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(8), 5810-5822; https://doi.org/10.3390/curroncol29080458
Submission received: 12 July 2022 / Revised: 8 August 2022 / Accepted: 9 August 2022 / Published: 16 August 2022
(This article belongs to the Section Breast Cancer)

Abstract

:
Breast cancer continues to be the most diagnosed cancer among women worldwide. Neoadjuvant chemotherapy is the standard of care for breast cancer patients with locally advanced disease and patients with poor pathological features, such as triple-negative (TN) or human epidermal growth factor receptor-2 (HER2)-positive subtypes. Neoadjuvant therapy offers several advantages, including better surgical outcomes, early systemic treatment for micro-metastases, and accurate tumor biology and chemosensitivity assessment. Multiple studies have shown that achieving pathological complete response (pCR) following neoadjuvant chemotherapy is associated with better prognosis and better treatment outcomes; almost half of such patients may fail to achieve pCR. Tumor proliferative index, hormone receptor (HR) status, and HER2 expression are the major predictors of pCR. Strategies to improve pCR have been dependent on augmenting neoadjuvant chemotherapy with the addition of taxanes and dual anti-HER2 targeted therapy in patients with HER2-positive tumor, and more recently, immunotherapy for patients with TN disease. The clinical management of patients with residual disease following neoadjuvant chemotherapy varies and depends mostly on the level of HR expression and HER2 status. Recent data have suggested that switching trastuzumab to trastuzumab-emtansine (T-DM1) in patients with HER2-positive disease and the addition of capecitabine for patients with HER2-negative and HR-negative subtype is associated with a better outcome; both strategies are incorporated into current clinical practice guidelines. This paper reviews available and ongoing studies addressing strategies to better manage patients who continue to have residual disease following neoadjuvant chemotherapy.

1. Introduction

Breast cancer is the most commonly diagnosed cancer among women worldwide and is considered a leading cause of cancer-related mortality [1,2]. Neoadjuvant therapy for breast cancer has long been considered to offer patients an early treatment for their tumors, especially for those who present with locally-advanced inoperable cancers, providing the benefit from downstaging the tumor and regional lymph nodes and improving the surgical outcomes by increasing the rate of breast-conserving surgery (BCS) [3]. Neoadjuvant therapy can help delay surgery for those awaiting genetic testing results or the resolution of an intercurrent illness and pregnancy. Additionally, neoadjuvant therapy allows the accurate assessment of tumor biology and chemosensitivity. It offers early systemic treatment for micro-metastases. It gives us the opportunity to modify adjuvant treatment according to pathology results, especially for high-risk patients, such as those with TN or HER2-positive disease [4]. Neoadjuvant therapy can also benefit patients with limited axillary nodal involvement (N1): such patients may avoid axillary lymph node dissection if converted to node-negative disease with neoadjuvant therapy. In such cases, sentinel lymph node sampling can be an acceptable alternative to full axillary dissection with its associated morbidity.
Despite the introduction of many targeted drugs and the utilization of immunotherapy, a relatively high percentage of patients fail to achieve pCR. Following neoadjuvant chemotherapy, patients with residual disease are at higher risk for local and systemic relapse [5,6]. While patients with HR-positive tumors receive adjuvant hormonal therapy, with or without radiation therapy, patients with TN disease may not receive any post-surgical systemic treatment.
This paper reviews available and ongoing studies addressing strategies to better manage patients who continue to have residual disease following neoadjuvant chemotherapy. We first describe pCR, which is the standard clinical measure of treatment outcome for breast cancer. This is followed by a review of the management of residual HER2-positive disease, the management of residual HER2-negative disease, immunotherapy, and PARP inhibitors in BRCA-positive patients.

2. Pathological Complete Response (pCR)

The definition of pCR varies across many clinical trials, but the most widely accepted definition includes an absence of residual invasive disease in the breast and axillary nodes (ypT0/is, ypN0). The most stringent definition, however, is the one that mandates no residual invasive or non-invasive (in situ) disease in both the breast and axillary lymph nodes (ypT0, ypN0) [7,8,9]. However, the presence of non-invasive carcinoma seems to have no negative impact on the long-term outcome of breast cancer patients [5].
Many studies have confirmed the association between pCR and event-free survival (EFS) and overall survival (OS), especially in patients with HR-negative, TN, or HER2-positive disease. Even from early neoadjuvant trials, such an association was very apparent [10]. Although the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-18 trial failed to demonstrate relapse-free survival (RFS) or OS advantages of neoadjuvant chemotherapy compared to the adjuvant one, it did show that patients who achieved pCR had better disease-free survival (DFS) (hazard ratio, 0.47; p = 0.0001) and OS (hazard ratio, 0.32; p = 0.0001) compared to those with residual disease [10]. Given the high correlation of pCR and clinical outcomes, both the European Medicines Agency (EMA) and the U.S. Food and Drug Administration (FDA) endorsed the use of pCR as an endpoint for the assessment of neoadjuvant treatment for HER2-positive and TN disease breast cancer patients [11,12].
The Collaborative Trials in Neoadjuvant Breast Cancer (CTNeoBC) working group collected data from 12 international trials with almost 12,000 patients enrolled. Eradication of tumor from both the axillary lymph nodes and the breast (ypT0, ypN0 or ypT0/is, ypN0) was associated with better EFS and OS. This association was strongest among patients with HER2-positive/HR-negative tumors treated with trastuzumab and among those with TN disease [5]. Another meta-analysis that included 36 studies with 5768 patients with HER2-positive disease concluded that patients who achieved pCR, compared to those with residual disease, had better EFS (hazard ratio, 0.37; 95% probability interval (PI), 0.32–0.43). The association was more noticeable in patients with HR-negative disease (hazard ratio, 0.29; 95% PI, 0.24–0.36) than in those with HR-positive disease (hazard ratio, 0.52; 95% PI, 0.40–0.66) [13]. A more recent and more extensive meta-analysis of data involving 52 studies that enrolled 27,895 patients with breast cancer confirmed that the higher the pCR following neoadjuvant therapy, the lower the risk of disease recurrence (hazard ratio, 0.31; 95% CI, 0.24–0.39) and death (hazard ratio, 0.22; 95% CI, 0.15–0.30) [14].

3. Management of Residual HER2-Positive Disease

3.1. Current Neoadjuvant Standards

Almost 20% of all breast cancers express HER2 [15,16]. This subtype of tumors is characterized by the amplification of the HER2 gene that encodes for a type of tyrosine kinase receptor among other types of genes within the same family: HER1 (sometimes referred to as EGFR), HER2, HER3, and HER4. Activation of these receptors results in the activation of downstream intracellular signaling pathways, resulting in continued cellular proliferation and survival. Targeting this oncogenic driver will offer a therapeutic advantage to tumors harboring it [17,18,19].
HER2-positive tumors tend to have more aggressive behavior and usually have worse outcomes than other subtypes [20]. Such worse outcomes were relatively offset since the introduction of anti-HER2 therapy, which improved the prognosis of breast cancer patients in both advanced and early disease settings [21].
In addition to adjuvant chemotherapy, trastuzumab for 1 year is the current standard of care for HER2-positive early-stage breast cancer patients. The benefit of adding trastuzumab to chemotherapy was proven in many large randomized trials: HERA [22,23,24,25], NSABP-B31, NCCTG N9831 [26,27,28], BCIRG 006 [29], and FNCLCC-PACS 04 trials [30]. Data from these trials, except the FNCLCC-PACS 04 [30], showed that trastuzumab when added to chemotherapy was associated with significantly better DFS and OS compared to chemotherapy alone, a benefit which was maintained with longer follow-up [25,29].
In an attempt to further improve the DFS of this special subtype of breast cancer patients, investigators studied an extended adjuvant treatment with neratinib, a pan-HER2 inhibitor, for one year after completing a full year of trastuzumab in the ExteNET trial. Patients (n = 2840) with HER2-positive early breast cancer were randomized to receive either neratinib or placebo. Patients in the neratinib group fared better in terms of 5-year DFS: 90.2% compared to 87.7% in the placebo group (hazard ratio, 0.73; 95% CI, 0.57–0.92; p = 0.008) [31]. The study was updated recently, and neratinib was associated with better OS at 8 years (hazard ratio, 0.79; 95% CI, 0.55–1.13). Additionally, central nervous system (CNS) events were fewer with neratinib [32].
The APHINITY trial evaluated the benefit of adding another anti-HER2 drug (pertuzumab) to a combination of both trastuzumab and chemotherapy in the adjuvant settings [33]. The results of this trial were not outstanding as was expected, since the addition of pertuzumab conferred only a small benefit in terms of 3-year invasive disease-free survival (iDFS): 94.1% versus 93.2% (hazard ratio, 0.81; 95% CI, 0.66–1.00; p = 0.045). Patients in the high-risk category (with node-positive disease) had the greatest benefit: a 3-year iDFS of 92.0% versus 90.2% (hazard ratio, 0.77; 95% CI, 0.62–0.96; p = 0.02) [33].
Taxane-based regimens, with or without anthracycline, plus dual HER2 blockade with pertuzumab and trastuzumab, remain the current standard of care for neoadjuvant treatment for patients with HER2-positive disease [34,35]. Over the years, many trials have involved different chemotherapy and anti-HER2 combinations with varying rates of pCR as high as 70% [36,37], as shown in Table 1.
The finding of residual disease after neoadjuvant treatment indicates chemotherapy-resistant tissues in the tumor [7,42]. Chemotherapy modification during neoadjuvant therapy, based on the failure of clinical response according to the physical examination, with or without imaging studies, showed no improvement in pCR, as was demonstrated in several studies, including the GeparTrio trial [43,44,45].
Over the years, multiple strategies have been tried to improve pCR rates and recurrence-free and long-term survivals. Such strategies involved the addition of new drugs in the neoadjuvant settings, extending the duration of neoadjuvant therapy, dose-intensification, and trying to use concurrent chemoradiation regimens; all failed to show significant survival differences [46,47,48,49,50,51].

3.2. Post Neoadjuvant Treatment

Another approach, termed the post-neoadjuvant treatment, offers additional adjuvant treatment for patients who do not achieve pCR after neoadjuvant therapy to overcome tumor resistance. The concept of post-neoadjuvant therapy has been studied with frequent failures or statistically insignificant results [52,53,54,55,56].
Chemotherapies have a narrow therapeutic index because they are tumor non-selective, causing multiple systemic toxicities. Antibody-drug conjugates (ADCs) are drugs that link a tumor-specific antibody to a cytotoxic drug. ADCs are designed to limit collateral damage to normal cells by specifically delivering cytotoxic agents to tumor cells, thus improving their therapeutic index.
Trastuzumab-emtansine (T-DM1) is an example of these antibody-drug conjugates, consisting of a monoclonal antibody against HER2 combined with a thioether linker and the antimicrotubule chemotherapy maytansine [57]. Its activity was first demonstrated in the metastatic settings in EMILIA and TH3RESA studies [58,59,60,61]. To study the activity of T-DM1 in the post-neoadjuvant settings, a phase III trial (the KATHERINE trial) involved 1486 HER2-positive patients with residual disease in the breast or axilla after neoadjuvant treatment (taxane-based chemotherapy, with or without anthracyclines) plus trastuzumab (with or without pertuzumab) who received either 14 cycles of T-DM1 or 14 cycles of trastuzumab; the primary endpoint was iDFS. Patients included were those with T1-T4 tumors, N0-N3 lymph nodes, and M0 (excluding T1aN0 or T1bN0) at presentation. The residual invasive disease was detected pathologically in the breast or axilla at the time of surgery after neoadjuvant treatment. Hormone receptors were positive in 72.3% of patients, and anthracyclines were given to 76.9%. The majority (80%) of patients received trastuzumab alone, while 18% were treated with dual anti-HER2: both trastuzumab and pertuzumab. Patients with positive margins after breast-conserving surgery, those with gross residual disease after mastectomy, and those who had progressed while taking neoadjuvant therapy were excluded. Additionally, patients with cardiopulmonary compromise, including patients with class II or higher, New York Heart Association (NYHA) heart failure, and patients with a history of deteriorating ejection fraction (EF) to less than 40% with previous therapies were excluded. After a median follow-up of 41.4 months, the 3-year iDFS rates were 88.3% in the T-DM1 group compared to 77% in the trastuzumab group (hazard ratio, 0.50; 95% CI, 0.39–0.64; p < 0.001), which represented a 50% reduction in the risk of recurrence of invasive disease or death. The rate of distant recurrences was 10.5% with T-DM1 versus 15.9% with trastuzumab, and all subgroups benefited from T-DM1 [62].
Criticism to the KATHERINE trial was the reliance on the pretreatment core biopsy to determine the HER2 status. There was an apparent loss of HER2 positivity in a subgroup of patients. The KATHERINE trial could not answer whether adjuvant T-DM1 was specifically beneficial in this subgroup. The most common grade 3 or more adverse events observed in this trial were thrombocytopenia and hypertension in the T-DM1 arm and hypertension and radiation-induced skin injury with trastuzumab. Despite the fact that more patients experienced thrombocytopenia in the T-DM1 arm, the percentages of patients with grade 3 or more hemorrhages were similar in both arms (0.4% in the T-DM1 group and 0.3% in the trastuzumab group). Based on this data, T-DM1 is widely accepted as the standard anti-HER2 treatment in patients who fail to achieve pCR after neoadjuvant anti-HER2 therapy.

3.3. Ongoing Trials Investigating Other Anti-HER2 Agents in the Post Neoadjuvant Settings

At least two additional clinical trials are ongoing to address residual disease following neoadjuvant therapy in HER2-positive breast cancer patients utilizing tumor vaccines. The first is recruiting patients with residual disease following taxanes and trastuzumab-based neoadjuvant therapy to receive nelipepimut-S, a vaccine derived from the E75 peptide. This antigen is expressed in the extracellular domain of HER2, coupled with granulocyte-macrophage colony-stimulating factor (GM-CSF). Nelipepimut-S will be administered for 2 years in the adjuvant settings with a primary endpoint of DFS [63]. Another randomized phase II trial investigates two dendritic cell vaccines (DC1 and WOKVAC) in HER2-positive patients who fail to achieve pCR after a trastuzumab-containing neoadjuvant therapy. The rationale is that the vaccines may induce an anti-tumor immune response. Patients are randomized to DC1 or WOKVAC, administered for 1 year, with the primary endpoint being DFS [64].

4. Management of Residual HER2-Negative Disease

4.1. Current Neoadjuvant Standards

The majority of breast cancers do not express HER2. This group can be divided into two subtypes. The first is HR-negative, commonly described as TN, representing 10–15% of all breast cancers. Several trials have attempted to improve pCR in this group of patients but with modest success [65,66]. The classical anthracycline, cyclophosphamide, and taxanes-based regimen resulted in pCR of 30–44%. The addition of carboplatin to this regimen improved pCR to around 55% [67]. Anthracycline-sparing regimens were also tried in patients with TN disease; carboplatin in combination with a taxane (without anthracycline) in regimens given for 12 to 18 weeks resulted in pCR rates ranging from 45% to 55% with a favorable toxicity profile, as shown in Table 2 [68,69].
The other subtype, the most common, is HR-positive disease, which is divided into two categories, luminal A and luminal B (HER2-negative). The difference between these breast cancer subtypes is the level of Ki67 expression, which indicates cell proliferation. These differences allow the classification of luminal A as having a low proliferation rate due to its low expression of Ki67 and luminal B (HER2-negative) as having a higher proliferation rate due to its higher level of Ki67 expression [78,79,80]. Ki67 can be considered an important biomarker in differentiating between subtypes and can serve as a prognostic marker for patients who fail to achieve pCR. In a study that enrolled 435 patients with stage IIB-IIIA disease treated with an anthracycline and taxanes with residual disease following neoadjuvant therapy, changes in Ki67 expression in the original core biopsy and the surgical specimen were evaluated in relation to DFS and OS. Forty-five percent of patients had luminal B-like tumors, 25% had luminal A-like, 14% had TN, 11% had triple-positive, and 5% had HER2-positive disease. Fifty-seven percent of participants had a decline in Ki67 percentage, and this decrease was directly correlated with a better DFS and OS, specifically in luminal B subtypes. However, no difference was observed in DFS or OS patients with luminal A, TN, HER2-positive, and luminal B/HER2-positive subtypes [81].

4.2. Residual Disease in HER2-Negative Breast Cancer

Despite the ultimate goal of achieving pCR in breast cancer, a high percentage of patients are left with residual disease post-treatment and surgery. The CREATE-X trial was designed to treat residual diseases in HER2-negative patients who did not achieve pCR. Breast cancer patients with residual disease following neoadjuvant chemotherapy were divided into two groups: one received the standard adjuvant care, and the other received standard adjuvant care plus capecitabine. The study included both hormone receptor-positive and negative patients and used DFS and OS as its endpoints. The study was terminated early, and final analysis showed that patients treated with capecitabine had a better 5-year DFS than the control group (74.1% versus 67.6%; hazard ratio for recurrence, second cancer, or death, 0.70; 95% CI, 0.53–0.92; p = 0.01). Overall survival was also longer in the capecitabine group (89.2%) compared to 83.6% in the control group (hazard ratio for death, 0.59; 95% CI, 0.39–0.90; p = 0.01). The difference was more significant in patients with TN disease: 5-year DFS was 69.8% in the capecitabine group compared to 56.1% in the control arm (hazard ratio for recurrence, second cancer, or death, 0.58; 95% CI, 0.39–0.87). The OS rate was also better: 78.8% compared to 70.3% (hazard ratio for death, 0.52; 95% CI, 0.30–0.90). Hand–foot syndrome was the most frequent adverse event, occurring in 73.4%, including 11.1% with a grade 3 event among the capecitabine group. Diarrhea, leukopenia, thrombocytopenia, neutropenia, and anemia were also encountered [56].
Additional clinical trials are being conducted to study the role of capecitabine as an adjuvant therapy following neoadjuvant chemotherapy in patients with TN breast cancer. The GEICAM/CIBOMA is a phase III trial that studied the value of adding capecitabine in patients with TN disease regardless of their neoadjuvant chemotherapy outcomes. There were 876 patients: 55.9% were lymph node-negative and 73.9% had a basal phenotype. After a median follow-up of 7.3 years (range: 0.0 to 11.1), DFS was not significantly prolonged with capecitabine compared to the control arm (hazard ratio, 0.82; 95% CI, 0.63 to 1.06; p = 0.136). Five-year DFS was 79.6% (95% CI, 75.8% to 83.4%) with capecitabine and 76.8% (95% CI, 72.7% to 80.9%) with observation. No statistical difference was observed in OS, either (hazard ratio, 0.92; 95% CI, 0.66 to 1.28; p = 0.623) [82,83].

5. Immunotherapy

5.1. Immunotherapy in Neoadjuvant Settings

Compared to other solid tumors or hematological malignancies, immunotherapy in breast cancer was delayed. Recently, promising data have started to appear, mostly in patients with the TN subtype [84]. The KEYNOTE-522 was a phase III randomized trial that assessed the use of the programmed cell death protein-1 (PD-1) inhibitor, pembrolizumab, and chemotherapy in the neoadjuvant therapy of patients with TN disease. Patients (n = 602) were randomized to receive chemotherapy (paclitaxel and carboplatin, then cyclophosphamide and anthracycline) plus pembrolizumab or chemotherapy plus placebo. The combination of pembrolizumab and chemotherapy resulted in a significant increase in pCR rates (64.8%; 95% CI, 59.9–69.5) compared to chemotherapy alone (51.2%; 95% CI, 44.1–58.3); the estimated treatment difference was 13.6% (95% CI, 5.4–21.8; p < 0.001). After a median follow-up of 15.5 months, 7.4% of patients in the pembrolizumab-chemotherapy group and 11.8% in the placebo-chemotherapy group had an event (hazard ratio, 0.63; 95% CI, 0.43–0.93) [77]. The study was also updated during the last annual meeting of the American Society of Clinical Oncology (ASCO). Compared with placebo, and after a median follow-up of 39.1 months, pembrolizumab added to neoadjuvant chemotherapy was associated with lower residual cancer burden (RCB) in patients with TN disease and with prolonged EFS even in patients who failed to achieve pCR [85].
The addition of atezolizumab to a 24-week carboplatin/nab-paclitaxel chemotherapy backbone was also tried in a phase III trial (NeoTRIP) in patients with locally advanced TN disease. The pCR rates were not significantly different between the two study arms (40.8% with chemotherapy compared to 43.5% with atezolizumab) [86]. Atezolizumab was also tried in the Impassion-031 trial, which randomized 333 previously untreated stage II-III patients with TN disease to atezolizumab combined with neoadjuvant chemotherapy (nab-paclitaxel, doxorubicin, and cyclophosphamide) versus chemotherapy alone. In the PD-L1-positive patients, pCR was reported in 69% in the atezolizumab plus chemotherapy group versus 49% in patients in the placebo plus chemotherapy group (rate difference, 20%; 95% CI, 4–35; p = 0.021) [87]. Regardless of PD-L1 expression, atezolizumab increased pCR rates from 41.1% in the chemotherapy-alone group to 57.6% in the combination group (rate difference, 17%; 95% CI, 6–27; p = 0.0044).

5.2. Immunotherapy for Residual Disease

Given the encouraging results of the upfront use of immunotherapy in the neoadjuvant setting of early-stage breast cancer, it was natural to try these agents in patients with residual disease following neoadjuvant therapy. This concept is being tested in the SWOG 1418, a randomized, open-label, phase III trial that will compare the iDFS of over 1100 patients with TNBC who fail to achieve pCR, with residual invasive breast cancer ≥ 1 cm, and/or positive lymph nodes after neoadjuvant therapy. Patients are randomized to receive one year of pembrolizumab as adjuvant therapy compared to none in the entire study population. The study will stratify patients by nodal stage (ypNo vs. ypN+), residual tumor (≥2 cm vs. <2 cm), PD-L1 status (positive vs. negative), and prior adjuvant chemotherapy [88].

6. PARP Inhibitors in BRCA-Positive Patients

Olaparib, a Poly ADP-ribose polymerase (PARP) inhibitor, was also tested in a subgroup of patients with BRCA1 or BRCA2 germline pathogenic or likely pathogenic variants who had residual disease following neoadjuvant chemotherapy or in high-risk patients following adjuvant therapy. The OlympiA is a phase III, double-blind, randomized trial that involved 1836 patients with high-risk, HER2-negative early breast cancer who had received local treatment plus adjuvant or neoadjuvant chemotherapy. Patients were randomized to receive oral olaparib or placebo for one full year. At a median follow-up of 2.5 years (event-driven analysis), the 3-year iDFS was 85.9% in the olaparib group and 77.1% in the placebo group (95% CI, 4.5–13.0; hazard ratio for invasive disease or death: 0.58; 99.5% CI, 0.41–0.82; p < 0.001). Similarly, the 3-year DFS was significantly higher in the olaparib group: 87.5% compared to 80.4% in the placebo group (95% CI, 3.0 to 11.1; hazard ratio for distant disease or death, 0.57; 99.5% CI, 0.39 to 0.83; p < 0.001) [89]. This study gives patients with BRCA1 or BRCA2 another option to improve treatment outcomes but adds a little confusion in choosing between the many newly introduced options in this setting.

7. Conclusions and Future Directions

While significant progress has been made in the neoadjuvant and adjuvant therapy of early-stage breast cancer [90,91,92,93], several questions remain unanswered: What is the impact of adjuvant immunotherapy, specifically for those who fail to achieve pCR? How would capecitabine then be incorporated? What is the role of other targeted agents such as PARP inhibitors? Would the other recently introduced antibody-drug conjugates, such as trastuzumab deruxtecan, be better than T-DM1 in patients with HER2-positive disease [94]? Can we better select patients for more appropriate therapy based on biomarkers? These questions, and many others, will hopefully be answered by many of the ongoing and planned clinical trials. Figure 1 summarizes the current knowledge related to the management of breast cancer post-neoadjuvant therapy.

Author Contributions

Conceptualization, H.A.-R.; methodology, H.A.-R.; validation, H.A.-R., H.K. and H.I.A.; resources, H.A.-R. and H.I.A.; data curation, H.A.-R., H.K., H.I.A. and T.B.D.; writing—original draft preparation, H.A.-R., H.K., H.I.A. and T.B.D.; writing—review and editing, H.A.-R., H.K., H.I.A. and T.B.D.; visualization, H.A.-R., H.K., H.I.A. and T.B.D.; supervision, H.A.-R.; project administration, H.A.-R.; funding acquisition, not applicable. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank Alice Haddadin, Rayan Bater, and Ameen Harb for their help in preparing this manuscript.

Conflicts of Interest

The authors declare that this review paper was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Lei, S.; Zheng, R.; Zhang, S.; Wang, S.; Chen, R.; Sun, K.; Zeng, H.; Zhou, J.; Wei, W. Global Patterns of Breast Cancer Incidence and Mortality: A Population-Based Cancer Registry Data Analysis from 2000 to 2020. Cancer Commun. 2021, 41, 1183–1194. [Google Scholar] [CrossRef] [PubMed]
  3. Mieog, S.; van der Hage, J.; van de Velde, C. Preoperative Chemotherapy for Women with Operable Breast Cancer. Cochrane Database Syst Rev. 2007, 18, CD005002. [Google Scholar] [CrossRef]
  4. Caparica, R.; Lambertini, M.; Pondé, N.; Fumagalli, D.; de Azambuja, E.; Piccart, M. Post-Neoadjuvant Treatment and the Management of Residual Disease in Breast Cancer: State of the Art and Perspectives. Ther. Adv. Med. Oncol. 2019, 11, 175883591982771. [Google Scholar] [CrossRef]
  5. 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]
  6. Kuroi, K.; Toi, M.; Ohno, S.; Nakamura, S.; Iwata, H.; Masuda, N.; Sato, N.; Tsuda, H.; Kurosumi, M.; Akiyama, F. Prognostic Significance of Subtype and Pathologic Response in Operable Breast Cancer; a Pooled Analysis of Prospective Neoadjuvant Studies of JBCRG. Breast Cancer 2013, 22, 486–495. [Google Scholar] [CrossRef] [PubMed]
  7. 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]
  8. Gianni, L.; Pienkowski, T.; Im, Y.-H.; Tseng, L.-M.; Liu, M.-C.; Lluch, A.; Starosławska, E.; la Haba-Rodriguez, J.D.; Im, S.-A.; Pedrini, J.L.; et al. 5-Year Analysis of Neoadjuvant Pertuzumab and Trastuzumab in Patients with Locally Advanced, Inflammatory, or Early-Stage HER2-Positive Breast Cancer (NeoSphere): A Multicentre, Open-Label, Phase 2 Randomised Trial. Lancet Oncol. 2016, 17, 791–800. [Google Scholar] [CrossRef]
  9. Mauri, D.; Pavlidis, N.; Ioannidis, J.P.A. Neoadjuvant Versus Adjuvant Systemic Treatment in Breast Cancer: A Meta-Analysis. JNCI J. Natl. Cancer Inst. 2005, 97, 188–194. [Google Scholar] [CrossRef]
  10. Rastogi, P.; Anderson, S.J.; Bear, H.D.; Geyer, C.E.; Kahlenberg, M.S.; Robidoux, A.; Margolese, R.G.; Hoehn, J.L.; Vogel, V.G.; Dakhil, S.R.; et al. Preoperative Chemotherapy: Updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27. J. Clin. Oncol. 2008, 26, 778–785. [Google Scholar] [CrossRef]
  11. Pathological Complete Response in Neoadjuvant Treatment of High-Risk Early-Stage Breast Cancer: Use as an Endpoint to Support Accelerated Approval (2020). Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/pathological-complete-response-neoadjuvant-treatment-high-risk-early-stage-breast-cancer-use (accessed on 1 May 2022).
  12. Agency, E.M. The Role of the Pathological Complete Response as an Endpoint in Neoadjuvant Breast Cancer Studies. 2014. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/draft-guidelinerole-pathological-complete-response-endpoint-neoadjuvant-breast-cancer-studies_en.pdf (accessed on 1 May 2022).
  13. Broglio, K.R.; Quintana, M.; Foster, M.; Olinger, M.; McGlothlin, A.; Berry, S.M.; Boileau, J.-F.; Brezden-Masley, C.; Chia, S.; Dent, S.; et al. Association of Pathologic Complete Response to Neoadjuvant Therapy in HER2 Positive Breast Cancer with Long Term Outcomes. JAMA Oncol. 2016, 2, 751. [Google Scholar] [CrossRef] [PubMed]
  14. Spring, L.M.; Fell, G.; Arfe, A.; Sharma, C.; Greenup, R.; Reynolds, K.L.; Smith, B.L.; Alexander, B.; Moy, B.; Isakoff, S.J.; 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, 2838–2848. [Google Scholar] [CrossRef] [PubMed]
  15. Slamon, D.J.; Godolphin, W.; Jones, L.A.; Holt, J.A.; Wong, S.G.; Keith, D.E.; Levin, W.J.; Stuart, S.G.; Udove, J.; Ullrich, A.; et al. Studies of the HER-2/neu Proto-Oncogene in Human Breast and Ovarian Cancer. Science 1989, 244, 707–712. [Google Scholar] [CrossRef] [PubMed]
  16. Wolff, A.C.; Hammond, M.E.H.; Hicks, D.G.; Dowsett, M.; McShane, L.M.; Allison, K.H.; Allred, D.C.; Bartlett, J.M.S.; Bilous, M.; Fitzgibbons, P.; et al. Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. J. Clin. Oncol. 2013, 31, 3997–4013. [Google Scholar] [CrossRef] [PubMed]
  17. Moasser, M.M. The Oncogene HER2: Its Signaling and Transforming Functions and Its Role in Human Cancer Pathogenesis. Oncogene 2007, 26, 6469–6487. [Google Scholar] [CrossRef] [PubMed]
  18. Rimawi, M.F.; Schiff, R.; Osborne, C.K. Targeting HER2 for the Treatment of Breast Cancer. Annu. Rev. Med. 2015, 66, 111–128. [Google Scholar] [CrossRef]
  19. Gutierrez, C.; Schiff, R. HER2: Biology, Detection, and Clinical Implications. Arch. Pathol. Lab. Med. 2011, 135, 55–62. [Google Scholar] [CrossRef]
  20. Lambertini, M.; Pondé, N.F.; Solinas, C.; de Azambuja, E. Adjuvant Trastuzumab: A 10-Year Overview of Its Benefit. Expert Rev. Anticancer. Ther. 2016, 17, 61–74. [Google Scholar] [CrossRef]
  21. Brandão, M.; Pondé, N.F.; Poggio, F.; Kotecki, N.; Salis, M.; Lambertini, M.; de Azambuja, E. Combination Therapies for the Treatment of HER2-Positive Breast Cancer: Current and Future Prospects. Expert Rev. Anticancer. Ther. 2018, 18, 629–649. [Google Scholar] [CrossRef]
  22. Piccart-Gebhart, M.J.; Procter, M.; Leyland-Jones, B.; Goldhirsch, A.; Untch, M.; Smith, I.; Gianni, L.; Baselga, J.; Bell, R.; Jackisch, C.; et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 2005, 353, 1659–1672. [Google Scholar] [CrossRef]
  23. Gianni, L.; Dafni, U.; Gelber, R.D.; Azambuja, E.; Muehlbauer, S.; Goldhirsch, A.; Untch, M.; Smith, I.; Baselga, J.; Jackisch, C.; et al. Treatment with Trastuzumab for 1 Year after Adjuvant Chemotherapy in Patients with HER2-Positive Early Breast Cancer: A 4-Year Follow-up of a Randomised Controlled Trial. Lancet Oncol. 2011, 12, 236–244. [Google Scholar] [CrossRef]
  24. Goldhirsch, A.; Gelber, R.D.; Piccart-Gebhart, M.J.; de Azambuja, E.; Procter, M.; Suter, T.M.; Jackisch, C.; Cameron, D.; Weber, H.A.; Heinzmann, D.; et al. 2 Years versus 1 Year of Adjuvant Trastuzumab for HER2-Positive Breast Cancer (HERA): An Open-Label, Randomised Controlled Trial. Lancet 2013, 382, 1021–1028. [Google Scholar] [CrossRef]
  25. Cameron, D.; Piccart-Gebhart, M.J.; Gelber, R.D.; Procter, M.; Goldhirsch, A.; de Azambuja, E.; Castro, G.; Untch, M.; Smith, I.; Gianni, L.; et al. 11 Years’ Follow-up of Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Early Breast Cancer: Final Analysis of the HERceptin Adjuvant (HERA) Trial. Lancet 2017, 389, 1195–1205. [Google Scholar] [CrossRef]
  26. Romond, E.H.; Perez, E.A.; Bryant, J.; Suman, V.J.; Geyer, C.E.; Davidson, N.E.; Tan-Chiu, E.; Martino, S.; Paik, S.; Kaufman, P.A.; et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 2005, 353, 1673–1684. [Google Scholar] [CrossRef]
  27. Perez, E.A.; Romond, E.H.; Suman, V.J.; Jeong, J.-H.; Davidson, N.E.; Geyer, C.E.; Martino, S.; Mamounas, E.P.; Kaufman, P.A.; Wolmark, N. Four-Year Follow-Up of Trastuzumab Plus Adjuvant Chemotherapy for Operable Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Joint Analysis of Data from NCCTG N9831 and NSABP B-31. J. Clin. Oncol. 2011, 29, 3366–3373. [Google Scholar] [CrossRef] [PubMed]
  28. Perez, E.A.; Romond, E.H.; Suman, V.J.; Jeong, J.-H.; Sledge, G.; Geyer, C.E.; Martino, S.; Rastogi, P.; Gralow, J.; Swain, S.M.; et al. Trastuzumab Plus Adjuvant Chemotherapy for Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Planned Joint Analysis of Overall Survival From NSABP B-31 and NCCTG N9831. J. Clin. Oncol. 2014, 32, 3744–3752. [Google Scholar] [CrossRef]
  29. Slamon, D.; Eiermann, W.; Robert, N.; Pienkowski, T.; Martin, M.; Press, M.; Mackey, J.; Glaspy, J.; Chan, A.; Pawlicki, M.; et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N. Engl. J. Med. 2011, 365, 1273–1283. [Google Scholar] [CrossRef]
  30. Spielmann, M.; Roché, H.; Delozier, T.; Canon, J.-L.; Romieu, G.; Bourgeois, H.; Extra, J.-M.; Serin, D.; Kerbrat, P.; Machiels, J.-P.; et al. Trastuzumab for Patients with Axillary-Node-Positive Breast Cancer: Results of the FNCLCC-PACS 04 Trial. J. Clin. Oncol. 2009, 27, 6129–6134. [Google Scholar] [CrossRef]
  31. Martin, M.; Holmes, F.A.; Ejlertsen, B.; Delaloge, S.; Moy, B.; Iwata, H.; von Minckwitz, G.; Chia, S.; Mansi, J.; Barrios, C.H.; et al. Neratinib after trastuzumab-based adjuvant therapy in HER2-positive breast cancer (ExteNET): 5-year analysis of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. Oncol. 2017, 18, 1688–1700. [Google Scholar] [CrossRef]
  32. Chan, A.; Delaloge, S.; Holmes, F.A.; Moy, B.; Iwata, H.; Harker, G.; Masuda, N.; Konstantinovic, Z.B.N.; Petrakova, K.; Zotano, A.L.G.; et al. Abstract S5-02: Neratinib after Trastuzumab-Based Adjuvant Therapy in Early-Stage HER2+ Breast Cancer: 3-Year Analysis from a Phase 3 Randomized, Placebo-Controlled, Double-Blind Trial (ExteNET). Cancer Res. 2016, 76, S5-02. [Google Scholar] [CrossRef]
  33. von Minckwitz, G.; Procter, M.; de Azambuja, E.; Zardavas, D.; Benyunes, M.; Viale, G.; Suter, T.; Arahmani, A.; Rouchet, N.; Clark, E.; et al. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N. Engl. J. Med. 2017, 377, 122–131. [Google Scholar] [CrossRef] [PubMed]
  34. Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2019, 30, 1674. [Google Scholar] [CrossRef] [PubMed]
  35. Denduluri, N.; Chavez-MacGregor, M.; Telli, M.L.; Eisen, A.; Graff, S.L.; Hassett, M.J.; Holloway, J.N.; Hurria, A.; King, T.A.; Lyman, G.H.; et al. Selection of Optimal Adjuvant Chemotherapy and Targeted Therapy for Early Breast Cancer: ASCO Clinical Practice Guideline Focused Update. J. Clin. Oncol. 2018, 36, 2433–2443. [Google Scholar] [CrossRef]
  36. Schneeweiss, A.; Chia, S.; Hickish, T.; Harvey, V.; Eniu, A.; Hegg, R.; Tausch, C.; Seo, J.H.; Tsai, Y.-F.; Ratnayake, J.; et al. Pertuzumab plus Trastuzumab in Combination with Standard Neoadjuvant Anthracycline-Containing and Anthracycline-Free Chemotherapy Regimens in Patients with HER2-Positive Early Breast Cancer: A Randomized Phase II Cardiac Safety Study (TRYPHAENA). Ann. Oncol. 2013, 24, 2278–2284. [Google Scholar] [CrossRef] [PubMed]
  37. NCCN Clinical Practice Guidelines in Oncology (Breast Cancer), Version 4-2022. Available online: https://www.nccn.org/professionals/physician_gls/pdf/breast (accessed on 8 August 2022).
  38. Baselga, J.; Bradbury, I.; Eidtmann, H.; Cosimo, S.D.; de Azambuja, E.; Aura, C.; Gómez, H.; Dinh, P.; Fauria, K.; Van Dooren, V.; et al. Lapatinib with Trastuzumab for HER2-Positive Early Breast Cancer (NeoALTTO): A Randomised, Open-Label, Multicentre, Phase 3 Trial. Lancet 2012, 379, 633–640. [Google Scholar] [CrossRef]
  39. Gianni, L.; Pienkowski, T.; Im, Y.-H.; Roman, L.; Tseng, L.-M.; Liu, M.-C.; Lluch, A.; Staroslawska, E.; la Haba-Rodriguez, J.d.; Im, S.-A.; et al. Efficacy and Safety of Neoadjuvant Pertuzumab and Trastuzumab in Women with Locally Advanced, Inflammatory, or Early HER2-Positive Breast Cancer (NeoSphere): A Randomised Multicentre, Open-Label, Phase 2 Trial. Lancet Oncol. 2012, 13, 25–32. [Google Scholar] [CrossRef]
  40. Carey, L.A.; Berry, D.A.; Cirrincione, C.T.; Barry, W.T.; Pitcher, B.N.; Harris, L.N.; Ollila, D.W.; Krop, I.E.; Henry, N.L.; Weckstein, D.J.; et al. Molecular Heterogeneity and Response to Neoadjuvant Human Epidermal Growth Factor Receptor 2 Targeting in CALGB 40601, a Randomized Phase III Trial of Paclitaxel Plus Trastuzumab with or Without Lapatinib. J. Clin. Oncol. 2016, 34, 542–549. [Google Scholar] [CrossRef]
  41. Robidoux, A.; Tang, G.; Rastogi, P.; Geyer, C.E.; Azar, C.A.; Atkins, J.N.; Fehrenbacher, L.; Bear, H.D.; Baez-Diaz, L.; Sarwar, S.; et al. Lapatinib as a Component of Neoadjuvant Therapy for HER2-Positive Operable Breast Cancer (NSABP Protocol B-41): An Open-Label, Randomised Phase 3 Trial. Lancet Oncol. 2013, 14, 1183–1192. [Google Scholar] [CrossRef]
  42. Karagiannis, G.S.; Pastoriza, J.M.; Wang, Y.; Harney, A.S.; Entenberg, D.; Pignatelli, J.; Sharma, V.P.; Xue, E.A.; Cheng, E.; D’Alfonso, T.M.; et al. Neoadjuvant Chemotherapy Induces Breast Cancer Metastasis through a TMEM-Mediated Mechanism. Sci. Transl. Med. 2017, 9, eaan0026, Erratum in Sci. Transl. Med. 2017, 9, aao3817. [Google Scholar] [CrossRef]
  43. von Minckwitz, G.; Kummel, S.; Vogel, P.; Hanusch, C.; Eidtmann, H.; Hilfrich, J.; Gerber, B.; Huober, J.; Costa, S.D.; Jackisch, C.; et al. Neoadjuvant Vinorelbine-Capecitabine Versus Docetaxel-Doxorubicin-Cyclophosphamide in Early Nonresponsive Breast Cancer: Phase III Randomized GeparTrio Trial. JNCI J. Natl. Cancer Inst. 2008, 100, 542–551. [Google Scholar] [CrossRef]
  44. Smith, I.C.; Heys, S.D.; Hutcheon, A.W.; Miller, I.D.; Payne, S.; Gilbert, F.J.; Ah-See, A.K.; Eremin, O.; Walker, L.G.; Sarkar, T.K.; et al. Neoadjuvant Chemotherapy in Breast Cancer: Significantly Enhanced Response with Docetaxel. J. Clin. Oncol. 2002, 20, 1456–1466. [Google Scholar] [CrossRef] [PubMed]
  45. von Minckwitz, G.; Blohmer, J.U.; Costa, S.D.; Denkert, C.; Eidtmann, H.; Eiermann, W.; Gerber, B.; Hanusch, C.; Hilfrich, J.; Huober, J.; et al. Response-Guided Neoadjuvant Chemotherapy for Breast Cancer. J. Clin. Oncol. 2013, 31, 3623–3630. [Google Scholar] [CrossRef] [PubMed]
  46. Moreno-Aspitia, A. Neoadjuvant Therapy in Early-Stage Breast Cancer. Crit. Rev. Oncol. Hematol. 2012, 82, 187–199. [Google Scholar] [CrossRef] [PubMed]
  47. Bear, H.D.; Tang, G.; Rastogi, P.; Geyer, C.E.; Robidoux, A.; Atkins, J.N.; Baez-Diaz, L.; Brufsky, A.M.; Mehta, R.S.; Fehrenbacher, L.; et al. Bevacizumab Added to Neoadjuvant Chemotherapy for Breast Cancer. N. Engl. J. Med. 2012, 366, 310–320. [Google Scholar] [CrossRef] [PubMed]
  48. Reinisch, M.; Ataseven, B.; Kümmel, S. Neoadjuvant Dose-Dense and Dose-Intensified Chemotherapy in Breast Cancer-Review of the Literature. Breast Care 2016, 11, 13–20. [Google Scholar] [CrossRef]
  49. von Minckwitz, G.; Rezai, M.; Loibl, S.; Fasching, P.A.; Huober, J.; Tesch, H.; Bauerfeind, I.; Hilfrich, J.; Eidtmann, H.; Gerber, B.; et al. Capecitabine in Addition to Anthracycline- and Taxane-Based Neoadjuvant Treatment in Patients with Primary Breast Cancer: Phase III GeparQuattro Study. J. Clin. Oncol. 2010, 28, 2015–2023. [Google Scholar] [CrossRef]
  50. Bear, H.D.; Tang, G.; Rastogi, P.; Geyer, C.E.; Liu, Q.; Robidoux, A.; Baez-Diaz, L.; Brufsky, A.M.; Mehta, R.S.; Fehrenbacher, L.; et al. Neoadjuvant plus Adjuvant Bevacizumab in Early Breast Cancer (NSABP B-40 [NRG Oncology]): Secondary Outcomes of a Phase 3, Randomised Controlled Trial. Lancet Oncol. 2015, 16, 1037–1048. [Google Scholar] [CrossRef]
  51. Halloran, N.O.; McVeigh, T.; Martin, J.; Keane, M.; Lowery, A.; Kerin, M. Neoadjuvant Chemoradiation and Breast Reconstruction: The Potential for Improved Outcomes in the Treatment of Breast Cancer. Ir. J. Med. Sci. (1971-) 2018, 188, 75–83. [Google Scholar] [CrossRef] [PubMed]
  52. Thomas, E.; Holmes, F.A.; Smith, T.L.; Buzdar, A.U.; Frye, D.K.; Fraschini, G.; Singletary, S.E.; Theriault, R.L.; McNeese, M.D.; Ames, F.; et al. The Use of Alternate, Non-Cross-Resistant Adjuvant Chemotherapy on the Basis of Pathologic Response to a Neoadjuvant Doxorubicin-Based Regimen in Women with Operable Breast Cancer: Long-Term Results From a Prospective Randomized Trial. J. Clin. Oncol. 2004, 22, 2294–2302. [Google Scholar] [CrossRef]
  53. Gonzalez-Angulo, A.M.; Lei, X.; Alvarez, R.H.; Green, M.C.; Murray, J.L.; Valero, V.; Koenig, K.B.; Ibrahim, N.K.; Litton, J.K.; Nair, L.; et al. Phase II Randomized Study of Ixabepilone Versus Observation in Patients with Significant Residual Disease After Neoadjuvant Systemic Therapy for HER2-Negative Breast Cancer. Clin. Breast Cancer 2015, 15, 325–331. [Google Scholar] [CrossRef]
  54. Kalra, M.; Tong, Y.; Jones, D.R.; Walsh, T.; Danso, M.A.; Ma, C.X.; Silverman, P.; King, M.-C.; Badve, S.S.; Perkins, S.M.; et al. Cisplatin +/− Rucaparib after Preoperative Chemotherapy in Patients with Triple-Negative or BRCA Mutated Breast Cancer. Npj Breast Cancer 2021, 7, 29. [Google Scholar] [CrossRef] [PubMed]
  55. von Minckwitz, G.; Rezai, M.; Tesch, H.; Huober, J.; Gerber, B.; Zahm, D.M.; Hilfrich, J.; Costa, S.D.; Dubsky, P.; Blohmer, J.U.; et al. Zoledronate for Patients with Invasive Residual Disease after Anthracyclines-Taxane-Based Chemotherapy for Early Breast Cancer\textendash The Phase III NeoAdjuvant Trial Add-oN (NaTaN) Study (GBG 36/ABCSG 29). Eur. J. Cancer 2016, 64, 12–21. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. Isakoff, S.J.; Baselga, J. Trastuzumab-DM1: Building a Chemotherapy-Free Road in the Treatment of Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. J. Clin. Oncol. 2011, 29, 351–354. [Google Scholar] [CrossRef] [PubMed]
  58. Diéras, V.; Miles, D.; Verma, S.; Pegram, M.; Welslau, M.; Baselga, J.; Krop, I.E.; Blackwell, K.; Hoersch, S.; Xu, J.; et al. Trastuzumab Emtansine versus Capecitabine plus Lapatinib in Patients with Previously Treated HER2-Positive Advanced Breast Cancer (EMILIA): A Descriptive Analysis of Final Overall Survival Results from a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2017, 18, 732–742. [Google Scholar] [CrossRef]
  59. Krop, I.E.; Kim, S.-B.; González-Martín, A.; LoRusso, P.M.; Ferrero, J.-M.; Smitt, M.; Yu, R.; Leung, A.C.F.; Wildiers, H. Trastuzumab Emtansine versus Treatment of Physician’s Choice for Pretreated HER2-Positive Advanced Breast Cancer (TH3RESA): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2014, 15, 689–699. [Google Scholar] [CrossRef]
  60. Krop, I.E.; Kim, S.-B.; Martin, A.G.; LoRusso, P.M.; Ferrero, J.-M.; Badovinac-Crnjevic, T.; Hoersch, S.; Smitt, M.; Wildiers, H. Trastuzumab Emtansine versus Treatment of Physician’s Choice in Patients with Previously Treated HER2-Positive Metastatic Breast Cancer (TH3RESA): Final Overall Survival Results from a Randomised Open-Label Phase 3 Trial. Lancet Oncol. 2017, 18, 743–754. [Google Scholar] [CrossRef]
  61. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.-Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef]
  62. von Minckwitz, G.; Huang, C.S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Can-cer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef]
  63. Medicine USNLo. Phase II Trial of Combination Immunotherapy with NeuVax and Trastuzumab in High-risk HER2+ Breast Cancer Patients (HER3+) (2014–2021). Available online: https://clinicaltrials.gov/ct2/show/NCT02297698 (accessed on 16 November 2021).
  64. Medicine USNLo. A Multicenter Phase II Study of Vaccines to Prevent Recurrence in Patients With HER-2Positive Breast Cancer (2018–2023). Available online: https://clinicaltrials.gov/ct2/history/NCT03384914?V_2=View (accessed on 12 October 2021).
  65. Sikov, W.M.; Berry, D.A.; Perou, C.M.; Singh, B.; Cirrincione, C.T.; Tolaney, S.M.; Kuzma, C.S.; Pluard, T.J.; Somlo, G.; Port, E.R.; et al. Impact of the addition of carboplatin and/or bevacizumab to Neoadjuvant Once per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete re-sponse rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance). J. Clin. Oncol. 2015, 33, 13–21. [Google Scholar]
  66. 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]
  67. 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]
  68. Sharma, P.; López-Tarruella, S.; García-Saenz, J.A.; Ward, C.; Connor, C.S.; Gómez, H.L.; Prat, A.; Moreno, F.; Jerez-Gilarranz, Y.; Barnadas, A.; et al. Efficacy of Neoadjuvant Carboplatin plus Docetaxel in Triple-Negative Breast Cancer: Combined Analysis of Two Cohorts. Clin. Cancer Res. 2017, 23, 649–657. [Google Scholar] [CrossRef] [PubMed]
  69. Gluz, O.; Nitz, U.; Liedtke, C.; Prat, A.; Christgen, M.; Feuerhake, F.; Garke, M.; Grischke, E.-M.; Forstbauer, H.; Braun, M.; et al. Abstract GS5-06: No Survival Benefit of Chemotherapy Escalation in Patients with pCR and “High-Immune” Triple-Negative Early Breast Cancer in the Neoadjuvant WSG-ADAPT-TN Trial. Cancer Res. 2019, 79, GS5-06. [Google Scholar] [CrossRef]
  70. Yamamoto, Y.; Iwase, H. Clinicopathological Features and Treatment Strategy for Triple-Negative Breast Cancer. Int. J. Clin. Oncol. 2010, 15, 341–351. [Google Scholar] [CrossRef]
  71. Bidard, F.-C.; Matthieu, M.-C.; Chollet, P.; Raoefils, I.; Abrial, C.; Dômont, J.; Spielmann, M.; Delaloge, S.; André, F.; Penault-Llorca, F. P53 Status and Efficacy of Primary Anthracyclines/Alkylating Agent-Based Regimen According to Breast Cancer Molecular Classes. Ann. Oncol. 2008, 19, 1261–1265. [Google Scholar] [CrossRef]
  72. Bergin, A.R.T.; Loi, S. Triple-Negative Breast Cancer: Recent Treatment Advances. F1000Research 2019, 8, 1342. [Google Scholar] [CrossRef]
  73. Arun, B.; Bayraktar, S.; Liu, D.D.; Barrera, A.M.G.; Atchley, D.; Pusztai, L.; Litton, J.K.; Valero, V.; Meric-Bernstam, F.; Hortobagyi, G.N.; et al. Response to Neoadjuvant Systemic Therapy for Breast Cancer in BRCA Mutation Carriers and Noncarriers: A Single-Institution Experience. J. Clin. Oncol. 2011, 29, 3739–3746. [Google Scholar] [CrossRef]
  74. Byrski, T.; Huzarski, T.; Dent, R.; Gronwald, J.; Zuziak, D.; Cybulski, C.; Kladny, J.; Gorski, B.; Lubinski, J.; Narod, S.A. Response to Neoadjuvant Therapy with Cisplatin in BRCA1-Positive Breast Cancer Patients. Breast Cancer Res. Treat. 2008, 115, 359–363. [Google Scholar] [CrossRef]
  75. Silver, D.P.; Richardson, A.L.; Eklund, A.C.; Wang, Z.C.; Szallasi, Z.; Li, Q.; Juul, N.; Leong, C.-O.; Calogrias, D.; Buraimoh, A.; et al. Efficacy of Neoadjuvant Cisplatin in Triple-Negative Breast Cancer. J. Clin. Oncol. 2010, 28, 1145–1153. [Google Scholar] [CrossRef]
  76. Telli, M.L.; Kurian, A.W.; Jensen, K.C.; Vinayak, S.; Flaherty, P.; Lipson, J.A.; Wapnir, I.; Daniel, B.; Carlson, R.W.; Mills, M.A.; et al. P3-14-08: A Phase II Study of Gemcitabine and Carboplatin (GC) Plus Iniparib (BSI-201) as Neoadjuvant Therapy for Triple-Negative and BRCA1/2 Mutation-Associated Breast Cancer. Cancer Res. 2011, 71, P3-14-08. [Google Scholar] [CrossRef]
  77. 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] [PubMed]
  78. Viale, G.; Bottiglieri, L. Pathological Definition of Triple Negative Breast Cancer. Eur. J. Cancer 2009, 45, 5–10. [Google Scholar] [CrossRef]
  79. Coates, A.S.; Winer, E.P.; Goldhirsch, A.; Gelber, R.D.; Gnant, M.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J.; André, F.; Baselga, J.; et al. 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, 1533–1546. [Google Scholar] [CrossRef] [PubMed]
  80. Ma, Y.; Zhang, S.; Zang, L.; Li, J.; Li, J.; Kang, Y.; Ren, W. Combination of Shear Wave Elastography and Ki-67 Index as a Novel Predictive Modality for the Pathological Response to Neoadjuvant Chemotherapy in Patients with Invasive Breast Cancer. Eur. J. Cancer 2016, 69, 86–101. [Google Scholar] [CrossRef] [PubMed]
  81. Cabrera-Galeana, P.; Muñoz-Montaño, W.; Lara-Medina, F.; Alvarado-Miranda, A.; Pérez-Sánchez, V.; Villarreal-Garza, C.; Quintero, R.M.; Porras-Reyes, F.; Bargallo-Rocha, E.; Carmen, I.D.; et al. Ki67 Changes Identify Worse Outcomes in Residual Breast Cancer Tumors After Neoadjuvant Chemotherapy. Oncologist 2018, 23, 670–678. [Google Scholar] [CrossRef]
  82. Muss, H.B.; Polley, M.C.; Berry, D.A.; Liu, H.; Cirrincione, C.T.; Theodoulou, M.; Mauer, A.M.; Kornblith, A.B.; Partridge, A.H.; Dressler, L.G.; et al. Randomized Trial of Standard Adjuvant Chemotherapy Regimens Versus Capecitabine in Older Women with Early Breast Cancer: 10-Year Update of the CALGB 49907 Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 2338–2348. [Google Scholar] [CrossRef]
  83. Lluch, A.; Gomez, H.; Ruiz-Borrego, M.; Bines, J.; Llombart, A.; Ramos, M.; Torres, R.; Martins-Segalla, J.G.; Torrecillas, L.; Barrios, C. Abstract P5-10-15: First Safety Data from a Randomised Phase III (CIBOMA 2004-01/GEICAM 2003-11) Trial Assessing Adjuvant Capecitabine Maintenance Therapy after Standard Chemotherapy for Triple-Negative Early Breast Cancer. Cancer Res. 2010, 70, P5-10-15. [Google Scholar] [CrossRef]
  84. Berger, E.R.; Park, T.; Saridakis, A.; Golshan, M.; Greenup, R.A.; Ahuja, N. Immunotherapy Treatment for Triple Negative Breast Cancer. Pharmaceuticals 2021, 14, 763. [Google Scholar] [CrossRef]
  85. Pusztai, L.; Denkert, C.; O’Shaughnessy, J.; Cortes, J.; Dent, R.A.; McArthur, H.L.; Kuemmel, S.; Bergh, J.C.S.; Park, Y.H.; Hui, R.; 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. [Google Scholar] [CrossRef]
  86. Gianni, L.; Huang, C.-S.; Egle, D.; Bermejo, B.; Zamagni, C.; Thill, M.; Anton, A.; Zambelli, S.; Bianchini, G.; Russo, S.; et al. Abstract GS3-04: Pathologic Complete Response (pCR) to Neoadjuvant Treatment with or without Atezolizumab in Triple Negative, Early High-Risk and Locally Advanced Breast Cancer. NeoTRIPaPDL1 Michelangelo Randomized Study. Cancer Res. 2020, 80, GS3-04. [Google Scholar] [CrossRef]
  87. 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]
  88. Medicine USNLo. Testing MK-3475 (Pembrolizumab) as Adjuvant Therapy for Triple Receptor-Negative Breast Cancer (2016-2021). Available online: https://clinicaltrials.gov/ct2/show/NCT02954874 (accessed on 5 May 2022).
  89. 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] [PubMed]
  90. Pelizzari, G.; Gerratana, L.; Basile, D.; Fanotto, V.; Bartoletti, M.; Liguori, A.; Fontanella, C.; Spazzapan, S.; Puglisi, F. Post-Neoadjuvant Strategies in Breast Cancer: From Risk Assessment to Treatment Escalation. Cancer Treat. Rev. 2019, 72, 7–14. [Google Scholar] [CrossRef]
  91. Patel, S.A.; DeMichele, A. Adding Adjuvant Systemic Treatment after Neoadjuvant Therapy in Breast Cancer: Review of the Data. Curr. Oncol. Rep. 2017, 19, 56. [Google Scholar] [CrossRef]
  92. Labrosse, J.; Osdoit, M.; Hamy, A.-S.; Coussy, F.; Pierga, J.-Y.; Reyal, F.; Laas, E. Adjuvant Chemotherapy for Breast Cancer after Preoperative Chemotherapy: A Propensity Score Matched Analysis. PLoS ONE 2020, 15, e0234173. [Google Scholar] [CrossRef]
  93. Foldi, J.; Rozenblit, M.; Park, T.S.; Knowlton, C.A.; Golshan, M.; Moran, M.; Pusztai, L. Optimal Management for Residual Disease Following Neoadjuvant Systemic Therapy. Curr. Treat. Options Oncol. 2021, 22, 79. [Google Scholar] [CrossRef]
  94. Ferraro, E.; Drago, J.Z.; Modi, S. Implementing Antibody-Drug Conjugates (ADCs) in HER2-Positive Breast Cancer: State of the Art and Future Directions. Breast Cancer Res. 2021, 23, 84. [Google Scholar] [CrossRef]
Figure 1. Suggested algorithm for management of residual disease following neoadjuvant therapy and surgery. * Continue immunotherapy if used prior to surgery. # May consider immunotherapy in clinical trials. HER2: human epidermal growth factor receptor 2; HR: hormone receptor; LN: lymph node; XRT: radiation therapy; T-DM1: trastuzumab emtansine.
Figure 1. Suggested algorithm for management of residual disease following neoadjuvant therapy and surgery. * Continue immunotherapy if used prior to surgery. # May consider immunotherapy in clinical trials. HER2: human epidermal growth factor receptor 2; HR: hormone receptor; LN: lymph node; XRT: radiation therapy; T-DM1: trastuzumab emtansine.
Curroncol 29 00458 g001
Table 1. Rates of pathologic complete response (pCR) in HER2-positive subtypes.
Table 1. Rates of pathologic complete response (pCR) in HER2-positive subtypes.
Clinical TrialDual
HER2
Therapy
pCR Rate with Single
Anti-HER2 Agent
(%)
pCR Rate with Dual
Anti-HER2 Therapy
(%)
NeoALTTO [38]L + T30 (95% CI, 22.4–37.5) with T
25 (95% CI, 18.1–32.3) with L
51 (95% CI, 43.1–59.5; p = 0.0001)
NeoSphere [39]T + P29 (95% CI, 20.6–38.5) with T
24 (95% CI, 15.8–33.7) with P
46 (95% CI, 36.1–55.7; p = 0.0141)
CALGB 40601 [40]L + T40 (95% CI, 32–49) with T
32 (95% CI, 22–44) with L
51 (95% CI, 42–60; p = 0.11)
NSABP B-41 [41]L + T53 (95% CI, 44.9–59.5) with T
53 (95% CI, 44.4–60.3) with L
62 (95% CI, 54.3–68.8; p = 0.095)
TRYPHAENA [36]T + PNA57–66
HER2: human epidermal growth factor receptor; L: lapatinib; T: trastuzumab; P: pertuzumab; pCR: pathologic complete response; NA: not available.
Table 2. Pathologic complete response (pCR) rates using different neoadjuvant therapies for triple-negative (TN) disease.
Table 2. Pathologic complete response (pCR) rates using different neoadjuvant therapies for triple-negative (TN) disease.
TreatmentPathologic Complete Response (pCR) Rates (%)
Anthracycline [70,71]14–47
Anthracycline + taxane [72]17–39
Anthracycline + cyclophosphamide followed by taxanes [65,66,73]30–44
Carboplatin to a backbone of anthracycline/taxanes [65,66,67,68]52–57
Platinum monotherapy (carboplatin, cisplatin) [74,75]23–90 *
Gemcitabine + carboplatin + iniparib [76]36
Pembrolizumab + standard chemotherapy [77]64.8
* Highest among patients with BRCA gene mutations.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abdel-Razeq, H.; Khalil, H.; Assi, H.I.; Dargham, T.B. Treatment Strategies for Residual Disease following Neoadjuvant Chemotherapy in Patients with Early-Stage Breast Cancer. Curr. Oncol. 2022, 29, 5810-5822. https://doi.org/10.3390/curroncol29080458

AMA Style

Abdel-Razeq H, Khalil H, Assi HI, Dargham TB. Treatment Strategies for Residual Disease following Neoadjuvant Chemotherapy in Patients with Early-Stage Breast Cancer. Current Oncology. 2022; 29(8):5810-5822. https://doi.org/10.3390/curroncol29080458

Chicago/Turabian Style

Abdel-Razeq, Hikmat, Hanan Khalil, Hazem I. Assi, and Tarek Bou Dargham. 2022. "Treatment Strategies for Residual Disease following Neoadjuvant Chemotherapy in Patients with Early-Stage Breast Cancer" Current Oncology 29, no. 8: 5810-5822. https://doi.org/10.3390/curroncol29080458

Article Metrics

Back to TopTop