Pyrotinib combined with trastuzumab and chemotherapy for the treatment of human epidermal growth factor receptor 2-positive metastatic breast cancer: a single-arm exploratory phase II trial

DOI: https://doi.org/10.21203/rs.3.rs-1752120/v1

Abstract

Purpose: A substantial need for effective and safe treatment options is still unmet for patients with heavily pre-treated human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC). Herein, we assessed the efficacy and safety of pyrotinib plus trastuzumab and chemotherapy in patients with heavily treated HER2-positive MBC.

Methods: In this single-arm exploratory phase II trial, patients with HER2-positive MBC previously treated with trastuzumab plus lapatinib or pertuzumab, received pyrotinib plus trastuzumab and chemotherapy. The primary end point was progression-free survival (PFS) in the total population (TP). Secondary end points included PFS in the subgroup with brain metastases (Sub-BrM), confirmed objective response rate (ORR), clinical benefit rate (CBR), disease control rate (DCR), exploration of predictive factors of PFS, and safety.

Results: Between November 1, 2018, and March 31, 2021, 40 patients were eligible for this study. The median PFS reached 7.5 months (95% confidence interval [CI], 4.7 to 9.9 months) and 9.4 months (95% CI, 6.6 to 12.1 months) in the TP and Sub-BrM, respectively. ORR was 50.5% (20/40). CBR was 75.5% (30/40) and DCR reached 97.5% (39/40). Cox univariate and multivariate analyses demonstrated that liver or/and lung metastases was the significant adverse prognostic factor for PFS (p = 0.018; p = 0.026; respectively). The most frequent grade 3 or 4 treatment-related adverse events were diarrhea, neutropenia and leukopenia. No new safety signals were observed.

Conclusion: Pyrotinib plus trastuzumab and chemotherapy offered a promising option with manageable safety profile for heavily pre-treated HER2-positive MBC, especially for those without liver or/and lung metastases.

Introduction

Breast cancer (BC) has surpassed lung cancer as the most commonly diagnosed malignancy in women according to Global Cancer statistics 2020 [1]. Approximately 15 to 20% of BC cases exhibit overexpression of the human epidermal growth factor receptor 2 (HER2) or/and amplification of the coding gene ERBB2, which is defined as HER2-positive subtype [2]. HER2-positive BC is more aggressive and prone to recurrence than HER2-negative tumors [3]. In the last decades, the clinical outcome of HER2-positive BC has been significantly improved since the introduction of HER2-targeted drugs mainly including monoclonal antibodies, tyrosine kinase inhibitors (TKIs) and antibody drug conjugates (ADCs). Currently, the standard of care for HER2-positive metastatic breast cancer (MBC) involves trastuzumab plus pertuzumab and a taxane as the first-line regimen, followed by second-line trastuzumab emtansine (T-DM1) or fam-trastuzumab deruxtecan-nxki (ds8201) for patients whose disease has progressed on prior dual HER2 blockade [4]. However, a large proportion of patients with HER2-positive MBC fail to be treated with standard second-line regimens due to economic and drug accessibility factors. Moreover, most patients inevitably experience disease progression on anti-HER2 targeted therapy due to de novo or acquired resistance. Therefore, it is necessary to explore novel approaches to overcome drug resistance. Key strategies include efficient suppression of the HER2 signaling pathway by dual blockade and development of more effective anti-HER2 therapies like antibody–drug conjugates, new anti-HER2 antibodies, bispecific antibodies and novel TKIs [5].

Brain metastases (BrM) is a common complication of advanced malignant disease and occurs in 1/3 of HER2-positive metastatic breast cancer [6]. Central nervous system-directed local therapies, including surgical resection and radiotherapy, are the foundations of BrM management [7]. Besides, TKIs and several chemotherapeutic drugs with blood-brain barrier penetrability have demonstrated efficacy in HER2-positive MBC patients with BrM. Despite sequential local and systemic treatment, resistance inevitably develops and options are limited for the control of BrM.

Studies have confirmed the activity of continued trastuzumab treatment beyond progression (TBP). The subgroup analysis of observational HERMINE study suggested that trastuzumab TBP offered a survival benefit to MBC patients treated with first-line trastuzumab [8]. Furthermore, a phase III study in HER2-positive BC showed that patients receiving continued trastuzumab TBP had a better post-progression survival than those not receiving (18.8m vs 13.3m, p = 0.02) [9]. Meanwhile, the potent anti-tumor efficacy of TKIs including lapatinib, neratinib, tucatinib and pyrotinib has been demonstrated in several pivotal studies [1014]. Pyrotinib is an orally administered irreversible pan-ErbB TKI which shows anti-tumor activity and acceptable safety profile in HER2-positive advanced and metastatic breast cancer [14]. In a phase II study, pyrotinib plus capecitabine yielded statistically significant better overall response rate (ORR) and progression-free survival (PFS) than lapatinib plus capecitabine in patients with HER2-positive MBC previously treated with taxanes, anthracyclines, and/or trastuzumab [15]. Based on this phase II study, pyrotinib in combination with capecitabine received its first conditional approval in China for the treatment of HER2-positive, advanced or metastatic breast cancer previously exposed to anthracycline or taxane chemotherapy. Afterwards, a multi-center, open-label, randomized, controlled, phase III trial (PHOEBE) confirmed the efficacy and safety of pyrotinib plus capecitabine in patients with disease progression on previous trastuzumab [16]. Another phase III trial PHENIX further verified that pyrotinib plus capecitabine significantly improved PFS and ORR compared with capecitabine monotherapy in trastuzumab-treated patients with HER2-postive advanced or metastatic breast cancer, including those with brain metastases [17].

Dual HER2 blockade by trastuzumab plus TKI, simultaneously targeting the extra- and intra-cellular domains of HER2, showed encouraging antitumor activity in BC, including early breast cancer and MBC with intracranial metastases [1821]. Our study aimed to explore the activity and safety of pyrotinib combined with trastuzumab and chemotherapy of physician’s choice in heavily pre-treated patients with HER2-positive MBC with or without BrM.

Methods

Patients and data collection

From November 1, 2018, to March 31, 2021, female patients aged ≥ 18 years old with histologically confirmed HER2-positive MBC at National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital (Shenzhen, Guangdong, China) were incorporated in this study. HER2 status was determined by central review based on immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH) examination using primary or metastatic lesion samples. Patients had been previously treated with trastuzumab, pertuzumab and/or lapatinib in the (neo)adjuvant/metastatic setting. Additional requirements included an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 or 1 (on a 5-point scale by which greater scores reflect higher degree of disability); measurable lesions; and adequate organ function. Patients were excluded if they had previously received treatment with pyrotinib for metastatic disease; had symptomatic brain metastasis which necessitates immediate local intervention; or had leptomeningeal disease. All procedures performed involving human participants were in accordance with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. This study was also approved by the institutional review board and ethics committee. All patients provided written informed consent for use of the medical information for research purpose.

Treatment

All eligible patients were treated with pyrotinib (240–400 mg orally once daily), trastuzumab (6 mg per kilogram of body weight intravenously per 21 days, with an initial loading dose of 8 mg per kilogram) and single chemotherapeutic agent (nab-paclitaxel, 260 mg per square meter of body-surface area intravenously on day 1 of each 21-day cycle; or capecitabine, 1000 mg per square meter of body-surface area orally twice daily on days 1 to 14 of each 21-day cycle; or gemcitabine, 1000 mg per square meter of body-surface area intravenously on days 1 and 8 of each 21-day cycle; or vinorelbine, 60 mg per square meter of body-surface area orally on days 1 and 8 of each 21-day cycle and dose elevation to 80 mg per square meter of body-surface area was allowed if well tolerated).

Follow-up and assessment

All patients were followed up until January 20, 2022. Disease response was evaluated according to imaging reports from serial clinical assessments. Patient/disease response assessments were performed at baseline, every 6 weeks for 24 weeks, and every 9 weeks thereafter, including performance status, history, laboratory examinations, electrocardiogram, contrast-enhanced spiral computed tomography (CT) or positron emission tomography/CT (PET/CT), and contrast-enhanced magnetic resonance imaging (MRI). MRI of the head was obtained for all patients at baseline and head scans were repeated at the frequency described above if BrM had been detected at baseline. MRI of the breast was not required. Disease was evaluated in accordance with Response Evaluation Criteria in Solid Tumors (RECIST) criteria, version 1.1 [22]. Safety was assessed mainly via the incidence of treatment-related adverse events (TRAEs) using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0.

End points

The primary end point was PFS (calculated from the date of first treatment with double targeted therapy to the date of documented disease progression or death from any cause or the last follow-up visit) in TP. The secondary end points included PFS in the subgroup with BrM (Sub-BrM) at baseline; objective response rate (ORR, defined as the percentage of patients who had a confirmed complete response or partial response); clinical benefit rate (CBR, defined as the percentage of patients who had a confirmed complete response or partial response or stable disease for at least 24 weeks); disease control rate (DCR, defined as the percentage of patients who had a confirmed complete response or partial response or stable disease for at least 4 weeks); exploration of predictive factors of PFS; and safety.

Statistical analysis

All data were analyzed using SPSS 23.0 statistical software (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). Descriptive analysis was utilized to display clinicopathological features. The Kaplan–Meier method was used to estimate PFS and 95% confidence intervals (CIs) for the total population and the subgroups. Cox univariate and multivariate models were used to determine the predictive value of variables for PFS. In the analysis of progression-free survival, data from patients who did not have any documented event, were lost to follow-up or died from any cause were censored at the last date when the patient was known to be event-free. All reported p values were two-sided, with p༜0.05 being regarded as statistically significant.

Results

Patient clinicopathological characteristics

Between November 1, 2018, and March 31, 2021, a total of 40 patients were eligible for this study and included in the final analysis. In TP, the median age at diagnosis was 46 years (range, 31–62 years). 33 patients (82.5%) were aged over 35 years old at diagnosis. ECOG performance status was 0 in 21 patients (52.5%) and 1 in 19 patients (47.5%). For HER2 status, HER2 3 + by IHC was recorded in 32 patients (80.0%), and HER2 2 + by IHC and amplification by FISH in 8 patients (20.0%). 21 patients (52.5%) were estrogen receptor (ER) and/or progesterone receptor (PR) positive and 19 patients (47.5%) were ER and PR negative. 39 patients (97.5%) had been treated with trastuzumab in the (neo)adjuvant or metastatic setting; 6 patients had received pertuzumab and trastuzumab as first-line therapeutic regimen; 21 patients (52.5%) received anti-HER2 therapy with TKI (lapatinib) for their metastatic disease; none of them was previously treated with TDM-1. 15 patients (37.5%) had brain metastases and 27 patients (67.5%) had lung or/and liver metastases at baseline. Of those patients with brain metastases, 7 patients had received central nervous system (CNS) local therapies including whole-brain radiotherapy (WBRT), stereotactic radiotherapy (SRT) and surgery plus SRT before study treatment; 3 patients whose intra-cranial disease progressed while remission of extra-cranial lesions lasted, received SRT with unchanged systemic therapy. 13 patients (32.5%) received at least 3 lines of therapy in the metastatic setting. Combined chemotherapeutic agents included vinorelbine (25, 62.5%), capecitabine (6, 15.0%), nab-paclitaxel (5, 12.5%) and gemcitabine (4, 10.0%). Patient clinicopathological characteristics at baseline were shown in Table 1.

Table 1

Patient Clinicopathological Characteristics at Baseline

Characteristics

cases (%)

ECOG

 

0

21 (52.5)

1

19 (47.5)

Age at diagnosis

 

≤ 35 years

7 (17.5)

> 35 years

33 (82.5)

Location

 

Left

27 (67.5)

Right

13 (32.5)

HER2 status

 

IHC 3+

32 (80.0)

IHC 2 + and FISH +

8 (20.0)

Hormone-receptor status

 

ER and/or PR positive

21 (52.5)

ER and PR negative

19 (47.5)

Previous anti-HER2 antibody treatment in metastatic setting

 

Yes

37 (92.5)

No

3 (7.5)

Previous anti-HER2 TKI treatment

 

Yes

21 (52.5)

No

19 (47.5)

Brain metastases

 

Yes

15 (37.5)

No

25 (62.5)

Liver or/and lung metastases

 

Yes

27 (67.5)

No

13 (32.5)

Number of previous treatment lines

 

≤ 2

27 (67.5)

> 2

13 (32.5)

Combined chemotherapeutic drug

 

Vinorelbine

25 (62.5)

Capecitabine

6 (15.0)

Nab-paclitaxel

5 (12.5)

Gemcitabine

4 (10.0)

Abbreviations:
ECOG Eastern Cooperative Oncology Group; HER2 human epidermal growth factor receptor 2; IHC immunohistochemistry; ER estrogen receptor; PR progesterone receptor.

Efficacy

By the end of follow-up, 32 patients had experienced disease progression and 3 patients had died. The median duration of PFS was 7.5 months in TP (95% CI, 4.7 to 9.9 months, Fig. 1) and was 9.4 months in Sub-BrM (95% CI, 6.6 to 12.1 months, Fig. 2). Objective response was seen in 20 of 40 patients (including 1 patient with complete response and 19 patients with partial response, 50.0%, Table 2). All patients with objective response achieved disease remission within four cycles (12 weeks) after treatment administered. Clinical benefit was observed in 30 of 40 patients (75.5%) and disease control in 39 of 40 patients (97.5%, Table 2).

Table 2

Efficacy outcomes in the total population and subgroup with brain metastases at baseline

End points

 

Median progression-free survival, months (95% CI)

7.5 (4.4 to 7.9)

Median progression-free survival in subgroup with brain metastases, months (95% CI)

9.4 (6.6 to 12.1)

Type of response, No. (%)

 

Complete

1 (2.5)

Partial

19 (45.0)

Stable disease

19 (50.0)

Disease progression

1 (2.5)

Objective response rate, No. (%)*

20 (50.0)

Clinical benefit rate, No. (%)**

30 (75.5)

Disease control rate, No. (%)***

39 (97.5)

*Include complete and partial response. **Include complete and partial response and stable disease lasting for at least 24 weeks. ***Include complete and partial response and stable disease lasting for at least 4 weeks. Abbreviations: CI confidence interval.

Univariate and multivariate analyses were performed to identify predictive factors of PFS for the 40 patients. By means of Cox univariate and multivariate analyses, we found that liver or/and lung metastases at baseline was the significant adverse prognostic factor for PFS (hazard ratio [HR], 0.322; 95% CI, 0.132 to 0.831; p = 0.018; HR, 0.281; 95% CI, 0.093 to 0.876; p = 0.026; respectively; Table 3). The median PFS was 6.0 months (95% CI, 2.8 to 9.2 months) in the subgroup with liver or/and lung metastases and was not reached in the subgroup without (Fig. 3).

Table 3

Cox regression analysis for progression-free survival (N = 40)

Variables

Univariate analysis

 

Multivariate analysis

 

HR

95% CI

P value

 

HR

95% CI

P value

Age at diagnosis, years (≤ 60 vs > 60)

1.277

0.484–3.368

0.622

       

Location (left vs right)

1.219

0.547–2.716

0.629

       

HER2 status (IHC 3 + vs IHC2 + and FISH +)

1.017

0.355–2.687

0.972

       

Hormone-receptor status (ER and/or PR positive vs ER and PR negative)

0.639

0.299–1.367

0.248

       

Previous anti-HER2 antibody treatment in metastatic setting (yes vs no)

0.947

0.225–4.037

0.953

       

Previous anti-HER2 TKI treatment (yes vs no)

0.958

0.451–2.033

0.911

       

Brain metastases (yes vs no)

1.877

0.843–4.179

0.123

       

Liver or/and lung metastases (yes vs no)

0.322

0.132–0.831

0.018

 

0.281

0.093–0.876

0.026

Number of previous treatment lines (≤ 2 vs > 2)

1.190

0.548–2.585

0.660

       

Combined chemotherapeutic drug (Vinorelbine vs Cepecitabine vs Nab-paclitaxel)

1.197

0.834–1.717

0.329

       

Abbreviations: HR hazard ratio; CI confidence interval; HER2 = human epidermal growth factor receptor; IHC immunohistochemistry; FISH fluorescence in situ hybridization; ER estrogen receptor; PR progesterone receptor; TKI tyrosine kinase inhibitor.

Safety

All treatment-related adverse events (TRAEs) reported in the study were summarized in Table 4. The most common adverse events (frequency ≥ 10%) were diarrhea (85.0%), leukopenia (42.5%), neutropenia (37.5%), fatigue (37.5%), vomiting (32.5%), and nausea (12.5%) in turn. Other recorded adverse events included hand-foot syndrome (10.0%), aspartate aminotransferase increase (10.0%), anorexia (7.5%), alanine aminotransferase increase (5.0%), anemia (5.0%), peripheral neurotoxicity (5.0%), rash (2.5%) and thrombocytopenia (2.5%). The most frequent grade 3 to 4 adverse events were diarrhea (25.0%), neutropenia (15.0%), and leukopenia (12.5%). Grade 4 adverse events occurred in 1 patient (leukopenia and febrile neutropenia). No grade 4 diarrhea or cardiac-related events were reported. No one discontinued study treatment or died owing to TRAEs. However, 14 patients (35.0%) experienced pyrotinib dose reduction because of diarrhea (10 patients reduced from 400 mg to 320 mg and 4 patients from 400 mg to 240 mg). 4 patients experienced chemotherapeutic drug dose reduction (nab-paclitaxel in 1 patient, capecitabine in 1 patient, and vinorelbine in 2 patients). Diarrhea, the most common adverse event, mostly occurred within the first two weeks after treatment initiation and could be managed by the administration of loperamide or/and pyrotinib dose reduction.

Table 4

Treatment-related adverse events in total population

Adverse events

Any grade (%)

Grade ≥ 3 (%)

Diarrhea

34 (85.0)

10 (25.0)

Leukopenia

17 (42.5)

5 (12.5)

Neutropenia

15 (37.5)

6 (15.0)

Fatigue

15 (37.5)

0

Vomiting

13 (32.5)

1 (2.5)

Nausea

5 (12.5)

0

Hand-foot syndrome

4 (10.0)

0

Aspartate aminotransferase increase

4 (10.0)

1 (2.5)

Anorexia

3 (7.5)

0

Alanine aminotransferase increase

2 (5.0)

0

Anemia

2 (5.0)

0

Peripheral neurotoxicity

2 (5.0)

0

Rash

1 (2.5)

0

Thrombocytopenia

1 (2.5)

1 (2.5)

Discussion

In this single-center study, pyrotinib combined with trastuzumab and chemotherapy demonstrated promising efficacy in heavily pre-treated HER2 positive MBC (92.5% of the patients with prior trastuzumab and 52.5% with lapatinib), with a median PFS of 7.5 months and an ORR of 50%, which implies that half of the patients could still respond to pyrotinib-based regimen even in later-line settings.

As is well known, efficacy and safety of pyrotinib in patients with HER2-positive MBC have been verified in several pivotal studies. In the phase I study pyrotinib monotherapy demonstrated an ORR of 83.3% in trastuzumab-naive patients and 33.3% in trastuzumab-pretreated patients [14]. The benefit of continued use of trastuzumab beyond disease progression was also confirmed in earlier studies [8, 9]. The anti-tumor activity and favorable tolerability of dual HER2 blockade with TKI plus trastuzumab has been reported previously and mechanisms of synergistic interaction may involve enhanced apoptosis of cancer cells, increased stabilization and degradation of HER2 receptors, and reversion of resistance to trastuzumab by accumulation of HER2 receptors on the surface of breast cancer cells [20, 21]. Impressively, Murthy and colleagues reported that tucatinib, which was an investigational, oral, highly selective inhibitor of the HER2 tyrosine kinase, was an alternative regimen in heavily pre-treated metastatic breast cancer when combined with trastuzumab and capecitabine, with a median PFS of 7.8 months in the study population and 7.6 months in the brain metastases subgroup [19]. Another single-arm exploratory phase II trial has demonstrated that pyrotinib plus trastuzumab and albumin-bound paclitaxel generated an encouraging pCR rate and an ORR of 100% in the neoadjuvant setting [18]. To our knowledge, there exists no published data investigating the activity and safety of pyrotinib plus trastuzumab and chemotherapy in MBC patients previously treated. Our findings are fairly consistent with and further consolidate the above-mentioned data. The PFS duration reported here is similar with that in the HER2CLIMB trial. Noteworthy, the median PFS of BrM subset that have presumably worse prognosis, was numerically superior to that of the total population. The previous therapeutic lines may predominantly account for this result, since 32.5% of the total population versus 20.0% of BrM subgroup received ≥ 2 lines of therapy previously. Besides, concerning the patients with intracranial metastasis, the PFS duration estimated here (9.4 months) was also longer than that in the HER2CLIMB trial (7.6 months). The potent activity of pyrotinib per se for intracranial metastases may partly account for this finding. Another possible explanation is that local therapy for intracranial metastases (46.7% of the patients with brain metastases received local therapy) improved blood brain barrier permeability and thus the intracranial concentration of anti-tumor drugs increased.

Up to now, evidences on head-to-head comparison of TKI plus trastuzumab versus TKI alone are lacking. A randomized phase II trial, which was designed to assess the efficacy and tolerability of pyrotinib plus capecitabine versus lapatinib plus capecitabine in women with HER2-positive MBC previously treated with taxanes, anthracyclines, and/or trastuzumab, reported that the ORR was 78.5% and the median PFS was 18.1 months in the pyrotinib arm [15]. Similarly, PHENIX study showed that the ORR and median PFS of pyrotinib plus capecitabine were 68.6% and 11.1 months, respectively [17]. Numerically, these findings were superior to the data reported in our study, which might be partly attributed to different distribution of patient characteristics. The phase II study and PHENIX study enrolled patients with no more than 2 lines of prior treatment in the metastatic setting, and nearly 70% of the patients were trastuzumab-naïve. However, 67.5% of the patients in our study received ≥ 2 lines of therapy previously, and almost all patients had received prior trastuzumab for their metastatic disease [15]. Considering that 52.5% of our patients had been exposed to prior lapatinib, our data further implies the activity of pyrotinib beyond progression on lapatinib treatment. It follows that pyrotinib combined with trastuzumab and chemotherapy may offer a promising alternative for patients with heavily treated HER2-positive MBC, including those with BrM.

In addition, we further analyzed the predictive factors of PFS and found that liver or/and lung metastases was independent adverse factor for PFS, which suggested that patients without liver or/and lung metastases might better benefit from this combination therapy. This finding could provide guidance for patient selection and optimize clinical management.

Referring to safety, the overall incidences of TRAEs were similar to that of previous report and no new TRAEs were reported. The majority of TRAEs were Grade 1 or 2 in severity and most of TRAEs could be managed by dose reduction and supportive treatment. No one discontinued study treatment or died due to TRAEs. Diarrhea, usually occurring on days 4–14 after the first dose, is the most common TRAE which is mainly induced by pyrotinib and could be managed by dose reduction and loperamide, turning mostly tolerable after treatment administered for one month. The majority of diarrhea events were Grade 1 or 2, and 25% of them were Grade 3 or 4. Prophylaxis use of loperamide could effectively reduce incidence of diarrhea caused by neratinib. However, evidence is scarce supporting the prophylaxis use of loperamide in pyrotinib treatment. Of note, no cardiac-related events were recorded in our study. The low incidence of severe adverse events demonstrated the safety of pyrotinib plus trastuzumab in heavily pretreated MBC patients.

After all, the study is an initial single-center investigation based on a small Chinese cohort. Another limitation is the relatively low rate of standard care administration in previous lines of treatment among the enrolled patients. Only 6 patients received pertuzumab and trastuzumab as first-line therapeutic regimen and none of them had received TDM-1 treatment owing to financial and drug accessibility factors. Multi-center randomized controlled trials in larger cohorts are needed to further validate the efficacy and safety of this combination regimen.

In conclusion, pyrotinib in combination with trastuzumab and chemotherapy offer an active option with a favorable safety profile in heavily pre-treated patients with HER2-positive MBC, including those with brain metastases. Multi-center randomized controlled trials are warranted to validate the results.

Declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

We are grateful to the patients and all the researchers, including the physicians, pathologists, and technicians, who participated in this study.

Funding

This work was supported by funds from ShenZhen basic research program (2018, JCYJ20180306171227129). Cai-Wen Du has received research support from Shenzhen Science and Technology Innovation Commission.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Author contributions statement

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by Xiao-Feng Xie, Qiu-Yi Zhang, Jia-Yi Huang, Li-Ping Chen, Si-Jia Guo, Shui-Ling Xiong and Xiao-Feng Lan. Writing-reviewing and editing were performed by Xie-Xiao Feng, Xue-Bai, Lin Song. The first draft of the manuscript was written by Xiao-Feng Xie and Cai-Wen Du. Supervision and project administration was performed by Cai-Wen Du. All authors are responsible for the contents and have read and approved the manuscript for submission.

Data availability

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

Ethical approval

All procedures performed involving human participants were in accordance with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. This study was also approved by institutional review boards and ethics committees (ethical code: 2019-35) of National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College.

Informed consent

Informed consent was obtained from all individual participants included in the study. The authors affirmed that human research participants provided informed consent for publication.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F (2021) Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Cancer J Clin 71:209–249. doi: 10.3322/caac.21660
  2. Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, de Azambuja E, Castro G, Untch M, Smith I, Gianni L, Baselga J, Al-Sakaff N, Lauer S, McFadden E, Leyland-Jones B, Bell R, Dowsett M, Jackisch C (2017) 11 years' follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. The Lancet 389:1195–1205. doi: 10.1016/s0140-6736(16)32616-2
  3. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL (1987) Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177–182
  4. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: breast cancer. Version 1.2022. November 24 (2021) https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf
  5. Pernas S, Tolaney SM (2019) HER2-positive breast cancer: new therapeutic frontiers and overcoming resistance. Therapeutic Adv Med Oncol 11:175883591983351. doi: 10.1177/1758835919833519
  6. Brosnan EM, Anders CK (2018) Understanding patterns of brain metastasis in breast cancer and designing rational therapeutic strategies. Annals of translational medicine 6:163. doi: 10.21037/atm.2018.04.35
  7. Sammons S, Van Swearingen AED, Chung C, Anders CK (2021) Advances in the management of breast cancer brain metastases. Neuro-oncology Adv 3:v63–v74. doi: 10.1093/noajnl/vdab119
  8. Extra JM, Antoine EC, Vincent-Salomon A, Delozier T, Kerbrat P, Bethune-Volters A, Guastalla JP, Spielmann M, Mauriac L, Misset JL, Serin D, Campone M, Hebert C, Remblier C, Bergougnoux L, Campana F, Namer M (2010) Efficacy of trastuzumab in routine clinical practice and after progression for metastatic breast cancer patients: the observational Hermine study. Oncologist 15:799–809. doi: 10.1634/theoncologist.2009-0029
  9. von Minckwitz G, Schwedler K, Schmidt M, Barinoff J, Mundhenke C, Cufer T, Maartense E, de Jongh FE, Baumann KH, Bischoff J, Harbeck N, L¨¹ck HJ, Maass N, Zielinski C, Andersson M, Stein RC, Nekljudova V, Loibl S (2011) Trastuzumab beyond progression: overall survival analysis of the GBG 26/BIG 3–05 phase III study in H ER2-positive breast cancer. Eur J Cancer 47:2273–2281 Gbg BIGsg, participating i
  10. Mukherjee A, Dhadda AS, Shehata M, Chan S (2007) Lapatinib: a tyrosine kinase inhibitor with a clinical role in breast cancer. Expert Opin Pharmacother 8:2189–2204
  11. Gomez HL, Doval DC, Chavez MA, Ang PC, Aziz Z, Nag S, Ng C, Franco SX, Chow LW, Arbushites MC, Casey MA, Berger MS, Stein SH, Sledge GW (2008) Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer. J clin oncol 26:2999–3005
  12. Rabindran SK, Discafani CM, Rosfjord EC, Baxter M, Floyd MB, Golas J, Hallett WA, Johnson BD, Nilakantan R, Overbeek E, Reich MF, Shen R, Shi X, Tsou HR, Wang YF, Wissner A (2004) Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer res 64:3958–3965
  13. Moulder SL, Borges VF, Baetz T, McSpadden T, Fernetich G, Murthy RK, Chavira R, Guthrie K, Barrett E, Chia SK (2017) Phase I Study of ONT-380, a HER2 Inhibitor, in Patients with HER2+-Advanced Solid Tumors, with an Expansion Cohort in HER2 + Metastatic Breast Cancer (MBC). Clin Cancer Res 23:3529–3536
  14. Ma F, Li Q, Chen S, Zhu W, Fan Y, Wang J, Luo Y, Xing P, Lan B, Li M, Yi Z, Cai R, Yuan P, Zhang P, Li Q, Xu B (2017) Phase I Study and Biomarker Analysis of Pyrotinib, a Novel Irreversible Pan-ErbB Receptor Tyrosine Ki nase Inhibitor, in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer. J Clin Oncol 35:3105–3112
  15. Ma F, Ouyang Q, Li W, Jiang Z, Tong Z, Liu Y, Li H, Yu S, Feng J, Wang S, Hu X, Zou J, Zhu X, Xu B (2019) Pyrotinib or Lapatinib Combined With Capecitabine in HER2-Positive Metastatic Breast Cancer With Prio r Taxanes, Anthracyclines, and/or Trastuzumab: A Randomized, Phase II Study. J Clin Oncol 37:2610–2619
  16. Xu B, Yan M, Ma F, Hu X, Feng J, Ouyang Q, Tong Z, Li H, Zhang Q, Sun T, Wang X, Yin Y, Cheng Y, Li W, Gu Y, Chen Q, Liu J, Cheng J, Geng C, Qin S, Wang S, Lu J, Shen K, Liu Q, Wang X, Wang H, Luo T, Yang J, Wu Y, Yu Z, Zhu X, Chen C, Zou J, Investigators P (2021) Pyrotinib plus capecitabine versus lapatinib plus capecitabine for the treatment of HER2-positive met astatic breast cancer (PHOEBE): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol 22:351–360
  17. Yan M, Bian L, Hu X, Zhang Q, Ouyang Q, Feng J, Yin Y, Sun T, Tong Z, Wang X, Yao H, Zou J, Zhu X, Jiang Z(2020) Pyrotinib plus capecitabine for human epidermal factor receptor 2-positive metastatic breast cancer after trastuzumab and taxanes (PHENIX): a randomized, double-blind, placebo-controlled phase 3 study.Translational Breast Cancer Research1
  18. Zhong X, He P, Chen J, Yan X, Wei B, Zhang Z, Bu H, Li J, Tian T, Lv Q, Wang X, Li H, Wang J, Huang J, Suo J, Liu X, Zheng H, Luo T (2022) Neoadjuvant pyrotinib plus trastuzumab and nab-paclitaxel for HER2-positive early or locally advanced breast cancer: an exploratory phase II trial. Gland Surg 11:216–225
  19. Murthy RK, Loi S, Okines A, Paplomata E, Hamilton E, Hurvitz SA, Lin NU, Borges V, Abramson V, Anders C, Bedard PL, Oliveira M, Jakobsen E, Bachelot T, Shachar SS, Muller V, Braga S, Duhoux FP, Greil R, Cameron D, Carey LA, Curigliano G, Gelmon K, Hortobagyi G, Krop I, Loibl S, Pegram M, Slamon D, Palanca-Wessels MC, Walker L, Feng W, Winer EP (2020) Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N Engl J Med 382:597–609. doi: 10.1056/NEJMoa1914609
  20. Blackwell KL, Burstein HJ, Storniolo AM, Rugo H, Sledge G, Koehler M, Ellis C, Casey M, Vukelja S, Bischoff J, Baselga J, O'Shaughnessy J (2010) Randomized study of Lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol 28:1124–1130
  21. Blackwell KL, Zaman K, Qin S, Tkaczuk KHR, Campone M, Hunt D, Bryce R, Goldstein LJ, Group S (2019) Neratinib in Combination With Trastuzumab for the Treatment of Patients With Advanced HER2-positive B reast Cancer: A Phase I/II Study. Clin Breast Cancer 19:97–104e104
  22. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45:228–247. doi: 10.1016/j.ejca.2008.10.026
  23. Statements & Declarations