
Anti‑HER2‑targeted therapies for the treatment of advanced HER2‑positive breast cancer with brain metastases (Review)
- Authors:
- Published online on: March 26, 2025 https://doi.org/10.3892/mco.2025.2840
- Article Number: 45
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].
Abstract
1. Introduction
In adult patients with cancer, brain metastases (BMs) most commonly originate from the lungs, breast, skin (melanoma) and gastrointestinal tract (1). Breast cancer (BC) is the second most common primary malignancy that is prone to BMs, which occurs in ~17% patients (2). In addition, the probability of BM is greater in human epidermal growth factor receptor 2 (HER2)-positive BC compared with that in other BC subtypes, ranging 30-55% (3). Breast cancer with brain metastases (BCBM) typically occurs at advanced stages (4,5), where the median survival time is <6 months (4,6-8).
Currently, the main treatment for BCBM (9) is local treatment, including surgery and radiotherapy. For single and large metastases, surgery can quickly relieve symptoms. However, the risk of developing leptomeningeal seeding from surgery is high, requiring comprehensive assessment of the patient and tumor status (10). The types of radiotherapy that can be applied for BCBM can be categorized into whole-brain radiotherapy (WBRT) or stereotactic radiotherapy (SRS). WBRT attempts to treat multiple metastases, but may cause cognitive impairments (11). By contrast, SRS is more precisely positioned and can cause significant oncolytic effects on small metastases with less damage, but it is inferior when targeting large metastases or multiple metastases (12). Reported overall survival (OS) in patients with breast cancer after treatment with either WBRT alone or SRS was 4-8 and 13-16 months previously found to be (3,13), respectively. In the retrospective study (14), which analyzed the data of 873 BCBM patients from 1999 to 2012, the median overall survival (OS) after diagnosis of BM was 9.1 months. By contrast, the OS of patients receiving WBRT was prolonged compared with those who had not received any treatment (95% CI, 0.52-0.88; P=0.004). Comparison in OS between SRS alone and surgery or SRS followed by WBRT (S/SRS + WBRT) yielded no difference (median OS, 14.9 vs. 17.2 months) (14).
In addition, with improvements in systemic therapy efficacy and the rapid development of imaging techniques, the incidence of BCBM has increased in recent years (15,16). Systemic therapy has led to significant survival benefit in HER2-positive BCBMs, but due to the blood-brain barrier (BBB), which partially excludes macromolecules from the CNS compartment, it serves the main role in the control of the extracranial disease. In addition, the use of imaging techniques contributed to improvements in detection rate of BM (17). The present review aims to discuss the current anti-HER2 targeted systemic therapies for patients with HER2-positive BCBMs on the basis of the latest literature (Table I).
2. Monoclonal antibodies (mAbs)
mAbs can bind not only to extracellular receptors but also to its epitope (18). They can block growth factor receptors, effectively preventing cell proliferation and indirectly recruiting cytotoxic cells, such as monocytes and macrophages, to mediate antibody-dependent cell-mediated cytotoxicity (ADCC) (19). Monoclonal antibodies can also bind to complement, leading to a type of toxicity known as complement-dependent cytotoxicity (CDC).
Trastuzumab (Herceptin)
Trastuzumab, a humanized mAb, was the first drug approved for the treatment of HER2-positive metastatic BC (20). Trastuzumab mediates ADCC, inhibits the PI3K/AKT signaling pathway, blocks the G1 phase of the cell cycle and inhibits DNA damage repair to serve an antitumor role (21,22). It is considered that the BBB allows substances of only small molecular weight (400-500 Da) (23) to pass through. Because of its large molecular mass (148,781 Da), trastuzumab is considered to be unable to cross the intact BBB. However, intracranial trastuzumab uptake and reduced BM have been observed in animal models of HER2-positive BCBM (24). The addition of trastuzumab to first-line treatment for HER2-positive metastatic disease has been documented to significantly increase the time to central nervous system (CNS) metastasis (25). The possible causes include disruption of the BBB during BM or after radiotherapy, in addition to increased vascular permeability due to neovascularization and the secretion of vascular endothelial growth factor (VEGF) (26). With tumor cell proliferation and new blood vessel formation, the blood-tumor barrier (BTB) forms between the tumor and blood vessels, where BTB permeability is significantly increased compared with the BBB. Therefore, some drugs that have difficulty passing through the BBB can enter brain metastases through intercellular bypass (27).
The registHER trial (28) previously demonstrated that patients with BM (n=258) who received trastuzumab after being diagnosed with CNS metastases had significantly longer OS compared with those who did not [n=119; 17.5 vs. 3.7 months, hazard ratio (HR)=0.25]. In addition, trastuzumab treatment (HR=0.33; 95% CI, 0.25-0.46; P=0.001) was found to be independently associated with a reduced risk of mortality after CNS metastasis. However, because of the low intracranial concentration of trastuzumab, these OS benefits were proposed to be due mainly to the long-term control of the extracranial disease (20).
At present, attempts have been made of using various biological techniques to increase drug permeability with the purpose of increasing the intracranial concentration of trastuzumab, such as combining MRI-guided focused ultrasound (MRgFUS) (29) with trastuzumab. MRgFUS was found to enhance drug uptake in 87±17% of sonicated voxels (>20% increase in standardized uptake value ratio), with ≤450% voxel-wise increase detected. In addition, nanoparticles can be used as carriers for macromolecular drugs to cross the BBB (30). However, the efficacy of nanoparticles remains in the experimental stage and there is no specific data.
Pertuzumab (Perjeta)
Pertuzumab is a recombinant humanized mAb that can binds to subdomain II of the extracellular domain of HER2, blocking the dimerization of HER2 with HER3 and HER2 with HER1, which inhibits the downstream PI3K and MAPK pathways (31). It also exhibits a complementary mechanism with trastuzumab, resulting in dual obstruction when used in combination (32). According to the CLEOPATRA trial results (33), the time to CNS progression was found to be delayed with pertuzumab (15 vs. 11.9 months; P=0.0049) (34), which may be due to the superior control of extracranial disease with pertuzumab. In a retrospective study (35), the combination of pertuzumab and trastuzumab significantly improved the OS to 44 months, compared with that after other-HER2-targeted therapy (17 months) and no-HER2-targeted therapy (3 months) in patients with HER2-positive BCBM. In addition, the PATRICIA study (36) showed that pertuzumab combined with high-dose trastuzumab (6 mg/kg weekly) achieved a CNS objective response rate (ORR) of only 11% (95% CI, 3.0-25.4, median duration of response, 4.6 months). However, a large proportion of patients achieved clinical benefit at 4 (68%) and 6 months (51%). Notably, two patients had stable intracranial and extracranial disease for >2 years. In the final efficacy analysis based on the latest follow-up, the median CNS-progression-free survival (PFS) was found to be 4.6 months, whereas the systemic PFS was also 4.6 months and the median OS was 27.2 months (37). These results suggest that it is possible to further optimize the dose and regimen of mAbs to combat BCBM.
Margetuximab (Margenza)
Margetuximab (MGAH22; margetuximab-cmkb) (38) is a human/mouse chimeric and Fc-optimized mAb against HER2. Margetuximab and trastuzumab share the same HER2 receptor-binding epitope. However, margetuximab exhibits increased binding capacity to CD16A (FcγRIIIA) and decreased binding capacity to CD32B (FcγRIIB), thereby enhancing the activation of innate and adaptive immune responses whilst maintaining the antiproliferative effect of trastuzumab. In the SOPHIA (NCT02492711) (39) clinical trial, a total of 71 patients (71/536) had BM at baseline. Margetuximab improved the primary PFS over trastuzumab [HR, 0.76; 95% CI, 0.59-0.98; P =0.03; median, 5.8 (95% CI, 5.5-7.0) months vs. 4.9 (95% CI, 4.2-5.6) months], whilst demonstrating an acceptable safety profile, making it an alternative option for late-line therapy. However, the overall OS analysis did not reveal the superiority of margetuximab over trastuzumab, where its intracranial response rate requires further exploration.
3. Tyrosine kinase inhibitors (TKIs)
Compared with those of mAbs, the physical characteristics of small-molecule TKIs allow them to serve an important role in crossing the BBB, thereby increasing drug concentrations in the brain (40). These findings suggest that TKIs may be a reasonable treatment strategy for CNS metastases (41,42). TKIs form a series of oral small-molecule drugs that promote apoptosis, inhibit the proliferation of cancer cells and act inside the cells (40). Because of its homologous structure, adenosine triphosphate (ATP) competitively binds to the intracellular ATP-binding domain of the epidermal growth factor receptor (EGFR) family, thereby inhibiting tyrosine kinase phosphorylation and subsequently blocking downstream signaling (43).
Lapatinib (Tykerb)
Lapatinib is a first-generation TKI for the treatment of HER2-positive BC and is a tyrosine kinase inhibitor of both HER1 and HER2. In 2013, a single-arm phase II multicenter study (LANDSCAPE) (44) evaluated the efficacy of lapatinib plus capecitabine in patients with HER2-positive BC who had not previously received WBRT. The CNS-ORR was calculated to be 57.1%, where the median PFS was 5.5 months. BM is reduced by >80% in 67% of patients. In addition, in the NRG Oncology-KROG/RTOG 1119(45) randomized trial, 143 patients were allocated to the following two groups: WBRT (37.5 Gy/3 weeks)/SRS (dose on the basis of lesion size) with or without lapatinib (1,000 mg per day for 6 weeks). The results reported that radiotherapy with lapatinib had a greater ORR (55 vs. 42%) at 4 weeks, suggesting that the combination may provide a short-term benefit. Therefore, the efficacy of lapatinib in the treatment of BM needs further clinical trial data for verification.
Pyrotinib (Irene)
Pyrotinib is a small-molecule irreversible TKI that can target HER1, HER2 and HER4(46). It was approved in China in 2018 in combination with capecitabine for the treatment of patients with advanced or metastatic BC (46). The PERMEATE trial (47) was designed to examine the efficacy of pyrotinib plus capecitabine in two separate cohorts of patients with HER2-positive BCBM. In cohort A (patients who had not previously received local radiotherapy), the intracranial ORR was 74.6% (median follow-up duration was 15.7 months). In cohort B (patients with active BM who progressed after radiotherapy), the intracranial ORR was 42.1% (median follow-up duration was 15.7 months). The latest follow-up data (48) (median follow-up, 40.5 months) revealed that cohort A had a median PFS of 10.7 months (95% CI, 7.6-14.9) and a median OS of 35.9 months (95% CI, 25.1-not reached). This previous study confirms the efficacy of the pyrotinib-capecitabine regimen for the treatment of HER2-positive BM.
In addition, the combination of pyrotinib, capecitabine and radiotherapy have also been assessed in another study in patients with HER2-positive BC and BM. According to BROPTIMA (49), the 1-year CNS-PFS rate was 74.9% (95% CI, 61.9-90.7), the median CNS-PFS was 18.0 months (95% CI, 15.5-not reached) and the CNS-ORR was 85%, with 17 patients (42.5%) achieving complete response (CR) and partial response (PR). In addition to effectively controlling intracranial lesions, pyrotinib effectively reversed extracranial lesions, with an overall median PFS of 17.6 months (95% CI, 12.8-34.1). The safety of this combination was acceptable and it did not significantly impair central nervous system function.
Neratinib (Nerlynx)
Neratinib is an oral irreversible TKI of HER1, HER2 and HER4(50). The NEfERT-T study (51) compared neratinib with trastuzumab for the treatment of previously untreated HER2-positive metastatic BC (MBC), where the rates of symptomatic and progressive CNS recurrence were 8.3 and 17.3%, respectively (P=0.002). Although neratinib plus paclitaxel was not found to be superior to trastuzumab plus paclitaxel in terms of PFS for patients with HER2-positive MBC as a first-line treatment, it may delay the occurrence and decrease the incidence of CNS metastasis. In the randomized controlled phase III NALA trial (52), neratinib plus capecitabine was demonstrated to be superior to lapatinib plus capecitabine in terms of 1-year PFS (29 vs. 15%) for patients who received HER2-targeted therapies with two or more lines. In addition, neratinib provided superior control of CNS metastases (CNS-ORR, 26.3 vs. 15.4%) and can delay the occurrence of symptomatic BM requiring intervention (cumulative incidence, 22.8 vs. 29.2%). In the TBCRC022 Co3 phase II trial (53) investigating prior CNS-directed therapy and neratinib plus capecitabine in patients with BCBM, cohort A (who did not receive prior lapatinib) had a higher CNS-ORR rate compared with that in cohort B (who received prior lapatinib), with the CNS-ORR reported to be 49 vs. 33%. The median PFS was also different between the two cohorts (cohort A, 5.5 months; cohort B, 3.1 months). These findings suggest that neratinib exhibits a definite control effect on HER2-positive BM in patients treated with lapatinib.
Tucatinib (Tukysa)
Tucatinib is an oral and reversible TKI that is highly selective for the HER2 domain (54). According to the HER2CLIMB study (55), the risk of intracranial progression was reduced by 68% (HR=0.32; 95% CI, 0.22-0.48; P<0.0001) in patients with HER2-positive BCBM. The median OS of the tucatinib group was 6.1 months longer compared with that of the control group (18.1 vs. 12.0 months). The CNS PFS was 9.9 months (95% CI, 8.4-11.7) in the tucatinib group, which was also superior to that observed in the control group (4.2 months; 95% CI, 3.6-5.7). According to follow-up (56) data (median follow-up of 26.9 months), the tucatinib combination group had greater clinical benefits in terms of CNS PFS and intracranial ORR. This subgroup analysis further supported the importance of tucatinib combination therapy for patients with HER2-positive brain metastases.
This trial also included a specialized subgroup of patients with untreated BM who used tucatinib plus trastuzumab and capecitabine in place of radiation therapy (66 patients). The median CNS-PFS in the tucatinib group was 8.1 months. Although the sample size was small, this analysis revealed that the combination of tucatinib with trastuzumab and capecitabine may provide an OS benefit for patients with untreated BM, possibly even delaying the duration of radiation therapy, which may cause cognitive impairments (11).
4. Antibody-drug conjugates (ADCs)
ADCs consist of mAbs, small-molecule drugs and linkers. The antibody binds to the target antigen on the surface of tumor cells. Through endocytosis and internalization, ADCs are transferred to lysosomes, where they release cytotoxic components and eventually lead to cell apoptosis (57).
Trastuzumab emtansine (T-DM1; Kadcyla)
T-DM1(58), which consists of trastuzumab and a cytotoxic component (the anti-microtubule drug DM1), exerts antitumor effects by inhibiting HER2 downstream signaling pathways, promoting ADCC, disrupting the microtubule network and causing apoptosis. Patients enrolled into the EMILIA study (59) were randomly allocated to the T-DM1 or lapatinib plus capecitabine groups. In addition, 45 and 50 patients had BM at baseline, respectively. In this subgroup, according to the RECIST 1.1 criteria (60), the median OS in the T-DM1 group was found to be 26.8 months, which was significantly superior compared with that in the lapatinib plus capecitabine group (12.9 months, HR=0.382; 95% CI, 0.18-0.80; P=0.0081).
The TH3RESA study (61) included patients with HER2-positive advanced BC who previously used trastuzumab and lapatinib. The subgroup analysis (RECIST V1.0) (62) revealed that T-DM1 treatment for patients with stable BM had a median OS of 17.3 months, which was superior compared with that of the investigator-selected treatment group (12.6 months, HR=0.62; 95% CI, 0.34-1.13), which was consistent with the beneficial trend noted in the overall population.
In another large-scale study, KAMILLA (63), among the 2,002 patients enrolled, 398 patients had BM at baseline. The median PFS was 5.5 months, whereas the median OS was 18.9 months (95% CI, 17.1-21.3). Among the 126 patients with measurable BM, the best overall response rate was 21.4%. Furthermore, 27 patients achieved stable disease (SD) for >6 months (assessed by RECIST V1.1). In total, 42.9% (54/126) patients had a >30% reduction in the sum of the maximum diameters of the target lesions. The clinical benefit rate was calculated to be as high as 42.9%. Therefore, notable antitumor activity was observed with T-DM1.
T-DXd (Enhertu)
T-DXd (64) is comprised of a humanized mAb against HER2 linked to a DNA topoisomerase I inhibitor (Dxd) through a cleavable tetrapeptide linker. The amino acid sequence of the antibody partially overlaps with that of trastuzumab, but DXd is 10-fold more potent than SN-38, the active metabolite of irinotecan (65). Upon binding to HER2, T-DXd disrupts HER2 signaling and triggers ADCC (64). In addition, endocytosis-mediated cleavage of T-DXd inside cells releases DXd, which induces DNA damage and apoptosis (66).
DESTINY-Breast03(67) was the first head-to-head clinical study of ADC agents designed to compare the therapeutic effect of T-DXd with that of the second-line standard treatment T-DM1. A total of 43 and 39 patients with stable BM were enrolled into the T-DXd and T-DM1 groups, respectively. The median PFS for patients with stable BM treated with T-DXd was 15 months (95% CI, 12.5-22.2), which was significantly superior compared with that of patients with stable BM treated with T-DM1 (3 months, 95% CI=2.8-5.8). According to RECIST V1.1, the intracranial ORR of the T-DXd group was 63.9% (67), which was approximately twice as high as that of the T-DM1 group (33.4%). The intracranial CR rates of the T-DXd and T-DM groups were 27.8 and 2.8%, respectively, where that of the T-DXd group was ~10X higher compared with that of the T-DM1 group (68). One of the limitations was that the number of patients with stable BM was small, whereas patients with active BM were excluded from this trial.
The TUXEDO-1 trial (69) was initiated to fill the gap in data on the potential activity of T-Dxd in active BMs. The median PFS was 14 months, 13.3% (2/15) achieved CR, 60% (9/15) achieved PR and 20% (3/15) achieved SD. The best CNS-ORR was 73.3% (RANO-BM criteria) (70), which met the predefined primary study endpoint. The results indicated that T-Dxd not only exhibited clinically relevant activity in HER2-positive BC with active BM but also prolonged disease control despite the presence of BM. Therefore, these findings suggest that T-Dxd can be safely used to treat patients with HER2-positive BCBM to delay the initiation of local therapy. According to the latest final outcome analysis (71), the median follow-up was 26.5 months (95% CI, 23.5 months-not reached) and the median PFS was 21 months (95% CI, 13.3-not reached). The median OS was not reached (95% CI 22.2-not reached). Although TUXEDO-1 has several limitations, such as a single-center design and a small sample size, the long-term outcomes suggest that despite its large molecular size, T-DXd prolonged both intra- and extracranial disease control with acceptable tolerability whilst maintaining QoL function. The most frequent AEs were mild and moderate, such as fatigue (66.7%), nausea (46.7%) and anemia (46.6%).
Another multicenter retrospective real-world study, the ROSET-BM study (72), was performed to evaluate the efficacy of T-Dxd in patients with active BM and leptomeningeal carcinomatosis (LMC). The results from the total population revealed that the median PFS was 16.1 months, where the 1-year OS was 74.9%. Notably, among the 19 patients with LMC, the 1-year PFS and OS rates were 60.7 (95% CI, 34.5-79.1%) and 87.1% (95% CI, 57.3-96.6%), respectively. The results demonstrated sustained systemic and CNS disease control in patients with LMC.
Cohort 5 (patients with HER2-positive or HER2-low advanced or metastatic BC and untreated leptomeningeal carcinomatosis) of the DEBBRAH study (73) also showed positive activity in previously untreated HER2-positive patients with pathologically proven LMC, with no new safety concerns. Since the study remains at the initial stages, a longer follow-up will provide prospective data for this rare subgroup. In symptomatic patients, the sequence of systemic vs. local treatment may also have an impact on outcomes. The ongoing DESTINY-Breast12 study (74) (NCT04739761) will confirm the efficacy of T-DXd in patients with active BM in BCBM and will provide the greatest amount of information to date (n=250).
ADCs in developmental stages
Novel HER2-targeted ADCs, such as SYD985, ARX788 and RC48, are being explored. ARX788 (Anviti) (75) is a locus-specific ADC drug that was independently developed in China and has been applied in phase III clinical research in the field of BC (76). It introduces para-acetyl-phenylalanine (pAF) into the HER2 mAb and binds to the cytotoxic tubulin inhibitor amberstatin 269 (AS269) to form a stable oxime bond. This structure minimizes untargeted toxicity caused by the shedding of cytotoxic drugs during circulation and effectively reduces the total amount of drug required for therapy (77). Upon endocytosis into cells, ARX788 then releases pAF-AS269 into lysosomes, which induces cell death by binding to microtubules (79). This agent is currently being explored in patients with HER2-positive advanced BM (NCT05018702).
RC48 (adastuximab) (79), which is comprised of the HER2-targeted antibody disitamab loaded with the toxin monomethyl auristatin E (MMAE) and the valine-citrulline linker, has more potent antineoplastic activity compared with T-DM1. RC48-ADC showed consistent efficacy in both the HER2-positive subgroup and the HER2-low expression subgroup (NCT02881138 and NCT03052634) (80). In addition, 2.0 mg/kg Q2W had a more favorable benefit-risk ratio compared with the other doses (81). In addition, in trastuzumab- and lapatinib-resistant xenograft tumor models in nude mice (79), this novel ADC drug has also shown superior antitumor activity compared with that by T-DM1, suggesting its potential as an improved therapy for HER2-positive BC.
5. Bispecific antibodies (bsAbs)
Zanidatamab (Zani, Ziihera), also known as ZW25(82), is a novel bispecific antibody that targets the extracellular II and IV domains of HER2. Its unique design yields multiple mechanisms of action, including dual HER2 signal blockade, increased clearance of HER2 proteins from the cell surface and enhanced antibody-dependent cytotoxic effects (83). In a previous phase I trial (NCT02892123) (84), Zani was well tolerated and showed preliminary antitumor activity in patients who had previously been treated with monotherapy or chemotherapy with advanced HER2-positive BC. The response rate of patients with BC (n=20) was 33% (median duration time, 5.1 months). A more recent phase Ib/2 trial (NCT04276493) (85), in which cohort A received 30 mg/kg Zani intravenously (IV) and cohort B received 1,800 mg Zani IV, both with docetaxel 75 mg/m2 IV Q3W. Preliminary results revealed that Zani combined with docetaxel had a manageable safety profile and an ORR of 90.9% (95% CI, 75.7-98.1; median follow-up time, 15.5 months) among the 33 patients evaluated for efficacy (EE), indicating promising antitumor activity in patients with advanced HER2-positive BC.
KN026 (Anbenitamab) (86) is a bsAb that binds to two different HER2 epitopes simultaneously, strongly inhibits the proliferation of HER2-overexpressing cancer cells and kills tumor cells that have developed resistance to the combination of trastuzumab and pertuzumab. According to the KN026-CHN-001 trial (87), 63 female patients with HER2-positive advanced BC who were previously treated with anti-HER2 targeted therapy (including trastuzumab plus pertuzumab, ADCs and TKI) were treated with IV KN026, with the dose increasing according to the ‘3 + 3 rule’ (88). A total of 23.8% (n=15) of the enrolled patients achieved a PR diagnosis. Additionally, CR was achieved in 1 patient (1.6%) and SD was achieved in 28 patients (44.4%). The confirmed ORR was 25.4% (95% CI, 15.3-37.9) and the disease control rate was 69.8% (95% CI, 57.0-80.8). The median PFS was 5.6 months.
Although there are no exact trial data on the application of ZANI and KN026 in patients with advanced BM, this novel class of drugs undoubtedly warrants further study.
6. Combination therapy
The majority of patients with advanced BM are treated with multiple lines of therapy and experience several problems, such as drug resistance, which greatly increases the difficulty of treatment (89). Combination therapy is particularly important. The combination and sequential treatment of multiple classes of drugs may provide improved therapeutic benefits.
Combinations of TKIs and ADCs
Although the use of ADCs can significantly improve the survival outcome of patients, the duration of tumor remission or benefit of ADC monotherapy remains limited because of the emergence of drug resistance mechanisms (90). Therefore, combination strategies with other antitumor drugs have become an important direction for drug development. In another cohort from the TBCRC022 study (91), the efficacy of neratinib plus T-DM1 was evaluated in patients with previously untreated BM (cohort 4A) and patients without prior T-DM1 therapy who progressed after local therapy (cohort 4B). Cohort 4C included patients who had received T-DM1 after local therapy progression. The CNS-ORRs in cohorts 4A, B and C were 33.3, 29.4 and 28.6%, respectively (median follow-up time: Cohort 4A, 33 months; cohort 4B, 28 months; cohort 4C, 28 months). In another randomized, double-blind, phase III study, HER2CLIMB02(92), revealed the advantages of ADCs plus small-molecule tyrosine kinase inhibitors in HER2-positive advanced BC. The results indicated that in both patients previously treated with trastuzumab and taxane and patients with locally advanced/metastatic BC with a past history of BM, the median PFS of T-DM1 plus tucatinib was significantly longer compared with that of T-DM1 alone (9.5 vs. 7.4 months; HR=0.76). In addition, patients with BM (40% of all enrolled patients) also had a significant PFS benefit (median PFS, 7.8 vs. 5.7 months; HR=0.64), where no additional adverse events were observed in the combination group compared with T-DM1 alone. The combination of tucatinib plus T-DXd and tucatinib plus T-DM1 has been investigated in HER2CLIMB-04 (NCT04539938) (93) and CompassHER2 RD (NCT04457596) (94) patients, the data of which may provide novel opportunities for the treatment of patients with advanced BC and BM.
ADCs combined with immunotherapy
According to studies on the synergistic mechanism of HER2-targeted ADCs combined with immunotherapy, HER2-targeted therapy and immunotherapy involve complex crosstalk mechanisms. Anti-HER2 therapy promotes CD8+ T-cell infiltration into tumor tissue (95). The secretion of IFN-γ (96) by CD8+ T cells then enhances the anti-HER2-inhibiting effect on cell proliferation and upregulates programmed death-ligand 1 (PD-L1) expression. The HER2-targeted ADC SHR-A1811(97) for injection can bind to and endocytose HER2-expressing tumor cells, cleave toxins through proteases in tumor cell lysosomes and induce cell cycle arrest and apoptosis. Adebrelimab (98) is a humanized anti-PD-L1 mAb that can specifically bind to PD-L1 molecules to block the programmed cell death protein 1/PD-L1 pathway, which leads to tumor immune tolerance and reactivates the antitumor activity of the immune system to achieve tumor treatment. Currently, clinical trials of adebrelimab combined with SHR-A1811 (NCT05353361) in BC have been approved.
7. Prospects and expectations
Protein degradation technology applied to targeted killing of the HER2 pathway
A novel targeted protein degradation system (99) has been recently designed based on phage-assisted continuous evolution technology. Degrons that can achieve targeted degradation, such as PROTAC, do not affect other non-target proteins and retain native genomic expression profiles (100), thereby minimizing interference with regulatory mechanisms that are critical to the natural biological functions of numerous proteins. In recent years, on the basis of the HER2-selective inhibitor tucatinib, a HER2 degrader was developed using PROTAC technology. The CH7C4(101) compound was obtained by optimizing the linker, E3 ligase ligand and binding site. CH7C4 effectively inhibits HER2-driven cancer cell proliferation through durable HER2 degradation and strong inhibition of downstream pathways, where its antitumor activity was stronger compared with that of tucatinib both in vitro and in vivo (101). This technology provides novel ideas for the development of new drugs for the treatment of HER2-positive BC.
Chimeric antigen receptor T-cell (CAR-T cell) immunotherapy exhibits numerous possibilities
CAR-T (102) cells are generated from T cells isolated from patients' peripheral blood and engineered in vitro to express synthetic receptors that recognize tumor antigens. CAR-T cells are subsequently cultured for expansion and infused back into patients (103). In the past, this type of immunotherapy was mainly used for treating hematologic tumors. Several clinical trials in different solid tumors are underway around the world (104), including for BC (NCT04650451, NCT03740256 and NCT03696030). Experimental data (105) have demonstrated that trastuzumab-resistant tumors can be effectively eliminated by HER2-CAR-T cells, indicating that the clinical use of trastuzumab-derived HER2-specific CAR-T cells represent an option for the treatment of trastuzumab-resistant tumors. In addition, other experimental data (106) have demonstrated that HER2-CAR-T cells containing the 4-1BB costimulatory domain exhibit superior tumor targeting ability, reducing the T-cell exhaustion phenotype and enhancing the proliferative capacity compared with those containing the CD28 costimulatory domain. Local intracranial delivery of HER2-CARs has shown potent antitumor activity in orthotopic xenograft models and in regional intraventricular delivery (106,107), which is undoubtedly worthy of further research.
8. Conclusion
On the basis of the current research progress in the treatment of BCBM, the overall efficacy of single macromolecular mAbs is not satisfactory, since they mainly serve a role in the extracranial control of this disease. Small molecule TKIs have demonstrated significant clinical benefits for patients with active BM, whereas ADC drugs have shown encouraging therapeutic activity and clinical benefit in both patients with BM and LMC. In summary, tucatinib and T-DXd are currently the two most promising therapeutic drugs in the field of BCBM. The direction of the next stage of development may target improving combination therapy. The application of Trop-2 targeted ADCs in the treatment of BCBM should be explored. With the continuous influx of research data, the rational deployment of drugs for HER2-positive BCBM will prolong PFS and improve the quality of life of patients (Table II).
Acknowledgements
Not applicable.
Funding
Funding: The present review was supported by Outstanding Young Scientific and Technological Talents Fund of Sichuan Province (grant no. 2022JDJQ0059) and The National Natural Science Foundation of China (grant nos. 82003198 and 82270196).
Availability of data and materials
Not applicable.
Authors' contributions
ZYW conducted literature searches, evaluated all relevant literature and wrote the first draft of the manuscript. HMH conceived the topic area, supervised the search strategy and writing, and reviewed and revised the manuscript drafts. All the authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Johnson JD and Young B: Demographics of brain metastasis. Neurosurg Clin N Am. 7:337–344. 1996.PubMed/NCBI | |
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022.PubMed/NCBI View Article : Google Scholar | |
Pasquier D, Darlix A, Louvel G, Fraisse J, Jacot W, Brain E, Petit A, Mouret-Reynier MA, Goncalves A, Dalenc F, et al: Treatment and outcomes in patients with central nervous system metastases from breast cancer in the real-life ESME MBC cohort. Eur J Cancer. 125:22–30. 2020.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Amiri-Kordestani L, Palmieri D, Liewehr DJ and Steeg PS: CNS metastases in breast cancer: Old challenge, new frontiers. Clin Cancer Res. 19:6404–6418. 2013.PubMed/NCBI View Article : Google Scholar | |
Watase C, Shiino S, Shimoi T, Noguchi E, Kaneda T, Yamamoto Y, Yonemori K, Takayama S and Suto A: Breast cancer brain metastasis-overview of disease state, treatment options and future perspectives. Cancers (Basel). 13(1078)2021.PubMed/NCBI View Article : Google Scholar | |
Gaspar L, Scott C, Rotman M, Asbell S, Phillips T, Wasserman T, McKenna WG and Byhardt R: Recursive partitioning analysis (RPA) of prognostic factors in three radiation therapy oncology group (RTOG) brain metastases trials. Int J Radiat Oncol Biol Phys. 37:745–751. 1997.PubMed/NCBI View Article : Google Scholar | |
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI View Article : Google Scholar | |
Tsukada Y, Fouad A, Pickren JW and Lane WW: Central nervous system metastasis from breast carcinoma. Autopsy study. Cancer. 52:2349–2354. 1983.PubMed/NCBI View Article : Google Scholar | |
Raghunath A, Desai K and Ahluwalia MS: Current treatment options for breast cancer brain metastases. Curr Treat Options Oncol. 20(19)2019.PubMed/NCBI View Article : Google Scholar | |
Soffietti R, Abacioglu U, Baumert B, Combs SE, Kinhult S, Kros JM, Marosi C, Metellus P, Radbruch A, Villa Freixa SS, et al: Diagnosis and treatment of brain metastases from solid tumors: Guidelines from the European association of neuro-oncology (EANO). Neuro Oncol. 19:162–174. 2017.PubMed/NCBI View Article : Google Scholar | |
Brown PD, Asher AL, Ballman KV, Farace E, Cerhan JH, Anderson SK, Carrero XW, Barker FG, Deming RL, Burri S, et al: NCCTG N0574 (Alliance): A phase III randomized trial of whole brain radiation therapy (WBRT) in addition to radiosurgery (SRS) in patients with 1 to 3 brain metastases. J Clin Oncol. 33 (18 Suppl)(LBA4)2015. | |
Sneed PK, Mendez J, Vemer-van den Hoek JG, Seymour ZA, Ma L, Molinaro AM, Fogh SE, Nakamura JL and McDermott MW: Adverse radiation effect after stereotactic radiosurgery for brain metastases: Incidence, time course, and risk factors. J Neurosurg. 123:373–386. 2015.PubMed/NCBI View Article : Google Scholar | |
Znidaric T, Gugic J, Marinko T, Gojkovic Horvat A, Paulin Kosir MS, Golo D, Ivanetic Pantar M and Ratosa I: Breast cancer patients with brain metastases or leptomeningeal disease: 10-year results of a national cohort with validation of prognostic indexes. Breast J. 25:1117–1125. 2019.PubMed/NCBI View Article : Google Scholar | |
Gao C, Wang F, Suki D, Strom E, Li J, Sawaya R, Hsu L, Raghavendra A, Tripathy D and Ibrahim NK: Effects of systemic therapy and local therapy on outcomes of 873 breast cancer patients with metastatic breast cancer to brain: MD Anderson cancer center experience. Int J Cancer. 148:961–970. 2021.PubMed/NCBI View Article : Google Scholar | |
Ferraro E, Reiner AS, Bou Nassif R, Tosi U, Brown S, Zeller S, Dang CT, Seidman AD and Moss NS: Survival among patients with ERBB2-positive metastatic breast cancer and central nervous system disease. JAMA Netw Open. 8(e2457483)2025.PubMed/NCBI View Article : Google Scholar | |
Bartsch R, Cameron D, Ciruelos E, Criscitiello C, Curigliano G, Duhoux FP, Foukakis T, Gligorov J, Harbeck N, LeVasseur N, et al: Expert recommendations on treatment sequencing and challenging clinical scenarios in human epidermal growth factor receptor 2-positive (HER2-positive) metastatic breast cancer. Cancer Treat Rev. 132(102853)2025.PubMed/NCBI View Article : Google Scholar | |
Rostami R, Mittal S, Rostami P, Tavassoli F and Jabbari B: Brain metastasis in breast cancer: A comprehensive literature review. J Neurooncol. 127:407–414. 2016.PubMed/NCBI View Article : Google Scholar | |
Voldborg BR, Damstrup L, Spang-Thomsen M and Poulsen HS: Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol. 8:1197–1206. 1997.PubMed/NCBI View Article : Google Scholar | |
Dalle S, Thieblemont C, Thomas L and Dumontet C: Monoclonal antibodies in clinical oncology. Anticancer Agents Med Chem. 8:523–532. 2008.PubMed/NCBI View Article : Google Scholar | |
Galanti D, Inno A, La Vecchia M, Borsellino N, Incorvaia L, Russo A and Gori S: Current treatment options for HER2-positive breast cancer patients with brain metastases. Crit Rev Oncol Hematol. 161(103329)2021.PubMed/NCBI View Article : Google Scholar | |
Valabrega G, Montemurro F and Aglietta M: Trastuzumab: Mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer. Ann Oncol. 18:977–984. 2007.PubMed/NCBI View Article : Google Scholar | |
Vu T and Claret FX: Trastuzumab: Updated mechanisms of action and resistance in breast cancer. Front Oncol. 2(62)2012.PubMed/NCBI View Article : Google Scholar | |
Pardridge WM: Drug delivery to the brain. J Cereb Blood Flow Metab. 17:713–731. 1997.PubMed/NCBI View Article : Google Scholar | |
Dijkers EC, Oude Munnink TH, Kosterink JG, Brouwers AH, Jager PL, de Jong JR, van Dongen GA, Schröder CP, Lub-de Hooge MN and de Vries EG: Biodistribution of 89Zr-trastuzumab and PET imaging of HER2-positive lesions in patients with metastatic breast cancer. Clin Pharmacol Ther. 87:586–592. 2010.PubMed/NCBI View Article : Google Scholar | |
Dawood S, Broglio K, Esteva FJ, Ibrahim NK, Kau SW, Islam R, Aldape KD, Yu TK, Hortobagyi GN and Gonzalez-Angulo AM: Defining prognosis for women with breast cancer and CNS metastases by HER2 status. Ann Oncol. 19:1242–1248. 2008.PubMed/NCBI View Article : Google Scholar | |
Garcia-Alvarez A, Papakonstantinou A and Oliveira M: Brain metastases in HER2-positive breast cancer: Current and novel treatment strategies. Cancers (Basel). 13(2927)2021.PubMed/NCBI View Article : Google Scholar | |
Lockman PR, Mittapalli RK, Taskar KS, Rudraraju V, Gril B, Bohn KA, Adkins CE, Roberts A, Thorsheim HR, Gaasch JA, et al: Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clin Cancer Res. 16:5664–5678. 2010.PubMed/NCBI View Article : Google Scholar | |
Brufsky AM, Mayer M, Rugo HS, Kaufman PA, Tan-Chiu E, Tripathy D, Tudor IC, Wang LI, Brammer MG, Shing M, et al: Central nervous system metastases in patients with HER2-positive metastatic breast cancer: Incidence, treatment, and survival in patients from registHER. Clin Cancer Res. 17:4834–4843. 2011.PubMed/NCBI View Article : Google Scholar | |
Meng Y, Reilly RM, Pezo RC, Trudeau M, Sahgal A, Singnurkar A, Perry J, Myrehaug S, Pople CB, Davidson B, et al: MR-guided focused ultrasound enhances delivery of trastuzumab to Her2-positive brain metastases. Sci Transl Med. 13(eabj4011)2021.PubMed/NCBI View Article : Google Scholar | |
Tosi G, Duskey JT and Kreuter J: Nanoparticles as carriers for drug delivery of macromolecules across the blood-brain barrier. Expert Opin Drug Deliv. 17:23–32. 2020.PubMed/NCBI View Article : Google Scholar | |
McKeage K and Perry CM: Trastuzumab: A review of its use in the treatment of metastatic breast cancer overexpressing HER2. Drugs. 62:209–243. 2002.PubMed/NCBI View Article : Google Scholar | |
Nahta R, Hung MC and Esteva FJ: The HER-2-targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res. 64:2343–2346. 2004.PubMed/NCBI View Article : Google Scholar | |
Swain SM, Miles D, Kim SB, Im YH, Im SA, Semiglazov V, Ciruelos E, Schneeweiss A, Loi S, Monturus E, et al: Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): End-of-study results from a double-blind, randomised, placebo-controlled, phase 3 study. Lancet Oncol. 21:519–530. 2020.PubMed/NCBI View Article : Google Scholar | |
Swain SM, Baselga J, Miles D, Im YH, Quah C, Lee LF and Cortés J: Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: Results from the randomized phase III study CLEOPATRA. Ann Oncol. 25:1116–1121. 2014.PubMed/NCBI View Article : Google Scholar | |
Bergen ES, Binter A, Starzer AM, Heller G, Kiesel B, Tendl-Schulz K, Bago-Horvath Z, Furtner J, Leitner J, Exner R, et al: Favourable outcome of patients with breast cancer brain metastases treated with dual HER2 blockade of trastuzumab and pertuzumab. Ther Adv Med Oncol. 13(17588359211009002)2021.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Pegram M, Sahebjam S, Ibrahim N, Fung A, Cheng A, Nicholas A, Kirschbrown W and Kumthekar P: Pertuzumab plus high-dose trastuzumab in patients with progressive brain metastases and HER2-positive metastatic breast cancer: Primary analysis of a phase II study. J Clin Oncol. 39:2667–2675. 2021.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Kumthekar P, Sahebjam S, Ibrahim N, Fung A, Cheng A, Nicholas A, Sussell J and Pegram M: Pertuzumab plus high-dose trastuzumab for HER2-positive breast cancer with brain metastases: PATRICIA final efficacy data. NPJ Breast Cancer. 9(94)2023.PubMed/NCBI View Article : Google Scholar | |
Nordstrom JL, Gorlatov S, Zhang W, Yang Y, Huang L, Burke S, Li H, Ciccarone V, Zhang T, Stavenhagen J, et al: Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res. 13(R123)2011.PubMed/NCBI View Article : Google Scholar | |
Rugo HS, Im SA, Cardoso F, Cortés J, Curigliano G, Musolino A, Pegram MD, Wright GS, Saura C, Escrivá-de-Romaní S, et al: Efficacy of margetuximab vs trastuzumab in patients with pretreated ERBB2-positive advanced breast cancer: A phase 3 randomized clinical trial. JAMA Oncol. 7:573–584. 2021.PubMed/NCBI View Article : Google Scholar | |
Angeli E and Bousquet G: Brain metastasis treatment: The place of tyrosine kinase inhibitors and how to facilitate their diffusion across the blood-brain barrier. Pharmaceutics. 13(1446)2021.PubMed/NCBI View Article : Google Scholar | |
Wang W, He H, Marín-Ramos NI, Zeng S, Swenson SD, Cho HY, Fu J, Beringer PM, Neman J, Chen L, et al: Enhanced brain delivery and therapeutic activity of trastuzumab after blood-brain barrier opening by NEO100 in mouse models of brain-metastatic breast cancer. Neuro Oncol. 23:1656–1667. 2021.PubMed/NCBI View Article : Google Scholar | |
Chien AJ and Rugo HS: Tyrosine kinase inhibitors for human epidermal growth factor receptor 2-positive metastatic breast cancer: Is personalizing therapy within reach? J Clin Oncol. 35:3089–3091. 2017.PubMed/NCBI View Article : Google Scholar | |
Traxler P: Tyrosine kinases as targets in cancer therapy-successes and failures. Expert Opin Ther Targets. 7:215–234. 2003.PubMed/NCBI View Article : Google Scholar | |
Bachelot T, Romieu G, Campone M, Diéras V, Cropet C, Dalenc F, Jimenez M, Le Rhun E, Pierga JY, Gonçalves A, et al: Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): A single-group phase 2 study. Lancet Oncol. 14:64–71. 2013.PubMed/NCBI View Article : Google Scholar | |
Kim IA, Winter KA, Sperduto PW, De Los Santos JF, Peereboom DM, Ogunleye T, Boulter D, Fritz JM, Cho KH, Shin KH, et al: Concurrent lapatinib with brain radiation therapy in patients with HER2+ breast cancer with brain metastases: NRG oncology-KROG/RTOG 1119 phase 2 randomized trial. Int J Radiat Oncol Biol Phys. 118:1391–1401. 2024.PubMed/NCBI View Article : Google Scholar | |
Blair HA: Pyrotinib: First global approval. Drugs. 78:1751–1755. 2018.PubMed/NCBI View Article : Google Scholar | |
Yan M, Ouyang Q, Sun T, Niu L, Yang J, Li L, Song Y, Hao C, Chen Z, Orlandi A, et al: Pyrotinib plus capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases (PERMEATE): A multicentre, single-arm, two-cohort, phase 2 trial. Lancet Oncol. 23:353–361. 2022.PubMed/NCBI View Article : Google Scholar | |
Yan M, Ouyang Q, Sun T, Niu L, Yang J, Li L, Song Y, Hao C and Chen Z: Pyrotinib plus capecitabine for patients with HER2-positive metastatic breast cancer and brain metastases: 3-year follow-up results from the phase 2 PERMEATE trial. J Clin Oncol. 41 (16 Suppl)(S1048)2023.PubMed/NCBI View Article : Google Scholar | |
Yang Z, Meng J, Mei X, Mo M, Xiao Q, Han X, Zhang L, Shi W, Chen X, Ma J, et al: Brain radiotherapy with pyrotinib and capecitabine in patients with ERBB2-positive advanced breast cancer and brain metastases: A nonrandomized phase 2 trial. JAMA Oncol. 10:335–341. 2024.PubMed/NCBI View Article : Google Scholar | |
Xuhong JC, Qi XW, Zhang Y and Jiang J: Mechanism, safety and efficacy of three tyrosine kinase inhibitors lapatinib, neratinib and pyrotinib in HER2-positive breast cancer. Am J Cancer Res. 9:2103–2119. 2019.PubMed/NCBI | |
Awada A, Colomer R, Inoue K, Bondarenko I, Badwe RA, Demetriou G, Lee SC, Mehta AO, Kim SB, Bachelot T, et al: Neratinib plus paclitaxel vs trastuzumab plus paclitaxel in previously untreated metastatic ERBB2-positive breast cancer: The NEfERT-T randomized clinical trial. JAMA Oncol. 2:1557–1564. 2016.PubMed/NCBI View Article : Google Scholar | |
Hurvitz SA, Saura C, Oliveira M, Trudeau ME, Moy B, Delaloge S, Gradishar W, Kim SB, Haley B, Ryvo L, et al: Efficacy of neratinib plus capecitabine in the subgroup of patients with central nervous system involvement from the NALA trial. Oncologist. 26:e1327–e1338. 2021.PubMed/NCBI View Article : Google Scholar | |
Freedman RA, Gelman RS, Anders CK, Melisko ME, Parsons HA, Cropp AM, Silvestri K, Cotter CM, Componeschi KP, Marte JM, et al: TBCRC 022: A phase II trial of neratinib and capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J Clin Oncol. 37:1081–1089. 2019.PubMed/NCBI View Article : Google Scholar | |
Moulder SL, Borges VF, Baetz T, Mcspadden T, Fernetich G, Murthy RK, Chavira R, Guthrie K, Barrett E and Chia SK: 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. 2017.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Borges V, Anders C, Murthy RK, Paplomata E, Hamilton E, Hurvitz S, Loi S, Okines A, Abramson V, et al: Intracranial efficacy and survival with tucatinib plus trastuzumab and capecitabine for previously treated HER2-positive breast cancer with brain metastases in the HER2CLIMB trial. J Clin Oncol. 38:2610–2619. 2020.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Murthy RK, Abramson V, Anders C, Bachelot T, Bedard PL, Borges V, Cameron D, Carey LA, Chien AJ, et al: Tucatinib vs placebo, both in combination with trastuzumab and capecitabine, for previously treated ERBB2 (HER2)-positive metastatic breast cancer in patients with brain metastases: Updated exploratory analysis of the HER2CLIMB randomized clinical trial. JAMA Oncol. 9:197–205. 2023.PubMed/NCBI View Article : Google Scholar | |
Chau CH, Steeg PS and Figg WD: Antibody-drug conjugates for cancer. Lancet. 394:793–804. 2019.PubMed/NCBI View Article : Google Scholar | |
Barok M, Joensuu H and Isola J: Trastuzumab emtansine: Mechanisms of action and drug resistance. Breast Cancer Res. 16(209)2014.PubMed/NCBI View Article : Google Scholar | |
Krop IE, Lin NU, Blackwell K, Guardino E, Huober J, Lu M, Miles D, Samant M, Welslau M and Diéras V: Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: A retrospective, exploratory analysis in EMILIA. Ann Oncol. 26:113–119. 2015.PubMed/NCBI View Article : Google Scholar | |
Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al: New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer. 45:228–247. 2009.PubMed/NCBI View Article : Google Scholar | |
Krop IE, Kim SB, González-Martín A, LoRusso PM, Ferrero JM, Smitt M, Yu R, Leung AC and Wildiers H: TH3RESA study collaborators. 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. 15:689–699. 2014.PubMed/NCBI View Article : Google Scholar | |
Schwartz LH, Litière S, de Vries E, Ford R, Gwyther S, Mandrekar S, Shankar L, Bogaerts J, Chen A, Dancey J, et al: RECIST 1.1-update and clarification: From the RECIST committee. Eur J Cancer. 62:132–137. 2016.PubMed/NCBI View Article : Google Scholar | |
Montemurro F, Delaloge S, Barrios CH, Wuerstlein R, Anton A, Brain E, Hatschek T, Kelly CM, Peña-Murillo C, Yilmaz M, et al: Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: Exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial☆. Ann Oncol. 31:1350–1358. 2020.PubMed/NCBI View Article : Google Scholar | |
Ogitani Y, Hagihara K, Oitate M, Naito H and Agatsuma T: Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 107:1039–1046. 2016.PubMed/NCBI View Article : Google Scholar | |
Keam SJ: Trastuzumab deruxtecan: First approval. Drugs. 80:501–508. 2020.PubMed/NCBI View Article : Google Scholar | |
Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, Soma M, Okamoto H, Oitate M, Arakawa S, et al: DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res. 22:5097–5108. 2016.PubMed/NCBI View Article : Google Scholar | |
Hurvitz S, Kim SB, Chung WP, Im SA, Park YH, Hegg R, Kim MH, Tseng LM, Petry V, Chung CF, et al: Abstract GS3-01: Trastuzumab deruxtecan (T-DXd; DS-8201a) vs trastuzumab emtansine (T-DM1) in patients (pts) with HER2+ metastatic breast cancer (mBC): Subgroup analyses from the randomized phase 3 study DESTINY-Breast03. Cancer Res. 82 (Suppl 4):GS3–01. 2022. | |
Hurvitz SA, Hegg R, Chung WP, Im SA, Jacot W, Ganju V, Chiu JWY, Xu B, Hamilton E, Madhusudan S, et al: Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: Updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 401:105–117. 2023.PubMed/NCBI View Article : Google Scholar | |
Bartsch R, Berghoff AS, Furtner J, Marhold M, Bergen ES, Roider-Schur S, Starzer AM, Forstner H, Rottenmanner B, Dieckmann K, et al: Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: A single-arm, phase 2 trial. Nat Med. 28:1840–1847. 2022.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Lee EQ, Aoyama H, Barani IJ, Barboriak DP, Baumert BG, Bendszus M, Brown PD, Camidge DR, Chang SM, et al: Response assessment criteria for brain metastases: Proposal from the RANO group. Lancet Oncol. 16:e270–e278. 2015.PubMed/NCBI View Article : Google Scholar | |
Bartsch R, Berghoff AS, Furtner J, Marhold M, Bergen ES, Roider-Schur S, Mair MJ, Starzer AM, Forstner H, Rottenmanner B, et al: Final outcome analysis from the phase II TUXEDO-1 trial of trastuzumab-deruxtecan in HER2-positive breast cancer patients with active brain metastases. Neuro Oncol. 26:2305–2315. 2024.PubMed/NCBI View Article : Google Scholar | |
Niikura N, Yamanaka T, Nomura H, Shiraishi K, Kusama H, Yamamoto M, Matsuura K, Inoue K, Takahara S, Kita S, et al: Treatment with trastuzumab deruxtecan in patients with HER2-positive breast cancer and brain metastases and/or leptomeningeal disease (ROSET-BM). NPJ Breast Cancer. 9(82)2023.PubMed/NCBI View Article : Google Scholar | |
Pérez-García JM, Vaz Batista M, Cortez P, Ruiz-Borrego M, Cejalvo JM, de la Haba-Rodriguez J, Garrigós L, Racca F, Servitja S, Blanch S, et al: Trastuzumab deruxtecan in patients with central nervous system involvement from HER2-positive breast cancer: The DEBBRAH trial. Neuro Oncol. 25:157–166. 2023.PubMed/NCBI View Article : Google Scholar | |
Lin NU, Ciruelos EM, Jerusalem G, Müller V, Niikura N, Viale G, Oscroft E, Anand S, Walker G and Harbeck N: CLRM-14. Open-label, multinational, multicenter, phase 3B/4 study of trastuzumab deruxtecan (T-DXD) in patients with or without baseline brain metastasis (BM) with previously treated advanced/metastatic human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC): Destiny-BREAST12. Neuro-Oncol Adv. 3 (Suppl 4)(iv4)2021. | |
Humphreys RC, Kirtely J, Hewit A, Biroc S, Knudsen N, Skidmore L and Wahl A: Abstract 639: Site specific conjugation of ARX-788, an antibody drug conjugate (ADC) targeting HER2, generates a potent and stable targeted therapeutic for multiple cancers. Cancer Res. 75 (15 Suppl)(S639)2015. | |
Hu X, Zhang Q, Wang L, Zhang J, Ouyang Q, Wang X, Li W, Xie W, Tong Z, Wang S, et al: ACE-breast-02: A randomized phase III trial of ARX788 versus lapatinib plus capecitabine for HER2-positive advanced breast cancer. Signal Transduct Target Ther. 10(56)2025.PubMed/NCBI View Article : Google Scholar | |
Barok M, Le Joncour V, Martins A, Isola J, Salmikangas M, Laakkonen P and Joensuu H: ARX788, a novel anti-HER2 antibody-drug conjugate, shows anti-tumor effects in preclinical models of trastuzumab emtansine-resistant HER2-positive breast cancer and gastric cancer. Cancer Lett. 473:156–163. 2020.PubMed/NCBI View Article : Google Scholar | |
Yu J, Fang T, Yun C, Liu X and Cai X: Antibody-drug conjugates targeting the human epidermal growth factor receptor family in cancers. Front Mol Biosci. 9(847835)2022.PubMed/NCBI View Article : Google Scholar | |
Yao X, Jiang J, Wang X, Huang C, Li D, Xie K, Xu Q, Li H, Li Z, Lou L and Fang J: A novel humanized anti-HER2 antibody conjugated with MMAE exerts potent anti-tumor activity. Breast Cancer Res Treat. 153:123–133. 2015.PubMed/NCBI View Article : Google Scholar | |
Wang J, Liu Y, Zhang Q, Feng J, Fang J, Chen X, Han Y, Li Q, Zhang P, Yuan P, et al: RC48-ADC, a HER2-targeting antibody-drug conjugate, in patients with HER2-positive and HER2-low expressing advanced or metastatic breast cancer: A pooled analysis of two studies. J Clin Oncol. 39 (15 Suppl)(S1022)2021. | |
Wang J, Liu Y, Zhang Q, Li W, Feng J, Wang X, Fang J, Han Y and Xu B: Disitamab vedotin, a HER2-directed antibody-drug conjugate, in patients with HER2-overexpression and HER2-low advanced breast cancer: A phase I/Ib study. Cancer Commun (Lond). 44:833–851. 2024.PubMed/NCBI View Article : Google Scholar | |
Meric-Bernstam F, Beeram M, Mayordomo JI, Hanna DL, Ajani JA, Murphy MAB, Murthy RK, Piha-Paul SA, Bauer TM, Bendell JC, et al: Single agent activity of ZW25, a HER2-targeted bispecific antibody, in heavily pretreated HER2-expressing cancers. J Clin Oncol. 36 (15 Suppl)(S2500)2018. | |
Bartsch R and Bergen E: ASCO 2018: Highlights in HER2-positive metastatic breast cancer. Memo. 11:280–283. 2018.PubMed/NCBI View Article : Google Scholar | |
Meric-Bernstam F, Beeram M, Hamilton E, Oh DY, Hanna DL, Kang YK, Elimova E, Chaves J, Goodwin R, Lee J, et al: Zanidatamab, a novel bispecific antibody, for the treatment of locally advanced or metastatic HER2-expressing or HER2-amplified cancers: A phase 1, dose-escalation and expansion study. Lancet Oncol. 23:1558–1570. 2022.PubMed/NCBI View Article : Google Scholar | |
Wang X, Lee KS, Zeng X, Sun T, Im YH, Li H, Wang K, Li H, Zhou P, Bao Y and Jiang Z: Zanidatamab (zani), a HER2-targeted bispecific antibody, in combination with docetaxel as first-line therapy (1L) for patients (pts) with advanced HER2-positive breast cancer (BC): Updated results from a phase 1b/2 study. J Clin Oncol. 41 (16 Suppl)(S1044)2023. | |
Wei H, Cai H, Jin Y, Wang P, Zhang Q, Lin Y, Wang W, Cheng J, Zeng N, Xu T and Zhou A: Structural basis of a novel heterodimeric Fc for bispecific antibody production. Oncotarget. 8:51037–51049. 2017.PubMed/NCBI View Article : Google Scholar | |
Zhang J, Ji D, Cai L, Yao H, Yan M, Wang X, Shen W, Du Y, Pang H, Lai X, et al: First-in-human HER2-targeted bispecific antibody KN026 for the treatment of patients with HER2-positive metastatic breast cancer: Results from a phase I study. Clin Cancer Res. 28:618–628. 2022.PubMed/NCBI View Article : Google Scholar | |
Liu M, Wang SJ and Ji Y: The i3+3 design for phase I clinical trials. J Biopharm Stat. 30:294–304. 2020.PubMed/NCBI View Article : Google Scholar | |
Goyette MA, Stevens LE, DePinho CR, Seehawer M, Nishida J, Li Z, Wilde CM, Li R, Qiu X, Pyke AL, et al: Cancer-stromal cell interactions in breast cancer brain metastases induce glycocalyx-mediated resistance to HER2-targeting therapies. Proc Natl Acad Sci USA. 121(e2322688121)2024.PubMed/NCBI View Article : Google Scholar | |
Drago JZ, Modi S and Chandarlapaty S: Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol. 18:327–344. 2021.PubMed/NCBI View Article : Google Scholar | |
Freedman R, Ren S, Tayob N, Gelman R, Smith KL, Davis R, Pereslete A, Attaya V, Cotter C, Chen WY, et al: Abstract PD7-03: Translational breast cancer research consortium trial 022: Neratinib and trastuzumab-emtansine for HER2+ breast cancer brain metastases (BCBM). Cancer Res. 83 (5 Suppl):PD7–03. 2023. | |
Hurvitz S, Loi S, O'Shaughnessy J, Okines A, Tolaney S, Sohn JH, Saura C, Zhu X, Cameron D, Bachelot T, et al: Abstract GS01-10: HER2CLIMB-02: Randomized, double-blind phase 3 trial of tucatinib and trastuzumab emtansine for previously treated HER2-positive metastatic breast caner. Cancer Res. 84 (Suppl 9):GS01–10. 2024.https://doi.org/10.1158/1538-7445.SABCS23-GS01-10. | |
Carey LA, Krop I, Ramos J, Feng W and Hamilton E: 331TiP HER2CLIMB-04: Phase II trial of tucatinib + trastuzumab deruxtecan in patients with HER2+ locally advanced or metastatic breast cancer with and without brain metastases. Ann Oncol. 32 (Suppl 5):S510–S511. 2021. | |
O'Sullivan CC, Ballman KV, McCall L, Kommalapati A, Zemla T, Weiss A, Mitchell M, Blinder V, Tung NM, Irvin WJ, et al: Alliance A011801 (compassHER2 RD): Postneoadjuvant T-DM1 + tucatinib/placebo in patients with residual HER2-positive invasive breast cancer. Future Oncol. 17:4665–4676. 2021.PubMed/NCBI View Article : Google Scholar | |
Xu M, Du X, Liu M, Li S, Li X, Fu YX and Wang S: The tumor immunosuppressive microenvironment impairs the therapy of anti-HER2/neu antibody. Protein Cell. 3:441–449. 2012.PubMed/NCBI View Article : Google Scholar | |
Stagg J, Loi S, Divisekera U, Ngiow SF, Duret H, Yagita H, Teng MW and Smyth MJ: Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA. 108:7142–7147. 2011.PubMed/NCBI View Article : Google Scholar | |
Yao H, Ryu MH, Park J, Voskoboynik M, Kim JH, Liu K, Barve M, Acuna-Villaorduna A, Im SA, Roy AC, et al: 656MO The HER2-targeting ADC SHR-A1811 in HER2-expressing/mutated advanced non-breast solid tumors (STs): Results from the global phase I study. Ann Oncol. 34 (Suppl 2):S461–S462. 2023. | |
Wang J, Zhou C, Yao W, Wang Q, Min X, Chen G, Xu X, Li X, Xu F, Fang Y, et al: Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment for extensive-stage small-cell lung cancer (CAPSTONE-1): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 23:739–747. 2022.PubMed/NCBI View Article : Google Scholar | |
Mercer JAM, DeCarlo SJ, Roy Burman SS, Sreekanth V, Nelson AT, Hunkeler M, Chen PJ, Donovan KA, Kokkonda P, Tiwari PK, et al: Continuous evolution of compact protein degradation tags regulated by selective molecular glues. Science. 383(eadk4422)2024.PubMed/NCBI View Article : Google Scholar | |
Gadd MS, Testa A, Lucas X, Chan KH, Chen W, Lamont DJ, Zengerle M and Ciulli A: Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat Chem Biol. 13:514–521. 2017.PubMed/NCBI View Article : Google Scholar | |
Hu M, Li Y, Li J, Zhou H, Liu C, Liu Z, Gong Y, Ying B and Xie Y: Discovery of potent and selective HER2 PROTAC degrader based Tucatinib with improved efficacy against HER2 positive cancers. Eur J Med Chem. 244(114775)2022.PubMed/NCBI View Article : Google Scholar | |
Feins S, Kong W, Williams EF, Milone MC and Fraietta JA: An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 94 (S1):S3–S9. 2019.PubMed/NCBI View Article : Google Scholar | |
Zhang BL, Qin DY, Mo ZM, Li Y, Wei W, Wang YS, Wang W and Wei YQ: Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors. Sci China Life Sci. 59:340–348. 2016.PubMed/NCBI View Article : Google Scholar | |
Chen T, Wang M, Chen Y and Liu Y: Current challenges and therapeutic advances of CAR-T cell therapy for solid tumors. Cancer Cell Int. 24(133)2024.PubMed/NCBI View Article : Google Scholar | |
Szöőr Á, Tóth G, Zsebik B, Szabó V, Eshhar Z, Abken H and Vereb G: Trastuzumab derived HER2-specific CARs for the treatment of trastuzumab-resistant breast cancer: CAR T cells penetrate and eradicate tumors that are not accessible to antibodies. Cancer Lett. 484:1–8. 2020.PubMed/NCBI View Article : Google Scholar | |
Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, Chang WC, Ostberg JR, Neman J, Jandial R, et al: Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2+ breast cancer metastasis to the brain. Clin Cancer Res. 24:95–105. 2018.PubMed/NCBI View Article : Google Scholar | |
Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, Ostberg JR, Blanchard MS, Kilpatrick J, Simpson J, et al: Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 375:2561–2569. 2016.PubMed/NCBI View Article : Google Scholar |