Next Article in Journal
Clinical Application of the Association between Genetic Alteration and Intraoperative Fluorescence Activity of 5-Aminolevulinic Acid during the Resection of Brain Metastasis of Lung Adenocarcinoma
Next Article in Special Issue
Reducing the Risk of Needle Tract Seeding or Tumor Cell Dissemination during Needle Biopsy Procedures
Previous Article in Journal
Assessment of the Psychosocial Impact of Pancreatic Cancer Surveillance in High-Risk Individuals
Previous Article in Special Issue
Disparities in Receipt of National Comprehensive Cancer Network Guideline-Adherent Care and Outcomes among Women with Triple-Negative Breast Cancer by Race/Ethnicity, Socioeconomic Status, and Insurance Type
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel HER-2 Targeted Therapies in Breast Cancer

by
Catarina Lopes Fernandes
1,*,
Diogo J. Silva
1 and
Alexandra Mesquita
1,2
1
Medical Oncology Department, Pedro Hispano Hospital, 4464-513 Matosinhos, Portugal
2
Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(1), 87; https://doi.org/10.3390/cancers16010087
Submission received: 18 November 2023 / Revised: 13 December 2023 / Accepted: 21 December 2023 / Published: 23 December 2023
(This article belongs to the Special Issue Advances in Invasive Breast Cancer: Treatment and Prognosis)

Abstract

:

Simple Summary

HER-2-targeting drugs changed the paradigm of HER-2-positive breast cancer, improving survival rates of an aggressive and lethal disease. Recently, a plethora of new drugs have been approved in different settings, leading to a continuous change in the treatment algorithm of HER-2-positive breast cancer. This current review aims to summarize all the new approved therapeutic options and highlight the new therapeutic options under development.

Abstract

Human epidermal growth factor 2 (HER-2)-positive breast cancer represents 15–20% of all breast cancer subtypes and has an aggressive biological behavior with worse prognosis. The development of HER-2-targeted therapies has changed the disease’s course, having a direct impact on survival rates and quality of life. Drug development of HER-2-targeting therapies is a prolific field, with numerous new therapeutic strategies showing survival benefits and gaining regulatory approval in recent years. Furthermore, the acknowledgement of the survival impact of HER-2-directed therapies on HER-2-low breast cancer has contributed even more to advances in the field. The present review aims to summarize the newly approved therapeutic strategies for HER-2-positive breast cancer and review the new and exploratory HER-2-targeted therapies currently under development.

1. Introduction

Breast cancer is estimated to be the type of cancer with the highest incidence in both sexes and the second most lethal all over the world in 2023, with an estimated 300,560 new cases and 43,700 deaths in the United States [1]. The human epidermal growth factor 2 (HER-2) overexpressing subtype accounts for 15–20% of all breast cancer cases [2] and, along with triple-negative, is historically associated with more aggressive behavior and a worse overall prognosis [3,4]. This paradigm has been changing ever since HER-2-targeted therapies were introduced in clinical practice, with excellent clinical responses and increasing survival rates.
HER-2 is one of the four transmembrane tyrosine kinase receptors in the HER family. Its activation leads to the activation of intracellular cell growth pathways, which results in abnormal cell proliferation and apoptosis evasion. Its overexpression in a subset of breast and ovary cancer patients was discovered in the late 1980s [5]. The search for an effective targeted therapy was promptly initiated, leading to the development of trastuzumab in the early 1990s and its later approval to treat HER-2-overexpressing metastatic breast cancer in 1998. Treatment with this targeted monoclonal antibody has been shown to almost double response rates and time to progression in this subset of patients [6]. Hence, ever since the approval of trastuzumab, the investigation of alternative HER-2 inhibitory agents has proliferated, mainly because trastuzumab demonstrated limited efficacy and resistance mechanisms started to come to light [7,8,9].
Regarding this, the search for better treatment options for HER-2-overexpressing breast cancer continues to this day. Current treatment options under investigation include not only HER-2-targeted therapies [monoclonal antibodies, tyrosine kinase inhibitors (TKIs) and antibody-drug conjugates (ADCs)], but also combination therapies with agents from other classes, such as cycline-dependent kinase (CDK) inhibitors, immune-checkpoint inhibitors (ICIs) and cell-therapies, among others. There are currently 662 ongoing interventional studies registered in clinicaltrials.gov for HER-2-positive breast cancer [10], and the number keeps growing.
The present review aims to summarize approved and in-development therapies for HER-2-overexpressing breast cancer and provide a comprehensive perspective on future directions in the treatment of this subset of patients.

2. HER-2 Biology and Cellular Mechanisms

2.1. HER-2 Receptor and Signaling Pathway

HER-2 belongs to the family of HER tyrosine kinase transmembrane receptors, which includes HER-1/epidermal growth factor (EGFR), HER-2, HER-3 and HER-4. Whenever activated by a ligand, these receptors stabilize in the cell membrane through homo- or heterodimerization, and hence activate intracellular signaling pathways—mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K), AKT and the mammalian target of rapamycin-dependent [11]. These pathways lead to cell division and growth, essential to cancer’s proliferation. The HER family of receptors was found to play an important role in physiological breast tissue growth and differentiation. HER-2 is composed of an extracellular ligand-binding domain (subdivided into four subdomains, I-IV), a transmembrane domain and an intracellular tyrosine kinase domain. Subdomain IV is the one closest to the cell membrane, making it essential to the stabilization of the receptor, while subdomain II is the one structurally more exposed, thus playing an important role in heterodimerization with other HER receptors [12]. HER-2 is the only HER family receptor that does not have a physiological ligand, so it relies on heterodimerization with other HER receptors, particularly HER-3, to activate downstream signaling pathways. The HER-2 receptor is encoded by the HER-2 (also known as neu or c-erbB-2) oncogene and was found to be overexpressed in a large number of solid tumors, such as breast, lung, colorectal and gastric cancers [13]. Its overexpression and abnormal ligand-independent activation in cancer cells leads to uncontrolled cell proliferation, evasion of apoptosis and high metastatic potential, thus fulfilling four of the fourteen described hallmarks of cancer [14].

2.2. HER-2 Pathological Classification

According to American Society of Clinical Oncology–College of American Pathologists (ASCO-CAP) guidelines, HER-2 positivity is defined as a 3+ score in immuno-histochemistry (IHC) staining in ≥10% of tumor cells or a 2+ score with HER-2 gene amplification through in situ hybridization (ISH) [15].
Recently, the concept of HER-2 low was established, regarding sensitivity to some anti-HER-2 therapies. It is defined as a 2+ score in IHC staining with absence of HER-2 amplification by ISH or simply a 1+ score in IHC, the latter with no need to search for ISH amplification [15].

3. Overview of Currently Approved HER-2-Targeted Therapies

Since the initial approval of trastuzumab in 1998, many other HER-2-targeted agents have been developed and some of them have been incorporated into clinical practice, integrating into an increasingly complex therapeutical algorithm.

3.1. Monoclonal Antibodies

3.1.1. Trastuzumab

Status: Approved worldwide for use in adjuvant, neoadjuvant and metastatic settings.
Trastuzumab was the first-ever HER-2-targeted monoclonal antibody (mAb) to be developed. It consists of a humanized immunoglobulin (Ig) G1 mAb that inhibits HER-2 by binding to its extracellular subdomain IV. Besides HER-2 signaling pathway inhibition, leading to cell cycle arrest, trastuzumab also induces antibody-dependent cell-mediated cytotoxicity (ADCC) [16,17] and antibody-dependent cell phagocytosis (ADCP) [18], both mediated by trastuzumab Fc-portion’s interaction with Fcγ receptors in the membrane of immune effector cells, such as natural killer (NK) cells and macrophages.
Early studies demonstrated a synergistic effect between trastuzumab and chemotherapy [19], which led to the first clinical trials testing this combination, eventually resulting in trastuzumab approval in combination with chemotherapy in HER-2-overexpressing metastatic breast cancer [6] and later in early breast cancer [20] as well.
Trastuzumab is currently approved (see Table 1):
(a)
In the metastatic setting, in the first line in combination with docetaxel for 6 cycles and then maintenance with pertuzumab until disease progression, regarding its overall survival benefit when compared with trastuzumab only with docetaxel (56.5 versus 40.8 months, hazard ratio (HR) 0.68) [21], or in monotherapy when the patient has contraindication to taxanes and/or pertuzumab [16];
(b)
In the neoadjuvant setting in combination with pertuzumab and taxane for locally advanced, inflammatory or early high-risk tumors, regarding its advantage in 5-year progression-free survival (86% versus 81%, HR 0.69) and in 5-year disease-free survival (84% versus 81%), when compared with trastuzumab plus chemotherapy [22];
(c)
In the adjuvant setting in combination with a taxane for 12 weeks and then as maintenance to complete 17 cycles, which demonstrated a 3-year invasive disease-free survival of 98,7% [23] or in monotherapy for one year after neoadjuvant therapy in patients achieving pathological complete response (pCR), which showed a 5-year disease-free survival of 84% and a 5-year overall survival of 92% [20].

3.1.2. Pertuzumab

Status: Approved worldwide for use in neoadjuvant, adjuvant and metastatic settings.
Pertuzumab is also a humanized mAb but, unlike trastuzumab, it binds the HER-2 subdomain II, preventing HER-2 dimerization with other HER family receptors, specially HER-3 [9]. This precludes HER-2 subsequent mitogenic signaling and thus cell growth and tumor proliferation. Like trastuzumab, pertuzumab also induces ADCC and ADCP, which intensifies its antitumoral activity.
Phase 2 studies showed a good safety profile but limited efficacy with pertuzumab in monotherapy [24]. Pertuzumab and trastuzumab combination therapy was then tested, revealing a favorable safety profile as well as major survival benefits [21].
Pertuzumab is currently approved (see Table 1):
(a)
In the metastatic setting, in first line in combination with docetaxel for 6 cycles and maintenance with trastuzumab until disease progression [21], as mentioned above;
(b)
In the neoadjuvant setting, in combination with trastuzumab and docetaxel to treat HER-2-positive early breast cancer with ≥2 cm [22];
(c)
In the adjuvant setting, in combination with trastuzumab for 1 year after combination with taxane in node-positive or high-risk node-negative disease, in which it demonstrated a 3-year invasive disease-free survival of 92% [25].
There is an ongoing phase 2 clinical trial testing the safety of omitting chemotherapy associated with trastuzumab and pertuzumab in the neoadjuvant setting (NCT01817452).

3.1.3. Margetuximab

Status: Approved in the United States of America (USA) for use in the metastatic setting.
Margetuximab, previously called MGHA22, is a mAb targeting subdomain IV of the HER-2 receptor, the same as trastuzumab. It has similar receptor affinity, but it differs in its engineered Fc-portion, which allows higher affinity to Fcγ receptor CD16A and lower affinity to inhibitory Fcγ receptor CD32B, leading to increased CD16A-mediated ADCC [26].
When compared to trastuzumab, margetuximab revealed a similar toxicity profile and progression-free survival (PFS) benefit, with no overall survival (OS) benefit in the third line and beyond in a phase 3 study for HER-2-overexpressing metastatic breast cancer [27].
Margetuximab is currently approved (see Table 1):
(a)
In the metastatic setting, in combination with chemotherapy in third line and beyond, showing a 9-month benefit in progression-free survival, although no benefit in overall survival when compared with trastuzumab [27].
An ongoing phase 2 study is testing the replacement of trastuzumab with margetuximab in the neoadjuvant setting, in association with pertuzumab and paclitaxel (NCT04425018).
Table 1. Summary of monoclonal antibodies (mAb) currently in use.
Table 1. Summary of monoclonal antibodies (mAb) currently in use.
mAbTargetApproved UseReference
TrastuzumabHER-2 subdomain IVNeoadjuvancy (+pertuzumab +ChT)
Adjuvancy (+ChT or monotherapy)
Palliative (±pertuzumab ±ChT)
[22]
[20,23]
[21]
PertuzumabHER-2 subdomain IINeoadjuvancy/Adjuvancy (+trastuzumab ± ChT)
Palliative (+trastuzumab ± ChT) in 1L
[22,25]
[21]
MargetuximabHER-2 subdomain IVPalliative (+ChT) in ≥3L[27]
ChT: Chemotherapy; 1L: First line; 3L: Third line.

3.2. Tyrosine Kinase Inhibitors (TKIs)

3.2.1. Lapatinib

Status: Approved worldwide for use in the metastatic setting.
Lapatinib is a potent and reversible tyrosine kinase inhibitor (TKI) of HER-2 and HER-1/EGFR. It competes with adenosine triphosphate (ATP) in its binding site to the two kinases, due to their similar structure, thereby inhibiting downstream signaling and cell proliferation [28]. Lapatinib also showed efficacy against truncated p95HER-2, which is resistant to trastuzumab extracellular binding and blockade [29].
Lapatinib proved to be effective in the treatment of HER-2-overexpressing metastatic breast cancer, in combination with capecitabine or trastuzumab, after progression with first-line trastuzumab [30]. It also showed benefits in association with endocrine therapy in hormonal receptors (HR)-positive and HER-2-positive breast cancer. Studies in the adjuvant setting showed no significant benefit and increased toxicity when adding or replacing trastuzumab with lapatinib [31,32]. In the neoadjuvant setting, lapatinib showed no benefit in the rate of pathological complete response (pCR) when compared with trastuzumab [33] but it showed a significant benefit when combined with trastuzumab and chemotherapy [34,35]. However, it is not yet approved in this setting.
Besides being active against part of trastuzumab-resistant tumor cells, lapatinib crosses the blood–brain barrier due to its small molecular size and has a significant intracranial antitumoral effect [36].
Lapatinib is currently approved (see Table 2):
(a)
In the metastastic setting, in combination with capecitabine, which proved a 4-month benefit in time to progression (8.4 versus 4.4 months, HR 0.49) and/or trastuzumab [30].

3.2.2. Neratinib

Status: Approved in the USA and Europe in the adjuvant setting and in the USA in the metastatic setting.
Neratinib, also called HKI-272, has an irreversible and pan-inhibitor effect on HER kinases, acting by covalently binding to a cysteine residue in the ATP binding site, specifically, residues Cys-773 in HER-2 and Cys-805 in HER-1 [37,38]. It also leads to the dissociation and degradation of HSP90.
Neratinib did not show any benefit in the metastatic setting in HER-2-overexpressing breast cancer when compared to lapatinib and capecitabine [39,40], but it was revealed to be effective when in combination with capecitabine [41], warranting its approval in the metastatic setting beyond third line. In the early-disease setting, neratinib showed a benefit in recurrence-free survival when used for one year in extended adjuvant therapy after 2-year neoadjuvant/adjuvant trastuzumab therapy, specifically in HR-positive patients [42]. In the neoadjuvant setting, neratinib showed benefits when compared to trastuzumab [43] and when added to it [44].
Neratinib is currently approved (see Table 2):
(a)
In the metastatic setting, in combination with capecitabine, in third line and beyond, showing a 2.2-month benefit in progression free-survival (8.8 versus 6.6 months, HR 0.76) [41].
(b)
In the adjuvant setting, for 1 year, after completing 1 year of adjuvant trastuzumab, regarding its 5-year invasive disease-free survival benefit of 2.5% (90.2% versus 87.7%, HR 0.73) [42].
Three phase 2 trials are currently evaluating neratinib in the neoadjuvant setting (NCT01042379; NCT04886531; NCT05919108) and one in the adjuvant setting in patients without pCR after neoadjuvant treatment (NCT05388149).

3.2.3. Tucatinib

Status: Approved worldwide for use in the metastatic setting.
Tucatinib is a reversible HER-2-selective TKI, with very little EGFR inhibition, which guarantees a more favorable tolerability profile than other TKIs [45]. Its exact mechanism of action is not yet clarified, but a molecular study points to a strong and stable binding to the ATP pocket in HER-2 tyrosine kinase [46]. Tucatinib demonstrated greater efficacy, in combination with capecitabine and trastuzumab, in third-line treatment of HER-2-overexpressing metastatic breast cancer with or without brain metastases [47].
Tucatinib is currently approved (see Table 2):
(a)
In the metastatic setting, beyond third-line treatment, in combination with capecitabine and trastuzumab. This combination proved to increase progression-free survival by 2.7 months (7.6 versus 4.9 months, HR 0.57) and overall survival by 5.5 months (24.7 versus 19.2 months, HR 0.73) when compared with trastuzumab plus capecitabine [47].
A phase 3 trial, HER2CLIMB-05, is currently evaluating the efficacy of tucatinib associated with trastuzumab and pertuzumab in patients not previously treated with a TKI (NCT05132582). There are four other phase 2 studies evaluating combinations of tucatinib, trastuzumab and different chemotherapy agents in the palliative setting (NCT05748834; NCT05458674; NCT05583110; NCT05955170). The previously mentioned phase 2 basket trial in the neoadjuvant setting also has one arm evaluating the efficacy of neoadjuvant tucatinib combined with trastuzumab and pertuzumab in HER-2-positive breast cancer patients (NCT01042379).

3.2.4. Pyrotinib

Status: Approved in China for use in the metastatic setting.
Pyrotinib is the most recently developed and approved TKI for HER-2-overexpressing metastatic breast cancer. It is an irreversible pan-HER TKI that covalently binds to the catalytic region of the HER-2 kinase [48]. It has proved, so far, to be effective in the metastatic setting in combination with capecitabine when compared to placebo or lapatinib in Chinese patients [49,50]. Pyrotinib is not yet approved for marketing in the European Union nor in the United States of America as a result of the lack of data on efficacy, tolerability and cost-effectiveness in a broader population [51].
Pyrotinib is currently approved (see Table 2):
(a)
In the metastatic setting in combination with capecitabine (only in China), showing a significantly higher progression-free survival (18.1 versus 7.0 months, HR 0.36) and higher overall response rate (78.5% versus 57.1%) when compared with lapatinib and capecitabine [49,50].
There are multiple ongoing clinical trials involving pyrotinib, mostly held in China. There are five studies in the metastatic setting (NCT05346861, NCT05255523, NCT04605575, NCT05429294, NCT04246502), with one phase 3 study and two studies in first-line treatment. There are six ongoing studies in the adjuvant setting (NCT04254263, NCT05841381, NCT05861271, NCT04659499, NCT05880927, NCT05834764), two of them phase 3, and seven studies in the neoadjuvant setting (NCT04929548, NCT05659056, NCT06000917, NCT04917900, NCT05430347, NCT04900311, NCT04290793).
Table 2. Summary of tyrosine kinase inhibitors approved in clinical practice.
Table 2. Summary of tyrosine kinase inhibitors approved in clinical practice.
TKITargetEffectApproved UseReference
LapatinibHER-1 and 2reversiblePalliative (+capecitabine and/or trastuzumab)[30]
NeratinibHER-1, 2 and 4irreversibleAdjuvant (after 1y trastuzumab ±pertuzumab)
Palliative (+capecitabine)
[42]
[41]
TucatinibHER-2irreversiblePalliative (+capecitabine +trastuzumab)[47]
PyrotinibHER-1, 2 and 4irreversiblePalliative (+capecitabine)[49,50]

3.3. Antibody-Drug Conjugates (ADCs)

3.3.1. Trastuzumab Emtansine

Status: Approved worldwide for use in the metastatic and adjuvant settings.
Trastuzumab emtansine (T-DM1) is the first-ever approved antibody–drug conjugate for solid tumors, approved for use in HER-2-positive metastatic breast cancer since 2013 [52] and in the adjuvant setting since 2019 [53]. It is composed of the IgG1 mAb trastuzumab connected to emtansine, a cytotoxic agent that acts by microtubule inhibition, through a non-cleavable linker [54].
Studies demonstrated T-DM1’s superiority in terms of rate of response, survival and toxicity profile, when compared with the previous standard second-line treatment, lapatinib and capecitabine, leading to its early approval in this setting [52]. Later studies also showed its efficacy in the adjuvant setting [53].
T-DM1 is currently approved (see Table 3):
(a)
In the metastatic setting, as second-line treatment for HER-2-overexpressing metastatic breast cancer after trastuzumab therapy, as the EMILIA trial showed a 3.2-month increase in progression-free survival (9.6 versus 6.4 months, HR 0.65) [52] and a 4-month increase in overall survival (29.9 versus 25.9 months, HR 0.75) [55] when compared with lapatinib plus capecitabine;
(b)
In the adjuvant setting, for residual disease after neoadjuvant chemotherapy combined with trastuzumab and pertuzumab, showing an increase in invasive disease free survival (88.3% versus 77.0% at 3 years, HR 0.50) when compared with trastuzumab [53].
A recent and still unpublished phase 2 trial, ATEMPT, evaluated the efficacy of adjuvant T-DM1 in stage I disease, compared with paclitaxel and trastuzumab (NCT01853748).

3.3.2. Trastuzumab Deruxtecan

Status: Approved in the USA and Europe for use in the metastatic setting.
Trastuzumab deruxtecan (T-DXd) is composed of the mAb trastuzumab connected to deruxtecan, a topoisomerase I inhibitor that causes cell apoptosis and double-stranded DNA breaks. These two compounds are connected by a linker that is cleaved by cathepsins, leading to its selective cleavage inside tumor cells, where cathepsins are upregulated. The cleavability of the ADC also allows a bystander killer effect, a mechanism that expands the drug’s cytotoxic effect to surrounding tumor cells. It also differs from T-DM1 in its higher antibody:drug ratio of 1:8 (T-DM1 has a 1:3.5) [56].
T-DXd proved to be effective in monotherapy in the treatment of HER-2-overexpressing refractory metastatic breast cancer [57,58], even when compared with T-DM1 [59]. More recent studies demonstrated that T-DXd is also effective in the treatment of HER-2-low metastatic breast cancer [60].
T-DXd is currently approved (see Table 3):
(a)
In the metastatic setting in HER-2-positive breast cancer, in the second line or further, regarding its benefit in delaying disease progression (75.8% of patients had no disease progression at 12 months versus 34.1% with T-DM1, HR 0.28) and delaying death (94.1% were alive at 12 months compared with 85.9%, HR 0.55) [59];
(b)
In the metastatic setting in HER-2-low breast cancer, regarding its 4.7-month benefit in progression-free survival when compared with physician’s choice chemotherapy (HR 0.51) and its 6.4-month benefit in overall survival (HR 0.64) [60].
There are currently two ongoing trials (phase 2 and 3) evaluating T-DXd as a first-line treatment in metastatic HER-2-positive breast cancer and one study evaluating it as a first-line treatment in HER-2-low patients (see Table 4). A phase 3 study is testing T-DXd as an adjuvant therapy after neoadjuvant treatment with residual disease. Three other studies, one phase 3, are evaluating the role of T-DXd in the neoadjuvant setting (see Table 4).
Table 3. Summary of antibody–drug conjugates approved in clinical practice.
Table 3. Summary of antibody–drug conjugates approved in clinical practice.
ADCCytotoxicLinkerDrug-to-Antibody RatioApproved UseReference
T-DM1Emtansine (DM1)Non-cleavable3.5:1Adjuvant HER-2+ EBC
HER-2+ MBC ≥ 2L
[53]
[52]
T-DXdDeruxtecan (DXd)Cleavable8:1HER-2+ MBC ≥ 2L
HER-2-low MBC
[58,59]
[60]
EBC: Early breast cancer; MBC: Metastatic breast cancer; 2L: Second line; 3L: Third line.
Table 4. Summary of ongoing clinical trials involving HER-2-targeted agents already in use in clinical practice.
Table 4. Summary of ongoing clinical trials involving HER-2-targeted agents already in use in clinical practice.
Anti-HER-2 AgentClinical Trials (Phase)PopulationTreatment in StudyStatus
Trastuzumab----
PertuzumabNCT01817452 (II)HER-2+ EBCNeoadjuvant P+T ±ChTRecruiting
MargetuximabNCT04262804 (II)
NCT04425018 (II)
HER-2+ MBC ≥ 3L
HER-2+ EBC
Margetuximab +ChT
Neoadj margetuximab +P +ChT
Completed; unpublished
Recruiting
Lapatinib----
NeratinibNCT01042379 (II)
NCT04886531 (II)
NCT05919108 (II)
NCT05388149 (II)
HER-2+ EBC
HER-2+ EBC
HER-2m EBC
HER-2+ EBC
Neoadj neratinib
Neoadj neratinib +T +ET
Neoadj neratinib
Adjuvant neratinib + T-DM1
Recruiting
Recruiting
Not yet recruiting
Recruiting
TucatinibNCT05132582 (III)
NCT05748834 (II)
NCT05458674 (II)
NCT05583110 (II)
NCT05955170 (II)
NCT01042379 (II)
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ EBC
Tucatinib +T+P
Tucatinib +ChT
Tucatinib +T + ChT
Tucatinib +T +ChT
Tucatinib +T +ChT
Neoadj tucatinib +T+P
Recruiting
Recruiting
Recruiting
Recruiting
Not yet recruiting
Recruiting
PyrotinibNCT05346861 (III)
NCT05255523 (II)
NCT04605575 (II)
NCT05429294 (II)
NCT04246502 (II)
NCT04254263 (III)
NCT05841381 (III)
NCT05861271 (II)
NCT04659499 (II)
NCT05880927 (II)
NCT05834764 (II)
NCT04929548 (II)
NCT05659056 (II)
NCT06000917 (II)
NCT04917900 (II)
NCT05430347 (II)
NCT04900311 (II)
NCT04290793 (II/III)
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
Pyrotinib rechallenge
Pyrotinib +T in ≥2L
Pyrotinib +ChT
Pyrotinib +T +ChT in 1L
Pyrotinib +ChT in 1L
Adj pyrotinib +T
Adj pyrotinib +T +ChT
Adj pyrotinib +ChT
Adj pyrotinib +ChT
Adj pyrotinib
Adj pyrotinib
Neoadj pyrotinib +T +P
Neoadj pyrotinib +T +ChT
Neoadj pyrotinib +T +ChT
Neoadj pyrotinib +T +ChT
Neoadj pyrotinib +T +ChT
Neoadj pyrotinib +T +ChT
Neoadj pyrotinib +ChT
Recruiting
Not yet recruiting
Recruiting
Recruiting
Not yet recruiting
Recruiting
Not yet recruiting
Not yet recruiting
Not yet recruiting
Recruiting
Recruiting
Not yet recruiting
Recruiting
Recruiting
Recruiting
Not yet recruiting
Not yet recruiting
Not yet recruiting
T-DM1NCT01853748 (II)HER-2+ EBCAdj T-DM1Active, not recruiting
T-DXdNCT04784715 (III)
NCT05744375 (II)
NCT04622319 (III)
NCT05113251 (III)
NCT05900206 (II)
NCT05704829 (II)
NCT05953168 (II)
HER-2+ MBC
HER-2+ MBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2+ EBC
HER-2-low MBC
T-DXd ±P in 1L
T-DXd in 1L
Adj T-DXd
Neoadj T-DXd ±T + P +ChT
Neoadj T-DXd
Neoadj T-DXd
T-DXd in 1L
Recruiting
Recruiting
Recruiting
Recruiting
Recruiting
Not yet recruiting
Not yet recruiting
Adj: Adjuvant; Neoadj: Neoadjuvant; ChT: Chemotherapy; EBC: Early breast cancer; ET: Endocrine therapy; MBC Metastatic breast cancer; P: Pertuzumab; T: Trastuzumab; 1L: First line; 2L: Second line.

4. Novel HER-2-Targeted Therapies

Despite the great evolution in HER-2-targeted therapies and the outstanding improvement in HER-2-overexpressing breast cancer prognosis, there is still a long way to go in HER-2 breast cancer treatment. In the early setting, there is still a considerable number of relapses, and, in the metastatic setting, most cases eventually progress under HER-2-targeted therapy, due to either suboptimal tumor cell growth inhibition or mechanisms of resistance. For these reasons, there is continuous research and new drug development in this area, which is summarized below.

4.1. Novel Monoclonal Antibodies

4.1.1. MM-302

Status: Phase 1 clinical trials.
MM-302 is a liposome containing approximately 20.000 molecules of doxorubicin, and it is coated by 45 surface anti-HER-2 antibodies [61]. Preclinical studies demonstrated the efficacy of this drug in HER-2-overexpressing breast and gastric cancer and showed synergy with trastuzumab. A phase 2 clinical trial testing MM-302 in combination with trastuzumab in heavily pretreated patients showed, however, no clinical benefit, leading to the study’s early termination [62].

4.1.2. Inetetamab

Status: Phase 2 clinical trials.
Inetetamab, also called cipterbin, is a novel anti-HER-2 mAb directed against subdomain IV of HER-2, with a slightly different aminoacid sequence in the Fc portion. Preclinical studies demonstrated efficacy against HER-2-overexpressing tumors [63,64]. A recently published retrospective study reported that inetetamab combined with vinorelbine and pyrotinib was effective in heavily pretreated HER-2-positive breast cancer patients [65].
An ongoing phase 2 trial is evaluating the role of inetetamab in the neoadjuvant setting, in combination with pertuzumab and chemotherapy. Five phase 2 studies are testing the combination of inetetamab with pyrotinib and chemotherapy in the metastatic setting, in first or subsequent lines (see Table 5).

4.2. Bispecific HER-2-Targeted Antibodies

Bispecific antibodies were developed with the aim of targeting different epitopes or domains in the HER-2 receptor, optimizing its blockade, or even targeting a different receptor besides HER-2, such as HER-3. This receptor plays an essential role in HER-2/HER-3 dimerization and downstream signaling of the PI3K pathway, one of the known mechanisms of resistance to HER-2-targeted therapy.

4.2.1. Zanidatamab

Status: Phase 2 clinical trials.
Zanidatamab (ZW25) is a humanized IgG1 mAb that simultaneously targets subdomains II and IV of the HER-2 extracellular component. Each HER-2 receptor can be targeted by two molecules of this drug, which enhances HER-2 internalization and downregulation [66].
Zanidatamab has proven its efficacy in second-line treatment of HER-2-positive MBC in two phase 1 studies, whether in monotherapy [67] or in combination with chemotherapy [68]. There are currently ongoing clinical trials testing zanidatamab’s efficacy in combination with ADCs (NCT05027139), with CDK4/6 inhibitors (NCT04224272) and in monotherapy in the neoadjuvant setting (NCT05035836).

4.2.2. MBS301

Status: Phase 1 clinical trials.
MBS301 is a bispecific IgG1 antibody targeting extracellular subdomains II and IV. It is glycol-engineered from trastuzumab and pertuzumab, consisting of one-half of each of these mAbs. In preclinical studies, this drug showed greater efficacy when compared to the trastuzumab and pertuzumab association, mostly due to an increased ADCC stimulation [69].
A phase 1 clinical trial is still ongoing in China (NCT03842085) and no preliminary data are yet available.

4.2.3. Anbenitamab

Status: Phase 2 clinical trials.
Anbenitamab, also called KN026, is a bispecific antibody (bsAb) derived from trastuzumab and pertuzumab which showed efficacy in tumor cells resistant to these two drugs [70]. Phase 1 clinical trials showed a similar efficacy to the trastuzumab and pertuzumab combination even in heavily pretreated patients [71].
Preliminary data from two recently concluded phase 1 studies in HER-2-overexpressing breast and gastric cancer patients revealed promising antitumoral activity with good tolerability [71,72]. Ongoing clinical trials are evaluating KN026’s efficacy in the neoadjuvant treatment of breast cancer and in the metastatic setting in combination with chemotherapy (see Table 5).

4.2.4. Zenocutuzumab

Status: Phase 1 clinical trials.
Zenocutuzumab (MCLA-128) is another IgG1 bsAb, but instead of targeting different subdomains of HER-2, it targets subdomain I of HER-2 and the ligand-binding site of HER-3, thereby impeding HER-2-HER-3 dimerization and downstream signaling. Besides blocking the HER-2 signaling pathway, zenocutuzumab also strongly stimulates ADCC [73].
Zenocutuzumab demonstrated efficacy in combination with trastuzumab and vinorelbine in the third line and beyond in HER-2-positive MBC [74]. A recently published phase 1 study testing this drug’s efficacy in patients with NGR1 fusion demonstrated robust and durable efficacy with a good tolerability profile [75].
There is currently one ongoing phase 2 trial testing a combination with chemotherapy, trastuzumab or endocrine therapy in HER-2-positive or HER-2-low/hormonal receptor-positive breast cancer patients (NCT03321981).

4.2.5. HER2(Per)-S-Fab

Status: Preclinical studies.
HER2(Per)-S-Fab is a bispecific antibody composed of pertuzumab antigen-binding fraction (Fab), which binds the HER-2 subdomain II and an anti-CD16/FcγRIIIa antibody [76]. The anti-CD16/FcγRIIIa portion exerts an immune stimulatory effect, by recruiting natural killer (NK) cells, which express CD16, to the tumoral microenvironment (TME) and promoting its cytotoxic effects against target HER-2-positive cells [76]. There are no ongoing clinical trials with this drug.

4.2.6. HER2-2XCD16

Status: Preclinical studies.
HER2-2XCD16 is a tribody which integrates an anti-HER-2 single chain fused with IFN- γ, leading to IFN-dependent cell death, even in tumors resistant to HER-2-blockage [77]. Besides an antitumoral effect in HER-2-overexpressing breast cancer cells, this molecule demonstrated an ability to interfere with the TME, switching it to an antitumoral environment [78].

4.2.7. Discontinued Bispecific Antibodies

There are three other bispecific antibodies that showed an antitumoral effect in HER-2-positive breast cancer but were, meanwhile, discontinued, by decision of the sponsor.
MM-111 targets both the extracellular component of HER-2 and the heregulin-binding site of HER-3, inhibiting both HER-2 and PI3K pathways [79]. Preclinical studies showed efficacy against HER-2-overexpressing breast cancer cells, and when combined with trastuzumab, it demonstrated more efficacy than trastuzumab with pertuzumab [79].
Ertumaxomab targets HER-2 and CD3 and has a trimodal action through the recognition of not only tumor cells, but also stromal cells and T-cells [80,81]. Preclinical studies demonstrated a moderate antitumoral effect in both HER-2-overexpressing and HER-2-low breast cancer, with inferior efficacy when compared with standard of care [81]. Phase 2 trials of ertumaxomab were early-terminated.
ISB1302 or GBR1302 also targets CD3 in T-cells and directs them to HER-2-overexpressing tumor cells, where they bind HER-2. Phase 1 studies confirmed activation of a T-cell-mediated immune response [82], but no further studies were conducted.

4.3. Novel Tyrosine Kinase Inhibitors (TKIs)

4.3.1. Poziotinib

Status: Phase 2 clinical trials.
Poziotinib is an irreversible pan-HER TKI that inhibits EGFR, HER-2 and HER-4. Preclinical studies revealed an antitumoral effect in HER-2-overexpressing or HER-2-mutant breast, lung and gastric cancer lines [83], and demonstrated that poziotinib upregulates HER-2 expression in the cell membrane and potentiates the antitumoral effect of other anti-HER-2 therapies, such as T-DM1 [84]. Phase 1 studies confirmed the efficacy of poziotinib in the treatment of HER-2-positive tumors [85]. A phase 2 trial tested poziotinib’s efficacy in the treatment of HER-2-overexpressing metastatic breast cancer patients in the third line and beyond, showing meaningful antitumoral efficacy with overall survival benefits and a toxicity profile similar to other TKIs [86,87]. A phase 1b (NCT03429101) and other phase 2 studies were recently conducted in metastatic breast cancer patients, in monotherapy or association with T-DM1, both in HER-2-positive (NCT02659514; NCT02418689) and HER-2-mutant patients (NCT02544997), and preliminary data is still awaited (see Table 6). There are no clinical trials currently ongoing.

4.3.2. DZD1516

Status: Phase 1 clinical trials.
DZD1516 is a reversible HER-2-specific TKI with full blood–brain barrier penetration [88]. In preclinical studies, it revealed an antitumoral effect in HER-2-positive tumors [88]. A phase 1 study from the same author revealed good disease control in heavily pretreated HER-2-positive breast cancer patients with brain metastases [88]. Another phase 1 study is currently ongoing (NCT04509596) and preliminary data confirms good intracranial disease control with a favorable toxicity profile [89].

4.3.3. Discontinued TKIs

Epertinib, also called S-222611, is a reversible pan-HER TKI. In preclinical studies, it revealed a superior antitumoral effect in HER-2-overexpressing tumors when compared with lapatinib and a more favorable toxicity profile when compared with irreversible TKIs [90]. Phase 1 clinical trials confirmed an antitumoral effect and good tolerability in HER-2-overexpressing breast, upper digestive, head and neck and renal tumors in monotherapy [91], and in combination with trastuzumab with or without capecitabine [92]. Despite these encouraging results, no further trials were performed.
BDTX-189 is an irreversible EGFR/HER-2 TKI. Preliminary data from a recently concluded phase 1 trial, MasterKey-01, revealed an antitumoral effect in patients with HER-2/HER-3-mutated and HER-2-amplified solid tumors [93]. Despite the promising results, the drug’s sponsor decided to discontinue further research.

4.4. Novel Antibody–Drug Conjugates (ADCs)

Despite the great efficacy of currently commercialized ADCs for HER-2-overexpressing breast cancer, mechanisms of resistance to T-DM1 and T-DXd have already been identified, and the development of further ADCs, with a greater therapeutic index and less toxicity, is needed, justifying continued research in this area.

4.4.1. Trastuzumab Duocarmycin (SYD985)

Status: Phase 3 clinical trials.
Trastuzumab duocarmycin is an IgG1 ADC composed of trastuzumab seco-duocarmycin-hydroxybenzamide-azaindole (seco-DUBA), a prodrug of DUBA, a DNA alkylator cytotoxic drug, in a drug-to-antibody ratio of 2.8:1, lower than both T-DM1 and T-DXd. After binding to HER-2 and being internalized, the cleavable linker binding both components of the ADC is cleaved in the lysosome, and the active drug DUBA is released in the cytoplasm [94]. SYD985 has considerable bystander killing effect for two reasons—DUBA itself is membrane-permeable after being released from the lysosome, and SYD985 cleaving proteases, like cathepsin B, are usually produced by tumor cells, meaning they are present in the extracellular milieu, thereby maximizing this ADC’s effect in surrounding tumor cells [94].
Preclinical studies demonstrated SYD985’s efficacy in HER-2-overexpressing breast cancer cells, with similar HER-2 binding, HER2-mediated internalization and ADCC stimulation when compared with T-DM1 [95]. Phase 1 studies confirmed efficacy not only in HER-2-overexpressing but also in HER-2-low metastatic breast cancer, with a greater efficacy in the HER-2-low subset of patients when compared with T-DM1, probably due its potent bystander killing effect [94]. The results of the phase 3 clinical trial TULIP are still to be published but preliminary data reveal a significant improvement in progression-free survival with no significant increase in overall response rate or overall survival, when compared with physician’s choice in third line and beyond HER-2-overexpressing metastatic breast cancer [96].
SYD985 is currently being tested along with other targeted therapies in the neoadjuvant setting (NCT01042379—see Table 7).

4.4.2. Trastuzumab Rezetecan

Status: Phase 2/3 clinical trials.
Trastuzumab rezetecan, also called SHR-A1811, is an ADC composed of trastuzumab and SHR9265, a novel topoisomerase I inhibitor derived from exatecan with a better liposolubility and cellular permeability. The two compounds are bonded by a stable and cleavable linker, in a drug-to-antibody ratio of 5.7:1 [97]. In preclinical studies, SHR-A1811 showed HER2-dependent antitumoral activity in breast and gastric cancer cell lines, resulting in a dramatic and sustained tumoral cell growth inhibition. It was also shown to have a significant bystander effect [97].
A recently published phase 1 study in HER2-expressing or -mutated solid tumors revealed that SHR-A1811 had a favorable antitumor effect with a manageable toxicity profile [98].
There are currently several ongoing trials testing SHR-A1811 in monotherapy or in combination with other agents—one phase 3 trial in HER-2-overexpressing metastatic breast cancer (NCT06057610), three in HER-2-low metastatic breast cancer (NCT05814354; NCT05845138; NCT05792410) and one in the neoadjuvant setting (NCT05635487).

4.4.3. ARX788

Status: Phase 2/3 clinical trials.
ARX788 is an ADC composed of an anti-HER-2 mAb conjugated with monomethyl auristatin F (MMAF), a highly potent synthetic auristatin derivative that acts as a microtubule inhibitor. These two components are connected by a non-cleavable linker, as in T-DM1, with a unique site-specific conjugation technology, unlike other ADCs, in which cytotoxic payloads are randomly assembled to antibodies. This ADC’s drug-to-antibody ratio is 1.9:1, lower than existent ADCs [99].
Preclinical studies revealed efficacy in HER-2-overexpressing and HER-2-low breast tumor cells and even in cells resistant to T-DM1. It also showed a stronger antitumoral activity when compared with T-DM1 [99,100]. A phase 1 clinical trial confirmed its efficacy in HER-2-overexpressing metastatic breast cancer patients who progressed on previous HER-2-targeted therapies, with a manageable safety profile [101].
There are currently ongoing one phase 2 trial in the metastatic setting [(NCT04829604), and three phase 2/3 studies in the neoadjuvant setting, two in combination with the TKI pyrotinib (NCT04983121; NCT05426486) and one in monotherapy in a previously mentioned multi-arm study (NCT01042379).

4.4.4. Disitamab Vedotin (RC48)

Status: Phase 2/3 clinical trials; approved in China, not approved by FDA or EMA.
Disitamab vedotin is an ADC that combines hertuzumab (a new anti-HER-2 mAb that targets HER-2 subdomain IV in a different epitope than trastuzumab) with monomethylauristatin E (MMAE), also known as vedotin, an inhibitor of tubuline polymerization during cell division [102,103]. This ADC integrates a cleavable linker and has an estimated drug-to-antibody ratio of 4:1.
In preclinical studies, RC48 demonstrated greater efficacy than trastuzumab and lapatinib against HER-2-overexpressing breast cancer cells and showed a greater antitumoral effect than T-DM1 in trastuzumab- and lapatinib-resistant tumor cells [102]. Ongoing phase 1 studies are assessing RC48’s efficacy in both HER-2-overexpressing and HER-2-low breast cancer (NCT02881138; NCT03052634).
There are several ongoing phase 2/3 trials in China, testing the effect of disitamab vedotin in different subsets of breast cancer: two studies in HER-2-positive metastatic breast cancer (NCT05331326; NCT03500380); three studies in HER-2-low metastatic breast cancer (NCT04400695; NCT05904964; NCT06105008); and finally, two studies in the neoadjuvant setting of HER-2-positive breast cancer (NCT05134519; NCT05726175) (see Table 7).
Disitamab vedotin received approval in China in June 2021 for treatment of HER-2-positive locally advanced/metastatic breast cancer beyond the third line [104].

4.4.5. Zanidatamab Zovodotin (ZW49)

Status: Phase 1 clinical trials.
Zanidatamab zovodotin is an ADC that combines zanidatamab, a bsAb targeting domains II and IV of the HER-2 extracellular component, and N-acyl sulfonamide auristatin, also called zovodotin, an auristatin that inhibits microtubule polymerization during cell division and that is associated with a more favorable toxicity profile than other auristatins such as MMAE and MMAF. It is bonded by a cleavable linker and has a drug-to-antibody ratio of 2:1 [105].
A preclinical study documented the antitumoral activity of ZW49 against HER-2-overexpressing and HER-2-low breast cancer cells and registered an increased binding and internalization of this ADC when compared with trastuzumab-based ADCs [105]. A phase 1 study testing ZW49 in HER-2-overexpressing solid tumors has been recently completed (NCT03821233). Preliminary data confirmed its efficacy in monotherapy, with a manageable toxicity profile in heavily pretreated HER-2-positive solid tumors [106].

4.4.6. MRG002

Status: Phase 2/3 clinical trials.
MRG002 is an ADC composed of a recombinant humanized anti-HER-2 mAb similar to trastuzumab but with less ADCC stimulation, called MAB802, and MMAE, an auristatin previously mentioned. These compounds are connected by a protease-cleavable linker and the ADC’s estimated drug-to-antibody ratio is 3.8:1 [107].
Preclinical studies showed an antitumoral effect in HER-2-overexpressing breast cancer cells and greater efficacy when compared with both trastuzumab and T-DM1, with an antitumoral effect in T-DM1-resistant cancer cells [107].
There are currently three phase 2/3 clinical trials ongoing in China, in the metastatic setting for HER-2-overexpressing (NCT05263869; NCT04924699) and HER-2-low (NCT04742153) breast cancer (see Table 7).

4.4.7. A166

Status: Preclinical studies.
A166 is a trastuzumab-based ADC that carries a duostatin-5 payload, a cytotoxic that acts by inhibiting microtubules. Duostatin-5 binds to trastuzumab by a protease-cleavable linker through a site-specific conjugation technology, in a drug-to-antibody ratio of 2:1.
Phase 1 studies revealed promising the efficacy and tolerable toxicity of A166 in HER-2-positive advanced solid tumors refractory to standard HER-2-targeted therapies [108]. There is another phase 1 trial currently ongoing in China in HER-2-overexpressing solid tumors (NCT05311397), but there are still no studies specifically in a breast cancer setting.

4.4.8. ALT-P7

Status: Phase 1 clinical trials.
ALT-P7 is another ADC composed by a trastuzumab biosimilar, HM2, and MMAE, a microtubule inhibitor, both connected by a cleavable linker in a ratio of 2:1 [109]. A phase 1 trial including HER-2 positive breast cancer patients that progressed under trastuzumab-based therapy was recently conducted (NCT03281824) and preliminary data reveals a favorable tolerability profile and indicates that there will probably be a phase 2 study [110].

4.4.9. XMT-1522

Status: Phase 1 clinical trials.
XMT-1522 is an ADC composed of an anti-HER-2 mAb, HT-19, which binds subdomain IV of HER-2 in a different epitope from trastuzumab, combined with a microtubule-inhibiting auristatin derivative, AF-HPA, in a proportion of 12:1. This ADC has this abnormally high drug-to-antibody ratio due to a biodegradable polymer-based conjugation platform that enables these high ratios without plasmatic aggregation or interference with the drug pharmacokinetics [109,111].
Preclinical studies demonstrated XMT-1522 to be effective against HER-2-overexpressing breast cancer cells, even in the presence of T-DM1 resistance, and with an approximately 100 times higher potency than T-DM1 [112]. It also showed a synergistic effect with trastuzumab and pertuzumab [113] and revealed an antitumoral effect in HER-2-low breast cancer cells [114]. A phase 1 clinical trial testing this ADC in the metastatic HER-2-overexpressing breast cancer setting (NCT02952729) was recently concluded, and preliminary data showed a good tolerability and antitumor activity [115].

4.4.10. Discontinued ADCs

PF-06804103 is composed of a trastuzumab-derived antibody associated with Aur0101, a potent microtubule polymerization inhibitor, with site-specific conjugation technology, in a proportion of 4:1 [116]. Preclinical studies showed antitumor activity against both HER-2-positive and HER-2-low breast cancer, showing greater response rates and more durable complete responses when compared with T-DM1 [117]. A recently published phase 1 trial confirmed antitumor activity in HER-2-positive and HER-2-low breast and gastric cancers but revealed significant toxicity in almost half of the patients [118], leading to the drug’s discontinuation.
Alta-ADC is an ADC composed of pertuzumab and MMAE in a proportion of 2:1. It has a particularly low affinity for HER-2 at low pH, allowing its dissociation in the endosome, and subsequent pertuzumab release to bind a different HER-2 receptor in the cell membrane. This particularity allows Alta-ADC to be effective in lower doses, which warrants a more manageable toxicity profile [119]. Despite preclinical studies showing superior efficacy when compared with T-DM1 and also efficacy in HER-2-low tumors, no more studies were conducted.
MEDI4276 is an ADC that combines a HER-2-bispecific antibody, which binds HER-2 in two different epitopes from trastuzumab and pertuzumab, and a novel microtubule inhibitor called AZ13599185, both connected by a cleavable linker, in a ratio of 4:1 [120,121]. Preclinical studies revealed a 10-fold more potent antitumoral effect than T-DM1 and also efficacy in T-DM1-resistant and HER-2-low tumor cells [121]. A phase 1/2 study conducted in 2016–2017, in heavily pretreated HER-2-overexpressing breast and gastric cancer patients, showed unacceptable toxicity [122], leading to its discontinuation.
DHES0815A is composed of an anti-HER-2 mAb linked to the cytotoxic drug pyrrolo [2,1-c][1,4] benzodiazepine monoamide (PBD-MA), which creates crosslinks in DNA minor grooves, leading to DNA strand breaks [123]. A phase 1 study revealed a modest antitumoral effect with significant toxicity [124], which led to drug discontinuation in the setting of breast cancer.

4.5. Other Antibody Conjugates

4.5.1. Immune-Stimulating Antibody Conjugates (ISAC)

BDC-1001 is not an ADC, as it includes no cytotoxic molecule, but is an immune-stimulating antibody conjugate, as it carries an immune-stimulating agent. It is composed of a trastuzumab biosimilar and a TLR-7/8 agonist that stimulates antigen-presenting cells to recognize and kill HER-2-overexpressing tumor cells, promoting a durable adaptive immune response. The two components are bound by a non-cleavable linker.
In preclinical studies, BDC-1001 showed an antitumoral effect through a solid immune response [125].
There are currently ongoing two phase 2 studies, one in HER-2-overexpressing metastatic breast cancer in combination with pertuzumab (NCT05954143), and one in HER-2-overexpressing solid tumors in combination with nivolumab (NCT04278144).

4.5.2. Radionuclide–Antibody Conjugates

Thorium-227 conjugates (TTCs) are a combination of monoclonal antibodies with α-emitting radionuclides (thorium-227) that induce DNA double strand breaks, resulting in cell death, and they also have the capacity to stimulate an immunogenic response [126]. BAY2701439 is a TTC composed of an anti-HER-2, trastuzumab-derived antibody, conjugated with thorium-227. Preclinical studies demonstrated an antitumoral effect in HER-2-overexpressing tumor cells, including breast cancer cells, and in this set of patients, BAY2701439 also revealed efficacy in HER-2-low and T-DM1-resistant tumors [127]. A phase 1 clinical trial testing BAY2701439 in HER-2-overexpressing solid tumors was recently concluded, and preliminary data is awaited (NCT04147819).

4.6. Combinations of HER-2-Targeted and Other Agents

4.6.1. Combination of HER-2-Targeted and Other Targeted Agents

HER-2-positive breast cancer is a heterogeneous disease, with tumors having different levels of hormonal receptor expression and a wide range of genomic alterations. Therefore, there is a growing interest in assessing the benefit of combining HER-2-targeted agents with other targeted therapies. Table 8 lists the main ongoing trials testing the combination of HER-2-targeted and other targeted agents.

4.6.2. Combination of HER-2-Targeted Agents and Immunotherapy

Besides the direct inhibition of HER-2 signaling pathways through HER-2-targeted therapies, leading to cell cycle arrest and cell growth inhibition, there is also an important role for immune-mediated cell death in HER-2-overexpressing tumors. Ever since the development of the first HER-2-targeted drug, trastuzumab, the immune system proved to play an important role in tumor growth inhibition and apoptosis through ADCC and ADCP [17,18]. This cytotoxic mechanism is so important that its blockade by HLA class I (such as HLA-G)-mediated NK cells’ inhibition constitutes an important resistance mechanism to trastuzumab [128]. Besides ADCC and ADCP, trastuzumab was also shown to stimulate the production of TGFβ and IFN-γ by tumor cells and NK cells, which leads to the expression of PD-1 on NK cells [128].
These parallel mechanisms of action prove that immune stimulation may be of great importance in HER-2-positive tumor cells’ death, and it may enhance HER-2 inhibition and help overcome resistance mechanisms to HER-2-targeted agents. Immune stimulation in this context has the additional advantage of triggering immune memory against HER-2 cancer cells and providing a more durable and sustained antitumoral response [129].
This premise has led to the conceptualization of novel immune-stimulating drugs and to the evaluation of the benefit of several combinations of immune checkpoint inhibitors (ICI) with HER-2-targeted agents. A phase 1 study published in 2019 confirmed that pembrolizumab in association with trastuzumab had antitumor efficacy in previously trastuzumab-resistant patients with both HER-2 and PD-L1 positivity [130]. A recently published phase 3 trial showed, however, no benefit in adding an ICI, atezolizumab, to neoadjuvant chemotherapy and double HER-2 blockade, regarding the rate of pCR [131]. Table 9 summarizes the main ongoing clinical trials testing combinations of ICI with HER-2-targeted agents.

5. Other exploratory Therapies in HER-2-Positive Breast Cancer

5.1. PROTACs

Proteolysis-targeting chimeras (PROTACs) are stimulators of the natural ubiquitin-protease system. They are composed of two ligands connected by a linker—one ligand that binds the target protein and another ligand that binds a ubiquitin ligase. The binding of PROTACs to their targets induces ubiquitylation and degradation of the target protein by the ubiquitin-proteasome system, after which the PROTAC regenerates to bind other targets [132].
Ab-PROTAC 3 is a trastuzumab-PROTAC conjugate, connected by a cleavable linker, that induces ubiquitin-proteasome-mediated degradation of bromodomain-containing 4 (BDR4), a protein that plays an essential role in DNA replication during cell division. Preclinical studies showed that this conjugate can selectively target BDR4 in HER-2-positive tumor cells [133].
ORM-5029 is a pertuzumab–PROTAC conjugate that targets GSTP1, a protein important in protein synthesis in the ribosome and thus cell survival. Preclinical studies revealed a ten- to a thousand-fold greater potency than trastuzumab in suppressing HER-2-positive cancer cells, and it also showed an effect in HER-2-low cancer. There is a phase 1 ongoing trial testing ORM-5029 in HER-2-positive solid tumors (NCT05511844).

5.2. Cell Therapies

Cell-based immunotherapy is an innovative therapeutic modality, currently in use only in hemato-oncology, that uses genetically engineered immune cells from the patient to target and eliminate specific tags in tumor cells. After removing immune cells (T cells, NK cells or macrophages) from the patient’s tumoral microenvironment to modify their receptors accordingly, these cells are expanded ex vivo and reimplanted in the patient’s circulation to boost the patient’s antitumoral immune response. Despite success in liquid tumors, implementation of cell-based therapies in solid tumors has been full of obstacles, regarding the increased difficulty of accessing immune cells in the tumor, the triggered systemic inflammatory response syndrome (SIRS) and the side effects profile [134,135].
There are currently a few ongoing phase 1 clinical trials in HER-2-positive tumors, including with CAR-T cells (NCT04511871), CAR-NK cells (NCT05385705) and with CAR-macrophages (NCT04660929).

5.3. Cancer Vaccines

Another early and exploratory therapeutical modality is cancer vaccines. Cancer vaccines grant the advantages of having a very mild and manageable toxicity profile when compared with any other antitumoral treatment, even targeted HER-2 therapies, a very favourable posology, with no need for frequent administrations, and of providing a long-term and durable immune response specifically targeted against the tumor [136,137]. Cancer vaccines can be peptide-based, protein-based, cell-based, made of dendritic cells, virus-based or made of recombinant DNA.
The first vaccine to show benefits in cancer was SipuleucelT, which proved to be effective in prostate cancer [138]. Nelipepimut-S is a vaccine that combines E75, a peptide that mimics the extracellular domain of the HER-2 receptor, and granulocyte-macrophage colony-stimulating factor. Preclinical studies demonstrated E75 recognition by CD8+ lymphocytes and subsequent HER-2-positive cells death [139]. Phase 1 and 2 studies confirmed its safety and efficacy in triggering an immune response [140,141,142]. A phase 3 study failed, however, to show a disease-free survival benefit in HER-2-positive early breast cancer patients, leading to the study’s early termination [143]. A vaccine integrating a different peptide, GLSI-100, revealed an effective immune response in a phase 2b trial [144] and is currently being evaluated in a phase 3 trial (NCT05232916).
There are several other molecules undergoing evaluation in ongoing phase 1 and 2 trials (for example, NCT04418219; NCT03384914; NCT03387553; NCT05378464; NCT05325632).

6. Conclusions and Future Directions

Ever since the discovery and description of HER-2, the landscape of HER-2-overexpressing breast cancer has been constantly evolving. Trastuzumab was the first-ever targeted drug developed, more than a quarter of a century ago, bringing a shift of paradigm in the prognosis of this subset of patients, so much that it is still part of the standard of care. Since then, several other effective targeted agents have been developed and successfully integrated into clinical practice, contributing to a continuous improvement in the treatment of this subset of patients. Despite this constant improvement, drug resistance still occurs, and the disease still progresses under HER-2-targeted therapy, mostly due to suboptimal HER-2 inhibition or to mechanisms of resistance. It makes it essential to keep investing in the development of new drugs and better therapeutical options.
Thanks to a joint effort of basic scientists, clinical investigators and the pharmaceutical industry, continued research in this area has been bringing to light multiple and promising novel HER-2-targeted drugs, which optimize HER-2 blockage and overcome some of the resistance mechanisms identified so far. There is a special focus on the recently discovered class of antibody–drug conjugates, which allow scientists to combine HER-2 targeting with a tailor-delivered cytotoxic molecule with limited toxicity, and progress in this area keeps growing. The focus has not only been on HER-2-targeted drugs; other classes of drugs have also shown a benefit and synergistic effect with anti-HER-2 therapies, thereby improving treatment efficacy and diversifying future therapeutical weapons. Even immunotherapy and other forms of immune stimulation have been showing a significant benefit in this disease previously considered “cold” and non-immune-sensitive.
Despite many encouraging discoveries in the last few years, there is still a long way to go towards improving HER-2-positive breast cancer patients’ lives. There are currently 662 ongoing clinical trials, and many more promising to come up soon. Other flourishing therapeutical possibilities, still very exploratory, include single-cell sequencing, transcriptomics, proteomics, theragnostics and even modulating the microbiome. Advances in molecular diagnosis could help identify tumor-specific resistance mechanisms and surrogate biomarkers, allowing for early resistance detection and the tailoring of therapeutical options.

Author Contributions

Original draft preparation and editing: C.L.F. and D.J.S.; Review: A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Giaquinto, A.N.; Sung, H.; Miller, K.D.; Kramer, J.L.; Newman, L.A.; Minihan, A.; Jemal, A.; Siegel, R.L. Breast Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 524–541. [Google Scholar] [CrossRef] [PubMed]
  3. Slamon, D.J.; Clark, G.M.; Wong, S.G.; Levin, W.J.; Ullrich, A.; McGuire, W.L. Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987, 235, 177–182. [Google Scholar] [CrossRef] [PubMed]
  4. Pegram, M.D.; Pauletti, G.; Slamon, D.J. HER-2/neu as a predictive marker of response to breast cancer therapy. Breast Cancer Res. Treat. 1998, 52, 65–77. [Google Scholar] [CrossRef] [PubMed]
  5. Slamon, D.J.; Godolphin, W.; Jones, L.A.; Holt, J.A.; Wong, S.G.; Keith, D.E.; Levin, W.J.; Stuart, S.G.; Udove, J.; Ullrich, A.; et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989, 244, 707–712. [Google Scholar] [CrossRef] [PubMed]
  6. Slamon, D.J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 2001, 344, 783–792. [Google Scholar] [CrossRef]
  7. Mukohara, T. Mechanisms of resistance to anti-human epidermal growth factor receptor 2 agents in breast cancer. Cancer Sci. 2011, 102, 1–8. [Google Scholar] [CrossRef]
  8. Garrett, J.T.; Arteaga, C.L. Resistance to HER2-directed antibodies and tyrosine kinase inhibitors: Mechanisms and clinical implications. Cancer Biol. Ther. 2011, 11, 793–800. [Google Scholar] [CrossRef]
  9. Rexer, B.N.; Arteaga, C.L. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: Mechanisms and clinical implications. Crit. Rev. Oncogene 2012, 17, 1–16. [Google Scholar] [CrossRef]
  10. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/search?distance=50&cond=Breast%20Cancer&term=HER-2positive%20Breast%20Cancer&aggFilters=ages:adult%20older,status:rec%20act,studyType:int (accessed on 7 November 2023).
  11. Xia, W.; Mullin, R.J.; Keith, B.R.; Liu, L.H.; Ma, H.; Rusnak, D.W.; Owens, G.; Alligood, K.J.; Spector, N.L. Anti-tumor activity of GW572016: A dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene 2002, 21, 6255–6263. [Google Scholar] [CrossRef]
  12. Franklin, M.C.; Carey, K.D.; Vajdos, F.F.; Leahy, D.J.; de Vos, A.M.; Sliwkowski, M.X. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 2004, 5, 317–328. [Google Scholar] [CrossRef] [PubMed]
  13. Koeppen, H.K.; Wright, B.D.; Burt, A.D.; Quirke, P.; McNicol, A.M.; Dybdal, N.O.; Sliwkowski, M.X.; Hillan, K.J. Overexpression of HER2/neu in solid tumours: An immunohistochemical survey. Histopathology 2001, 38, 96–104. [Google Scholar] [CrossRef] [PubMed]
  14. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  15. Wolff, A.C.; Somerfield, M.R.; Dowsett, M.; Hammond, M.E.H.; Hayes, D.F.; McShane, L.M.; Saphner, T.J.; Spears, P.A.; Allison, K.H. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: ASCO-College of American Pathologists Guideline Update. J. Clin. Oncol. 2023, 41, 3867–3872. [Google Scholar] [CrossRef] [PubMed]
  16. Baselga, J. Clinical trials of Herceptin(R) (trastuzumab). Eur. J. Cancer 2001, 37 (Suppl. S1), 18–24. [Google Scholar] [CrossRef] [PubMed]
  17. Park, S.; Jiang, Z.; Mortenson, E.D.; Deng, L.; Radkevich-Brown, O.; Yang, X.; Sattar, H.; Wang, Y.; Brown, N.K.; Greene, M.; et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 2010, 18, 160–170. [Google Scholar] [CrossRef]
  18. Shi, Y.; Fan, X.; Deng, H.; Brezski, R.J.; Rycyzyn, M.; Jordan, R.E.; Strohl, W.R.; Zou, Q.; Zhang, N.; An, Z. Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcgamma receptors on macrophages. J. Immunol. 2015, 194, 4379–4386. [Google Scholar] [CrossRef]
  19. Baselga, J.; Norton, L.; Albanell, J.; Kim, Y.M.; Mendelsohn, J. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res. 1998, 58, 2825–2831. [Google Scholar]
  20. Slamon, D.; Eiermann, W.; Robert, N.; Pienkowski, T.; Martin, M.; Press, M.; Mackey, J.; Glaspy, J.; Chan, A.; Pawlicki, M.; et al. Adjuvant trastuzumab in HER-2 positive breast cancer. N. Engl. J. Med. 2011, 365, 1273–1283. [Google Scholar] [CrossRef]
  21. Swain, S.M.; Kim, S.B.; Cortes, J.; Ro, J.; Semiglazov, V.; Campone, M.; Ciruelos, E.; Ferrero, J.M.; Schneeweiss, A.; Knott, A.; et al. Pertuzumab, trastuzumab, and docetaxel for HER-2 positive metastatic breast cancer (CLEOPATRA study): Overall survival results from a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2013, 14, 461–471. [Google Scholar] [CrossRef]
  22. Gianni, L.; Pienkowski, T.; Im, Y.H.; Roman, L.; Tseng, L.M.; Liu, M.C.; Lluch, A.; Staroslawska, E.; Haba Rodriguez, J.; Im, S.A.; et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER-2 positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012, 13, 25–32. [Google Scholar] [CrossRef] [PubMed]
  23. Tolaney, S.M.; Barry, W.T.; Dang, C.T.; Yardley, D.A.; Moy, B.; Marcom, P.K.; Albain, K.S.; Rugo, H.S.; Ellis, M.; Chir, B.; et al. Adjuvant paclitaxel and trastuzumab for node-negative, HER-2 positive breast cancer. N. Engl. J. Med. 2015, 372, 134–141. [Google Scholar] [CrossRef] [PubMed]
  24. Gianni, L.; Llado, A.; Bianchi, G.; Cortes, J.; Kellokumpu-Lehtinen, P.L.; Cameron, D.A.; Miles, D.; Salvagni, S.; Wardley, A.; Goeminne, J.C.; et al. Open-label, phase II, multicenter, randomized study of the efficacy and safety of two dose levels of Pertuzumab, a human epidermal growth factor receptor 2 dimerization inhibitor, in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer. J. Clin. Oncol. 2010, 28, 1131–1137. [Google Scholar] [PubMed]
  25. Von Minckwitz, G.; Procter, M.; de Azambuja, E.; Zardavas, D.; Benyunes, M.; Viale, G.; Suter, T.; Arahmani, A.; Rouchet, N.; Clark, E.; et al. Adjuvant Pertuzumab and Trastuzumab in Early HER-2 positive Breast Cancer. N. Engl. J. Med. 2017, 377, 122–131. [Google Scholar] [CrossRef] [PubMed]
  26. Nordstrom, J.L.; 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. 2011, 13, R123. [Google Scholar] [CrossRef] [PubMed]
  27. Rugo, H.S.; Im, S.A.; Cardoso, F.; Cortes, J.; Curigliano, G.; Musolino, A.; Pegram, M.D.; Bachelot, T.; Wright, G.S.; Saura, C.; et al. Margetuximab versus Trastuzumab in Patients with Previously Treated HER-2 positive Advanced Breast Cancer (SOPHIA): Final Overall Survival Results From a Randomized Phase 3 Trial. J. Clin. Oncol. 2023, 41, 198–205. [Google Scholar] [CrossRef] [PubMed]
  28. Traxler, P. Tyrosine kinases as targets in cancer therapy—Successes and failures. Expert Opin. Ther. Targets 2003, 7, 215–234. [Google Scholar] [CrossRef] [PubMed]
  29. Scaltriti, M.; Chandarlapaty, S.; Prudkin, L.; Aura, C.; Jimenez, J.; Angelini, P.; Sánchez, G.; Guzman, M.; Parra, J.L.; Ellis, C.; et al. Clinical benefit of lapatinib-based therapy in patients with human epidermal growth factor receptor 2–positive breast tumors coexpressing the truncated p95HER2 receptor. Clin. Cancer Res. 2010, 16, 2688–2695. [Google Scholar] [CrossRef]
  30. Geyer, C.E.; Forster, J.; Lindquist, D.; Chan, S.; Romieu, C.G.; Pienkowski, T.; Jagiello-Gruszfeld, A.; Crown, J.; Chan, A.; Kaufman, B.; et al. Lapatinib plus capecitabine for HER-2 positive advanced breast cancer. N. Engl. J. Med. 2006, 355, 2733–2743. [Google Scholar] [CrossRef]
  31. Goss, P.E.; Smith, I.E.; O’Shaughnessy, J.; Ejlertsen, B.; Kaufmann, M.; Boyle, F.; Buzdar, A.U.; Fumoleau, P.; Gradishar, W.; Martin, M.; et al. Adjuvant lapatinib for women with early-stage HER-2 positive breast cancer: A randomised, controlled, phase 3 trial. Lancet Oncol. 2013, 14, 88–96. [Google Scholar] [CrossRef]
  32. Piccart-Gebhart, M.; Holmes, E.; Baselga, J.; de Azambuja, E.; Dueck, A.C.; Viale, G.; Zujewski, J.A.; Goldhirsch, A.; Armour, A.; Pritchard, K.; et al. Adjuvant Lapatinib and Trastuzumab for Early Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Results From the Randomized Phase III Adjuvant Lapatinib and/or Trastuzumab Treatment Optimization Trial. J. Clin. Oncol. 2016, 34, 1034–1042. [Google Scholar] [CrossRef] [PubMed]
  33. Untch, M.; Loibl, S.; Bischoff, J.; Eidtmann, H.; Kaufmann, M.; Blohmer, J.U.; Hilfrich, J.; Strumberg, D.; Fasching, P.A.; Kreienberg, R.; et al. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy (GeparQuinto, GBG 44): A randomised phase 3 trial. Lancet Oncol. 2012, 13, 135–144. [Google Scholar] [CrossRef] [PubMed]
  34. Guarneri, V.; Dieci, M.V.; Griguolo, G.; Miglietta, F.; Girardi, F.; Bisagni, G.; Generali, D.G.; Cagossi, K.; Sarti, S.; Frassoldati, A.; et al. Trastuzumab-lapatinib as neoadjuvant therapy for HER-2 positive early breast cancer: Survival analyses of the CHER-Lob trial. Eur. J. Cancer 2021, 153, 133–141. [Google Scholar] [CrossRef] [PubMed]
  35. De Azambuja, E.; Holmes, A.P.; Piccart-Gebhart, M.; Holmes, E.; Di Cosimo, S.; Swaby, R.F.; Untch, M.; Jackisch, C.; Lang, I.; Smith, I.; et al. Lapatinib with trastuzumab for HER-2 positive early breast cancer (NeoALTTO): Survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response. Lancet Oncol. 2014, 15, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
  36. Lin, N.U.; Carey, L.A.; Liu, M.C.; Younger, J.; Come, S.E.; Ewend, M.; Harris, G.J.; Bullitt, E.; Abbeele, A.D.; Henson, J.W.; et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J. Clin. Oncol. 2008, 26, 1993–1999. [Google Scholar] [CrossRef] [PubMed]
  37. Rabindran, S.K.; Discafani, C.M.; Rosfjord, E.C.; Baxter, M.; Floyd, M.B.; Golas, J.; Hallett, W.A.; Johnson, B.D.; Nilakantan, R.; Overbeek, E.; et al. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Res. 2004, 64, 3958–3965. [Google Scholar] [CrossRef] [PubMed]
  38. Segovia-Mendoza, M.; Gonzalez-Gonzalez, M.E.; Barrera, D.; Diaz, L.; Garcia-Becerra, R. Efficacy and mechanism of action of the tyrosine kinase inhibitors gefitinib, lapatinib and neratinib in the treatment of HER-2 positive breast cancer: Preclinical and clinical evidence. Am. J. Cancer Res. 2015, 5, 2531–2561. [Google Scholar]
  39. Martin, M.; Bonneterre, J.; Geyer, C.E., Jr.; Ito, Y.; Ro, J.; Lang, I.; Kim, S.B.; Germa, C.; Vermette, J.; Wang, K.; et al. A phase two randomised trial of neratinib monotherapy versus lapatinib plus capecitabine combination therapy in patients with HER2+ advanced breast cancer. Eur. J. Cancer 2013, 49, 3763–3772. [Google Scholar] [CrossRef]
  40. Freedman, R.A.; Gelman, R.S.; Wefel, J.S.; Melisko, M.E.; Hess, K.R.; Connolly, R.M.; Poznak, C.H.; Niravath, P.A.; Puhalla, S.L.; Ibrahim, N.; et al. Translational Breast Cancer Research Consortium (TBCRC) 022: A Phase II Trial of Neratinib for Patients with Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. J. Clin. Oncol. 2016, 34, 945–952. [Google Scholar] [CrossRef]
  41. Saura, C.; Oliveira, M.; Feng, Y.H.; Dai, M.S.; Chen, S.W.; Hurvitz, S.A.; Kim, S.B.; Moy, B.; Delaloge, S.; Gradishar, W.; et al. Neratinib Plus Capecitabine versus Lapatinib Plus Capecitabine in HER-2 positive Metastatic Breast Cancer Previously Treated with ≥2 HER2-Directed Regimens: Phase III NALA Trial. J. Clin. Oncol. 2020, 38, 3138–3149. [Google Scholar] [CrossRef]
  42. Martin, M.; Holmes, F.A.; Ejlertsen, B.; Delaloge, S.; Moy, B.; Iwata, H.; Minckwitz, G.; Chia, S.K.L.; Mansi, J.; Barrios, C.H.; et al. Neratinib after trastuzumab-based adjuvant therapy in HER-2 positive breast cancer (ExteNET): 5-year analysis of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1688–1700. [Google Scholar] [CrossRef] [PubMed]
  43. Park, J.W.; Liu, M.C.; Yee, D.; Yau, C.; van ‘t Veer, L.J.; Symmans, W.F.; Paoloni, M.; Perlmutter, J.; Hylton, N.M.; Hogarth, M.; et al. Adaptive Randomization of Neratinib in Early Breast Cancer. N. Engl. J. Med. 2016, 375, 11–22. [Google Scholar] [CrossRef] [PubMed]
  44. Jacobs, S.A.; Robidoux, A.; Abraham, J.; Perez-Garcia, J.M.; La Verde, N.; Orcutt, J.M.; Cazzaniga, M.E.; Piette, F.; Antolín, S.; Aguirre, E.; et al. NSABP FB-7: A phase II randomized neoadjuvant trial with paclitaxel + trastuzumab and/or neratinib followed by chemotherapy and postoperative trastuzumab in HER2(+) breast cancer. Breast Cancer Res. 2019, 21, 133. [Google Scholar] [CrossRef] [PubMed]
  45. Kulukian, A.; Lee, P.; Taylor, J.; Rosler, R.; de Vries, P.; Watson, D.; Forero-Torres, A.; Peterson, S. Preclinical Activity of HER2-Selective Tyrosine Kinase Inhibitor Tucatinib as a Single Agent or in Combination with Trastuzumab or Docetaxel in Solid Tumor Models. Mol. Cancer Ther. 2020, 19, 976–987. [Google Scholar] [CrossRef] [PubMed]
  46. Kaur, S.; Bhattacharyya, R.; Banerjee, D.; Shukla, J. Application of Tucatinib and Trastuzumab: Dual Anti HER2 Therapy Against HER2 Positive Breast Cancer. Indian J. Clin Biochem. 2021, 36, 124–125. [Google Scholar] [CrossRef] [PubMed]
  47. Curigliano, G.; Mueller, V.; Borges, V.; Hamilton, E.; Hurvitz, S.; Loi, S.; Murthy, R.; Okines, A.; Paplomata, E.; Cameron, D.; et al. Tucatinib versus placebo added to trastuzumab and capecitabine for patients with pretreated HER2+ metastatic breast cancer with and without brain metastases (HER2CLIMB): Final overall survival analysis. Ann. Oncol. 2022, 33, 321–329. [Google Scholar] [CrossRef] [PubMed]
  48. Li, X.; Yang, C.; Wan, H.; Zhang, G.; Feng, J.; Zhang, L.; Chen, X.; Zhong, D.; Lou, L.; Tao, W.; et al. Discovery and development of pyrotinib: A novel irreversible EGFR/HER2 dual tyrosine kinase inhibitor with favorable safety profiles for the treatment of breast cancer. Eur. J. Pharm. Sci. 2017, 110, 51–61. [Google Scholar] [CrossRef]
  49. Ma, F.; Ouyang, Q.; Li, W.; Jiang, Z.; Tong, Z.; Liu, Y.; Li, H.; Yu, S.; Feng, J.; Wang, S.; et al. Pyrotinib or Lapatinib Combined with Capecitabine in HER-2 positive Metastatic Breast Cancer with Prior Taxanes, Anthracyclines, and/or Trastuzumab: A Randomized, Phase II Study. J. Clin. Oncol. 2019, 37, 2610–2619. [Google Scholar] [CrossRef]
  50. Jiang, Z.; Yan, M.; Hu, X.; Zhang, Q.; Ouyang, Q.; Feng, J.; Yin, Y.; Sun, T.; Tong, Z.; Wang, X.; et al. Pyrotinib combined with capecitabine in women with HER2+ metastatic breast cancer previously treated with trastuzumab and taxanes: A randomized phase III study. J. Clin. Oncol. 2019, 37, 1001. [Google Scholar] [CrossRef]
  51. Bao, Y.; Zhang, Z.; He, X.; Cai, L.; Wang, X.; Li, X. Cost-Effectiveness of Pyrotinib Plus Capecitabine versus Lapatinib Plus Capecitabine for the Treatment of HER2-Positive Metastatic Breast Cancer in China: A Scenario Analysis of Health Insurance Coverage. Curr. Oncol. 2022, 29, 6053–6067. [Google Scholar] [CrossRef]
  52. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab emtansine for HER-2 positive advanced breast cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [PubMed]
  53. Von Minckwitz, G.; Huang, C.S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER-2 positive Breast Cancer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef] [PubMed]
  54. Lewis Phillips, G.D.; Li, G.; Dugger, D.L.; Crocker, L.M.; Parsons, K.L.; Mai, E.; Blattler, W.A.; Lambert, J.M.; Chari, R.V.J.; Lutz, R.J.; et al. Targeting HER-2 positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290. [Google Scholar] [CrossRef] [PubMed]
  55. Dieras, V.; Miles, D.; Verma, S.; Pegram, M.; Welslau, M.; Baselga, J.; Krop, I.E.; Blackwell, K.; Hoersch, S.; Xu, J.; et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER-2 positive advanced breast cancer (EMILIA): A descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 732–742. [Google Scholar] [CrossRef] [PubMed]
  56. Yver, A.; Agatsuma, T.; Soria, J.C. The art of innovation: Clinical development of trastuzumab deruxtecan and redefining how antibody-drug conjugates target HER-2 positive cancers. Ann. Oncol. 2020, 31, 430–434. [Google Scholar] [CrossRef] [PubMed]
  57. Tamura, K.; Tsurutani, J.; Takahashi, S.; Iwata, H.; Krop, I.E.; Redfern, C.; Sagara, Y.; Doi, T.; Park, H.; Murthy, R.K.; et al. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER-2 positive breast cancer previously treated with trastuzumab emtansine: A dose-expansion, phase 1 study. Lancet Oncol. 2019, 20, 816–826. [Google Scholar] [CrossRef] [PubMed]
  58. Modi, S.; Saura, C.; Yamashita, T.; Park, Y.H.; Kim, S.B.; Tamura, K.; Andre, F.; Iwata, H.; Ito, Y.; Tsurutani, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER-2 positive Breast Cancer. N. Engl. J. Med. 2020, 382, 610–621. [Google Scholar] [CrossRef]
  59. Cortes, J.; Kim, S.B.; Chung, W.P.; Im, S.A.; Park, Y.H.; Hegg, R.; Kim, M.H.; Tseng, L.M.; Petry, V.; Chung, C.F.; et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N. Engl. J. Med. 2022, 386, 1143–1154. [Google Scholar] [CrossRef]
  60. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab Deruxtecan in Previously Treated HER-2 low Advanced Breast Cancer. N. Engl. J. Med. 2022, 387, 9–20. [Google Scholar] [CrossRef]
  61. Espelin, C.W.; Leonard, S.C.; Geretti, E.; Wickham, T.J.; Hendriks, B.S. Dual HER2 Targeting with Trastuzumab and Liposomal-Encapsulated Doxorubicin (MM-302) Demonstrates Synergistic Antitumor Activity in Breast and Gastric Cancer. Cancer Res. 2016, 76, 1517–1527. [Google Scholar] [CrossRef]
  62. Miller, K.; Cortes, J.; Hurvitz, S.A.; Krop, I.E.; Tripathy, D.; Verma, S.; Riahi, K.; Reynolds, J.G.; Wickham, T.J.; Molnar, I.; et al. HERMIONE: A randomized Phase 2 trial of MM-302 plus trastuzumab versus chemotherapy of physician’s choice plus trastuzumab in patients with previously treated, anthracycline-naive, HER-2 positive, locally advanced/metastatic breast cancer. BMC Cancer 2016, 16, 352. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, T.; Zhang, P.; Di, L.; Wang, X.; Yang, J.; Tong, Z.; Liu, J.; Feng, J.; Liu, D.; Yu, Q.; et al. Efficacy and safety of inetetamab in combination with chemotherapy as first-line treatment of HER-2 positive metastatic breast cancer: A subgroup analysis in the HOPES study. Transl. Breast Cancer Res. 2022, 3, 1136380. [Google Scholar] [CrossRef]
  64. Bian, L.; Xu, B.H.; Di, L.J.; Wang, T.; Wang, X.J.; Jiao, S.C.; Yang, J.L.; Tong, Z.S.; Liu, J.; Feng, J.F.; et al. [Phase Ⅲ randomized controlled, multicenter, prospective study of recombinant anti-HER2 humanized monoclonal antibody (Cipterbin) combined with vinorelbine in patients with HER2 positive metastatic breast cancer: The HOPES Study]. Zhonghua Yi Xue Za Zhi 2020, 100, 2351–2357. [Google Scholar] [PubMed]
  65. Liu, X.; Zhang, P.; Li, C.; Song, X.; Liu, Z.; Shao, W.; Li, S.; Wang, X.; Yu, Z. Efficacy and safety of inetetamab-containing regimens in patients with HER-2 positive metastatic breast cancer: A real-world retrospective study in China. Front. Oncol. 2023, 13, 1136380. [Google Scholar] [CrossRef] [PubMed]
  66. Weisser, N.E.; Wickman, G.; Abraham, L.; O’Toole, J.; Harbourne, B.; Guedia, J.; Cheng, C.W.; Chan, P.; Browman, D.; Gold, M.R.; et al. The bispecific antibody zanidatamab’s (ZW25’s) unique mechanisms of action and durable anti-tumor activity in HER2-expressing cancers. In Proceedings of the AACR Annual Meeting 2021, Philadelphia, PA, USA, 9–14 July 2021; p. 1005. [Google Scholar]
  67. Meric-Bernstam, F.; Hanna, D.; Beeram, M.; Lee, K.W.; Kang, Y.K.; Chaves, J.; Lee, J.; Goodwin, R.; Vaklavas, C.; Oh, D.Y.; et al. Safety, anti-tumour activity, and biomarker results of the HER2-targeted bispecific antibody ZW25 in HER2-expressing solid tumours. Ann. Oncol. 2019, 30, v167–v168. [Google Scholar] [CrossRef]
  68. Bedard, P.L.; Im, S.A.; Elimova, E.; Rha, S.Y.; Goodwin, R.; Ferrario, C.; Lee, K.W.; Hanna, D.; Meric-Bernstam, F.; Mayordomo, J.; et al. Zanidatamab (ZW25), a HER2-targeted bispecific antibody, in combination with chemotherapy (chemo) for HER-2 positive breast cancer (BC): Results from a phase 1 study. In Proceedings of the San Antonio Breast Cancer Symposium 2021, San Antonio, TX, USA, 7–10 December 2021; Cancer Research: San Antonio, TX, USA, 2022. [Google Scholar]
  69. Huang, S.; Li, F.; Liu, H.; Ye, P.; Fan, X.; Yuan, X.; Wu, Z.; Chen, J.; Jin, C.; Shen, B.; et al. Structural and functional characterization of MBS301, an afucosylated bispecific anti-HER2 antibody. MAbs 2018, 10, 864–875. [Google Scholar] [CrossRef] [PubMed]
  70. Wei, H.; Cai, H.; Jin, Y.; Wang, P.; Zhang, Q.; Lin, Y.; Wang, W.; Cheng, J.; Zeng, N.; Xu, T.; et al. Structural basis of a novel heterodimeric Fc for bispecific antibody production. Oncotarget 2017, 8, 51037–51049. [Google Scholar] [CrossRef]
  71. 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 HER-2 positive Metastatic Breast Cancer: Results from a Phase I Study. Clin. Cancer Res. 2022, 28, 618–628. [Google Scholar] [CrossRef]
  72. Ji, D.; Zhang, J.; Shen, W.; Du, Y.; Xu, J.; Yang, J.; Luo, X.; Kong, P.; Yang, F.; Hu, X. Preliminary safety, efficacy and pharmacokinetics (PK) results of KN026 (a HER2 bispecific antibody) monotherapy in advanced solid tumor patients with HER2 expression. J. Clin. Oncol. 2020, 38, 1041. [Google Scholar] [CrossRef]
  73. Geuijen, C.A.W.; De Nardis, C.; Maussang, D.; Rovers, E.; Gallenne, T.; Hendriks, L.J.A.; Visser, T.; Nijhuis, R.; Logtenberg, T.; Kruif, J.; et al. Unbiased Combinatorial Screening Identifies a Bispecific IgG1 that Potently Inhibits HER3 Signaling via HER2-Guided Ligand Blockade. Cancer Cell 2018, 33, 922–936.e10. [Google Scholar] [CrossRef]
  74. Hamilton, E.P.; Petit, T.; Pistilli, B.; Goncalves, A.; Ferreira, A.; Dalenc, F.; Cardoso, F.; Mita, M.M.; Dezentjé, V.O.; Manso, L.; et al. Clinical activity of MCLA-128 (zenocutuzumab), trastuzumab, and vinorelbine in HER2 amplified metastatic breast cancer (MBC) patients (pts) who had progressed on anti-HER2 ADCs. J. Clin. Oncol. 2020, 38, 3093. [Google Scholar] [CrossRef]
  75. Schram, A.M.; Goto, K.; Kim, D.; Martin-Romano, P.; Ou, S.I.; O’Kane, G.M.; O’Reilly, E.M.; Umemoto, K.; Duruisseaux, M.; Neuzillet, C.; et al. Efficacy and safety of zenocutuzumab, a HER2 x HER3 bispecific antibody, in advanced NRG1 fusion-positive (NRG1+) cancer. J. Clin. Oncol. 2022, 40, 105. [Google Scholar] [CrossRef]
  76. Deng, W.; Liu, J.; Pan, H.; Li, L.; Zhou, C.; Wang, X.; Shu, R.; Dong, B.; Cao, D.; Li, Q.; et al. A Bispecific Antibody Based on Pertuzumab Fab Has Potent Antitumor Activity. J. Immunother. 2018, 41, 1–8. [Google Scholar] [CrossRef] [PubMed]
  77. Oberg, H.H.; Kellner, C.; Gonnermann, D.; Sebens, S.; Bauerschlag, D.; Gramatzki, M.; Kabelitz, D.; Peipp, M.; Wesch, D. Tribody [(HER2)(2)xCD16] Is More Effective Than Trastuzumab in Enhancing gammadelta T Cell and Natural Killer Cell Cytotoxicity Against HER2-Expressing Cancer Cells. Front. Immunol. 2018, 9, 814. [Google Scholar] [CrossRef] [PubMed]
  78. Zhang, H.; Lam, L.; Nagai, Y.; Zhu, Z.; Chen, X.; Ji, M.Q.; Greene, M.I. A targeted immunotherapy approach for HER2/neu transformed tumors by coupling an engineered effector domain with interferon-gamma. Oncoimmunology 2018, 7, e1300739. [Google Scholar] [CrossRef] [PubMed]
  79. McDonagh, C.F.; Huhalov, A.; Harms, B.D.; Adams, S.; Paragas, V.; Oyama, S.; Zhang, B.; Luss, L.; Overland, R.; Nguyen, S.; et al. Antitumor activity of a novel bispecific antibody that targets the ErbB2/ErbB3 oncogenic unit and inhibits heregulin-induced activation of ErbB3. Mol. Cancer Ther. 2012, 11, 582–593. [Google Scholar] [CrossRef]
  80. Kiewe, P.; Hasmuller, S.; Kahlert, S.; Heinrigs, M.; Rack, B.; Marme, A.; Korfel, A.; Jager, M.; Lindhofer, H.; Sommer, H.; et al. Phase I trial of the trifunctional anti-HER2 x anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin. Cancer Res. 2006, 12, 3085–3091. [Google Scholar] [CrossRef]
  81. Jager, M.; Schoberth, A.; Ruf, P.; Hess, J.; Lindhofer, H. The trifunctional antibody ertumaxomab destroys tumor cells that express low levels of human epidermal growth factor receptor 2. Cancer Res. 2009, 69, 4270–4276. [Google Scholar] [CrossRef]
  82. Wermke, M.; Alt, J.; Kauh, J.; Back, J.; Salhi, Y.; Reddy, V.; Bayever, E.; Ochsenreither, S. Preliminary biomarker and pharmacodynamic data from a phase I study of single-agent bispecific antibody T-cell engager GBR 1302 in subjects with HER-2 positive cancers. J. Clin. Oncol. 2018, 36, 69. [Google Scholar] [CrossRef]
  83. Kim, H.J.; Kim, H.P.; Yoon, Y.K.; Kim, M.S.; Lee, G.S.; Han, S.W.; Im, S.A.; Kim, T.Y.; Oh, D.Y.; Bang, Y.J. Antitumor activity of HM781-36B, a pan-HER tyrosine kinase inhibitor, in HER2-amplified breast cancer cells. Anticancer Drugs 2012, 23, 288–297. [Google Scholar] [CrossRef]
  84. Robichaux, J.P.; Elamin, Y.Y.; Vijayan, R.S.K.; Nilsson, M.B.; Hu, L.; He, J.; He, J.; Zhang, F.; Pisegna, M.; Poteete, A.; et al. Pan-Cancer Landscape and Analysis of ERBB2 Mutations Identifies Poziotinib as a Clinically Active Inhibitor and Enhancer of T-DM1 Activity. Cancer Cell 2020, 37, 420. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, T.M.; Lee, K.W.; Oh, D.Y.; Lee, J.S.; Im, S.A.; Kim, D.W.; Han, S.W.; Kim, Y.J.; Kim, T.Y.; Kim, J.H.; et al. Phase 1 Studies of Poziotinib, an Irreversible Pan-HER Tyrosine Kinase Inhibitor in Patients with Advanced Solid Tumors. Cancer Res. Treat. 2018, 50, 835–842. [Google Scholar] [CrossRef] [PubMed]
  86. Park, Y.H.; Lee, K.H.; Sohn, J.H.; Lee, K.S.; Jung, K.H.; Kim, J.H.; Lee, K.H.; Ahn, J.S.; Kim, T.Y.; Kim, G.M.; et al. A phase II trial of the pan-HER inhibitor poziotinib, in patients with HER-2 positive metastatic breast cancer who had received at least two prior HER2-directed regimens: Results of the NOV120101-203 trial. Int. J. Cancer 2018, 143, 3240–3247. [Google Scholar] [CrossRef] [PubMed]
  87. Park, Y.H.; Jung, K.H.; Sohn, J.; Lee, K.S.; Kim, J.H.; Yang, Y.; Beak, E.; Han, H.; Im, S.-A. Poziotinib for HER-2 positive metastatic breast cancer (MBC): Final clinical efficacy and safety results for long-term follow-up of the phase II NOV120101-203 trial. In Annals of Oncology, Proceedings of the ESMO Congress 2022, Berlin, Germany, 3–5 May 2022; Elsevier: Amsterdam, The Netherlands, 2022; pp. S207–S208. [Google Scholar]
  88. Zhang, J.; McAndrew, N.P.; Wang, X.; Du, Y.; DiCarlo, B.; Wang, M.; Chen, K.; Yu, W.; Hu, X. Preclinical and clinical activity of DZD1516, a full blood-brain barrier-penetrant, highly selective HER2 inhibitor. Breast Cancer Res. 2023, 25, 81. [Google Scholar] [CrossRef] [PubMed]
  89. Hu, X.; Zhang, J.; McAndrew, N.P. Early Clinical Safety and Pharmacokinetics Data of DZD1516, an BBB-Penetrant Selective HER2 Inhibitor for the Treatment of HER-2 positive Metastatic Breast Cancer. J. Clin. Oncol. 2022, 40, 1038. [Google Scholar] [CrossRef]
  90. Tanaka, H.; Hirata, M.; Shinonome, S.; Wada, T.; Iguchi, M.; Dohi, K.; Inoue, M.; Ishioka, Y.; Hojo, K.; Yamada, T.; et al. Preclinical antitumor activity of S-222611, an oral reversible tyrosine kinase inhibitor of epidermal growth factor receptor and human epidermal growth factor receptor 2. Cancer Sci. 2014, 105, 1040–1048. [Google Scholar] [CrossRef] [PubMed]
  91. Spicer, J.; Baird, R.; Suder, A.; Cresti, N.; Corbacho, J.G.; Hogarth, L.; Frenkel, E.; Matsumoto, S.; Kawabata, I.; Donaldson, K.; et al. Phase 1 dose-escalation study of S-222611, an oral reversible dual tyrosine kinase inhibitor of EGFR and HER2, in patients with solid tumours. Eur. J. Cancer 2015, 51, 137–145. [Google Scholar] [CrossRef]
  92. Macpherson, I.R.; Spiliopoulou, P.; Rafii, S.; Saggese, M.; Baird, R.D.; Garcia-Corbacho, J.; Italiano, A.; Bonneterre, J.; Campone, M.; Cresti, N.; et al. A phase I/II study of epertinib plus trastuzumab with or without chemotherapy in patients with HER-2 positive metastatic breast cancer. Breast Cancer Res. 2019, 22, 1. [Google Scholar] [CrossRef]
  93. Schram, A.M.; Ahnert, J.R.; Patel, M.R.; Jauhari, S.; Sachdev, J.C.; Zhu, V.W.; LoRusso, P.; Nguyen, D.; Le, X.; O’Connor, M.; et al. Safety and preliminary efficacy from the phase 1 portion of MasterKey-01: A First-in-human dose-escalation study to determine the recommended phase 2 dose (RP2D), pharmacokinetics (PK) and preliminary antitumor activity of BDTX-189, an inhibitor of allosteric ErbB mutations, in patients (pts) with advanced solid malignancies. J. Clin. Oncol. 2021, 39, 3086. [Google Scholar]
  94. Banerji, U.; van Herpen, C.M.L.; Saura, C.; Thistlethwaite, F.; Lord, S.; Moreno, V.; Macpherson, I.R.; Boni, V.; Rolfo, C.; de Vries, E.G.E.; et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: A phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019, 20, 1124–1135. [Google Scholar] [CrossRef]
  95. Dokter, W.; Ubink, R.; van der Lee, M.; van der Vleuten, M.; van Achterberg, T.; Jacobs, D.; Loosveld, E.; van den Dobbelsteen, D.; Egging, D.; Mattaar, E.; et al. Preclinical profile of the HER2-targeting ADC SYD983/SYD985: Introduction of a new duocarmycin-based linker-drug platform. Mol. Cancer Ther. 2014, 13, 2618–2629. [Google Scholar] [CrossRef] [PubMed]
  96. Manich, C.S.; O’Shaughnessy, J.; Aftimos, P.G.; van den Tweel, E.; Oesterholt, M.; Escrivá-de-Romaní, S.I.; Tueux, N.Q.; Tan, T.J.; Lim, J.S.; Ladoire, S.; et al. Primary outcome of the phase III SYD985.002/TULIP trial comparing [vic-]trastuzumab duocarmazine to physician’s choice treatment in patients with pre-treated HER-2 positive locally advanced or metastatic breast cancer. Ann. Oncol. 2021, 32, 1283–1346. [Google Scholar]
  97. Zhang, T.; You, L.; Xu, J.; Yin, J.; Qu, B.; Mao, Y.; Fu, B.; Cao, D.; Zhao, L.; Feng, J.; et al. SHR-A1811, a novel anti-HER2 ADC with superior bystander effect, optimal DAR and favorable safety profiles. Cancer Res. 2023, 83 (Suppl. S8), LB031. [Google Scholar] [CrossRef]
  98. Yao, H.; Ryu, M.H.; Park, J.; Voskoboynik, M.; Kim, J.H.; Liu, K.; Barve, M.; Acuna-Villaorduna, A.; Im, S.-A.; Roy, A.C.; et al. 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. 2023, 34, S458–S497. [Google Scholar] [CrossRef]
  99. Skidmore, L.; Sakamuri, S.; Knudsen, N.A.; Hewet, A.G.; Milutinovic, S.; Barkho, W.; Biroc, S.L.; Kirtley, J.; Marsden, R.; Storey, K.; et al. ARX788, a Site-specific Anti-HER2 Antibody-Drug Conjugate, Demonstrates Potent and Selective Activity in HER-2 low and T-DM1-resistant Breast and Gastric Cancers. Mol. Cancer Ther. 2020, 19, 1833–1843. [Google Scholar] [CrossRef]
  100. Barok, M.; Le Joncour, V.; Martins, A.; Isola, J.; Salmikangas, M.; Laakkonen, P.; Joensuu, H. ARX788, a novel anti-HER2 antibody-drug conjugate, shows anti-tumor effects in preclinical models of trastuzumab emtansine-resistant HER-2 positive breast cancer and gastric cancer. Cancer Lett. 2020, 473, 156–163. [Google Scholar] [CrossRef]
  101. Zhang, J.; Ji, D.; Shen, W.; Xiao, Q.; Gu, Y.; O’Shaughnessy, J.; Hu, X. Phase I Trial of a Novel Anti-HER2 Antibody-Drug Conjugate, ARX788, for the Treatment of HER-2 positive Metastatic Breast Cancer. Clin. Cancer Res. 2022, 28, OF1–OF10. [Google Scholar]
  102. Shi, F.; Liu, Y.; Zhou, X.; Shen, P.; Xue, R.; Zhang, M. Disitamab vedotin: A novel antibody-drug conjugates for cancer therapy. Drug Deliv. 2022, 29, 1335–1344. [Google Scholar] [CrossRef]
  103. Yao, X.; Jiang, J.; Wang, X.; Huang, C.; Li, D.; Xie, K.; Xu, Q.; Li, H.; Li, Z.; Lou, L.; et al. A novel humanized anti-HER2 antibody conjugated with MMAE exerts potent anti-tumor activity. Breast Cancer Res. Treat 2015, 153, 123–133. [Google Scholar] [CrossRef]
  104. Deeks, E.D. Disitamab Vedotin: First Approval. Drugs 2021, 81, 1929–1935. [Google Scholar] [CrossRef]
  105. Hamblett, K.J.; Barnscher, S.D.; Davies, R.H.; Hammon, P.W.; Hernandez, A.; Wickman, G.R.; Fung, V.K.; Ding, T.; Garnett, G.; Galey, A.S.; et al. ZW49, a HER2 targeted biparatopic antibody drug conjugate for the treatment of HER2 expressing cancers. In Proceedings of the 2018 San Antonio Breast Cancer Symposium, San Antonio, TX, USA, 4–8 December 2018; Cancer Research: San Antonio, TX, USA, 2019. [Google Scholar]
  106. Jhaveri, K.; Dotan, E.; Oh, D.; Ferrario, C.; Tolcher, A.W.; Lee, K.; Liao, C.; Kang, Y.; Kim, Y.H.; Hamilton, E.P.; et al. Preliminary results from a phase I study using the bispecific, human epidermal growth factor 2 (HER2)-targeting antibody-drug conjugate (ADC) zanidatamab zovodotin (ZW49) in solid cancers. Ann. Oncol. 2022, 33, S197–S224. [Google Scholar] [CrossRef]
  107. Li, H.; Zhang, X.; Xu, Z.; Li, L.; Liu, W.; Dai, Z.; Zhao, Z.; Xiao, L.; Li, H.; Hu, C. Preclinical evaluation of MRG002, a novel HER2-targeting antibody-drug conjugate with potent antitumor activity against HER-2 positive solid tumors. Antib. Ther. 2021, 4, 175–184. [Google Scholar] [PubMed]
  108. Zhang, J.; Liu, R.; Gao, S.; Li, W.; Chen, Y.; Meng, Y.; Liu, C.; Jin, W.; Wu, J.; Wang, Y.; et al. Phase I study of A166, an antibody–drug conjugate in advanced HER2-expressing solid tumours. NPJ Breast Cancer 2023, 9, 28. [Google Scholar] [CrossRef] [PubMed]
  109. Rinnerthaler, G.; Gampenrieder, S.P.; Greil, R. HER2 Directed Antibody-Drug-Conjugates beyond T-DM1 in Breast Cancer. Int. J. Mol. Sci. 2019, 20, 1115. [Google Scholar] [CrossRef] [PubMed]
  110. Park, Y.H.; Kim, J.-Y.; Ahn, J.S.; Im, Y.-H.; Kim, S.-H.; Lee, S.; Chung, Y.S.; Park, S.J. First-in-human phase I study of ALT-P7, a HER2-targeting antibody-drug conjugate in patients with HER-2 positive advanced breast cancer. J. Clin. Oncol. 2020, 38, 3551. [Google Scholar] [CrossRef]
  111. Yurkovetskiy, A.; Ter-Ovanesyan, E.; Conlon, P.; Devit, M.; Bu, C.; Bodyak, N.; Lowinger, T.; Bergstrom, D. Non-clinical pharmacokinetics of XMT-1522, a HER2 targeting auristatin-based antibody drug conjugate. Cancer Res. 2017, 77 (Suppl. S13), 48. [Google Scholar] [CrossRef]
  112. Le Joncour, V.; Martins, A.; Puhka, M.; Isola, J.; Salmikangas, M.; Laakkonen, P.; Joensuu, H.; Barok, M. A Novel Anti-HER2 Antibody-Drug Conjugate XMT-1522 for HER-2 positive Breast and Gastric Cancers Resistant to Trastuzumab Emtansine. Mol. Cancer Ther. 2019, 18, 1721–1730. [Google Scholar] [CrossRef] [PubMed]
  113. Bodyak, N.; Park, P.; Gumerov, D.; DeVit, M.; Yin, M.; Thomas, J.D.; Qin, L.; Lowinger, T.B.; Bergstrom, D.A. Trastuzumab-Dolaflexin, a Highly Potent Fleximer-Based Antibody-Drug Conjugate, Demonstrates a Favorable Therapeutic Index in Exploratory Toxicology Studies in Multiple Species. Cancer Res. 2015, 75, 641. [Google Scholar] [CrossRef]
  114. Traore, T.; Khattar, M. Synergy of an anti-HER2 ADC TAK-522 (XMT-1522) in combination with anti-PD1 monoclonal antibody (mAb) in a syngeneic breast cancer model expressing human HER2. J. Immunol. 2018, 200, 122–129. [Google Scholar] [CrossRef]
  115. Hamilton, E.P.; Bardia, A.; Beeram, M.; Bendell, J.; Mosher, R.; Hailman, E.; Bergstrom, D.; Burris, H.; Soliman, H. Phase 1 dose escalation of XMT-1522, a novel HER2-targeting antibody-drug conjugate (ADC), in patients (pts) with HER2-expressing breast, lung and gastric tumors. J. Clin. Oncol. 2018, 36, 2546. [Google Scholar] [CrossRef]
  116. Meric-Bernstam, F.; Calvo, E.; Moreno, V.; Chung, H.C.; Park, Y.H.; Bang, Y.-J.; Rosen, L.S.; Mita, M.M.; Garrido-Laguna, I.; Leung, A.C.F.; et al. A phase I dose escalation study evaluating the safety and tolerability of a novel anti-HER2 antibody-drug conjugate (PF-06804103) in patients with HER-2 positive solid tumors. J. Oncol. 2020, 38, 1039. [Google Scholar] [CrossRef]
  117. Graziani, E.I.; Sung, M.; Ma, D.; Narayanan, B.; Marquette, K.; Puthenveetil, S.; Tumey, L.N.; Bikker, J.; Casavant, J.; Bennett, E.M.; et al. PF-06804103, A Site-specific Anti-HER2 Antibody-Drug Conjugate for the Treatment of HER2-expressing Breast, Gastric, and Lung Cancers. Mol. Cancer Ther. 2020, 19, 2068–2078. [Google Scholar] [CrossRef] [PubMed]
  118. Meric-Bernstam, F.; Calvo, E.; Lee, K.S.; Moreno, V.; Park, Y.H.; Rha, S.Y.; Chalasani, P.; Zhong, W.; Zhou, L.; Pirie-Shepherd, S.; et al. Safety and Tolerability of a Novel Anti-HER2 Antibody-Drug Conjugate (PF-06804103) in Patients with HER2-Expressing Solid Tumors: A Phase 1 Dose-Escalation Study. Mol. Cancer Ther. 2023, 22, 1191–1203. [Google Scholar] [CrossRef] [PubMed]
  119. Kang, J.C.; Sun, W.; Khare, P.; Karimi, M.; Wang, X.; Shen, Y.; Ober, R.J.; Ward, E.S. Engineering a HER2-specific antibody-drug conjugate to increase lysosomal delivery and therapeutic efficacy. Nat. Biotechnol. 2019, 37, 523–526. [Google Scholar] [CrossRef] [PubMed]
  120. Oganesyan, V.; Peng, L.; Bee, J.S.; Li, J.; Perry, S.R.; Comer, F.; Xu, L.; Cook, K.; Senthil, K.; Clarke, L. Structural insights into the mechanism of action of a biparatopic anti-HER2 antibody. J. Biol. Chem. 2018, 293, 8439–8448. [Google Scholar] [CrossRef] [PubMed]
  121. Li, J.Y.; Perry, S.R.; Muniz-Medina, V.; Wang, X.; Wetzel, L.K.; Rebelatto, M.C.; Hinrichs, M.J.; Bezabeh, B.Z.; Fleming, R.L.; Dimasi, N.; et al. A Biparatopic HER2-Targeting Antibody-Drug Conjugate Induces Tumor Regression in Primary Models Refractory to or Ineligible for HER2-Targeted Therapy. Cancer Cell 2016, 29, 117–129. [Google Scholar] [CrossRef] [PubMed]
  122. Pegram, M.D.; Hamilton, E.P.; Tan, A.R.; Storniolo, A.M.; Balic, K.; Rosenbaum, A.I.; Liang, M.; He, P.; Marshall, S.; Scheuber, A.; et al. First-in-Human, Phase 1 Dose-Escalation Study of Biparatopic Anti-HER2 Antibody-Drug Conjugate MEDI4276 in Patients with HER-2 positive Advanced Breast or Gastric Cancer. Mol. Cancer Ther. 2021, 20, 1442–1453. [Google Scholar] [CrossRef] [PubMed]
  123. Phillips, G.; Li, G.; Guo, J.; Yu, S.F.; Lee, G.; Zhang, D.; Dragovich, P.S.; Pillow, T.; Wei, B.; Lee, M.V.; et al. Preclinical development of DHES0815A: A HER2-directed antibody-drug conjugate comprised of a reduced potency PBD dimer linked to a domain I binding HER2 antibody. In Proceedings of the San Antonio Breast Cancer Symposium 2021, San Antonio, TX, USA, 7–10 December 2021; Cancer Research: San Antonio, TX, USA, 2022. [Google Scholar]
  124. Krop, I.; Hamilton, E.; Jung, K.H.; Modi, S.; Kalinsky, K.M.; Phillips, G. A phase I dose-escalation study of DHES0815A, a HER2-targeting antibody-drug conjugate with a DNA monoalkylator payload, in patients with HER-2 positive breast cancer. In Proceedings of the San Antonio Breast Cancer Symposium 2021, San Antonio, TX, USA, 7–10 December 2021; Cancer Research: San Antonio, TX, USA, 2022. [Google Scholar]
  125. Pegram, M.; Rasco, D.; Spira, A.; Wang, D.; Weinberg, B.A.; Alonso, M.; Fang, L.; Husain, A.; Kowanetz, M.; Perez, E.A.; et al. Preliminary results from a phase I/II study of BDC-1001, a novel HER2 targeting TLR7/8 immune-stimulating antibody conjugate (ISAC), in patients (pts) with advanced HER2-expressing solid tumors. In Annals of Oncology, Proceedings of the ESMO Congress 2021, Online, 16–21 September 2021; Elsevier: Amsterdam, The Netherlands, 2021. [Google Scholar]
  126. Hagemann, U.B.; Ellingsen, C.; Schuhmacher, J.; Kristian, A.; Mobergslien, A.; Cruciani, V.; Wickstroem, K.; Schatz, C.A.; Kneip, C.; Golfier, S.; et al. Mesothelin-Targeted Thorium-227 Conjugate (MSLN-TTC): Preclinical Evaluation of a New Targeted Alpha Therapy for Mesothelin-Positive Cancers. Clin. Cancer Res. 2019, 25, 4723–4734. [Google Scholar] [CrossRef]
  127. Karlsson, J. HER2-TTC: A Targeted thorium conjugate to treat HER2 expressing cancers with potent alpha radiation [abstract]. Cancer Res. 2021, 81 (Suppl. S13), ND07. [Google Scholar] [CrossRef]
  128. Zheng, G.; Guo, Z.; Li, W.; Xi, W.; Zuo, B.; Zhang, R.; Wen, W.; Yang, A.G.; Jia, L. Interaction between HLA-G and NK cell receptor KIR2DL4 orchestrates HER-2 positive breast cancer resistance to trastuzumab. Signal Transduct. Target. Ther. 2021, 6, 236. [Google Scholar] [CrossRef]
  129. Yang, T.; Kang, L.; Li, D.; Song, Y. Immunotherapy for HER-2 positive breast cancer. Front. Oncol. 2023, 13, 1097983. [Google Scholar] [CrossRef] [PubMed]
  130. Loi, S.; Giobbie-Hurder, A.; Gombos, A.; Bachelot, T.; Hui, R.; Curigliano, G.; Campone, M.; Biganzoli, L.; Bonnefoi, H.; Jerusalem, G.; et al. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER-2 positive breast cancer (PANACEA): A single-arm, multicentre, phase 1b-2 trial. Lancet Oncol. 2019, 20, 371–382. [Google Scholar] [CrossRef] [PubMed]
  131. Huober, J.; Barrios, C.H.; Niikura, N.; Jarzab, M.; Chang, Y.C.; Huggins-Puhalla, S.L.; Pedrini, J.; Zhukova, L.; Graupner, V.; Eiger, D.; et al. Atezolizumab with Neoadjuvant Anti-Human Epidermal Growth Factor Receptor 2 Therapy and Chemotherapy in Human Epidermal Growth Factor Receptor 2-Positive Early Breast Cancer: Primary Results of the Randomized Phase III IMpassion050 Trial. J. Clin. Oncol. 2022, 40, 2946–2956. [Google Scholar] [CrossRef] [PubMed]
  132. Bekes, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200. [Google Scholar] [CrossRef]
  133. Maneiro, M.A.; Forte, N.; Shchepinova, M.M.; Kounde, C.S.; Chudasama, V.; Baker, J.R.; Tate, E.W. Antibody-PROTAC Conjugates Enable HER2-Dependent Targeted Protein Degradation of BRD4. ACS Chem. Biol. 2020, 15, 1306–1312. [Google Scholar] [CrossRef]
  134. Sloas, C.; Gill, S.; Klichinsky, M. Engineered CAR-Macrophages as Adoptive Immunotherapies for Solid Tumors. Front. Immunol. 2021, 12, 783305. [Google Scholar] [CrossRef]
  135. Marofi, F.; Al-Awad, A.S.; Sulaiman Rahman, H.; Markov, A.; Abdelbasset, W.K.; Ivanovna Enina, Y.; Mahmoodi, M.; Hassanzadeh, A.; Yazdanifar, M.; Stanley Chartrand, M.; et al. CAR-NK Cell: A New Paradigm in Tumor Immunotherapy. Front. Oncol. 2021, 11, 673276. [Google Scholar] [CrossRef]
  136. Al-Awadhi, A.; Lee Murray, J.; Ibrahim, N.K. Developing anti-HER2 vaccines: Breast cancer experience. Int. J. Cancer 2018, 143, 2126–2132. [Google Scholar] [CrossRef]
  137. Lollini, P.L.; Cavallo, F.; Nanni, P.; Forni, G. Vaccines for tumour prevention. Nat. Rev. Cancer 2006, 6, 204–216. [Google Scholar] [CrossRef]
  138. Handy, C.E.; Antonarakis, E.S. Sipuleucel-T for the treatment of prostate cancer: Novel insights and future directions. Future Oncol. 2018, 14, 907–917. [Google Scholar] [CrossRef]
  139. Dillon, P.M.; Brenin, C.M.; Slingluff, C.L., Jr. Evaluating Nelipepimut-S in the Treatment of Breast Cancer: A Short Report on the Emerging Data. Breast Cancer 2020, 12, 69–75. [Google Scholar] [CrossRef] [PubMed]
  140. Murray, J.L.; Gillogly, M.E.; Przepiorka, D.; Brewer, H.; Ibrahim, N.K.; Booser, D.J.; Hortobagyi, G.N.; Kudelka, A.P.; Grabstein, K.H.; Cheever, M.A.; et al. Toxicity, immunogenicity, and induction of E75-specific tumor-lytic CTLs by HER-2 peptide E75 (369–377) combined with granulocyte macrophage colony-stimulating factor in HLA-A2+ patients with metastatic breast and ovarian cancer. Clin. Cancer Res. 2002, 8, 3407–3418. [Google Scholar] [PubMed]
  141. Clifton, G.T.; Hale, D.; Vreeland, T.J.; Hickerson, A.T.; Litton, J.K.; Alatrash, G.; Murthy, R.K.; Qiao, N.; Philips, A.; Lukas, J.; et al. Results of a Randomized Phase IIb Trial of Nelipepimut-S + Trastuzumab versus Trastuzumab to Prevent Recurrences in Patients with High-Risk HER2 Low-Expressing Breast Cancer. Clin. Cancer Res. 2020, 26, 2515–2523. [Google Scholar] [CrossRef] [PubMed]
  142. Mittendorf, E.A.; Clifton, G.T.; Holmes, J.P.; Schneble, E.; van Echo, D.; Ponniah, S.; Peoples, G.E. Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients. Ann. Oncol. 2014, 25, 1735–1742. [Google Scholar] [CrossRef]
  143. Mittendorf, E.A.; Lu, B.; Melisko, M.; Price Hiller, J.; Bondarenko, I.; Brunt, A.M.; Sergii, G.; Petrakova, K.; Peoples, G.E. Efficacy and Safety Analysis of Nelipepimut-S Vaccine to Prevent Breast Cancer Recurrence: A Randomized, Multicenter, Phase III Clinical Trial. Clin. Cancer Res. 2019, 25, 4248–4254. [Google Scholar] [CrossRef]
  144. Patel, S.; McWilliams, D.; Thompson, J.; Patel, M.; Daugherty, F.J. Evaluation of booster injections in maintaining peak immunity in a phase IIb study evaluating HER2/neu peptide GP2 (GLSI-100) versus GM-CSF alone after adjuvant trastuzumab in women with HER2-positive breast cancer [abstract]. 2022 ASCO Annual Meeting. J. Clin. Oncol. 2022, 40, LBA550. [Google Scholar] [CrossRef]
Table 5. Summary of ongoing or recent clinical trials involving monoclonal antibodies (mAb) and bispecific antibodies (bsAb).
Table 5. Summary of ongoing or recent clinical trials involving monoclonal antibodies (mAb) and bispecific antibodies (bsAb).
mAb/bsAbOngoing Clinical Trials (Phase)PopulationTreatment in StudyStatus
MM-302None---
InetetamabNCT05749016 (II)
NCT05823623 (II)
NCT04681911 (II)
NCT05823623 (II)
NCT05621434 (II)
NCT04963595 (II)
HER-2+ EBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
Neoadj inetetamab +P +ChT
Inetetamab +pyrotinib +ChT
Inetetamab +pyrotinib +ChT
Inetetamab +pyrotinib +ChT
Inetetamab +pyrotinib +ChT in 1L
Inetetamab +pyrotinib +ChT in 1L
Recruiting
Recruiting
Recruiting
Recruiting
Recruiting
Not yet recruiting
Zanidatamab (ZW25)NCT05027139 (I)
NCT04224272 (II)
NCT05027139 (Ib/II)
NCT05035836 (II)
NCT01042379 (II)
HER-2+ MBC
HER-2+/HR+ MBC
HER-2+ MBC
HER-2+ EBC
HER-2+ EBC
ZW25
ZW25 + palbociclib+fulvestrant
ZW25 +ALX148
Neoadj ZW25
Neoadj ZW25
Concluded; unpublished
Concluded; unpublished
Recruiting
Recruiting
Recruiting
MBS301NCT03842085 (I)HER-2+ MBCMBS301Recruiting
Anbenitamab (KN026)NCT03847168 (I)
NCT04165993 (II)
NCT04881929 (II)
HER-2+ MBC
HER-2+/low MBC
HER-2+ EBC
KN026
KN026 ±ChT
Neoadj KN026 +ChT
Concluded; unpublished
Active, not recruiting
Recruiting
Zenocutuzumab (MCLA-128)NCT02912949 (I)
NCT03321981 (II)
HER-2+
HER-2+/HER-2-low/HR+ MBC
MCLA-128
MCLA-128 +T+ChT/+ET
Published
Active, not recruiting
HER(Per)-S-FabNone---
HER2-2XCD16None---
Adj: Adjuvant; ChT: Chemotherapy; EBC: Early breast cancer; MBC: Metastatic breast cancer; Neoadj: Neoadjuvant; P: Pertuzumab; T: Trastuzumab.
Table 6. Summary of ongoing or recent clinical trials involving tyrosine kinase inhibitors (TKIs).
Table 6. Summary of ongoing or recent clinical trials involving tyrosine kinase inhibitors (TKIs).
TKITargetEffectClinical Trials (Phase)PopulationStatus
PoziotinibPan-HER (HER-1,2,4)IrreversibleNCT02418689 (II)
NCT02659514 (II)
NCT03429101 (Ib)
NCT02544997 (II)
HER2+ MBC
HER+ MBC
HER2+ MBC
HER2m MBC
Published
Completed; unpublished
Completed; unpublished
Completed; unpublished
DZD1516HER-2ReversibleNCT04509596 (I)HER2+ MBCOngoing
MBC: Metastatic breast cancer; HER2m: HER-2-mutated; HER2+: HER-2 positive.
Table 7. Summary of antibody–drug conjugates (ADCs) and other antibody conjugates currently in study.
Table 7. Summary of antibody–drug conjugates (ADCs) and other antibody conjugates currently in study.
ADC/Antibody
Conjugates
PayloadAntibody-to-Drug RatioLinkerClinical Trial (Phase)SettingStatus
Trastuzumab
duocarmycin (SYD985)
Seco-DUBA2.8:1CleavableNCT03262935 (III)
NCT01042379 (II)
HER-2+ MBC in ≥3L
HER-2+ EBC (neoadj)
Closed; unpublished
Recruiting
Trastuzumab rezetecan (SHR-A1811)SHR92655.7:1CleavableNCT06057610 (III)
NCT05814354 (III)
NCT05845138 (I/II)
NCT05792410 (Ib/II)
NCT05635487 (II)
HER-2+ MBC
HER-2-low MBC
HER-2-low MBC
HER-2-low MBC
HER-2+ EBC (neoadj)
Recruiting
Recruiting
Recruiting
Recruiting
Recruiting
ARX788MMAF1.9:1Non-
cleavable
NCT04829604 (II)
NCT04983121 (II)
NCT05426486 (II/III)
NCT01042379 (II)
HER-2+ MBC
HER-2+ EBC (neoadj)
HER-2+ EBC (neoadj)
HER-2+ EBC (neoadj)
Recruiting
Recruiting
Recruiting
Recruiting
Disitamab vedotin
(RC48)
MMAE4:1CleavableNCT03052634 (I/II)
NCT05331326 (II)
NCT03500380 (II/III)
NCT04400695 (III)
NCT05904964 (III)
NCT06105008 (II)
NCT05134519 (II)
NCT05726175 (II)
HER-2+/low MBC
HER-2+ MBC
HER-2+ MBC
HER-2-low MBC
HR+/HER-2-low MBC
HR+/HER-2-low MBC
HER-2+ EBC (neoadj)
HER-2+ EBC (neoadj)
Closed; unpublished
Recruiting
Recruiting
Recruiting
Recruiting
Not yet recruiting
Not yet recruiting
Not yet recruiting
Zanidatamab zovodotin (ZW49)Zovodotin2:1CleavableNCT03821233 (I)HER-2+ solid tumorsClosed; unpublished
MRG002MMAE3.8:1CleavableNCT05263869 (II)
NCT04924699 (II/III)
NCT04742153 (II)
HER-2+ MBC
HER-2+ MBC
HER-2-low MBC
Recruiting
Recruiting
Recruiting
A166Duostatin-52:1CleavableNCT03602079 (I/II)
NCT05311397 (I)
HER-2+ solid tumorsPublished
Recruiting
ALT-P7MMAE2:1CleavableNCT03281824 (I)HER-2+ MBCClosed; unpublished
XMT-1522AF-HPA12:1CleavableNCT02952729 (I)HER-2+ MBCClosed; unpublished
BAY2701439Thorium-227UnknownUnknownNCT04147819 (I)HER-2+ solid tumorsClosed; unpublished
BDC-1001TLR-7/8 agonistUnknownNon-
cleavable
NCT04278144 (II)HER-2+ solid tumors
Metastatic HER-2+ BC
Recruiting
Recruiting
Adj: Adjuvant; EBC: Early breast cancer; MBC: Metastatic breast cancer; Neoadj: Neoadjuvant.
Table 8. Summary of ongoing clinical trials testing combinations of HER-2-targeted agents with other targeted agents.
Table 8. Summary of ongoing clinical trials testing combinations of HER-2-targeted agents with other targeted agents.
Combination AgentClinical Trials (Phase)PopulationTreatment in StudyStatus
CDK4/6 inhibitorsNCT05577442 (II)
NCT05969184 (II)
NCT03304080 (I/II)
NCT02448420 (II)
NCT03530696 (II)
NCT03054363 (II)
NCT05076695 (II)
NCT05076695 (II)
NCT04858516 (II)
NCT03913234 (Ib/II)
NCT05319873 (I/II)
NCT02657343 (I/II)
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ EBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
HR+/HER-2+ MBC
Dalpiciclib +pyrotinib +ET
Palbociclib + T +P +ET
Palbociclib +T +P +ET
Palbociclib +T +ET
Palbociclib +T-DM1
Palbociclib +tucatinib +ET
Palbociclib +pyrotinib +T +F
Neoadj palbociclib +pyrotinib +T +F
Neoadj palbociclib +pyrotinib +T +ET
Ribociclib +T +ET
Ribociclib +tucatinib +T
Ribociclib +T/T-DM1
Not yet recruiting
Recruiting
Active, not recruiting
Active, not recruiting
Completed; unpublished
Completed; unpublished
Recruiting
Recruiting
Not yet recruiting
Recruiting
Recruiting
Completed; unpublished
PARP inhibitorsNCT03368729 (I/II)HER-2+ mBRCA MBCNiraparib +TRecruiting
PIK3CA inhibitorsNCT03765983 (II)
NCT04108858 (I/II)
NCT02705859 (I)
NCT05230810 (I/II)
NCT04208178 (III)
NCT05063786 (III)
HER-2+ mPIK3CA MBC
HER-2+ mPIK3CA MBC
HER-2+ mPIK3CA MBC
HER-2+ mPIK3CA MBC
HER-2+ mPIK3CA MBC
HER-2+ mPIK3CA MBC
GDC-0084 +T
Copanlisib +T +P
Copanlisib +T
Alpelisib +tucatinib
Alpelisib +P +T in 1L
Alpelisib +T ±F
Recruiting
Recruiting
Completed
Recruiting
Active, not recruiting
Recruiting
ATK inhibitorsNCT04253561 (I)HER-2+ mAKT MBCIpatasertib +T +PRecruiting
Adj: Adjuvant; Neoadj: Neoadjuvant; ChT: Chemotherapy; EBC: Early breast cancer; ET: Endocrine therapy; F: fulvestrant; HR: Hormone receptors; MBC Metastatic breast cancer; mAKT: AKT mutated; mBRCA: BRCA mutated; mPIK3CA: PIK3CA mutated; P: Pertuzumab; T: Trastuzumab.
Table 9. Summary of ongoing clinical trials evaluating the combination of HER-2-targeted agents with immune-checkpoint inhibitors (ICI).
Table 9. Summary of ongoing clinical trials evaluating the combination of HER-2-targeted agents with immune-checkpoint inhibitors (ICI).
Combination AgentClinical Trials (Phase)PopulationTreatment in StudyStatus
AtezolizumabNCT03199885 (III)
NCT03125928 (II)
NCT04759248 (II)
NCT04740918 (III)
NCT04873362 (III)
NCT03595592 (III)
HER-2+ MBC
HER-2+ MBC
HER-2+ MBC
HER-2+ PD-L1+ MBC
HER-2+ EBC
HER-2+ EBC
Atezolizumab +T +P +ChT
Atezolizumab +T +P +ChT
Atezolizumab +T +ChT
Atezolizumab +T-DM1
Adj atezolizumab +T-DM1
Neoadj atezolizumab +T +P
Active, not recruiting
Active, not recruiting
Recruiting
Recruiting
Recruiting
Active, not recruiting
DurvalumabNCT03820141 (II)
NCT05795101 (II)
NCT02649686 (I)
HER-2+ MBC
HER-2+/low MBC
HER-2+ MBC
Durvalumab +T +P in 1L
Durvalumab +T-DXd
Durvalumab +T
Recruiting
Recruiting
Completed
PembrolizumabNCT04789096 (II)
NCT03032107 (Ib)
NCT03988036 (II)
NCT03747120 (II)
HER-2+ MBC
HER-2+ MBC
HER-2+ EBC
HER-2+ EBC
Pembrolizumab +tucatinib +T ±ChT
Pembrolizumab +T-DM1
Neoadj pembrolizumab +T +P
Neoadj pembrolizumab +T +P +ChT
Recruiting
Active, not recruiting
Completed
Recruiting
Adj: Adjuvant; Neoadj: Neoadjuvant; ChT: Chemotherapy; EBC: Early breast cancer; ET: Endocrine therapy; MBC Metastatic breast cancer; P: Pertuzumab; T: Trastuzumab; 1L: First line.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fernandes, C.L.; Silva, D.J.; Mesquita, A. Novel HER-2 Targeted Therapies in Breast Cancer. Cancers 2024, 16, 87. https://doi.org/10.3390/cancers16010087

AMA Style

Fernandes CL, Silva DJ, Mesquita A. Novel HER-2 Targeted Therapies in Breast Cancer. Cancers. 2024; 16(1):87. https://doi.org/10.3390/cancers16010087

Chicago/Turabian Style

Fernandes, Catarina Lopes, Diogo J. Silva, and Alexandra Mesquita. 2024. "Novel HER-2 Targeted Therapies in Breast Cancer" Cancers 16, no. 1: 87. https://doi.org/10.3390/cancers16010087

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop