Next Article in Journal
Exhaled Nitric Oxide as Biomarker of Type 2 Diseases
Previous Article in Journal
Plasma-Derived Exosome Proteins as Novel Diagnostic and Prognostic Biomarkers in Neuroblastoma Patients
Previous Article in Special Issue
Physical Training Chronically Stimulates the Motor Neuron Cell Nucleus in the Ts65Dn Mouse, a Model of Down Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer

Department of Medicine, University of California, San Francisco, CA 94158, USA
Cells 2023, 12(21), 2517; https://doi.org/10.3390/cells12212517
Submission received: 24 August 2023 / Revised: 14 October 2023 / Accepted: 20 October 2023 / Published: 25 October 2023
(This article belongs to the Collection Feature Papers in Cell Nuclei: Function, Transport and Receptors)

Abstract

:
Human epidermal growth factor receptor 3 (HER3) is the only family member of the EGRF/HER family of receptor tyrosine kinases that lacks an active kinase domain (KD), which makes it an obligate binding partner with other receptors for its oncogenic role. When HER3 is activated in a ligand-dependent (NRG1/HRG) or independent manner, it can bind to other receptors (the most potent binding partner is HER2) to regulate many biological functions (growth, survival, nutrient sensing, metabolic regulation, etc.) through the PI3K–AKT–mTOR pathway. HER3 has been found to promote tumorigenesis, tumor growth, and drug resistance in different cancer types, especially breast and non-small cell lung cancer. Given its ubiquitous expression across different solid tumors and role in oncogenesis and drug resistance, there has been a long effort to target HER3. As HER3 cannot be targeted through its KD with small-molecule kinase inhibitors via the conventional method, pharmaceutical companies have used various other approaches, including blocking either the ligand-binding domain or extracellular domain for dimerization with other receptors. The development of treatment options with anti-HER3 monoclonal antibodies, bispecific antibodies, and different combination therapies showed limited clinical efficiency for various reasons. Recent reports showed that the extracellular domain of HER3 is not required for its binding with other receptors, which raises doubt about the efforts and applicability of the development of the HER3-antibodies for treatment. Whereas HER3-directed antibody–drug conjugates showed potentiality for treatment, these drugs are still under clinical trial. The currently understood model for dimerization-induced signaling remains incomplete due to the absence of the crystal structure of HER3 signaling complexes, and many lines of evidence suggest that HER family signaling involves more than the interaction of two members. This review article will significantly expand our knowledge of HER3 signaling and shed light on developing a new generation of drugs that have fewer side effects than the current treatment regimen for these patients.

1. Introduction

Although after 2020, the diagnosis and treatment of cancer were adversely affected due to the coronavirus disease 2019 (COVID-19) pandemic, in 2022, 1,918,030 new cancer cases and 609,360 cancer deaths are projected in the United States [1]. Despite the enormous efforts in advancing anti-cancer agents, it is still challenging to treat cancer due to the development of drug resistance. Accumulation of mutations in solid tumors imposes even more pharmacological challenges. The ability of tumor cells to survive, grow, migrate, and invade depends on the interaction of cell surface receptors and many growth factors [2]. Some of these growth factors were found to bind with cell surface localized receptor tyrosine kinases (RTKs), as shown in Figure 1. RTKs have been an attractive target for developing anti-cancer agents due to their accessibility and the ability to block the catalytic kinase function using small molecules [3,4,5]. There are four types of RTKs: human epidermal growth factor receptor (HER) 1 (EGFR, ErbB1), HER2 (Neu, ErbB2), HER3 (ErbB3), and HER4 (ErbB4). These RTKs are generally expressed in epithelial, mesenchymal, and neuronal tissues and are found to regulate cell division, proliferation, and differentiation [6,7]. Among these four members of RTKs, EGFR and HER2 are the most studied targeted molecules in cancer therapy [8].
Although HER3 has long been underestimated for targeting, recent studies found its emerging role in oncogenesis, tumor progression, and drug resistance [9]. HER3 is a unique family member of HER family proteins for many reasons. Unlike other HER family members, HER3 cannot form a homodimer, lacks/almost no intracellular kinase activity, and can form a heterodimer with other non-HER family proteins [10,11,12,13]. When HER3 is activated via binding with other receptors, it primarily activates PI3K/Akt signaling [14,15]. Additionally, HER3 was also reported to activate the MAPK cascade, Janus kinase (JAK), and proto-oncogene c-Src (SRC) signaling pathways, all of these pathways are tumorigenic [16,17]. It is now well recognized that HER3 can restore the signaling function via the PI3K/Akt axis under EGFR-targeted inhibition [18].
Even though it is now recognized that HER3 is a prime target for therapy, all the drugs available in the market are targeted against only two members of the HER/ERBB family proteins, namely EGFR and HER2. As it is impossible to target HER3 via conventional methods through inhibition of kinase activity, many pharmaceutical companies have been trying to develop different agents, including monoclonal antibodies and drug conjugates. Nonetheless, many drugs are developed targeting HER3 and are currently in various stages of clinical development. Interestingly, some studies also suggest that the extracellular domain of HER3 is not required for signaling, which raises an important question about the applicability of that specific therapy that targets the extracellular domain of HER3 [19].
It is suggested that signal generation in the HER3 occurs through an asymmetric kinase dimerization, where one kinase allosterically activates the other kinase [20]. Although it has been assumed that signaling and oncogenic functions of HER3 involve heterodimerization with another receptor, no crystal structure has been developed for a fully active HER3 signaling complex, so it is still known whether HER3 forms a dimer or there are more than two binding partners involved for this interaction. In fact, higher-order oligomerization is the widely believed mode of signaling in the HER family. The involvement of oligomeric assemblies in HER family signaling has been suggested by several studies involving fluorescent tracing and other techniques [21,22,23,24,25,26,27]. However, the interfaces involved in these interactions and their functional relevance remain to be defined. Hence, it is essential to critically review the 30 years of bench-to-bedside research to develop novel pharmacological interceptors of HER3. Such understanding can lay the foundation for a newer generation of drugs that can significantly enhance the efficacy of TKIs and prove highly effective without the need for cytotoxic agents.

2. HER3 Structure and Mechanism of Activation

HER3 was discovered by Kraus et al. in 1989, and it maps to chromosome 12q13 [28]. HER3 protein mainly has five domains: the extracellular domain, transmembrane domain, juxta membrane domain, kinase domain, and C-terminal tail, as shown in Figure 2. The extracellular domain consists of four subdomains, from domain I to domain IV. Upon ligand binding, a structural conformational change occurs that converts an untethered/inactive form to a tethered/active form, which enables it for heterodimerization with other HER receptors. Although the HER3 kinase domain (KD) shares 83% amino acid sequence identity with both EGFR and HER2, the presence of specific nonconservative amino acid residues (Cys-721, His-740, and Asn-815) in the HER3 KD makes its KD inactive [29].
Similar to other receptor tyrosine kinases (RTKs), the extracellular domain of HER3 exists in a reversible equilibrium between open (active) and closed (inactive) conformations. HER3 is active when this equilibrium shifts towards open conformation, allowing its dimerization arm within domain II to be exposed for dimerization (through the cysteine-rich CR1 region). So, when HER3 is in the open conform, it can dimerize with another HER family receptor. HER3 is the only family member of the HER family that does not form a homodimer. HER3 activation can occur in a ligand-dependent or ligand-independent manner. Conventionally, HER3 becomes activated via binding of its ligand NRG1 (also known as HRG) to the extracellular domain of HER3, which exposes its dimerization interface to be available for binding with other receptors. However, when any of its dimerization partners are present at sufficient concentration, it stabilizes HER3 in the open conformation transiently, known as ligand-independent activation.
Signal generation in the HER family occurs through an asymmetric kinase dimerization, where one kinase allosterically activates the other kinase. In the case of the HER2-HER3 dimer, HER2 acts as a receiver kinase, phosphorylating tyrosine residues on the C-terminal tails of the activator kinases, i.e., HER3. After ligand binding, HER3 heterodimerizes with other HER family proteins that cross the phosphorylate C-terminal tail of HER3. This transphosphorylation at the HER3 C-terminal tail creates docking sites for p85, a regulatory subunit of protein phosphatidylinositol 3-kinase (PI3K) [30]. Studies reported that six tyrosine residues of the HER3 C-terminal tail are phosphorylated to recruit and activate PI3K [15,31]. Once activated, PI3K phosphorylates phosphatidylinositol bisphosphate (PIP2) and is converted to phosphatidylinositol trisphosphate (PIP3) in the plasma membrane [32]. PKB/Akt binds to PIP3, allowing PDK1 to phosphorylate the T308 residue of Akt to activate it [32]. The PI3K/Akt pathway also activates the mammalian target of rapamycin (mTOR) to control various biological processes, including survival, translation, nutrient sensing, cell cycle control, and metabolic regulation [32].

3. Expressions of HER3 in Different Types of Cancers

HER3 expression and activation levels go up during organogenesis in the postnatal maturation period [33,34]. Additionally, embryonic HER3 knockdown mice showed severely underdeveloped sympathetic ganglia and a partial lack of Schwann cells [35]; these suggest the importance of HER3 in the development of the fetal mouse brain. HER3 transcripts were reported in the human liver, kidney, and brain but not in heart or lung fibroblasts [28]. Like EGFR and HER2 expressions, HER3 expression was also reported in normal keratinocyte and glandular epithelium tissues; however, unlike EGFR and HER2, HER3 expression was not detected in fibroblasts, skeletal muscle, or lymphoid cells [28], suggesting the tissue-specific function of HER3 in ectodermal development.
As physiological HER3 expression was reported in normal tissue, similarly, abnormal expression of HER3 has often been linked to a variety of different cancers, including breast cancer, colorectal carcinoma, squamous cell carcinoma of the head and neck, uveal melanoma, and gastric, ovarian, prostate, and bladder cancers, as shown in Table 1 [36,37,38,39]. Overexpression of HER3 protein has been linked to 50–70% of cases of breast cancer [40,41,42]. Upregulation of HER3 has been associated with metastasis, tumor volume, and risk of recurrence [43,44]. In colon cancer, HER3 overexpression has been associated with lymph node metastasis and poor progression [45,46,47,48]. Similarly, HER3 expression has been associated with increased metastasis and decreased overall survival in squamous cell carcinoma of the head and neck [49,50]. The above reports suggest the importance of HER3 in tumorigenesis and the need for targeting to inhibit tumor growth.
Although HER3 is a membrane protein (like EGFR), it has also been reported to localize in the nucleus. A study reported in human breast cancer cells that HER3 predominantly localizes in the nucleus; however, upon ligand stimulation, it is transported to the cytoplasm [51]. Previous studies suggested that HER3 localization also has tissue-specific functions. In gastric carcinomas, HER3 nuclear expression is associated with vascular and lymphatic invasion [52]. Similarly, in prostate cancer, HER3 nuclear expression is correlated with tumor progression [53]. In contrast, nuclear HER3 expression is associated with favorable overall survival in uveal melanoma [54]. In contrast, membranous expression of HER3 is associated with decreased survival in head and neck squamous cell carcinoma [55].
Table 1. Expression of HER3 in different types of cancer detected by immunohistochemistry (IHC) in the selected studies.
Table 1. Expression of HER3 in different types of cancer detected by immunohistochemistry (IHC) in the selected studies.
Cancer Type% of HER3 OverexpressionAntibody Used in IHCCutoff for OverexpressionReference
Pancreatic41.3%Nanotools, Teningen, GermanyModerate staining is observed in >10% of tumor cells (score 2+), and strong staining is observed in >10% of tumor cells (score 3)[56]
Breast43.0%Clone 2F12, Labvision, Cheshire, UKPositive: Optimal cutoffs for HER2:HER3 dimers were assessed by performing a minimum P value estimation using approximate 5% cutoffs across the entire dataset using relapse-free survival as an endpoint[57]
17.5%IgG1, Neomarkers, UK4-point scale, where 0 = no staining, 1 = light staining, 2 = moderate staining, and 3 = strong staining [58]
Colorectal17.0%MAb-MS-725-P, Neomarkers, Fremont, CAMembranous staining: >1% of tumor cells stained. Cytoplasmic staining: 2+: moderate immunostaining in >10% of tumor cells and 3+: strong immunostaining in >10% of tumor cells [59]
69.7%Lab Vision, Fremont, CA Cytoplasmic and membrane CytoplasmicCytoplasmic staining: 0: no staining or weak staining in <10% of tumor cells.
membranous staining: 0: no staining in <10% of tumor cells; 1: weak staining in >10% tumor cells; 2+: moderate staining in > 10% tumor cells; 3: strong staining in >10% tumor cells
[59]
20.9%Clone C-17, 1:50; Santa Cruz, CADepending on the intensity of staining, HER3 expression was classified as weak, intermediate, or strong [60]
Gastric59.0%Mouse monoclonal antibody, Neomarkers2+ = moderate staining, and 3+ = strong staining[61]
34.0%RB-9211 rabbit polyclonal, dilution 1:100, N terminal; Neomarkers, Fremont, CA0 = <10% of positively stained cells; 1 = 10–25%; 2 = 26–50%; 3 = 51–75%; 4 = >75%[52]
Melanomaapproximately 65.0%Clone C-17, 1:50 dilution; SantaCruz, CAPositive: high GIS > 6 (GIS: German immunohistochemical scoring)[60]
Ovary53.4%C-17, rabbit polyclonal antibody; dilution: 1:25; Santa Cruz, CAPositive: scores >8 [62]
Head and Neck8.8%RTJ.2, mouse monoclonal antibody; Santa Cruz, CAScores of 0, +1, +2, and +3 for increasing intensity [55]
Cervix55.5%MS-725-P, mouse antibody; NeomarkersIntensity scale 0–4 based on pixel density[63]

4. Role of HER3 in the Genesis and Progression of Different Types of Cancer

As the KD of HER3 is inactive, it is an obligate binding partner with multiple HER family proteins to activate PI3K/Akt signaling [6]. PI3K/Akt signaling contributes to many biological functions (translation, survival, nutrient sensing, metabolic regulation, and cell cycle control) [64,65,66], which are also involved in tumorigenesis, suggesting the importance of HER3 in cancer development. This HER3/PI3 K/Akt signaling cascade has been linked to breast, ovarian, colon, gastric, and lung cancer [67].
Interestingly, a sequence analysis suggested the binding activity of HER3 cytoplasmic domains with not only PI3K but also other proteins such as GRB7, GRB2, SHC, and SRC [15]. GRB7 and GRB2 interact with HER3 through their SH2 domain, whereas SRC SHC interacts with HER3 through the PTB domain [67,68,69,70]. Studies found that GRB2 binds to HER3 only in the absence of GRB7 [69]. The SHC–HER3 interaction is essential for MAPK signaling [70].
Because HER3 function depends on binding with other receptors, HER3 cannot transform a normal cell into cancer cells [71,72]. The most favorable binding partner of HER3 is HER2. A study showed that introducing HER3 into NIH3T3 fibroblast cells results in a low level of colony growth, but when HER3 is transfected with HER2, it significantly induces colony growth compared to HER2 and HER3 alone [73]. Similarly, in vivo studies showed that the introduction of both EGFR and HER3 was not tumorigenic, whereas the introduction of HER2 and HER3 yielded significant tumorigenic growth compared to the combination of other HER family proteins. These suggest that the most potent binding partner of HER3 is HER2.
HER3 is similarly important to its most potent binding partner, HER2 [38]. Studies also found that the knockdown of HER3 expression is more effective in inhibiting breast cancer cell growth than the knockdown of EGFR [38,74]. Interestingly, HER3 was found to preferentially dimerize with EGFR to induce cell proliferation, invasion, and migration in melanoma and pancreatic cancer [60,75]. Activation of HER3 via neuregulin-1 (NRG-1) was found to be enriched in a subset of SCCHN, and HER3 expression was associated with reduced survival in SCCHN [55,76]. Moreover, our previous studies in a panel of cancer cell lines of breast, bladder, colon, gastric, esophageal, lung, tongue, and endometrium cancer showed constitutive phosphorylation of HER3 in most of the HER2-amplified cancers [77]. Also, we noticed upon HER3 knockdown that overall tumor growth was substantially reduced in three HER2-amplified cancers from non-breast origin, suggesting HER3 has a vital role in tumorigenesis and tumor growth beyond breast cancer [77].

5. Role of HER3 Ligands in Tumor Growth and Resistance to Different Anti-Cancer Therapies

As discussed above, HER3, when activated via its ligand, dimerizes with another receptor (the most potent binding partner of HER3 is HER2) and transduces signals through the PI3K–AKT–mTOR pathway. Overexpression of HER3 ligands heregulin/neuregulin was shown to induce breast cancer progression. Studies also showed that resistance to TKIs may be due to compensatory upregulation of HER3 and that prerequisite for ligand-mediated activation of HER3. In breast cancer, it was found that high expression HER family receptor ligands play an essential role in developing resistance to anti-HER2 inhibitors [78,79,80,81]. Similarly, a study showed that the EGF ligand betacellulin and the HER3 ligand heregulin reduced the antiproliferative effect of anti-HER2/HER3 therapy [82]. The HER3 ligand neuregulin β1 was found to induce resistance to T-DM1 (a combination of Herceptin and the chemotherapy medicine emtansine). This study showed that neuregulin β1-mediated T-DM1 resistance was reversed by pertuzumab [83]. This study also found that HER2 non-overexpressing breast cancer cells (MDA-MB-175VII) that release autocrine heregulin are also resistant to T-DM1 [83]. Similarly, another study showed that neuregulin1 was upregulated after treatment with a p110α-specific inhibitor (BYL719), and this treatment failed to reduce tumor growth [84]. Heregulin has also been reported to induce anchorage-independent growth of breast cancer cells more potently than EGF [85]. As heregulin/neuregulin1 induces lapatinib resistance in HER2-amplified breast and gastric cancer cells, HER3 inhibition via siRNA was found to reverse this effect [81,86]. Additionally, activation of HER2 signaling by increased heregulin production was noted to be associated with acquired resistance to cetuximab in colorectal cancer cells [87].

6. Role of HER3 in Resistance to Different Anti-Cancer Therapies

6.1. HER3 as a Mediator of Resistance to Targeted Therapies

Many pharmaceutical companies developed small-molecule inhibitors and monoclonal antibodies against EGFR and HER2 because of their pro-oncogenic function in different types of cancer [6,88]. A monoclonal antibody, cetuximab, has been approved for SCCHN in combination with radiation therapy for locally advanced disease and platinum-based chemotherapy as a standard first-line systemic therapy. A clinical trial (EXTREME trial) showed that cetuximab had reduced the risk of death by 20% compared to patients receiving chemotherapy alone, and the median survival rate increased to 10.1 months [89]. Small-molecule inhibitors against EGFR, such as erlotinib and gefitinib, have also been shown to be significantly effective against EGFR-mutant lung cancer [90,91]. Trastuzumab (Herceptin; Genentech), a humanized monoclonal antibody against HER2, has shown clinical success [92]. Additionally, a phase III clinical trial in gastric cancer (ToGA trial) showed that trastuzumab and chemotherapy significantly improve overall survival without affecting the quality of life [93].
Multiple mechanisms have been proposed to associate HER3-mediated with drug resistance, including upregulation of HER3 expression [94], higher production of NRG1 [95,96], and HER2 amplification [87,97]. Studies found that NRG1-induced HER3 activation induced resistance to BRAF V600E inhibitor vemurafenib in colon cancer [98,99]. In accordance with these findings, anti-HER3 antibodies restore sensitivity to vemurafenib in BRAF-V600E mutant colon cancer [99]. Similarly, a study showed that triple blockade of HER2/HER3 signaling using trastuzumab, pertuzumab, and patritumab could overcome resistance to trastuzumab therapy in heregulin-expressing and HER2-positive breast cancer [100].
A previous report showed that HER3 plays a crucial role in HER2-mediated resistance to tamoxifen, and inhibition of HER3 expression reverses this resistance [101]. Similarly, HER3 expression and activation were also reported to induce resistance to fulvestrant [102]. Various studies noted that HER3 is a major player in trastuzumab and lapatinib resistance through the PI3K/AKT and Src signaling pathways [16,95,103,104]. HER3 was even reported to be linked to resistance to adjuvant chemotherapy in triple-negative breast cancer (TNBC) [105]. Likewise, a study showed that HER3 overexpression caused paclitaxel resistance in HER2-overexpressing BC cell lines [106]. HER3-mediated signaling was also found to be involved in inducing resistance in several other EGFR-mediated targeted therapies, including gefitinib and cetuximab [87,107]. Also, in lung cancer, MET amplification is reported to be involved in gefitinib and erlotinib resistance through upregulation of the HER3/PI3K signaling axis [12]. Similarly, upregulation of NRG1 and HER3 expression was found to induce resistance to anaplastic lymphoma kinase (ALK) and BRAF inhibitors in melanoma and thyroid cancer [94,99,108].
There are multiple antibodies targeting EGFR that have been approved for clinical use. However, patients developed resistance over time. Drug-induced compensatory upregulation of HER3 and sustained PI3 K/Akt activation have been reported to play an essential role in resistance to HER2-targeted therapy in breast cancer [18,30]. Similarly, in our previous studies in a panel of cancer cell lines of eight different types of cancer, we noticed that after treatment with lapatinib (an inhibitor of EGFR and HER2), almost all HER2-amplified cancers show only a transient HER3 inactivation and HER3 is eventually re-phosphorylated over time [77]. Studies also showed that prolonged exposure to EGFR inhibitors gefitinib, erlotinib, or the HER2 inhibitor AG-825 caused upregulation of HER3 and Akt phosphorylation [12,18]. This increased membrane expression of HER3 under prolonged treatment of HER TKIs regulated through Akt-mediated negative-feedback signaling [18]. After prolonged treatment with HER2 antibody, trastuzumab showed upregulation of EGFR and HER3 expression in breast cancer [109]. Another study showed that the upregulation of HER3 ligand heregulin is also a possible mechanism of cetuximab resistance in colorectal cancer patients [87]. Additionally, amplification of proto-oncogene MET causes gefitinib resistance by driving HER3-dependent activation of PI3K [12].
HER3 and HER2 expression was significantly correlated with gefitinib resistance but not cetuximab in non-small cell lung cancer and head and neck squamous cell carcinoma (HNSCC) [107]. A combination treatment of gefitinib and the HER2-HER3 dimerization inhibitor pertuzumab showed more effective growth inhibition than gefitinib alone on gefitinib-resistant HNSCC cell lines [107]. As in pancreatic cancer, HER3 dimerized with EGFR and siRNA-mediated inhibition of HER3 expression showed acquired resistance to erlotinib, suggesting that lack of HER3 expression makes these cells less dependent on EGFR [75]. This result suggests that this resistance-promoting function of HER3 may be caused by activated HER3, not the total HER3 expression.

6.2. HER3 as a Mediator of Resistance to Hormonal Therapy, Chemotherapy, and Radiotherapy

HER3 is not only connected to resistance to targeted therapy; many studies also found HER3-mediated resistance to hormonal therapy, chemotherapy, and radiotherapy. One study found the critical role of HER3 in the progression of breast cancer cells, whereas inhibition of HER3 expression reduces resistance to reversed anti-estrogen receptor (ER) tamoxifen resistance in breast cancer cell lines [101]. Additionally, co-expression of HER2 and HER3 was reported to develop resistance to tamoxifen in breast cancer patients [110,111]. Similarly, elevated signaling via EGFR, HER2, and HER3 was found to induce resistance to ER agonist fulvestrant [112]. As a mechanism, another study showed that fulvestrant treatment enhances the HER3 expression as well as activation in an NRG1-dependent manner in breast cancer cells [102]. In triple-negative breast cancer, patients with high expression of both HER3 and EGFR were found to have worse 10-year survival after adjuvant chemotherapy compared to patients without adjuvant chemotherapy [105]. In castration-resistant prostate cancer (CRPC), NRG1 released from stromal cells induced antiandrogen resistance [113].
HER2 and HER3-mediated PI3K/AKT signaling was also reported to increase resistance for several chemotherapeutic agents, including 5-fluorouracil, paclitaxel, camptothecin, and etoposide in breast cancer cells [114]. Enhanced HER3 expression was also reported to cause paclitaxel resistance in erbB2-overexpressing breast cancer cells via the upregulation of Survivin [106]. A study found that a DNA-damaging agent, doxorubicin-induced HER3-PI3K-AKT signaling in ovarian cancer cells, and a combination of HER3 inhibition and doxorubicin reported increased apoptosis in the chemoresistant cells [115].
Moreover, using both in vitro and in vivo models, some studies also reported that ionizing radiation (IR) induces activation of EGFR, HER2, HER3, and HER4, and interestingly, silencing of HER3 inhibits the viability of cancer cells after treatment with IR [116,117].

7. Targeting HER3 and Its Challenges

HER family members are found to be expressed in endothelial and cardiac cells and play an essential role in proliferation and differentiation [118,119]. Similarly, HER3 KO was reported to exhibit ED13.5 lethality due to defective valve formation, pronounced heart defects, vasculature abnormalities, and specifically hypoplastic cardiac cushions with decreased mesenchyme [120,121]. In contrast to other HER family members, HER3 is also noted to be expressed in endocardial cushion cells and mesenchymal cells undergoing EMT [119]. HER3 is essential for developing and maintaining normal tissue and is intensely involved in developing a wide range of cancers [32]. HER3 has been shown to be involved in intrinsic or developed resistance against HER-targeting agents, and there is a considerable focus on optimizing this therapy [18,122]. Although EGFR and HER2 are widely known targets for therapy in cancer, HER3 has long been underestimated for cancer therapy. HER3 is the only family member of the HER family that lacks an active kinase domain, making it an obligate binding partner with other receptors for function. Studies showed that EGFR and HER2 are the most preferred dimerization partners of HER3 and also the most active heterodimers [74,123,124,125]. In view of the aberrant expression of HER3 and/or activation of HER3 and its ligand, NRG1 is associated with tumor progression and acquired resistance to EGFR and HER2-targeted therapies, and HER3 emerges as an interesting target [126,127]. As the HER3 kinase domain binds ATP with a Kd of approximately 1.1 μM and showed very little catalysis of autophosphorylation, it would not be beneficial to make small-molecule inhibitors against this KD [128]. Different pharmaceutical companies have been trying to target HER3 with a wide variety of approaches.
Although HER3 knockout mice do not survive, inhibition of HER3 expression was found to be broadly safe in the clinic, with a low grade of side effects [121,129]. The pharmaceutical industry has been trying to develop antibodies against HER3, but they have shown limited clinical efficiency for several reasons. Some developed antibodies (e.g., seribantumab) block the NRG1-binding region. However, they do not consider the ligand-independent heterodimer formation [130]. Similarly, some antibodies (e.g., elgemtumab) were developed to target the closed conformation of HER3, but they do not address the conformation-dependent binding [131].

8. Development of HER3-Directed Therapy and Its Clinical Stages

Monoclonal antibodies (MoAbs) and antibody–drug conjugates have been widely used in targeting EGFR and HER2 in different cancers. Although HER3 has been ignored for a long time for its pseudo-KD, recently, HER3 came into the limelight due to its emerging role in tumor progression and drug resistance in various types of cancers.

8.1. HER3-Targeting Monoclonal Antibodies and Clinical Trials

Pharmaceutical companies have been developing HER3 inhibitors and pan-ErbB inhibitors, which target HER3 as well as other HER family proteins. Many of these drugs are in the early clinical stages. As the HER3 KD is inactive, most of the TKIs cannot inhibit the ATP binding site of HER3. This challenge makes researchers develop antibodies that target the extracellular domain of HER3. Although several HER3-targeting monoclonal antibodies have been developed that showed limited toxicity profiles in clinical trials, objective responses were rarely observed (Table 2). Only three antibodies moved to phase II to III clinical trials: which include (I) Patritumab (U3-1287), a fully human HER3-directed monoclonal antibody that binds to the extracellular domain of HER3 and promotes receptor internalization [132]; (II) Seribantumab (MM-121), a fully human immunoglobulin G2 that binds to the ligand-binding domain of HER3 and inhibits HRG-mediated downstream PI3K/AKT signaling [133]; (III) Lumretuzumab (RO5479599), an immunoconjugate containing a humanized HER3-directed monoclonal antibody that binds to HER3 extracellular domain, inhibiting HER3 dimerization and EGFR-dependent signaling, and activates the immune system to exert antibody-dependent cellular cytotoxicity [134].
MM-121 is one of the fully humanized HER3 monoclonal antibodies that block the binding of HRG1 (a neuregulin-1 type I polypeptide) to HER3 [130]. MM-121 was found to inhibit tumor growth in pancreatic ductal adenocarcinoma (PDAC) through the Akt axis when combined with erlotinib [135]. Similarly, MM-121 was also found to induce sensitivity to gefitinib (an EGFR inhibitor) in gefitinib-resistant lung cancer cell lines [136]. In the lung cancer model, MM-121 showed an additive effect to reduce tumor growth when combined with the anti-EGFR antibody cetuximab [136]. In comparison, MM-111 is a bispecific antibody that binds both HER2 and HER3, shown to inhibit PI3K signaling. U3-1287 (AMG888) is the first fully humanized HER3 monoclonal antibody that induces rapid internalization of HER3 [137]. U3-1287 was reported to inhibit the growth of breast, lung, and colorectal cancer cell lines and inhibit tumor growth in pancreatic, NSCLC, and colorectal cancer xenograft models [138]. Similarly, pharmaceutical companies developed many antibodies targeting HER2 HER3 dimerization interfaces. Pertuzumab is one of these antibodies that inhibit HER2 and HER3 binding, and many clinical trials showed a significant benefit for HER2-positive breast cancer patients [68,139]. There have been many clinical trials with pertuzumab in different other cancer types as well. In addition to the above antibodies, many multitarget inhibitors (such as MEHD7945A, MP-470, and AZD8931) are still under clinical development.
Table 2. Development of HER3-directed monoclonal antibodies.
Table 2. Development of HER3-directed monoclonal antibodies.
Monoclonal AntibodiesStudy PopulationClinical Trial
Phase
Adverse EventsStatus
Finding
Reference
LumretuzumabAdvanced or metastatic NSCLCNCT02204345
phase I + II
Gastrointestinal, hematological, and nervous system toxicities, but generally mild and manageableTerminated
Efficacy of lumretuzumab + carboplatin
+ paclitaxel is similar to chemotherapy alone
[140]
Metastatic BC expressing HER3 and HER2NCT01918254
phase I
Diarrhea and hypokalemiaCompleted
Lumretuzumab + pertuzumab + paclitaxel was correlated with a serious incidence of diarrhea
[141]
Metastatic and/or locally advanced malignant HER3 + solid tumors of epithelial cell originNCT01482377
phase I
Gastrointestinal and skin toxicitiesCompleted
Moderate clinical activity with toxicity manageable
[142,143]
ISU104Advanced solid tumorsNCT03552406
phase I
Dose escalation study (PART I)
Dose-expansion study (PART II)
PART I: oral mucositis, pruritus, diarrhea, and fatigue
PART II: anorexia, mucositis oral and diarrhea in monotherapy and diarrhea and acneiform rash in combination with cetuximab
Status unknown
ISU104 was well tolerated up to 20 mg/kg/day without DLT and showed a disease control rate of 60%
ISU104 monotherapy or with cetuximab was safe with promising clinical outcomes in recurrent or metastatic HNSCC treated with the combination
[144]
CDX-3379Advanced cancerNCT02014909
phase I
Diarrhea, fatigue, nausea, and rashCompleted
CDX-3379 can be combined safely with cetuximab, erlotinib, vemurafenib, or trastuzumab at 15 to 20 mg/kg
[145]
HNSCCNCT02473731, phase IDiarrhea, fatigue, and acneiform dermatitis, but mild or moderateCompleted
CDX-3379 was well tolerated and associated with tumor regression
[146]
Advanced stage NRAS mutant and BRAF/NRAS wild-type melanomaNCT03580382 phase I + II Terminated
Advanced HNSCCNCT03254927 phase II Terminated
CDX-3379 in combination with cetuximab is well tolerated with signs of antitumor activity
[147]
Thyroid cancerNCT02456701 phase I Completed
Vemurafenib + CDX-3379 is safe and enhances efficacy for RAI uptake
[148]
AV-203Metastatic or advanced solid tumorsNCT01603979 phase I Completed
AV-203 was well tolerated. RP2D is 20 mg/kg IV every 2 weeks. The PR in a patient with squamous NSCLC guarantees future testing of AV-203 in this indication
[149]
GSK2849330Advanced HER3 + solid tumorsNCT01966445 phase IDrug tolerated with no major issuesCompleted
GSK2849330 has a durable response in an exceptional responder with an advanced CD74–NRG1-rearranged IMA
[150]
Advanced HER3 + solid tumorsNCT02345174 phase IDecreased appetite and diarrheaCompleted
Despite the restricted number of patients, an exploratory ID50 of 2 mg/kg and ID90 of 18 mg/kg have been reported
[151]
SeribantumabAdvanced NSCLCNCT00994123 phase I + IIDiarrhea, rash, decreased appetite, fatigue, and nauseaCompleted
Phase I: no maximum tolerated dose was determined and the AE profile was similar between comparative treatment
Phase II: there was no significant difference in PFS between monotherapy and combination therapy. However, retrospective analyses suggest that detectable NRG mRNA levels identified patients who may benefit from MM-121
[152]
NSCLC expressing NRGNCT02387216 phase IIDiarrhea, fatigue, and neutropenia in the combination treatmentTerminated
Seribantumab does not improve PFS when added to docetaxel
[153]
CRC, HNSCC, NSCLC, TNBC, and other tumors with EGFR dependenceNCT01451632
phase I
Part 1: fatigue, dermatitis acneiform, hypomagnesemia, diarrhea, decreased appetite, and hypokalemia
Part 2: diarrhea, hypokalemia, nausea, fatigue, hypomagnesemia, decreased appetite, dermatitis acneiform, mucosal inflammation, dehydration, and weight decrease
Completed
Unlike doublet treatment, seribantumab + cetuximab + irinotecan was difficult to tolerate. However, MM121 + cetuximab with and without irinotecan had no activity in the vast majority of patients with prior exposure to EGFR-directed therapy
[154]
Advanced gynecologic and breast cancersNCT01209195 phase I Completed
ER +, HER2- BC, and TNBCNCT01421472
phase II
Completed
Platinum-resistant or refractory recurrent/advanced ovarian cancersNCT01447706, phase IIDiarrhea, vomiting, stomatitis, and mucosal inflammationCompleted[155]
Locally advanced or metastatic ER + and/or PR + and HER2- BCNCT01151046 phase IIDiarrhea, nausea, fatigue, and arthralgiaCompleted
The addition of MM-121 to exemestane did not significantly prolong PFS in the unselected population
[156]
CRC, NSCLC, and HNSCCNCT02538627 phase I Terminated
Advanced solid tumorsNCT00734305 phase I Completed
Advanced solid NCT01447225 Diarrhea, nausea, and fatigue, Completed
MM-121 can be administrated with
[157]
Tumorsphase IAnemia, vomiting, hypokalemia, decreased appetite, thrombocytopenia, peripheral edema, neutropenia, and constipationGemcitabine, pemetrexed, cabazitaxel, and carboplatin
Postmenopausal women with metastatic BCNCT03241810 phase II Terminated
Locally advanced or metastatic solid tumorsNCT01436565 phase I Completed
NRG1 gene fusion-positive advanced solid tumorsNCT04383210
phase II
Active, not recruiting
An NRG1 fusion-positive metastatic pancreatic cancer patientNCT04790695
phase II
Completed
PatritumabAdvanced, refractory solid tumorsNCT01957280 phase IThe most frequently reported treatment-related AEs were gastrointestinal
Completed
Well tolerated with no anti-patritumab neutralizing antibodies formation and with normal bioavailability
[158]
EGFR wild-type subjects with locally advanced or metastatic NSCLC who have progressed on at least one prior systemic therapyNCT02134015 phase IIIIn placebo + erlotinib, the most frequent AEs were rash, diarrhea, and fatigue, in patritumab + erlotinib were diarrhea, rash, and decreased appetiteTerminated
Patritumab + erlotinib apparently do not have better results than placebo + erlotinib
Recurrent or metastatic HNSCCNCT02633800 phase IIRash, anemia, neutropenia, hypomagnesemia, and nauseaTerminated
Patritumab + cetuximab + platinum was safe but not more efficacious than cetuximab + platinum
[159]
EGFR treatment naïve subjects with advanced NSCLC who have progressed on at least one prior chemotherapyNCT01211483 phase I + IIAE grade > 3 included diarrhea and rashCompleted
Patritumab improved PFS in the NRG high, but not in the ITT population
Recurrent or metastatic HNSCCNCT02350712
phase I
Skin and subcutaneous tissue disordersCompleted
Patritumab (18 mg/kg loading dose, 9 mg/kg maintenance dose) with cetuximab, and platinum therapy was tolerated and active in HNSCC
[160]
Advanced solid tumorsNCT01479023, phase IDiarrhea, dizziness, fatigue, headache, hypertension, and weight lossTerminated
[64Cu]DOTA-patritumab and unlabeled patritumab are safe and well tolerated
[161]
Newly diagnosed HER2 + metastatic BCNCT01512199
phase I + II
Terminated
Advanced solid tumorsNCT00730470 phase IFatigue, diarrhea, nausea, decreased appetite, and dysgeusiaCompleted
Patritumab treatment was well tolerated and some evidence of disease stabilization was observed
[132]
Elgemtumab (LJM716)Platinum-pretreated recurrent/metastatic HNSCCNCT02143622
phase I + II
Withdrawn
Advanced HER2 + BC or gastric cancerNCT01602406
phase I 
Diarrhea, nausea, fatigue, and chillsCompleted
As of 4 October 2013, LJM716 demonstrated clinical activity in combination with trastuzumab in trastuzumab-resistant patients with an acceptable safety profile
[162]
Metastatic HER2 + BCNCT02167854
phase I
Diarrhea, hyperglycemia, hypokalemia, mucositis, and transaminitisCompleted
The combination treatment of LJM716, BYL719 (PI3K inhibitor) and trastuzumab has antitumor activity in these pretreated HER2 + metastatic BC with PIK3CA mutations
[163]
Patients with previously treated ESCCNCT01822613
phase I + II
Completed
HER2 + BC, HER2 + gastric cancer, HNSCC and ESCCNCT01598077
phase I
Diarrhea, decreased appetite, pyrexia, fatigue, nausea, infusion-related reactions, vomiting, constipation and dyspnea and anemia and hypomagnesemiaCompleted
LJM716 was well tolerated, with a manageable safety profile
[164]
Japanese patients with advanced solid tumorsNCT01911936
phase I
Diarrhea, stomatitis, fatigue, pyrexia and paronychiaCompleted
LJM716 was well tolerated and a degree of tumor shrinkage was reported
[165]
REGN1400Patients with advanced NSCLC, CRC, or HNSCC who progressed on prior erlotinib or cetuximabNCT01727869
phase I
Rash, diarrhea, nausea, and hypomagnesemia Completed
REGN1400 as monotherapy or combined with erlotinib or cetuximab was generally tolerated
[166]
Sym013Patients with advanced epithelial malignanciesNCT02906670
phase I + II
Terminated
NSCLC: non-small cell lung cancer, BC: breast cancer, HNSCC: head and neck squamous cell carcinoma; AEs: adverse events; PFS: progression-free survival; ITT: intention to treat; ESCC: esophageal squamous cell carcinoma; CRC: colorectal cancer.

8.1.1. HER3-Directed Monoclonal Antibodies in Breast Cancer

A phase 1 trial with sarilumab, paclitaxel, and trastuzumab in pretreated HER2-positive metastatic breast cancer confirmed manageable toxicities and encouraged preliminary activity [167]. A phase Ib trial, when lumretuzumab was added with pertuzumab and paclitaxel in 35 patients with HER3-positive HER2-low breast cancer, demonstrated a high incidence of diarrhea and a narrow therapeutic window [141]. In a biomarker analysis, a phase II study of seribantumab plus exemestane in patients with hormone receptor (HR)-positive HER2-negative metastatic breast cancer found a clinical benefit in the HRG-high subgroup [156]; however, this trial (SHERBOC trial) was prematurely terminated.

8.1.2. HER3-Directed Monoclonal Antibodies in NSCLC

In a phase II HERALD study, when patritumab was added to erlotinib, it did not prolong progression-free survival in 215 NSCLC patients and increased the risk of gastrointestinal toxicity [168]. At the same time, a phase III HER-3 lung trial did not confirm the efficacy of patritumab and erlotinib in the subgroup of EGFR wild-type NSCLC patients with high HRG expression [169]. Similarly, a randomized phase II trial of seribantumab in combination with erlotinib in patients with EGFR wild-NSCLC demonstrated progression-free survival benefits in patients with detectable HRG mRNA in the tumor [152]. Therefore, seribantumab received a fast-track designation for patients with HRG-positive NSCLC.

8.1.3. HER3-Directed Monoclonal Antibodies in Other Cancers

A phase II trial of an anti-HER3 monoclonal antibody, CDX-3379, and cetuximab in a population of heavily pretreated patients with head and neck squamous cell carcinoma (HNSCC) demonstrated antitumor activity with an acceptable safety profile [147]. Additionally, a first-in-human phase I study with ISU104 (ErbB3 monoclonal antibody) in patients with advanced solid tumors showed mucositis and diarrhea represented the most frequent treatment-related adverse events (TRAEs) [170].

8.2. HER3-Targeting Bispecific Antibodies and Clinical Trials

Blocking HER3 signals via monoclonal antibodies is challenging, so different bispecific antibodies have been developed for clinical tests, as summarized in Table 3. Among all these bispecific antibodies, the most promising one is the HER2/HER3-directed zenocutuzumab (MCLA-128), which is currently under a phase II clinical trial (NCT02912949). Zenocutuzumab was shown to reduce HRG-stimulated HER3 tumor growth and recruit natural killer (NK) cells into the tumor. Zenocutuzumab was evaluated in patients with NRG1 fusion-positive solid tumors, including NSCLC, breast cancer, and pancreatic cancer [171,172].
Another important bispecific antibody is duligotuzumab (MEHD7954A), which is an EGFR/HER3-directed bispecific monoclonal antibody. In a randomized phase II trial (MEHGAN study), duligotuzumab failed to improve clinical outcomes compared with cetuximab in squamous cell carcinoma of the head and neck (HNSCC) due to high incidence of gastrointestinal TRAEs [173]. In another phase II trial, the addition of duligotuzumab to FOLFIRI was not found to improve the clinical outcomes in patients with RAS exon 2/3 wild-type metastatic colorectal cancer compared with cetuximab plus FOLFIRI [174].
Moreover, the development of another bispecific HER3/IGF-1R-directed MoAb istiratumab (MM-141) was discontinued due to the negative results of the phase II CARRIE trial. This study showed that the addition of istiratumab to first-line nab-paclitaxel and gemcitabine did not show a clinical benefit in patients with metastatic pancreatic cancer with high IGF-1 serum levels [175].
Table 3. Development of HER3-directed bispecific antibodies.
Table 3. Development of HER3-directed bispecific antibodies.
Bispecific AntibodiesStudy PopulationClinical Trial
Phase
Adverse EventsStatus
Finding
Reference
Zenocutuzumab (Zeno, MCLA-128)Solid tumors harboring an NRG1 fusionNCT02912949
phase I + II
Infusion-related reactions, diarrhea, rash, and fatigueRecruiting
As of January 2017, MCLA-128 reported a safety profile and antitumor activity in pretreated metastatic breast cancer patients progressing on HER2 therapies
[176]
A patient with advanced NRG1-fusion-positive solid tumorNCT04100694
not given
Status and findings are not posted on www.clinicaltrials.gov (accessed on 20 October 2023)
Metastatic BCNCT03321981
phase II
Neutropenia/neutrophil count decrease, diarrhea, asthenia/fatigue, and nauseaActive, not recruiting
The combination of MCLA-128 + trastuzumab + vinorelbine is active in pretreated patients with HER2 + metastatic BC. The treatment is safe with manageable AEs
[177]
SI-B001Locally advanced or metastatic epithelial tumorsNCT04603287
phase I
Recruiting
Recurrent and metastatic HNSCCNCT05054439
phase II
Recruiting
Recurrent metastatic ESCCNCT05022654
phase II
Recruiting
Recurrent and metastatic NSCLCNCT05020769
phase II + III
Recruiting
EGFR/ALK wild-type recurrent or metastatic NSCLCNCT05020457
phase II
Recruiting
Unresectable or metastatic digestive system malignancies (colorectal and gastric cancer)NCT05039944
phase II
Terminated
MM-111Advanced, refractory HER2 amplified, NRG + BCNCT01097460
phase I
Fatigue, diarrhea, and dyspnoeaCompleted
Advanced, refractory HER2 amplified, NRG + cancersNCT00911898
phase I
Completed
Advanced HER2 + solid tumorsNCT01304784
phase I
Anemia, acute renal failure (assessed as cisplatin-related), chest pain, decreased appetite, diarrhea, febrile neutropenia, hyperuricemia, hypokalemia, hyponatremia, hypophosphatemia, mucosal inflammation, nausea, neutropenia, stomatitis, thrombocytopenia, and vomitingCompleted
Treatment with MM-111 and standard-of-care HER2-directed regimens was viable
[178]
HER2 + carcinomas of the distal esophagus, gastroesophageal junction, and stomachNCT01774851
phase II
Diarrhea, anemia, decreased appetite, alopecia, fatigue, nausea, vomiting, asthenia, neutropenia, constipation, and coughTerminated
MM-111 did not improve PFS or OS when added to paclitaxel + trastuzumab
[179]
Istiratumab (MM-141)Advanced solid tumorsNCT01733004
phase I
Vomiting, nausea, fatigue, abdominal pain, increased AP, dyspnea, diarrhea, anemia, increased AST, and rashCompleted
MM-141 was well tolerated as monotherapy and in combination with everolimus or paclitaxel + gemcitabine in patients with relapsed/refractory solid tumors
[180]
Metastatic pancreatic cancerNCT02399137
phase II
Neutropenia, alopecia, diarrhea, fatigue, thrombocytopenia, anemia, and decreased appetiteCompleted
Istiratumab failed to improve the efficacy of chemotherapy
[175]
CRC, NSCLC, and HNSCCNCT02538627
phase I
Terminated
DuligotuzumabLocally advanced or metastatic solid tumors with mutant KRASNCT01986166
phase I
Diarrhea, general disorders, dermatitis acneiform, rash, rash erythematous, rash maculo-papular, and nauseaCompleted
The combination of cobimetinib and duligotuzumab was correlated with increased toxicity and limited efficacy
[181]
Locally advanced or metastatic epithelial tumorsNCT01207323
phase I
Headache, rash, and diarrheaCompleted
Duligotuzumab was well tolerated with evidence of tumor pharmacodynamic modulation and antitumor activity in HNSCC
[182]
Recurrent/metastatic HNSCCNCT01911598
phase I
Neutropenia, hypokalemia, dehydration, anemia, and diarrhea in arm A and neutropenia, anemia, febrile neutropenia, leukopenia, thrombocytopenia, and hypomagnesemia in arm BCompleted
Duligotuzumab with cisplatin + 5-fluorouracil (arm A) or carboplatin + paclitaxel (arm B) demonstrated promising activity despite chemotherapy dose reductions and could be maintained with duligotuzumab alone
[183]
KRAS wild-type metastatic CRCNCT01652482 phase IIRash, diarrhea, fatigue, and nausea. There were fewer rash events of any grade in the duligotuzumab arm but more diarrheaCompleted
The combination of FOLFIRI with duligotuzumab did not improve clinical outcomes compared with the cetuximab combination
[174]
Recurrent/metastatic HNSCCNCT01577173
phase II
Rash, infections, diarrhea, fatigue, and nauseaCompleted
Duligotuzumab demonstrated similar activity to cetuximab, but not superior
[173]
NSCLC: non-small cell lung cancer, BC: breast cancer, HNSCC: head and neck squamous cell carcinoma; AEs: adverse events; PFS: progression-free survival; ESCC: esophageal squamous cell carcinoma; CRC: colorectal cancer; OS: overall survival.

8.3. HER3-Targeting Antibody–Drug Conjugates and Clinical Trials

The new era of therapy is antibody–drug conjugates (ADCs), where cytotoxic small-molecule inhibitors conjugated on a monoclonal antibody scaffold; upon binding with the cell surface antigen, the ADC is internalized by the tumor cell and processed by the endo-lysosomal system. Patritumab deruxtecan (U3 1402; HER3-DXd) is one of the ADCs (Table 4), where the topoisomerase I inhibitor deruxtecan is attached with patritumab via a tetrapeptide-based cleavable linker. This highly cytotoxic patritumab deruxtecan is found to inhibit DNA replication and trigger immune cells. A study evaluated its antitumor activity against HER3-expressing tumors with tolerable safety profiles [184]. In a preclinical study, patritumab deruxtecan significantly sensitized the tumor to PD-1 blockade, which further suggests patritumab deruxtecan is a promising candidate as a partner of immunotherapy for patients with tumor-specific HER3 expression [185].

8.3.1. Development of Antibody–Drug Conjugates in Breast Cancer

In a first-in-human study (U31402-A-J101), HER3-DXd was evaluated in heavily pretreated HER3-expressing metastatic breast cancer patients [191]. This study found promising antitumor activity in HR+/HER2− and HER2+ metastatic breast cancer patients as well as TNBC patients. HER3-DXd also showed a manageable toxicity profile and a low rate of discontinuation (9.9%) due to treatment-emergent adverse events (TEAEs), most commonly gastrointestinal and hematologic.
Based on the promising results of the first-in-human study (U31402-A-J101), HER3-DXd was investigated in HER2-positive breast cancer treated with chemo-free dual HER2 blockade. This study showed a combined score of TILs, and tumor cellularity (CelTIL score) was correlated with a pathological complete response at day 15 [192]. SOLTI TOT-HER3 window of opportunity trial (NCT04610528) showed a single dose of HER3-DXd (6.4 mg/kg) led to a clinically meaningful response, increased immune infiltration, and suppression of proliferation across varied levels of baseline ERBB3 mRNA [193]. The TOT-HER3 trial is still enrolling patients to evaluate the biological activity of HER3-DXd, measured as the CelTIL score increased post-treatment (C1D21) in patients with HR+/HER2-negative breast cancer and patients with TNBC tumors. Additionally, another phase II ICARUS-BREAST trial (NCT04965766) is investigating the HER3-DXd in hormone receptor-positive (HR+) unresectable locally advanced or metastatic breast cancer who are resistant to endocrine therapy and cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors.

8.3.2. Development of Antibody–Drug Conjugates in NSCLC

In a dose-expansion cohort (DEC) of a phase I trial (clinical trial information: NCT03260491), HER3-DXd exhibited an objective response rate (ORR) of 39% (95% CI 24.4–54.5%) in patients with EGFR-mutated NSCLC progressed after osimertinib and platinum-based chemotherapy [186]. In this study, HER3-DXd showed antitumor activity independent of EGFR TKI resistance mechanisms, suggesting that HER3-DXd could be considered a treatment option agnostic to the EGFR TKI resistance mechanism. After the progression of this study, another phase II trial, HERTHENA-Lung01 (clinical trial information: NCT04619004), is currently undergoing, which is investigating the effect of the HER3-DXd in previously treated patients with metastatic or locally advanced EGFR-mutated NSCLC after failure of third-generation TKIs. In a phase I trial (clinicalTrials.gov (accessed on 20 October 2023) Id # NCT04676477), HER3-DXd combined with osimertinib was also administered in patients with locally advanced or metastatic EGFR-mutated NSCLC. Interestingly, HER3-DXd was found to show effectiveness in many other oncogenic mutations, including KRAS/NRAS mutations and ALK fusions with limited TRAE and drug-related deaths [194].

9. Emerging Treatment Strategies

Many non-target strategies have been proposed to kill cancer cells [195,196]. Similarly, many strategies have been proposed that inhibit HER3 indirectly. Since some proteins such as HER3 are challenging to target, PROteolysis TArgeting Chimeras (PROTACs) has emerged as a possible solution that inhibits protein function by degrading target proteins instead of inhibiting them [197]. Although PROTAC-mediated HER3 degradation has not been reported, partial HER3 degradation was found via the treatment of monoclonal antibody NG33 [198]. Crosslinked trastuzumab binding to HER2 also caused HER2 and HER3 endocytosis and degradation in breast cancer cells [199]. Another study showed that treatment with TX2-121-1 covalently binds to HER3 and causes partial degradation of HER3, and inhibits heterodimerization of HER3 with HER2 or c-Met [200]. Different treatment strategies have emerged using noncoding RNAs [201,202,203]. Some studies also proposed antisense oligonucleotides or micro-RNA-mediated degradation of HER3 mRNA; however, these strategies have yet to be tested in clinical trials [204,205].
HER3-targeting vaccine (Ad-HER3-FL) was also reported to activate HER3-specific T-cells and induce anti-HER3-specific antibodies. This vaccine also enhanced intratumoral T-cell infiltration when combined with an immune checkpoint inhibitor compared to the vaccine alone [2]. Similarly, another study proposed antitumor immune responses via treatment with an immunoreacting HER3 epitope (HER-3872-868) in lung and head and neck cancer models [3].

10. Conclusions

The recent improvements in cancer research showed the importance of HER3 tumorigenesis, progression, and primary/acquired resistance to HER2- or EGFR-targeted therapy. Unlike other HER family receptors, inactive KD makes HER3 an obligate binding partner with other receptors. This feature gives researchers a challenging job in creating drugs against HER3. Many approaches have been proposed to inhibit the oncogenic function of HER3. First, due to the correlation of HER3 expression with sensitivity and resistance to HER TKIs, HER3 can be used as an important biomarker. Second, there is much progress in combination therapy with HER3 and EGFR/HER2-targeted agents, showing the way of overcoming the resistance. Thirdly, because HER3 signals through the PI3 K/Akt pathway, the combination therapy with HER3 and PI3 K/Akt inhibitors should have an anti-cancer function. Lastly, inhibition of HER3 function can also be achieved via inhibition of HER3-ligand (heregulin and neuregulin) production.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADCAntibody–drug conjugate
AE(s)Adverse event(s)
bAbsBispecific antibodies
BCBreast cancer
ECDExtracellular domain
EGFREpidermal growth factor receptor
ER + Estrogen receptor positive
ESCCEsophageal squamous cell carcinoma
HER2 + HER2-amplified
HNSCCHead and neck squamous cell carcinoma
HRGHeregulin
IGF1RInsulin-like growth factor 1 receptor
IMAInvasive mucinous adenocarcinomas
MoAbsMonoclonals antibodies
NRG(s)Neuregulin(s)
NSCLCNon-small cell lung cancers
OSOverall survival
PD-1Programmed cell death-1
PFSProgression-free survival
PRPartial response
RAIRadioactive iodine
RP2DRecommended phase 2 dose
RTKReceptor tyrosine kinases
scDbBispecific single-chain diabody
T-DM1Trastuzumab-emtansine
TKIsTyrosine kinase inhibitors
TNBCTriple-negative breast cancer

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer, J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Witsch, E.; Sela, M.; Yarden, Y. Roles for grOsada, T.; Morse, M.A.; Hobeika, A.; Diniz, M.A.; Gwin, W.R.; Hartman, Z.; Wei, J.; Guo, H.; Yang, X.Y.; Liu, C.X.; et al. Vaccination targeting human HER3 alters the phenotype of infiltrating T cells and responses to immune checkpoint inhibition. Oncoimmunology 2017, 6, e1315495. [Google Scholar] [CrossRef]
  3. Kumai, T.; Ohkuri, T.; Nagato, T.; Matsuda, Y.; Oikawa, K.; Aoki, N.; Kimura, S.; Celis, E.; Harabuchi, Y.; Kobayashi, H. Targeting HER-3 to elicit antitumor helper T cells against head and neck squamous cell carcinoma. Sci. Rep. 2015, 5, 16280. [Google Scholar] [CrossRef] [PubMed]
  4. Witsch, E.; Sela, M.; Yarden, Y. Roles for growth factors in cancer progression. Physiology 2010, 25, 85–101. [Google Scholar] [CrossRef] [PubMed]
  5. Robinson, D.R.; Wu, Y.M.; Lin, S.F. The protein tyrosine kinase family of the human genome. Oncogene 2000, 19, 5548–5557. [Google Scholar] [CrossRef]
  6. Hynes, N.E.; Lane, H.A. ERBB receptors and cancer: The complexity of targeted inhibitors. Nat. Rev. Cancer 2005, 5, 341–354. [Google Scholar] [CrossRef] [PubMed]
  7. Mendelsohn, J.; Baselga, J. Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J. Clin. Oncol. 2003, 21, 2787–2799. [Google Scholar] [CrossRef]
  8. Yarden, Y.; Pines, G. The ERBB network: At last, cancer therapy meets systems biology. Nat. Rev. Cancer 2012, 12, 553–563. [Google Scholar] [CrossRef]
  9. Haikala, H.M.; Jänne, P.A. Thirty Years of HER3: From Basic Biology to Therapeutic Interventions. Clin. Cancer Res. 2021, 27, 3528–3539. [Google Scholar] [CrossRef]
  10. Jura, N.; Shan, Y.; Cao, X.; Shaw, D.E.; Kuriyan, J. Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3. Proc. Natl. Acad. Sci. USA 2009, 106, 21608–21613. [Google Scholar] [CrossRef]
  11. Berger, M.B.; Mendrola, J.M.; Lemmon, M.A. ErbB3/HER3 does not homodimerize upon neuregulin binding at the cell surface. FEBS Lett. 2004, 569, 332–336. [Google Scholar] [CrossRef] [PubMed]
  12. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  13. Kunii, K.; Davis, L.; Gorenstein, J.; Hatch, H.; Yashiro, M.; Di Bacco, A.; Elbi, C.; Lutterbach, B. FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res. 2008, 68, 2340–2348. [Google Scholar] [CrossRef] [PubMed]
  14. Soltoff, S.P.; Carraway, K.L., 3rd; Prigent, S.A.; Gullick, W.G.; Cantley, L.C. ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor. Mol. Cell Biol. 1994, 14, 3550–3558. [Google Scholar] [CrossRef]
  15. Prigent, S.A.; Gullick, W.J. Identification of c-erbB-3 binding sites for phosphatidylinositol 3’-kinase and SHC using an EGF receptor/c-erbB-3 chimera. EMBO J. 1994, 13, 2831–2841. [Google Scholar] [CrossRef] [PubMed]
  16. Huang, X.; Gao, L.; Wang, S.; McManaman, J.L.; Thor, A.D.; Yang, X.; Esteva, F.J.; Liu, B. Heterotrimerization of the growth factor receptors erbB2, erbB3, and insulin-like growth factor-i receptor in breast cancer cells resistant to herceptin. Cancer Res. 2010, 70, 1204–1214. [Google Scholar] [CrossRef]
  17. Liu, J.; Kern, J.A. Neuregulin-1 activates the JAK-STAT pathway and regulates lung epithelial cell proliferation. Am. J. Respir. Cell Mol. Biol. 2002, 27, 306–313. [Google Scholar] [CrossRef]
  18. Sergina, N.V.; Rausch, M.; Wang, D.; Blair, J.; Hann, B.; Shokat, K.M.; Moasser, M.M. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 2007, 445, 437–441. [Google Scholar] [CrossRef]
  19. Campbell, M.R.; Ruiz-Saenz, A.; Zhang, Y.; Peterson, E.; Steri, V.; Oeffinger, J.; Sampang, M.; Jura, N.; Moasser, M.M. Extensive conformational and physical plasticity protects HER2-HER3 tumorigenic signaling. Cell Rep. 2022, 38, 110285. [Google Scholar] [CrossRef]
  20. Thakkar, D.; Sancenon, V.; Taguiam, M.M.; Guan, S.; Wu, Z.; Ng, E.; Paszkiewicz, K.H.; Ingram, P.J.; Boyd-Kirkup, J.D. 10D1F, an Anti-HER3 Antibody that Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models. Mol. Cancer Ther. 2020, 19, 490–501. [Google Scholar] [CrossRef]
  21. Clayton, A.H.; Orchard, S.G.; Nice, E.C.; Posner, R.G.; Burgess, A.W. Predominance of activated EGFR higher-order oligomers on the cell surface. Growth Factors 2008, 26, 316–324. [Google Scholar] [CrossRef] [PubMed]
  22. Hofman, E.G.; Bader, A.N.; Voortman, J.; van den Heuvel, D.J.; Sigismund, S.; Verkleij, A.J.; Gerritsen, H.C.; van Bergen en Henegouwen, P.M. Ligand-induced EGF receptor oligomerization is kinase-dependent and enhances internalization. J. Biol. Chem. 2010, 285, 39481–39489. [Google Scholar] [CrossRef]
  23. van Lengerich, B.; Agnew, C.; Puchner, E.M.; Huang, B.; Jura, N. EGF and NRG induce phosphorylation of HER3/ERBB3 by EGFR using distinct oligomeric mechanisms. Proc. Natl. Acad. Sci. USA 2017, 114, E2836–E2845. [Google Scholar] [CrossRef]
  24. Landgraf, R.; Eisenberg, D. Heregulin reverses the oligomerization of HER3. Biochemistry 2000, 39, 8503–8511. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, Y.; Bharill, S.; Karandur, D.; Peterson, S.M.; Marita, M.; Shi, X.; Kaliszewski, M.J.; Smith, A.W.; Isacoff, E.Y.; Kuriyan, J. Molecular basis for multimerization in the activation of the epidermal growth factor receptor. eLife 2016, 5. [Google Scholar] [CrossRef]
  26. Needham, S.R.; Roberts, S.K.; Arkhipov, A.; Mysore, V.P.; Tynan, C.J.; Zanetti-Domingues, L.C.; Kim, E.T.; Losasso, V.; Korovesis, D.; Hirsch, M.; et al. EGFR oligomerization organizes kinase-active dimers into competent signalling platforms. Nat. Commun. 2016, 7, 13307. [Google Scholar] [CrossRef]
  27. Saffarian, S.; Li, Y.; Elson, E.L.; Pike, L.J. Oligomerization of the EGF receptor investigated by live cell fluorescence intensity distribution analysis. Biophys. J. 2007, 93, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  28. Kraus, M.H.; Issing, W.; Miki, T.; Popescu, N.C.; Aaronson, S.A. Isolation and characterization of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: Evidence for overexpression in a subset of human mammary tumors. Proc. Natl. Acad. Sci. USA 1989, 86, 9193–9197. [Google Scholar] [CrossRef] [PubMed]
  29. Plowman, G.D.; Whitney, G.S.; Neubauer, M.G.; Green, J.M.; McDonald, V.L.; Todaro, G.J.; Shoyab, M. Molecular cloning and expression of an additional epidermal growth factor receptor-related gene. Proc. Natl. Acad. Sci. USA 1990, 87, 4905–4909. [Google Scholar] [CrossRef]
  30. Baselga, J.; Swain, S.M. Novel anticancer targets: Revisiting ERBB2 and discovering ERBB3. Nat. Rev. Cancer 2009, 9, 463–475. [Google Scholar] [CrossRef]
  31. Hellyer, N.J.; Cheng, K.; Koland, J.G. ErbB3 (HER3) interaction with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem. J. 1998, 333 Pt 3, 757–763. [Google Scholar] [CrossRef] [PubMed]
  32. Campbell, M.R.; Amin, D.; Moasser, M.M. HER3 comes of age: New insights into its functions and role in signaling, tumor biology, and cancer therapy. Clin. Cancer Res. 2010, 16, 1373–1383. [Google Scholar] [CrossRef] [PubMed]
  33. Darcy, K.M.; Zangani, D.; Wohlhueter, A.L.; Huang, R.Y.; Vaughan, M.M.; Russell, J.A.; Ip, M.M. Changes in ErbB2 (her-2/neu), ErbB3, and ErbB4 during growth, differentiation, and apoptosis of normal rat mammary epithelial cells. J. Histochem. Cytochem. 2000, 48, 63–80. [Google Scholar] [CrossRef]
  34. Stern, D.F. ErbBs in mammary development. Exp. Cell Res. 2003, 284, 89–98. [Google Scholar] [CrossRef]
  35. Levi, A.D.; Bunge, R.P.; Lofgren, J.A.; Meima, L.; Hefti, F.; Nikolics, K.; Sliwkowski, M.X. The influence of heregulins on human Schwann cell proliferation. J. Neurosci. 1995, 15, 1329–1340. [Google Scholar] [CrossRef]
  36. Maurer, C.A.; Friess, H.; Kretschmann, B.; Zimmermann, A.; Stauffer, A.; Baer, H.U.; Korc, M.; Büchler, M.W. Increased expression of erbB3 in colorectal cancer is associated with concomitant increase in the level of erbB2. Hum. Pathol. 1998, 29, 771–777. [Google Scholar] [CrossRef] [PubMed]
  37. Beji, A.; Horst, D.; Engel, J.; Kirchner, T.; Ullrich, A. Toward the prognostic significance and therapeutic potential of HER3 receptor tyrosine kinase in human colon cancer. Clin. Cancer Res. 2012, 18, 956–968. [Google Scholar] [CrossRef]
  38. Lee-Hoeflich, S.T.; Crocker, L.; Yao, E.; Pham, T.; Munroe, X.; Hoeflich, K.P.; Sliwkowski, M.X.; Stern, H.M. A central role for HER3 in HER2-amplified breast cancer: Implications for targeted therapy. Cancer Res. 2008, 68, 5878–5887. [Google Scholar] [CrossRef]
  39. 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]
  40. Quinn, C.M.; Ostrowski, J.L.; Lane, S.A.; Loney, D.P.; Teasdale, J.; Benson, F.A. c-erbB-3 protein expression in human breast cancer: Comparison with other tumour variables and survival. Histopathology 1994, 25, 247–252. [Google Scholar] [CrossRef]
  41. Naidu, R.; Yadav, M.; Nair, S.; Kutty, M.K. Expression of c-erbB3 protein in primary breast carcinomas. Br. J. Cancer 1998, 78, 1385–1390. [Google Scholar] [CrossRef]
  42. Barnes, N.L.; Khavari, S.; Boland, G.P.; Cramer, A.; Knox, W.F.; Bundred, N.J. Absence of HER4 expression predicts recurrence of ductal carcinoma in situ of the breast. Clin. Cancer Res. 2005, 11, 2163–2168. [Google Scholar] [CrossRef] [PubMed]
  43. Lemoine, N.R.; Barnes, D.M.; Hollywood, D.P.; Hughes, C.M.; Smith, P.; Dublin, E.; Prigent, S.A.; Gullick, W.J.; Hurst, H.C. Expression of the ERBB3 gene product in breast cancer. Br. J. Cancer 1992, 66, 1116–1121. [Google Scholar] [CrossRef] [PubMed]
  44. Travis, A.; Pinder, S.E.; Robertson, J.F.; Bell, J.A.; Wencyk, P.; Gullick, W.J.; Nicholson, R.I.; Poller, D.N.; Blamey, R.W.; Elston, C.W.; et al. C-erbB-3 in human breast carcinoma: Expression and relation to prognosis and established prognostic indicators. Br. J. Cancer 1996, 74, 229–233. [Google Scholar] [CrossRef]
  45. Ciardiello, F.; Kim, N.; Saeki, T.; Dono, R.; Persico, M.G.; Plowman, G.D.; Garrigues, J.; Radke, S.; Todaro, G.J.; Salomon, D.S. Differential expression of epidermal growth factor-related proteins in human colorectal tumors. Proc. Natl. Acad. Sci. USA 1991, 88, 7792–7796. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, J.C.; Wang, S.T.; Chow, N.H.; Yang, H.B. Investigation of the prognostic value of coexpressed erbB family members for the survival of colorectal cancer patients after curative surgery. Eur. J. Cancer 2002, 38, 1065–1071. [Google Scholar] [CrossRef]
  47. Kountourakis, P.; Pavlakis, K.; Psyrri, A.; Rontogianni, D.; Xiros, N.; Patsouris, E.; Pectasides, D.; Economopoulos, T. Prognostic significance of HER3 and HER4 protein expression in colorectal adenocarcinomas. BMC Cancer 2006, 6, 46. [Google Scholar] [CrossRef]
  48. Grivas, P.D.; Antonacopoulou, A.; Tzelepi, V.; Sotiropoulou-Bonikou, G.; Kefalopoulou, Z.; Papavassiliou, A.G.; Kalofonos, H. HER-3 in colorectal tumourigenesis: From mRNA levels through protein status to clinicopathologic relationships. Eur. J. Cancer 2007, 43, 2602–2611. [Google Scholar] [CrossRef]
  49. Shintani, S.; Funayama, T.; Yoshihama, Y.; Alcalde, R.E.; Matsumura, T. Prognostic significance of ERBB3 overexpression in oral squamous cell carcinoma. Cancer Lett. 1995, 95, 79–83. [Google Scholar] [CrossRef]
  50. Funayama, T.; Nakanishi, T.; Takahashi, K.; Taniguchi, S.; Takigawa, M.; Matsumura, T. Overexpression of c-erbB-3 in various stages of human squamous cell carcinomas. Oncology 1998, 55, 161–167. [Google Scholar] [CrossRef]
  51. Alaoui-Jamali, M.A.; Song, D.J.; Benlimame, N.; Yen, L.; Deng, X.; Hernandez-Perez, M.; Wang, T. Regulation of multiple tumor microenvironment markers by overexpression of single or paired combinations of ErbB receptors. Cancer Res. 2003, 63, 3764–3774. [Google Scholar] [PubMed]
  52. Begnami, M.D.; Fukuda, E.; Fregnani, J.H.; Nonogaki, S.; Montagnini, A.L.; da Costa, W.L., Jr.; Soares, F.A. Prognostic implications of altered human epidermal growth factor receptors (HERs) in gastric carcinomas: HER2 and HER3 are predictors of poor outcome. J. Clin. Oncol. 2011, 29, 3030–3036. [Google Scholar] [CrossRef]
  53. Koumakpayi, I.H.; Diallo, J.S.; Le Page, C.; Lessard, L.; Gleave, M.; Bégin, L.R.; Mes-Masson, A.M.; Saad, F. Expression and nuclear localization of ErbB3 in prostate cancer. Clin. Cancer Res. 2006, 12, 2730–2737. [Google Scholar] [CrossRef] [PubMed]
  54. Trocmé, E.; Mougiakakos, D.; Johansson, C.C.; All-Eriksson, C.; Economou, M.A.; Larsson, O.; Seregard, S.; Kiessling, R.; Lin, Y. Nuclear HER3 is associated with favorable overall survival in uveal melanoma. Int. J. Cancer 2012, 130, 1120–1127. [Google Scholar] [CrossRef] [PubMed]
  55. Takikita, M.; Xie, R.; Chung, J.Y.; Cho, H.; Ylaya, K.; Hong, S.M.; Moskaluk, C.A.; Hewitt, S.M. Membranous expression of Her3 is associated with a decreased survival in head and neck squamous cell carcinoma. J. Transl. Med. 2011, 9, 126. [Google Scholar] [CrossRef] [PubMed]
  56. Hirakawa, T.; Nakata, B.; Amano, R.; Kimura, K.; Shimizu, S.; Ohira, G.; Yamada, N.; Ohira, M.; Hirakawa, K. HER3 overexpression as an independent indicator of poor prognosis for patients with curatively resected pancreatic cancer. Oncology 2011, 81, 192–198. [Google Scholar] [CrossRef]
  57. Spears, M.; Taylor, K.J.; Munro, A.F.; Cunningham, C.A.; Mallon, E.A.; Twelves, C.J.; Cameron, D.A.; Thomas, J.; Bartlett, J.M. In situ detection of HER2:HER2 and HER2:HER3 protein-protein interactions demonstrates prognostic significance in early breast cancer. Breast Cancer Res. Treat. 2012, 132, 463–470. [Google Scholar] [CrossRef]
  58. Witton, C.J.; Reeves, J.R.; Going, J.J.; Cooke, T.G.; Bartlett, J.M. Expression of the HER1-4 family of receptor tyrosine kinases in breast cancer. J. Pathol. 2003, 200, 290–297. [Google Scholar] [CrossRef]
  59. Baiocchi, G.; Lopes, A.; Coudry, R.A.; Rossi, B.M.; Soares, F.A.; Aguiar, S.; Guimarães, G.C.; Ferreira, F.O.; Nakagawa, W.T. ErbB family immunohistochemical expression in colorectal cancer patients with higher risk of recurrence after radical surgery. Int. J. Color. Dis. 2009, 24, 1059–1068. [Google Scholar] [CrossRef]
  60. Reschke, M.; Mihic-Probst, D.; van der Horst, E.H.; Knyazev, P.; Wild, P.J.; Hutterer, M.; Meyer, S.; Dummer, R.; Moch, H.; Ullrich, A. HER3 is a determinant for poor prognosis in melanoma. Clin. Cancer Res. 2008, 14, 5188–5197. [Google Scholar] [CrossRef]
  61. Hayashi, M.; Inokuchi, M.; Takagi, Y.; Yamada, H.; Kojima, K.; Kumagai, J.; Kawano, T.; Sugihara, K. High expression of HER3 is associated with a decreased survival in gastric cancer. Clin. Cancer Res. 2008, 14, 7843–7849. [Google Scholar] [CrossRef]
  62. Tanner, B.; Hasenclever, D.; Stern, K.; Schormann, W.; Bezler, M.; Hermes, M.; Brulport, M.; Bauer, A.; Schiffer, I.B.; Gebhard, S.; et al. ErbB-3 predicts survival in ovarian cancer. J. Clin. Oncol. 2006, 24, 4317–4323. [Google Scholar] [CrossRef]
  63. Lee, C.M.; Shrieve, D.C.; Zempolich, K.A.; Lee, R.J.; Hammond, E.; Handrahan, D.L.; Gaffney, D.K. Correlation between human epidermal growth factor receptor family (EGFR, HER2, HER3, HER4), phosphorylated Akt (P-Akt), and clinical outcomes after radiation therapy in carcinoma of the cervix. Gynecol. Oncol. 2005, 99, 415–421. [Google Scholar] [CrossRef] [PubMed]
  64. Majumder, A.; Singh, M.; Behera, J.; Theilen, N.T.; George, A.K.; Tyagi, N.; Metreveli, N.; Tyagi, S.C. Hydrogen sulfide alleviates hyperhomocysteinemia-mediated skeletal muscle atrophy via mitigation of oxidative and endoplasmic reticulum stress injury. Am. J. Physiol. Cell Physiol. 2018, 315, C609–C622. [Google Scholar] [CrossRef] [PubMed]
  65. Majumder, A.; Singh, M.; George, A.K.; Behera, J.; Tyagi, N.; Tyagi, S.C. Hydrogen sulfide improves postischemic neoangiogenesis in the hind limb of cystathionine-β-synthase mutant mice via PPAR-γ/VEGF axis. Physiol. Rep. 2018, 6, e13858. [Google Scholar] [CrossRef] [PubMed]
  66. Majumder, A.; Singh, M.; George, A.K.; Tyagi, S.C. Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/ER stress condition (1). Can. J. Physiol. Pharmacol. 2019, 97, 441–456. [Google Scholar] [CrossRef]
  67. Sithanandam, G.; Anderson, L.M. The ERBB3 receptor in cancer and cancer gene therapy. Cancer Gene Ther. 2008, 15, 413–448. [Google Scholar] [CrossRef]
  68. Fiddes, R.J.; Campbell, D.H.; Janes, P.W.; Sivertsen, S.P.; Sasaki, H.; Wallasch, C.; Daly, R.J. Analysis of Grb7 recruitment by heregulin-activated erbB receptors reveals a novel target selectivity for erbB3. J. Biol. Chem. 1998, 273, 7717–7724. [Google Scholar] [CrossRef] [PubMed]
  69. Schulze, W.X.; Deng, L.; Mann, M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol. Syst. Biol. 2005, 1, 2005.0008. [Google Scholar] [CrossRef]
  70. Vijapurkar, U.; Cheng, K.; Koland, J.G. Mutation of a Shc binding site tyrosine residue in ErbB3/HER3 blocks heregulin-dependent activation of mitogen-activated protein kinase. J. Biol. Chem. 1998, 273, 20996–21002. [Google Scholar] [CrossRef]
  71. Zhang, K.; Sun, J.; Liu, N.; Wen, D.; Chang, D.; Thomason, A.; Yoshinaga, S.K. Transformation of NIH 3T3 cells by HER3 or HER4 receptors requires the presence of HER1 or HER2. J. Biol. Chem. 1996, 271, 3884–3890. [Google Scholar] [CrossRef] [PubMed]
  72. Jeong, E.G.; Soung, Y.H.; Lee, J.W.; Lee, S.H.; Nam, S.W.; Lee, J.Y.; Yoo, N.J.; Lee, S.H. ERBB3 kinase domain mutations are rare in lung, breast and colon carcinomas. Int. J. Cancer 2006, 119, 2986–2987. [Google Scholar] [CrossRef]
  73. Alimandi, M.; Romano, A.; Curia, M.C.; Muraro, R.; Fedi, P.; Aaronson, S.A.; Di Fiore, P.P.; Kraus, M.H. Cooperative signaling of ErbB3 and ErbB2 in neoplastic transformation and human mammary carcinomas. Oncogene 1995, 10, 1813–1821. [Google Scholar] [PubMed]
  74. Holbro, T.; Beerli, R.R.; Maurer, F.; Koziczak, M.; Barbas, C.F., 3rd; Hynes, N.E. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2003, 100, 8933–8938. [Google Scholar] [CrossRef]
  75. Liles, J.S.; Arnoletti, J.P.; Tzeng, C.W.; Howard, J.H.; Kossenkov, A.V.; Kulesza, P.; Heslin, M.J.; Frolov, A. ErbB3 expression promotes tumorigenesis in pancreatic adenocarcinoma. Cancer Biol. Ther. 2010, 10, 555–563. [Google Scholar] [CrossRef]
  76. Wilson, T.R.; Lee, D.Y.; Berry, L.; Shames, D.S.; Settleman, J. Neuregulin-1-mediated autocrine signaling underlies sensitivity to HER2 kinase inhibitors in a subset of human cancers. Cancer Cell 2011, 20, 158–172. [Google Scholar] [CrossRef]
  77. Majumder, A.; Sandhu, M.; Banerji, D.; Steri, V.; Olshen, A.; Moasser, M.M. The role of HER2 and HER3 in HER2-amplified cancers beyond breast cancers. Sci. Rep. 2021, 11, 9091. [Google Scholar] [CrossRef]
  78. Chakrabarty, A.; Rexer, B.N.; Wang, S.E.; Cook, R.S.; Engelman, J.A.; Arteaga, C.L. H1047R phosphatidylinositol 3-kinase mutant enhances HER2-mediated transformation by heregulin production and activation of HER3. Oncogene 2010, 29, 5193–5203. [Google Scholar] [CrossRef]
  79. Ritter, C.A.; Perez-Torres, M.; Rinehart, C.; Guix, M.; Dugger, T.; Engelman, J.A.; Arteaga, C.L. Human breast cancer cells selected for resistance to trastuzumab in vivo overexpress epidermal growth factor receptor and ErbB ligands and remain dependent on the ErbB receptor network. Clin. Cancer Res. 2007, 13, 4909–4919. [Google Scholar] [CrossRef] [PubMed]
  80. Révillion, F.; Lhotellier, V.; Hornez, L.; Bonneterre, J.; Peyrat, J.P. ErbB/HER ligands in human breast cancer, and relationships with their receptors, the bio-pathological features and prognosis. Ann. Oncol. 2008, 19, 73–80. [Google Scholar] [CrossRef]
  81. Wilson, T.R.; Fridlyand, J.; Yan, Y.; Penuel, E.; Burton, L.; Chan, E.; Peng, J.; Lin, E.; Wang, Y.; Sosman, J.; et al. Widespread potential for growth-factor-driven resistance to anti-cancer kinase inhibitors. Nature 2012, 487, 505–509. [Google Scholar] [CrossRef]
  82. Motoyama, A.B.; Hynes, N.E.; Lane, H.A. The efficacy of ErbB receptor-targeted anti-cancer therapeutics is influenced by the availability of epidermal growth factor-related peptides. Cancer Res. 2002, 62, 3151–3158. [Google Scholar] [PubMed]
  83. Phillips, G.D.; Fields, C.T.; Li, G.; Dowbenko, D.; Schaefer, G.; Miller, K.; Andre, F.; Burris, H.A., 3rd; Albain, K.S.; Harbeck, N.; et al. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: Critical role for neuregulin blockade in antitumor response to combination therapy. Clin. Cancer Res. 2014, 20, 456–468. [Google Scholar] [CrossRef] [PubMed]
  84. Elkabets, M.; Vora, S.; Juric, D.; Morse, N.; Mino-Kenudson, M.; Muranen, T.; Tao, J.; Campos, A.B.; Rodon, J.; Ibrahim, Y.H.; et al. mTORC1 inhibition is required for sensitivity to PI3K p110α inhibitors in PIK3CA-mutant breast cancer. Sci. Transl. Med. 2013, 5, 196ra199. [Google Scholar] [CrossRef]
  85. Lin, M.C.; Rojas, K.S.; Cerione, R.A.; Wilson, K.F. Identification of mTORC2 as a necessary component of HRG/ErbB2-dependent cellular transformation. Mol. Cancer Res. 2014, 12, 940–952. [Google Scholar] [CrossRef] [PubMed]
  86. Sato, Y.; Yashiro, M.; Takakura, N. Heregulin induces resistance to lapatinib-mediated growth inhibition of HER2-amplified cancer cells. Cancer Sci. 2013, 104, 1618–1625. [Google Scholar] [CrossRef]
  87. Yonesaka, K.; Zejnullahu, K.; Okamoto, I.; Satoh, T.; Cappuzzo, F.; Souglakos, J.; Ercan, D.; Rogers, A.; Roncalli, M.; Takeda, M.; et al. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci. Transl. Med. 2011, 3, 99ra86. [Google Scholar] [CrossRef]
  88. Ono, M.; Kuwano, M. Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin. Cancer Res. 2006, 12, 7242–7251. [Google Scholar] [CrossRef]
  89. Vermorken, J.B.; Mesia, R.; Rivera, F.; Remenar, E.; Kawecki, A.; Rottey, S.; Erfan, J.; Zabolotnyy, D.; Kienzer, H.R.; Cupissol, D.; et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N. Engl. J. Med. 2008, 359, 1116–1127. [Google Scholar] [CrossRef]
  90. Leighl, N.B. Treatment paradigms for patients with metastatic non-small-cell lung cancer: First-, second-, and third-line. Curr. Oncol. 2012, 19, S52–S58. [Google Scholar] [CrossRef]
  91. Vincent, M.D.; Kuruvilla, M.S.; Leighl, N.B.; Kamel-Reid, S. Biomarkers that currently affect clinical practice: EGFR, ALK, MET, KRAS. Curr. Oncol. 2012, 19, S33–S44. [Google Scholar] [CrossRef]
  92. Murphy, C.G.; Morris, P.G. Recent advances in novel targeted therapies for HER2-positive breast cancer. Anti-Cancer Drugs 2012, 23, 765–776. [Google Scholar] [CrossRef] [PubMed]
  93. Van Cutsem, E.; Bang, Y.J.; Feng-Yi, F.; Xu, J.M.; Lee, K.W.; Jiao, S.C.; Chong, J.L.; López-Sanchez, R.I.; Price, T.; Gladkov, O.; et al. HER2 screening data from ToGA: Targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 2015, 18, 476–484. [Google Scholar] [CrossRef] [PubMed]
  94. Abel, E.V.; Basile, K.J.; Kugel, C.H., 3rd; Witkiewicz, A.K.; Le, K.; Amaravadi, R.K.; Karakousis, G.C.; Xu, X.; Xu, W.; Schuchter, L.M.; et al. Melanoma adapts to RAF/MEK inhibitors through FOXD3-mediated upregulation of ERBB3. J. Clin. Investig. 2013, 123, 2155–2168. [Google Scholar] [CrossRef] [PubMed]
  95. Xia, W.; Petricoin, E.F., 3rd; Zhao, S.; Liu, L.; Osada, T.; Cheng, Q.; Wulfkuhle, J.D.; Gwin, W.R.; Yang, X.; Gallagher, R.I.; et al. An heregulin-EGFR-HER3 autocrine signaling axis can mediate acquired lapatinib resistance in HER2+ breast cancer models. Breast Cancer Res. 2013, 15, R85. [Google Scholar] [CrossRef]
  96. Gwin, W.R.; Spector, N.L. Pertuzumab protects the achilles’ heel of trastuzumab–emtansine. Clin. Cancer Res. 2014, 20, 278–280. [Google Scholar] [CrossRef]
  97. Vlacich, G.; Coffey, R.J. Resistance to EGFR-targeted therapy: A family affair. Cancer Cell 2011, 20, 423–425. [Google Scholar] [CrossRef]
  98. Prahallad, A.; Sun, C.; Huang, S.; Di Nicolantonio, F.; Salazar, R.; Zecchin, D.; Beijersbergen, R.L.; Bardelli, A.; Bernards, R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012, 483, 100–103. [Google Scholar] [CrossRef]
  99. Prasetyanti, P.R.; Capone, E.; Barcaroli, D.; D’Agostino, D.; Volpe, S.; Benfante, A.; van Hooff, S.; Iacobelli, V.; Rossi, C.; Iacobelli, S.; et al. ErbB-3 activation by NRG-1β sustains growth and promotes vemurafenib resistance in BRAF-V600E colon cancer stem cells (CSCs). Oncotarget 2015, 6, 16902–16911. [Google Scholar] [CrossRef]
  100. Watanabe, S.; Yonesaka, K.; Tanizaki, J.; Nonagase, Y.; Takegawa, N.; Haratani, K.; Kawakami, H.; Hayashi, H.; Takeda, M.; Tsurutani, J.; et al. Targeting of the HER2/HER3 signaling axis overcomes ligand-mediated resistance to trastuzumab in HER2-positive breast cancer. Cancer Med. 2019, 8, 1258–1268. [Google Scholar] [CrossRef]
  101. Liu, B.; Ordonez-Ercan, D.; Fan, Z.; Edgerton, S.M.; Yang, X.; Thor, A.D. Downregulation of erbB3 abrogates erbB2-mediated tamoxifen resistance in breast cancer cells. Int. J. Cancer 2007, 120, 1874–1882. [Google Scholar] [CrossRef]
  102. Hutcheson, I.R.; Goddard, L.; Barrow, D.; McClelland, R.A.; Francies, H.E.; Knowlden, J.M.; Nicholson, R.I.; Gee, J.M. Fulvestrant-induced expression of ErbB3 and ErbB4 receptors sensitizes oestrogen receptor-positive breast cancer cells to heregulin β1. Breast Cancer Res. 2011, 13, R29. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, S.; Huang, W.C.; Li, P.; Guo, H.; Poh, S.B.; Brady, S.W.; Xiong, Y.; Tseng, L.M.; Li, S.H.; Ding, Z.; et al. Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways. Nat. Med. 2011, 17, 461–469. [Google Scholar] [CrossRef]
  104. Chandarlapaty, S.; Sakr, R.A.; Giri, D.; Patil, S.; Heguy, A.; Morrow, M.; Modi, S.; Norton, L.; Rosen, N.; Hudis, C.; et al. Frequent mutational activation of the PI3K-AKT pathway in trastuzumab-resistant breast cancer. Clin. Cancer Res. 2012, 18, 6784–6791. [Google Scholar] [CrossRef] [PubMed]
  105. Ogden, A.; Bhattarai, S.; Sahoo, B.; Mongan, N.P.; Alsaleem, M.; Green, A.R.; Aleskandarany, M.; Ellis, I.O.; Pattni, S.; Li, X.B.; et al. Combined HER3-EGFR score in triple-negative breast cancer provides prognostic and predictive significance superior to individual biomarkers. Sci. Rep. 2020, 10, 3009. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, S.; Huang, X.; Lee, C.K.; Liu, B. Elevated expression of erbB3 confers paclitaxel resistance in erbB2-overexpressing breast cancer cells via upregulation of Survivin. Oncogene 2010, 29, 4225–4236. [Google Scholar] [CrossRef]
  107. Erjala, K.; Sundvall, M.; Junttila, T.T.; Zhang, N.; Savisalo, M.; Mali, P.; Kulmala, J.; Pulkkinen, J.; Grenman, R.; Elenius, K. Signaling via ErbB2 and ErbB3 associates with resistance and epidermal growth factor receptor (EGFR) amplification with sensitivity to EGFR inhibitor gefitinib in head and neck squamous cell carcinoma cells. Clin. Cancer Res. 2006, 12, 4103–4111. [Google Scholar] [CrossRef]
  108. Wilson, F.H.; Johannessen, C.M.; Piccioni, F.; Tamayo, P.; Kim, J.W.; Van Allen, E.M.; Corsello, S.M.; Capelletti, M.; Calles, A.; Butaney, M.; et al. A functional landscape of resistance to ALK inhibition in lung cancer. Cancer Cell 2015, 27, 397–408. [Google Scholar] [CrossRef]
  109. Narayan, M.; Wilken, J.A.; Harris, L.N.; Baron, A.T.; Kimbler, K.D.; Maihle, N.J. Trastuzumab-induced HER reprogramming in “resistant” breast carcinoma cells. Cancer Res. 2009, 69, 2191–2194. [Google Scholar] [CrossRef]
  110. Tovey, S.; Dunne, B.; Witton, C.J.; Forsyth, A.; Cooke, T.G.; Bartlett, J.M. Can molecular markers predict when to implement treatment with aromatase inhibitors in invasive breast cancer? Clin. Cancer Res. 2005, 11, 4835–4842. [Google Scholar] [CrossRef]
  111. Tovey, S.M.; Witton, C.J.; Bartlett, J.M.; Stanton, P.D.; Reeves, J.R.; Cooke, T.G. Outcome and human epidermal growth factor receptor (HER) 1-4 status in invasive breast carcinomas with proliferation indices evaluated by bromodeoxyuridine labelling. Breast Cancer Res. 2004, 6, R246–R251. [Google Scholar] [CrossRef] [PubMed]
  112. Frogne, T.; Benjaminsen, R.V.; Sonne-Hansen, K.; Sorensen, B.S.; Nexo, E.; Laenkholm, A.V.; Rasmussen, L.M.; Riese, D.J., 2nd; de Cremoux, P.; Stenvang, J.; et al. Activation of ErbB3, EGFR and Erk is essential for growth of human breast cancer cell lines with acquired resistance to fulvestrant. Breast Cancer Res. Treat. 2009, 114, 263–275. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Z.; Karthaus, W.R.; Lee, Y.S.; Gao, V.R.; Wu, C.; Russo, J.W.; Liu, M.; Mota, J.M.; Abida, W.; Linton, E.; et al. Tumor Microenvironment-Derived NRG1 Promotes Antiandrogen Resistance in Prostate Cancer. Cancer Cell 2020, 38, 279–296.e279. [Google Scholar] [CrossRef] [PubMed]
  114. Knuefermann, C.; Lu, Y.; Liu, B.; Jin, W.; Liang, K.; Wu, L.; Schmidt, M.; Mills, G.B.; Mendelsohn, J.; Fan, Z. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene 2003, 22, 3205–3212. [Google Scholar] [CrossRef] [PubMed]
  115. Bezler, M.; Hengstler, J.G.; Ullrich, A. Inhibition of doxorubicin-induced HER3-PI3K-AKT signalling enhances apoptosis of ovarian cancer cells. Mol. Oncol. 2012, 6, 516–529. [Google Scholar] [CrossRef]
  116. Yan, Y.; Hein, A.L.; Greer, P.M.; Wang, Z.; Kolb, R.H.; Batra, S.K.; Cowan, K.H. A novel function of HER2/Neu in the activation of G2/M checkpoint in response to γ-irradiation. Oncogene 2015, 34, 2215–2226. [Google Scholar] [CrossRef]
  117. He, G.; Di, X.; Yan, J.; Zhu, C.; Sun, X.; Zhang, S. Silencing human epidermal growth factor receptor-3 radiosensitizes human luminal A breast cancer cells. Cancer Sci. 2018, 109, 3774–3782. [Google Scholar] [CrossRef]
  118. Casalini, P.; Iorio, M.V.; Galmozzi, E.; Ménard, S. Role of HER receptors family in development and differentiation. J. Cell Physiol. 2004, 200, 343–350. [Google Scholar] [CrossRef]
  119. Armstrong, E.J.; Bischoff, J. Heart valve development: Endothelial cell signaling and differentiation. Circ. Res. 2004, 95, 459–470. [Google Scholar] [CrossRef]
  120. Fouladkou, F.; Lu, C.; Jiang, C.; Zhou, L.; She, Y.; Walls, J.R.; Kawabe, H.; Brose, N.; Henkelman, R.M.; Huang, A.; et al. The ubiquitin ligase Nedd4-1 is required for heart development and is a suppressor of thrombospondin-1. J. Biol. Chem. 2010, 285, 6770–6780. [Google Scholar] [CrossRef]
  121. Erickson, S.L.; O’Shea, K.S.; Ghaboosi, N.; Loverro, L.; Frantz, G.; Bauer, M.; Lu, L.H.; Moore, M.W. ErbB3 is required for normal cerebellar and cardiac development: A comparison with ErbB2-and heregulin-deficient mice. Development 1997, 124, 4999–5011. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. Graus-Porta, D.; Beerli, R.R.; Daly, J.M.; Hynes, N.E. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997, 16, 1647–1655. [Google Scholar] [CrossRef]
  124. Prenzel, N.; Fischer, O.M.; Streit, S.; Hart, S.; Ullrich, A. The epidermal growth factor receptor family as a central element for cellular signal transduction and diversification. Endocr. Relat. Cancer 2001, 8, 11–31. [Google Scholar] [CrossRef]
  125. Tzahar, E.; Waterman, H.; Chen, X.; Levkowitz, G.; Karunagaran, D.; Lavi, S.; Ratzkin, B.J.; Yarden, Y. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol. Cell Biol. 1996, 16, 5276–5287. [Google Scholar] [CrossRef] [PubMed]
  126. Lee, Y.; Ma, J.; Lyu, H.; Huang, J.; Kim, A.; Liu, B. Role of erbB3 receptors in cancer therapeutic resistance. Acta Biochim. Biophys. Sinica 2014, 46, 190–198. [Google Scholar] [CrossRef]
  127. Karachaliou, N.; Lazzari, C.; Verlicchi, A.; Sosa, A.E.; Rosell, R. HER3 as a Therapeutic Target in Cancer. BioDrugs 2017, 31, 63–73. [Google Scholar] [CrossRef] [PubMed]
  128. Shi, F.; Telesco, S.E.; Liu, Y.; Radhakrishnan, R.; Lemmon, M.A. ErbB3/HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc. Natl. Acad. Sci. USA 2010, 107, 7692–7697. [Google Scholar] [CrossRef]
  129. Jacob, W.; James, I.; Hasmann, M.; Weisser, M. Clinical development of HER3-targeting monoclonal antibodies: Perils and progress. Cancer Treat. Rev. 2018, 68, 111–123. [Google Scholar] [CrossRef]
  130. Schoeberl, B.; Pace, E.A.; Fitzgerald, J.B.; Harms, B.D.; Xu, L.; Nie, L.; Linggi, B.; Kalra, A.; Paragas, V.; Bukhalid, R.; et al. Therapeutically targeting ErbB3: A key node in ligand-induced activation of the ErbB receptor-PI3K axis. Sci. Signal 2009, 2, ra31. [Google Scholar] [CrossRef]
  131. Garner, A.P.; Bialucha, C.U.; Sprague, E.R.; Garrett, J.T.; Sheng, Q.; Li, S.; Sineshchekova, O.; Saxena, P.; Sutton, C.R.; Chen, D.; et al. An antibody that locks HER3 in the inactive conformation inhibits tumor growth driven by HER2 or neuregulin. Cancer Res. 2013, 73, 6024–6035. [Google Scholar] [CrossRef] [PubMed]
  132. LoRusso, P.; Jänne, P.A.; Oliveira, M.; Rizvi, N.; Malburg, L.; Keedy, V.; Yee, L.; Copigneaux, C.; Hettmann, T.; Wu, C.Y.; et al. Phase I study of U3-1287, a fully human anti-HER3 monoclonal antibody, in patients with advanced solid tumors. Clin. Cancer Res. 2013, 19, 3078–3087. [Google Scholar] [CrossRef] [PubMed]
  133. Denlinger, C.S.; Keedy, V.L.; Moyo, V.; MacBeath, G.; Shapiro, G.I. Phase 1 dose escalation study of seribantumab (MM-121), an anti-HER3 monoclonal antibody, in patients with advanced solid tumors. Investig. New Drugs 2021, 39, 1604–1612. [Google Scholar] [CrossRef]
  134. Meulendijks, D.; Jacob, W.; Martinez-Garcia, M.; Taus, A.; Lolkema, M.P.; Voest, E.E.; Langenberg, M.H.; Fleitas Kanonnikoff, T.; Cervantes, A.; De Jonge, M.J.; et al. First-in-Human Phase I Study of Lumretuzumab, a Glycoengineered Humanized Anti-HER3 Monoclonal Antibody, in Patients with Metastatic or Advanced HER3-Positive Solid Tumors. Clin. Cancer Res. 2016, 22, 877–885. [Google Scholar] [CrossRef]
  135. Liles, J.S.; Arnoletti, J.P.; Kossenkov, A.V.; Mikhaylina, A.; Frost, A.R.; Kulesza, P.; Heslin, M.J.; Frolov, A. Targeting ErbB3-mediated stromal-epithelial interactions in pancreatic ductal adenocarcinoma. Br. J. Cancer 2011, 105, 523–533. [Google Scholar] [CrossRef] [PubMed]
  136. Schoeberl, B.; Faber, A.C.; Li, D.; Liang, M.C.; Crosby, K.; Onsum, M.; Burenkova, O.; Pace, E.; Walton, Z.; Nie, L.; et al. An ErbB3 antibody, MM-121, is active in cancers with ligand-dependent activation. Cancer Res. 2010, 70, 2485–2494. [Google Scholar] [CrossRef]
  137. Arnett, S.O.; Teillaud, J.L.; Wurch, T.; Reichert, J.M.; Dunlop, C.; Huber, M. IBC’s 21st Annual Antibody Engineering and 8th Annual Antibody Therapeutics International Conferences and 2010 Annual Meeting of the Antibody Society, 5–9 December 2010, San Diego, CA, USA. MAbs 2011, 3, 133–152. [Google Scholar] [CrossRef]
  138. Jathal, M.K.; Chen, L.; Mudryj, M.; Ghosh, P.M. Targeting ErbB3: The New RTK(id) on the Prostate Cancer Block. Immunol. Endocr. Metab. Agents Med. Chem. 2011, 11, 131–149. [Google Scholar] [CrossRef]
  139. Sithanandam, G.; Fornwald, L.W.; Fields, J.; Anderson, L.M. Inactivation of ErbB3 by siRNA promotes apoptosis and attenuates growth and invasiveness of human lung adenocarcinoma cell line A549. Oncogene 2005, 24, 1847–1859. [Google Scholar] [CrossRef]
  140. Cejalvo, J.M.; Jacob, W.; Fleitas Kanonnikoff, T.; Felip, E.; Navarro Mendivil, A.; Martinez Garcia, M.; Taus Garcia, A.; Leighl, N.; Lassen, U.; Mau-Soerensen, M.; et al. A phase Ib/II study of HER3-targeting lumretuzumab in combination with carboplatin and paclitaxel as first-line treatment in patients with advanced or metastatic squamous non-small cell lung cancer. ESMO Open 2019, 4, e000532. [Google Scholar] [CrossRef]
  141. Schneeweiss, A.; Park-Simon, T.W.; Albanell, J.; Lassen, U.; Cortés, J.; Dieras, V.; May, M.; Schindler, C.; Marmé, F.; Cejalvo, J.M.; et al. Phase Ib study evaluating safety and clinical activity of the anti-HER3 antibody lumretuzumab combined with the anti-HER2 antibody pertuzumab and paclitaxel in HER3-positive, HER2-low metastatic breast cancer. Investig. New Drugs 2018, 36, 848–859. [Google Scholar] [CrossRef]
  142. Kim, H.S.; Han, J.Y.; Shin, D.H.; Lim, K.Y.; Lee, G.K.; Kim, J.Y.; Jacob, W.; Ceppi, M.; Weisser, M.; James, I. EGFR and HER3 signaling blockade in invasive mucinous lung adenocarcinoma harboring an NRG1 fusion. Lung Cancer 2018, 124, 71–75. [Google Scholar] [CrossRef]
  143. Meulendijks, D.; Jacob, W.; Voest, E.E.; Mau-Sorensen, M.; Martinez-Garcia, M.; Taus, A.; Fleitas, T.; Cervantes, A.; Lolkema, M.P.; Langenberg, M.H.G.; et al. Phase Ib Study of Lumretuzumab Plus Cetuximab or Erlotinib in Solid Tumor Patients and Evaluation of HER3 and Heregulin as Potential Biomarkers of Clinical Activity. Clin. Cancer Res. 2017, 23, 5406–5415. [Google Scholar] [CrossRef]
  144. Kim, S.; Keam, B.; Shin, S.; Chae, Y.; Seo, S.; Park, K.; Kim, T.; Park, L.; Hong, S.; Lim, E. 928P Phase I dose-expansion (part II) study of ISU104 (a novel anti-ErbB3 monoclonal antibody) alone and combination with cetuximab (CET), in patients (pts) with recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). Ann. Oncol. 2020, 31, S667–S668. [Google Scholar] [CrossRef]
  145. Falchook, G.S.; Bauer, T.M.; LoRusso, P.; McLaughlin, J.F.; LaVallee, T.; Peck, R.A.; Eder, J.P. Safety, pharmacokinetics (PK), pharmacodynamics (Pd), and antitumor activity in a phase 1b study evaluating anti-ErbB3 antibody KTN3379 in adults with advanced tumors alone and with targeted therapies. J. Clin. Oncol. 2016, 34, 2501. [Google Scholar] [CrossRef]
  146. Duvvuri, U.; George, J.; Kim, S.; Alvarado, D.; Neumeister, V.M.; Chenna, A.; Gedrich, R.; Hawthorne, T.; LaVallee, T.; Grandis, J.R.; et al. Molecular and Clinical Activity of CDX-3379, an Anti-ErbB3 Monoclonal Antibody, in Head and Neck Squamous Cell Carcinoma Patients. Clin. Cancer Res. 2019, 25, 5752–5758. [Google Scholar] [CrossRef]
  147. Bauman, J.E.; Saba, N.F.; Wise-Draper, T.M.; Adkins, D.; O’Brien, P.E.; Heath-Chiozzi, M.; Golden, P.; Drescher, J.; Alvarado, D.; Gedrich, R. CDX3379-04: Phase II evaluation of CDX-3379 in combination with cetuximab in patients with advanced head and neck squamous cell carcinoma (HNSCC). J. Clin. Oncol. 2019, 37, 6025. [Google Scholar] [CrossRef]
  148. Tchekmedyian, V.; Dunn, L.; Sherman, E.; Baxi, S.S.; Grewal, R.K.; Larson, S.M.; Pentlow, K.S.; Haque, S.; Tuttle, R.M.; Sabra, M.M.; et al. Enhancing Radioiodine Incorporation in BRAF-Mutant, Radioiodine-Refractory Thyroid Cancers with Vemurafenib and the Anti-ErbB3 Monoclonal Antibody CDX-3379: Results of a Pilot Clinical Trial. Thyroid 2022, 32, 273–282. [Google Scholar] [CrossRef]
  149. Sarantopoulos, J.; Gordon, M.S.; Harvey, R.D.; Sankhala, K.K.; Malik, L.; Mahalingam, D.; Owonikoko, T.K.; Lewis, C.M.; Payumo, F.; Miller, J. First-in-human phase 1 dose-escalation study of AV-203, a monoclonal antibody against ERBB3, in patients with metastatic or advanced solid tumors. J. Clin. Oncol. 2014, 32, 11113. [Google Scholar] [CrossRef]
  150. Drilon, A.; Somwar, R.; Mangatt, B.P.; Edgren, H.; Desmeules, P.; Ruusulehto, A.; Smith, R.S.; Delasos, L.; Vojnic, M.; Plodkowski, A.J.; et al. Response to ERBB3-Directed Targeted Therapy in NRG1-Rearranged Cancers. Cancer Discov. 2018, 8, 686–695. [Google Scholar] [CrossRef]
  151. Menke-van der Houven van Oordt, C.W.; McGeoch, A.; Bergstrom, M.; McSherry, I.; Smith, D.A.; Cleveland, M.; Al-Azzam, W.; Chen, L.; Verheul, H.; Hoekstra, O.S.; et al. Immuno-PET Imaging to Assess Target Engagement: Experience from (89)Zr-Anti-HER3 mAb (GSK2849330) in Patients with Solid Tumors. J. Nucl. Med. 2019, 60, 902–909. [Google Scholar] [CrossRef]
  152. Sequist, L.V.; Gray, J.E.; Harb, W.A.; Lopez-Chavez, A.; Doebele, R.C.; Modiano, M.R.; Jackman, D.M.; Baggstrom, M.Q.; Atmaca, A.; Felip, E.; et al. Randomized Phase II Trial of Seribantumab in Combination with Erlotinib in Patients with EGFR Wild-Type Non-Small Cell Lung Cancer. Oncol. 2019, 24, 1095–1102. [Google Scholar] [CrossRef]
  153. Sequist, L.V.; Janne, P.A.; Huber, R.M.; Gray, J.E.; Felip, E.; Perol, M.; Hirsch, F.R.; Tan, D.S.-W.; Kuesters, G.; Zalutskaya, A. SHERLOC: A phase 2 study of MM-121 plus with docetaxel versus docetaxel alone in patients with heregulin (HRG) positive advanced non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2019, 37, 9036. [Google Scholar] [CrossRef]
  154. Cleary, J.M.; McRee, A.J.; Shapiro, G.I.; Tolaney, S.M.; O’Neil, B.H.; Kearns, J.D.; Mathews, S.; Nering, R.; MacBeath, G.; Czibere, A.; et al. A phase 1 study combining the HER3 antibody seribantumab (MM-121) and cetuximab with and without irinotecan. Investig. New Drugs 2017, 35, 68–78. [Google Scholar] [CrossRef]
  155. Liu, J.F.; Ray-Coquard, I.; Selle, F.; Poveda, A.M.; Cibula, D.; Hirte, H.; Hilpert, F.; Raspagliesi, F.; Gladieff, L.; Harter, P.; et al. Randomized Phase II Trial of Seribantumab in Combination with Paclitaxel in Patients with Advanced Platinum-Resistant or -Refractory Ovarian Cancer. J. Clin. Oncol. 2016, 34, 4345–4353. [Google Scholar] [CrossRef] [PubMed]
  156. Higgins, M.J.; Doyle, C.; Paepke, S.; Azaro, A.; Martin, M.; Semiglazov, V.; Smirnova, I.; Krasnozhon, D.; Manikhas, A.; Harb, W.A. A randomized, double-blind phase II trial of exemestane plus MM-121 (a monoclonal antibody targeting ErbB3) or placebo in postmenopausal women with locally advanced or metastatic ER+/PR+, HER2-negative breast cancer. J. Clin. Oncol. 2014, 32, 15. [Google Scholar] [CrossRef]
  157. Arnedos, M.; Denlinger, C.S.; Harb, W.A.; Rixe, O.; Morris, J.C.; Dy, G.K.; Adjei, A.A.; Pearlberg, J.; Follows, S.; Czibere, A.G. A phase I study of MM-121 in combination with multiple anticancer therapies in patients with advanced solid tumors. J. Clin. Oncol. 2013, 31, 2609. [Google Scholar] [CrossRef]
  158. Papadopoulos, K.P.; Moore, K.N.; Lush, R.; Desai, M.; Mahmood, S.; Beckman, R.A.; Mendell-Harary, J. Pharmacokinetics, safety, and tolerability of a new patritumab formulation in patients with advanced, refractory solid tumors. J. Clin. Oncol. 2015, 33, e14026. [Google Scholar] [CrossRef]
  159. Forster, M.D.; Dillon, M.T.; Kocsis, J.; Remenár, É.; Pajkos, G.; Rolland, F.; Greenberg, J.; Harrington, K.J. Patritumab or placebo, with cetuximab plus platinum therapy in recurrent or metastatic squamous cell carcinoma of the head and neck: A randomised phase II study. Eur. J. Cancer. 2019, 123, 36–47. [Google Scholar] [CrossRef]
  160. Dillon, M.T.; Grove, L.; Newbold, K.L.; Shaw, H.; Brown, N.F.; Mendell, J.; Chen, S.; Beckman, R.A.; Jennings, A.; Ricamara, M.; et al. Patritumab with Cetuximab plus Platinum-Containing Therapy in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: An Open-Label, Phase Ib Study. Clin. Cancer Res. 2019, 25, 487–495. [Google Scholar] [CrossRef] [PubMed]
  161. Lockhart, A.C.; Liu, Y.; Dehdashti, F.; Laforest, R.; Picus, J.; Frye, J.; Trull, L.; Belanger, S.; Desai, M.; Mahmood, S.; et al. Phase 1 Evaluation of [(64)Cu]DOTA-Patritumab to Assess Dosimetry, Apparent Receptor Occupancy, and Safety in Subjects with Advanced Solid Tumors. Mol. Imaging Biol. 2016, 18, 446–453. [Google Scholar] [CrossRef]
  162. Im, S.-A.; Juric, D.; Baselga, J.; Kong, A.; Martin, P.; Lin, C.-C.; Dees, E.C.; Schellens, J.H.; De Braud, F.G.; Delgado, L. A phase 1 dose-escalation study of anti-HER3 monoclonal antibody LJM716 in combination with trastuzumab in patients with HER2-overexpressing metastatic breast or gastric cancer. J. Clin. Oncol. 2014, 32, 2519. [Google Scholar] [CrossRef]
  163. Shah, P.D.; Chandarlapaty, S.; Dickler, M.N.; Ulaner, G.; Zamora, S.J.; Sterlin, V.; Iasonos, A.; Coughlin, C.M.; Morozov, A.; Ero, J. Phase I study of LJM716, BYL719, and trastuzumab in patients (pts) with HER2-amplified (HER2+) metastatic breast cancer (MBC). J. Clin. Oncol. 2015, 33, 590. [Google Scholar] [CrossRef]
  164. Reynolds, K.L.; Bedard, P.L.; Lee, S.H.; Lin, C.C.; Tabernero, J.; Alsina, M.; Cohen, E.; Baselga, J.; Blumenschein, G., Jr.; Graham, D.M.; et al. A phase I open-label dose-escalation study of the anti-HER3 monoclonal antibody LJM716 in patients with advanced squamous cell carcinoma of the esophagus or head and neck and HER2-overexpressing breast or gastric cancer. BMC Cancer 2017, 17, 646. [Google Scholar] [CrossRef]
  165. Takahashi, S.; Kobayashi, T.; Tomomatsu, J.; Ito, Y.; Oda, H.; Kajitani, T.; Kakizume, T.; Tajima, T.; Takeuchi, H.; Maacke, H.; et al. LJM716 in Japanese patients with head and neck squamous cell carcinoma or HER2-overexpressing breast or gastric cancer. Cancer Chemother. Pharmacol. 2017, 79, 131–138. [Google Scholar] [CrossRef] [PubMed]
  166. Papadopoulos, K.P.; Adjei, A.A.; Rasco, D.W.; Liu, L.; Kao, R.J.; Brownstein, C.M.; DiCioccio, A.T.; Lowy, I.; Trail, P.; Wang, D. Phase 1 study of REGN1400 (anti-ErbB3) combined with erlotinib or cetuximab in patients (pts) with advanced non-small cell lung cancer (NSCLC), colorectal cancer (CRC), or head and neck cancer (SCCHN). J. Clin. Oncol. 2014, 32, 2516. [Google Scholar] [CrossRef]
  167. Mukai, H.; Saeki, T.; Aogi, K.; Naito, Y.; Matsubara, N.; Shigekawa, T.; Ueda, S.; Takashima, S.; Hara, F.; Yamashita, T.; et al. Patritumab plus trastuzumab and paclitaxel in human epidermal growth factor receptor 2-overexpressing metastatic breast cancer. Cancer Sci. 2016, 107, 1465–1470. [Google Scholar] [CrossRef]
  168. von Pawel, J.; Jotte, R.; Spigel, D.R.; O’Brien, M.E.; Socinski, M.A.; Mezger, J.; Steins, M.; Bosquée, L.; Bubis, J.; Nackaerts, K.; et al. Randomized phase III trial of amrubicin versus topotecan as second-line treatment for patients with small-cell lung cancer. J. Clin. Oncol. 2014, 32, 4012–4019. [Google Scholar] [CrossRef]
  169. Paz-Arez, L.; Serwatowski, P.; Szczęsna, A.; Von Pawel, J.; Toschi, L.; Tibor, C.; Morabito, A.; Zhang, L.; Shuster, D.; Chen, S. P3. 02b-045 Patritumab plus erlotinib in EGFR wild-type advanced non–small cell lung cancer (NSCLC): Part a results of HER3-Lung Study: Topic: EGFR Clinical. J. Thorac. Oncol. 2017, 12, S1214–S1215. [Google Scholar] [CrossRef]
  170. Kim, S.-B.; Keam, B.; Shin, S.; Chae, Y.; Kim, T.; Kim, M.-S.; Kim, J.; Park, K.; Ahn, J.; Park, L. First in human, a phase I study of ISU104, a novel ErbB3 monoclonal antibody, in patients with advanced solid tumours. Ann. Oncol. 2019, 30, v168. [Google Scholar] [CrossRef]
  171. Schram, A.M.; O’Reilly, E.M.; Somwar, R.; Benayed, R.; Shameem, S.; Chauhan, T.; Torrisi, J.; Ford, J.; Maussang, D.; Wasserman, E. Abstract PR02: Clinical proof of concept for MCLA-128, a bispecific HER2/3 antibody therapy, in NRG1 fusion-positive cancers. Mol. Cancer Ther. 2019, 18, PR02. [Google Scholar] [CrossRef]
  172. Schram, A.M.; O’Reilly, E.; O’Kane, G. Efficacy and safety of zenocutuzumab in advanced pancreatic cancer and other solid tumors harboring NRG1 fusions. J. Clin. Oncol. 2021, 39, 3003. [Google Scholar] [CrossRef]
  173. Fayette, J.; Wirth, L.; Oprean, C.; Udrea, A.; Jimeno, A.; Rischin, D.; Nutting, C.; Harari, P.M.; Csoszi, T.; Cernea, D.; et al. Randomized Phase II Study of Duligotuzumab (MEHD7945A) vs. Cetuximab in Squamous Cell Carcinoma of the Head and Neck (MEHGAN Study). Front. Oncol. 2016, 6, 232. [Google Scholar] [CrossRef]
  174. Hill, A.G.; Findlay, M.P.; Burge, M.E.; Jackson, C.; Alfonso, P.G.; Samuel, L.; Ganju, V.; Karthaus, M.; Amatu, A.; Jeffery, M.; et al. Phase II Study of the Dual EGFR/HER3 Inhibitor Duligotuzumab (MEHD7945A) versus Cetuximab in Combination with FOLFIRI in Second-Line RAS Wild-Type Metastatic Colorectal Cancer. Clin. Cancer Res. 2018, 24, 2276–2284. [Google Scholar] [CrossRef] [PubMed]
  175. Kundranda, M.; Gracian, A.C.; Zafar, S.F.; Meiri, E.; Bendell, J.; Algül, H.; Rivera, F.; Ahn, E.R.; Watkins, D.; Pelzer, U.; et al. Randomized, double-blind, placebo-controlled phase II study of istiratumab (MM-141) plus nab-paclitaxel and gemcitabine versus nab-paclitaxel and gemcitabine in front-line metastatic pancreatic cancer (CARRIE). Ann. Oncol. 2020, 31, 79–87. [Google Scholar] [CrossRef] [PubMed]
  176. Alsina, M.; Boni, V.; Schellens, J.H.; Moreno, V.; Bol, K.; Westendorp, M.; Sirulnik, L.A.; Tabernero, J.; Calvo, E. First-in-human phase 1/2 study of MCLA-128, a full length IgG1 bispecific antibody targeting HER2 and HER3: Final phase 1 data and preliminary activity in HER2+ metastatic breast cancer (MBC). J. Clin. Oncol. 2017, 35 (Suppl. S15). [Google Scholar] [CrossRef]
  177. Hamilton, E.P.; Petit, T.; Pistilli, B.; Goncalves, A.; Ferreira, A.A.; Dalenc, F.; Cardoso, F.; Mita, M.M.; Dezentjé, V.O.; Manso, L. 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]
  178. Richards, D.A.; Braiteh, F.S.; Garcia, A.; Denlinger, C.S.; Conkling, P.R.; Edenfield, W.J.; Anthony, S.P.; Hellerstedt, B.A.; Raju, R.N.; Becerra, C. A phase 1 study of MM-111, a bispecific HER2/HER3 antibody fusion protein, combined with multiple treatment regimens in patients with advanced HER2-positive solid tumors. J. Clin. Oncol. 2014, 32, 651. [Google Scholar] [CrossRef]
  179. Denlinger, C.S.; Alsina Maqueda, M.; Watkins, D.J.; Sym, S.J.; Bendell, J.C.; Park, S.H.; Arkenau, H.-T.; Bekaii-Saab, T.S.; Kudla, A.J.; McDonagh, C.F. Randomized phase 2 study of paclitaxel (PTX), trastuzumab (T) with or without MM-111 in HER2 expressing gastroesophageal cancers (GEC). J. Clin. Oncol. 2016, 34, 4043. [Google Scholar] [CrossRef]
  180. Isakoff, S.; Bahleda, R.; Saleh, M.; Bordoni, R.; Shields, A.; Dauer, J.; Curley, M.; Baum, J.; McClure, T.; Louis, C. A phase 1 study of MM-141, a novel tetravalent monoclonal antibody targeting IGF-1R and ErbB3, in relapsed or refractory solid tumors. Eur. J. Cancer 2016, 69, S137–S138. [Google Scholar] [CrossRef]
  181. Lieu, C.H.; Hidalgo, M.; Berlin, J.D.; Ko, A.H.; Cervantes, A.; LoRusso, P.; Gerber, D.E.; Eder, J.P.; Eckhardt, S.G.; Kapp, A.V.; et al. A Phase Ib Dose-Escalation Study of the Safety, Tolerability, and Pharmacokinetics of Cobimetinib and Duligotuzumab in Patients with Previously Treated Locally Advanced or Metastatic Cancers with Mutant KRAS. Oncol. 2017, 22, 1024-e1089. [Google Scholar] [CrossRef]
  182. Juric, D.; Dienstmann, R.; Cervantes, A.; Hidalgo, M.; Messersmith, W.; Blumenschein, G.R., Jr.; Tabernero, J.; Roda, D.; Calles, A.; Jimeno, A.; et al. Safety and Pharmacokinetics/Pharmacodynamics of the First-in-Class Dual Action HER3/EGFR Antibody MEHD7945A in Locally Advanced or Metastatic Epithelial Tumors. Clin. Cancer Res. 2015, 21, 2462–2470. [Google Scholar] [CrossRef]
  183. Jimeno, A.; Machiels, J.P.; Wirth, L.; Specenier, P.; Seiwert, T.Y.; Mardjuadi, F.; Wang, X.; Kapp, A.V.; Royer-Joo, S.; Penuel, E.; et al. Phase Ib study of duligotuzumab (MEHD7945A) plus cisplatin/5-fluorouracil or carboplatin/paclitaxel for first-line treatment of recurrent/metastatic squamous cell carcinoma of the head and neck. Cancer 2016, 122, 3803–3811. [Google Scholar] [CrossRef]
  184. Hashimoto, Y.; Koyama, K.; Kamai, Y.; Hirotani, K.; Ogitani, Y.; Zembutsu, A.; Abe, M.; Kaneda, Y.; Maeda, N.; Shiose, Y.; et al. A Novel HER3-Targeting Antibody-Drug Conjugate, U3-1402, Exhibits Potent Therapeutic Efficacy through the Delivery of Cytotoxic Payload by Efficient Internalization. Clin. Cancer Res. 2019, 25, 7151–7161. [Google Scholar] [CrossRef] [PubMed]
  185. Haratani, K.; Yonesaka, K.; Takamura, S.; Maenishi, O.; Kato, R.; Takegawa, N.; Kawakami, H.; Tanaka, K.; Hayashi, H.; Takeda, M.; et al. U3-1402 sensitizes HER3-expressing tumors to PD-1 blockade by immune activation. J. Clin. Invest. 2020, 130, 374–388. [Google Scholar] [CrossRef] [PubMed]
  186. Jänne, P.A.; Baik, C.; Su, W.C.; Johnson, M.L.; Hayashi, H.; Nishio, M.; Kim, D.W.; Koczywas, M.; Gold, K.A.; Steuer, C.E.; et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer. Cancer Discov. 2022, 12, 74–89. [Google Scholar] [CrossRef]
  187. Yu, H.; Baik, C.; Gold, K.; Hayashi, H.; Johnson, M.; Koczywas, M.; Murakami, H.; Nishio, M.; Steuer, C.; Su, W. LBA62 Efficacy and safety of patritumab deruxtecan (U3-1402), a novel HER3 directed antibody drug conjugate, in patients (pts) with EGFR-mutated (EGFRm) NSCLC. Ann. Oncol. 2020, 31, S1189–S1190. [Google Scholar] [CrossRef]
  188. Janne, P.A.; Yu, H.A.; Johnson, M.L.; Steuer, C.E.; Vigliotti, M.; Iacobucci, C.; Chen, S.; Yu, C.; Sellami, D.B. Safety and preliminary antitumor activity of U3-1402: A HER3-targeted antibody drug conjugate in EGFR TKI-resistant, EGFRm NSCLC. J. Clin. Oncol. 2019, 37, 9010. [Google Scholar] [CrossRef]
  189. Masuda, N.; Yonemori, K.; Takahashi, S.; Kogawa, T.; Nakayama, T.; Iwase, H.; Takahashi, M.; Toyama, T.; Saeki, T.; Saji, S. Abstract PD1-03: Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in HER3-overexpressing metastatic breast cancer: Updated results of a phase 1/2 trial. Cancer Res. 2019, 79 (Suppl. S4), PD1-03-PD01-03. [Google Scholar] [CrossRef]
  190. Kogawa, T.; Yonemori, K.; Masuda, N.; Takahashi, S.; Takahashi, M.; Iwase, H.; Nakayama, T.; Saeki, T.; Toyama, T.; Takano, T. Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in breast cancer patients: Phase 1 dose escalation study. J. Clin. Oncol. 2018, 36, 2512. [Google Scholar] [CrossRef]
  191. Krop, I.E.; Masuda, N.; Mukohara, T.; Takahashi, S.; Nakayama, T.; Inoue, K.; Iwata, H.; Toyama, T.; Yamamoto, Y.; Hansra, D.M. Results from the phase 1/2 study of patritumab deruxtecan, a HER3-directed antibody-drug conjugate (ADC), in patients with HER3-expressing metastatic breast cancer (MBC). J. Clin. Oncol. 2022, 40, 16. [Google Scholar] [CrossRef]
  192. Nuciforo, P.; Pascual, T.; Cortés, J.; Llombart-Cussac, A.; Fasani, R.; Paré, L.; Oliveira, M.; Galvan, P.; Martínez, N.; Bermejo, B. A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade. Ann. Oncol. 2018, 29, 170–177. [Google Scholar] [CrossRef] [PubMed]
  193. Prat, A.; Falato, C.; Brunet, L.P.; Saez, O.M.; Andujar, J.C.; Vila, M.M.; Tolosa, P.; Bofill, F.S.; Jurado, J.C.; Gonzalez-Farre, B. LBA3 Patritumab deruxtecan (HER3-DXd) in early-stage HR+/HER2-breast cancer: Final results of the SOLTI TOT-HER3 window of opportunity trial. Ann. Oncol. 2022, 33, S164. [Google Scholar] [CrossRef]
  194. Steuer, C.E.; Hayashi, H.; Su, W.-C.; Nishio, M.; Johnson, M.L.; Kim, D.-W.; Koczywas, M.; Felip, E.; Gold, K.A.; Murakami, H. Efficacy and safety of patritumab deruxtecan (HER3-DXd) in advanced/metastatic non-small cell lung cancer (NSCLC) without EGFR-activating mutations. J. Clin. Oncol. 2022, 40, 9017. [Google Scholar] [CrossRef]
  195. Majumder, A. Targeting Homocysteine and Hydrogen Sulfide Balance as Future Therapeutics in Cancer Treatment. Antioxidants 2023, 12, 1520. [Google Scholar] [CrossRef] [PubMed]
  196. Sedillo, J.C.; Cryns, V.L. Targeting the methionine addiction of cancer. Am. J. Cancer Res. 2022, 12, 2249–2276. [Google Scholar]
  197. Sun, X.; Gao, H.; Yang, Y.; He, M.; Wu, Y.; Song, Y.; Tong, Y.; Rao, Y. PROTACs: Great opportunities for academia and industry. Signal Transduct. Target. Ther. 2019, 4, 64. [Google Scholar] [CrossRef]
  198. Gaborit, N.; Abdul-Hai, A.; Mancini, M.; Lindzen, M.; Lavi, S.; Leitner, O.; Mounier, L.; Chentouf, M.; Dunoyer, S.; Ghosh, M.; et al. Examination of HER3 targeting in cancer using monoclonal antibodies. Proc. Natl. Acad. Sci. USA 2015, 112, 839–844. [Google Scholar] [CrossRef]
  199. Wymant, J.M.; Sayers, E.J.; Muir, D.; Jones, A.T. Strategic Trastuzumab Mediated Crosslinking Driving Concomitant HER2 and HER3 Endocytosis and Degradation in Breast Cancer. J. Cancer 2020, 11, 3288–3302. [Google Scholar] [CrossRef]
  200. Xie, T.; Lim, S.M.; Westover, K.D.; Dodge, M.E.; Ercan, D.; Ficarro, S.B.; Udayakumar, D.; Gurbani, D.; Tae, H.S.; Riddle, S.M.; et al. Pharmacological targeting of the pseudokinase Her3. Nat. Chem. Biol. 2014, 10, 1006–1012. [Google Scholar] [CrossRef]
  201. Singh, M.; George, A.K.; Homme, R.P.; Majumder, A.; Laha, A.; Sandhu, H.S.; Tyagi, S.C. Expression Analysis of the Circular RNA Molecules in the Human Retinal Cells Treated with Homocysteine. Curr. Eye Res. 2019, 44, 287–293. [Google Scholar] [CrossRef] [PubMed]
  202. Singh, M.; George, A.K.; Homme, R.P.; Majumder, A.; Laha, A.; Sandhu, H.S.; Tyagi, S.C. Circular RNAs profiling in the cystathionine-β-synthase mutant mouse reveals novel gene targets for hyperhomocysteinemia induced ocular disorders. Exp. Eye Res. 2018, 174, 80–92. [Google Scholar] [CrossRef]
  203. George, A.K.; Master, K.; Majumder, A.; Homme, R.P.; Laha, A.; Sandhu, H.S.; Tyagi, S.C.; Singh, M. Circular RNAs constitute an inherent gene regulatory axis in the mammalian eye and brain (1). Can. J. Physiol. Pharmacol. 2019, 97, 463–472. [Google Scholar] [CrossRef] [PubMed]
  204. Wu, Y.; Zhang, Y.; Wang, M.; Li, Q.; Qu, Z.; Shi, V.; Kraft, P.; Kim, S.; Gao, Y.; Pak, J.; et al. Downregulation of HER3 by a novel antisense oligonucleotide, EZN-3920, improves the antitumor activity of EGFR and HER2 tyrosine kinase inhibitors in animal models. Mol. Cancer Ther. 2013, 12, 427–437. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, Y.; Qu, Z.; Kim, S.; Shi, V.; Liao, B.; Kraft, P.; Bandaru, R.; Wu, Y.; Greenberger, L.M.; Horak, I.D. Down-modulation of cancer targets using locked nucleic acid (LNA)-based antisense oligonucleotides without transfection. Gene Ther. 2011, 18, 326–333. [Google Scholar] [CrossRef]
Figure 1. Cartoon diagram showing four receptor tyrosine kinases (RTKs) members and their corresponding ligands (HER2 does not have any ligand) on the left-hand side. On the right-hand side, it shows ligand-dependent heterodimerization of HER2 and HER3 and their downstream signaling cascade.
Figure 1. Cartoon diagram showing four receptor tyrosine kinases (RTKs) members and their corresponding ligands (HER2 does not have any ligand) on the left-hand side. On the right-hand side, it shows ligand-dependent heterodimerization of HER2 and HER3 and their downstream signaling cascade.
Cells 12 02517 g001
Figure 2. Cartoon diagram showing the extracellular domain, transmembrane domain, juxta membrane domain, kinase domain, and C-terminal tail of HER3.
Figure 2. Cartoon diagram showing the extracellular domain, transmembrane domain, juxta membrane domain, kinase domain, and C-terminal tail of HER3.
Cells 12 02517 g002
Table 4. Development of HER3-directed antibody–drug conjugates.
Table 4. Development of HER3-directed antibody–drug conjugates.
Antibody–Drug ConjugatesStudy PopulationClinical Trial
Phase
Adverse EventsStatus
Finding
Reference
U3-1402Advanced or metastatic CRCNCT04479436
phase II
Terminated
Naïve patients with HR + /HER2-early BCNCT04610528
phase I
Active, not recruiting
Metastatic or unresectable NSCLCNCT03260491
phase I
Nausea, vomiting, fatigue, decreased appetite, and alopeciaActive, not recruiting
U3-1402 has antitumor activity and a manageable safety profile
[186,187,188]
HER3 + metastatic BCNCT02980341
phase I + II
Nausea, vomiting, and decreased appetiteActive, not recruiting
In a preliminary analysis, U3-1402 demonstrated antitumor activity and a manageable safety profile
[189,190]
Metastatic or locally advanced EGFR-mutated NSCLCNCT04619004
phase II
Active, not recruiting
Locally advanced or metastatic EGFR-mutated NSCLCNCT04676477
phase I
Recruiting
Metastatic BCNCT04699630
phase II
Recruiting
Advanced BCNCT04965766
phase II
Recruiting
Metastatic or locally advanced EGFR-mutated NSCLC after failure of EGFR TKI therapyNCT05338970
phase III
Recruiting
NSCLC: non-small cell lung cancer, BC: breast cancer, HNSCC: head and neck squamous cell carcinoma; AEs: adverse events; PFS: progression-free survival; ESCC: esophageal squamous cell carcinoma; CRC: colorectal cancer; OS: overall survival; TKI: tyrosine kinase inhibitor.
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

Majumder, A. HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer. Cells 2023, 12, 2517. https://doi.org/10.3390/cells12212517

AMA Style

Majumder A. HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer. Cells. 2023; 12(21):2517. https://doi.org/10.3390/cells12212517

Chicago/Turabian Style

Majumder, Avisek. 2023. "HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer" Cells 12, no. 21: 2517. https://doi.org/10.3390/cells12212517

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