Skip to main content
Genes & Diseases logoLink to Genes & Diseases
. 2024 Jun 17;12(4):101354. doi: 10.1016/j.gendis.2024.101354

HER3: Unmasking a twist in the tale of a previously unsuccessful therapeutic pursuit targeting a key cancer survival pathway

Omkar Desai a,b,c,1, Moeez Rathore a,b,1, Christina S Boutros a,b,c, Michel'le Wright a,b, Elizabeth Bryson a,b, Kimberly Curry a,b, Rui Wang a,b,c,
PMCID: PMC12022662  PMID: 40290122

Abstract

HER3, formally referred to as ERB-B2 receptor tyrosine kinase 3, is a member of the ErbB receptor tyrosine kinases (also known as EGFR) family. HER3 plays a significant pro-cancer role in various types of cancer due to its overexpression and abnormal activation, which initiates downstream signaling pathways crucial in cancer cell survival and progression. As a result, numerous monoclonal antibodies have been developed to block HER3 activation and subsequent signaling pathways. While pre-clinical investigations have effectively showcased significant anti-cancer effects of HER3-targeted therapies, these therapies have had little impact on cancer patient outcomes in the clinic, except for patients with rare NRG1 fusion mutations. This review offers a comprehensive description of the oncogenic functions of HER3, encompassing its structure and mediating signaling pathways. More importantly, it provides an in-depth exploration of past and ongoing clinical trials investigating HER3-targeted therapies for distinct types of cancer and discusses the tumor microenvironment and other critical determinants that may contribute to the observed suboptimal outcomes in most clinical studies using HER3-targeted therapies. Lastly, we suggest alternative approaches and the exploration of novel strategies to potentially improve the efficacy of targeting the pivotal oncogenic HER3 signaling pathway in future translational investigations.

Keywords: Cancer, ErbB3, HER3, Neuregulin, Tumor microenvironment

Introduction

The ErbB family of receptors (also known as human epidermal growth factor receptor family) is one of the most extensively studied receptor tyrosine kinase (RTK) families. It consists of four distinctive receptors: i) epidermal growth factor receptor (EGFR, also known as HER1/ErbB1), ii) ERB-B2 receptor tyrosine kinase 2 (ErbB2, also known as HER2), iii) ERB-B2 receptor tyrosine kinase 3 (ErbB3, also known as HER3), and iv) ERB-B2 receptor tyrosine kinase 4 (ErbB4, also known as HER4).1 ErbB family receptors are structurally akin, being single-chain modular glycoproteins featuring an extracellular ligand-binding domain, a singular transmembrane domain, a juxta-membrane domain, and an intracellular tyrosine kinase domain followed by C-terminal regulatory regions (Fig. 1A).2 ErbB receptors are expressed in both mesenchymal and epithelial cells and are activated by receptor-specific soluble ligands, following which they undergo a conformational change, inducing homodimerization or heterodimerization (mainly at the juxta-membrane domain regions),3 leading to the activation of downstream signaling intracellularly. Importantly, HER2 lacks a receptor domain while displaying robust tyrosine kinase activity, and in contrast, HER3 encompasses a ligand-binding domain but the kinase domain has weak activity and is often considered as “kinase-dead”.4 Consequently, these two receptors often do not operate independently and demonstrate receptor cooperativity, wherein “kinase-dead” HER3 and HER2 often heterodimerize with each other or other ErbB receptors, and occasionally with other RTKs, to mediate downstream signaling.5 The binding of ligands to EGFR or HER4 induces dimerization either with itself to form homodimers, or with other ErbB receptors to form heterodimers (Fig. 1B).6 EGFR exhibits binding affinity towards a minimum of seven ligands, including epidermal growth factor (EGF), transforming growth factor-alpha (TGFα), heparin-binding EGF-like growth factor, amphiregulin, epiregulin, betacellulin, and epigen, whereas HER3 and HER4 are activated by neuregulin 1/2 (NRG1/2) and neuregulin 3/4 (NRG3/4), respectively.7,8 In contrast, HER2 lacks identified ligands and remains persistently in an open conformation with an exposed dimerization loop, necessitating a ligand-bound heterodimerization partner for signaling to occur.2

Figure 1.

Fig. 1

Schematic illustration of ErbB receptor structures and dimerization patterns. ErbB, ERB-B2 receptor tyrosine kinase.

The ErbB family downstream signaling pathways are strongly associated with cell proliferation, survival, and cancer development.1,9 Mutation or up-regulation of EGFR and HER2 are associated with the development of many types of malignancies and have been reviewed extensively by others.10 The crosstalk between the ErbB family of receptors which enhance and diversify downstream signal transduction is becoming a major survival mechanism of cancer cells, and contexts such as the tumor microenvironment are critical for the activation of HER3 and other ErbB family receptors. This review will focus on discussing the pro-cancer role of HER3. HER3 overexpression has been observed in various types of solid tumors, including breast cancer,11 cervical cancer,12 colorectal cancer (CRC),13 gastric cancer,14 melanoma,15 non-small cell lung cancer (NSCLC),16 ovarian cancer,17 and pancreatic cancer.18 In this review, we will discuss key aspects of HER3 signaling, including the structure of HER3 and the involved downstream signaling axis, HER3 gene mutations, and more importantly, focus on reviewing pro-cancer roles of HER3 in various cancer types and associated pre-clinical and clinical studies. We will also discuss recent developments in HER3-directed therapies and the new roles of HER3 in tumor microenvironment-mediated signaling pathways. Lastly, we will provide our take on the future directions of developing novel HER3-targeted therapies.

Structure of HER3

HER3 was first discovered in 1989 by Kraus et al, with the human encoding gene ERBB3 being identified on chromosome 12q13.19 As mentioned earlier, HER3 consists of a large extracellular ligand-binding ectodomain, a transmembrane domain, and a short juxta membrane segment, followed by an intracellular tyrosine kinase domain and a tyrosine-containing carboxy-terminal tail (Fig. 2A).1,20 The ectodomain is further divided into four sub-domains (I–IV), comprising two cysteine-rich regions (II and IV) and two flanking domains (I and III).21 In the absence of a ligand, HER3 remains inactive due to the binding between subdomains II and IV.22 Upon ligand binding at the binding pocket in domains I and IV,21,22 the inactive conformation is altered to expose these domains for subsequent activation.21 Distinct from other RTKs, HER3 is considered a “kinase-dead” receptor with 1000-fold weaker kinase activity than fully activated EGFR.1,23,24 As a result, HER3 predominantly heterodimerizes with EGFR and HER2, and to a lesser extent with HER4,1 which have significant kinase activity upon activation. However, it is worth noting that Wang and colleagues have reported that K742 in HER3 has weak but detectable pseudokinase activity and leads to auto-phosphorylation activation without other receptors.23,25,26

Figure 2.

Fig. 2

Schematics of HER3 structure and downstream signaling. (A) Full-length HER3 (amino acid residues 1–1342) with four receptor domains (I–IV, yellow), a transmembrane and a short juxta membrane domain (TM and JM, respectively, gray), and a pseudo kinase domain (Pseudo-K, green). Y104 and V333 are for ligand binding (red). Numbers indicate amino acid positions. (B) Upon ligand binding with NRG, HER3 dimerizes with either EGFR or HER2, activating downstream MAPK and PI3K signaling pathways. HER2/3, human epidermal growth factor receptor 2/3; NRG, neuregulin; EGFR, epidermal growth factor receptor; MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase.

HER3 signaling pathways

Currently, three HER3 ligands have been identified: neuregulin 1/1b (NRG1/1b, also known as heregulins) and neuregulin 2 (NRG2).27 NRG-activated HER3 leads to activations of two downstream signaling pathways: the mitogen-activated protein kinase (MAPK) and the phosphatidylinositol 3-kinase (PI3K) pathways (Fig. 2B).28 Signaling through the MAPK pathway begins with the activation of RAS by an RTK (in this case HER3), leading to the phosphorylation and activation of RAF, MEK, and MAPK in turn. MAPK regulates the activities of transcription factors including AP-1 (activator protein 1), NF-κB (nuclear factor-kappa B), and p53, all of which play critical roles in regulating cellular functions including proliferation, differentiation, apoptosis, and stress response.29 Constitutively activating mutations in RAS genes are commonly found in cancer. These mutations lead to the activation of the MAPK cascade signaling independently of HER3 and other RTKs, promoting cancer development and progression.30 However, a recent study reported that HER3 inhibition attenuates cell and tumor growth in both RAS wild-type and mutant CRC,31 suggesting HER3-targeted therapies may be used for treating patients with RAS-mutant CRC who could not receive EGFR-targeted therapies.

For activating the PI3K pathway, the C-terminal intracellular domain of HER3 has six consensus phosphor-tyrosine sites that bind to the regulatory p85 subunit of PI3K.32 This interaction leads to the phosphorylation of membrane phosphoinositides, which leads to the recruitment and activation of Akt (protein kinase B) and PDK1 (pyruvate dehydrogenase kinase 1).19 Akt regulates a variety of protein functions that are involved in cell survival, apoptosis, cell cycle progression, and cell migration, all of which contribute to cancer development and progression.33 Together, HER3 is a key upstream effector of MAPK and PI3K signaling cascades that are involved in cancer cell growth and survival. Therefore, many HER3-targeted therapies aim to inhibit receptor activation to attenuate these pro-cancer signaling cascades to block cancer cell growth and survival.19

Mutations

Prior studies by Jaiswal et al found that HER3 mutations were found in approximately 12% of gastric, 11% of colon, 4% of breast, 1% of NSCLC, and other types of cancer, primarily occurring in the extracellular domain34 (summarized in Table 1). For example, HER3 mutations such as V104M, P262H, or G284R, promoted pro-tumor phenotypes including migration and anchorage-independent growth in colonic or breast non-neoplastic epithelial cell lines.34, 35, 36 Additional studies showed that these mutations activated downstream signaling pathways, particularly Akt, which led to cell growth and survival.37,38 Jaiswal and team also investigated the oncogenic properties of these mutations by screening cohorts with different tumor types for synonymous mutations of HER3.34 Many of the mutations were in regions that were conserved among HER3 orthologs, suggesting functional relevance. Moreover, HER3 mutations were also found to be ligand-independent and promote tumor growth. The T355 to isoleucine (T355I) mutation was particularly noteworthy, as it was associated with increased levels of HER3 and ERK1/2 phosphorylation and downstream cyclin D1 activation in estrogen receptor-positive and HER2-overexpressing breast cancer.28,39,40 Other mutations such as tyrosine kinase domain (TKD) mutations were also found to have high oncogenicity.34 Collier et al found that the E909G TKD mutation increased HER2/HER3 heterodimer kinase activity in vitro with purified proteins. On the other hand, no frameshift, nonsense, or intronic HER3 mutations were reported in cancer-related studies.40

Table 1.

HER3 and NRG1 gene alterations in distinct types of cancer.

Cancer type HER3 amplification HER3 mutations NRG1 fusions Reference
Breast Yes E928G, V104L, G284R, T355I, T389K, M91I, D297Y/A/H/N/V, S846I, E1261A ADAM9, COX10-AS, AKAP13 34,40,41, 42, 43
Non-small cell lung cancer Yes E928G, A232V, V714M, E332K/Q, D297Y/A/H/N/V, S846I CD74, SDC4, SLC3A2, TNC, MDK, DIP2B, RBPMS, MRPL13, ROCK1, DPYSL2, PARP8 34,40,41,43, 44, 45, 46
Colorectal cancer Yes V104L/M, P262H/S, G284R, A232V/T, G325R, D297Y, S846I, E928G/K/Q, M60K, V295A, T355A/I, M60R, R103G/H, N126K, R475W POMK 34,40,41,43,45
Pancreatic cancer Unknown Unknown ATP1B1, APP, CDH1, VTCN1 43,45
Gynecologic cancer (ovarian) Yes L536L, V104M, V438I, D1149E SETD4, TSHZ2, ZYMYM2, RAB3IL1 43,45,47
Head and neck cancer Unknown M91I THBS1, PDE7A 45,48
Prostate cancer Unknown Unknown STMN2 49
Gastric cancer Yes A232V, E928G, P262H/S, T389K, V104L, 1218delE, A172P, D297Y, V654G, K926R, L930W, Q809R, H228R, G284R, D297Y/A/H/N/V, T355I/P, R426W, S846I Unknown 34,40,50, 51, 52

Note: HER3, human epidermal growth factor receptor 3; NRG1, neuregulin 1.

Role of HER3 in distinct cancer types and related clinical studies

Breast cancer

Breast cancer affects one in eight women across the United States and is associated with high mortality, metastasis, and relapse. It often exhibits overexpression of estrogen, progesterone, or HER2 receptors. Among these, triple-negative breast cancer stands out as the most aggressive phenotype, characterized by the absence of receptor expression.53 Breast cancer is a heterogeneous disease, with distinct receptor expression profiles in different patients.54 The overexpression of ErbB family receptors, specifically HER3, is a strong indicator of worse patient overall clinical outcomes, including metastasis, tumor growth, and recurrence.55, 56, 57 HER3 activation promotes cell growth and survival of HER2-positive tumors and usually leads to resistance to HER2-targeted therapy in patients with breast cancer.55,58, 59, 60 High expression of HER3 has been shown to cause resistance to tamoxifen and trastuzumab therapies in HER2 overexpressing breast cancer cells.61 To tackle this issue, therapeutic approaches targeting HER3 have been created. These approaches involve the use of monoclonal antibodies (mAbs) to inhibit the HER3 receptor, leading to its internalization and subsequent degradation.28,61 Numerous clinical trials have been undertaken to assess the effectiveness of HER3-targeted therapies in the management of patients with breast cancer, and these investigations are discussed herein.

Trastuzumab is currently the standard of care for treating patients with HER2-positive breast cancer. However, as discussed earlier, HER3 overexpression and ligand-dependent HER2-HER3 interactions can lead to treatment resistance and evasion of drug-induced apoptosis.61 To address this, a humanized HER3-specific mAb, patritumab (U3-1287), was developed, which inhibits ligand binding and triggers receptor internalization for degradation.62 Patritumab blocked cancer cell growth in vitro and re-sensitized tumors to trastuzumab therapy in HER2-positive breast cancer in pre-clinical models.61,63 However, findings from a phase Ib/II clinical trial evaluating the combination of patritumab with trastuzumab and paclitaxel in patients with HER2-positive metastatic breast cancer showed no improvement compared with standard-of-care therapy (NCT01512199). Alternatively, an antibody drug conjugate (ADC) version of patritumab with a cytotoxic payload, U3-1402 (also known as HER3-Dxd), is being assessed in combination with DS-1062 (anti-TROP2 mAb) for treating patients with HER3-positive metastatic breast cancer (NCT04699630). Additional discoveries in the anti-cancer effects of this HER3-targeted ADC will be elaborated upon later in this review.

Meanwhile, multiple other HER3-targeting mAbs were developed and assessed in clinical studies for treating patients with breast cancer. Seribantumab (previously known as MM-121) was developed to block the ligand neuregulin and HER3 binding (NRG-HER3) and was developed as a full-length IgG2 mAb to avoid toxicities in non-cancerous tissues. It exhibited anti-proliferative effects in vitro and in vivo in cells expressing HER2 and HER3.64,65 However, a phase II clinical trial (NCT01151046) used the combination of seribantumab with exemestane (endocrine therapy) in patients with HER2-negative metastatic breast cancer showed negligible effect on progression-free survival (PFS). Interestingly, a subset of patients in the trial with high levels of NRGs in tumor tissues had increased PFS, suggesting HER3-targeted therapies can be effective when the targeted pathway is highly active.65 The effects of seribantumab and other specific antibodies in patients with high levels of NRG-induced HER3 activation will be further discussed in subsequent sections. Elgemtumab (LJM716) is another HER3-specific mAb which blocks phosphorylation of HER3/Akt. Investigated in patients with HER2-positive metastatic breast cancer, its efficacy was limited when co-administered with BYL719, a PI3K inhibitor (NCT02167854). However, favorable outcomes were observed when combined with trastuzumab.66

In contrast to the aforementioned mAbs, bispecific antibodies simultaneously targeting HER3 and a different ErbB receptor have also been developed. MM-111 is a novel HER2/HER3 bispecific antibody that has demonstrated a good safety profile in a phase I study but had little additional benefit when combined with standard-of-care chemotherapy in treating patients with advanced HER2-positive breast cancer (NCT01304784).67,68 Zenocutuzumab (MCLA-128) is another HER2/HER3 bispecific antibody that has been used in combination with trastuzumab in patients with HER2-positive breast cancer and in combination with endocrine treatment in patients with ER-positive/low HER2-expressing breast cancer (NCT03321981). Although these treatments were well tolerated, their effects on outcomes of patients with breast cancer remain limited or unclear. Sapitinib (AZD8931), an EGFR/HER2/HER3 inhibitor, was employed in conjunction with endocrine therapy to address endocrine resistance in patients with hormone-sensitive advanced breast cancer.69 Nonetheless, it did not augment endocrine responsiveness and was associated with greater toxicity. It is worth mentioning that other than the recent trial using HER3-ADC (NCT04699630), most prior clinical studies in breast and other types of cancer did not evaluate HER3 expression before recruiting participants, potentially leading to unfavorable effects on the study outcomes. This limitation observed in most clinical studies concerning HER3-targeted therapy will be further discussed in this review article.

Non-small cell lung cancer

Lung cancer is the leading cause of cancer-related death worldwide, with NSCLC accounting for 82% of all cases.70 Although HER3 expression has only been found in 30%–40% of NSCLC tumors,44,71,72 it is considered a key mediator of the development of resistance to EGFR-targeted therapies with tyrosine kinase inhibitors (TKIs), partially via activation of the PI3K/Akt pathway.44,72,73 Consequently, HER3-targeted mAbs, such as patritumab, seribantumab, and lumretuzumab have been assessed for treating patients with NSCLC.

Patritumab blocked lung cancer cell growth in vitro and in vivo, and in a phase II clinical trial, combining the HER3 antibody patritumab with a pan-HER inhibitor erlotinib led to increased PFS in patients with advanced NSCLC with high NRG expression (NCT01211483).74, 75, 76 In a separate phase I study, the use of patritumab and erlotinib combination in patients with NSCLC also led to stable disease as the best response without dose-limiting toxicities.77 However, as this line of investigation continued in a phase III clinical trial (NCT02134015), the study was prematurely terminated as it failed to reach the pre-defined criteria. Similar to patritumab, seribantumab has exhibited robust anti-cancer efficacy in pre-clinical studies of NSCLC.78 Furthermore, the recent CRESTONE trial (NCT04383210) showcased an objective response rate (ORR) of 36% in patients with NSCLC. However, a phase II study evaluating seribantumab in combination with docetaxel was terminated due to the lack of significant improvement in PFS (NCT02387216). In contrast, another phase II trial revealed a potential benefit when employing seribantumab in conjunction with erlotinib. This observation was based on a prespecified retrospective subgroup analysis of patients whose tumors exhibited high levels of NRG expression.78 This underscores that elevated levels of NRG and the consequent activation of HER3 represent a potential predictive biomarker for the responsiveness of patients to HER3-targeted therapy.

In NSCLC, another HER3-targeted mAb, lumretuzumab (RG7116), has also been used and demonstrated significant antibody-dependent cell-mediated cytotoxicity.79 However, the combination of lumretuzumab with carboplatin and paclitaxel as first-line treatment of patients with NSCLC showed limited effect and most patients discontinued the treatment due to disease progression (NCT02204345).79 Similarly, combining lumretuzumab with cetuximab (EGFR-specific antibody) had little impact on patients with HER3-expressing NSCLC (NCT01482377), although the combination blocked NSCLC cell growth in pre-clinical studies.80 Similar to breast cancer, although HER3-specific antibodies significantly blocked NSCLC cell growth and survival in pre-clinical studies, findings from clinical trials suggest HER3-targeted therapies have limited efficacy in patients, except for patients with NRG-high and HER3-activated phenotypes. While most HER3-targeted clinical trials have been unsuccessful in NSCLC, it is crucial to acknowledge that, like studies for breast cancer, HER3 expression levels in NSCLC tumors were not examined and used for patient recruitment, which is a major pitfall in the study design given that only approximately 30%–40% of patients with NSCLC exhibit HER3 expression.

Colorectal cancer

CRC is the second most common cause of cancer-related death in the United States.81 HER3 overexpression has been observed in a significant subset of patients with CRC and has been associated with poor prognosis.82, 83, 84, 85, 86 A preclinical study analyzed patient samples and found that 80% of primary CRC tumors and 82% of CRC liver metastases had high levels of HER3 proteins, determined by immunohistochemistry.85 In another study, immunohistochemistry analysis showed that HER3 expression was detected in 93% of metastatic CRC tumors.87 These findings suggest that HER3 may be a potential therapeutic target in treating patients with CRC, particularly those with advanced disease. Our laboratory's pre-clinical investigations have shown that the inhibition of HER3 significantly attenuates the survival of CRC cells.31,88 Several HER3-targeted mAbs, including seribantumab, REGN1400, Av-203, and KTN3379 have been tested either alone or in combination with EGFR-targeted therapies in phase I and phase II trials in patients with CRC, but the outcomes of these trials have shown severe toxicities.55,89 Meanwhile, in a phase I clinical trial, the use of patritumab resulted in stable disease as the best response with no dose-limiting toxicities.90 Duligotuzumab, a dual-action antibody targeting HER3/EGFR, was employed in a phase II trial (NCT01652482). However, this trial yielded no PFS or overall survival (OS) advantages.91 Duligotuzumab has also been tested in combination with cobimetinib (MEK inhibitor) in patients with locally advanced or metastatic CRC-harboring mutant KRAS (NCT01986166). Nevertheless, this combination was associated with increased toxicity and limited efficacy in comparison to cobimetinib or duligotuzumab monotherapy. A phase I/II study examined the toxicity and efficacy of the pan-HER inhibitor sapitinib combined with standard-of-care chemotherapy in patients with metastatic CRC (NCT01862003). Although sapitinib showed a low toxicity profile, no improvement was seen in the ORR.92

Pancreatic cancer

Pancreatic ductal adenocarcinoma (PDAC) presently ranks as the third highest cause of cancer-related death in the United States, and its incidence is projected to increase in the next decade.93 Most patients with PDAC present with metastasis at the time of diagnosis, and almost all patients eventually develop distant metastasis.2, 3, 4 Unfortunately, treatment options for patients with metastatic PDAC are limited, and the 5-year survival is only 3%.4 Recent research suggests that NRG secreted by liver endothelial cells can activate the HER3-Akt signaling pathway in metastatic PDAC cells that express HER3. This suggests that HER3-targeted therapies may be effective in treating patients with HER3-positive metastatic PDAC.5 Pre-clinical studies have demonstrated that HER3-targeted therapies are effective in attenuating tumor growth in PDAC.6 Based on these findings, numerous clinical trials have been initiated to further evaluate the efficacy of HER3-targeted therapies in PDAC. As mentioned earlier, the CRESTONE phase II clinical trial is currently evaluating the safety and efficacy of seribantumab in patients with advanced solid tumors that have NRG1 gene fusion mutations (NCT04383210). Preliminary data from the trial suggests that seribantumab has a favorable safety profile and has demonstrated a durable response in patients with NRG1 gene fusion, including a 33% ORR in patients with PDAC. Another ongoing phase II clinical trial is investigating the efficacy of zenocutuzumab (MCLA-128) in patients with solid tumors harboring an NRG1 gene fusion (eNRGy) (NCT02912949). Among the patients with pancreatic cancer enrolled in the trial, an objective response was observed in 39% of patients.94 The data derived from these clinical trials substantiates the efficacy of HER3-targeted therapies in treating patients with PDAC, particularly those exhibiting HER3 expression or NRG1 gene fusion. More information regarding NRG1 gene fusion in HER3-targeted therapies will be discussed later in this review.

Gynecologic cancer

Gynecologic cancers are a significant health concern for women worldwide, with ovarian cancer ranking as the fifth leading cause of cancer-related death in women, with a 5-year relative survival rate of only 49%.95 In pre-clinical investigations, overexpression of HER3 is commonly observed in gynecologic cancers.96,97 Up to 68% of ovarian tumors have been detected with HER3 overexpression (either at protein or mRNA levels), which is correlated with an unfavorable patient prognosis and an increased prevalence of metastasis.17,47,96,97 Similar to other types of cancer, elevated HER3 expression is linked to the development of acquired resistance to standard-of-care treatment in ovarian cancer.28,98,99 Additional investigations examined the correlation between HER3 expression and patient survival in cervical cancer and found that HER3 expression was significantly associated with reduced disease-free survival (DFS) and OS.100 For HER3-targeted therapies, seribantumab has been tested in gynecologic cancers in combination with EGFR-targeted mAb, as well as other chemotherapeutic agents such as gemcitabine, carboplatin, and pemetrexed (NCT01447225, NCT02538627). These studies have largely shown a lack of success.55,101 Interestingly, within a phase II clinical trial, the addition of seribantumab alongside paclitaxel demonstrated an elevated median PFS in patients diagnosed with platinum-resistant/refractory ovarian cancer exhibiting high NRG levels (NCT01447706).102 These results again imply that the assessment of NRG/HER3 expression could serve as a crucial biomarker for identifying individuals with gynecologic cancers who may derive benefits from HER3-targeted therapies.

Head and neck cancers

Head and neck cancers account for 4% of all cancers in the United States and have caused an estimated 277,597 deaths worldwide in 2020.103 A few but significant studies in head and neck squamous cell carcinoma (HNSCC) indicate that positive HER3 expression is linked to worse outcomes.104,105 Meanwhile, pre-clinical studies using multiple HER3-targeted mAbs (LMJ716, RG7116, REGN1400, and seribantumab) have shown potent inhibition of cancer cell and tumor growth, but a clinical trial using LMJ716 failed to provide conclusive information (NCT01598077).44,106 Similarly, the EGFR/HER3 dual antibody, duligotuzumab, also showed limited efficacy in a phase IB clinical trial when used in combination with cisplatin/5-fluorouracil or carboplatin/paclitaxel for recurrent or metastatic HNSCC.107 Likewise, in another phase IB clinical trial of patients with HNSCC, the combination of patritumab with cetuximab plus a platinum-containing agent demonstrated a tumor response rate of 47%, but 40% of patients discontinued treatment due to disease progression or adverse events.108

Prostate cancer

Prostate cancer is the most diagnosed cancer and the second leading cause of cancer-related death in men.70 In a study that examined the tumor samples of castration-resistant prostate cancer, it was found that HER2 and HER3 were highly expressed.49 Additionally, in vivo studies have shown that activation of the NRG-HER3 signaling has been linked to anti-androgen therapy resistance in prostate cancer.109 Although blocking EGFR and HER2, which are essential for HER3 activation, by afatinib did not improve outcomes of patients with prostate cancer in the clinic,110 a recent pre-clinical study using prostate cell lines showed that direct inhibition of HER3 by patritumab (antibodies) or U3-1402 (patritumab ADC) exhibited anti-tumor effects in vitro, suggesting HER3 inhibition could impede tumor growth in patients with prostate cancer.49 As such, further evaluation of HER3-targeting in patients with prostate cancer is warranted in prospective clinical trials.

NRG1 gene fusion mutations

The identification of oncogenic gene fusions or rearrangements has been observed in various solid tumors. This has led to the successful identification and targeting of oncogenic gene fusions as one of the most significant advancements in oncology.111,112 NRG1 fusion mutations were first identified in NSCLC.113 They are caused by DNA rearrangements, resulting in the creation of a “chimera protein” by the fusion of NRG1 domains with partner proteins (such as ATP1B1 (ATPase Na+/K+ transporting subunit beta 1), APP (amyloid beta precursor protein), CD74 (leukocyte differentiation antigen 74), and SDC4 (syndecan 4); summarized in Table 1).43,114 As a result, NRG1 fusion proteins are highly expressed on the cell membrane and induce HER3 activation. Although the occurrence of NRG1 rearrangements is rare, with the incidence of 0.5% in pancreatic cancer, 0.3% in NSCLC, 0.2% in breast cancer, and 0.1% in colorectal cancer,43 multiple studies in distinct cancer types discussed above have determined that the presence of NRG1 fusion mutations is associated with enhanced anti-cancer effects caused by HER3-targeted therapies.

In addition, the eNRGy1 global multicenter registry for NRG1 gene fusions has shown an objective response rate of 25% in lung cancer patients treated by afatinib (clinically available EGFR/HER2 inhibitors),115 and a pre-clinical study demonstrated that NRG1 gene fusion and rearranged cancer cell lines exhibited heightened sensitivity to tarloxotinib, a prodrug of a potent irreversible inhibitor of EGFR/HER2.116 In recent clinical studies, a phase II clinical trial evaluating the efficacy of zenocutuzumab (MCLA-128) in patients with NRG1 gene fusion cancer showed an ORR of 34% and a median duration of response of 9 months.94 In a study analyzing patients with advanced PDAC, those with NRG1 gene fusion-positive tumors that were treated with afatinib demonstrated a significant and rapid response while on therapy.117

As a result, a phase II clinical trial was initiated to specifically assess the safety and efficacy of seribantumab in patients with advanced solid tumors harboring NRG1 gene fusion mutations (NCT04383210, CRESTONE). Preliminary data from this clinical trial indicates that seribantumab has a favorable safety profile with few dose-limiting toxicities and has produced a durable response in patients with NRG1 gene fusion, with an ORR of 36% in NSCLC and 33% in PDAC.118 Also, another phase II clinical trial is currently underway to study the efficacy of afatinib in advanced NRG1-rearranged malignancies (NCT04410653). These findings emphasize the significance of NRG1 fusions as a predictive biomarker, suggesting their utility in identifying patients likely to benefit from HER3-targeted therapy. Together, these observations collectively suggest HER3-targeted therapies can be effective when precisely directed toward the specific population such as with NRG1 gene fusion mutations, or HER3 expression as discussed above for lung cancer.

Antibody–drug conjugates

ADCs provide targeted delivery of cytotoxic drugs (referred to as “payload”) to tumor cells via antibody carriers, ensuring precision in drug delivery. Over the last decade, more than ten ADCs have gained approval, with several others in clinical development, reshaping the landscape of cancer therapy.119 A notable example is U3-1402, a pioneering HER3-targeted ADC utilizing patritumab as the carrier and DXd, a derivative of exatecan (topoisomerase I inhibitor), as the payload. Early reports from a phase I/II clinical trial revealed promising anti-tumor efficacy in patients with HER3-expressing metastatic breast cancer, boasting a 47% ORR and a disease control rate of 94% (NCT02980341).120 In a separate clinical study, the utilization of HER3-DXd in patients with EGFR TKI-resistant NSCLC exhibiting HER3 expression demonstrated clinical efficacy, showcasing a 39% ORR and a median PFS of 8 months.121,122 Recent findings also indicate that HER3-DXd displayed in vitro anti-tumor activity against HER3-positive cells, regardless of clinically relevant HER3 mutations.123 Furthermore, in a pre-clinical study using xenograft tumors, co-administration of U3-1402 with a PD-L1/PD-1 antibody in HER3-positive colorectal and lung cancer cell lines exhibited significantly enhanced anti-tumor immunity by sensitizing tumors to PD-1 blockade.124 In those studies, the authors speculated that the de novo antigens caused by the cytotoxic payload, Dxd, sensitized cancer cells to immunotherapies which had limited clinical impact as a monotherapy in solid tumors.124 Additionally, clinical analyses revealed a high frequency of tumor-specific HER3 expression among patients with solid tumors resistant to PD-1 blockade.124 These promising results suggest that exploring the combination of HER3-targeted ADC with immune checkpoint blockade could yield beneficial effects, warranting further investigation in future clinical trials. Moreover, in pre-clinical models of multidrug-resistant triple-mutant lung cancer and double-mutant CRC, a novel self-immolation T moiety and exatecan ADC effectively overcame chemoresistance and showed improved intra-tumoral pharmacodynamic response.125 These findings highlight the potential of ADCs to broaden the spectrum of responsive patient populations and applicable tumor types.

Tumor microenvironment and HER3 signaling

In recent years, extensive research has been conducted to characterize the effects of the tumor microenvironment on cancer cells. The tumor microenvironment consists of cellular components such as the tumor cells and non-neoplastic stromal cells including immune cells, fibroblasts, endothelial cells, and a non-cellular component of the extracellular matrix.126 In addition to oncogenic alterations in cancer cells, crosstalk between cancer and stromal cells in the tumor microenvironment has also been identified as a key factor in activating HER3 for the development and progression of cancer (Fig. 3). For example, fibroblasts express and secrete NRGs to activate the HER2-HER3 pathway in breast cancer.127 Moreover, our recent studies suggest that HER3 is a key mediator of the pro-survival effects of the surrounding microenvironment. With a focus on liver metastases, which occur in over 80% of advanced CRC and PDAC cases, we collectively discovered that the surrounding liver microenvironment activates cancer-associated HER3, promoting cell growth and chemoresistance in CRC and PDAC cells.88,128, 129, 130, 131 Specifically, we determined that the liver-specific endothelial cell-rich microenvironment secretes soluble factors to activate HER3 in a paracrine fashion, and we found that blocking HER3 activity effectively blocked liver endothelial cell-induced CRC tumor growth. Interestingly, we found that although liver endothelial cells secrete NRGs, the canonical HER3 ligands are not the primary mediator of endothelial cell-induced HER3 activation.88 Based on these findings it is also noteworthy that the efficacy of HER3-targeted therapy relies on the HER3 expression in cancer cells, suggesting future clinical trials should employ HER3 expression as a key predictive marker for HER3-targeted therapy efficacy.

Figure 3.

Fig. 3

Schematic illustration of cancer-associated HER3 activated by the microenvironment components. HER3, human epidermal growth factor receptor 3.

Aside from endothelial cells and cancer-associated fibroblasts,128,132 HER3 also plays a role in immune cell biology and functions. Breast cancer cell-secreted NRGs activate macrophage-associated HER3 which in turn promotes the expression of JAG1 (jagged 1) and increases cancer cell invasion.133 Similarly, HER3 signaling regulates MHC I (major histocompatibility complex class I)-related chains A and B (MICA and MICB) in breast cancer cells and leads to natural killer-mediated toxicity in breast cancer.134 Moreover, studies in cervical cancer suggest that HER3 methylation in cancer cells is associated with the presence of macrophages, effector memory CD8 T-cells, activated CD8 T-cells, regulatory T cells, immature B cells, T helper 1 cells, and myeloid-derived suppressor cells but the mechanism remains unclear.135 Meanwhile, as HER3 expression is commonly detected in cancer, it has been assessed as a potential antigen for engineered CD4 T targeting in breast cancer and head and neck cancer,136,137 and the HER3 vaccine is being assessed as a potential strategy for cancer treatment/prevention either alone or in combination with immunotherapies.138 Together, these studies suggest HER3 mediates immune cell functions in various types of cancer, although much work is needed to further elucidate the involved mechanisms.

Discussion

The role of HER3 overexpression and oncogenic mutations in various solid tumor types is well documented. Both pre-clinical and clinical studies have shown that overexpression of HER3 is associated with tumor cell proliferation, chemoresistance, and overall worse survival. Most pre-clinical investigations employed either subcutaneous xenografts or primary orthotopic models and demonstrated the anti-cancer effects of HER3 inhibition principally on primary tumors, as described above for each cancer type. On the other hand, clinical trials investigating HER3-targeted therapies were mostly conducted in patients with advanced metastatic tumors that failed to respond to prior standard-of-care therapies. For instance, in breast cancer, the HER3-specific antibody patritumab demonstrated significant anti-cancer effects on xenografts and primary tumors in preclinical studies and in patients with metastatic breast cancer in clinical trials.61,139 However, there is no information on the effects of the antibodies on primary tumors in those patients or in patients only with primary tumors. Meanwhile, based on the preclinical studies from our laboratory that demonstrated that the liver microenvironment secretes soluble factors to activate cancer-associated HER3, we hypothesize that HER3 is highly active in liver metastases compared with primary tumors, suggesting that therapeutic interventions may yield greater efficacy in patients with liver metastases.88,128

In contrast, most clinical studies assessing HER3 inhibitors and antibodies, mostly in patients with metastatic tumors, showed limited impact in improving patient survival. One potential key reason for the failed outcomes is that, in most trials, HER3 expression in patients was not assessed and used as a selection criterion for patient recruitment. Our laboratory has shown that HER3-targeted therapies are only effective in cells and tumors that express HER3,128 implying that previous clinical trials failed potentially because a substantial number of enrolled patients had HER3-negative tumors, especially the patients with lung cancer wherein only 30%–40% tumors are HER3-positive.44,71,72 Thus, the results of those “negative clinical trial results” are debatable and call for further investigation. The significance of incorporating relevant biomarkers into HER3-targeted therapies is further supported by the consistent finding across various studies that individuals with NRG1 gene fusion mutations exhibit notable positive responses to HER3-targeted treatments.94,115, 116, 117 While significant studies used cancer cell lines and retrospectively analyzed archived patient tumor tissues and detected HER3 overexpression at mRNA levels (either by quantitative PCR or RNA sequencing), most clinical trials overlooked the assessment of HER3 expression. Among those few clinical studies in metastatic breast cancer, NSCLC, and HNSCC, HER3 expression was assessed by immunohistochemistry staining using biopsy tissues, followed by established histological scoring of staining intensity and distribution across tumor samples (e.g., 0 for no staining or membrane staining in ≤10%, 1+ for faint or barely perceptible incomplete membrane staining in >10%, 2+ for weak-to-moderate complete membrane staining in >10%, and 3+ for circumferential membrane staining that is complete, intense, and in >10%).140, 141, 142, 143 Despite that, determining HER3 expression in clinical studies remains a challenge partially due to variations in antibody specificity for immunohistochemistry staining and potential spatial heterogeneity in HER3 cell surface localization. Hence, establishing HER3 expression as a predictive biomarker for patient response to HER3-targeted therapies and developing reliable and rapid methods for assessing HER3 expression in cancer will have significant implications in future clinical trials for treating patients with solid tumors.

Within the scientific community, a prevailing belief is emerging that resistance to established HER3-targeted therapies could stem from alternative mechanisms of HER3 activation. In our laboratory, we identified that soluble factors released by the tumor microenvironment could activate HER3 independently of NRG1, and the subsequent HER3 activation does not rely on the conventional heterodimerization of HER3 with other ErbB receptors such as EGFR and HER2.88,128 Recent preliminary unpublished data from our laboratory indicates that leucine-rich α-2-glycoprotein 1 (LRG1), secreted by the EC microenvironment, can directly bind to and activate HER3 as a novel ligand, distinct from NRGs.144 While the precise mechanism of LRG1-HER3 binding remains unknown, our findings suggest alternative activation mechanisms of HER3, which may contribute to the development of resistance to HER3 antibodies, and warrant further investigations for developing strategies to block HER3 activation.

Lastly, significant findings from pre-clinical and clinical investigations indicate that cancer cells may counteract ErbB receptor inhibitions by up-regulating other RTKs (non-targeted ErbB receptors or others) as adaptive resistance mechanisms. For example, lung cancer develops resistance to EGFR inhibitors by over-activation of HER3.145 As a result, combining EGFR and HER3 inhibitions had superior anti-cancer effects compared with EGFR inhibitors alone. Similarly, in breast cancer, developed resistance to trastuzumab is associated with increased HER3 expression.146 According to this paradigm, when exposed to HER3-targeted antibodies and inhibitors, cancer cells may activate alternative survival pathways to sustain proliferation in a HER3-independent manner. Concurrent inhibition of multiple ErbB family receptors has been attempted through the administration of sapitinib, a pan-HER inhibitor, in clinical trials for patients with breast cancer and CRC, but the interventions did not yield improvements in patient outcomes.69,92 This implies that sapitinib may not be effective, necessitating the development of a novel drug with comparable action, or unidentified resistance mechanisms may contribute to this outcome. Notably, in contrast to previous unsuccessful clinical trials, the assessment of HER3 expression/activation emerges as a pivotal predictive biomarker for identifying patients who could derive benefits from combination therapies in future clinical trials.

Conclusion

As outlined in this review, HER3 plays a critical role in the oncogenesis of various cancer types. The tumor microenvironment significantly influences the activation of HER3 and the promotion of cancer cell survival. While HER3 as a therapeutic target holds promise, the way it has been addressed in prior clinical trials was suboptimal. The lack of a predictive biomarker for patient selection in these trials could be a contributing factor. Additionally, alternative mechanisms of HER3 activation, such as by a new ligand, LRG1, and the potential involvement of other acquired resistance mechanisms, could also contribute to this phenomenon. Therefore, identifying HER3 expression as a predictive biomarker, developing reliable and feasible assays to evaluate HER3 expression, and elucidating alternative HER3 activation mechanisms will improve the effectiveness of current HER3-targeted therapies. Furthermore, improving existing therapies by developing novel ADCs and combining HER3-targeted therapies with other standard-of-care regimens may enhance patient outcomes in future clinical trials. In summary, HER3 stands out as a pivotal target in cutting-edge cancer therapies, offering the potential for enhanced outcomes through a deeper comprehension of the tumor microenvironment and the molecular mechanisms underlying cancer progression.

CRediT authorship contribution statement

Conceptualization, O.D., M.R., C.B., and R.W.; data curation, O.D., M.R., C.B., E. B., and R.W.; writing – original draft and revising, O.D., M.R., C.B., M. W.M., C. K., and E. B.; R.W.; visualization, E. B. and R. W.; supervision, R.W.; funding acquisition, R.W.; review of the manuscript, all authors.

Funding

This work was supported by the National Institutes of Health (No. R00CA225756, R37CA278982 to R.W.) and the U.S. Department of Defense (No. HT9425-23-1-0657 to R.W.).

Conflict of interests

R.W. is a member of the editorial board for Genes & Diseases but was not involved in the editorial review or the decision to publish this article. All other authors declare that there are no competing interests.

Footnotes

Peer review under responsibility of Chongqing Medical University.

References

  • 1.Haikala H.M., Jänne P.A. Thirty years of HER3: from basic biology to therapeutic interventions. Clin Cancer Res. 2021;27(13):3528–3539. doi: 10.1158/1078-0432.CCR-20-4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Burgess A.W., Cho H.S., Eigenbrot C., et al. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell. 2003;12(3):541–552. doi: 10.1016/s1097-2765(03)00350-2. [DOI] [PubMed] [Google Scholar]
  • 3.Jura N., Endres N.F., Engel K., et al. Mechanism for activation of the EGF receptor catalytic domain by the juxtamembrane segment. Cell. 2009;137(7):1293–1307. doi: 10.1016/j.cell.2009.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Guy P.M., Platko J.V., Cantley L.C., Cerione R.A., Carraway K.L., 3rd Insect cell-expressed p180erbB3 possesses an impaired tyrosine kinase activity. Proc Natl Acad Sci U S A. 1994;91(17):8132–8136. doi: 10.1073/pnas.91.17.8132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Holbro T., Civenni G., Hynes N.E. The ErbB receptors and their role in cancer progression. Exp Cell Res. 2003;284(1):99–110. doi: 10.1016/s0014-4827(02)00099-x. [DOI] [PubMed] [Google Scholar]
  • 6.El-Gamal M.I., Mewafi N.H., Abdelmotteleb N.E., et al. A review of HER4 (ErbB4) kinase, its impact on cancer, and its inhibitors. Molecules. 2021;26(23):7376. doi: 10.3390/molecules26237376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Freed D.M., Bessman N.J., Kiyatkin A., et al. EGFR ligands differentially stabilize receptor dimers to specify signaling kinetics. Cell. 2017;171(3):683–695.e18. doi: 10.1016/j.cell.2017.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Montero J.C., Rodríguez-Barrueco R., Ocaña A., Díaz-Rodríguez E., Esparís-Ogando A., Pandiella A. Neuregulins and cancer. Clin Cancer Res. 2008;14(11):3237–3241. doi: 10.1158/1078-0432.CCR-07-5133. [DOI] [PubMed] [Google Scholar]
  • 9.Sheng Q., Liu J. The therapeutic potential of targeting the EGFR family in epithelial ovarian cancer. Br J Cancer. 2011;104(8):1241–1245. doi: 10.1038/bjc.2011.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Roskoski R., Jr. The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol Res. 2014;79:34–74. doi: 10.1016/j.phrs.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 11.Witton C.J., Reeves J.R., Going J.J., Cooke T.G., Bartlett J.M.S. Expression of the HER1-4 family of receptor tyrosine kinases in breast cancer. J Pathol. 2003;200(3):290–297. doi: 10.1002/path.1370. [DOI] [PubMed] [Google Scholar]
  • 12.Lee C.M., Shrieve D.C., Zempolich K.A., et al. 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(2):415–421. doi: 10.1016/j.ygyno.2005.05.045. [DOI] [PubMed] [Google Scholar]
  • 13.Baiocchi G., Lopes A., Coudry R.A., et al. ErbB family immunohistochemical expression in colorectal cancer patients with higher risk of recurrence after radical surgery. Int J Colorectal Dis. 2009;24(9):1059–1068. doi: 10.1007/s00384-009-0702-6. [DOI] [PubMed] [Google Scholar]
  • 14.Hayashi M., Inokuchi M., Takagi Y., et al. High expression of HER3 is associated with a decreased survival in gastric cancer. Clin Cancer Res. 2008;14(23):7843–7849. doi: 10.1158/1078-0432.CCR-08-1064. [DOI] [PubMed] [Google Scholar]
  • 15.Reschke M., Mihic-Probst D., van der Horst E.H., et al. HER3 is a determinant for poor prognosis in melanoma. Clin Cancer Res. 2008;14(16):5188–5197. doi: 10.1158/1078-0432.CCR-08-0186. [DOI] [PubMed] [Google Scholar]
  • 16.Scharpenseel H., Hanssen A., Loges S., et al. EGFR and HER3 expression in circulating tumor cells and tumor tissue from non-small cell lung cancer patients. Sci Rep. 2019;9(1):7406. doi: 10.1038/s41598-019-43678-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tanner B., Hasenclever D., Stern K., et al. ErbB-3 predicts survival in ovarian cancer. J Clin Oncol. 2006;24(26):4317–4323. doi: 10.1200/JCO.2005.04.8397. [DOI] [PubMed] [Google Scholar]
  • 18.Hirakawa T., Nakata B., Amano R., et al. HER3 overexpression as an independent indicator of poor prognosis for patients with curatively resected pancreatic cancer. Oncology. 2011;81(3–4):192–198. doi: 10.1159/000333825. [DOI] [PubMed] [Google Scholar]
  • 19.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 U S A. 1989;86(23):9193–9197. doi: 10.1073/pnas.86.23.9193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wieduwilt M.J., Moasser M.M. The epidermal growth factor receptor family: biology driving targeted therapeutics. Cell Mol Life Sci. 2008;65(10):1566–1584. doi: 10.1007/s00018-008-7440-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Diwanji D., Trenker R., Thaker T.M., et al. Structures of the HER2-HER3-NRG1β complex reveal a dynamic dimer interface. Nature. 2021;600(7888):339–343. doi: 10.1038/s41586-021-04084-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cho H.S., Leahy D.J. Structure of the extracellular region of HER3 reveals an interdomain tether. Science. 2002;297(5585):1330–1333. doi: 10.1126/science.1074611. [DOI] [PubMed] [Google Scholar]
  • 23.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 U S A. 2010;107(17):7692–7697. doi: 10.1073/pnas.1002753107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.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 U S A. 2009;106(51):21608–21613. doi: 10.1073/pnas.0912101106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Steinkamp M.P., Low-Nam S.T., Yang S., Lidke K.A., Lidke D.S., Wilson B.S. erbB3 is an active tyrosine kinase capable of homo- and heterointeractions. Mol Cell Biol. 2014;34(6):965–977. doi: 10.1128/MCB.01605-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Li Y., Liu Z., Zhao Y., et al. PD-L1 expression is regulated by ATP-binding of the ERBB3 pseudokinase domain. Genes Dis. 2022;10(4):1702–1713. doi: 10.1016/j.gendis.2022.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kawakami H., Yonesaka K. HER3 and its ligand, heregulin, as targets for cancer therapy. Recent Pat Anti-Cancer Drug Discov. 2016;11(3):267–274. doi: 10.2174/1574892811666160418123221. [DOI] [PubMed] [Google Scholar]
  • 28.Mishra R., Patel H., Alanazi S., Yuan L., Garrett J.T. HER3 signaling and targeted therapy in cancer. Oncol Rev. 2018;12(1):355. doi: 10.4081/oncol.2018.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yue J., López J.M. Understanding MAPK signaling pathways in apoptosis. Int J Mol Sci. 2020;21(7):2346. doi: 10.3390/ijms21072346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hobbs G.A., Der C.J., Rossman K.L. RAS isoforms and mutations in cancer at a glance. J Cell Sci. 2016;129(7):1287–1292. doi: 10.1242/jcs.182873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rathore M., Zhang W., Wright M., et al. Liver endothelium promotes HER3-mediated cell survival in colorectal cancer with wild-type and mutant KRAS. Mol Cancer Res. 2022;20(6):996–1008. doi: 10.1158/1541-7786.MCR-21-0633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ruiz-Saenz A., Dreyer C., Campbell M.R., Steri V., Gulizia N., Moasser M.M. HER2 amplification in tumors activates PI3K/akt signaling independent of HER3. Cancer Res. 2018;78(13):3645–3658. doi: 10.1158/0008-5472.CAN-18-0430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Somanath P.R., Razorenova O.V., Chen J., Byzova T.V. Akt1 in endothelial cell and angiogenesis. Cell Cycle. 2006;5(5):512–518. doi: 10.4161/cc.5.5.2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Jaiswal B.S., Kljavin N.M., Stawiski E.W., et al. Oncogenic ERBB3 mutations in human cancers. Cancer Cell. 2013;23(5):603–617. doi: 10.1016/j.ccr.2013.04.012. [DOI] [PubMed] [Google Scholar]
  • 35.Wee P., Wang Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers. 2017;9(5):52. doi: 10.3390/cancers9050052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lindsey S., Langhans S.A. Epidermal growth factor signaling in transformed cells. Int Rev Cell Mol Biol. 2015;314:1–41. doi: 10.1016/bs.ircmb.2014.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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(6):3550–3558. doi: 10.1128/mcb.14.6.3550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liang J., Slingerland J.M. Multiple roles of the PI3K/PKB (Akt) pathway in cell cycle progression. Cell Cycle. 2003;2(4):339–345. [PubMed] [Google Scholar]
  • 39.Mishra R., Alanazi S., Yuan L., et al. Activating HER3 mutations in breast cancer. Oncotarget. 2018;9(45):27773–27788. doi: 10.18632/oncotarget.25576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kiavue N., Cabel L., Melaabi S., et al. ERBB3 mutations in cancer: biological aspects, prevalence and therapeutics. Oncogene. 2020;39(3):487–502. doi: 10.1038/s41388-019-1001-5. [DOI] [PubMed] [Google Scholar]
  • 41.Kilroy M.K., Park S., Feroz W., et al. HER3 alterations in cancer and potential clinical implications. Cancers. 2022;14(24):6174. doi: 10.3390/cancers14246174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Stern D.F. ERBB3/HER3 and ERBB2/HER2 duet in mammary development and breast cancer. J Mammary Gland Biol Neoplasia. 2008;13(2):215–223. doi: 10.1007/s10911-008-9083-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jonna S., Feldman R.A., Swensen J., et al. Detection of NRG1 gene fusions in solid tumors. Clin Cancer Res. 2019;25(16):4966–4972. doi: 10.1158/1078-0432.CCR-19-0160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mota J.M., Collier K.A., Barros Costa R.L., et al. A comprehensive review of heregulins, HER3, and HER4 as potential therapeutic targets in cancer. Oncotarget. 2017;8(51):89284–89306. doi: 10.18632/oncotarget.18467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Laskin J., Liu S.V., Tolba K., et al. NRG1 fusion-driven tumors: biology, detection, and the therapeutic role of afatinib and other ErbB-targeting agents. Ann Oncol. 2020;31(12):1693–1703. doi: 10.1016/j.annonc.2020.08.2335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Recondo G., Bahcall M., Spurr L.F., et al. Molecular mechanisms of acquired resistance to MET tyrosine kinase inhibitors in patients with MET exon 14-mutant NSCLC. Clin Cancer Res. 2020;26(11):2615–2625. doi: 10.1158/1078-0432.CCR-19-3608. [DOI] [PubMed] [Google Scholar]
  • 47.Chung Y.W., Kim S., Hong J.H., et al. Overexpression of HER2/HER3 and clinical feature of ovarian cancer. J Gynecol Oncol. 2019;30(5):e75. doi: 10.3802/jgo.2019.30.e75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Stransky N., Egloff A.M., Tward A.D., et al. The mutational landscape of head and neck squamous cell carcinoma. Science. 2011;333(6046):1157–1160. doi: 10.1126/science.1208130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gil V., Miranda S., Riisnaes R., et al. HER3 is an actionable target in advanced prostate cancer. Cancer Res. 2021;81(24):6207–6218. doi: 10.1158/0008-5472.CAN-21-3360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kwon H.J., Park Y., Nam S.K., et al. Genetic and immune microenvironment characterization of HER2-positive gastric cancer: their association with response to trastuzumab-based treatment. Cancer Med. 2023;12(9):10371–10384. doi: 10.1002/cam4.5769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mujoo K., Choi B.K., Huang Z., Zhang N., An Z. Regulation of ERBB3/HER3 signaling in cancer. Oncotarget. 2014;5(21):10222–10236. doi: 10.18632/oncotarget.2655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ding K., Chen X., Li Y., et al. Gastric cancer harboring an ERBB3 mutation treated with a pyrotinib-irinotecan combo: a case study. OncoTargets Ther. 2021;14:545–550. doi: 10.2147/OTT.S286024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Afifi N., Barrero C.A. Understanding breast cancer aggressiveness and its implications in diagnosis and treatment. J Clin Med. 2023;12(4):1375. doi: 10.3390/jcm12041375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Testa U., Castelli G., Pelosi E. Breast cancer: a molecularly heterogenous disease needing subtype-specific treatments. Med Sci. 2020;8(1):18. doi: 10.3390/medsci8010018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lyu H., Han A., Polsdofer E., Liu S., Liu B. Understanding the biology of HER3 receptor as a therapeutic target in human cancer. Acta Pharm Sin B. 2018;8(4):503–510. doi: 10.1016/j.apsb.2018.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Black L.E., Longo J.F., Carroll S.L. Mechanisms of receptor tyrosine-protein kinase ErbB-3 (ERBB3) action in human neoplasia. Am J Pathol. 2019;189(10):1898–1912. doi: 10.1016/j.ajpath.2019.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hamburger A.W. The role of ErbB3 and its binding partners in breast cancer progression and resistance to hormone and tyrosine kinase directed therapies. J Mammary Gland Biol Neoplasia. 2008;13(2):225–233. doi: 10.1007/s10911-008-9077-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Liu X., Liu S., Lyu H., Riker A.I., Zhang Y., Liu B. Development of effective therapeutics targeting HER3 for cancer treatment. Biol Proced Online. 2019;21:5. doi: 10.1186/s12575-019-0093-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Dey N., Williams C., Leyland-Jones B., De P. A critical role for HER3 in HER2-amplified and non-amplified breast cancers: function of a kinase-dead RTK. Am J Transl Res. 2015;7(4):733–750. [PMC free article] [PubMed] [Google Scholar]
  • 60.Spector N.L., Blackwell K.L. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2009;27(34):5838–5847. doi: 10.1200/JCO.2009.22.1507. [DOI] [PubMed] [Google Scholar]
  • 61.Watanabe S., Yonesaka K., Tanizaki 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(3):1258–1268. doi: 10.1002/cam4.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kawakami H., Okamoto I., Yonesaka K., et al. The anti-HER3 antibody patritumab abrogates cetuximab resistance mediated by heregulin in colorectal cancer cells. Oncotarget. 2014;5(23):11847–11856. doi: 10.18632/oncotarget.2663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yang L., Li Y., Shen E., et al. NRG1-dependent activation of HER3 induces primary resistance to trastuzumab in HER2-overexpressing breast cancer cells. Int J Oncol. 2017;51(5):1553–1562. doi: 10.3892/ijo.2017.4130. [DOI] [PubMed] [Google Scholar]
  • 64.Malm M., Frejd F.Y., Ståhl S., Löfblom J. Targeting HER3 using mono- and bispecific antibodies or alternative scaffolds. mAbs. 2016;8(7):1195–1209. doi: 10.1080/19420862.2016.1212147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Schoeberl B., Kudla A., Masson K., et al. Systems biology driving drug development: from design to the clinical testing of the anti-ErbB3 antibody seribantumab (MM-121) NPJ Syst Biol Appl. 2017;3 doi: 10.1038/npjsba.2016.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Garner A.P., Bialucha C.U., Sprague E.R., et al. An antibody that locks HER3 in the inactive conformation inhibits tumor growth driven by HER2 or neuregulin. Cancer Res. 2013;73(19):6024–6035. doi: 10.1158/0008-5472.CAN-13-1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Richards D., Braiteh F., Anthony S., et al. 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. Ann Oncol. 2012;23:ix170. [Google Scholar]
  • 68.Higgins M.J., Gabrail N.Y., Miller K., et al. A phase I/II study of MM-111, a novel bispecific antibody that targets the ErB2/ErB3 heterodimer, in combination with trastuzumab in advanced refractory HER2-positive breast cancer. J Clin Oncol. 2011;29(15_suppl):TPS119. [Google Scholar]
  • 69.Johnston S., Basik M., Hegg R., et al. Inhibition of EGFR, HER2, and HER3 signaling with AZD8931 in combination with anastrozole as an anticancer approach: phase II randomized study in women with endocrine-therapy-naïve advanced breast cancer. Breast Cancer Res Treat. 2016;160(1):91–99. doi: 10.1007/s10549-016-3979-5. [DOI] [PubMed] [Google Scholar]
  • 70.Sung H., Ferlay J., Siegel R.L., et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
  • 71.Timotheadou E., Skarlos D.V., Samantas E., et al. Evaluation of the prognostic role of a panel of biomarkers in stage IB-IIIA non-small cell lung cancer patients. Anticancer Res. 2007;27(6C):4481–4489. [PubMed] [Google Scholar]
  • 72.Manickavasagar T., Yuan W., Carreira S., et al. HER3 expression and MEK activation in non-small-cell lung carcinoma. Lung Cancer Manag. 2021;10(2) doi: 10.2217/lmt-2020-0031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Liu Q., Yu S., Zhao W., Qin S., Chu Q., Wu K. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer. 2018;17(1):53. doi: 10.1186/s12943-018-0793-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yuan M., Huang L.L., Chen J.H., Wu J., Xu Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct Target Ther. 2019;4:61. doi: 10.1038/s41392-019-0099-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Li C., Brand T.M., Iida M., et al. Human epidermal growth factor receptor 3 (HER3) blockade with U3-1287/AMG888 enhances the efficacy of radiation therapy in lung and head and neck carcinoma. Discov Med. 2013;16(87):79–92. [PMC free article] [PubMed] [Google Scholar]
  • 76.LoRusso P., Jänne P.A., Oliveira M., 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(11):3078–3087. doi: 10.1158/1078-0432.CCR-12-3051. [DOI] [PubMed] [Google Scholar]
  • 77.Shimizu T., Yonesaka K., Hayashi H., et al. Phase 1 study of new formulation of patritumab (U3-1287) process 2, a fully human anti-HER3 monoclonal antibody in combination with erlotinib in Japanese patients with advanced non-small cell lung cancer. Cancer Chemother Pharmacol. 2017;79(3):489–495. doi: 10.1007/s00280-016-3231-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sequist L.V., Gray J.E., Harb W.A., et al. Randomized phase II trial of seribantumab in combination with erlotinib in patients with EGFR wild-type non-small cell lung cancer. Oncologist. 2019;24(8):1095–1102. doi: 10.1634/theoncologist.2018-0695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cejalvo J.M., Jacob W., Fleitas Kanonnikoff T., 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(4) doi: 10.1136/esmoopen-2019-000532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kim H.S., Han J.Y., Shin D.H., et al. EGFR and HER3 signaling blockade in invasive mucinous lung adenocarcinoma harboring an NRG1 fusion. Lung Cancer. 2018;124:71–75. doi: 10.1016/j.lungcan.2018.07.026. [DOI] [PubMed] [Google Scholar]
  • 81.Siegel R.L., Miller K.D., Goding Sauer A., et al. Colorectal cancer statistics, 2020. CA Cancer J Clin. 2020;70(3):145–164. doi: 10.3322/caac.21601. [DOI] [PubMed] [Google Scholar]
  • 82.Loree J.M., Bailey A.M., Johnson A.M., et al. Molecular landscape of ERBB2/ERBB3 mutated colorectal cancer. J Natl Cancer Inst. 2018;110(12):1409–1417. doi: 10.1093/jnci/djy067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Appert-Collin A., Hubert P., Crémel G., Bennasroune A. Role of ErbB receptors in cancer cell migration and invasion. Front Pharmacol. 2015;6:283. doi: 10.3389/fphar.2015.00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yan Q., Guo K., Feng G., et al. Association between the overexpression of Her3 and clinical pathology and prognosis of colorectal cancer: a meta-analysis. Medicine. 2018;97(37) doi: 10.1097/MD.0000000000012317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lédel F., Stenstedt K., Hallström M., Ragnhammar P., Edler D. HER3 expression in primary colorectal cancer including corresponding metastases in lymph node and liver. Acta Oncol. 2015;54(4):480–486. doi: 10.3109/0284186X.2014.983654. [DOI] [PubMed] [Google Scholar]
  • 86.Kountourakis P., Pavlakis K., Psyrri A., et al. Prognostic significance of HER3 and HER4 protein expression in colorectal adenocarcinomas. BMC Cancer. 2006;6:46. doi: 10.1186/1471-2407-6-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bent A.H., Maru D.M., Vauthey J.N., et al. HER3 expression in metastatic colorectal cancer: defining the clinicomolecular profile of an emerging target. J Clin Oncol. 2022;40(16_suppl l) 3588–3588. [Google Scholar]
  • 88.Wang R., Bhattacharya R., Ye X., Fan F., Boulbes D.R., Ellis L.M. Endothelial cells promote colorectal cancer cell survival by activating the HER3-AKT pathway in a paracrine fashion. Mol Cancer Res. 2019;17(1):20–29. doi: 10.1158/1541-7786.MCR-18-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.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. doi: 10.1016/j.ctrv.2018.06.011. [DOI] [PubMed] [Google Scholar]
  • 90.Wakui H., Yamamoto N., Nakamichi S., et al. Phase 1 and dose-finding study of patritumab (U3-1287), a human monoclonal antibody targeting HER3, in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2014;73(3):511–516. doi: 10.1007/s00280-014-2375-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hill A.G., Findlay M.P., Burge M.E., 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(10):2276–2284. doi: 10.1158/1078-0432.CCR-17-0646. [DOI] [PubMed] [Google Scholar]
  • 92.Propper D.J., Gao F., Saunders M.P., et al. PANTHER: AZD8931,inhibitor of EGFR, ERBB2 and ERBB3 signalling, combined with FOLFIRI: a phase I/II study to determine the importance of schedule and activity in colorectal cancer. Br J Cancer. 2023;128(2):245–254. doi: 10.1038/s41416-022-02015-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Siegel R.L., Miller K.D., Fuchs H.E., Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
  • 94.Schram A.M., Goto K., Kim D.W., et al. Efficacy and safety of zenocutuzumab, a HER2 x HER3 bispecific antibody, across advanced NRG1 fusion (NRG1+) cancers. J Clin Oncol. 2022;40(16_suppl):105. [Google Scholar]
  • 95.American Cancer Society. Cancer facts and figures 2022. Available at: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2022/2022-cancer-facts-and-figures.pdf. Accessed on March, 2023.
  • 96.Cîrstea A.E., Stepan A.E., Mărgăritescu C., Zăvoi R.E., Olimid D.A., Simionescu C.E. The immunoexpression of EGFR, HER2 and HER3 in malignant serous ovarian tumors. Rom J Morphol Embryol. 2017;58(4):1269–1273. [PubMed] [Google Scholar]
  • 97.Kojima Y., Sudo K., Yoshida H., et al. Changes in HER3 expression profiles between primary and recurrent gynecological cancers. Cancer Cell Int. 2023;23(1):18. doi: 10.1186/s12935-022-02844-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Li X., Duan Y., Qiao C., et al. Anti-HER3 monoclonal antibody inhibits acquired trastuzumab-resistant gynecologic cancers. Technol Cancer Res Treat. 2016;15(4):573–582. doi: 10.1177/1533034615588422. [DOI] [PubMed] [Google Scholar]
  • 99.Davies S., Holmes A., Lomo L., et al. High incidence of ErbB3, ErbB4, and MET expression in ovarian cancer. Int J Gynecol Pathol. 2014;33(4):402–410. doi: 10.1097/PGP.0000000000000081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chang C.S., Shim J.I., Byeon S.J., et al. Prognostic significance of HER3 expression in patients with cervical cancer. Cancers. 2022;14(9):2139. doi: 10.3390/cancers14092139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hafeez U., Parslow A.C., Gan H.K., Scott A.M. New insights into ErbB3 function and therapeutic targeting in cancer. Expert Rev Anticancer Ther. 2020;20(12):1057–1074. doi: 10.1080/14737140.2020.1829485. [DOI] [PubMed] [Google Scholar]
  • 102.Liu J.F., Ray-Coquard I., Selle F., 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(36):4345–4353. doi: 10.1200/JCO.2016.67.1891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.American Society of Clinical Oncology. Head and neck cancer: statistics. Available at: https://www.cancer.net/cancer-types/head-and-neck-cancer/statistics. Accessed 13 December 2022.
  • 104.Takikita M., Xie R., Chung J.Y., et al. Membranous expression of Her3 is associated with a decreased survival in head and neck squamous cell carcinoma. J Transl Med. 2011;9:126. doi: 10.1186/1479-5876-9-126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wei Q., Sheng L., Shui Y., Hu Q., Nordgren H., Carlsson J. EGFR, HER2, and HER3 expression in laryngeal primary tumors and corresponding metastases. Ann Surg Oncol. 2008;15(4):1193–1201. doi: 10.1245/s10434-007-9771-3. [DOI] [PubMed] [Google Scholar]
  • 106.Reynolds K.L., Bedard P.L., Lee S.H., 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(1):646. doi: 10.1186/s12885-017-3641-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jimeno A., Machiels J.P., Wirth L., 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(24):3803–3811. doi: 10.1002/cncr.30256. [DOI] [PubMed] [Google Scholar]
  • 108.Dillon M.T., Grove L., Newbold K.L., 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(2):487–495. doi: 10.1158/1078-0432.CCR-18-1539. [DOI] [PubMed] [Google Scholar]
  • 109.Zhang Z., Karthaus W.R., Lee Y.S., et al. Tumor microenvironment-derived NRG1 promotes antiandrogen resistance in prostate cancer. Cancer Cell. 2020;38(2):279–296.e9. doi: 10.1016/j.ccell.2020.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Molife L.R., Omlin A., Jones R.J., et al. Randomized phase II trial of nintedanib, afatinib and sequential combination in castration-resistant prostate cancer. Future Oncol. 2014;10(2):219–231. doi: 10.2217/fon.13.250. [DOI] [PubMed] [Google Scholar]
  • 111.Drilon A., Somwar R., Mangatt B.P., et al. Response to ERBB3-directed targeted therapy in NRG1-rearranged cancers. Cancer Discov. 2018;8(6):686–695. doi: 10.1158/2159-8290.CD-17-1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Schram A.M., Chang M.T., Jonsson P., Drilon A. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol. 2017;14(12):735–748. doi: 10.1038/nrclinonc.2017.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Fernandez-Cuesta L., Plenker D., Osada H., et al. CD74-NRG1 fusions in lung adenocarcinoma. Cancer Discov. 2014;4(4):415–422. doi: 10.1158/2159-8290.CD-13-0633. [DOI] [PubMed] [Google Scholar]
  • 114.Aguirre A.J. Oncogenic NRG1 fusions: a new hope for targeted therapy in pancreatic cancer. Clin Cancer Res. 2019;25(15):4589–4591. doi: 10.1158/1078-0432.CCR-19-1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Drilon A., Duruisseaux M., Han J.Y., et al. Clinicopathologic features and response to therapy of NRG1 fusion-driven lung cancers: the eNRGy1 global multicenter registry. J Clin Oncol. 2021;39(25):2791–2802. doi: 10.1200/JCO.20.03307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tirunagaru V.G., Estrada-Bernal A., Yu H., et al. Abstract 2202: tarloxotinib exhibits potent activity in NRG1 fusion and rearranged cancers. Cancer Res. 2019;79(13_suppl):2202. [Google Scholar]
  • 117.Jones M.R., Williamson L.M., Topham J.T., et al. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin Cancer Res. 2019;25(15):4674–4681. doi: 10.1158/1078-0432.CCR-19-0191. [DOI] [PubMed] [Google Scholar]
  • 118.Carrizosa D.R., Burkard M.E., Elamin Y.Y., et al. CRESTONE: initial efficacy and safety of seribantumab in solid tumors harboring NRG1 fusions. J Clin Oncol. 2022;40(16_suppl l) 3006–3006. [Google Scholar]
  • 119.Joubert N., Beck A., Dumontet C., Denevault-Sabourin C. Antibody-drug conjugates: the last decade. Pharmaceuticals. 2020;13(9):245. doi: 10.3390/ph13090245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yonemori K., Masuda N., Takahashi S., et al. Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in HER3-overexpressing metastatic breast cancer: updated results from a phase I/II trial. Ann Oncol. 2019;30:iii48. [Google Scholar]
  • 121.Jänne P.A., Baik C., Su W.C., 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(1):74–89. doi: 10.1158/2159-8290.CD-21-0715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yonesaka K., Tanizaki J., Maenishi O., et al. HER3 augmentation via blockade of EGFR/AKT signaling enhances anticancer activity of HER3-targeting patritumab deruxtecan in EGFR-mutated non-small cell lung cancer. Clin Cancer Res. 2022;28(2):390–403. doi: 10.1158/1078-0432.CCR-21-3359. [DOI] [PubMed] [Google Scholar]
  • 123.Koyama K., Ishikawa H., Abe M., et al. Patritumab deruxtecan (HER3-DXd), a novel HER3 directed antibody drug conjugate, exhibits in vitro activity against breast cancer cells expressing HER3 mutations with and without HER2 overexpression. PLoS One. 2022;17(5) doi: 10.1371/journal.pone.0267027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Haratani K., Yonesaka K., Takamura S., et al. U3-1402 sensitizes HER3-expressing tumors to PD-1 blockade by immune activation. J Clin Investig. 2020;130(1):374–388. doi: 10.1172/JCI126598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Weng W., Meng T., Zhao Q., et al. Antibody-exatecan conjugates with a novel self-immolative moiety overcome resistance in colon and lung cancer. Cancer Discov. 2023;13(4):950–973. doi: 10.1158/2159-8290.CD-22-1368. [DOI] [PubMed] [Google Scholar]
  • 126.Anderson N.M., Simon M.C. The tumor microenvironment. Curr Biol. 2020;30(16):R921–R925. doi: 10.1016/j.cub.2020.06.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Guardia C., Bianchini G., Arpí-LLucià O., et al. Preclinical and clinical characterization of fibroblast-derived neuregulin-1 on trastuzumab and pertuzumab activity in HER2-positive breast cancer. Clin Cancer Res. 2021;27(18):5096–5108. doi: 10.1158/1078-0432.CCR-20-2915. [DOI] [PubMed] [Google Scholar]
  • 128.Rathore M., Zhang W., Wright M., et al. Liver endothelium microenvironment promotes HER3-mediated cell growth in pancreatic ductal adenocarcinoma. J Cancer Sci Clin Ther. 2022;6(4):431–445. doi: 10.26502/jcsct.5079182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ryan D.P., Hong T.S., Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371(22):2140–2141. doi: 10.1056/NEJMc1412266. [DOI] [PubMed] [Google Scholar]
  • 130.Mielgo A., Schmid M.C. Liver tropism in cancer: the hepatic metastatic niche. Cold Spring Harb Perspect Med. 2020;10(3) doi: 10.1101/cshperspect.a037259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Siebenhüner A.R., Güller U., Warschkow R. Population-based SEER analysis of survival in colorectal cancer patients with or without resection of lung and liver metastases. BMC Cancer. 2020;20(1):246. doi: 10.1186/s12885-020-6710-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Ogier C., Colombo P.E., Bousquet C., et al. Targeting the NRG1/HER3 pathway in tumor cells and cancer-associated fibroblasts with an anti-neuregulin 1 antibody inhibits tumor growth in pre-clinical models of pancreatic cancer. Cancer Lett. 2018;432:227–236. doi: 10.1016/j.canlet.2018.06.023. [DOI] [PubMed] [Google Scholar]
  • 133.Cabrera R.M., Mao S.P.H., Surve C.R., Condeelis J.S., Segall J.E. A novel neuregulin - jagged1 paracrine loop in breast cancer transendothelial migration. Breast Cancer Res. 2018;20(1):24. doi: 10.1186/s13058-018-0960-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Okita R., Mougiakakos D., Ando T., et al. HER2/HER3 signaling regulates NK cell-mediated cytotoxicity via MHC class I chain-related molecule A and B expression in human breast cancer cell lines. J Immunol. 2012;188(5):2136–2145. doi: 10.4049/jimmunol.1102237. [DOI] [PubMed] [Google Scholar]
  • 135.Yang X., Chen Y., Li M., Zhu W. ERBB3 methylation and immune infiltration in tumor microenvironment of cervical cancer. Sci Rep. 2022;12(1):8112. doi: 10.1038/s41598-022-11415-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kumai T., Ohkuri T., Nagato T., et al. Targeting HER-3 to elicit antitumor helper T cells against head and neck squamous cell carcinoma. Sci Rep. 2015;5 doi: 10.1038/srep16280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Basu A., Albert G.K., Awshah S., et al. Identification of immunogenic MHC class II human HER3 peptides that mediate anti-HER3 CD4+ Th1 responses and potential use as a cancer vaccine. Cancer Immunol Res. 2022;10(1):108–125. doi: 10.1158/2326-6066.CIR-21-0454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Osada T., Morse M.A., Hobeika A., et al. Vaccination targeting human HER3 alters the phenotype of infiltrating T cells and responses to immune checkpoint inhibition. OncoImmunology. 2017;6(6) doi: 10.1080/2162402X.2017.1315495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Mukai H., Saeki T., Aogi K., et al. Patritumab plus trastuzumab and paclitaxel in human epidermal growth factor receptor 2-overexpressing metastatic breast cancer. Cancer Sci. 2016;107(10):1465–1470. doi: 10.1111/cas.13017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ocana A., Vera-Badillo F., Seruga B., Templeton A., Pandiella A., Amir E. HER3 overexpression and survival in solid tumors: a meta-analysis. J Natl Cancer Inst. 2013;105(4):266–273. doi: 10.1093/jnci/djs501. [DOI] [PubMed] [Google Scholar]
  • 141.Krop I.E., Masuda N., Mukohara T., et al. Patritumab deruxtecan (HER3-DXd), a human epidermal growth factor receptor 3-directed antibody-drug conjugate, in patients with previously treated human epidermal growth factor receptor 3-expressing metastatic breast cancer: a multicenter, phase I/II trial. J Clin Oncol. 2023;41(36):5550–5560. doi: 10.1200/JCO.23.00882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gan H.K., Millward M., Jalving M., et al. A phase I, first-in-human study of GSK2849330, an anti-HER3 monoclonal antibody, in HER3-expressing solid tumors. Oncologist. 2021;26(10):e1844–e1853. doi: 10.1002/onco.13860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Yu H.A., Goto Y., Hayashi H., et al. HERTHENA-Lung01, a phase II trial of patritumab deruxtecan (HER3-DXd) in epidermal growth factor receptor-mutated non-small-cell lung cancer after epidermal growth factor receptor tyrosine kinase inhibitor therapy and platinum-based chemotherapy. J Clin Oncol. 2023;41(35):5363–5375. doi: 10.1200/JCO.23.01476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Rathore M.G., Zhang W., Wright M., et al. Abstract 3177: liver endothelium secreted LRG1 is a novel ligand of HER3 to promote metastatic colorectal cancer growth. Cancer Res. 2022;82(12_suppl):3177. [Google Scholar]
  • 145.Romaniello D., Marrocco I., Belugali Nataraj N., et al. Targeting HER3, a catalytically defective receptor tyrosine kinase, prevents resistance of lung cancer to a third-generation EGFR kinase inhibitor. Cancers. 2020;12(9):2394. doi: 10.3390/cancers12092394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Schlam I., Tarantino P., Tolaney S.M. Overcoming resistance to HER2-directed therapies in breast cancer. Cancers. 2022;14(16):3996. doi: 10.3390/cancers14163996. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Genes & Diseases are provided here courtesy of Chongqing Medical University

RESOURCES