Skip to main content

HER2-targeted therapies in cancer: a systematic review

Abstract

Abnormal alterations in human epidermal growth factor receptor 2 (HER2, neu, and erbB2) are associated with the development of many tumors. It is currently a crucial treatment for multiple cancers. Advanced in molecular biology and further exploration of the HER2-mediated pathway have promoted the development of medicine design and combination drug regimens. An increasing number of HER2-targeted drugs including specific monoclonal antibodies, tyrosine kinase inhibitors (TKIs), and antibody-drug conjugates (ADCs) have been approved by the U.S. Food and Drug Administration. The emergence of ADCs, has significantly transformed the treatment landscape for various tumors, such as breast, gastric, and bladder cancer. Classic monoclonal antibodies and novel TKIs have not only demonstrated remarkable efficacy, but also expanded their indications, with ADCs in particular exhibiting profound clinical applications. Moreover the concept of low HER2 expression signifies a breakthrough in HER2-targeted therapy, indicating that an increasing number of tumors and patients will benefit from this approach. This article, provides a comprehensive review of the underlying mechanism of action, representative drugs, corresponding clinical trials, recent advancements, and future research directions pertaining to HER2-targeted therapy.

Introduction

Human growth factor receptor 2(HER2/erbB2) belongs to the epidermal growth factor receptor(EGFR) family and comprises four members: erbB1(EGFR/HER1), erbB2(HER2), erbB3(HER3), and erbB4(HER4) [1]. These family members mainly consist of three domains: an extracellular domain (ECD), a transmembrane domain (TMD), and an intracellular region [1]. The ECD consists of four subdomains (I-IV) [2]. The ECD adopts a closed conformation involving domains II and IV in the absence of ligand. Ligand binding between domains I and III untethers the dimerization arm in domain II, leading to receptor homodimers or heterodimerization, allosteric kinase activation, and downstream signaling [3]. Overexpressing of HER2 proteins facilitates the formation of either homodimers or heterodimers [4]. Therefore promoting growth and proliferation are promoted primarily through the MAPK/ERK and PI3K/AKT/mTOR pathways [5].

HER2 alterations in diverse cancers

Alterations in HER2 expression in tumor cells primarily arise from the mutation, amplification or overexpression of the HER2 gene (erbB2). Mutations in HER2 are most commonly observed within the intracellular tyrosine kinase structural domain, encompassing exon 20 (20%), exon 19 (11%) and exon 21 (9%). The mutation hotspot varies across different types of cancer [6]. The presence of HER2 mutations does not always coincide with HER2 amplification [7], which is often associated with the overexpression of the HER2 protein [8]. HER2 amplification and overexpression have been observed in various types of cancers, including but not limited to breast cancer, gastric cancer, non-small cell lung cancer (NSCLC), bile duct cancer, bladder cancer, and colorectal cancer [9,10,11,12,13]. This association may indicate an unfavorable prognosis or the development of drug resistance in the treatment of various malignancies [14].

The overexpression of HER2 is associated with aggressive behavior and a poorer prognosis in patients with breast cancer and bladder cancer [9, 15,16,17]. Additionally, amplification or overexpression of the HER2 gene is linked to unfavorable clinicopathological features and prognosis in biliary tract cancers [18]. HER2 alterations have been identified as oncogenic drivers in lung cancer, all of which are correlated with an unfavorable prognosis [19]. While the prognostic significance of HER2 in gastric/gastroesophageal junction adenocarcinoma and colorectal cancer remains debated, some studies suggest that HER2 amplification may be indicative of a poor prognosis [20,21,22,23]. However, the QUASAR,FUCOS,and PICCOLO trials have shown no significant correlation between overall survival(OS) or progression-free survival(PFS) in patients with HER2-amplified mCRC [24]. However, studies by Sarah B Fisher [25] and Shen et al. [26] indicated no significant relationship between HER2 expression and gastric cancer prognosis. It is important to note that differences in population characteristics included in the studies may account for the discrepancy in findings [27]. Furthermore, the frequency of amplification is greater among patients with KRAS/BRAF wild-type mutations than among other patients [28, 29]. Patients with either HER2-amplified mCRC or those who have exon 20 insertions(ex20ins) may also face a greater risk of developing brain metastases [29].

HER2-targeted therapies

The current classification of HER2-targeted drugs includes antibodies, tyrosine kinase inhibitors(TKIs) and antibody-drug conjugates (ADCs) (Fig. 1). The mechanism of action of antibodies involves two primary aspects: binding to the extracellular domain of the HER2 protein, preventing the formation of HER2-containing heterodimers, modulating the downstream effectors of ERBB2 signaling, and recruiting extracellular immune cells for antibody-dependent cell-mediated cytotoxic effects (ADCCs) [30,31,32]. Representative drugs include trastuzumab, pertuzumab and ZW25. ZW25, a bispecific antibody, can target both HER2 extracellular region II (pertuzumab binding site) and IV (trastuzumab binding site) and activate ADCC [33,34,35,36].

Fig. 1
figure 1

Classification and mechanisms of prevalent anti-HER2-targeting medications. (PTEN: phosphatase and tensin homolog; PDK1: phosphoinositide-dependent protein kinase 1; AKT: protein kinase B; TSC1/2: tuberous sclerosis complex 1/2; GTP: guanosine triphosphate; GDP: guanosine diphosphate; mTORC: mammalian target of rapamycin; GRB2: growth factor receptor-bound protein 2; SOS: guanosine release protein; MEK: mitogen-activated extracellular signal-regulated kinase; ERK:extracellular signal-regulated kinase; PLC: phospholipase C; PKC: protein kinase C; STAT: signal transducer and activator of transcription)

Unlike antibodies, small molecule tyrosine kinase inhibitors (TKIs) can Unlike catalyze the kinase structure within cells and compete with ATP to inhibit the downstream signaling of the HER2 family [37]. TKI drugs block the phosphorylation of tyrosine kinase residues in the PI3K/AKT and MAPK pathways, which regulate tumor cell proliferation, migration, angiogenesis, drug resistance, and apoptosis [38]. In addition, TKI drugs with small molecular weights and high lipid solubilities can effectively penetrate the blood-brain barrier [39], which makes them promising for application in tumors with brain metastases.

ADCs are composed of an antibody, a linker and a cytotoxic small molecule, as exemplified by T-DM1, RC48, and DS-8201 [39]. After the antibody binds to the HER2 protein, the molecules are endocytosed into the cell and subsequently cleaved. The release of cytotoxic small molecules causes damage to DNA, tubulin, or other substances, inhibiting the growth, proliferation, survival, and development of cancer cells [27]. Moreover, these cytotoxic small molecules can diffuse to neighboring target cells and cause nonspecific tumor cell death, known as the bystander effect [40] (Fig. 1). ADCs not only preserve monoclonal antibody efficacy but also enhance cytotoxicity through the incorporation of a small molecule payload, thereby significantly augmenting the therapeutic potential of antitumor drugs [41].

HER2 targeted therapies in different cancer

Breast cancer

Antibodies

The introduction of HER2-targeted therapy has revolutionized the landscape of breast cancer treatment. The currently approved HER2 monoclonal antibodies (mAb) for breast cancer treatment include trastuzumab and pertuzumab. Trastuzumab (Herceptin) was the first anti-HER2 humanized mAb developed and was approved for marketing by the U.S. Food and Drug Administration (FDA) in 1998 [33]. Preclinical data demonstrating the synergistic effects of cytotoxic agents and trastuzumab have been obtained, subsequently, many clinical studies have confirmed the powerful therapeutic efficacy of these agents [42,43,44,45,46,47,48,49]. It is currently used for neoadjuvant, adjuvant and advanced salvage therapy for HER2-overexpressed breast cancer. Pertuzumab (Perjeta) was approved for marketing by the FDA in 2012. This is attributed to the findings of several pivotal studies, including the NeoSphere, APHINITY, and CLEOPATRA trials [50,51,52]. These investigations have demonstrated that the pertuzumab combined with trastuzumab (HP) regimen can further enhance outcomes and prolong survival in patients with HER2-positive breast cancer across neoadjuvant and adjuvant therapy as well as advanced first-line treatment. HP has become an option for neoadjuvant and adjuvant postoperative adjuvant therapy in patients with HER2-positive early breast cancer ≥T2 or ≥N1 and is a standard recommendation for first-line treatment of patients with metastatic breast cancer [53].

TKIs

Currently, lapatinib, pyrotinib, tucatinib and neratinib are employed in clinical settings. Lapatinib was approved by the FDA on March 13, 2007 for second-line treatment in patients with advanced or metastatic breast cancer with HER2 overexpression [54]. The updated findings from the ALTERNATIVE Phase III trial demonstrated that in postmenopausal women with HR-positive/HER2-positive advanced breast cancer, the combination of aromatase inhibitors (AIs) with lapatinib significantly extended the median progression-free survival (mPFS) compared to that of patients receiving AI monotherapy (11 vs. 5.6 months, P = 0.0063) [54]. The NMPA approved pyrotinib on August 16, 2018, for the treatment of patients with recurrent or metastatic HER2-positive breast cancer who had have not received prior trastuzumab therapy. The PHEDRA and PHILA randomized phase III studies demonstrated the benefits of pyrotinib as a neoadjuvant and advanced first-line therapy for HER2-positive breast cancer [55, 56]. Consequently, in 2020, the Chinese Society of Clinical Oncology (CSCO) has classified pyrotinib as recommended for these patients [57]. In 2020, the FDA approved tucatinib in combination with trastuzumab and capecitabine as a treatment option for patients with advanced HER2-positive breast cancer [58]. These findings are based on the results from the phase III clinical trial (HER2CLIMB) [59]. Neratinib, a dual EGFR/HER2 inhibitor, was FDA-approved for sale on July 17, 2017. Based on the ExteNET study results: patients with HER2 overexpression and high-risk factors after surgery can receive neratinib as intensive adjuvant therapy for one year [60]. This finding highlights the need for intensive anti-HER2 adjuvant therapy in early-stage breast cancer patients at high risk of recurrence.

ADCs

Currently, there are two FDA-approved ADCs for the treatment of breast cancer: Trastuzumab Emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd). T-DM1 consists of trastuzumab and the cytotoxic agent mertansine [61]. DS-8201 comprises trastuzumab and a topoisomerase I inhibitor (Deruxtecan) [62]. Compared to T-DM1, T-Dxd has lysable linkers and has a greater drug-to-antibody ratio (8:1), resulting in greater antitumor effects [63]. According to the EMILIA trial [64], TDM1 was associated with an increase in progression-free survival (PFS) to 9.6 months in patients with HER2-positive advanced breast cancer who previously failed trastuzumab and paclitaxel treatment. Furthermore, the DESTINY BREAST03 study confirmed that T-DXd further extended the median PFS to 28.8 months under the same inclusion criteria [63]. The effectiveness of T-DXd in breast cancer patients with low expression of HER2 expression (IHC1+ or IHC2+ and FISH-) was confirmed by the randomized controlled phase III study, DESTINY-Breast04 [65]. This finding provides a novel justification for the precise treatment of patients exhibiting low HER2 expression, thereby compelling the use of FDA-approved T-DXd for the treatment of patients with low HER2 expression in breast cancer.

As of 2021, there are curruntly 34 ADC drugs in clinical trials targeting HER2. Notable examples include SYD985 and RC48. SYD985 is a combination of trastuzumab and docaramazine analogues linked by cuttable connectors(DARs). Preclinical studies have demonstrated that SYD985 has superior antitumor activity compared to T-DM1 [66]. Preliminary results from the CRISTINA SAURA study demonstrated that treatment with SYD985 was effective (ORR of 33% and mPFS of 9.4 months) in HER2-positive metastatic breast cancer patients [67]. Based on these data, the FDA has granted Fast Track designation to SYD985. A Phase III clinical trial, TULIP, is currently underway. RC48-ADC is a domestic original Chinese ADCs consisting of hertuzumab, a histone cleavable linker and monomethyl auristatin E (MMAE) coupling [68]. A phase III clinical trial of RC48 in the treatment of locally advanced or metastatic breast cancer with low expression of HER2 is underway (Tables 1 and 2). ZW-49 is a double-antibody ADC that can target two different sites of ECD4 and ECD2 [69], and is currently undergoing phase I clinical trials. Many promising ADC drug-related studies are underway (Table 2).

Table 1 Expression of HER2 in different cancers
Table 2 Recent agents targeting HER2 in different cancer

Gastric cancer

Antibodies

Gastric cancer is a prevalent malignancy that ranks fifth in terms of global incidence. Regrettably, the prognosis for advanced or metastatic gastric cancer remains dismal, with a mere 5% to 10% five-year survival rate [106]. Based on the results of the phase III clinical study ToGA, the FDA approved trastuzumab for treating HER2-overexpressing gastric/gastroesophageal junction(G/GEJ) adenocarcinoma in January 2010 [107]. In the phase III clinical trial KEYNOTE-811, pembrolizumab plus trastuzumab resulted in a significant reduction of tumor size and improved objective remission rates in patients with HER2-positive adenocarcinoma of the G/GEJ adenocarcinoma (ORR 74.4%) [108]. A phase II trial demonstrated that using trastuzumab/trastuzumab+pertuzumab combination chemotherapy during the perioperative phase increased the pathologic response rate from 25% to 45% [109]. An ongoing extension phase III clinical trial (INNOVATION), is currently underway. Margetuximab, a monoclonal antibody optimized for the Fc structural domain, has demonstrated improved ADCC effects and antitumor immune activity. In the phase II trial, margetuximab was combined with pablizumab as a second-line treatment for HER2-positive (and/or PD-L1 positive) patients with gastroesophageal cancer (GEA), which resulted in significant improvement in patient survival (HER2-positive: ORR 28.2%, DCR 63.4%, mPFS 4.3 months, mOS 13.9 months) [110]. As a result of these findings, the FDA has approved the use of margetuximab for the treatment of adenocarcinoma of GEA. A phase IB/II trial revealed that combining trastuzumab with the ramucirumab (VEGF-2-targeting agent) improved the prognosis of patients with gastric cancer who had progressed after trastuzumab treatment (mPFS 7.2 months, ORR 33%, DCR 95.6%) [111].

Zanidatamab (ZW25) and KN026 are considered promising bispecific antibodies that target HER2 for the treatment of gastric cancer. In 2019, Zanidatatamab was granted fast track status by the FDA due to its demonstrated efficacy when combined with standard chemotherapy in the first-line treatment of advanced GEA (ORR 54%, DCR 79%) [112]. In a previous phase I clinical trial, KN026 was administered to patients with adenocarcinoma and HER2 overexpression in the gastroesophageal junction, revealing an objective response rate of 55.6% and manageable adverse reactions [113]. Several ongoing clinical trials investigating KN026 and ZW25 for advanced/metastatic gastric cancer hold great promise for their results.

TKIs

Although TKIs have demonstrated efficacy in the treatment of breast cancer, their effectiveness in treating gastric cancer is remians limited. The combination of lapatinib with chemotherapeutic agents, as observed in the tytan and logic trials, did not result in improved overall survival in patients with HER2-positive gastroesophageal adenocarcinoma [114, 115]. This lack of improvement may be attributed to factors such as lapatinib's toxicity, and patient demographic factors, including age and region. While afatinib and tucatinib have shown some antitumor activity in patients with HER2-positive gastroesophageal cancer [116, 117], further research is necessary to determine their specific efficacy.

ADCs

The FDA has approved trastuzumab deruxtecan (DS-8201) for use in patients with locally advanced or metastatic HER2-positive G/GEJ adenocarcinoma who have been treated with trastuzumab based on the results of the DESTINY-Gastric01 trial [118]. The results of the second-phase clinical trial DESTINY-Gastric02 demonstrated that T-DXd monotherapy exhibited remarkable efficacy as a second-line treatment for patients with HER2-positive advanced gastric cancer(ORR 41.8%, overall survival (OS)>1 year). This is currently the highest recorded outcome among second-line treatment regimens [119]. T-DXd has shown clinical activity in patients with previously treated HER2-low expression (IHC 2+/ISH-, IHC 1+) G/GEJ adenocarcinoma, without any reported new adverse effects [118]. A phase II trial demonstrated positive results in the treatment of locally advanced or metastatic gastric cancer with HER2 IHC 2+/3+ in patients who had undergone two or more prior systemic chemotherapies (ORR 25%, DCR 42%, mOS 7.6 months) [120]. In a phase I trial (ACE-Gastric-01), the novel ADC drug ARX788 exhibited good tolerability and antitumor activity in treating patients with HER2-positive advanced gastric cancer and GEJ adenocarcinoma [91]. ARX788 has several advantages due to its Zanidatamab-specific double antibody components and ADC drugs. Currently, clinical trials are underway to explore the use of ARX788 and RC48.

NSCLC

Antibodies

HER2-targeting monoclonal antibodies, like trastuzumab and pertuzumab, have not demonstrated significant antitumor activity when used alone in patients with HER2-mutated NSCLC [121,122,123]. A recent phase I-II study revealed that the combination of trastuzumab and pertuzumab exhibited only modest antitumor activity in advanced HER2-mutated NSCLC patients after multiple treatments [124]. Further investigation are warranted to determine the reasons for the poor efficacy of monoclonal antibody in treating NSCLC.

TKIs

Non-selective tyrosine kinase inhibitors (TKIs), such as afatinib, dacomitinib, and neratinib, exhibit poor antitumor activity against non-small cell lung cancer (NSCLC), and their efficacy may be linked to specific HER2 mutation types [125,126,127,128]. Selective TKIs such as poziotinib and pyrotinib have demonstrated promising antitumor activity in recent studies. In particular, poziotinib has shown superior activity against NSCLC patients with HER2 exon 20 mutations in ex vivo experiments and several phase II clinical trials (ORR 39%, DCR 73%) [129]. It is important to note that major adverse effects such as rash and diarrhea have been reported [130, 131]. Despite these adverse effects, poziotinib is considered to be one of the most efficacious selective TKIs currently available. Preclinical studies have demonstrated that the combination of poziotinib and T-DM1 results in complete tumor regression [6]. Furthermore, in patients with metastatic NSCLC with HER2 mutation or HER2 amplification, the combination of pyrotinib and apatinib has exhibited favorable efficacy (mPFS 5.8-8.5 months) [132]. In addition to the aforementioned TKIs, tarloxotinib and mobocertinib have shown potential effectiveness in treating NSCLC patients with HER2 mutations [133,134,135]. However, further research is needed of the preliminary stage of development of these drugs.

ADCs

T-DXd has been approved by the FDA for the treatment of patients with advanced NSCLC. Other ADC drugs, such as T-DM1, RC48, and SHR-A1811 are also being explored for drug efficacy and safety. The multicenter clinical phase II trial DESTINY-Lung01 [136] demonstrated that T-DXd had effective antitumor activity in patients with HER2 mutations and overexpression (mPFS 6.4-14 vs. 2.8-7 months) [137]. Subsequently, the follow-up study DESTINY-Lung02 revealed that administering T-DXd at a lower dosage significantly decreased the occurrence of interstitial pneumonia (ILD) and other adverse events, while sustaining an objective response rate (ORR) (5.9% vs. 14% for ILD incidence; 53.8% vs. 42.9% for ORR) [138]. The FDA-approved T-DXd for the backline treatment of patients with advanced NSCLC with HER2 mutations was based on the DESTINY-Lung02 study. The ongoing phase Ib clinical trial DESTINY-Lung03 is investigating the safety and efficacy of T-DXd in combination with immunotherapy for patients with HER2-positive advanced NSCLC. However, T-DM1 has limited efficacy in NSCLC patients, with only a few studies showing mild antitumor activity [131, 139]. Additionally, other types of ADC drugs, such as RC48 and SHR-A1811, are undergoing clinical trials for patients with HER2-abnormal NSCLC.

Bladder cancer

Antibodies

HER2 monoclonal antibodies are not yet approved for urothelial carcinoma treatment by the FDA. A multicenter phase II trial conducted by Hussain et al. showed that trastuzumab combined with chemotherapy significantly improved outcomes in advanced urothelial carcinoma patients with HER2-positive disease (DFS 9.3 month, OS 14.1 month) [140]. Another phase I/II trial confirmed that trastuzumab combined with radiotherapy + paclitaxel enhanced the treatment efficacy in patients with muscle-invasive urothelial carcinoma who were unsuitable for bladder resection, but these treatment resulted in significant gastrointestinal toxicity (35%) [141].

TKIs

Current research on TKI in bladder cancer involves lapatinib, afatinib and neratinib, which have shown promising results. In a phase II trial, lapatinib was used as second-line treatment for locally advanced or metastatic cell carcinoma with EGFR and/or HER2 overexpression (median time to progression (TTP) 8.6 weeks, mOS 17.9 weeks) [142]. The phase II trial of afatinib for platinum-refractory metastatic uroepithelial carcinoma demonstrated a significantly longer PFS in patients harboring HER2 or HER3 mutations than those without alterations (6.6 months vs 1.4 months, P<0.001) [143]. Dacomitinib, a second-generation of TKIs, not only inhibits HER2, HER4 and other associated proteins but also has the potential in mitigate resistance issues arising from bypass activation pathways such as HER2 [144]. An early study revealed effective inhibition of daclatinib in HER2-expressing bladder cancer cells [145], and a late-stage study is being prepared. A study investigating the efficacy of neratinib in patients with metastatic bladder cancer harboring a HER2-GRB7 gene fusion is underway.

ADCs

The currently approved ADC for uroepithelial carcinoma is RC48. T-DM1 and TDX-d are being studied. The results from a phase II trial, RC48-C005, demonstrated that RC48-ADC significantly improved objective remission rates and survival in patients with locally advanced or metastatic uroepithelial cancer(ORR 51.2%, mPFS 6.9 months, mOS 13.9 months) [41]. Based on this study, the FDA granted RC48 "Breakthrough Therapy Designation". Preliminary results from the latest study(RC48-C104) demonstrated an ORR of 80% for RC48 in combination with tremelimumab in previously untreated patients with first-line metastatic uroepithelial cancer. The ORR was also reported as follows based on the result of HER2 immunohistochemistry grouping: 100% for HER2-3+, 77.8% for HER2-2+, 66.7% for HER2-1+, and 50% for HER2-negative cases [146] . Therefore, RC48 was approved by CFDA for use in uroepithelial cancer treatment and was recommended by the CSCO guidelines. In preclinical research, T-DM1 was shown to demonstrate strong inhibitory effects on the growth of HER2-positive bladder cancer cell line RT4V6 [147]. A phase IB study, T-Dxd-A-U105, presented preliminary results at the 2022 American Society of Clinical Oncology ASCO congress. The combination of T-Dxd and nivolumab demonstrated significant efficacy in second- and later-line treatment of HER2-expressing urothelial carcinoma(DCR 76.6%, mPFS 6.9 months, mOS 11.0 months); however, it was associated with a notable incidence of serious adverse events (AEs ) [148]. While the combination of ADC and immune checkpoint inhibitors is a promising antitumor strategy, it is important to note that the side effects may be more severe.

Biliary tract cancer (BTC)

Antibodies

While no HER2-targeted drug has been approved for biliary malignancies yet, clinical trials have demonstrated the effectiveness of HER2-targeted therapy in gallbladder cancer. A phase II clinical trial found that trastuzumab had an ORR of 66.6% in the first or second-line treatment of patients with HER2-amplified gallbladder cancer [149]. Another phase IIA trial, MyPathway, demonstrated that combining trastuzumab with pertuzumab improved the treatment efficacy in patients with HER2-positive advanced metastatic biliary tract cancer(ORR 23%) [150]. As a result, the National Comprehensive Cancer Network (NCCN) guidelines recommend trastuzumab and pertuzumab for the treatment of advanced biliary tract tumors with HER2 amplification [151].The bispecific antibody ZW25 has been granted breakthrough therapy status by the FDA for patients with biliary tract cancer who have previously received other treatments and exhibit HER2 gene amplification. The findings from a phase I trial of Zanidatamab as a second-line therapy for biliary tract cancer were presented at the 2021 ASCO meeting, revealing an ORR of 40% and a DCR of 65% [152]. These results were significantly outperformed historical data on second-line chemotherapy. Currently, a global phase IIB study is underway [153].

TKIs

The reversible TKI, lapatinib, effectively inhibits the activation of MAPK, PI3K-AKT (protein kinase B), and phospholipaseCγ (PLCγ) downstream signaling pathways by blocking both HER1 and HER2 [154]. However, despite being evaluated in various clinical settings, lapatinib has not shown efficacy for the management of mUC [155,156,157]. Additionally, the TKI drug neratinib was shown to be safe and well-tolerable for the second-line treatment of HER2-mutated BTC patients(ORR 16%, mPFS 2.8 months, mOS 5.4 months) [158]. However, further research is needed to validate these findings.

ADCs

The exploration of ADC drugs in HER2-positive biliary tract cancer has shown promising results in recent years. A phase II study(HERB) was conducted by ASCO in 2022 to treat HER2-positive BTC patients (ORR 36.4%, DCR 81.8%, mPFS 5.7 months) [159]. Notably, positive outcomes were also observed in patients with low HER2 expression (ORR 12.5%, DCR 75%, mPFS 3.5 months) [159]. A combined analysis of two phase II clinical trials, namely RC48-C005 and RC48-C009, was conducted in a cohort of 107 patients with HER2-positive metastatic urothelial carcinoma (mUC) who had previously failed at least one line of systemic chemotherapy(ORR 50.5%, PFS 5.9 months) [160]. Trastuzumab duocarmazine (SYD985) is composed of trastuzumab, a cleavable linker, and duocarmycin (a DNA alkylator) [161]. Recently, a phase I trial assessed the safety and activity of SYD985 in advanced tumors patients with local or advanced urothelial cancer (partial response rate 25%) [67].

Colorectal cancer

Antibodies and TKIs

Research has shown that the use of HER2 monoclonal antibodies alone is not effective in treating colorectal cancer. However, combining monoclonal antibodies with TKIs has been found to be an effective approach. The combination of trastuzumab and pertuzumab was tested in a phase IIA trial (MyPathway) as first-line treatment for metastatic colorectal cancer (mCRC), Which resulting in an ORR of 32%. Consequently, the NCCN guidelines now recommend using trastuzumab combined with pertuzumab and lapatinib as initial therapies for mCRC [162]. Another phase II trial(HERACLES) demonstrated that lapatinib combined with trastuzumab was active and well tolerated in patients with refractory HER2-positive metastatic colorectal cancer (ORR 35%) [163]. The combination of tucatinib and trastuzumab demonstrated good efficacy in a phase II clinical trial (MOUNTAINEER) (ORR 55%, mPFS 6.2 months, OS 17.3 months) [164]. Similarly, the ongoing phase II trial (HER2-FUSCC-G) demonstrated that the combination of pyrotinib and trastuzumab also demonstrated promising antitumor activity(mPFS 7.53 months, OS 16.8 month) [165].

ADCs

The ADCs that have shown better efficacy in colorectal cancer patients include T-DM1 and T-dxd. According to a phase II clinical study called HERACLES-B, the combination of T-DM1 and pertuzumab has been shown to have low toxicity and high disease control rate (80% in 4 months) in the treatment of HER2-positive mCRC [165]. This combination is presents a viable option for patients with low tumor burden as an active anti-HER2 therapy, while T-DXd monotherapy has shown some antitumor activity in HER2-positive mCRC patients according to the phase II clinical trial (Destiny-CRC01) [166]. Therefore, NCCN guidelines recommend T-Dxd monotherapy as a treatment for mCRC patients with HER2 amplification. However, it is crucial to monitor and intervene in interstitial lung disease (ILD), which remains a potential adverse reaction among patients received T-DXd monotherapy.

Other malignancies

HER2 gene abnormalities (mutation/deletion/amplification) have alse been detected in ovarian cancer, cervical cancer, endometrial cancer, bladder cancer, and salivary gland cancer [167, 168]. However, the efficacy of classic HER2 monoclonal antibodies such as trastuzumab and pertuzumab is not satisfactory for treating uterine or ovarian carcinosarcoma. Preclinical trial data has shown that T-DM1, exhibits potent antitumor activity against chemotherapy-resistant epithelial ovarian cancer (EOC) with HER2-overexpressing [169]. In a phase II clinical trial NCI-MATCH, T-DM1 has demonstrated activity in patients with salivary gland tumors [168]. Additionally, the novel ADC drug SYD985 has shown promising results in preclinical research on uterine and ovarian carcinoma [170]. The specific efficacy of these treatments needs to be confirmed in subsequent clinical trials. The phase II trial DESTINY-PanTumor02 demonstrated that T-DXd exhibited favorable efficacy and safety profiles in various tumor types with HER2 overexpression, including patients with cervical cancer, endometrial cancer, ovarian cancer, biliary tract cancer, and bladder cancer (ORR 37.1%, DCR 83.2%, median duration of response 11.8 months). Importantly, patients with IHC3+ expression had a higher ORR of 61.3% and a longer DOR of 22.1 months [171]. Currently, there are several ongoing clinical trials related to this matter (Table 2).

Conclusions and perspectives

HER2-targeted drugs have been effective at treating tumors for nearly two decades and have become a significant milestone in the field. Trastuzumab, a monoclonal antibody, represents the most advanced drug targeting anti-HER2 therapy and has gained approval the treatment of breast cancer(BC), GC and mCRC treatment [172]. The TKIs targeting HER2 are primarily utilized in BC, NSCLC, and mCRC. In contrast, ADCs have gained FDA approval for the treatment of breast cancer, bladder cancer, and NSCLC. They have also exhibited remarkable efficacy in the treatment of ovarian, bladder, gastric, colon, cervical, endometrial cancers as well as biliary tract cancer; thus holding a promising future.

However the presence of primary and acquired resistance presents significant clinical challenges. The mechanisms underlying primary resistance to anti-HER2 therapy include the following: (1) target receptor inactivation [173]; (2) abnormal activation of downstream components within the PI3K/Akt/mTOR and Ras-Raf-MAPK signaling pathways [174]; (3)overexpression of other HER ligands or receptors [175, 176]; (4) alternative signaling generated by other receptors, such as the insulin-like growth factor-1 receptor (IGF1R) [177]; and (5) the exertion of influence by the tumor microenvironment [178]. Recent studies suggest that miRNA-mediated alterations in gene expression are involve in the acquisition of drug resistance [179]. The membrane-associated glycoprotein mucin-4 (MUC4) may potentially mask the extracellular domain of HER2, thereby impeding effective binding with antibodies [180]. Acquired resistance primarily arises from alterations in the level of target signaling or active target receptors [181]. Furthermore, HER2 gene mutations can result in altered or enhanced interactions between HER2 and trastuzumab, which could manifest as either resistance or sensitivity [182, 183].

In response to resistance to HER2-targeted therapy, two main strategies have been employed. One approach involves the development of drugs with a novel structure and enhanced efficacy. For instance, lapatinib and pyrotinib can act on the intracellular domain of both the HER1 and HER2 proteins. And ADCs can cleave, internalize, and release payloads to HER2-positive cancer cells through their special structure, resulting in improved efficacy. Another strategy focuses on exploring drug combinations to overcome drug resistance, such as utilizing extracellular domain-binding trastuzumab and TKIs in breast and colon cancer treatment. Additionally, the combination of inhibitors targeting the PI3K-AKT-mTOR pathway or immune checkpoints with anti-HER2 agents has demonstrated promising results [184, 185]. A recent study showed that inhibiting the EGFR/HER2 signaling network affects cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinoma (PDAC) organoids and mouse models. Specifically, it reveals that activated myofibroblastic CAFs (myCAFs) through EGFR play a crucial role in promoting PDAC metastasis in mice. These findings highlight the importance of anti-HER2 therapy and immunotherapy as potential treatment strategies for PDAC [186]. A similar strategy can be applied for acquired resistance. Moreover, the HER2 status should be retested to predict the effect of HER2-targeted therapy.

The determination of HER2 status plays a crucial role in the appropriateness of HER2-targeted therapy. However, there is currently a lack of consensus and standardized definitions for HER2-positive across different tumors, thus breast cancer serves as the reference criterion. The diagnostic criteria included immunohistochemical(IHC)+++ or IHC++ and fluorescence in situ hybridization (FISH)-positive. It is important to note that HER2 overexpression and amplification patterns vary among different tumors [67, 187], resulting in varying levels of effectiveness of anti-HER2 therapy in patients with different cancer types. Therefore, there is an urgent need to establish detailed positive criteria for different tumors. Furthermore, performing repeated biopsies to assess HER2 status during disease progression is crucial. The inaccessibility of lesion biopsy remains a challenge in current clinical progress. Clinical trials led by the DESTINY series have recently used liquid biopsy techniques such as ctDNA to detect the HER2 status of patients [188]. This noninvasive method captures the precise expression profile of dynamic and heterogeneous tumor genomes while mitigating the risks associated with bleeding, infection, and tumor dissemination caused by traditional needle biopsy [188]. However, issues such as detection accuracy, positive cutoff value, and cost need to be resolved.

HER2-targeted therapy is a valuable antitumor treatment that merits further enhancement and advancement. While several mechanisms of resistance have been identified, the impact of new drug combinations on resistance profiles has not been determined [189,190,191,192,193]. However, further research is needed to explore the underlying mechanism of these drugs resistance and develop more effective drugs and treatment regimens.

Availability of data and materials

The datasets generated during the current study are available in the cBioPortal repository, cBioPortal for Cancer Genomics.

Abbreviations

ADCs:

Antibody-drug conjugates

ADCC:

Antibody-dependent cell-mediated cytotoxic effects

AKT:

Protein kinase B

BC:

Breast cancer

CSCO:

Chinese society of clinical oncology

CAFs:

Cancer-associated fibroblasts

EGFR:

Epidermal growth factor receptor

ECD:

Extracellular structural domain

EOC:

Epithelial ovarian cancer

ERK:

Extracellular signal-regulated kinase

FDA:

Food and Drug Administration

FISH:

Fluorescence in situ hybridization

GC:

Gastric cancer

G/GEJ:

Gastric/gastroesophageal junction

GEA:

Gastroesophageal cancer

GTP:

Guanosine triphosphate

GDP:

Guanosine diphosphate

GRB2:

Growth factor receptor-bound protein 2

HER2:

Human epidermal growth factor receptor2

IHC:

Immunohistochemical

ILD:

Interstitial pneumonia

mTORC:

Mammalian target of rapamycin

mAbs:

Monoclonal antibodies

mCRC:

Metastatic colorectal cancer

MEK:

Mitogen-activated extracellular signal-regulated kinase

MUC4:

Membrane-associated glycoprotein mucin-4

NSCLC:

Non-small cell lung cancer

NCCN:

National comprehensive cancer network

OS:

Overall survival

ORR:

Objective remission rate

PFS:

Progression-free survival

PTEN:

Phosphatase and tensin homolog

PDK1:

Phosphoinositide-dependent protein kinase 1

PDAC:

Pancreatic ductal adenocarcinoma

PLC:

Phospholipase C

PKC:

Protein kinase C

SOS:

Guanosine release protein

STAT:

Signal transducer and activator of transcription

TKI:

Tyrosine kinase inhibitors

TTP:

Time to progression

TMD:

Transmembrane domain

TSC1/2:

Tuberous sclerosis complex1/2

UC:

Urothelial carcinoma

References

  1. Kovacs E, Zorn JA, Huang Y, Barros T, Kuriyan J. A structural perspective on the regulation of the epidermal growth factor receptor. Ann Rev Biochem. 2015;84:739–64.

    Article  CAS  PubMed  Google Scholar 

  2. Zagozdzon R, Gallagher WM, Crown J. Truncated HER2: implications for HER2-targeted therapeutics. Drug Discov Today. 2011;16(17–18):810–6.

    Article  CAS  PubMed  Google Scholar 

  3. Pahuja KB, Nguyen TT, Jaiswal BS, Prabhash K, Thaker TM, Senger K, et al. Actionable Activating Oncogenic ERBB2/HER2 Transmembrane and Juxtamembrane Domain Mutations. Cancer Cell. 2018;34(5):792-806.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lee-Hoeflich ST, Crocker L, Yao E, Pham T, Munroe X, Hoeflich KP, et al. A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy. Cancer Res. 2008;68(14):5878–87.

    Article  CAS  PubMed  Google Scholar 

  5. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37.

    Article  CAS  PubMed  Google Scholar 

  6. Robichaux JP, Elamin YY, Vijayan RSK, Nilsson MB, Hu L, He J, et al. Pan-cancer landscape and analysis of ERBB2 mutations identifies Poziotinib as a clinically active inhibitor and enhancer of T-DM1 Activity. Cancer Cell. 2019;36(4):444-57.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Desmedt C, Zoppoli G, Gundem G, Pruneri G, Larsimont D, Fornili M, et al. Genomic Characterization of Primary Invasive Lobular Breast Cancer. J Clin Oncol. 2016;34(16):1872–81.

    Article  CAS  PubMed  Google Scholar 

  8. Akbari V, Chou CP, Abedi D. New insights into affinity proteins for HER2-targeted therapy: Beyond trastuzumab. Biochim Biophys Acta Rev Cancer. 2020;1874(2):188448.

    Article  CAS  PubMed  Google Scholar 

  9. Loibl S, Gianni L. HER2-positive breast cancer. Lancet (London, England). 2017;389(10087):2415–29.

    Article  CAS  PubMed  Google Scholar 

  10. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet (London, England). 2010;376(9742):687–97.

    Article  CAS  PubMed  Google Scholar 

  11. Bob T, Zheng T, Ni A, Hellmann M, Jordan E, Barron D, et al. Identifying HER2 mutation, amplification, and HER2 protein overexpression as therapeutic targets in lung cancers. J Clin Oncol. 2016;34:e20666-e.

    Article  Google Scholar 

  12. Raghav K, Overman M, Yu R, Meric-Bernstam F, Menter D, Kee B, et al. HER2 amplification as a negative predictive biomarker for anti-epidermal growth factor receptor antibody therapy in metastatic colorectal cancer. J Clin Oncol. 2016;34:3517.

    Article  Google Scholar 

  13. Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev. 2015;34(1):157–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, et al. HER2 amplification: a potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2012;2(10):922–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet (London, England). 2011;378(9805):1812–23.

    Article  CAS  PubMed  Google Scholar 

  16. Simmons C, Miller N, Geddie W, Gianfelice D, Oldfield M, Dranitsaris G, et al. Does confirmatory tumor biopsy alter the management of breast cancer patients with distant metastases? Ann Oncol. 2009;20(9):1499–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Laé M, Couturier J, Oudard S, Radvanyi F, Beuzeboc P, Vieillefond A. Assessing HER2 gene amplification as a potential target for therapy in invasive urothelial bladder cancer with a standardized methodology: results in 1005 patients. Ann Oncol. 2010;21(4):815–9.

    Article  PubMed  Google Scholar 

  18. Nakazawa K, Dobashi Y, Suzuki S, Fujii H, Takeda Y, Ooi A. Amplification and overexpression of c-erbB-2, epidermal growth factor receptor, and c-met in biliary tract cancers. J Pathol. 2005;206(3):356–65.

    Article  CAS  PubMed  Google Scholar 

  19. Ren S, Wang J, Ying J, Mitsudomi T, Lee DH, Wang Z, et al. Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open. 2022;7(1):100395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. García I, Vizoso F Fau - Martín A, Martín A Fau - Sanz L, Sanz L Fau - Abdel-Lah O, Abdel-Lah O Fau - Raigoso P, Raigoso P Fau - García-Muñiz JL, et al. Clinical significance of the epidermal growth factor receptor and HER2 receptor in resectable gastric cancer. (1068-9265).

  21. Kurokawa Y, Matsuura N, Kimura Y, Adachi S, Fujita J, Imamura H, et al. Multicenter large-scale study of prognostic impact of HER2 expression in patients with resectable gastric cancer. Gastric Cancer. 2015;18(4):691–7.

    Article  CAS  PubMed  Google Scholar 

  22. Mondaca S, Nervi B, Pinto M, Abou-Alfa GK. Biliary tract cancer prognostic and predictive genomics. Chin Clin Oncol. 2019;8(4):42.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ingold Heppner B, Behrens HM, Balschun K, Haag J, Krüger S, Becker T, et al. HER2/neu testing in primary colorectal carcinoma. Br J Cancer. 2014;111(10):1977–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Richman SD, Southward K, Chambers P, Cross D, Barrett J, Hemmings G, et al. HER2 overexpression and amplification as a potential therapeutic target in colorectal cancer: analysis of 3256 patients enrolled in the QUASAR, FOCUS and PICCOLO colorectal cancer trials. J Pathol. 2016;238(4):562–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fisher SB, Fisher KE, Squires MH 3rd, Patel SH, Kooby DA, El-Rayes BF, et al. HER2 in resected gastric cancer: Is there prognostic value? J Surg Oncol. 2014;109(2):61–6.

    Article  PubMed  Google Scholar 

  26. Shen GS, Zhao JD, Zhao JH, Ma XF, Du F, Kan J, et al. Association of HER2 status with prognosis in gastric cancer patients undergoing R0 resection: A large-scale multicenter study in China. (2219-2840 (Electronic)).

  27. Li W, Zhang X, Du Y, Zhang Y, Lu J, Hu W, et al. HER2-targeted advanced metastatic gastric/gastroesophageal junction adenocarcinoma: treatment landscape and future perspectives. Biomark Res. 2022;10:71.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Raghav KPS, Overman MJ, Yu R, Meric-Bernstam F, Menter D, Kee BK, et al. HER2 amplification as a negative predictive biomarker for anti-epidermal growth factor receptor antibody therapy in metastatic colorectal cancer. J Clin Oncol. 2016;34(15_suppl):3517.

    Article  Google Scholar 

  29. Zhao S, Fang W, Pan H, Yang Y, Liang Y, Yang L, et al. Conformational Landscapes of HER2 Exon 20 Insertions Explain Their Sensitivity to Kinase Inhibitors in Lung Adenocarcinoma. J Thorac Oncol. 2020;15(6):962–72.

    Article  CAS  PubMed  Google Scholar 

  30. Hudis CA. Trastuzumab–mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39–51.

    Article  CAS  PubMed  Google Scholar 

  31. Gennari R, Menard S, Fagnoni F, Ponchio L, Scelsi M, Tagliabue E, et al. Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res. 2004;10(17):5650–5.

    Article  CAS  PubMed  Google Scholar 

  32. Ménard S, Casalini P, Campiglio M, Pupa SM, Tagliabue E. Role of HER2/neu in tumor progression and therapy. Cell Mol Life Sci. 2004;61(23):2965–78.

    Article  PubMed  Google Scholar 

  33. Dokmanovic M, Wu Y, Shen Y, Chen J, Hirsch DS, Wu WJ. Trastuzumab-induced recruitment of Csk-homologous kinase (CHK) to ErbB2 receptor is associated with ErbB2-Y1248 phosphorylation and ErbB2 degradation to mediate cell growth inhibition. Cancer Biol Ther. 2014;15(8):1029–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Agus DB, Gordon MS, Taylor C, Natale RB, Karlan B, Mendelson DS, et al. Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer. J Clin Oncol. 2005;23(11):2534–43.

    Article  CAS  PubMed  Google Scholar 

  35. Howie LJ, Scher NS, Amiri-Kordestani L, Zhang L, King-Kallimanis BL, Choudhry Y, et al. FDA Approval Summary: Pertuzumab for Adjuvant Treatment of HER2-Positive Early Breast Cancer. Clin Cancer Res. 2019;25(10):2949–55.

    Article  CAS  PubMed  Google Scholar 

  36. Meric-Bernstam F, Beeram M, Mayordomo J, Hanna D, Ajani J, Murphy M, et al. Single agent activity of ZW25, a HER2-targeted bispecific antibody, in heavily pretreated HER2-expressing cancers. J Clin Oncol. 2018;36:2500.

    Article  Google Scholar 

  37. Singla H, Munshi A, Banipal RPS, Kumar V. Recent Updates on the Therapeutic Potential of HER2 Tyrosine Kinase Inhibitors for the Treatment of Breast Cancer. Curr Cancer Drug Targets. 2018;18(4):306–27.

    Article  CAS  PubMed  Google Scholar 

  38. Yang X, Wu D, Yuan S. Tyrosine Kinase Inhibitors in the Combination Therapy of HER2 Positive Breast Cancer. Technol Cancer ResTreat. 2020;19:1533033820962140.

    CAS  Google Scholar 

  39. Lin NU, Carey LA, Liu MC, Younger J, Come SE, Ewend M, et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2008;26(12):1993–9.

    Article  CAS  PubMed  Google Scholar 

  40. Khera E, Cilliers C, Smith MD, Ganno ML, Lai KC, Keating TA, et al. Quantifying ADC bystander payload penetration with cellular resolution using pharmacodynamic mapping. Neoplasia (New York, NY). 2021;23(2):210–21.

    Article  CAS  Google Scholar 

  41. Sheng X, Yan X, Wang L, Shi Y, Yao X, Luo H, et al. Open-label, Multicenter, Phase II Study of RC48-ADC, a HER2-Targeting Antibody-Drug Conjugate, in Patients with Locally Advanced or Metastatic Urothelial Carcinoma. Clin Cancer Res. 2021;27(1):43–51.

    Article  CAS  PubMed  Google Scholar 

  42. Baselga J, Norton L, Albanell J, Kim YM, Mendelsohn J. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res. 1998;58:2825–31.

    CAS  PubMed  Google Scholar 

  43. Pegram M, et al. Inhibitory efects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers. Oncogene. 1999;18:2241–51.

    Article  CAS  PubMed  Google Scholar 

  44. Pietras RJ, et al. Antibody to HER-2/neu receptor blocks DNA repair after cisplatin in human breast and ovarian cancer cells. Oncogene. 1994;9:1829–38.

    CAS  PubMed  Google Scholar 

  45. Pietras RJ, Pegram MD, Finn RS, Maneval DA, Slamon DJ. Remission of human breast cancer xenografts on therapy with humanized monoclonal antibody to HER-2 receptor and DNA-reactive drugs. Oncogene. 1998;17:2235–49.

    Article  CAS  PubMed  Google Scholar 

  46. Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72.

    Article  CAS  PubMed  Google Scholar 

  47. Romond EH, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–84.

    Article  CAS  PubMed  Google Scholar 

  48. Slamon D, et al. Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med. 2011;365:1273–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Moja, L. et al. Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst Rev. 2012; (2012): CD006243 .

  50. Gianni L, Pienkowski T, Im YH, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17(6):791–800.

    Article  CAS  PubMed  Google Scholar 

  51. von Minckwitz G, Procter M, de Azambuja E, et al. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N Engl J Med. 2017;377(2):122–31.

    Article  Google Scholar 

  52. Swain SM, Kim SB, Cortés J, et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA study): overall survival results from a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2013;14(6):461–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gradishar WJ, Moran MS, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Comprehensive Cancer Netw. 2022;20(6):691–722.

    Article  Google Scholar 

  54. Johnston SRD, Hegg R, Im SA, Park IH, Burdaeva O, Kurteva G, et al. Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade With Lapatinib Plus Trastuzumab in Combination With an Aromatase Inhibitor in Postmenopausal Women With HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer: Updated Results of ALTERNATIVE. J Clin Oncol. 2021;39(1):79–89.

    Article  CAS  PubMed  Google Scholar 

  55. Wu J, Jiang Z, Liu Z, et al. Neoadjuvant pyrotinib, trastuzumab, and docetaxel for HER2-positive breast cancer (PHEDRA): a double-blind, randomized phase 3 trial. BMC Med. 2022;20(1):498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ma F, Yan M, Li W, et al. Pyrotinib versus placebo in combination with trastuzumab and docetaxel as first line treatment in patients with HER2 positive metastatic breast cancer (PHILA): randomised, double blind, multicentre, phase 3 trial. BMJ. 2023;16(383):p2665.

    Google Scholar 

  57. Li Q, Liu J, Jiang Z, Liu Q. CSCO breast cancer guideline: precise, economical and oriental. Sci China Life Sci. 2020;63(9):1410–2.

    Article  PubMed  Google Scholar 

  58. Cortés J, Kim SB, Chung WP, Im SA, Park YH, Hegg R, et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N Engl J Med. 2022;386(12):1143–54.

    Article  PubMed  Google Scholar 

  59. Lin NU, Borges V, Anders C, Murthy RK, Paplomata E, Hamilton E, et al. Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial. J Clin Oncol. 2020;38(23):2610–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Giordano SH, Franzoi MAB, Temin S, Anders CK, Chandarlapaty S, Crews JR, et al. Systemic Therapy for Advanced Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: ASCO Guideline Update. J Clin Oncol. 2022;40(23):2612–35.

    Article  CAS  PubMed  Google Scholar 

  61. Montemurro F, Delaloge S, Barrios CH, Wuerstlein R, Anton A, Brain E, et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial(). Ann Oncol. 2020;31(10):1350–8.

    Article  CAS  PubMed  Google Scholar 

  62. Shitara K, Bang YJ, Iwasa S, Sugimoto N, Ryu MH, Sakai D, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med. 2020;382(25):2419–30.

    Article  CAS  PubMed  Google Scholar 

  63. Cortes J, Kim SB, Chung W-P, Park Y, Hegg R, Kim MH, et al. LBA1 Trastuzumab deruxtecan (T-DXd) vs trastuzumab emtansine (T-DM1) in patients (Pts) with HER2+ metastatic breast cancer (mBC): Results of the randomized phase III DESTINY-Breast03 study. Ann Oncol. 2021;32:S1287–8.

    Article  Google Scholar 

  64. Diéras V, Miles D, Verma S, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial [published correction appears in Lancet Oncol. Lancet Oncol. 2017;18(6):732–42.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N Engl J Med. 2022;387(1):9–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Xu Z, Guo D, Jiang Z, Tong R, Jiang P, Bai L, et al. Novel HER2-Targeting Antibody-Drug Conjugates of Trastuzumab Beyond T-DM1 in Breast Cancer: Trastuzumab Deruxtecan(DS-8201a) and (Vic-)Trastuzumab Duocarmazine (SYD985). Eur J Med Chem. 2019;183:111682.

    Article  CAS  PubMed  Google Scholar 

  67. Banerji U, van Herpen CML, Saura C, Thistlethwaite F, Lord S, Moreno V, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019;20(8):1124–35.

    Article  CAS  PubMed  Google Scholar 

  68. Zhang J, Ji D, Shen W, Xiao Q, Gu Y, O'Shaughnessy J, et al. Phase I Trial of a Novel Anti-HER2 Antibody-drug Conjugate, ARX788, for the Treatment of HER2-positive Metastatic Breast Cancer. Clin Cancer Res. 2022.

  69. Proctor JR, Gartner EM, Gray TE, Davies RH. Population pharmacokinetics of zanidatamab, an anti-HER2 biparatopic antibody, in patients with advanced or metastatic cancer. Cancer Chemother Pharmacol. 2022;90(5):399–408.

    Article  CAS  PubMed  Google Scholar 

  70. Li Q, Jiang B, Guo J, Shao H, Del Priore IS, Chang Q, et al. INK4 Tumor Suppressor Proteins Mediate Resistance to CDK4/6 Kinase Inhibitors. Cancer Discov. 2022;12(2):356–71.

    Article  CAS  PubMed  Google Scholar 

  71. Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, et al. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell. 2018;34(3):427-38.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Smith AE, Ferraro E, Safonov A, Morales CB, Lahuerta EJA, Li Q, et al. HER2 + breast cancers evade anti-HER2 therapy via a switch in driver pathway. Nat Commun. 2021;12(1):6667.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  73. Bang YJ. Advances in the management of HER2-positive advanced gastric and gastroesophageal junction cancer. J Clin Gastroenterol. 2012;46(8):637–48.

    Article  CAS  PubMed  Google Scholar 

  74. Cappellesso R, Fassan M, Hanspeter E, Bornschein J, d’Amore ES, Cuorvo LV, et al. HER2 status in gastroesophageal cancer: a tissue microarray study of 1040 cases. Hum Pathol. 2015;46(5):665–72.

    Article  CAS  PubMed  Google Scholar 

  75. Matsusaka S, Nashimoto A, Nishikawa K, Miki A, Miwa H, Yamaguchi K, et al. Clinicopathological factors associated with HER2 status in gastric cancer: results from a prospective multicenter observational cohort study in a Japanese population (JFMC44-1101). Gastric Cancer. 2016;19(3):839–51.

    Article  CAS  PubMed  Google Scholar 

  76. Valtorta E, Martino C, Sartore-Bianchi A, Penaullt-Llorca F, Viale G, Risio M, et al. Assessment of a HER2 scoring system for colorectal cancer: results from a validation study. Modern Pathol. 2015;28(11):1481–91.

    Article  CAS  Google Scholar 

  77. Integrated genomic characterization of oesophageal carcinoma. Nature. 2017;541(7636):169-75.

  78. Sihag S, Nussenzweig SC, Walch HS, Hsu M, Tan KS, Sanchez-Vega F, et al. Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways. Clin Cancer Res. 2021;27(12):3491–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yoshizawa A, Sumiyoshi S, Sonobe M, Kobayashi M, Uehara T, Fujimoto M, et al. HER2 status in lung adenocarcinoma: a comparison of immunohistochemistry, fluorescence in situ hybridization (FISH), dual-ISH, and gene mutations. Lung Cancer (Amsterdam, Netherlands). 2014;85(3):373–8.

    Article  PubMed  Google Scholar 

  80. Jee J, Lebow ES, Yeh R, Das JP, Namakydoust A, Paik PK, et al. Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer. Nat Med. 2022;28(11):2353–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zuiverloon TCM, Theodorescu D. Re: Genomic Differences Between “Primary” and “Secondary” Muscle-invasive Bladder Cancer as a Basis for Disparate Outcomes to Cisplatin-based Neoadjuvant Chemotherapy. Eur Urol. 2019;75(4):694.

    Article  PubMed  Google Scholar 

  82. Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell. 2017;171(3):540-56.e25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Sohal DP, Shrotriya S, Abazeed M, Cruise M, Khorana A. Molecular characteristics of biliary tract cancer. Crit Rev Oncol/Hematol. 2016;107:111–8.

    Article  PubMed  Google Scholar 

  84. Harder J, Waiz O, Otto F, Geissler M, Olschewski M, Weinhold B, et al. EGFR and HER2 expression in advanced biliary tract cancer. World J Gastroenterol. 2009;15(36):4511–7.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Shafizadeh N, Grenert JP, Sahai V, Kakar S. Epidermal growth factor receptor and HER-2/neu status by immunohistochemistry and fluorescence in situ hybridization in adenocarcinomas of the biliary tree and gallbladder. Hum Pathol. 2010;41(4):485–92.

    Article  CAS  PubMed  Google Scholar 

  86. Giraldo NA, Drill E, Satravada BA, Dika IE, Brannon AR, Dermawan J, et al. Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention. Clin Cancer Res. 2022;28(24):5359–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Oaknin A, Friedman CF, Roman LD, D’Souza A, Brana I, Bidard FC, et al. Neratinib in patients with HER2-mutant, metastatic cervical cancer: Findings from the phase 2 SUMMIT basket trial. Gynecol Oncol. 2020;159(1):150–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Narayan RR, Creasy JM, Goldman DA, Gönen M, Kandoth C, Kundra R, et al. Regional differences in gallbladder cancer pathogenesis: Insights from a multi-institutional comparison of tumor mutations. Cancer. 2019;125(4):575–85.

    Article  CAS  PubMed  Google Scholar 

  89. Seo AN, Kwak Y, Kim DW, Kang SB, Choe G, Kim WH, et al. HER2 status in colorectal cancer: its clinical significance and the relationship between HER2 gene amplification and expression. PloS One. 2014;9(5):e98528.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  90. Mondaca S, Walch H, Nandakumar S, Chatila WK, Schultz N, Yaeger R. Specific Mutations in APC, but Not Alterations in DNA Damage Response, Associate With Outcomes of Patients With Metastatic Colorectal Cancer. Gastroenterology. 2020;159(5):1975-8.e4.

    Article  CAS  PubMed  Google Scholar 

  91. Yaeger R, Chatila WK, Lipsyc MD, Hechtman JF, Cercek A, Sanchez-Vega F, et al. Clinical Sequencing Defines the Genomic Landscape of Metastatic Colorectal Cancer. Cancer Cell. 2018;33(1):125-36.e3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Maximiano S, Magalhães P, Guerreiro MP, Morgado M. Trastuzumab in the Treatment of Breast Cancer. BioDrugs. 2016;30(2):75–86.

    Article  CAS  PubMed  Google Scholar 

  93. Wang FH, Zhang XT, Li YF, Tang L, Qu XJ, Ying JE, et al. The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer, 2021. Cancer Commun (Lond). 2021;41(8):747–95.

    Article  PubMed  Google Scholar 

  94. Gradishar WJ, Moran MS, Abraham J, Aft R, Agnese D, Allison KH, et al. NCCN Guidelines® Insights: Breast Cancer, Version 4.2021. J Natl Compr Canc Netw. 2021;19(5):484–93.

    Article  PubMed  Google Scholar 

  95. Zhang X, Chen J, Weng Z, Li Q, Zhao L, Yu N, et al. A new anti-HER2 antibody that enhances the antitumor efficacy of trastuzumab and pertuzumab with a distinct mechanism of action. Mol Immunol. 2020;119:48–58.

    Article  CAS  PubMed  Google Scholar 

  96. Nur Husna SM, Wong KK. Margetuximab and trastuzumab deruxtecan: New generation of anti-HER2 immunotherapeutic agents for breast cancer. Mol Immunol. 2022;152:45–54.

    Article  CAS  PubMed  Google Scholar 

  97. Molinelli C, Parisi F, Razeti MG, Arecco L, Cosso M, Fregatti P, et al. Trastuzumab emtansine (T-DM1) as adjuvant treatment of HER2-positive early breast cancer: safety and efficacy. Expert Rev Anticancer Ther. 2021;21(3):241–50.

    Article  CAS  PubMed  Google Scholar 

  98. Lee J, Park YH. Trastuzumab deruxtecan for HER2+ advanced breast cancer. Future Oncol. 2022;18(1):7–19.

    Article  CAS  PubMed  Google Scholar 

  99. Zhang H, Karakas C, Tyburski H, Turner BM, Peng Y, Wang X, et al. HER2-low breast cancers: Current insights and future directions. Semin Diagn Pathol. 2022;39(5):305–12.

    Article  PubMed  Google Scholar 

  100. Grieb BC, Agarwal R. HER2-Directed Therapy in Advanced Gastric and Gastroesophageal Adenocarcinoma: Triumphs and Troubles. Curr Treat Options Oncol. 2021;22(10):88.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Riudavets M, Sullivan I, Abdayem P, Planchard D. Targeting HER2 in non-small-cell lung cancer (NSCLC): a glimpse of hope? An updated review on therapeutic strategies in NSCLC harbouring HER2 alterations. ESMO Open. 2021;6(5):100260.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Neratinib for breast cancer. Aust Prescr. 2019;42(6):209–10.

    Article  Google Scholar 

  103. Bilancia D, Rosati G, Dinota A, Germano D, Romano R, Manzione L. Lapatinib in breast cancer. Ann Oncol. 2007;18 Suppl 6:vi26-30.

  104. Lee A. Tucatinib: First Approval. Drugs. 2020;80(10):1033–8.

    Article  CAS  PubMed  Google Scholar 

  105. Blair HA. Pyrotinib: First Global Approval. Drugs. 2018;78(16):1751–5.

    Article  PubMed  Google Scholar 

  106. Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376(9742):687–97.

    Article  PubMed  Google Scholar 

  107. Tabernero J, Hoff PM, Shen L, Ohtsu A, Shah MA, Cheng K, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018;19(10):1372–84.

    Article  CAS  PubMed  Google Scholar 

  108. Janjigian YY, Kawazoe A, Yañez P, Li N, Lonardi S, Kolesnik O, et al. Combined PD-1 and HER2 blockade for HER2-positive gastric cancer. Nature. 2021;600(7890):727–30.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  109. Wagner AD, Grabsch HI, Mauer M, Marreaud S, Caballero C, Thuss-Patience P, et al. EORTC-1203-GITCG - the “INNOVATION”-trial: Effect of chemotherapy alone versus chemotherapy plus trastuzumab, versus chemotherapy plus trastuzumab plus pertuzumab, in the perioperative treatment of HER2 positive, gastric and gastroesophageal junction adenocarcinoma on pathologic response rate: a randomized phase II-intergroup trial of the EORTC-Gastrointestinal Tract Cancer Group, Korean Cancer Study Group and Dutch Upper GI-Cancer group. BMC Cancer. 2019;19(1):494.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Catenacci DVT, Kang YK, Park H, Uronis HE, Lee KW, Ng MCH, et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22-05): a single-arm, phase 1b–2 trial. Lancet Oncol. 2020;21(8):1066–76.

    Article  CAS  PubMed  Google Scholar 

  111. Rha S, Kim C, Jung M, Kim H, Lee C-k, Jeung H-C, et al. Multicenter phase Ib/II study of second-line trastuzumab, ramucirumab, and paclitaxel in patients with HER2-positive advanced gastric or gastroesophageal junction cancer: Updated HER-RAM study with biomarker analysis. J Clin Oncol. 2022;40:330.

  112. Meric-Bernstam F, Hamilton EP, Beeram M, Hanna DL, El-Khoueiry AB, Kang Y-K, et al. Zanidatamab (ZW25) in HER2-expressing gastroesophageal adenocarcinoma (GEA): Results from a phase I study. J Clin Oncol. 2021;39(3_suppl):164.

  113. Xu J, Zhang Y, Wu J, Xu N, Ying J, Xiang X, et al. The preliminary efficacy of KN026 (Anti-HER2 BsAb) in advanced gastric and gastroesophageal junction cancer patients with HER2 expression. J Clin Oncol. 2021;39(15_suppl):e16005-e.

  114. Hecht JR, Bang YJ, Qin SK, Chung HC, Xu JM, Park JO, et al. Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC–A Randomized Phase III Trial. J Clin Oncol. 2016;34(5):443–51.

    Article  CAS  PubMed  Google Scholar 

  115. Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014;32(19):2039-49.

  116. Janjigian YY, Ku GY, Ilson DH, Boyar MS, Capanu M, Chou JF, et al. A phase II study of afatinib in patients (pts) with metastatic human epidermal growth factor receptor (HER2)-positive trastuzumab refractory esophagogastric (EG) cancer. J Clin Oncol. 2015;33(3_suppl):59.

  117. Catenacci DVT, Strickler JH, Nakamura Y, Shitara K, Janjigian YY, Barzi A, et al. MOUNTAINEER-02: Phase 2/3 study of tucatinib, trastuzumab, ramucirumab, and paclitaxel in previously treated HER2+ gastric or gastroesophageal junction adenocarcinoma—Trial in progress. J Clin Oncol. 2022;40(4_suppl):TPS371-TPS.

  118. Yamaguchi K, Bang Y-J, Iwasa S, Sugimoto N, Ryu M, Sakai D, et al. 1422MO Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-low, advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma: Results of the exploratory cohorts in the phase II, multicenter, open-label DESTINY-Gastric01 study. Ann Oncol. 2020;31:S899–900.

    Article  Google Scholar 

  119. Ku GY, Di Bartolomeo M, Smyth E, Chau I, Park H, Siena S, et al. 1205MO Updated analysis of DESTINY-Gastric02: A phase II single-arm trial of trastuzumab deruxtecan (T-DXd) in western patients (Pts) with HER2-positive (HER2+) unresectable/metastatic gastric/gastroesophageal junction (GEJ) cancer who progressed on or after trastuzumab-containing regimen. Ann Oncol. 2022;33:S1100.

    Article  Google Scholar 

  120. Peng Z, Liu T, Wei J, et al. Efficacy and safety of a novel anti-HER2 therapeutic antibody RC48 in patients with HER2-overexpressing, locally advanced or metastatic gastric or gastroesophageal junction cancer: a single-arm phase II study. Cancer Commun (Lond). 2021;41(11):1173–82.

    Article  PubMed  Google Scholar 

  121. Langer CJ, Stephenson P, Thor A, Vangel M, Johnson DH. Trastuzumab in the treatment of advanced non-small-cell lung cancer: is there a role? Focus on Eastern Cooperative Oncology Group study 2598. J Clin Oncol. 2004;22(7):1180–7.

    Article  CAS  PubMed  Google Scholar 

  122. Gatzemeier U, Groth G, Butts C, Van Zandwijk N, Shepherd F, Ardizzoni A, et al. Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in HER2-positive non-small-cell lung cancer. Ann Oncol. 2004;15(1):19–27.

    Article  CAS  PubMed  Google Scholar 

  123. Herbst RS, Davies AM, Natale RB, Dang TP, Schiller JH, Garland LL, et al. Efficacy and safety of single-agent pertuzumab, a human epidermal receptor dimerization inhibitor, in patients with non small cell lung cancer. Clin Cancer Res. 2007;13(20):6175–81.

    Article  CAS  PubMed  Google Scholar 

  124. van Berge Henegouwen JM, Jebbink M, Hoes LR, van der Wijngaart H, Zeverijn LJ, van der Velden DL, et al. Trastuzumab and pertuzumab combination therapy for advanced pre-treated HER2 exon 20-mutated non-small cell lung cancer. Eur J Cancer (Oxford, England: 1990). 2022;171:114-23.

  125. Dziadziuszko R, Smit EF, Dafni U, Wolf J, Wasąg B, Biernat W, et al. Afatinib in NSCLC With HER2 Mutations: Results of the Prospective, Open-Label Phase II NICHE Trial of European Thoracic Oncology Platform (ETOP). J Thorac Oncol. 2019;14(6):1086–94.

    Article  CAS  PubMed  Google Scholar 

  126. Gandhi L, Besse B, Mazieres J, Waqar S, Md A, Barlesi F, et al. MA04.02 Neratinib ± Temsirolimus in HER2-Mutant Lung Cancers: An International, Randomized Phase II Study. J Thorac Oncol. 2017;12:S358-S9.

  127. Kris MG, Camidge DR, Giaccone G, Hida T, Li BT, O’Connell J, et al. Targeting HER2 aberrations as actionable drivers in lung cancers: phase II trial of the pan-HER tyrosine kinase inhibitor dacomitinib in patients with HER2-mutant or amplified tumors. Ann Oncol. 2015;26(7):1421–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Lai WV, Lebas L, Barnes TA, Milia J, Ni A, Gautschi O, et al. Afatinib in patients with metastatic or recurrent HER2-mutant lung cancers: a retrospective international multicentre study. Eur J Cancer (Oxford, England : 1990). 2019;109:28–35.

    Article  CAS  PubMed Central  Google Scholar 

  129. Cornelissen R, Prelaj A, Sun S, et al. Poziotinib in Treatment-Naive NSCLC Harboring HER2 Exon 20 Mutations: ZENITH20-4, A Multicenter, Multicohort, Open-Label, Phase 2 Trial (Cohort 4). J Thorac Oncol. 2023;18(8):1031–41.

    Article  CAS  PubMed  Google Scholar 

  130. Elamin YY, Robichaux JP, Carter BW, Altan M, Gibbons DL, Fossella FV, et al. Poziotinib for Patients With HER2 Exon 20 Mutant Non-Small-Cell Lung Cancer: Results From a Phase II Trial. J Clin Oncol. 2022;40(7):702–9.

    Article  CAS  PubMed  Google Scholar 

  131. Peters S, Stahel R, Bubendorf L, Bonomi P, Villegas A, Kowalski DM, et al. Trastuzumab Emtansine (T-DM1) in Patients with Previously Treated HER2-Overexpressing Metastatic Non-Small Cell Lung Cancer: Efficacy, Safety, and Biomarkers. Clin Cancer Res. 2019;25(1):64–72.

    Article  CAS  PubMed  Google Scholar 

  132. Yang G, Xu H, Yang Y, Zhang S, Xu F, Hao X, et al. Pyrotinib combined with apatinib for targeting metastatic non-small cell lung cancer with HER2 alterations: a prospective, open-label, single-arm phase 2 study (PATHER2). (1741-7015 (Electronic)).

  133. Estrada-Bernal A, Le AT, Doak AE, Tirunagaru VG, Silva S, Bull MR, et al. Tarloxotinib Is a Hypoxia-Activated Pan-HER Kinase Inhibitor Active Against a Broad Range of HER-Family Oncogenes. Clin Cancer Res. 2021;27(5):1463–75.

    Article  CAS  PubMed  Google Scholar 

  134. Liu SV, Villaruz LC, Lee V, Zhu VW, Baik C, Sacher A, et al. LBA61 First analysis of RAIN-701: Study of tarloxotinib in patients with non-small cell lung cancer (NSCLC) EGFR Exon 20 insertion, HER2-activating mutations & other solid tumours with NRG1/ERBB gene fusions. Ann Oncol. 2020;31:S1189.

    Article  Google Scholar 

  135. Riely GJ, Neal JW, Camidge DR, Spira A, Piotrowska Z, Horn L, et al. 1261MO Updated results from a phase I/II study of mobocertinib (TAK-788) in NSCLC with EGFR exon 20 insertions (exon20ins). Ann Oncol. 2020;31:S815–6.

    Article  Google Scholar 

  136. Azar I, Alkassis S, Fukui J, Alsawah F, Fedak K, Al Hallak MN, et al. Spotlight on Trastuzumab Deruxtecan (DS-8201, T-DXd) for HER2 Mutation Positive Non-Small Cell Lung Cancer. Lung Cancer (Auckland, NZ). 2021;12:103–14.

    Google Scholar 

  137. Li BT, Smit EF, Goto Y, Nakagawa K, Udagawa H, Mazières J, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer. N Engl J Med. 2022;386(3):241–51.

    Article  CAS  PubMed  Google Scholar 

  138. Goto K, Sang-We K, Kubo T, Goto Y, Ahn MJ, Planchard D, et al. LBA55 Trastuzumab deruxtecan (T-DXd) in patients (Pts) with HER2-mutant metastatic non-small cell lung cancer (NSCLC): Interim results from the phase 2 DESTINY-Lung02 trial. Ann Oncol. 2022;33.

  139. Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers. Cancer Discov. 2020;10(5):674–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Hussain MH, MacVicar GR, Petrylak DP, Dunn RL, Vaishampayan U, Lara PN Jr, et al. Trastuzumab, paclitaxel, carboplatin, and gemcitabine in advanced human epidermal growth factor receptor-2/neu-positive urothelial carcinoma: results of a multicenter phase II National Cancer Institute trial. J Clin Oncol. 2007;25(16):2218–24.

    Article  CAS  PubMed  Google Scholar 

  141. Michaelson MD, Hu C, Pham HT, Dahl DM, Lee-Wu C, Swanson GP, et al. A Phase 1/2 Trial of a Combination of Paclitaxel and Trastuzumab With Daily Irradiation or Paclitaxel Alone With Daily Irradiation After Transurethral Surgery for Noncystectomy Candidates With Muscle-Invasive Bladder Cancer (Trial NRG Oncology RTOG 0524). Int J Rad Oncol Biol Phys. 2017;97(5):995–1001.

    Article  CAS  Google Scholar 

  142. Wülfing C, Machiels JP, Richel DJ, Grimm MO, Treiber U, De Groot MR, et al. A single-arm, multicenter, open-label phase 2 study of lapatinib as the second-line treatment of patients with locally advanced or metastatic transitional cell carcinoma. Cancer. 2009;115(13):2881–90.

    Article  PubMed  Google Scholar 

  143. Choudhury NJ, Campanile A, Antic T, Yap KL, Fitzpatrick CA, Wade JL 3rd, et al. Afatinib Activity in Platinum-Refractory Metastatic Urothelial Carcinoma in Patients With ERBB Alterations. J Clin Oncol. 2016;34(18):2165–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Du X, Yang B, An Q, Assaraf YG, Cao X, Xia J. Acquired resistance to third-generation EGFR-TKIs and emerging next-generation EGFR inhibitors. Innovation (Cambridge (Mass)). 2021;2(2):100103.

  145. Grivas PD, Day KC, Karatsinides A, Paul A, Shakir N, Owainati I, et al. Evaluation of the antitumor activity of dacomitinib in models of human bladder cancer. Mol Med (Cambridge, Mass). 2013;19(1):367–76.

    Article  Google Scholar 

  146. Sheng X, He Z, Han W, Zhou A-P, Luo H, Shi Y, et al. An open-label, single-arm, multicenter, phase II study of RC48-ADC to evaluate the efficacy and safety of subjects with HER2 overexpressing locally advanced or metastatic urothelial cancer (RC48-C009). Journal of Clinical Oncology. 2021;39(15_suppl):4584-.

  147. Hayashi T, Seiler R, Oo HZ, Jäger W, Moskalev I, Awrey S, et al. Targeting HER2 with T-DM1, an Antibody Cytotoxic Drug Conjugate, is Effective in HER2 Over Expressing Bladder Cancer. J Urol. 2015;194(4):1120–31.

    Article  CAS  PubMed  Google Scholar 

  148. Matt D. Galsky, Gianluca Del Conte, et al. Primary analysis from DS8201-A-U105: A phase 1b, two-part, open-label study of trastuzumab deruxtecan (T-DXd) with nivolumab (nivo) in patients (pts) with HER2-expressing urothelial carcinoma (UC). (Meeting Abstract) 2022 ASCO Genitourinary Cancers Symposium.

  149. Trastuzumab in Treating Patients With Locally Advanced or Metastatic Gallbladder Cancer or Bile Duct Cancer That Cannot Be Removed by Surgery. 2007.

  150. Javle M, Borad MJ, Azad NS, Kurzrock R, Abou-Alfa GK, George B, et al. Pertuzumab and trastuzumab for HER2-positive, metastatic biliary tract cancer (MyPathway): a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2021;22(9):1290–300.

    Article  CAS  PubMed  Google Scholar 

  151. Benson AB, D'Angelica MI, Abbott DE, Anaya DA, Anders R, Are C, et al. Hepatobiliary Cancers, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Comprehensive Cancer Netw. 2021;19(5):541-65.

  152. Meric-Bernstam F, Hanna DL, El-Khoueiry AB, Kang Y-K, Oh D-Y, Chaves JM, et al. Zanidatamab (ZW25) in HER2-positive biliary tract cancers (BTCs): Results from a phase I study. J Clin Oncol. 2021;39(3_suppl):299.

  153. Pant S, Ducreux M, Harding JJ, Javle MM, Oh D-Y, Wasan HS, et al. A phase IIb, open-label, single-arm study of zanidatamab (ZW25) monotherapy in subjects with advanced or metastatic HER2-amplified biliary tract cancers. J Clin Oncol. 2021;39(3_suppl):TPS352-TPS.

  154. Nagano M, Kohsaka S, Ueno T, et al. High-Throughput Functional Evaluation of Variants of Unknown Significance in ERBB2. Clin Cancer Res. 2018;24(20):5112–22.

    Article  CAS  PubMed  Google Scholar 

  155. Cerbone L, Sternberg CN, Sengeløv L, et al. Results from a Phase I Study of Lapatinib with Gemcitabine and Cisplatin in Advanced or Metastatic Bladder Cancer: EORTC Trial 30061. Oncology. 2016;90(1):21–8.

    Article  CAS  PubMed  Google Scholar 

  156. Chang SS. Re: Phase III, Double-Blind, Randomized Trial that Compared Maintenance Lapatinib versus Placebo after First-Line Chemotherapy in Patients with Human Epidermal Growth Factor Receptor 1/2-Positive Metastatic Bladder Cancer. J Urol. 2018;199(4):891–2.

    CAS  PubMed  Google Scholar 

  157. Galsky MD, Von Hoff DD, Neubauer M, et al. Target-specific, histology-independent, randomized discontinuation study of lapatinib in patients with HER2-amplified solid tumors. Invest New Drugs. 2012;30(2):695–701.

    Article  CAS  PubMed  Google Scholar 

  158. Tao Z, Li SX, Shen K, Zhao Y, Zeng H, Ma X. Safety and Efficacy Profile of Neratinib: A Systematic Review and Meta-Analysis of 23 Prospective Clinical Trials. Clin Drug Investig. 2019;39(1):27–43.

    Article  CAS  PubMed  Google Scholar 

  159. Yoshino T, Bartolomeo MD, Raghav KPS, Masuishi T, Kawakami H, Yamaguchi K, et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressing metastatic colorectal cancer (mCRC): Final results from a phase 2, multicenter, open-label study (DESTINY-CRC01). J Clin Oncol. 2022;40(4_suppl):119.

  160. Sheng X, Wang L, He Z, et al. Efficacy and Safety of Disitamab Vedotin in Patients With Human Epidermal Growth Factor Receptor 2-Positive Locally Advanced or Metastatic Urothelial Carcinoma: A Combined Analysis of Two Phase II Clinical Trials. J Clin Oncol. Published online November 21, 2023.

  161. Tarantino P, Carmagnani Pestana R, Corti C, et al. Antibody-drug conjugates: Smart chemotherapy delivery across tumor histologies. CA Cancer J Clin. 2022;72(2):165–82.

    Article  PubMed  Google Scholar 

  162. Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, et al. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Comprehensive Cancer Netw. 2021;19(3):329-59.

  163. Sartore-Bianchi A, Lonardi S, Aglietta M, Martino C, Ciardiello F, Marsoni S, et al. Central Nervous System as Possible Site of Relapse in ERBB2-Positive Metastatic Colorectal Cancer: Long-term Results of Treatment With Trastuzumab and Lapatinib. JAMA Oncol. 2020;6(6):927–9.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Strickler J, Zemla T, Ou FS, Cercek A, Wu C, Sanchez F, et al. 527PDTrastuzumab and tucatinib for the treatment of HER2 amplified metastatic colorectal cancer (mCRC): initial results from the MOUNTAINEER trial. Ann Oncol. 2019;30:v200.

  165. Chang J, Xu M, Wang C, Huang D, Zhang Z, Chen Z, et al. Dual HER2 Targeted Therapy With Pyrotinib and Trastuzumab in Refractory HER2 Positive Metastatic Colorectal Cancer: A Result From HER2-FUSCC-G Study. Clin Colorectal Cancer. 2022;21(4):347–53.

    Article  PubMed  Google Scholar 

  166. Siena S, Di Bartolomeo M, Raghav K, Masuishi T, Loupakis F, Kawakami H, et al. Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2021;22(6):779–89.

    Article  CAS  PubMed  Google Scholar 

  167. YW C, S K, JH H, JK L, NW L, YS L, et al. Overexpression of HER2/HER3 and clinical feature of ovarian cancer. 2019;30(5):e75.

  168. KL J, XV W, V M, SW L, EP M, JA Z, et al. Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q. 2019;30(11):1821-30.

  169. G M, E B, S B, G A, JD B, K D, et al. Superior in vitro and in vivo activity of trastuzumab-emtansine (T-DM1) in comparison to trastuzumab, pertuzumab and their combination in epithelial ovarian carcinoma with high HER2/neu expression. 2017;147(1):145-52.

  170. Menderes G, Bonazzoli E, Bellone S, Black J, Predolini F, Pettinella F, et al. SYD985, a Novel Duocarmycin-Based HER2-Targeting Antibody-Drug Conjugate, Shows Antitumor Activity in Uterine and Ovarian Carcinosarcoma with HER2/Neu Expression. Clin Cancer Res. 2017;23(19):5836–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Meric-Bernstam F, Makker V, Oaknin A, et al. Efficacy and Safety of Trastuzumab Deruxtecan in Patients With HER2-Expressing Solid Tumors: Primary Results From the DESTINY-PanTumor02 Phase II Trial. J Clin Oncol. 2024;42(1):47–58.

    Article  CAS  PubMed  Google Scholar 

  172. Markham A. Margetuximab: First Approval. Drugs. 2021;81(5):599–604.

    Article  CAS  PubMed  Google Scholar 

  173. Scaltriti M, Rojo F, Ocaña A, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst. 2007;99(8):628–38.

    Article  CAS  PubMed  Google Scholar 

  174. Gewinner C, Wang ZC, Richardson A, et al. Evidence that inositol polyphosphate 4-phosphatase type II is a tumor suppressor that inhibits PI3K signaling. Cancer Cell. 2009;16(2):115–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Oh DY, Bang YJ. HER2-targeted therapies - a role beyond breast cancer. Nat Rev Clin Oncol. 2020;17(1):33–48.

    Article  CAS  PubMed  Google Scholar 

  176. Hansel DE, Swain E, Dreicer R, Tubbs RR. HER2 overexpression and amplification in urothelial carcinoma of the bladder is associated with MYC coamplification in a subset of cases. Am J Clin Pathol. 2008;130(2):274–81.

    Article  PubMed  Google Scholar 

  177. Gallardo A, Lerma E, Escuin D, et al. Increased signalling of EGFR and IGF1R, and deregulation of PTEN/PI3K/Akt pathway are related with trastuzumab resistance in HER2 breast carcinomas. Br J Cancer. 2012;106(8):1367–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Musolino A, Naldi N, Bortesi B, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26(11):1789–96.

    Article  CAS  PubMed  Google Scholar 

  179. Vo TH, El-Sherbieny Abdelaal E, Jordan E, et al. miRNAs as biomarkers of therapeutic response to HER2-targeted treatment in breast cancer: a systematic review. Biochem Biophys Rep. 2023;37:101588.

    PubMed  PubMed Central  Google Scholar 

  180. Price-Schiavi SA, et al. Rat Muc4 (sialomucin complex) reduces binding of anti-ErbB2 antibodies to tumor cell surfaces, a potential mechanism for herceptin resistance. Int J Cancer. 2002;99:783–91.

    Article  CAS  PubMed  Google Scholar 

  181. Luque-Cabal M, García-Teijido P, Fernández-Pérez Y, Sánchez-Lorenzo L, Palacio-Vázquez I. Mechanisms Behind the Resistance to Trastuzumab in HER2-Amplified Breast Cancer and Strategies to Overcome It. Clin Med Insights Oncol. 2016;10(Suppl 1):21–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Nagy P, et al. Decreased accessibility and lack of activation of ErbB2 in JIMT-1, a herceptinresistant, MUC4-expressing breast cancer cell line. Cancer Res. 2005;65:473–82.

    Article  CAS  PubMed  Google Scholar 

  183. Paez JG, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–500.

    Article  ADS  CAS  PubMed  Google Scholar 

  184. Gambardella V, Gimeno-Valiente F, Tarazona N, Ciarpaglini CM, Roda D, Fleitas T, et al. NRF2 through RPS6 Activation Is Related to Anti-HER2 Drug Resistance in HER2 -Amplified Gastric Cancer. Clin Cancer Res. 2019;25(5):1639–49.

    Article  CAS  PubMed  Google Scholar 

  185. Iwata TN, Ishii C, Ishida S, Ogitani Y, Wada T, Agatsuma T. A HER2-Targeting Antibody-Drug Conjugate, Trastuzumab Deruxtecan (DS-8201a), Enhances Antitumor Immunity in a Mouse Model. Mol Cancer Ther. 2018;17(7):1494–503.

    Article  CAS  PubMed  Google Scholar 

  186. Mucciolo G, Araos Henríquez J, Jihad M, et al. EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell. Published online December 19, 2023.

  187. Hanna WM, Rüschoff J, Bilous M, Coudry RA, Dowsett M, Osamura RY, et al. HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Modern Pathol. 2014;27(1):4-18.

  188. Godoy-Ortiz A, Alba-Bernal A, Pascual J, Comino-Méndez I, Alba E. Unveiling the Potential of Liquid Biopsy in HER2-Positive Breast Cancer Management. Cancers (Basel). 2022;14(3):587.

    Article  CAS  PubMed  Google Scholar 

  189. Bao Y, Oguz G, Lee WC, Lee PL, Ghosh K, Li J, et al. EZH2-mediated PP2A inactivation confers resistance to HER2-targeted breast cancer therapy. Nat Commun. 2020;11(1):5878.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  190. Sanchez-Vega F, Hechtman JF, Castel P, Ku GY, Tuvy Y, Won H, et al. EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2 -Amplified Esophagogastric Cancer. Cancer Discov. 2019;9(2):199–209.

    Article  CAS  PubMed  Google Scholar 

  191. Lee C-k, Rha SY, Kim HS, Jung M, Kang B, Che J, et al. A single arm phase Ib/II trial of first-line pembrolizumab, trastuzumab and chemotherapy for advanced HER2-positive gastric cancer. Nat Commun. 2022;13(1):6002.

  192. Wang D-S, Liu Z-X, Lu Y-X, Bao H, Wu X, Zeng Z-L, et al. Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer. Gut. 2019;68(7):1152–61.

    Article  CAS  PubMed  Google Scholar 

  193. Makiyama A, Sukawa Y, Kashiwada T, Kawada J, Hosokawa A, Horie Y, et al. Randomized, Phase II Study of Trastuzumab Beyond Progression in Patients With HER2-Positive Advanced Gastric or Gastroesophageal Junction Cancer: WJOG7112G (T-ACT Study). J Clin Oncol.10.

Download references

Acknowledgements

Not applicable.

Funding

There is no funding for this review.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: K.Z. and H.Z.; Supervision: K.Z., H.Z. and T.L.; Visualization: K.Z., X.Y. and H.T; Editing and revision: K.Z. and H.Z. and X.Z.; Illustration and charting (Fig. 1 and Tables 1 and 2): K.Z., X.Y. and H.T. Manuscript writing: All authors. Manuscript writing: All authors. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Ting Luo or Hong Zheng.

Ethics declarations

Ethics approval and consent to participate

Not applicable. This review does not report on or involve the use of any animal or human data or tissue

Consent for publication

Not applicable. This review does not contain data from any individual person.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, K., Yang, X., Tai, H. et al. HER2-targeted therapies in cancer: a systematic review. Biomark Res 12, 16 (2024). https://doi.org/10.1186/s40364-024-00565-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-024-00565-1

Keywords