Abstract
HER2 activating mutations act as oncogenic drivers in various cancer types. In the clinic, they can be identified by next generation sequencing (NGS) in either tumor biopsies or circulating cell-free DNA (cfDNA). Preclinical data indicate that HER2 “hot spot” mutations are constitutively active, have transforming capacity in vitro and in vivo and show variable sensitivity to anti-HER2 based therapies. Recent clinical trials also revealed activity of HER2-targeted drugs against a variety of tumors harboring HER2 mutations. Here, we review the prevalence and type of HER2 mutations identified in different human cancers, their biochemical and biological characterization, and their sensitivity to anti HER2-based therapies in both preclinical and clinical settings.
Keywords: HER2 mutations, next-generation sequencing (NGS), Afatinib, Neratinib, TCGA
1. Introduction
Over the past decade, genomic profiling has enabled the identification of numerous actionable oncogenic drivers that are causative for cancer initiation and progression in multiple tumor types (Kandoth, et al., 2013) (Cancer Genome Atlas Research, et al., 2013) (Bailey, Tokheim, Porta-Pardo, Sengupta, Bertrand, Weerasinghe, Colaprico, Wendl, Kim, Reardon, Ng, et al., 2018). The same oncogenic drivers can promote the development and the growth of tumors with different histologies (Bailey, Tokheim, Porta-Pardo, Sengupta, Bertrand, Weerasinghe, Colaprico, Wendl, Kim, Reardon, Kwok-Shing Ng, et al., 2018). This observation has provided the rationale for the design of “basket trials”, in which the efficacy of a specific targeted therapy is tested against tumors that harbor the qualifying genomic aberrations, independent of tumor histology. Basket trials have been proven to be a particularly suitable approach when the targeted genomic alterations are rare driver events and occur across different cancer types (Hyman, et al., 2015) (Hainsworth, et al., 2018). In this setting, even if the pharmacological target is present in small percentages of patients, the studies allow the rapid enrolment of a sufficient number of patients to evaluate the efficacy of the tested drug. Responses to therapy can be either tissue agnostic or histology-dependent. As an example, of the former, objective clinical responses were obtained in up to 80% of patients with adult and pediatric tumors of different origin harboring oncogenic fusions involving the neurotrophin receptors tyrosine kinase genes NTRK1, NTRK2 or NTRK3 (encoding TRKA, TRKB and TRKC, respectively) treated with the TRK inhibitor Larotrectinib (Drilon, et al., 2018). By contrast, the efficacy of the irreversible pan-ERBB inhibitor Neratinib against tumors harboring activating mutations in erbB2, the gene encoding for the human epidermal growth factor receptor 2 (HER2), has been shown to be largely dependent on tumor histology, with clinical responses achieved in breast, cervical and biliary cancer and intrinsic refractoriness in all the other tumor types tested including colorectal, bladder and ovarian cancer (Hyman, et al., 2018).
2. HER2 proto-oncogene
The erbB2 gene, located on the short arm of chromosome 17, was discovered and cloned in 1983 (Figure 1) (Sato, et al., 1983). The product of this gene, HER2, is a transmembrane glycoprotein of 185 KDa that consists of three main domains: an extracellular, a transmembrane and an intracellular domain with tyrosine kinase activity (Cho, et al., 2003). The epidermal growth factor receptor family of tyrosine kinases consists of 4 receptor tyrosine kinases (EGFR/erbB1/HER1, erbB2/HER2, erbB3/HER3, and erbB4/HER4) (Cho et al., 2003) that are activated by ligand-induced dimerization (Ushiro & Cohen, 1980). HER2, despite being the only receptor of the family with no known ligand, is the preferred partner for heterodimerization with other erbB members (Graus-Porta, Beerli, Daly, & Hynes, 1997). Receptors dimerization induces transphosphorylation of tyrosine residues within the kinase domains and consequent activation of downstream signaling cascades (Zhang, Gureasko, Shen, Cole, & Kuriyan, 2006) (Wagner, Stacey, Liu, & Pawson, 2013).
Figure 1. Timeline of key advances relating to the biology and therapeutic targeting of HER2 signaling.
Milestone discoveries that are relevant to HER2 amplified/overexpressing (boxes above the timeline arrow) and HER2 mutant tumors (boxes below the timeline arrow). Key events relating to the following fields of study are color coded as follows: FDA approval of anti-HER2 drugs (green); identification of HER2 mutation in cancers (yellow); clinical trial enrolling HER2 mutated cancers (blue).
The identification of HER2 amplification/overexpression in a subset of breast cancer and the consequent development of specific anti-HER2 therapies had a tremendous impact on the clinical management of breast cancer patients. In 1998, the US Food and Drug Administration (FDA) approved Trastuzumab, a monoclonal antibody targeting the extracellular domain of HER2, in combination with paclitaxel as first-line treatment in HER2-positive metastatic breast cancer, and as a single agent for second and third-line therapy (Figure 1). Trastuzumab approval resulted in the inclusion of HER2 testing in the guidelines of the American Society of Clinical Oncology (ASCO) as a recommended test for all breast cancer one year later (Smith, et al., 1999). Several studies have shown improvements in progression-free survival (PFS) and overall survival (OS) in HER2-positive breast cancer patients treated with Trastuzumab-based therapies (Vogel, et al., 2002) (Marty, et al., 2005). Many other anti HER2 drugs have been further developed and nine are now FDA-approved for breast cancer treatment: Pertuzumab (a recombinant humanized monoclonal antibody that targets the extracellular dimerization domain of the HER2), Adotrastuzumab Emtansine (TDM-1, a HER2-targeting antibody drug conjugate (ADC)), Neratinib, Lapatinib (a reversible pan-ERBB inhibitor) and five Trastuzumab biosimilars (Figure 1) (Amiri-Kordestani, et al., 2014) (Gradishar, 2012; Martin, et al., 2017) (Blumenthal, et al., 2013). (Lee, et al., 2018) (Voelker, 2018). More recently, HER2 amplification/overexpression has also been identified in other tumor types such as gastric, where trastuzumab-based therapy is also standard of care, colon, salivary gland, bladder and uterine serous carcinoma (Grabsch, Sivakumar, Gray, Gabbert, & Muller, 2010) (Buza, English, Santin, & Hui, 2013).
2.1. Prevalence of HER2 mutations in human cancers
TCGA and other available databases show that HER2 mutations are present in a variety of human cancer types (Figure 2A). Bladder cancer represents the histology with the highest prevalence of HER2 mutations (9–18%), followed by uterine cervix (6%), colorectal (5.8%), lung (4%) and breast (4%) (Robertson, et al., 2017) (Cancer Genome Atlas Research, 2014a) (Ojesina, et al., 2014) (Giannakis, et al., 2016) (Stephens, et al., 2004) (Razavi, et al., 2018). More recently, Pahuja et al. analyzed 111,176 tumors and found that about 3.5% of them harbored HER2 mutations (Pahuja, et al., 2018). While the vast majority of these mutations mapped to the ECD and to the kinase domain (KD) of the receptor, about 3% and 7% were found in the transmembrane domain (TM) (G660D, R678Q, E693K) or in the juxtamembrane domain (JM) (Q709L), respectively. These newly identified mutations were mainly found in colorectal, breast and lung cancer and were confirmed to be oncogenic and sensitive to anti-HER2 therapies as part of this study (Pahuja, et al., 2018) (Figure 3).
Figure 2. Distribution and frequency of HER2 mutations in solid tumors.
A: HER2 mutations identified across multiple cancer histologies. Only published studies on solid tumors with more than 100 samples were included. The frequency of HER2 mutations varies from 1% in cancer types such as melanoma and up to 12% in bladder cancer. Similar histologies are depicted with the same colors. B: Lollipop diagram depicting activating HER2 mutations identified in several cancer types. The most prevalent mutation, S310F/Y, is localized in the ECD and has been frequently reported in bladder, uterine cervix, breast and stomach cancer. HER2 mutations that mapped in the KD are also depicted. These are frequently found in lung, colorectal, breast and uterine tumors. Mutations in the TM and the JM domains recently identified in lung and colorectal cancer are also indicated. The most common mutated residues in the ECD, the JM and the KD modelled using PyMOL molecular visualization software are presented.
Figure 3. Sensitivity of HER2 mutants to anti-HER2 therapy.
A heat map of the half maximal inhibitory concentration (IC50) values for different pan-ERBB inhibitors is shown. These drugs have different levels of activity against mutant HER2 proteins in vitro (box colors indicate the range of sensitivity/resistance for each mutant). The symbols in each box indicate the clinical results obtained from case reports or clinical trials. (C: clinical response; R: clinical resistance). Of note, some mutants that have been described to be sensitive to specific inhibitors in preclinical analyses were instead found to be resistance to the same drugs in patients.
2.2. Lung cancers
Lung cancer was the first tumor type in which HER2 mutations were described. Stephens et al. sequenced the ERBB2 gene from 120 primary lung tumors and found that 4% of these cases harbored mutations within the kinase domain. The percentage raised up to 10% in the adenocarcinoma subtype (Stephens, et al., 2004). Similar results were obtained by TCGA in a cohort of 178 lung squamous cell carcinomas (4% of cases) (Cancer Genome Atlas Research, 2012) and in 230 resected lung adenocarcinomas, where 3% of the sample carried HER2 mutations) (Cancer Genome Atlas Research, 2014b). The most prevalent alteration in this cancer types is the in-frame insertion/duplication G776YVMA within the exon 20 (Figure 2B).
2.2.1. Preclinical data
Wang et al. showed that the G766YVMA insertion was more potent than wild-type HER2 in activating signal transducers, phosphorylating EGFR, and promoting survival, invasiveness and tumorigenicity in lung cancer models (Wang, et al., 2006). Additional work by Perera et. al. found that Trastuzumab treatment in combination with Rapamycin reduced tumor growth in transgenic mice expressing the HER2 exon 20 YVMA insertion in the lung epithelium (Perera, et al., 2009). Later, Greulich et al. reported that NIH 3T3 cells overexpressing wild-type HER2 exhibited a weak anchorage-independent phenotype while the G309E, S310F, and S310Y mutants (that map to the ECD and are exclusive of the adenocarcinoma subtype) resulted in robust colony formation in soft agar (Greulich, et al., 2012) (Figure 2B). They further showed that the amino acid change from serine to phenylalanine in the 310 codon (S310F) induced hyperactivation of the kinase. These HER2 ECD mutants were activated by either elevated C-terminal tail phosphorylation or by covalent dimerization mediated by intermolecular disulfide bond formation. These distinct mechanisms of receptor activation converged to tyrosine phosphorylation of cellular effectors such as SHP-2, a phosphatase that has been shown to induce the activation of the Ras-Erk signaling, thus promoting growth and metastasis of multiple cancer types (Aceto, et al., 2012; Bentires-Alj, et al., 2004; Hellmuth, et al., 2008; Schneeberger, et al., 2014). The same authors showed that Ba/F3 cells transformed with HER2 ECD mutants were sensitive to Neratinib and Afatinib (another irreversible pan-ERBB inhibitor) with IC50s in the low nanomolar range (Figure 3), resulting in effective abrogation of cell survival. Serra et al. reported a case of a 29 years old patient with a germline TP53 mutation (Li-Fraumeni Syndrome) who developed two primary tumors in breast and lung, respectively. Adjuvant treatment with Tamoxifen completely resolved the breast tumor and the lung cancer was treated with adjuvant chemotherapy that kept the disease under control for about 4 years. Upon progression, NGS on a newly identified lung nodule found a novel HER2 mutation in the TM domain (V659E) (Serra, et al., 2013). The patient was treated with Lapatinib and Paclitaxel and achieved an objective clinical response that lasted for 9 months. Additional studies by Liu et al. showed that Ba/F3 cells stably expressing the A775_G776insYVMA insertion were sensitive to Osimertinib, a third-generation tyrosine kinase inhibitor (TKI). Osimertinib was more potent than Erlotinib in reducing pHER2 levels and inhibiting cells growth. Interestingly, they observed decreased levels of MYC after Osimertinib exposure and found that the combination of Osimertinib and JQ1 (a potent inhibitor of the BET family of bromodomain proteins which include BRD2, BRD3, BRD4) was highly effective in reducing HER2, AKT and ERK phosphorylation in these models (Liu, et al., 2018). Together, their results provide the rationale to test the efficacy of Osimertinib as single agent or in combination with BRD4/MYC inhibitors in patients harboring HER2-driven (mutated or amplified) lung cancers.
2.2.2. Clinical responses in HER2 mutant lung cancer patients to anti HER2-based therapy
De Greve et al. reported the first evidences of clinical benefit from Afatinib treatment in HER2 mutant lung cancer patients in 2011 (De Greve, et al., 2012). Three patients with advanced lung adenocarcinoma bearing HER2 mutations were treated with Afatinib single agent or in combination with Paclitaxel and all achieved significant tumor regression. Chuang et al. later reported the outcomes of nine patients with metastatic lung adenocarcinoma harboring HER2 mutations treated with HER2-targeted therapies. Four of the nine patients experienced clinical benefits with responses that lasted up to 10 months. All four responders harbored an exon 20 insertion and they were treated with Trastuzumab, Afatinib or TDM-1 (Chuang, et al., 2017). In a recent case report, a patient with the recurrent YVMA insertion treated with Afatinib first-line achieved a PR that lasted 12 months (Shi & Wang, 2018). Another group reported the case of a woman with NSCLC harboring a HER2 exon 20 mutation (c.2437A>G) who previously failed cytotoxic chemotherapy and Gefitinib treatment but responded to Afatinib (Park, et al., 2018). In 2018, Hyman et al. reported the data of a global multicenter, multi-histology basket trial to test Neratinib in HER2 and HER3 mutated cancers of any tumor type (Hyman, Piha-Paul, Rodon, et al., 2017). Twenty-six HER2 mutated lung cancers, mostly harboring the exon 20 insertion, were enrolled and only one objective response was observed. The median PFS was 5.5 months and the objective response rate was 3.8% (clinical benefit rate (CBR) was 42.3%). Importantly, HER2 exon 20 insertions are paralogous of EGFR exon 20 insertions, which are resistant to first- and second-generation EGFR tyrosine kinase inhibitors (Yasuda, et al., 2013). More recently, in a phase II trial, Li et al. assessed the activity of TDM-1 in a cohort of heavily pretreated patients with advanced HER2-mutant lung cancers (Li, et al., 2018). Eighteen patients were enrolled and responses were seen across patients with HER2 exon 20 insertions and point mutations in the kinase, transmembrane, and extracellular domains. Interestingly, mass spectrometry confirmed low or undetectable levels of HER2 protein expression among HER2 mutant responders, suggesting that HER2 mutations may promote the internalization of TDM-1 regardless of the receptor levels on the cell membrane. A plausible explanation for this phenomenon is that the constitutively active mutant receptors are more efficient than the wild type in forming homo/hetero dimers, which would result in an increased internalization turnover (Li, et al., 2018). Finally, it was reported a case of a lung cancer patient who was initially treated with Carboplatin and Pemetrexed followed by Erlotinib. At progression, genomic testing revealed the presence of a germline HER2 G660D mutation and the patient experienced a quick reduction of tumor volume after 12 weeks of Afatinib (Pahuja, et al., 2018).
2.3. Breast cancer
HER2 somatic mutations are present in about 4% of breast cancer patients (Banerji, et al., 2012; Cancer Genome Atlas Network, 2012; Ellis, et al., 2012) (Figure 2 panel A). In invasive lobular breast cancer, which composes about 15% of the ER-positive subtype, the prevalence of HER2 mutations is higher (about 10% including the three largest studies available on cBioPortal).
Approximately 70% of HER2 mutations occur in the kinase domain, between amino acids 755 and 781 (exons 19 and 20) (Figure 2 panel B). Another 20% are found in the extracellular domain (ECD) at either amino acid 309 or 310 (exon 8) (Figure 2B).
2.3.1. Preclinical data
In 2006, Wang et al. showed that the HER2 exon 20 YVMA insertion mutation was also a “driver” mutation in breast tumors (Wang, et al., 2006). They found that, despite this alteration mediated resistance to EGFR TKIs, both Trastuzumab and Lapatinib remained active against MCF10A cells transduced with this mutant. Bose et al. later investigated the impact of 13 additional HER2 mutations using the same model. They found that three mutations (R678Q, I767M, and Y835F) exerted no functional effects, while the others, including HER2 G309A, L755S, D769H/Y, V777L, P780_781insGSP, V842I, and R896Q, resulted in increased HER2 signaling and increased tumor growth when compared to HER2 wild type transduced controls. The HER2 L755S mutation, the most frequent in breast cancer, induced a weak oncogenic phenotype. Computational modeling suggests that Lapatinib cannot bind to the L755S substitution due to steric hindrance. It is worth noting that Neratinib was effective against all the HER2 mutants (Bose, et al., 2013; Trowe, et al., 2008) (Figure 3).
Trastuzumab activity was also investigated against HER2 mutants. Trastuzumab treatment was able to reduce the formation of colonies in Matrigel-based assays but did not show a cytotoxic effect (Bose, et al., 2013). More recently, a study conducted on both preclinical models and patients by our group showed that the presence of HER2 mutations can limit the sensitivity to Trastuzumab and Lapatinib in HER2-amplified breast cancer patients. However, in this context, tumors with concomitant mutation of HER2 and amplification of the ERBB2 gene remain sensitive to Neratinib monotherapy (Cocco, et al., 2018). Together, these data suggest that HER2 mutated breast cancers are sensitive to Neratinib regardless the mutant allele and the copy number status of the ERBB2 gene.
Interestingly, a recent study reported that the vast majority of HER2 mutations identified in breast cancer (about 70%) is found in metastatic ER+ tumors, suggesting that the emergence of HER2 mutations may represent a mechanism of acquired resistance to endocrine therapy (Croessmann, et al., 2019). Therefore, the use of anti-HER2 agents may partially restore the sensitivity of these tumors to endocrine therapy. In support of this hypothesis, HER2 mutations (L755S and V777L) were found to over-activate the PIK/AKT/mTOR pathway inducing endocrine resistance, while addition of Neratinib or Alpelisib or Everolimus restored the sensitivity to Fulvestrant in HER2 mutant breast cancer cells. Interestingly, triple therapy with Neratinib, Everolimus and Fulvestrant induced a more potent tumor growth inhibition achieving a complete tumor regression in a preclinical model of breast cancer xenograft harboring the V777L HER2 mutation (Croessmann, et al., 2019). Although the authors did not investigate the molecular mechanisms that link HER2 inhibition and ER activity in these models, a previous study showed that PI3K inhibition in PIK3CA mutated ER+ breast cancer cells induced a robust ER-dependent transcriptional program mediated by the activation of KMT2D, a histone H3 lysine 4 methyltransferase (Toska, et al., 2017). Perhaps a similar phenomenon is taking place in this setting, partially explain why HER2 inhibition combined with Fulvestrant is effective in ER+ HER2 mutated cancer. Further studies are needed to elucidate this mechanism and evaluate which agent (Neratinib, TDM-1 or other potent ADCs conjugated to a derivative of the camptothecin analog exatecan targeting HER2 such as DS-8201a) in combination with Fulvestrant may be more effective against HER2 mutated, ER+ breast tumors.
2.3.2. Clinical responses of HER2 mutant breast tumors to anti HER2-based therapies
The first case report of a HER2 mutated breast cancer was published in 2014 (Ali, et al., 2014). A 58 years old woman was diagnosed with triple negative, inflammatory breast cancer and, after progressing to multiple standard chemotherapies, two HER2 mutations (V777L and S310F) were detected by NGS in the absence of ERBB2 gene amplification. The patient experienced quick symptoms improvements upon Lapatinib and Trastuzumab-based therapies that lasted 6 months (Ali, et al., 2014). A second case reported a 51 years old woman with a ER+, HER2 negative breast cancer who developed metastatic disease refractory to many chemotherapy regimens (Chumsri, et al., 2015). NGS on a progressing lesion identified a HER2 S310F mutation and the patient was treated with the combination of Trastuzumab, Pertuzumab, and Fulvestrant. Her liver metastases and malignant ascites markedly improved and she remained on this therapy for 12 months (Chumsri, et al., 2015). An additional case reported a 43 years old patient with ER+, HER2 negative, metastatic breast cancer with a HER2 L755S mutation. She was treated with Neratinib monotherapy and experienced dramatic improvement in symptoms and tumor markers that lasted 11 months (Ben-Baruch, Bose, Kavuri, Ma, & Ellis, 2015). At progression, the addition of Capecitabine to Neratinib resulted in a second response that lasted more than 6 months (Ben-Baruch, et al., 2015). Similarly, another report described a HER2 (D769H) mutant patient with metastatic HER2 negative breast cancer who achieved a partial response upon Trastuzumab, Pertuzumab, and chemotherapy (Shih, et al., 2015).
In the phase II MutHER trial, they evaluated the activity of Neratinib in HER2 mutant non-amplified metastatic breast cancer. Clinical endpoints included CBR, PFS, toxicity, and HER2 mutations detection in circulating tumor DNA (ctDNA) (Ma, et al., 2017). CBR was observed in 31% of patients. This included one CR, one PR, and three SD > 24 weeks. Median PFS was 16 weeks and the on-target effect of Neratinib was supported by early decreases of ctDNA HER2mut variant allele frequency, which then increased at progression. In the SUMMIT trial conducted by Hyman and colleagues, breast cancer patients were the ones who experienced the greatest antitumor activity with a median PFS of 3.5 months and an overall response of 32% (Hyman, Piha-Paul, Rodon, et al., 2017). Given that the vast majority of HER2-mutant tumors are also ER+, and that dual inhibition of HER2 and ER+ has been shown to be more effective than each of the single agent in inhibiting tumor growth in preclinical models (Croessmann, et al., 2019), the SUMMIT trial has subsequently tested the combination of Neratinib plus Fulvestrant in HER2 mutated, ER+ metastatic breast cancer patients. Initial results on the first 12 patients treated with the combination showed a CBR of 58%, OOR of 42%, and median PFS of 3.7 months (Hyman, Piha-Paul, Saura, et al., 2017). These findings are in line with the preliminary results obtained by the SOLAR-1 trial that is currently evaluating the efficacy and safety of Alpelisib (α-specific PI3K inhibitor) plus Fulvestrant in patients with HR+, HER2– negative advanced breast cancer (Fabrice, 2018) and confirm the preclinical results obtained by our group in this setting (Toska, et al., 2017). Specifically, the addition of Fulvestrant to Neratinib or to Alpelisib may prevent the increase of ER activity thus inhibiting tumor growth (Figure 4). Given these premises, it would be of interest to evaluate the feasibility of a triple combination with Neratinib, Alpelisib and Fulvestrant in patients harboring ER+, PIK3CA mutated or HER2 mutated breast cancer in the near future.
Figure 4. Model of HER2 dependent ER regulation.
Proposed model of regulation of ER activity by HER2 signaling in HER2 mutant ER+ breast tumors. In the absence of HER2 inhibition, active HER2 signaling results in suppression of ER dependent transcription. Inhibition of HER2 (with Neratinib or antibody-drug-conjugates such as TDM-1 or DS-8201) promotes increased ER activity as a result of an adaptive mechanism of cellular defense. The activation of ER-dependent transcription may be prevented through dual HER2/ER inhibition (Neratinib plus Fulvestrant or TDM-1/DS-8201 plus Fulvestrant).
2.4. Bladder cancers, gastrointestinal and cervical cancer
Bladder cancer represents the tumor type with the highest prevalence of HER2 mutations ranging from 9 to 12 % (Figure 2 panel A). The S310F hotspot in the ECD is the most common substitution in this tumor type, and the same mutation is also the most prevalent in stomach/esophageal and cervical cancer, malignancies in which HER2 mutations are found in about 5% and 6% of cases, respectively (Ojesina, et al., 2014) (Cancer Genome Atlas Research, et al., 2017) (Figure 2 panel B). TCGA data showed that about 4% of colorectal cancer patients harbor HER2 somatic mutations, with the substitution V842I (Cancer Genome Atlas, 2012).
2.4.1. Preclinical data
De Martino et al. showed that bladder cancer cells with the mutations S310F, S653C, R678Q, or the double D277H/S310F mutation had enhanced phosphorylation of HER2 and were sensitive to Lapatinib (de Martino, et al., 2014). In another study, a HER2 mutated (S310F) bladder cancer cell line was also found to be sensitive to Afatinib and Dacomitinib (an irreversible pan-ERBB inhibitor) (Tamura, et al., 2018). In colorectal cancer, Kavouri et al. showed that the HER2 mutations V842I, V777L, and L755S, as well as the ECD mutation S310F, were oncogenic in this malignancy (Kavuri, et al., 2015). They also demonstrated that, although the V842I substitution induced resistance to Cetuximab, Panitumumab and Trastuzumab, cells expressing this mutant retained sensitivity to Neratinib and Afatinib. A more recent work evaluated the efficacy of pan-HER2 inhibitors in Lynch and Lynch-like HER2 mutated colorectal cancer cell lines (Kloth, et al., 2016). The authors found that both the L755S and the V842I substitutions were recurrent events in this setting and sensitive to Afatinib, Neratinib and Dacomitinib. While Lapatinib and Trastuzumab were active against the V842I, both agents failed to target the L755S, thus confirming the results previously obtained in breast cancer (Bose, et al., 2013).
2.4.2. Sensitivity to anti HER2-based therapies
Among the 16 bladder cancer patients enrolled in the SUMMIT trial, no responses were observed (median PFS of 1.8 months) (Hyman, et al., 2018). Genomic characterization showed the presence of a higher number of TP53 and PIK3CA mutations, as well as Cyclin E amplification when compared to tumors that experienced clinical benefits in the same cohort, thus suggesting that these genomic alterations may have contributed to the intrinsic resistance of these tumors to Neratinib therapy. Interestingly, TCGA data showed that about 52% and 22% of HER2 mutated bladder cancers harbored a TP53 or a PIK3CA mutation, respectively, and up to 10% had Cyclin E amplification (Robertson, et al., 2017). The SUMMIT trial also included 12 colorectal cancer and 5 gastro-esophageal cancer patients with no responses observed (PFS of 1.8 and 1.7 months, respectively) (Hyman, et al., 2018). Better outcomes were observed in cervical cancer and biliary tract cancer patients, with objective responses of 20% and 22.2% and a PFS of 20.1 and 2.8 months, respectively (Hyman, et al., 2018).
3. Ongoing trials
Sixteen clinical trials investigating the efficacy of HER2 targeted therapies in HER2 mutant cancers are currently ongoing (Table 1). Two phase II studies are investigating the efficacy of Neratinib plus Trastuzumab or plus Cetuximab ( NCT03457896) and the activity of the ADC DS-8201a ( NCT03505710) in non-small lung cancers. Multiple trials investigating the efficacy of Afatinib in HER2 mutant tumors of various histologies are currently enrolling ( NCT03810872, NCT02183883, NCT02780687, NCT02597946). In addition, the NCI MATCH trial subprotocol B ( NCT02465060) is testing Afatinib in HER2 mutated cancers of any type and is currently enrolling patients nationwide. More trials are investigating the role of Neratinib in combination with other agents such as Fulvestrant ( NCT01670877, NCT01953926) in breast cancer or Everolimus, Palbociclib or Trametinib in patients with advanced solid tumors harboring EGFR or HER2 mutation/amplification or HER3/4 mutations ( NCT03065387). Additionally, new TKIs are now under clinical investigation: Pyrotinib (an oral irreversible pan-ERBB receptor TKI with activity against HER1, HER2 and HER4) in non-small cell lung cancer and in breast cancer ( NCT02834936 and NCT03412383) and Poziotinib (a pan-ERBB inhibitor) in metastatic breast cancer and in patients with stage IV lung adenocarcinoma ( NCT02544997, NCT02979821) harboring HER2 mutations.
Table 1.
Active clinical trials enrolling HER2 mutated cancers
| NCT # | Title | Status | Conditions | Interventions | Phases |
|---|---|---|---|---|---|
| NCT02834936 | A Clinical Study of Pyrotinib in Patients of Advanced Non-Small Cell Lung Cancer With HER2 Mutation | Recruiting | NSCLC | Pyrotinib | Phase 2 |
| NCT03065387 | Study of the Pan-ERBB Inhibitor Neratinib Given in Combination With Everolimus, Palbociclib or Trametinib in Advanced Cancer Subjects With EGFR Mutation/Amplification, HER2 Mutation/Amplification, HER3/4 Mutation or KRAS Mutation |
Recruiting | Solid Tumors | Neratinib Everolimus Palbociclib Trametinib | Phase 1 |
| NCT01670877 | Neratinib +/− Fulvestrant in Metastatic HER2 Nonamplified But HER2 Mutant Breast Cancer | Recruiting | Breast Neoplasms | Neratinib Fulvestrant Trastuzumab | Phase 2 |
| NCT03810872 | An Explorative Study of Afatinib in the Treatment of Advanced Cancer Carrying an EGFR, a HER2 or a HER3 Mutation | Recruiting | Cancers Harbouring an EGFR Mutation, a HER2 Mutation or a HER3 Mutation | Afatinib Paclitaxel | Phase 2 |
| NCT02716116 | A Trial of TAK-788 (AP32788) in Non-small Cell Lung Cancer (NSCLC) | Recruiting | NSCLC | TAK-788 | Phase1| Phase 2 |
| NCT02183883 | Deciphering Afatinib Response and Resistance With INtratumour Heterogeneity | Recruiting | NSCLC | Afatinib | Phase 2 |
| NCT02544997 | A Phase II, Single-Arm Trial of Poziotinib as Salvage Treatment in Patients With Metastatic Breast Cancer Who Has HER2 or EGFR Mutation or Activated AR or EGFR Pathway | Recruiting | Metastatic Breast Cancer | Poziotinib | Phase 2 |
| NCT03410927 | A Study of TAS0728 in Patients With Solid Tumors With HER2 or HER3 Abnormalities | Recruiting | Advanced Solid Tumors | TAS0728 | Phase 1|Phase 2 |
| NCT03505710 | DS-8201a in Human Epidermal Growth Factor Receptor 2 (HER2)-Expressing or -Mutated Non-Small Cell Lung Cancer | Recruiting | NSCLC | Trastuzumab deruxtecan (DS-8201a) | Phase 2 |
| NCT03318939 | Phase 2 Study of Poziotinib in Patients With NSCLC Having EGFR or HER2 Exon 20 Insertion Mutation | Recruiting | NSCLC | Poziotinib | Phase 2 |
| NCT01953926 | Neratinib HER Mutation Basket Study (SUMMIT) | Recruiting | Malignant Solid Tumor|Fibrolamellar Carcinoma | Neratinib Paclitaxel Fulvestrant Trastuzumab | Phase 2 |
| NCT03066206 | Poziotinib in EGFR Exon 20 Mutant Advanced Non-Small Cell Lung Cancer (NSCLC) and HER2 Exon 20 Mutant NSCLC | Recruiting | Malignant Neoplasm of Respiratory and Intrathoracic Organ|NSCLC | Poziotinib | Phase 2 |
| NCT03457896 | Study of Neratinib +Trastuzumab or Neratinib + Cetuximab in Patients With KRAS/NRAS/BRAF/PIK3CA Wild-Type Metastatic Colorectal Cancer by HER2 Status | Recruiting | Metastatic Colorectal Cancer | Trastuzumab Cetuximab Neratinib | Phase 2 |
| NCT03412383 | Pyrotinib in Metastatic HER2 Non-amplified But HER2 Mutant Breast Cancer | Recruiting | Breast Cancer | Pyrotinib | Phase 2 |
| NCT02780687 | Afatinib Monotherapy in Patients With ERBB-deregulated Metastatic Urothelial Tract Carcinoma After Failure of Platinum Based Chemotherapy | Recruiting | Urologic Neoplasms | Afatinib | Phase 2 |
| NCT02465060 | Targeted Therapy Directed by Genetic Testing in Treating Patients With Advanced Refractory Solid Tumors, Lymphomas, or Multiple Myeloma (The MATCH Screening Trial) | Recruiting | Solid Tumors | Adavosertib Afatinib Binimetinib Capivasertib Crizotinib Dabrafenib Dasatinib Defactinib Larotrectinib Nivolumab Osimertinib Palbociclib Pertuzumab GSK2636771 Sapanisertib Sunitinib Malate Taselisib Trametinib Trastuzumab TDM-1 Vismodegib | Phase 2 |
4. Resistance to targeted therapy
The idea of treating HER2 mutant tumors with specific therapies is relatively new and, as a consequence, little is known about the potential mechanisms of resistance to anti-HER2 agents in this setting. Primary resistance may be mediated by the presence of on-target mutations that hamper the binding of first generation TKIs such as Lapatinib. Studies have shown that three point mutations (L755S, L755P and T798M) confer primary as well as acquired resistance to this agent (Kancha, et al., 2011) (Bose, et al., 2013). Acquired resistance can also be mediated by the emergence of de novo mutations in HER2. Hanker et al reported a case in which the emergence of the gatekeeper HER2 T798I was identified in the cfDNA of a HER2 L869R-mutant breast cancer patient who initially responded and then progressed to Neratinib. Structural modeling of this acquired mutation suggested that the increased bulk of isoleucine in HER2 T798I reduces Neratinib binding by generating steric hindrance. Interestingly, while this mutation induced Neratinib resistance, Afatinib and the Osimertinib metabolite AZ5104 were still able to suppress cell growth in transduced preclinical models (Hanker, et al., 2017).
In addition to on-target mechanisms, it has been described that TP53, PIK3CA and K-RAS mutations may contribute to intrinsic resistance to anti-HER2 therapy in HER2 overexpressing or EGFR mutated tumors (Canale, et al., 2017; Ma, et al., 2017) (Ma, et al., 2017) (Black, et al., 2015) (Linardou, et al. 2008). Cyclin E gene amplification also has been reported as mechanism of resistance to Trastuzumab in several cancer models (Scaltriti, et al., 2011) (Cocco, et al., 2016). These genomic alterations may therefore also contribute to confer resistance to anti-HER2 therapies in the context of HER2 mutant tumors. As mentioned before, TP53, PIK3CA and CCNE are among the most altered genes in HER2 mutated bladder cancers, thus potentially explaining why these tumors are refractory to anti HER2 therapies. Moreover, among the 66 total patients of the SUMMIT trial that did not respond to Neratinib monotherapy, HER3 mutations were found in 14% of their tumors. Although additional studies are needed to evaluate whether these mutations can confer resistance to Neratinib, it has recently been demonstrated that breast cancer cell lines harboring HER3 mutations (F94L, G284R, D297Y, T355I, and E1261A) have enhanced HER2-HER3 heterodimerization that induces a gain-of-function phenotype and results in Lapatinib resistance (Mishra, et al., 2018). Furthermore, it has been recently demonstrated that the FDA-approved pan-ERBB inhibitors Afatinib and Neratinib effectively reduced the expression of mutant K-RAS proteins in several tumor types (Moll HP, et al., 2018) (Booth et al., 2018) (Booth et al., 2019), thus supporting the use of these agents to treat K-RAS mutated cancers.
5. Conclusions
Despite their low frequency, activating HER2 mutations have recently emerged as novel therapeutic targets for a variety of human tumors. Although anti-HER2 therapies have shown promising clinical results in a subset of HER2-mutated breast, lung and cervical cancer patients, the same therapies were largely inefficacious in other tumor types such as bladder and colorectal cancers. Whether the lack of responses is related to concurrent presence of genomic alterations that can obliterate the dependency to HER2-mutations remains to be elucidated, but the available data from basket trials suggest that ad hoc registrational clinical studies may be successfully designed for specific histologies. In this context, the use of novel emerging anti-HER2 agents or combinatorial treatments may represent meaningful advancements for the treatment of HER2 mutated cancer patients.
Acknowledgments:
This work was founded by the NIH grants R03CA187094–01 and P30CA008748 and the Breast Cancer Research Foundation. EC is a recipient of a MSK Society Scholar Prize.
Lists of abbreviation:
- NGS
next generation sequencing
- cfDNA
circulating cell-free DNA
- NTRK
neurotrophic receptor tyrosine kinase
- PFS
progression-free survival
- OS
overall survival
- ECD
extracellular domain
- JM
juxtamembrane domain
- TM
transmembrane domain
- KD
kinase domain
- TKI
tyrosine kinase inhibitor
- TCGA
The Cancer Genome Atlas
- TDM-1
Trastuzumab Emtansine
- IHC
immunohistochemistry
- ER
estrogen receptor
- CBR
clinical benefit rate
- ADC
antibody drug conjugate
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Statement:
M.S. is in the Advisory Board of Bioscience Institute and Menarini Ricerche, received research funds from Puma Biotechnology, Daiichi-Sankio, Targimmune, Immunomedics and Menarini Ricerche, is a co-founder of Medendi Medical Travel and in the past two years he received honoraria from Menarini Ricerche and ADC Pharma. ADS has received research funds from Merck, Genentech/Roche, Puma Biotechnology, Gilead Science and Immunomedics Inc.
References
- Aceto N, Sausgruber N, Brinkhaus H, Gaidatzis D, Martiny-Baron G, Mazzarol G, Confalonieri S, Quarto M, Hu G, Balwierz PJ, Pachkov M, Elledge SJ, van Nimwegen E, Stadler MB, & Bentires-Alj M (2012). Tyrosine phosphatase SHP2 promotes breast cancer progression and maintains tumor-initiating cells via activation of key transcription factors and a positive feedback signaling loop. Nat Med, 18, 529–537. [DOI] [PubMed] [Google Scholar]
- Ali SM, Alpaugh RK, Downing SR, Stephens PJ, Yu JQ, Wu H, Buell JK, Miller VA, Lipson D, Palmer GA, Ross JS, & Cristofanilli M (2014). Response of an ERBB2-Mutated Inflammatory Breast Carcinoma to Human Epidermal Growth Factor Receptor 2-Targeted Therapy. J Clin Oncol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amiri-Kordestani L, Blumenthal GM, Xu QC, Zhang L, Tang SW, Ha L, Weinberg WC, Chi B, Candau-Chacon R, Hughes P, Russell AM, Miksinski SP, Chen XH, McGuinn WD, Palmby T, Schrieber SJ, Liu Q, Wang J, Song P, Mehrotra N, Skarupa L, Clouse K, Al-Hakim A, Sridhara R, Ibrahim A, Justice R, Pazdur R, & Cortazar P (2014). FDA approval: ado-trastuzumab emtansine for the treatment of patients with HER2-positive metastatic breast cancer. Clin Cancer Res, 20, 4436–4441. [DOI] [PubMed] [Google Scholar]
- Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, Colaprico A, Wendl MC, Kim J, Reardon B, Kwok-Shing Ng P, Jeong KJ, Cao S, Wang Z, Gao J, Gao Q, Wang F, Liu EM, Mularoni L, Rubio-Perez C, Nagarajan N, Cortes-Ciriano I, Zhou DC, Liang WW, Hess JM, Yellapantula VD, Tamborero D, Gonzalez-Perez A, Suphavilai C, Ko JY, Khurana E, Park PJ, Van Allen EM, Liang H, Group, M. C. W., Cancer Genome Atlas Research, N., Lawrence MS, Godzik A, Lopez-Bigas N, Stuart J, Wheeler D, Getz G, Chen K, Lazar AJ, Mills GB, Karchin R, & Ding L (2018). Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell, 174, 1034–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, Colaprico A, Wendl MC, Kim J, Reardon B, Ng PK, Jeong KJ, Cao S, Wang Z, Gao J, Gao Q, Wang F, Liu EM, Mularoni L, Rubio-Perez C, Nagarajan N, Cortes-Ciriano I, Zhou DC, Liang WW, Hess JM, Yellapantula VD, Tamborero D, Gonzalez-Perez A, Suphavilai C, Ko JY, Khurana E, Park PJ, Van Allen EM, Liang H, Group, M. C. W., Cancer Genome Atlas Research, N., Lawrence MS, Godzik A, Lopez-Bigas N, Stuart J, Wheeler D, Getz G, Chen K, Lazar AJ, Mills GB, Karchin R, & Ding L (2018). Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell, 173, 371–385 e318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, Lawrence MS, Sivachenko AY, Sougnez C, Zou L, Cortes ML, Fernandez-Lopez JC, Peng S, Ardlie KG, Auclair D, Bautista-Piña V, Duke F, Francis J, Jung J, Maffuz-Aziz A, Onofrio RC, Parkin M, Pho NH, Quintanar-Jurado V, Ramos AH, Rebollar-Vega R, Rodriguez-Cuevas S, Romero-Cordoba SL, Schumacher SE, Stransky N, Thompson KM, Uribe-Figueroa L, Baselga J, Beroukhim R, Polyak K, Sgroi DC, Richardson AL, Jimenez-Sanchez G, Lander ES, Gabriel SB, Garraway LA, Golub TR, Melendez-Zajgla J, Toker A, Getz G, Hidalgo-Miranda A, & Meyerson M (2012). Sequence analysis of mutations and translocations across breast cancer subtypes. Nature, 486, 405–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J, Lehle M, Ruschoff J, Kang YK, & To GATI (2010). 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, 376, 687–697. [DOI] [PubMed] [Google Scholar]
- Ben-Baruch NE, Bose R, Kavuri SM, Ma CX, & Ellis MJ (2015). HER2-Mutated Breast Cancer Responds to Treatment With Single-Agent Neratinib, a Second-Generation HER2/EGFR Tyrosine Kinase Inhibitor. J Natl Compr Canc Netw, 13, 1061–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bentires-Alj M, Paez JG, David FS, Keilhack H, Halmos B, Naoki K, Maris JM, Richardson A, Bardelli A, Sugarbaker DJ, Richards WG, Du J, Girard L, Minna JD, Loh ML, Fisher DE, Velculescu VE, Vogelstein B, Meyerson M, Sellers WR, & Neel BG (2004). Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res, 64, 8816–8820. [DOI] [PubMed] [Google Scholar]
- Black JD, Lopez S, Cocco E, Bellone S, Altwerger G, Schwab CL, English DP, Bonazzoli E, Predolini F, Ferrari F, Ratner E, Silasi DA, Azodi M, Schwartz PE, & Santin AD (2015). PIK3CA oncogenic mutations represent a major mechanism of resistance to trastuzumab in HER2/neu overexpressing uterine serous carcinomas. Br J Cancer, 113, 1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blumenthal GM, Scher NS, Cortazar P, Chattopadhyay S, Tang S, Song P, Liu Q, Ringgold K, Pilaro AM, Tilley A, King KE, Graham L, Rellahan BL, Weinberg WC, Chi B, Thomas C, Hughes P, Ibrahim A, Justice R, & Pazdur R (2013). First FDA approval of dual anti-HER2 regimen: pertuzumab in combination with trastuzumab and docetaxel for HER2-positive metastatic breast cancer. Clin Cancer Res, 19, 4911–4916. [DOI] [PubMed] [Google Scholar]
- Booth L, Roberts JL, Poklepovic A, Kirkwood J, Sander C, Avogadri-Connors F, Cutler RE Jr., Lalani AS, & Dent P (2018). The levels of mutant K-RAS and mutant N-RAS are rapidly reduced in a Beclin1 / ATG5 -dependent fashion by the irreversible ERBB1/2/4 inhibitor neratinib. Cancer Biol Ther, 19, 132–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Booth L, Roberts JL, Rais R, Cutler RE Jr., Diala I, Lalani AS, Hancock JF, Poklepovic A, & Dent P (2019). Neratinib augments the lethality of [regorafenib + sildenafil]. J Cell Physiol, 234, 4874–4887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, Goel N, Aronson AB, Li S, Ma CX, Ding L, Mardis ER, & Ellis MJ (2013). Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov, 3, 224–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buza N, English DP, Santin AD, & Hui P (2013). Toward standard HER2 testing of endometrial serous carcinoma: 4-year experience at a large academic center and recommendations for clinical practice. Mod Pathol, 26, 1605–1612. [DOI] [PubMed] [Google Scholar]
- Canale M, Petracci E, Delmonte A, Chiadini E, Dazzi C, Papi M, Capelli L, Casanova C, De Luigi N, Mariotti M, Gamboni A, Chiari R, Bennati C, Calistri D, Ludovini V, Crino L, Amadori D, & Ulivi P (2017). Impact of TP53 Mutations on Outcome in EGFR-Mutated Patients Treated with First-Line Tyrosine Kinase Inhibitors. Clin Cancer Res, 23, 2195–2202. [DOI] [PubMed] [Google Scholar]
- Cancer Genome Atlas, N. (2012). Comprehensive molecular characterization of human colon and rectal cancer. Nature, 487, 330–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Network, T. (2012). Comprehensive molecular portraits of human breast tumors. Nature, 490, 61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research, N. (2012). Comprehensive genomic characterization of squamous cell lung cancers. Nature, 489, 519–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research, N. (2014a). Comprehensive molecular characterization of urothelial bladder carcinoma. Nature, 507, 315–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research, N. (2014b). Comprehensive molecular profiling of lung adenocarcinoma. Nature, 511, 543–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research, N., Analysis Working Group: Asan, U., Agency, B. C. C., Brigham, Women’s, H., Broad, I., Brown, U., Case Western Reserve, U., Dana-Farber Cancer, I., Duke, U., Greater Poland Cancer, C., Harvard Medical, S., Institute for Systems, B., Leuven, K. U., Mayo, C., Memorial Sloan Kettering Cancer, C., National Cancer, I., Nationwide Children’s, H., Stanford, U., University of, A., University of, M., University of North, C., University of, P., University of, R., University of Southern, C., University of Texas, M. D. A. C. C., University of, W., Van Andel Research, I., Vanderbilt, U., Washington, U., Genome Sequencing Center: Broad, I., Washington University in St, L., Genome Characterization Centers, B. C. C. A., Broad, I., Harvard Medical, S., Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, U., University of North, C., University of Southern California Epigenome, C., University of Texas, M. D. A. C. C., Van Andel Research, I., Genome Data Analysis Centers: Broad, I., Brown, U., Harvard Medical, S., Institute for Systems, B., Memorial Sloan Kettering Cancer, C., University of California Santa, C., University of Texas, M. D. A. C. C., Biospecimen Core Resource: International Genomics, C., Research Institute at Nationwide Children’s, H., Tissue Source Sites: Analytic Biologic, S., Asan Medical, C., Asterand, B., Barretos Cancer, H., BioreclamationIvt, Botkin Municipal, C., Chonnam National University Medical, S., Christiana Care Health, S., Cureline, Duke, U., Emory, U., Erasmus, U., Indiana University School of, M., Institute of Oncology of, M., International Genomics, C., Invidumed, Israelitisches Krankenhaus, H., Keimyung University School of, M., Memorial Sloan Kettering Cancer, C., National Cancer Center, G., Ontario Tumour, B., Peter MacCallum Cancer, C., Pusan National University Medical, S., Ribeirao Preto Medical, S., St. Joseph’s, H., Medical, C., St. Petersburg Academic, U., Tayside Tissue, B., University of, D., University of Kansas Medical, C., University of, M., University of North Carolina at Chapel, H., University of Pittsburgh School of, M., University of Texas, M. D. A. C. C., Disease Working Group: Duke, U., Memorial Sloan Kettering Cancer, C., National Cancer, I., University of Texas, M. D. A. C. C., Yonsei University College of, M., Data Coordination Center, C. I., & Project Team: National Institutes of, H. (2017). Integrated genomic characterization of oesophageal carcinoma. Nature, 541, 169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research, N., Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, & Stuart JM (2013). The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet, 45, 1113–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW Jr., & Leahy DJ (2003). Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature, 421, 756–760. [DOI] [PubMed] [Google Scholar]
- Chuang JC, Stehr H, Liang Y, Das M, Huang J, Diehn M, Wakelee HA, & Neal JW (2017). ERBB2-Mutated Metastatic Non-Small Cell Lung Cancer: Response and Resistance to Targeted Therapies. J Thorac Oncol, 12, 833–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chumsri S, Weidler J, Ali S, Balasubramanian S, G. W, DeFazio-Eli L, Chenna A, Huang W, DeRiddle A,L,G, & Perez EA (2015). Prolonged response to trastuzumab in HER2 non-amplified patient with elevated HER2 dimerization harboring ERBB2 S310F mutation J Natl Compr Canc Netw, 13. [DOI] [PubMed] [Google Scholar]
- Cocco E, Javier Carmona F, Razavi P, Won HH, Cai Y, Rossi V, Chan C, Cownie J, Soong J, Toska E, Shifman SG, Sarotto I, Savas P, Wick MJ, Papadopoulos KP, Moriarty A, Cutler RE Jr., Avogadri-Connors F, Lalani AS, Bryce RP, Chandarlapaty S, Hyman DM, Solit DB, Boni V, Loi S, Baselga J, Berger MF, Montemurro F, & Scaltriti M (2018). Neratinib is effective in breast tumors bearing both amplification and mutation of ERBB2 (HER2). Sci Signal, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cocco E, Lopez S, Black J, Bellone S, Bonazzoli E, Predolini F, Ferrari F, Schwab CL, Menderes G, Zammataro L, Buza N, Hui P, Wong S, Zhao S, Bai Y, Rimm DL, Ratner E, Litkouhi B, Silasi DA, Azodi M, Schwartz PE, & Santin AD (2016). Dual CCNE1/PIK3CA targeting is synergistic in CCNE1-amplified/PIK3CA-mutated uterine serous carcinomas in vitro and in vivo. Br J Cancer, 115, 303–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Croessmann S, Formisano L, Kinch LN, Gonzalez-Ericsson PI, Sudhan DR, Nagy RJ, Mathew A, Bernicker EH, Cristofanilli M, He J, Cutler RE Jr., Lalani AS, Miller VA, Lanman RB, Grishin NV, & Arteaga CL (2019). Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer. Clin Cancer Res, 25, 277–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Greve J, Moran T, Graas MP, Galdermans D, Vuylsteke P, Canon JL, Schallier D, Decoster L, Teugels E, Massey D, Chand VK, & Vansteenkiste J (2015). Phase II study of afatinib, an irreversible ErbB family blocker, in demographically and genotypically defined lung adenocarcinoma. Lung Cancer, 88, 63–69. [DOI] [PubMed] [Google Scholar]
- De Greve J, Teugels E, Geers C, Decoster L, Galdermans D, De Mey J, Everaert H, Umelo I, In’t Veld P, & Schallier D (2012). Clinical activity of afatinib (BIBW 2992) in patients with lung adenocarcinoma with mutations in the kinase domain of HER2/neu. Lung Cancer, 76, 123–127. [DOI] [PubMed] [Google Scholar]
- de Martino M, Zhuang D, Klatte T, Rieken M, Roupret M, Xylinas E, Clozel T, Krzywinski M, Elemento O, & Shariat SF (2014). Impact of ERBB2 mutations on in vitro sensitivity of bladder cancer to lapatinib. Cancer Biol Ther, 15, 1239–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, Nathenson M, Doebele RC, Farago AF, Pappo AS, Turpin B, Dowlati A, Brose MS, Mascarenhas L, Federman N, Berlin J, El-Deiry WS, Baik C, Deeken J, Boni V, Nagasubramanian R, Taylor M, Rudzinski ER, Meric-Bernstam F, Sohal DPS, Ma PC, Raez LE, Hechtman JF, Benayed R, Ladanyi M, Tuch BB, Ebata K, Cruickshank S, Ku NC, Cox MC, Hawkins DS, Hong DS, & Hyman DM (2018). Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med, 378, 731–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, Van Tine BA, Hoog J, Goiffon RJ, Goldstein TC, Ng S, Lin L, Crowder R, Snider J, Ballman K, Weber J, Chen K, Koboldt DC, Kandoth C, Schierding WS, McMichael JF, Miller CA, Lu C, Harris CC, McLellan MD, Wendl MC, DeSchryver K, Allred DC, Esserman L, Unzeitig G, Margenthaler J, Babiera GV, Marcom PK, Guenther JM, Leitch M, Hunt K, Olson J, Tao Y, Maher CA, Fulton LL, Fulton RS, Harrison M, Oberkfell B, Du F, Demeter R, Vickery TL, Elhammali A, Piwnica-Worms H, McDonald S, Watson M, Dooling DJ, Ota D, Chang LW, Bose R, Ley TJ, Piwnica-Worms D, Stuart JM, Wilson RK, & Mardis ER (2012). Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature, 486, 353–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fabrice A (2018). LBA3_PR - Alpelisib (ALP) + fulvestrant (FUL) for advanced breast cancer (ABC): results of the Phase 3 SOLAR-1 trial. In ESMO. [Google Scholar]
- Fader AN, Roque DM, Siegel E, Buza N, Hui P, Abdelghany O, Chambers SK, Secord AA, Havrilesky L, O’Malley DM, Backes F, Nevadunsky N, Edraki B, Pikaart D, Lowery W, ElSahwi KS, Celano P, Bellone S, Azodi M, Litkouhi B, Ratner E, Silasi DA, Schwartz PE, & Santin AD (2018). Randomized Phase II Trial of Carboplatin-Paclitaxel Versus Carboplatin-Paclitaxel-Trastuzumab in Uterine Serous Carcinomas That Overexpress Human Epidermal Growth Factor Receptor 2/neu. J Clin Oncol, 36, 2044–2051. [DOI] [PubMed] [Google Scholar]
- Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, & Sliwkowski MX (2004). Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell, 5, 317–328. [DOI] [PubMed] [Google Scholar]
- Giannakis M, Mu XJ, Shukla SA, Qian ZR, Cohen O, Nishihara R, Bahl S, Cao Y, Amin-Mansour A, Yamauchi M, Sukawa Y, Stewart C, Rosenberg M, Mima K, Inamura K, Nosho K, Nowak JA, Lawrence MS, Giovannucci EL, Chan AT, Ng K, Meyerhardt JA, Van Allen EM, Getz G, Gabriel SB, Lander ES, Wu CJ, Fuchs CS, Ogino S, & Garraway LA (2016). Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma. Cell Rep, 17, 1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grabsch H, Sivakumar S, Gray S, Gabbert HE, & Muller W (2010). HER2 expression in gastric cancer: Rare, heterogeneous and of no prognostic value - conclusions from 924 cases of two independent series. Cell Oncol, 32, 57–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gradishar WJ (2012). HER2 therapy--an abundance of riches. N Engl J Med, 366, 176–178. [DOI] [PubMed] [Google Scholar]
- Graus-Porta D, Beerli RR, Daly JM, & Hynes NE (1997). ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J, 16, 1647–1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greulich H, Kaplan B, Mertins P, Chen TH, Tanaka KE, Yun CH, Zhang X, Lee SH, Cho J, Ambrogio L, Liao R, Imielinski M, Banerji S, Berger AH, Lawrence MS, Zhang J, Pho NH, Walker SR, Winckler W, Getz G, Frank D, Hahn WC, Eck MJ, Mani DR, Jaffe JD, Carr SA, Wong KK, & Meyerson M (2012). Functional analysis of receptor tyrosine kinase mutations in lung cancer identifies oncogenic extracellular domain mutations of ERBB2. Proc Natl Acad Sci U S A, 109, 14476–14481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hainsworth JD, Meric-Bernstam F, Swanton C, Hurwitz H, Spigel DR, Sweeney C, Burris H, Bose R, Yoo B, Stein A, Beattie M, & Kurzrock R (2018). Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results From MyPathway, an Open-Label, Phase IIa Multiple Basket Study. J Clin Oncol, 36, 536–542. [DOI] [PubMed] [Google Scholar]
- Hanker AB, Brewer MR, Sheehan JH, Koch JP, Sliwoski GR, Nagy R, Lanman R, Berger MF, Hyman DM, Solit DB, He J, Miller V, Cutler RE Jr., Lalani AS, Cross D, Lovly CM, Meiler J, & Arteaga CL (2017). An Acquired HER2(T798I) Gatekeeper Mutation Induces Resistance to Neratinib in a Patient with HER2 Mutant-Driven Breast Cancer. Cancer Discov, 7, 575–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hellmuth K, Grosskopf S, Lum CT, Wurtele M, Roder N, von Kries JP, Rosario M, Rademann J, & Birchmeier W (2008). Specific inhibitors of the protein tyrosine phosphatase Shp2 identified by high-throughput docking. Proc Natl Acad Sci U S A, 105, 7275–7280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussain MH, MacVicar GR, Petrylak DP, Dunn RL, Vaishampayan U, Lara PN Jr., Chatta GS, Nanus DM, Glode LM, Trump DL, Chen H, Smith DC, & National Cancer I. (2007). 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, 25, 2218–2224. [DOI] [PubMed] [Google Scholar]
- Hyman DM, Piha-Paul SA, Rodon J, Saura C, Shapiro GI, Quinn DI, Moreno V, & al e. (2017). Abstract CT001: Neratinib in HER2 or HER3 mutant solid tumors: SUMMIT, a global, multi-histology, open-label, phase 2 “basket” study. Cancer Research, 77(13 Supplement), CT001. [Google Scholar]
- Hyman DM, Piha-Paul SA, Saura C, Arteaga C, Mayer I, Shapiro GI, & al e. (2017). Abstract PD2–08: Neratinib + fulvestrant in ERBB2 mutant, HER2–non-amplified, estrogen receptor (ER) positive, metastatic breast cancer (MBC): Preliminary analysis from the phase II SUMMIT trial. Cancer Research, 77(4 Supplement), PD2–08. [Google Scholar]
- Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, Juric D, Quinn DI, Moreno V, Doger B, Mayer IA, Boni V, Calvo E, Loi S, Lockhart AC, Erinjeri JP, Scaltriti M, Ulaner GA, Patel J, Tang J, Beer H, Selcuklu SD, Hanrahan AJ, Bouvier N, Melcer M, Murali R, Schram AM, Smyth LM, Jhaveri K, Li BT, Drilon A, Harding JJ, Iyer G, Taylor BS, Berger MF, Cutler RE Jr., Xu F, Butturini A, Eli LD, Mann G, Farrell C, Lalani AS, Bryce RP, Arteaga CL, Meric-Bernstam F, Baselga J, & Solit DB (2018). HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature, 554, 189–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, Wolf J, Raje NS, Diamond EL, Hollebecque A, Gervais R, Elez-Fernandez ME, Italiano A, Hofheinz RD, Hidalgo M, Chan E, Schuler M, Lasserre SF, Makrutzki M, Sirzen F, Veronese ML, Tabernero J, & Baselga J (2015). Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med, 373, 726–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kancha RK, von Bubnoff N, Bartosch N, Peschel C, Engh RA, & Duyster J (2011). Differential sensitivity of ERBB2 kinase domain mutations towards lapatinib. PLoS One, 6, e26760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, Leiserson MDM, Miller CA, Welch JS, Walter MJ, Wendl MC, Ley TJ, Wilson RK, Raphael BJ, & Ding L (2013). Mutational landscape and significance across 12 major cancer types. Nature, 502, 333–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kavuri SM, Jain N, Galimi F, Cottino F, Leto SM, Migliardi G, Searleman AC, Shen W, Monsey J, Trusolino L, Jacobs SA, Bertotti A, & Bose R (2015). HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov, 5, 832–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kloth M, Ruesseler V, Engel C, Koenig K, Peifer M, Mariotti E, Kuenstlinger H, Florin A, Rommerscheidt-Fuss U, Koitzsch U, Wodtke C, Ueckeroth F, Holzapfel S, Aretz S, Propping P, Loeffler M, Merkelbach-Bruse S, Odenthal M, Friedrichs N, Heukamp LC, Zander T, & Buettner R (2016). Activating ERBB2/HER2 mutations indicate susceptibility to pan-HER inhibitors in Lynch and Lynch-like colorectal cancer. Gut, 65, 1296–1305. [DOI] [PubMed] [Google Scholar]
- Lee J, Kang HA, Bae JS, Kim KD, Lee KH, Lim KJ, Choo MJ, & Chang SJ (2018). Evaluation of analytical similarity between trastuzumab biosimilar CT-P6 and reference product using statistical analyses. MAbs, 10, 547–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li BT, Shen R, Buonocore D, Olah ZT, Ni A, Ginsberg MS, Ulaner GA, Offin M, Feldman D, Hembrough T, Cecchi F, Schwartz S, Pavlakis N, Clarke S, Won HH, Brzostowski EB, Riely GJ, Solit DB, Hyman DM, Drilon A, Rudin CM, Berger MF, Baselga J, Scaltriti M, Arcila ME, & Kris MG (2018). Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J Clin Oncol, 36, 2532–2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linardou H, Dahabreh IJ, Kanaloupiti D, Siannis F, Bafaloukos D, Kosmidis P, Papadimitriou CA, Murray S (2008). Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: A systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol, 9, 962–972. [DOI] [PubMed] [Google Scholar]
- Liu S, Li S, Hai J, Wang X, Chen T, Quinn MM, Gao P, Zhang Y, Ji H, Cross DAE, & Wong KK (2018). Targeting HER2 Aberrations in Non-Small Cell Lung Cancer with Osimertinib. Clin Cancer Res, 24, 2594–2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma CX, Bose R, Gao F, Freedman RA, Telli ML, Kimmick G, Winer E, Naughton M, Goetz MP, Russell C, Tripathy D, Cobleigh M, Forero A, Pluard TJ, Anders C, Niravath PA, Thomas S, Anderson J, Bumb C, Banks KC, Lanman RB, Bryce R, Lalani AS, Pfeifer J, Hayes DF, Pegram M, Blackwell K, Bedard PL, Al-Kateb H, & Ellis MJC (2017). Neratinib Efficacy and Circulating Tumor DNA Detection of HER2 Mutations in HER2 Nonamplified Metastatic Breast Cancer. Clin Cancer Res, 23, 5687–5695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin M, Holmes FA, Ejlertsen B, Delaloge S, Moy B, Iwata H, von Minckwitz G, Chia SKL, Mansi J, Barrios CH, Gnant M, Tomasevic Z, Denduluri N, Separovic R, Gokmen E, Bashford A, Ruiz Borrego M, Kim SB, Jakobsen EH, Ciceniene A, Inoue K, Overkamp F, Heijns JB, Armstrong AC, Link JS, Joy AA, Bryce R, Wong A, Moran S, Yao B, Xu F, Auerbach A, Buyse M, & Chan A (2017). Neratinib after trastuzumab-based adjuvant therapy in HER2-positive breast cancer (ExteNET): 5-year analysis of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol, 18, 1688–1700. [DOI] [PubMed] [Google Scholar]
- Marty M, Cognetti F, Maraninchi D, Snyder R, Mauriac L, Tubiana-Hulin M, Chan S, Grimes D, Anton A, Lluch A, Kennedy J, O’Byrne K, Conte P, Green M, Ward C, Mayne K, & Extra JM (2005). Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J Clin Oncol, 23, 4265–4274. [DOI] [PubMed] [Google Scholar]
- Menderes G, Lopez S, Han C, Altwerger G, Gysler S, Varughese J, Schwartz PE, & Santin AD (2018). Mechanisms of resistance to HER2-targeted therapies in HER2-amplified uterine serous carcinoma, and strategies to overcome it. Discov Med, 26, 39–50. [PubMed] [Google Scholar]
- Mishra R, Alanazi S, Yuan L, Solomon T, Thaker TM, Jura N, & Garrett JT (2018). Activating HER3 mutations in breast cancer. Oncotarget, 9, 27773–27788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moll HP, Pranz K, Musteanu M, Grabner B, Hruschka N, Mohrherr J, Aigner P, Stiedl P, Brcic L, Laszlo V, Schramek D, Moriggl R, Eferl R, Moldvay J, Dezso K, Lopez-Casas PP, Stoiber D, Hidalgo M, Penninger J, Sibilia M, Gyorffy B, Barbacid M, Dome B, Popper H, & Casanova E (2018). Afatinib restrains K-RAS-driven lung tumorigenesis. Sci Transl Med, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nayar U, Cohen O, Kapstad C, Cuoco MS, Waks AG, Wander SA, Painter C, Freeman S, Persky NS, Marini L, Helvie K, Oliver N, Rozenblatt-Rosen O, Ma CX, Regev A, Winer EP, Lin NU, & Wagle N (2019). Acquired HER2 mutations in ER(+) metastatic breast cancer confer resistance to estrogen receptor-directed therapies. Nat Genet, 51, 207–216. [DOI] [PubMed] [Google Scholar]
- Ojesina AI, Lichtenstein L, Freeman SS, Pedamallu CS, Imaz-Rosshandler I, Pugh TJ, Cherniack AD, Ambrogio L, Cibulskis K, Bertelsen B, Romero-Cordoba S, Trevino V, Vazquez-Santillan K, Guadarrama AS, Wright AA, Rosenberg MW, Duke F, Kaplan B, Wang R, Nickerson E, Walline HM, Lawrence MS, Stewart C, Carter SL, McKenna A, Rodriguez-Sanchez IP, Espinosa-Castilla M, Woie K, Bjorge L, Wik E, Halle MK, Hoivik EA, Krakstad C, Gabino NB, Gomez-Macias GS, Valdez-Chapa LD, Garza-Rodriguez ML, Maytorena G, Vazquez J, Rodea C, Cravioto A, Cortes ML, Greulich H, Crum CP, Neuberg DS, Hidalgo-Miranda A, Escareno CR, Akslen LA, Carey TE, Vintermyr OK, Gabriel SB, Barrera-Saldana HA, Melendez-Zajgla J, Getz G, Salvesen HB, & Meyerson M (2014). Landscape of genomic alterations in cervical carcinomas. Nature, 506, 371–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oudard S, Culine S, Vano Y, Goldwasser F, Theodore C, Nguyen T, Voog E, Banu E, Vieillefond A, Priou F, Deplanque G, Gravis G, Ravaud A, Vannetzel JM, Machiels JP, Muracciole X, Pichon MF, Bay JO, Elaidi R, Teghom C, Radvanyi F, & Beuzeboc P (2015). Multicentre randomised phase II trial of gemcitabine+platinum, with or without trastuzumab, in advanced or metastatic urothelial carcinoma overexpressing Her2. Eur J Cancer, 51, 45–54. [DOI] [PubMed] [Google Scholar]
- Pahuja KB, Nguyen TT, Jaiswal BS, Prabhash K, Thaker TM, Senger K, Chaudhuri S, Kljavin NM, Antony A, Phalke S, Kumar P, Mravic M, Stawiski EW, Vargas D, Durinck S, Gupta R, Khanna-Gupta A, Trabucco SE, Sokol ES, Hartmaier RJ, Singh A, Chougule A, Trivedi V, Dutt A, Patil V, Joshi A, Noronha V, Ziai J, Banavali SD, Ramprasad V, DeGrado WF, Bueno R, Jura N, & Seshagiri S (2018). Actionable Activating Oncogenic ERBB2/HER2 Transmembrane and Juxtamembrane Domain Mutations. Cancer Cell, 34, 792–806 e795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park CK, Hur JY, Choi CM, Kim TO, Cho HJ, Shin HJ, Lim JH, Choi YD, Kim YC, & Oh IJ (2018). Efficacy of Afatinib in a Previously-Treated Patient with Non-Small Cell Lung Cancer Harboring HER2 Mutation: Case Report. J Korean Med Sci, 33, e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perera SA, Li D, Shimamura T, Raso MG, Ji H, Chen L, Borgman CL, Zaghlul S, Brandstetter KA, Kubo S, Takahashi M, Chirieac LR, Padera RF, Bronson RT, Shapiro GI, Greulich H, Meyerson M, Guertler U, Chesa PG, Solca F, Wistuba II, & Wong KK (2009). HER2YVMA drives rapid development of adenosquamous lung tumors in mice that are sensitive to BIBW2992 and rapamycin combination therapy. Proc Natl Acad Sci U S A, 106, 474–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, Cai Y, Bielski CM, Donoghue MTA, Jonsson P, Penson A, Shen R, Pareja F, Kundra R, Middha S, Cheng ML, Zehir A, Kandoth C, Patel R, Huberman K, Smyth LM, Jhaveri K, Modi S, Traina TA, Dang C, Zhang W, Weigelt B, Li BT, Ladanyi M, Hyman DM, Schultz N, Robson ME, Hudis C, Brogi E, Viale A, Norton L, Dickler MN, Berger MF, Iacobuzio-Donahue CA, Chandarlapaty S, Scaltriti M, Reis-Filho JS, Solit DB, Taylor BS, & Baselga J (2018). The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell, 34, 427–438 e426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rexer BN, & Arteaga CL (2012). Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications. Crit Rev Oncog, 17, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, Hinoue T, Laird PW, Hoadley KA, Akbani R, Castro MAA, Gibb EA, Kanchi RS, Gordenin DA, Shukla SA, Sanchez-Vega F, Hansel DE, Czerniak BA, Reuter VE, Su X, de Sa Carvalho B, Chagas VS, Mungall KL, Sadeghi S, Pedamallu CS, Lu Y, Klimczak LJ, Zhang J, Choo C, Ojesina AI, Bullman S, Leraas KM, Lichtenberg TM, Wu CJ, Schultz N, Getz G, Meyerson M, Mills GB, McConkey DJ, Network TR, Weinstein JN, Kwiatkowski DJ, & Lerner SP (2017). Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell, 171, 540–556 e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato JD, Kawamoto T, Le AD, Mendelsohn J, Polikoff J, & Sato GH (1983). Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Mol Biol Med, 1, 511–529. [PubMed] [Google Scholar]
- Scaltriti M, Eichhorn PJ, Cortes J, Prudkin L, Aura C, Jimenez J, Chandarlapaty S, Serra V, Prat A, Ibrahim YH, Guzman M, Gili M, Rodriguez O, Rodriguez S, Perez J, Green SR, Mai S, Rosen N, Hudis C, & Baselga J (2011). Cyclin E amplification/overexpression is a mechanism of trastuzumab resistance in HER2+ breast cancer patients. Proc Natl Acad Sci U S A, 108, 3761–3766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneeberger VE, Luetteke N, Ren Y, Berns H, Chen L, Foroutan P, Martinez GV, Haura EB, Chen J, Coppola D, & Wu J (2014). SHP2E76K mutant promotes lung tumorigenesis in transgenic mice. Carcinogenesis, 35, 1717–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serra V, Vivancos A, Puente XS, Felip E, Silberschmidt D, Caratu G, Parra JL, De Mattos-Arruda L, Grueso J, Hernandez-Losa J, Arribas J, Prudkin L, Nuciforo P, Scaltriti M, Seoane J, & Baselga J (2013). Clinical response to a lapatinib-based therapy for a Li-Fraumeni syndrome patient with a novel HER2V659E mutation. Cancer Discov, 3, 1238–1244. [DOI] [PubMed] [Google Scholar]
- Shi Y, & Wang M (2018). Afatinib as first-line treatment for advanced lung adenocarcinoma patients harboring HER2 mutation: A case report and review of the literature. Thorac Cancer, 9, 1788–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shih J, Bashir B, Gustafson KS, Andrake M, Dunbrack RL, Goldstein LJ, & Boumber Y (2015). Cancer Signature Investigation: ERBB2 (HER2)-Activating Mutation and Amplification-Positive Breast Carcinoma Mimicking Lung Primary. J Natl Compr Canc Netw, 13, 947–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith TJ, Davidson NE, Schapira DV, Grunfeld E, Muss HB, Vogel VG 3rd, & Somerfield MR (1999). American Society of Clinical Oncology 1998 update of recommended breast cancer surveillance guidelines. J Clin Oncol, 17, 1080–1082. [DOI] [PubMed] [Google Scholar]
- Toska E, Osmanbeyoglu HU, Castel P, Chan C, Hendrickson RC, Elkabets M, Dickler MN, Scaltriti M, Leslie CS, Armstrong SA, & Baselga J (2017). PI3K pathway regulates ER-dependent transcription in breast cancer through the epigenetic regulator KMT2D. Science, 355, 1324–1330. [DOI] [PMC free article] [PubMed] [Google Scholar]




