Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2016 Aug 1;6(8):1609–1623.

A perspective on anti-EGFR therapies targeting triple-negative breast cancer

Katsuya Nakai 1,2, Mien-Chie Hung 1,3,4, Hirohito Yamaguchi 1
PMCID: PMC5004067  PMID: 27648353

Abstract

Triple-negative breast cancer (TNBC), which lacks estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), accounts for about 15-20% of breast cancers and is the most aggressive breast cancer subtype. There are currently no effective therapies against metastatic TNBC. Compared with other breast cancer subtypes, EGFR is frequently overexpressed in TNBC and a potential therapeutic target for this disease. There are two types of EGFR inhibitors, small molecular tyrosine kinase inhibitor (TKI) and monoclonal antibody (mAb), for the treatment of cancers, such as non-small cell lung cancer and colorectal cancer. For breast cancer, however, the clinical trials of EGFR inhibitors have failed due to low response rates. Because a small portion of patients do demonstrate response to EGFR inhibitors, it may be necessary to stratify patients to enhance the efficacy of EGFR inhibitors in TNBC and to develop the effective combination therapy for this patient population. In this review, we describe some of the molecular mechanisms underlying EGFR inhibitor sensitivity and further discuss the possible therapeutic strategies to increase the efficacy of EGFR inhibitors in TNBC.

Keywords: TNBC, EGFR, EGFR inhibitors, drug resistance

Introduction

Breast cancer is the most common cancer type and the second leading cause of cancer-related deaths in women in the United States. The significant increase in the 5-year survival rate of breast cancer patients from 75 to 90% can be attributed to the advances in early detection and therapeutic approaches [1]. Although early-stage breast cancer is highly curable, the 5-year survival rate of patients with metastatic breast cancer is about 20%. Breast cancer is also a heterogeneous disease with multiple subtypes. About 15-20% of breast cancer patients are diagnosed with triple-negative breast cancer (TNBC), which is defined by the lack of expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor (HER2). TNBC is generally characterized by aggressive clinical course and poor prognosis compared with other breast cancer subtypes [2]. Moreover, due to the lack of druggable targets, such as ER and HER2, chemotherapy is still a primary option for systemic treatment. While TNBC patients respond better to chemotherapy than do non-TNBC patients, TNBC patients who do not respond eventually develop the metastatic form of the disease, which is virtually incurable [3,4]. Moreover, because the current treatment regimens are ineffective against a significant number of TNBC patients, new effective treatment strategies are urgently needed. In TNBC, the epidermal growth factor receptor (EGFR) is frequently overexpressed, and anti-EGFR therapies, including small molecule tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (mAbs), have been developed and are currently used for treatment of some cancers such as non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). Unfortunately, no EGFR therapies are currently approved for treatment of breast cancer. In this review, we briefly summarize the results from clinical studies of EGFR therapies and potential resistant mechanisms against anti-EGFR therapies in various cancer types and discuss the perspective on anti-EGFR therapies in TNBC.

Breast cancer subtypes

Immunohistochemistry (IHC) classification by ER, PR, and HER2 is currently used as the standard assessment of this disease in clinic. However, breast cancer is a heterogeneous disease that exhibits distinct clinical behavior, and recent advances in new technologies for gene and protein expression profiling further revealed the complex features of breast cancer. On the basis of gene expression profiling, breast cancer can be classified into five different subtypes that include basal-like, HER2-enriched, luminal A and B, and normal-like [5,6]. These subtypes, referred to as intrinsic subtypes, well match IHC classification [7], and among them, basal-like breast cancer (BLBC) is the most aggressive subtype with a high histologic grade, and accounts for about 15% of invasive breast cancer. About 80% of TNBC overlaps with BLBC, both of which are highly aggressive and exhibit poor clinical outcomes. Furthermore, TNBC harbors substantial genetic heterogeneity, and is further divided into six subtypes, including basal-like 1 (BL1), BL2, immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (M), MSL (mesenchymal-stem-like), and luminal androgen receptor (LAR) [8]. These TNBC subtypes have been reported to differ in drug sensitivity and require different therapeutic approaches based on their characteristics.

TNBC and BLBC are also closely associated with hereditary breast and ovarian cancer syndrome caused by germline mutations in the BRCA1/2 genes [9,10]. BRCA1 and BRCA2 are involved in double-stranded DNA damage repair and play an essential role in DNA integrity. The incidence of breast cancer in individuals with BRCA1/2 germline mutations is 20-30 times higher than those without the mutations, and 75% of breast cancer patients with BRCA1 mutation turned out to have TNBC [11]. Recently, inhibitors against poly (ADP-ribose) polymerase (PARP) have been shown to induce synthetic lethality in BRCA-deficient ovarian and breast cancers. One PARP inhibitor, olaparib, is approved for the treatment of BRCA mutated advanced ovarian cancer. Several other PARP inhibitors are being tested in clinical trials and are expected to receive approval for treatment for patients with TNBC and BLBC.

EGFR in human cancers

EGFR is a receptor tyrosine kinase (RTK) that belongs to the ErbB family, and a transmembrane protein comprising an extracellular ligand binding domain, transmembrane domain, and cytoplasmic tyrosine kinase domain [12-14]. When a ligand binds to the extracellular region of EGFR, the receptor forms a dimer, turning on its kinase activity, followed by autophosphorylation at multiple tyrosine residues in the intracellular region to recruit various substrates. The receptor activation promotes cell proliferation, motility, and survival via activation of various downstream signaling pathways, such as Ras-Raf-MEK-ERK, PI3K-AKT-mTOR, and Src-STAT3 [15]. Ligand-activated EGFR molecules are then ubiquitinated, internalized, and isolated in endosomes. There are two major pathways of internalized EGFR, lysosome-mediated degradation pathway, which transports EGFR to the lysosomes for degradation, and receptor recycling pathway, which sorts EGFR to cell surface again [14,15]. EGFR is also known to translocate into the nucleus, where it is involved in transcriptional regulation, DNA replication, and DNA repair [16]. The EGFR gene is frequently mutated or overexpressed lung, colon, head and neck, brain, pancreatic, and breast cancers and promotes tumor progression and drug resistance in these cancers [17-20]. Therefore, EGFR is an attractive drug target, and the inhibitors of EGFR, including TKIs and mAbs, have been developed and some are currently used in the clinic.

Overexpression of EGFR in cancer is partly due to gene amplification [21], but the underlying mechanisms are not yet fully understood. It has been reported that EGFR degradation through endocytosis is critical for upregulation of EGFR protein in some types of cancer cells, including breast cancer cells [22,23]. Moreover, inhibition of BRCA1 has been shown to induce upregulation of EGFR mRNA and protein in breast and ovarian cancer cells [24,25] although the molecular mechanisms are still uncertain. Because TNBC is closely associated with BRCA-mutant breast cancer, the BRCA1-mediated upregulation of EGFR mRNA and protein may partially explain the overexpression of EGFR in TNBC.

EGFR expression and mutation in human TNBC tissues

Compared to other subtypes of breast cancer, EGFR is more frequently overexpressed in TNBC [11], and EGFR expression has been recognized as a factor of poor prognosis for TNBC [26]. EGFR expression, gene amplification, and mutation status have been broadly studied in TNBC. IHC-based interrogation of the frequency of EGFR protein expression in TNBC indicated a range between 13-76%, largely depending on the methods of the evaluation and antibodies (Table 1) [27-34]. For example, Choi et al. [27] and Rakha et al. [29] reported a frequency of 13% and 37% of EGFR overexpression, respectively, in TNBC using an antibody from Novocastra (2+ to 3+ membranous staining in ≥ 10% tumor cells for evaluation). Tan et al. reported a rate of 52% of EGFR overexpression in TNBC using an antibody from Zymed (2+ to 3+ membranous staining in ≥ 10% tumor cells) [30]. Using EGFR PharmDx Kit (Dako), which is widely used to assess EGFR expression in CRC, Martin et al. have indicated a frequency of 76% of EGFR overexpression in TNBC [31]. EGFR protein overexpression has been detected in 72% of TNBC by EGFR PhrmDx, but a frequency of 11% and 47%, respectively, of EGFR mRNA overexpression and normal expression was also observed in the same study [32], suggesting that EGFR protein overexpression in TNBC is partly due to post-transcriptional regulation, such as protein stabilization or enhanced recycling. Amplification of EGFR gene was observed in 2-24% of TNBC patient tumor tissue samples whereas high polysomy of EGFR gene was reported to be between 8-27% (Table 1) [31,35-38]. Therefore, gene amplification only partly accounts for expression of EGFR protein.

Table 1.

EGFR protein expression, gene mutations, and copy number in TNBC

Year Authors Country Patient Number Methods Results
2007 Rakha et al. [29] UK 282 IHC (Novocastra) Positive: 37%
2008 Tan et al. [30] China 31 IHC (Zymed) Positive: 52%
2008 Toyama et al. [44] Japan 110 IHC (PhamaDx) Positive: 31%
qPCR (TaqMan) No activating mutations
2009 Viale et al. [28] Italy 284 IHC (Zymed) Positive: 13%
2010 Gumuskaya et al. [35] Turkey 62 IHC (Zymed, Novocastra) Positive: 61% (Zymed), 78% (Novocastra)
FISH (Abbot) Gene amplification: 2%
2011 Jacot et al. [41] France 229 Direct sequencing No activating mutations
2011 Shao et al. [38] China 59 FISH (GP medical) Gene amplification: 12%
2011 Teng et al. [42] China 70 Direct sequencing Exon 19 deletions and L858R mutation: 11%
2012 Choi et al. [27] Korea 122 IHC (Novocastra) Positive: 13%
2012 Liu et al. [33] China 287 IHC (Dako) Positive: 36%
2012 Meseure et al. [32] France 18 IHC (PharmDx) Positive: 72%
2012 Grob et al. [36] Germany 65 FISH (Abbott) Gene amplification: 2%
Direct sequencing No activating mutations
2012 Martin et al. [31] Switzerland 38 IHC (PharmDx) Positive: 76%
FISH (Abbot) Gene amplification: 24%
Direct sequencing No activating mutations
2012 Lv et al. [43] China 13 qPCR (TaqMan) L858R mutation: 7.7%
2012 Santarpia et al. [39] USA 267 Sequenom technology L858R mutation: 2.6%
2014 Nakajima et al. [45] Japan 84 IHC (Ventana) Positive: 33%
DISH (Ventana) No gene amplifications
PCR (DNAFORM) No activating mutations
2014 Park et al. [34] Korea 151 IHC (PharmDx) High EGFR copy number: 64%
FISH (Abbot) Gene amplification: 2%
High polysomy: 31%
Direct sequencing L858R mutation: 2%
2014 Tilch et al. [40] Australia 107 OncoCarta Assay Panel No activating mutations
2015 Cao et al. [46] China 50 ARMS assay No activating mutations
2015 Bemanian et al. [48] Norway 17 Direct sequencing T790M mutation: 12%

Abbreviations: TNBC, triple-negative breast cancer; IHC, Immunohistochemistry; FISH, fluorescence in situ hybridization; DISH, dual-color in situ hybridization;

primary breast cancer patients.

Many studies have been conducted to investigate the activating mutations in EGFR genes, but the results are controversial (Table 1). Santarpia et al. have documented that the EGFR-activating mutation (L858R) exists in 3.4% of TNBC (4 out of 116) [39]. However, several reports from Europe and Australia indicated that no activating mutations were identified in TNBC patient samples [31,40,41]. In contrast with the European and Australian studies, report from Asia by Teng et al. indicated the presence of activating mutations (4 of exon 19 deletion and 1 of L858R out of 70 TNBC samples) in a patient cohort in Singapore [42]. Furthermore, Lv et al. reported that two activating mutations, exon 19 deletion and L858R mutation, were detected in all tumor samples from a breast cancer patient cohort (N=143) in China [43] while several studies did not find any activating mutations in EGFR in Japanese and Chinese cohorts [44-46]. It is well known that the incidence of EGFR activating mutations in NSCLC is significantly higher in Asian population [47]. Although some studies have indicated that the incidence of EGFR mutations in TNBC may be related to ethnicity, it is clear that more studies will be required to demonstrate the significance of ethnicity in EGFR-activating mutation(s) in TNBC. A recent Norwegian breast cancer study revealed a 12% frequency (2 out of 17) of the T790M mutation in the tumors of TNBC patients [48], but because these tumors do not have primary activating mutations, there may not be any clinical significance.

Anti-EGFR therapies for treatment of various cancers

Specific anti-EGFR agents currently used in clinic include TKIs for NSCLC (gefitinib, erlotinib, afatinib, and osimertinib) and pancreatic cancer (erlotinib) as well as mAbs for CRC (cetuximab and panitumumab), head and neck squamous cell carcinoma (HNSCC) (cetuximab), and squamous cell lung cancer (necitumumab). EGFR TKIs bind to the ATP binding site of the EGFR tyrosine kinase to compete with ATP, thereby inhibiting EGFR kinase activity [49]. EGFR TKIs are currently approved for and highly effective against NSCLC with EGFR-activating mutations, e.g., exon 19 deletion and L858R mutation [50,51]. Although some patients with NSCLC with wild type EGFR gene amplification and wild type KRAS also respond to EGFR TKIs, the TKIs have yet to receive approval for those patient [52,53]. NSCLC patients with EGFR mutations dramatically respond to initial TKIs treatment but most patients eventually develop acquired resistance after long-term treatment. Consequently, understanding the mechanisms of EGFR TKI resistance in NSCLC is an important subject being studied extensively in preclinical and clinical studies. About half of acquired resistance to gefitinib and erlotinib is attributed to a secondary EGFR mutation in exon 20, T790M. A third-generation EGFR TKI, osimertinib, has demonstrated efficacy against EGFR with primary exon 19 deletion or L858R and secondary T790M mutations. Nonetheless, more recent studies have identified yet another mutation (C797S) contributing to the resistance of NSCLC to osimertinib [54]. A newer generation TKI was recently reported to inhibit EGFR with T790M and C797S mutations in NSCLC [55]. In addition to secondary mutations, there are many possible mechanisms of resistance that have been reported, including activation of alternative receptor kinases (c-Met, HER2, FGF and Axl overexpression), activation of downstream bypass signaling pathways (BRAF mutation, PTEN loss, NF-κB activation, etc.), and phenotypic changes (small-cell lung cancer transformation and epithelial-to-mesenchymal transition) [56-64]. Although these mechanisms partially explain TKI-resistance, the clinical significance of some of these mechanisms is not well established. Since activating mutation of EGFR in breast cancer is rare, it is uncertain whether some of the above-mentioned mechanisms in NSCLC are involved in the failure of clinical trials of TKI in TNBC.

EGFR mAbs bind to the ligand binding site on the cell surface EGFR in such a manner to compete with EGFR ligands, thereby inhibiting EGFR activation and dimerization [65]. Following antibody binding, cell surface EGFR undergoes internalization and subsequent degradation [66]. Interestingly, in addition to inhibition of EGFR signaling pathway, EGFR mAbs have been shown to induce antibody-mediated immune response to cancer cells, such as antibody-dependent cell-mediated cytotoxicity and T-cell-mediated immune response, which are essential in the efficacy of the EGFR mAb therapy [67,68]. Aside from EGFR-specific inhibitors, EGFR/HER2 dual kinase inhibitor lapatinib and VEGFR/EGFR/RET inhibitor vandetanib are also being used to treat patients with HER2 positive breast cancer and medullary thyroid cancer, respectively.

The mechanisms underlying EGFR mAb resistance have been widely studied in CRC. Mutant RAS, which is able to bypass the EGFR signaling pathway, is reported to be the primary mechanism driving resistance to EGFR mAbs. Consequently, EGFR mAbs are only used to treat CRC patients with wild type KRAS and NRAS tumors [69]. In addition to RAS mutations, various intrinsic resistance mechanisms have been reported, including activation of EGFR downstream signaling by activating BRAF and PIC3CA mutation, and PTEN deletion, as well as alternative receptor activation, such as HER2 and c-Met [70-73]. Furthermore, multiple mechanisms of acquired resistance, such as acquired mutations in KRAS, NRAS, and BRAF, and amplification of HER2 and c-Met genes have been reported [73-76]. Besides upregulation of EGFR downstream signaling, the mechanisms that affect mAb binding to EGFR have been identified, including the S492R mutation and arginine methylation (R198/200) in extracellular domain of EGFR [77,78]. As for predictive factors for therapeutic efficacy of EGFR mAb, so far, the status of RAS mutation is the only one used in the clinic. However, among nonresponders to EGFR mAb, only 30-40% of patients with CRC have KRAS mutations, suggesting that other mechanisms also play an important role in cetuximab response, and that identification of additional predictive markers is urgently needed. Although several clinical studies shown that increased EGFR gene copy number correlates with the clinical outcome of EGFR mAb therapy in CRC patients, the significance of EGFR gene copy number in the efficacy of EGFR mAb therapy remains controversial [79-81]. In TNBC patients, mutations in KRAS gene are rare (0-7.7%) [36,40,82,83], suggesting that other mechanisms may be involved in EGFR mAb sensitivity.

Anti-EGFR therapies in breast cancer clinical trials

Several clinical trials investigating the toxicity and efficacy of TKIs in breast cancer have been conducted, but the results thus far have been disappointing for both TKI monotherapy and in combination with chemotherapy (Table 2). Outcomes in phase II clinical trials of gefitinib and erlotinib as a monotherapy in metastatic and recurrent breast cancer demonstrated only a partial response (PR) of 0-3% [84-86]. The phase I clinical trial of erlotinib combined with chemotherapy (Bendamustine) for stage III and IV TNBC patients was conducted with results indicating an objective response rate (ORR) of 0% but with severe lymphopenia as an adverse effect [87]. A clinical trial to test the additive effect of gefitinib in preoperative chemotherapy in ER-negative breast cancer patients reported no significant difference in pathologic complete response (pCR) between patients treated with gefitinib (17%, 7 out of 41 patients) and placebo (12%, 5 out of 41 patients). Interestingly, however, a significant difference between TNBC and non-TNBC (P=0.03) was observed for gefitinib [88]. In a phase II trial of lapatinib monotherapy for patients with recurrent and anthracycline-refractory inflammatory breast cancer (IBC), the response rate was 50% among the 30 patients with HER2-positive tumors but only 7% among the 15 patients with HER2-negative IBC [89]. In a phase II trial with second-generation irreversible EGFR TKI afatinib in patients with metastatic TNBC, no objective responses were observed [90]. Currently, five EGFR-TKI clinical trials are ongoing in the United States, including TKI monotherapy and TKI in combination with chemotherapy as well as the combination of mTOR inhibitor, AMPK activator (Metformin), or an anti-VEGF mAb with chemotherapy.

Table 2.

Clinical studies of EGFR inhibitors in TNBC

Year Author Phase Regimen Outcome Patient Number and Population
2012 Bernsdorf et al. [88] II NAC: EC vs EC + Gefinitib ORR: No significance ER negative early BC (n=181)
2012 Schuler et al. [90] II Afanitib alone ORR: 0% Metastatic TNBC (n=29)
2013 Layman et al. [87] I Bendamustine and erlotinib ORR: 9% Metastatic TNBC (n=11)
2012 Carey et al. [91] II Carboplatin vs Carboplatin + Cetuximab ORR: 6% (Carb), 16% (Carb + cetux), TTP - 2.1 month Stage IV TNBC (n=102)
2013 Baselga et al. [92] II Cisplatin vs Cisplatin + Cetuximab ORR: 10% (cis), 20% (cis + cetux) P=0.032 Metastatic TNBC (n=115)
2014 Nabholtz et al. [95] II NAC: EFC100 + DOC100 + Panitumumab pCR rate 47% Operable TNBC (n=60)
2015 Tredan et al. [93] II Ixapepilone vs Ixabepilone + Cetuximab ORR: No significance Advanced/Metastatic TNBC (n=79)
2016 Nabholtz et al. [96] II NAC: Docetaxel + Cetuximab pCR: 24% Operable TNBC (n=33)
2016 Crozier et al. [94] II Irinotecane + Cetuximab ORR: 11% Metastatic TNBC (n=19)

Abbreviations: EGFR: epidermal growth factor receptor; NAC: neoadjuvant chemotherapy; EC: epirubicin and cyclophosphamide; EFC: epirubicin, fluorouracil and cyclophosphamide; DOC: docetaxel; ORR: overall response rate; TTP: time to progression; pCR: pathological complete response; PFS: progression-free survival; ER: estrogen receptor; BC: breast cancer; TNBC: triple negative breast cancer.

To date, 6 phase II clinical trials to investigate the efficacy and safety of anti-EGFR mAbs in patients with TNBC have been reported (Table 2). Carey et al. compared cetuximab and cetuximab plus carboplatin in a metastatic advanced recurrent breast cancer clinical trial [91]. The response rate (RR) was 6% with cetuximab alone, and 16% with cetuximab plus carboplatin. During the course of this study [91], a repeat biopsy at the tumor sites was carried out in 16 patients 1 week after the initiation of the treatment; EGFR activation was detected in tumor specimens from 13 patients, and the treatment-mediated inhibition of the EGFR pathway was observed in 5 of these patients. However, the PR outcome was obtained in only one treated patient. Meanwhile, because cetuximab failed to inhibit EGFR signaling in 72% (13 of 18) of the patients, the authors of the study suggested that an alternative pathway may be present. Baselga et al. reported that an RR of 20% in the cisplatin-cetuximab combination group and an RR of 10% in the cisplatin alone group in a clinical trial for advanced TNBC [92]. However, no statistically significant difference was found between the two treatment groups. Progression-free survival (PFS) was 1.5 months for the cisplatin alone group and 3.7 months for the cisplatin plus cetuximab group, showing an extension of 2.2 months. Similarly, Tredan et al. conducted a phase II trial of ixabepilone alone and ixabepilone plus cetuximab in patients with advanced/metastatic TNBC with results indicating no improvement in the RR or PFS [93]. Meanwhile, a slightly higher response rate to irinotecan and cetuximab was reported by Croziert et al. in patients with TNBC compared to one with other subtypes of breast cancer but the difference was not of statistical significance (TNBC 18% vs non-TNBC 0%: P=0.49) [94].

Two studies have investigated EGFR mAbs in neoadjuvant setting in operable TNBC patients [95,96]. One study examined the standard FEC (5-fluorouracil, epidoxorubicin, and cyclophosphamide) therapy and preoperative chemotherapy with docetaxel combined with panitumumab in TNBC patients, and the other cetuximab combined with docetaxel. Both of those single-arm studies reported modest activities of the therapies administered [95,96]. Interestingly, the studies also showed that high CD8+ tumor-infiltrating lymphocytes (TILs) in the tumor microenvironment can predict response to EGFR mAb neoadjuvant therapy. Although it has been known that the status of TILs is a general prognostic factor in cancer, including TNBC [97], it may be particularly critical for EGFR mAb therapies since T-cell-mediated immune response plays an essential role in the efficacy of the EGFR mAb therapy [67]. Overall, in TNBC, the outcomes of clinical trials of EGFR mAbs seem slightly better than the ones of EGFR TKIs. In addition to the above-mentioned trials, there are currently 3 ongoing clinical trials evaluating anti EGFR-mAbs combined with chemotherapy in TNBC (Table 3).

Table 3.

Ongoing clinical trials of EGFR inhibitors in breast cancer

Phase Intervention Target NCT Number Patient Number and Disease
I Metformin AMPK etc. NCT01650506 8 (TNBC)
Erlotinib TKI (1st)
II Paclitaxel Chemo NCT02511847 40 (TNBC)
Afatinib TKI (2nd)
II Icotinib EGFR TKI NCT02362230 67 (TNBC)
II Nab-paclitaxel Chemo NCT00733408 63 (TNBC)
Erlotinib EGFR TKI
Bevacizumab VGEF mAb
II Ixabepilone Chemo NCT01097642 40 (TNBC)
Cetuximab EGFR mAb
II Nab-paclitaxel Chemo NCT01036087 40 (IBC, include TNBC)
Carboplatin Chemo
5-Fluorouracil Chemo
Epirubicin Chemo
Cyclophosphamide Chemo
Panitumumab EGFR mAb
II Docetaxel Chemo NCT01939054 90 (TNBC)
Capecitabine Chemo
Nimotuzumab EGFR mAb

Abbreviations: EGFR, epidermal growth factor receptor; Chemo, Chemotherapy; TKI, tyrosine kinase inhibitor; mAb, monoclonal antibody; mTORi, mammalian target of rapamycin inhibitor; VEGF, vascular endothelial growth factor; NCT, national clinical trial; TNBC, triple negative breast cancer; IBC, inflammatory breast cancer.

Conclusions and future directions

TNBC patients initially respond to conventional chemotherapy, but the disease frequently relapses and leads to worse outcome than patients with other subtypes of breast cancer. Therefore, effective therapeutic strategies for TNBC are urgently needed. A significant number of TNBC is associated with EGFR overexpression, and EGFR-targeted therapies, including TKIs and mAbs, are already available to treat various types of cancers, making EGFR inhibitors attractive options for treatment of TNBC patients. While results from clinical trials of EGFR TKI have been disappointing for breast cancer, a small portion of patients may still respond to EGFR inhibitors. It is unfortunate that some breast cancer patients who actually respond to EGFR inhibitors cannot benefit from the therapies already used in clinic due to the low overall response rate of the entire population of breast cancer patients in the clinical trials. Thus, it would be crucial to identify the subpopulation of EGFR-overexpressing breast cancer patients that will respond to treatment with EGFR inhibitors so that they can benefit immediately from existing drugs without waiting for development of new drugs that usually takes over a decade.

One plausible reason for the lack of response to current targeted therapies is that most TNBCs are not exclusively dependent on EGFR signaling for their survival. EGFR TKIs are effective for NSCLC with activating mutations in EGFR, whose survival is largely dependent on EGFR. Although EGFR activating mutations in TNBC are rare, these tumors with EGFR activating mutations may still respond to EGFR TKIs via the aforementioned mechanisms of resistance for EGFR inhibitors other than secondary mutations, including activation of alternative receptors and pathways. Because some patients with NSCLC with wild type EGFR gene amplification and wild type KRAS demonstrate response to EGFR TKIs [52,53], these alternative resistant pathways may need to be blocked in wild type EGFR-overexpressing TNBC to increase efficacy. Identification of the biomarkers that are associated with the mechanisms of resistance and potential combination of EGFR TKIs and other inhibitors that attenuate these mechanisms are required.

For anti-EGFR mAbs, certain levels of therapeutic effects have been reported in some studies, but the efficacy has not been satisfactory. Similar to TKIs, it is necessary to dissect the potential pathways associated with intrinsic resistance to EGFR mAbs. Recently, we have reported that extracellular domain of EGFR is methylated by protein arginine methyltransfrase 1 (PRMT1), and that EGFR methylation is involved in mAb resistance in CRC [77]. It would be of interest to determine whether EGFR methylation also plays a role in primary resistance to EGFR mAbs in TNBC. If so, EGFR methylation may help stratify patients to maximize response to EGFR mAb. Inhibitors against PRMT1 are currently under development [98], and the combination of PRMT1 inhibitors and EGFR mAbs may be the effective therapeutic strategy for TNBC with high levels of EGFR methylation.

In addition, EGFR inhibitors may be combined other types of targeted drugs. Currently, the most promising clinical target for TNBC is PARP. Inhibitors against PARP are known to induce synthetic lethal effects in cancer cells with BRCA mutations [99]. There are multiple PARP inhibitors that are in clinical trials, and one PARP inhibitor, olaparib, has been approved for the treatment of BRCA-deficient metastatic ovarian cancer. Interestingly, inhibition of EGFR was reported to sensitize TNBC and HNSCC cells to PARP inhibitors [100,101]. Moreover, we recently demonstrated that PARP is directly phosphorylated by c-Met kinase, and that the PARP phosphorylation is involved in PARP inhibitor resistance [102]. Thus, EGFR may also be involved in PARP inhibitor resistance in TNBC, and the combination of EGFR inhibitors and PARP inhibitors may be an effective therapeutic strategy against TNBC.

Finally, immune checkpoint inhibitors such as PD-1 and CTLA-4 antibodies have drawn considerable attention in cancer treatment development due to the outstanding efficacy in some cancers, such as melanoma and lung cancer [103]. There are multiple trials of immune checkpoint inhibitors in TNBC ongoing. Given that T-cell-mediated immune response is associated with EGFR mAb efficacy [68], the combination of EGFR mAb and immune checkpoint inhibitors may be promising therapeutic approaches for TNBC with high EGFR copy numbers.

Acknowledgements

This work was funded in part by the following: Cancer Prevention and Research Institute of Texas (RP150245); National Breast Cancer Foundation, Inc. (to M.-C.H.).

Disclosure of conflict of interest

None.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
  • 2.Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–1948. doi: 10.1056/NEJMra1001389. [DOI] [PubMed] [Google Scholar]
  • 3.Liedtke C, Mazouni C, Hess KR, Andre F, Tordai A, Mejia JA, Symmans WF, Gonzalez-Angulo AM, Hennessy B, Green M, Cristofanilli M, Hortobagyi GN, Pusztai L. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2008;26:1275–1281. doi: 10.1200/JCO.2007.14.4147. [DOI] [PubMed] [Google Scholar]
  • 4.Carey LA, Dees EC, Sawyer L, Gatti L, Moore DT, Collichio F, Ollila DW, Sartor CI, Graham ML, Perou CM. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res. 2007;13:2329–2334. doi: 10.1158/1078-0432.CCR-06-1109. [DOI] [PubMed] [Google Scholar]
  • 5.Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  • 6.Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lonning PE, Borresen-Dale AL. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dai X, Li T, Bai Z, Yang Y, Liu X, Zhan J, Shi B. Breast cancer intrinsic subtype classification, clinical use and future trends. Am J Cancer Res. 2015;5:2929–2943. [PMC free article] [PubMed] [Google Scholar]
  • 8.Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750–2767. doi: 10.1172/JCI45014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Foulkes WD, Stefansson IM, Chappuis PO, Begin LR, Goffin JR, Wong N, Trudel M, Akslen LA. Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst. 2003;95:1482–1485. doi: 10.1093/jnci/djg050. [DOI] [PubMed] [Google Scholar]
  • 10.Burgess M, Puhalla S. BRCA 1/2-Mutation Related and Sporadic Breast and Ovarian Cancers: More Alike than Different. Front Oncol. 2014;4:19. doi: 10.3389/fonc.2014.00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Reis-Filho JS, Tutt AN. Triple negative tumours: a critical review. Histopathology. 2008;52:108–118. doi: 10.1111/j.1365-2559.2007.02889.x. [DOI] [PubMed] [Google Scholar]
  • 12.Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol. 2009;21:177–184. doi: 10.1016/j.ceb.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 13.Yarden Y. The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8. doi: 10.1016/s0959-8049(01)00230-1. [DOI] [PubMed] [Google Scholar]
  • 14.Burgess AW. EGFR family: structure physiology signalling and therapeutic targets. Growth Factors. 2008;26:263–274. doi: 10.1080/08977190802312844. [DOI] [PubMed] [Google Scholar]
  • 15.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  • 16.Lee HH, Wang YN, Hung MC. Non-canonical signaling mode of the epidermal growth factor receptor family. Am J Cancer Res. 2015;5:2944–2958. [PMC free article] [PubMed] [Google Scholar]
  • 17.Rimawi MF, Shetty PB, Weiss HL, Schiff R, Osborne CK, Chamness GC, Elledge RM. Epidermal growth factor receptor expression in breast cancer association with biologic phenotype and clinical outcomes. Cancer. 2010;116:1234–1242. doi: 10.1002/cncr.24816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Shigematsu H, Gazdar AF. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. Int J Cancer. 2006;118:257–262. doi: 10.1002/ijc.21496. [DOI] [PubMed] [Google Scholar]
  • 19.Gan HK, Kaye AH, Luwor RB. The EGFRvIII variant in glioblastoma multiforme. J Clin Neurosci. 2009;16:748–754. doi: 10.1016/j.jocn.2008.12.005. [DOI] [PubMed] [Google Scholar]
  • 20.Bronte G, Terrasi M, Rizzo S, Sivestris N, Ficorella C, Cajozzo M, Di Gaudio F, Gulotta G, Siragusa S, Gebbia N, Russo A. EGFR genomic alterations in cancer: prognostic and predictive values. Front Biosci (Elite Ed) 2011;3:879–887. doi: 10.2741/e296. [DOI] [PubMed] [Google Scholar]
  • 21.Hanawa M, Suzuki S, Dobashi Y, Yamane T, Kono K, Enomoto N, Ooi A. EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus. Int J Cancer. 2006;118:1173–1180. doi: 10.1002/ijc.21454. [DOI] [PubMed] [Google Scholar]
  • 22.Zhang J, Antonyak MA, Singh G, Cerione RA. A mechanism for the upregulation of EGF receptor levels in glioblastomas. Cell Rep. 2013;3:2008–2020. doi: 10.1016/j.celrep.2013.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vivanco I, Rohle D, Versele M, Iwanami A, Kuga D, Oldrini B, Tanaka K, Dang J, Kubek S, Palaskas N, Hsueh T, Evans M, Mulholland D, Wolle D, Rajasekaran S, Rajasekaran A, Liau LM, Cloughesy TF, Dikic I, Brennan C, Wu H, Mischel PS, Perera T, Mellinghoff IK. The phosphatase and tensin homolog regulates epidermal growth factor receptor (EGFR) inhibitor response by targeting EGFR for degradation. Proc Natl Acad Sci U S A. 2010;107:6459–6464. doi: 10.1073/pnas.0911188107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Burga LN, Hu H, Juvekar A, Tung NM, Troyan SL, Hofstatter EW, Wulf GM. Loss of BRCA1 leads to an increase in epidermal growth factor receptor expression in mammary epithelial cells, and epidermal growth factor receptor inhibition prevents estrogen receptor-negative cancers in BRCA1-mutant mice. Breast Cancer Res. 2011;13:R30. doi: 10.1186/bcr2850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Li D, Bi FF, Cao JM, Cao C, Li CY, Yang Q. Effect of BRCA1 on epidermal growth factor receptor in ovarian cancer. J Exp Clin Cancer Res. 2013;32:102. doi: 10.1186/1756-9966-32-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004;10:5367–5374. doi: 10.1158/1078-0432.CCR-04-0220. [DOI] [PubMed] [Google Scholar]
  • 27.Choi J, Jung WH, Koo JS. Clinicopathologic features of molecular subtypes of triple negative breast cancer based on immunohistochemical markers. Histol Histopathol. 2012;27:1481–1493. doi: 10.14670/HH-27.1481. [DOI] [PubMed] [Google Scholar]
  • 28.Viale G, Rotmensz N, Maisonneuve P, Bottiglieri L, Montagna E, Luini A, Veronesi P, Intra M, Torrisi R, Cardillo A, Campagnoli E, Goldhirsch A, Colleoni M. Invasive ductal carcinoma of the breast with the “triple-negative” phenotype: prognostic implications of EGFR immunoreactivity. Breast Cancer Res Treat. 2009;116:317–328. doi: 10.1007/s10549-008-0206-z. [DOI] [PubMed] [Google Scholar]
  • 29.Rakha EA, El-Sayed ME, Green AR, Lee AH, Robertson JF, Ellis IO. Prognostic markers in triple-negative breast cancer. Cancer. 2007;109:25–32. doi: 10.1002/cncr.22381. [DOI] [PubMed] [Google Scholar]
  • 30.Tan DS, Marchio C, Jones RL, Savage K, Smith IE, Dowsett M, Reis-Filho JS. Triple negative breast cancer: molecular profiling and prognostic impact in adjuvant anthracycline-treated patients. Breast Cancer Res Treat. 2008;111:27–44. doi: 10.1007/s10549-007-9756-8. [DOI] [PubMed] [Google Scholar]
  • 31.Martin V, Botta F, Zanellato E, Molinari F, Crippa S, Mazzucchelli L, Frattini M. Molecular characterization of EGFR and EGFR-downstream pathways in triple negative breast carcinomas with basal like features. Histol Histopathol. 2012;27:785–792. doi: 10.14670/HH-27.785. [DOI] [PubMed] [Google Scholar]
  • 32.Meseure D, Vacher S, Drak Alsibai K, Trassard M, Susini A, Le Ray C, Lerebours F, Le Scodan R, Spyratos F, Marc Guinebretiere J, Lidereau R, Bieche I. Profiling of EGFR mRNA and protein expression in 471 breast cancers compared with 10 normal tissues: a candidate biomarker to predict EGFR inhibitor effectiveness. Int J Cancer. 2012;131:1009–1010. doi: 10.1002/ijc.26434. [DOI] [PubMed] [Google Scholar]
  • 33.Liu D, He J, Yuan Z, Wang S, Peng R, Shi Y, Teng X, Qin T. EGFR expression correlates with decreased disease-free survival in triple-negative breast cancer: a retrospective analysis based on a tissue microarray. Med Oncol. 2012;29:401–405. doi: 10.1007/s12032-011-9827-x. [DOI] [PubMed] [Google Scholar]
  • 34.Park HS, Jang MH, Kim EJ, Kim HJ, Lee HJ, Kim YJ, Kim JH, Kang E, Kim SW, Kim IA, Park SY. High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod Pathol. 2014;27:1212–1222. doi: 10.1038/modpathol.2013.251. [DOI] [PubMed] [Google Scholar]
  • 35.Gumuskaya B, Alper M, Hucumenoglu S, Altundag K, Uner A, Guler G. EGFR expression and gene copy number in triple-negative breast carcinoma. Cancer Genet Cytogenet. 2010;203:222–229. doi: 10.1016/j.cancergencyto.2010.07.118. [DOI] [PubMed] [Google Scholar]
  • 36.Grob TJ, Heilenkotter U, Geist S, Paluchowski P, Wilke C, Jaenicke F, Quaas A, Wilczak W, Choschzick M, Sauter G, Lebeau A. Rare oncogenic mutations of predictive markers for targeted therapy in triple-negative breast cancer. Breast Cancer Res Treat. 2012;134:561–567. doi: 10.1007/s10549-012-2092-7. [DOI] [PubMed] [Google Scholar]
  • 37.Pintens S, Neven P, Drijkoningen M, Van Belle V, Moerman P, Christiaens MR, Smeets A, Wildiers H, Vanden Bempt I. Triple negative breast cancer: a study from the point of view of basal CK5/6 and HER-1. J Clin Pathol. 2009;62:624–628. doi: 10.1136/jcp.2008.061358. [DOI] [PubMed] [Google Scholar]
  • 38.Shao MM, Zhang F, Meng G, Wang XX, Xu H, Yu XW, Chen LY, Tse GM. Epidermal growth factor receptor gene amplification and protein overexpression in basal-like carcinoma of the breast. Histopathology. 2011;59:264–273. doi: 10.1111/j.1365-2559.2011.03921.x. [DOI] [PubMed] [Google Scholar]
  • 39.Santarpia L, Qi Y, Stemke-Hale K, Wang B, Young EJ, Booser DJ, Holmes FA, O’Shaughnessy J, Hellerstedt B, Pippen J, Vidaurre T, Gomez H, Valero V, Hortobagyi GN, Symmans WF, Bottai G, Di Leo A, Gonzalez-Angulo AM, Pusztai L. Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers. Breast Cancer Res Treat. 2012;134:333–343. doi: 10.1007/s10549-012-2035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tilch E, Seidens T, Cocciardi S, Reid LE, Byrne D, Simpson PT, Vargas AC, Cummings MC, Fox SB, Lakhani SR, Chenevix Trench G. Mutations in EGFR, BRAF and RAS are rare in triple-negative and basal-like breast cancers from Caucasian women. Breast Cancer Res Treat. 2014;143:385–392. doi: 10.1007/s10549-013-2798-1. [DOI] [PubMed] [Google Scholar]
  • 41.Jacot W, Lopez-Crapez E, Thezenas S, Senal R, Fina F, Bibeau F, Romieu G, Lamy PJ. Lack of EGFR-activating mutations in European patients with triple-negative breast cancer could emphasise geographic and ethnic variations in breast cancer mutation profiles. Breast Cancer Res. 2011;13:R133. doi: 10.1186/bcr3079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Teng YH, Tan WJ, Thike AA, Cheok PY, Tse GM, Wong NS, Yip GW, Bay BH, Tan PH. Mutations in the epidermal growth factor receptor (EGFR) gene in triple negative breast cancer: possible implications for targeted therapy. Breast Cancer Res. 2011;13:R35. doi: 10.1186/bcr2857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lv N, Lin S, Xie Z, Tang J, Ge Q, Wu M, Xie X, Wei W. Absence of evidence for epidermal growth factor receptor and human homolog of the Kirsten rat sarcoma-2 virus oncogene mutations in breast cancer. Cancer Epidemiol. 2012;36:341–346. doi: 10.1016/j.canep.2012.01.004. [DOI] [PubMed] [Google Scholar]
  • 44.Toyama T, Yamashita H, Kondo N, Okuda K, Takahashi S, Sasaki H, Sugiura H, Iwase H, Fujii Y. Frequently increased epidermal growth factor receptor (EGFR) copy numbers and decreased BRCA1 mRNA expression in Japanese triple-negative breast cancers. BMC Cancer. 2008;8:309. doi: 10.1186/1471-2407-8-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nakajima H, Ishikawa Y, Furuya M, Sano T, Ohno Y, Horiguchi J, Oyama T. Protein expression, gene amplification, and mutational analysis of EGFR in triple-negative breast cancer. Breast Cancer. 2014;21:66–74. doi: 10.1007/s12282-012-0354-1. [DOI] [PubMed] [Google Scholar]
  • 46.Cao WM, Gao Y, Wang XJ. Lack of epidermal growth factor receptor (EGFR)-activating mutations in triple-negative breast cancer in China. Breast Cancer Res. 2015;17:115. doi: 10.1186/s13058-015-0628-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Janne PA, Engelman JA, Johnson BE. Epidermal growth factor receptor mutations in non-small-cell lung cancer: implications for treatment and tumor biology. J. Clin. Oncol. 2005;23:3227–3234. doi: 10.1200/JCO.2005.09.985. [DOI] [PubMed] [Google Scholar]
  • 48.Bemanian V, Sauer T, Touma J, Lindstedt BA, Chen Y, Odegard HP, Vetvik KM, Bukholm IR, Geisler J. The epidermal growth factor receptor (EGFR/HER-1) gatekeeper mutation T790M is present in European patients with early breast cancer. PLoS One. 2015;10:e0134398. doi: 10.1371/journal.pone.0134398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Carey KD, Garton AJ, Romero MS, Kahler J, Thomson S, Ross S, Park F, Haley JD, Gibson N, Sliwkowski MX. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer Res. 2006;66:8163–8171. doi: 10.1158/0008-5472.CAN-06-0453. [DOI] [PubMed] [Google Scholar]
  • 50.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 51.Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–1500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
  • 52.Cappuzzo F, Finocchiaro G, Grossi F, Bidoli P, Favaretto A, Marchetti A, Valente ML, Cseh A, Clementi L, Massey D, Santoro A. Phase II study of afatinib, an irreversible ErbB family blocker, in EGFR FISH-positive non-small-cell lung cancer. J Thorac Oncol. 2015;10:665–672. doi: 10.1097/JTO.0000000000000442. [DOI] [PubMed] [Google Scholar]
  • 53.Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, Liu N, Zhang T, Marrano P, Whitehead M, Squire JA, Kamel-Reid S, Seymour L, Shepherd FA, Tsao MS. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR. 21. J. Clin. Oncol. 2008;26:4268–4275. doi: 10.1200/JCO.2007.14.8924. [DOI] [PubMed] [Google Scholar]
  • 54.Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D, Matthews SE, Cantarini M, Barrett JC, Janne PA, Oxnard GR. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med. 2015;21:560–562. doi: 10.1038/nm.3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jia Y, Yun CH, Park E, Ercan D, Manuia M, Juarez J, Xu C, Rhee K, Chen T, Zhang H, Palakurthi S, Jang J, Lelais G, DiDonato M, Bursulaya B, Michellys PY, Epple R, Marsilje TH, McNeill M, Lu W, Harris J, Bender S, Wong KK, Janne PA, Eck MJ. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature. 2016;534:129–132. doi: 10.1038/nature17960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–1043. doi: 10.1126/science.1141478. [DOI] [PubMed] [Google Scholar]
  • 57.Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, Ohashi K, Janjigian YY, Spitzler PJ, Melnick MA, Riely GJ, Kris MG, Miller VA, Ladanyi M, Politi K, Pao W. 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;2:922–933. doi: 10.1158/2159-8290.CD-12-0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Terai H, Soejima K, Yasuda H, Nakayama S, Hamamoto J, Arai D, Ishioka K, Ohgino K, Ikemura S, Sato T, Yoda S, Satomi R, Naoki K, Betsuyaku T. Activation of the FGF2-FGFR1 autocrine pathway: a novel mechanism of acquired resistance to gefitinib in NSCLC. Mol Cancer Res. 2013;11:759–767. doi: 10.1158/1541-7786.MCR-12-0652. [DOI] [PubMed] [Google Scholar]
  • 59.Ohashi K, Sequist LV, Arcila ME, Moran T, Chmielecki J, Lin YL, Pan Y, Wang L, de Stanchina E, Shien K, Aoe K, Toyooka S, Kiura K, Fernandez-Cuesta L, Fidias P, Yang JC, Miller VA, Riely GJ, Kris MG, Engelman JA, Vnencak-Jones CL, Dias-Santagata D, Ladanyi M, Pao W. Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. Proc Natl Acad Sci U S A. 2012;109:E2127–2133. doi: 10.1073/pnas.1203530109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bivona TG, Hieronymus H, Parker J, Chang K, Taron M, Rosell R, Moonsamy P, Dahlman K, Miller VA, Costa C, Hannon G, Sawyers CL. FAS and NF-kappaB signalling modulate dependence of lung cancers on mutant EGFR. Nature. 2011;471:523–526. doi: 10.1038/nature09870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T, Abdel-Rahman M, Wang X, Levine AD, Rho JK, Choi YJ, Choi CM, Kim SW, Jang SJ, Park YS, Kim WS, Lee DH, Lee JS, Miller VA, Arcila M, Ladanyi M, Moonsamy P, Sawyers C, Boggon TJ, Ma PC, Costa C, Taron M, Rosell R, Halmos B, Bivona TG. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 2012;44:852–860. doi: 10.1038/ng.2330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sos ML, Koker M, Weir BA, Heynck S, Rabinovsky R, Zander T, Seeger JM, Weiss J, Fischer F, Frommolt P, Michel K, Peifer M, Mermel C, Girard L, Peyton M, Gazdar AF, Minna JD, Garraway LA, Kashkar H, Pao W, Meyerson M, Thomas RK. PTEN loss contributes to erlotinib resistance in EGFR-mutant lung cancer by activation of Akt and EGFR. Cancer Res. 2009;69:3256–3261. doi: 10.1158/0008-5472.CAN-08-4055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, Shen L, Fan YH, Giri U, Tumula P, Nilsson MB, Gudikote J, Tran HT, Cardnell RJ, Bearss DJ, Warner SL, Foulks JM, Kanner SB, Gandhi V, Krett NL, Rosen ST, Kim ES, Herbst RS, Blumenschein GR, Lee JJ, Lippman SM, Ang KK, Mills GB, Hong WK, Weinstein JN, Wistuba II, Coombes KR, Minna JD, Heymach JV. An epithelial-mesenchymal transition (EMT) gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res. 2013;19:279–90. doi: 10.1158/1078-0432.CCR-12-1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zakowski MF, Ladanyi M, Kris MG. EGFR mutations in small-cell lung cancers in patients who have never smoked. N Engl J Med. 2006;355:213–215. doi: 10.1056/NEJMc053610. [DOI] [PubMed] [Google Scholar]
  • 65.Kim ES, Khuri FR, Herbst RS. Epidermal growth factor receptor biology (IMC-C225) Curr Opin Oncol. 2001;13:506–513. doi: 10.1097/00001622-200111000-00014. [DOI] [PubMed] [Google Scholar]
  • 66.Doody JF, Wang Y, Patel SN, Joynes C, Lee SP, Gerlak J, Rolser RL, Li Y, Steiner P, Bassi R, Hicklin DJ, Hadari YR. Inhibitory activity of cetuximab on epidermal growth factor receptor mutations in non small cell lung cancers. Mol Cancer Ther. 2007;6:2642–2651. doi: 10.1158/1535-7163.MCT-06-0506. [DOI] [PubMed] [Google Scholar]
  • 67.Trivedi S, Jie HB, Ferris RL. Tumor antigen-specific monoclonal antibodies and induction of T-cell immunity. Semin Oncol. 2014;41:678–684. doi: 10.1053/j.seminoncol.2014.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yang X, Zhang X, Mortenson ED, Radkevich-Brown O, Wang Y, Fu YX. Cetuximab-mediated tumor regression depends on innate and adaptive immune responses. Mol Ther. 2013;21:91–100. doi: 10.1038/mt.2012.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, Schilsky RL. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J. Clin. Oncol. 2016;34:179–185. doi: 10.1200/JCO.2015.63.9674. [DOI] [PubMed] [Google Scholar]
  • 70.Yonesaka K, Zejnullahu K, Okamoto I, Satoh T, Cappuzzo F, Souglakos J, Ercan D, Rogers A, Roncalli M, Takeda M, Fujisaka Y, Philips J, Shimizu T, Maenishi O, Cho Y, Sun J, Destro A, Taira K, Takeda K, Okabe T, Swanson J, Itoh H, Takada M, Lifshits E, Okuno K, Engelman JA, Shivdasani RA, Nishio K, Fukuoka M, Varella-Garcia M, Nakagawa K, Janne PA. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci Transl Med. 2011;3:99ra86. doi: 10.1126/scitranslmed.3002442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Frattini M, Saletti P, Romagnani E, Martin V, Molinari F, Ghisletta M, Camponovo A, Etienne LL, Cavalli F, Mazzucchelli L. PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients. Br J Cancer. 2007;97:1139–1145. doi: 10.1038/sj.bjc.6604009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P, Penault-Llorca F, Rougier P, Vincenzi B, Santini D, Tonini G, Cappuzzo F, Frattini M, Molinari F, Saletti P, De Dosso S, Martini M, Bardelli A, Siena S, Sartore-Bianchi A, Tabernero J, Macarulla T, Di Fiore F, Gangloff AO, Ciardiello F, Pfeiffer P, Qvortrup C, Hansen TP, Van Cutsem E, Piessevaux H, Lambrechts D, Delorenzi M, Tejpar S. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753–762. doi: 10.1016/S1470-2045(10)70130-3. [DOI] [PubMed] [Google Scholar]
  • 73.Bardelli A, Corso S, Bertotti A, Hobor S, Valtorta E, Siravegna G, Sartore-Bianchi A, Scala E, Cassingena A, Zecchin D, Apicella M, Migliardi G, Galimi F, Lauricella C, Zanon C, Perera T, Veronese S, Corti G, Amatu A, Gambacorta M, Diaz LA Jr, Sausen M, Velculescu VE, Comoglio P, Trusolino L, Di Nicolantonio F, Giordano S, Siena S. Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013;3:658–673. doi: 10.1158/2159-8290.CD-12-0558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, Kinzler KW, Oliner KS, Vogelstein B. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540. doi: 10.1038/nature11219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, Cora D, Di Nicolantonio F, Buscarino M, Petti C, Ribero D, Russolillo N, Muratore A, Massucco P, Pisacane A, Molinaro L, Valtorta E, Sartore-Bianchi A, Risio M, Capussotti L, Gambacorta M, Siena S, Medico E, Sapino A, Marsoni S, Comoglio PM, Bardelli A, Trusolino L. A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 2011;1:508–523. doi: 10.1158/2159-8290.CD-11-0109. [DOI] [PubMed] [Google Scholar]
  • 76.Misale S, Arena S, Lamba S, Siravegna G, Lallo A, Hobor S, Russo M, Buscarino M, Lazzari L, Sartore-Bianchi A, Bencardino K, Amatu A, Lauricella C, Valtorta E, Siena S, Di Nicolantonio F, Bardelli A. Blockade of EGFR and MEK intercepts heterogeneous mechanisms of acquired resistance to anti-EGFR therapies in colorectal cancer. Sci Transl Med. 2014;6:224ra226. doi: 10.1126/scitranslmed.3007947. [DOI] [PubMed] [Google Scholar]
  • 77.Liao HW, Hsu JM, Xia W, Wang HL, Wang YN, Chang WC, Arold ST, Chou CK, Tsou PH, Yamaguchi H, Fang YF, Lee HJ, Lee HH, Tai SK, Yang MH, Morelli MP, Sen M, Ladbury JE, Chen CH, Grandis JR, Kopetz S, Hung MC. PRMT1-mediated methylation of the EGF receptor regulates signaling and cetuximab response. J Clin Invest. 2015;125:4529–4543. doi: 10.1172/JCI82826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Montagut C, Dalmases A, Bellosillo B, Crespo M, Pairet S, Iglesias M, Salido M, Gallen M, Marsters S, Tsai SP, Minoche A, Seshagiri S, Serrano S, Himmelbauer H, Bellmunt J, Rovira A, Settleman J, Bosch F, Albanell J. Identification of a mutation in the extracellular domain of the Epidermal Growth Factor Receptor conferring cetuximab resistance in colorectal cancer. Nat Med. 2012;18:221–223. doi: 10.1038/nm.2609. [DOI] [PubMed] [Google Scholar]
  • 79.Sartore-Bianchi A, Moroni M, Veronese S, Carnaghi C, Bajetta E, Luppi G, Sobrero A, Barone C, Cascinu S, Colucci G, Cortesi E, Nichelatti M, Gambacorta M, Siena S. Epidermal growth factor receptor gene copy number and clinical outcome of metastatic colorectal cancer treated with panitumumab. J. Clin. Oncol. 2007;25:3238–3245. doi: 10.1200/JCO.2007.11.5956. [DOI] [PubMed] [Google Scholar]
  • 80.Moroni M, Veronese S, Benvenuti S, Marrapese G, Sartore-Bianchi A, Di Nicolantonio F, Gambacorta M, Siena S, Bardelli A. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol. 2005;6:279–286. doi: 10.1016/S1470-2045(05)70102-9. [DOI] [PubMed] [Google Scholar]
  • 81.Personeni N, Fieuws S, Piessevaux H, De Hertogh G, De Schutter J, Biesmans B, De Roock W, Capoen A, Debiec-Rychter M, Van Laethem JL, Peeters M, Humblet Y, Van Cutsem E, Tejpar S. Clinical usefulness of EGFR gene copy number as a predictive marker in colorectal cancer patients treated with cetuximab: a fluorescent in situ hybridization study. Clin Cancer Res. 2008;14:5869–5876. doi: 10.1158/1078-0432.CCR-08-0449. [DOI] [PubMed] [Google Scholar]
  • 82.Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili G, Ding J, Tse K, Haffari G, Bashashati A, Prentice LM, Khattra J, Burleigh A, Yap D, Bernard V, McPherson A, Shumansky K, Crisan A, Giuliany R, Heravi-Moussavi A, Rosner J, Lai D, Birol I, Varhol R, Tam A, Dhalla N, Zeng T, Ma K, Chan SK, Griffith M, Moradian A, Cheng SW, Morin GB, Watson P, Gelmon K, Chia S, Chin SF, Curtis C, Rueda OM, Pharoah PD, Damaraju S, Mackey J, Hoon K, Harkins T, Tadigotla V, Sigaroudinia M, Gascard P, Tlsty T, Costello JF, Meyer IM, Eaves CJ, Wasserman WW, Jones S, Huntsman D, Hirst M, Caldas C, Marra MA, Aparicio S. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486:395–399. doi: 10.1038/nature10933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pereira CB, Leal MF, de Souza CR, Montenegro RC, Rey JA, Carvalho AA, Assumpcao PP, Khayat AS, Pinto GR, Demachki S, de Arruda Cardoso Smith M, Burbano RR. Prognostic and predictive significance of MYC and KRAS alterations in breast cancer from women treated with neoadjuvant chemotherapy. PLoS One. 2013;8:e60576. doi: 10.1371/journal.pone.0060576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.von Minckwitz G, Jonat W, Fasching P, du Bois A, Kleeberg U, Luck HJ, Kettner E, Hilfrich J, Eiermann W, Torode J, Schneeweiss A. A multicentre phase II study on gefitinib in taxane- and anthracycline-pretreated metastatic breast cancer. Breast Cancer Res Treat. 2005;89:165–172. doi: 10.1007/s10549-004-1720-2. [DOI] [PubMed] [Google Scholar]
  • 85.Baselga J, Albanell J, Ruiz A, Lluch A, Gascon P, Guillem V, Gonzalez S, Sauleda S, Marimon I, Tabernero JM, Koehler MT, Rojo F. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J. Clin. Oncol. 2005;23:5323–5333. doi: 10.1200/JCO.2005.08.326. [DOI] [PubMed] [Google Scholar]
  • 86.Dickler MN, Cobleigh MA, Miller KD, Klein PM, Winer EP. Efficacy and safety of erlotinib in patients with locally advanced or metastatic breast cancer. Breast Cancer Res Treat. 2009;115:115–121. doi: 10.1007/s10549-008-0055-9. [DOI] [PubMed] [Google Scholar]
  • 87.Layman RM, Ruppert AS, Lynn M, Mrozek E, Ramaswamy B, Lustberg MB, Wesolowski R, Ottman S, Carothers S, Bingman A, Reinbolt R, Kraut EH, Shapiro CL. Severe and prolonged lymphopenia observed in patients treated with bendamustine and erlotinib for metastatic triple negative breast cancer. Cancer Chemother Pharmacol. 2013;71:1183–1190. doi: 10.1007/s00280-013-2112-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bernsdorf M, Ingvar C, Jorgensen L, Tuxen MK, Jakobsen EH, Saetersdal A, Kimper-Karl ML, Kroman N, Balslev E, Ejlertsen B. Effect of adding gefitinib to neoadjuvant chemotherapy in estrogen receptor negative early breast cancer in a randomized phase II trial. Breast Cancer Res Treat. 2011;126:463–470. doi: 10.1007/s10549-011-1352-2. [DOI] [PubMed] [Google Scholar]
  • 89.Johnston S, Trudeau M, Kaufman B, Boussen H, Blackwell K, LoRusso P, Lombardi DP, Ben Ahmed S, Citrin DL, DeSilvio ML, Harris J, Westlund RE, Salazar V, Zaks TZ, Spector NL. Phase II study of predictive biomarker profiles for response targeting human epidermal growth factor receptor 2 (HER-2) in advanced inflammatory breast cancer with lapatinib monotherapy. J. Clin. Oncol. 2008;26:1066–1072. doi: 10.1200/JCO.2007.13.9949. [DOI] [PubMed] [Google Scholar]
  • 90.Schuler M, Awada A, Harter P, Canon JL, Possinger K, Schmidt M, De Greve J, Neven P, Dirix L, Jonat W, Beckmann MW, Schutte J, Fasching PA, Gottschalk N, Besse-Hammer T, Fleischer F, Wind S, Uttenreuther-Fischer M, Piccart M, Harbeck N. A phase II trial to assess efficacy and safety of afatinib in extensively pretreated patients with HER2-negative metastatic breast cancer. Breast Cancer Res Treat. 2012;134:1149–1159. doi: 10.1007/s10549-012-2126-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, Liu MC, Storniolo AM, Rimawi MF, Forero-Torres A, Wolff AC, Hobday TJ, Ivanova A, Chiu WK, Ferraro M, Burrows E, Bernard PS, Hoadley KA, Perou CM, Winer EP. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J. Clin. Oncol. 2012;30:2615–2623. doi: 10.1200/JCO.2010.34.5579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Baselga J, Gomez P, Greil R, Braga S, Climent MA, Wardley AM, Kaufman B, Stemmer SM, Pego A, Chan A, Goeminne JC, Graas MP, Kennedy MJ, Ciruelos Gil EM, Schneeweiss A, Zubel A, Groos J, Melezinkova H, Awada A. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J. Clin. Oncol. 2013;31:2586–2592. doi: 10.1200/JCO.2012.46.2408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tredan O, Campone M, Jassem J, Vyzula R, Coudert B, Pacilio C, Prausova J, Hardy-Bessard AC, Arance A, Mukhopadhyay P, Aloe A, Roche H. Ixabepilone alone or with cetuximab as first-line treatment for advanced/metastatic triple-negative breast cancer. Clin Breast Cancer. 2015;15:8–15. doi: 10.1016/j.clbc.2014.07.007. [DOI] [PubMed] [Google Scholar]
  • 94.Crozier JA, Advani PP, LaPlant B, Hobday T, Jaslowski AJ, Moreno-Aspitia A, Perez EA. N0436 (Alliance): A Phase II Trial of Irinotecan With Cetuximab in Patients With Metastatic Breast Cancer Previously Exposed to Anthracycline and/or Taxane-Containing Therapy. Clin Breast Cancer. 2016;16:23–30. doi: 10.1016/j.clbc.2015.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Nabholtz JM, Abrial C, Mouret-Reynier MA, Dauplat MM, Weber B, Gligorov J, Forest AM, Tredan O, Vanlemmens L, Petit T, Guiu S, Van Praagh I, Jouannaud C, Dubray-Longeras P, Tubiana-Mathieu N, Benmammar KE, Kullab S, Bahadoor MR, Radosevic-Robin N, Kwiatkowski F, Desrichard A, Cayre A, Uhrhammer N, Chalabi N, Chollet P, Penault-Llorca F. Multicentric neoadjuvant phase II study of panitumumab combined with an anthracycline/taxane-based chemotherapy in operable triple-negative breast cancer: identification of biologically defined signatures predicting treatment impact. Ann Oncol. 2014;25:1570–1577. doi: 10.1093/annonc/mdu183. [DOI] [PubMed] [Google Scholar]
  • 96.Nabholtz JM, Chalabi N, Radosevic-Robin N, Dauplat MM, Mouret-Reynier MA, Van Praagh I, Servent V, Jacquin JP, Benmammar KE, Kullab S, Bahadoor MR, Kwiatkowski F, Cayre A, Abrial C, Durando X, Bignon YJ, Chollet P, Penault-Llorca F. Multicentric neoadjuvant pilot Phase II study of cetuximab combined with docetaxel in operable triple negative breast cancer. Int J Cancer. 2016;138:2274–2280. doi: 10.1002/ijc.29952. [DOI] [PubMed] [Google Scholar]
  • 97.Pruneri G, Vingiani A, Bagnardi V, Rotmensz N, De Rose A, Palazzo A, Colleoni AM, Goldhirsch A, Viale G. Clinical validity of tumor-infiltrating lymphocytes analysis in patients with triple-negative breast cancer. Ann Oncol. 2016;27:249–256. doi: 10.1093/annonc/mdv571. [DOI] [PubMed] [Google Scholar]
  • 98.Mai A, Cheng D, Bedford MT, Valente S, Nebbioso A, Perrone A, Brosch G, Sbardella G, De Bellis F, Miceli M, Altucci L. epigenetic multiple ligands: mixed histone/protein methyltransferase, acetyltransferase, and class III deacetylase (sirtuin) inhibitors. J Med Chem. 2008;51:2279–2290. doi: 10.1021/jm701595q. [DOI] [PubMed] [Google Scholar]
  • 99.Sonnenblick A, de Azambuja E, Azim HA Jr, Piccart M. An update on PARP inhibitors--moving to the adjuvant setting. Nat Rev Clin Oncol. 2015;12:27–41. doi: 10.1038/nrclinonc.2014.163. [DOI] [PubMed] [Google Scholar]
  • 100.Nowsheen S, Bonner JA, Lobuglio AF, Trummell H, Whitley AC, Dobelbower MC, Yang ES. Cetuximab augments cytotoxicity with poly (adp-ribose) polymerase inhibition in head and neck cancer. PLoS One. 2011;6:e24148. doi: 10.1371/journal.pone.0024148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nowsheen S, Cooper T, Stanley JA, Yang ES. Synthetic lethal interactions between EGFR and PARP inhibition in human triple negative breast cancer cells. PLoS One. 2012;7:e46614. doi: 10.1371/journal.pone.0046614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Du Y, Yamaguchi H, Wei Y, Hsu JL, Wang HL, Hsu YH, Lin WC, Yu WH, Leonard PG, Lee GRt, Chen MK, Nakai K, Hsu MC, Chen CT, Sun Y, Wu Y, Chang WC, Huang WC, Liu CL, Chang YC, Chen CH, Park M, Jones P, Hortobagyi GN, Hung MC. Blocking c-Met-mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors. Nat Med. 2016;22:194–201. doi: 10.1038/nm.4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015;33:1974–1982. doi: 10.1200/JCO.2014.59.4358. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES