Purpose: This study investigated the safety, clinical activity, and target-associated biomarkers of lumretuzumab, a humanized, glycoengineered, anti-HER3 monoclonal antibody (mAb), in combination with the EGFR-blocking agents erlotinib or cetuximab in patients with advanced HER3-positive carcinomas.

Experimental Design: The study included two parts: dose escalation and dose extension phases with lumretuzumab in combination with either cetuximab or erlotinib, respectively. In both parts, patients received lumretuzumab doses from 400 to 2,000 mg plus cetuximab or erlotinib according to standard posology, respectively. The effect of HRG mRNA and HER3 mRNA and protein expression were investigated in a dedicated extension cohort of squamous non–small cell lung cancer (sqNSCLC) patients treated with lumretuzumab and erlotinib.

Results: Altogether, 120 patients were treated. One dose-limiting toxicity (DLT) in the cetuximab part and two DLTs in the erlotinib part were reported. The most frequent adverse events were gastrointestinal and skin toxicities, which were manageable. The objective response rate (ORR) was 6.1% in the cetuximab part and 4.2% in the erlotinib part. In the sqNSCLC extension cohort of the erlotinib part, higher tumor HRG and HER3 mRNA levels were associated with a numerically higher disease control rate but not ORR.

Conclusions: The toxicity profile of lumretuzumab in combination with cetuximab and erlotinib was manageable, but only modest clinical activity was observed across tumor types. In the sqNSCLC cohort, there was no evidence of meaningful clinical benefit despite enriching for tumors with higher HRG mRNA expression levels. Clin Cancer Res; 23(18); 5406–15. ©2017 AACR.

Translational Relevance

Preclinical models have demonstrated superior antitumor activity when HER3- and EGFR-targeting therapies are combined as compared with single-agent activity. Furthermore, preclinical and clinical data suggest that response to HER3-targeting therapy is positively correlated to expression of the HER3 ligand heregulin (HRG).

Across tumor types, the clinical activity of the anti-HER3 antibody lumretuzumab when given in combination with the EGFR-blocking agents cetuximab or erlotinib was modest, with a manageable safety profile. To enrich for tumors with higher HRG mRNA levels, we evaluated the combination of lumretuzumab and erlotinib in a dedicated cohort of squamous non–small cell lung cancer patients. Here, neither HRG nor HER3 mRNA expression levels were associated with an increased response rate, questioning (i) the relevance of HER3 blockade in combination with EGFR-targeting therapy per se and (ii) the relevance of HRG and HER3 as response prediction biomarkers for this therapy.

HER3 is a key dimerization partner of HER family members that activates several oncogenic signal transduction pathways, particularly the PI3K/Akt pathway (1). Recent studies indicate that HER3 pathway activation is important in the development of resistance to EGFR- and HER2-targeting treatments (2–6). The role of HER3 as a prognostic marker remains controversial. Increased HER3 protein expression as measured by immunohistochemistry (IHC) has been described as an adverse prognostic factor in many solid tumor types, including breast, gastric, lung, ovarian, and colon cancers (7–11), and targeting HER3 can sensitize refractory tumor models to EGFR inhibitors (12). In contrast, higher expression levels of HER3 mRNA were associated with increased progression-free survival (PFS) in HER2-positive metastatic breast cancer treated with pertuzumab plus trastuzumab plus docetaxel (13) and in patients with platinum-resistant ovarian cancer treated with gemcitabine plus pertuzumab (14). However, adding pertuzumab to chemotherapy in platinum-resistant ovarian cancer patients selected on low HER3 mRNA expression did not increase PFS significantly (15). Autocrine loops involving the HER3 ligand HRG and leading to HER3 activation have been described in squamous cell carcinoma of the head and neck (SCCHN), non–small cell lung cancer (NSCLC), and ovarian cancer (8, 16–20).

Lumretuzumab is a humanized, glycoengineered immunoglobulin G1 antibody that selectively binds with high affinity to the extracellular domain of HER3. Prevention of HRG binding to HER3 by lumretuzumab resulted in almost complete inhibition of HER3 heterodimerization and phosphorylation as well as inhibition of tumor growth in cell line–based mouse xenograft models (21). In a phase I study, the safety of lumretuzumab was evaluated in patients with advanced solid tumors. No DLTs were observed at doses of 100 to 2,000 mg every 2 weeks (as a flat dose) and an MTD was not reached. Downregulation of HER3 membranous protein was observed in on-treatment tumor biopsies from 200 mg and higher doses, and a target-independent pharmacokinetic profile was observed at ≥400 mg doses (22).

Multiple preclinical models have shown that combinations of anti-HER3 antibodies with anti-EGFR therapies lead to enhanced antitumor activity as compared with the single agents, and even complete tumor regression has been described in different studies (12, 21, 23, 24). Preclinical studies have also shown that HER3 pathway activation and overexpression of HRG predicted response to HER3-targeting therapy (25, 26), and preliminary clinical data from early studies with anti-HER3 antibodies are consistent with these reports (22, 27, 28). We therefore hypothesized that (i) the combination of lumretuzumab with EGFR inhibitors could improve clinical activity versus EGFR inhibitors alone, and (ii) higher tumor expression levels of HRG mRNA could indicate improved clinical outcome and could ultimately serve as a predictive biomarker for HER3-targeting therapy.

The current study evaluated the safety and clinical activity, and potential biomarkers of lumretuzumab treatment when combined with the EGFR-targeting agents cetuximab or erlotinib. To evaluate the clinical activity of lumretuzumab plus erlotinib in a biomarker-enriched population, we recruited an extension cohort of sqNSCLC patients, as sqNSCLC has been shown to express higher HRG mRNA levels compared with non-sqNSCLC (data on file; refs. 29, 30).

Study design

This was a multicenter, phase Ib, open-label, nonrandomized, dose escalation and extension study (ClinicalTrials.gov Identifier: NCT01482377) investigating the safety (including the MTD and/or optimal biological dose), pharmacokinetics, pharmacodynamics, and clinical activity of lumretuzumab in combination with cetuximab or erlotinib in patients with metastatic or advanced HER3-positive carcinomas. The study consisted of a dose escalation phase following a standard “3 + 3” study design and an extension phase conducted for the cetuximab plus lumretuzumab as well as the erlotinib plus lumretuzumab combination.

For lumretuzumab monotherapy, pharmacokinetics was linear at dosages ≥400 mg, indicative of target-mediated drug disposition saturation, and clinical pharmacodynamic activity was demonstrated (22). Therefore, the dose of 400 mg was used as the starting dose for the combination treatments with cetuximab and erlotinib. Two administration schedules were applied: every 2 weeks and every 3 weeks; the latter only applied for a subset of patients in the lumretuzumab plus erlotinib part.

Standard doses of cetuximab (400 mg/m2 for the first infusion followed by 250 mg/m2 weekly infusions) and erlotinib (daily dose of 150 mg orally) were used.

Patients continued treatment until disease progression, unacceptable toxicity, or withdrawal of consent.

Ethics

Local ethics committee approval was obtained, and all patients provided written informed consent. The study was conducted in accordance with Good Clinical Practice guidelines and the Declaration of Helsinki in nine centers in Spain, the Netherlands, Denmark, and South Korea.

Patients

Patients had to have a histologically confirmed diagnosis of an advanced or metastatic HER3-expressing carcinoma that was refractory to standard treatment or for which no standard therapy existed. Eligible patients were ≥18 years of age, had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 to 2 and had adequate hematology, blood chemistry, and renal and liver function. Patients eligible for enrollment underwent a fresh (pretreatment) tumor biopsy that was used to assess the level of HER3 protein expression by IHC using central pathology review. Discernible HER3 membrane staining in any neoplastic cell, provided that a minimum of 100 tumor cells were present in the biopsy specimen, was considered diagnostically positive for HER3 protein expression.

For the extension phase of the cetuximab combination part, only patients with advanced HER3-expressing SCCHN, NSCLC, and colorectal cancer (proven to be KRAS wild-type and expressing EGFR as per local assessment by the investigator) were recruited. For the extension phase of the erlotinib combination part, only patients with advanced HER3-expressing sqNSCLC and patients with nonsquamous lung carcinomas with a documented NRG1 somatic gene fusion (determined by local PCR and sequencing assays) were recruited to enrich for patients more likely to benefit from lumretuzumab. Higher expression of HRG mRNA in sqNSCLC as compared with non-sqNSCLC had been demonstrated through mining of data extracted from TCGA and analysis of HRG by qRT-PCR in formalin-fixed, paraffin-embedded (FFPE) tumor specimens from the Roche clinical tumor bank and from preclinical HER3 xenograft response modeling studies. In addition, higher HRG levels positively correlated with response to lumretuzumab treatment in preclinical models (data not shown). Recently, gene fusions were identified involving the gene encoding for HRG (NRG1; ref. 31) activating HER3 in an autocrine and paracrine manner as a novel mechanism for HER pathway dysregulation in lung adenocarcinomas (32). Tumors harboring these NRG1 fusions may be particularly susceptible to HER3-targeting therapy. The protocol was therefore amended to allow inclusion of these rare patients to explore signals of clinical activity. Because of the differences in the underlying biology, it was decided to analyze the patients with NRG1-rearranged tumors separately and not include them into the analysis of the sqNSCLC cohort.

Responding patients were tested for EGFR mutations with Qiagen therascreen EGFR Pyro assay (Source BioScience).

Study drug administration

Patients received premedication 30 minutes prior to the start of the first lumretuzumab infusion consisting of paracetamol (500–1,000 mg orally) and diphenhydramine (25–50 mg orally or inravenously, or an alternative antihistamine). Corticosteroids were allowed in case of ≥ grade 2 infusion-related reactions (IRR). Lumretuzumab was administered as an intravenous infusion. Cetuximab and erlotinib were administered according to standard posology, and dose reductions were allowed according to the label. Study drugs could be delayed to assess or treat adverse events (AE) for up to 14 days. One reduction to the previously assessed lower dose level was allowed for lumretuzumab.

Tumor response and safety

Tumor response assessment using RECIST version 1.1 (33) was conducted at screening and every 8 or 9 weeks thereafter for the every-2-week or every-3-week schedule, respectively.

Safety assessments included physical (ECOG performance status, vital signs) and laboratory examinations and electrocardiogram. AEs were defined according to the Common Terminology Criteria for Adverse Events, version 4.0 (CTC-AE v4.0).

Definition of dose-limiting toxicity

A dose-limiting toxicity (DLT) was defined as an AE occurring during the first two cycles of treatment (i.e., 28 days) with lumretuzumab that was considered to be study drug related and was either: grade 4 neutropenia (i.e., absolute neutrophil count <0.5 × 109 cells/L for a minimal duration of 7 days); grade 3/4 febrile neutropenia; grade 4 thrombocytopenia; grade 3 thrombocytopenia associated with bleeding episodes; or grade ≥ 3 nonhematologic toxicity. IRRs and grade 3 skin and/or epithelial toxicities associated with EGFR inhibition were not considered DLTs for the cetuximab combination. For the erlotinib combination, IRRs, grade 3 skin, and/or epithelial toxicities associated with EGFR inhibition and grade 3 diarrhea, which reduced to less than grade 2 within 14 days by supportive treatment or dose reduction of erlotinib or interruption of erlotinib, were not considered DLTs.

Biomarker assessments

Fresh tumor biopsies were collected during screening and on day 14 of cycle 1. HER3 protein expression was assessed using an IHC assay and scored semiquantitatively and reported as an immunoreactive score (IRS, range 0–300) as described previously (22).

HRG mRNA expression was measured by a qRT-PCR assay in FFPE sections obtained from fresh tumor biopsies collected from all patients at screening prior to initiation of treatment. In addition, HER3 mRNA expression was determined using a separate qRT-PCR assay. Both assays were performed at Roche Molecular Systems.

Total RNA was isolated from FFPE tumor tissue sections using the Cobas RNA Isolation Kit. TaqMan probes were designed to detect HRG or HER3 mRNA and respective reference genes simultaneously. All reagents were prepared at Roche Molecular Systems, and qRT-PCR was performed using the Cobas 4800 system. Calculation of the cycle-to-threshold (Ct) for each fluorescent channel was done using LC480 software, and the relative HRG or HER3 mRNA log expression was reported as ΔCt, where ΔCt = Ct(reference) − Ct(target). Where feasible, biopsies with less than 50% tumor content underwent macrodissection guided by pathologist annotation of adjacent H&E-stained sections. No reference ranges were defined a priori for HRG or HER3 mRNA expression using the initial research-grade assay.

During the course of the clinical study, the HRG mRNA assay was transitioned to a prototype diagnostic assay, which differed in that reagents were prepared in a GMP facility, the assay run using a z480 PCR system, and calculation of Ct values was performed using validated diagnostic software (Heregulin Expression Test Analysis Package for use with Cobas 4800 System Release 2.1). Predose HRG mRNA expression data derived using both the research and prototype diagnostic assays were available for 37 of 120 patients. Overall, the research assay reported higher expression than the diagnostic prototype due to differences in the data analysis parameter settings, albeit there was strong correlation between the two assays (mean difference 1.53 ΔCtR2 = 0.96).

The prototype diagnostic assay was used to define a reference range for HRG expression using 150 primary FFPE tumor biopsy samples from sqNSCLC patients (obtained from a Roche tumor bank and an external vendor). Median HRG log expression corresponded to a ΔCt of −1.92 with a 75th percentile of −0.92. The median was used as a threshold above which tumor biopsies derived from patients were defined as HRG-high.

Statistical analysis

All patients who received at least one dose of study medication were included in the safety population. Descriptive statistics were used for demographics and safety.

To explore biomarker levels and correlations, a pharmacodynamic population was also defined, comprising of all patients receiving at least one dose of study medication and providing a pharmacodynamic sample at baseline. Because of the distinct histology, invasive mucinous NSCLC, and potentially distinct biology, lung cancer patients with NRG1 gene alterations were included in the sqNSCLC cohort but were analyzed separately. For some patients, biomarker data were not available, either due to lack of evaluable tumor tissue or for technical reasons. Therefore, for each analysis, the number of evaluable patients (n) is specified.

Exploratory statistical analyses were used to evaluate differences in biomarker levels for patients with different tumor histologies (nonsquamous vs. squamous carcinoma) or lung cancer diagnosis and changes from baseline levels using the Wilcoxon rank sum test or Wilcoxon signed rank test as appropriate.

The Spearman rank correlation coefficient (ρ) was used to determine the association between biomarker levels.

The information on patients' histology (squamous and adenocarcinoma) was available for the vast majority of the patients or could be inferred through the tumor type (e.g., patients with colorectal cancer have adenocarcinoma): Only 4 patients had to be excluded in the analyses of biomarker differences between adeno and squamous carcinoma patients as the histology was either unknown, both histologies were present, or the histology was of transitional cell type, although all available data were utilized to assess biomarker correlations.

For efficacy, RECIST response was evaluated and summarized by study part (cetuximab/erlotinib combinations), and the objective response from the sqNSCLC patients of the extension cohort was related to biomarker levels via a multinomial logit model. All available patients were included in the analyses, as all dose levels were considered pharmacodynamically active.

Patients

Patient demographics and baseline characteristics are presented in Table 1. In the dose escalation phase of the cetuximab part, 27 patients were enrolled into five dose cohorts, that is, 400 (n = 5), 800 (n = 6), 1,200 (n = 5), 1,600 (n = 5), and 2,000 mg (n = 6) of lumretuzumab every 2 weeks. In the extension phase of the cetuximab part, 22 patients received 2,000 mg. In the dose escalation phase of the erlotinib part, 37 patients were enrolled into four every-2-week dose cohorts, that is, 400 (n = 6), 800 (n = 6), 1,600 (n = 7), and 2,000 mg (n = 8) of lumretuzumab, and 10 patients were enrolled into two every-3-week dose cohorts, that is, 800 (n = 7) and 1,600 mg (n = 3). In the extension phase of the erlotinib part, 34 patients received 800 mg of lumretuzumab every 2 weeks (Fig. 1). Two of those patients had NSCLC adenocarcinoma with an NRG1 fusion, and 32 were sqNSCLC patients.

Table 1.

Baseline patient demographics and characteristics

Cetuximab partErlotinib part
Characteristicsn = 49n = 71
Sex, n (%) 
 Male 31 (63.3) 46 (64.8) 
 Female 18 (36.7) 25 (35.2) 
Age (years), median (range) 59 (35, 81) 63 (30, 78) 
ECOG score, n (%) 
 0 12 (24.5) 25 (35.2) 
 1 36 (73.5) 41 (57.7) 
 2 1 (2.0) 5 (7.9) 
Prior chemotherapy, n (%) 46 (93.9) 69 (97.2) 
 Median number (range) 2 (0–6) 1 (0–5) 
Prior EGFR-targeting therapy, n (%) 27 (55.1) 20 (28.2) 
Prior surgery, n (%) 30 (61.2) 42 (59.2) 
Prior radiotherapy 27 (55.1) 41 (57.7) 
Tumor type, n (%) 
 Colorectal 22 13 
 Head and neck 11 
 NSCLC 10 43 
 Carcinoma of unknown primary origin 
 Pancreatic cancer 
 Gastric cancer 
 Thymus cancer 
 Other 4a 5b 
Cetuximab partErlotinib part
Characteristicsn = 49n = 71
Sex, n (%) 
 Male 31 (63.3) 46 (64.8) 
 Female 18 (36.7) 25 (35.2) 
Age (years), median (range) 59 (35, 81) 63 (30, 78) 
ECOG score, n (%) 
 0 12 (24.5) 25 (35.2) 
 1 36 (73.5) 41 (57.7) 
 2 1 (2.0) 5 (7.9) 
Prior chemotherapy, n (%) 46 (93.9) 69 (97.2) 
 Median number (range) 2 (0–6) 1 (0–5) 
Prior EGFR-targeting therapy, n (%) 27 (55.1) 20 (28.2) 
Prior surgery, n (%) 30 (61.2) 42 (59.2) 
Prior radiotherapy 27 (55.1) 41 (57.7) 
Tumor type, n (%) 
 Colorectal 22 13 
 Head and neck 11 
 NSCLC 10 43 
 Carcinoma of unknown primary origin 
 Pancreatic cancer 
 Gastric cancer 
 Thymus cancer 
 Other 4a 5b 

aOther primary tumors included one patient each with esophageal cancer, breast cancer, urothelial pelvic cancer, and spiradenocarcinoma, respectively.

bOther primary tumors included one patient each with cervical cancer, esophageal cancer, ovarian cancer, gastroesophageal junction cancer, and anal cancer, respectively.

Figure 1.

Flow diagram of study design, patient enrollment, and lumretuzumab dose. CRC, colorectal cancer; N, number of patients; NSCLC, non–small cell lung cancer; q2w, every 2 weeks; q3w, every 3 weeks; SCCHN, squamous cell carcinoma of the head and neck. aFor all patients, a biopsy was taken at screening and on treatment 14 days after cycle 1. Solely for the extension phase of the erlotinib part, no on-treatment biopsy was taken, and for these patients, the prototype diagnostic RT-PCR assay was used.

Figure 1.

Flow diagram of study design, patient enrollment, and lumretuzumab dose. CRC, colorectal cancer; N, number of patients; NSCLC, non–small cell lung cancer; q2w, every 2 weeks; q3w, every 3 weeks; SCCHN, squamous cell carcinoma of the head and neck. aFor all patients, a biopsy was taken at screening and on treatment 14 days after cycle 1. Solely for the extension phase of the erlotinib part, no on-treatment biopsy was taken, and for these patients, the prototype diagnostic RT-PCR assay was used.

Close modal

Overall, 102 patients (85.0%) discontinued the study due to progressive disease or death, 6 patients (5.0%) were withdrawn due to an AE, 6 patients (5.0%) refused further treatment, 1 patient (0.8%) withdrew consent, and 5 patients (4.2%) discontinued for other reasons.

Safety

Three patients with DLTs were reported, one in the cetuximab part (dehydration grade 3 in the 800-mg cohort) and two in the erlotinib part (diarrhea grade 3 and hypokalemia grade 3 in one patient of the 1,600 mg cohort and blood bilirubin increased grade 3 in one patient of the 2,000 mg cohort). The MTD was not reached up to the highest dose tested (i.e., 2,000 mg) in both treatment parts. A total of 1,452 AEs of any grade were reported in 120 patients (100%; Table 2). Most AEs (87.3%) were grade 1 or 2 in intensity. A total of 184 grade ≥3 AEs were reported in 88 patients (73.3%). No patients in the cetuximab part and 3 patients (4.2%) in the erlotinib part died from an AE (pneumonia, cerebrovascular accident, and hemoptysis with one patient each, all considered unrelated to lumretuzumab treatment). The most frequent AEs included diarrhea [38 patients (77.6%) in the cetuximab part and 54 patients (76.1%) in the erlotinib part] and rash [26 patients (53.1%) in the cetuximab part and 32 patients (45.1%) in the erlotinib part]. The most frequent ≥grade 3 AEs included diarrhea [6 patients (12.2%) in the cetuximab part and 11 patients (15.5%) in the erlotinib part], hypomagnesemia [5 patients (10.2%) in the cetuximab part and no patients in the erlotinib part], and dermatitis acneiform [5 patients (10.2%) in the cetuximab part and 4 patients (5.6%) in the erlotinib part]. Diarrhea was the only AE for which a potential dose dependency was observed during dose escalation. In the cetuximab part, 1 of 6 patients had grade 3 diarrhea only at the top dose of 2,000 mg lumretuzumab. In the erlotinib part, 1 of 6 patients at 800 mg, 3 of 7 patients at 1,600 mg, and 2 of 8 patients at 2,000 mg lumretuzumab had grade 3 diarrhea.

Table 2.

Summary of AEs of any grade and of grade ≥3 regardless of relationship to study drug

Patients having an AE, n (%)
Cetuximab partErlotinib part
n = 49n = 71
AEAll gradesGrade ≥3All gradesGrade ≥3
Diarrhea 38 (77.6) 6 (12.2) 54 (76.1) 11 (15.5) 
Rash 26 (53.1) 4 (8.2) 32 (45.1) 2 (2.8) 
Dry skin 23 (46.9) 26 (36.6) 
Decreased appetite 21 (42.9) 2 (4.1) 27 (38.0) 5 (7.0) 
Hypomagnesemia 19 (38.8) 5 (10.2) 15 (21.1) 
Fatigue 12 (24.5) 2 (4.1) 26 (36.6) 2 (2.8) 
Nausea 16 (32.7) 1 (2.0) 16 (22.5) 
Paronychia 16 (32.7) 2 (4.1) 3 (4.2) 
Weight decreased 6 (12.2) 21 (29.6) 2 (2.8) 
Mucosal inflammation 14 (28.6) 20 (28.2) 1 (1.4) 
Vomiting 13 (26.5) 1 (2.0) 12 (16.9) 
Dermatitis acneiform 12 (24.5) 5 (10.2) 18 (25.4) 4 (5.6) 
Stomatitis 7 (14.3) 2 (4.1) 16 (22.5) 1 (1.4) 
Hypokalemia 5 (10.2) 2 (4.1) 16 (22.5) 7 (9.9) 
Dyspnea 6 (12.2) 2 (4.1) 16 (22.5) 3 (4.2) 
Pyrexia 9 (18.4) 15 (21.1) 
Asthenia 8 (16.3) 1 (2.0) 15 (21.1) 1 (1.4) 
Conjunctivitis 9 (18.4) 4 (5.6) 
Infusion-related reaction 9 (18.4) 3 (6.1)a 5 (7.0) 
Anemia 6 (12.2) 3 (6.1) 11 (15.5) 3 (4.2) 
Cough 7 (14.3) 8 (11.3) 
Rash pustular 7 (14.3) 1 (2.0) 1 (1.4) 
Urinary tract infection 3 (6.1) 9 (12.7) 
Infection 6 (12.2) 3 (6.1) 3 (4.2) 1 (1.4) 
Dry mouth 6 (12.2) 3 (4.2) 
Abdominal pain 5 (10.2) 2 (4.1) 6 (8.5) 
Skin fissures 5 (10.2) 3 (4.2) 
Epistaxis 5 (10.2) 1 (1.4) 
Myalgia 5 (10.2) 
Anxiety 5 (10.2) 1 (1.4) 
Dysphonia 7 (9.9) 
Palmar-plantar erythrodysesthesia syndrome 1 (2.0) 7 (9.9) 
Dizziness 4 (8.2) 6 (8.5) 
Dehydration 4 (8.2) 3 (6.1) 5 (7.0) 2 (2.8) 
Edema peripheral 4 (8.2) 4 (5.6) 1 (1.4) 
Ascites 4 (8.2) 2 (4.1) 2 (2.8) 1 (1.4) 
Eye infection 4 (8.2) 2 (2.8) 
Nasopharyngitis 4 (8.2) 3 (4.2) 
Pneumonia 4 (8.2) 1 (2.0) 3 (4.2) 2 (2.8) 
Dysgeusia 4 (8.2) 1 (1.4) 
Blood bilirubin increased 1 (2.0) 1 (2.0) 5 (7.0) 1 (1.4) 
Rash maculopapular 1 (2.0) 5 (7.0) 
Constipation 3 (6.1) 3 (4.2) 
Nail infection 3 (6.1) 1 (1.4) 
Erythema 3 (6.1) 1 (1.4) 
Hypocalcemia 3 (6.1) 2 (4.1) 
Musculoskeletal pain 3 (6.1) 3 (4.2) 
Pulmonary embolism 3 (6.1) 3 (6.1) 1 (1.4) 1 (1.4) 
Dry eye 3 (6.1) 2 (2.8) 
Headache 3 (6.1) 2 (2.8) 
Pneumothorax 3 (6.1) 
Angular cheilitis 3 (6.1) 
Influenza-like illness 3 (6.1) 3 (4.2) 
Respiratory tract infection 2 (4.1) 4 (5.6) 3 (4.2) 
Blood creatinine increased 2 (4.1) 4 (5.6) 
Hypercalcemia 4 (5.6) 2 (2.8) 
Hyponatremia 4 (5.6) 4 (5.6) 
Alanine aminotransferase increased 4 (5.6) 1 (1.4) 
Pruritus 1 (2.0) 4 (5.6) 
Alopecia 4 (5.6) 
Dysphagia 2 (4.1) 2 (4.1) 4 (5.6) 
Patients having an AE, n (%)
Cetuximab partErlotinib part
n = 49n = 71
AEAll gradesGrade ≥3All gradesGrade ≥3
Diarrhea 38 (77.6) 6 (12.2) 54 (76.1) 11 (15.5) 
Rash 26 (53.1) 4 (8.2) 32 (45.1) 2 (2.8) 
Dry skin 23 (46.9) 26 (36.6) 
Decreased appetite 21 (42.9) 2 (4.1) 27 (38.0) 5 (7.0) 
Hypomagnesemia 19 (38.8) 5 (10.2) 15 (21.1) 
Fatigue 12 (24.5) 2 (4.1) 26 (36.6) 2 (2.8) 
Nausea 16 (32.7) 1 (2.0) 16 (22.5) 
Paronychia 16 (32.7) 2 (4.1) 3 (4.2) 
Weight decreased 6 (12.2) 21 (29.6) 2 (2.8) 
Mucosal inflammation 14 (28.6) 20 (28.2) 1 (1.4) 
Vomiting 13 (26.5) 1 (2.0) 12 (16.9) 
Dermatitis acneiform 12 (24.5) 5 (10.2) 18 (25.4) 4 (5.6) 
Stomatitis 7 (14.3) 2 (4.1) 16 (22.5) 1 (1.4) 
Hypokalemia 5 (10.2) 2 (4.1) 16 (22.5) 7 (9.9) 
Dyspnea 6 (12.2) 2 (4.1) 16 (22.5) 3 (4.2) 
Pyrexia 9 (18.4) 15 (21.1) 
Asthenia 8 (16.3) 1 (2.0) 15 (21.1) 1 (1.4) 
Conjunctivitis 9 (18.4) 4 (5.6) 
Infusion-related reaction 9 (18.4) 3 (6.1)a 5 (7.0) 
Anemia 6 (12.2) 3 (6.1) 11 (15.5) 3 (4.2) 
Cough 7 (14.3) 8 (11.3) 
Rash pustular 7 (14.3) 1 (2.0) 1 (1.4) 
Urinary tract infection 3 (6.1) 9 (12.7) 
Infection 6 (12.2) 3 (6.1) 3 (4.2) 1 (1.4) 
Dry mouth 6 (12.2) 3 (4.2) 
Abdominal pain 5 (10.2) 2 (4.1) 6 (8.5) 
Skin fissures 5 (10.2) 3 (4.2) 
Epistaxis 5 (10.2) 1 (1.4) 
Myalgia 5 (10.2) 
Anxiety 5 (10.2) 1 (1.4) 
Dysphonia 7 (9.9) 
Palmar-plantar erythrodysesthesia syndrome 1 (2.0) 7 (9.9) 
Dizziness 4 (8.2) 6 (8.5) 
Dehydration 4 (8.2) 3 (6.1) 5 (7.0) 2 (2.8) 
Edema peripheral 4 (8.2) 4 (5.6) 1 (1.4) 
Ascites 4 (8.2) 2 (4.1) 2 (2.8) 1 (1.4) 
Eye infection 4 (8.2) 2 (2.8) 
Nasopharyngitis 4 (8.2) 3 (4.2) 
Pneumonia 4 (8.2) 1 (2.0) 3 (4.2) 2 (2.8) 
Dysgeusia 4 (8.2) 1 (1.4) 
Blood bilirubin increased 1 (2.0) 1 (2.0) 5 (7.0) 1 (1.4) 
Rash maculopapular 1 (2.0) 5 (7.0) 
Constipation 3 (6.1) 3 (4.2) 
Nail infection 3 (6.1) 1 (1.4) 
Erythema 3 (6.1) 1 (1.4) 
Hypocalcemia 3 (6.1) 2 (4.1) 
Musculoskeletal pain 3 (6.1) 3 (4.2) 
Pulmonary embolism 3 (6.1) 3 (6.1) 1 (1.4) 1 (1.4) 
Dry eye 3 (6.1) 2 (2.8) 
Headache 3 (6.1) 2 (2.8) 
Pneumothorax 3 (6.1) 
Angular cheilitis 3 (6.1) 
Influenza-like illness 3 (6.1) 3 (4.2) 
Respiratory tract infection 2 (4.1) 4 (5.6) 3 (4.2) 
Blood creatinine increased 2 (4.1) 4 (5.6) 
Hypercalcemia 4 (5.6) 2 (2.8) 
Hyponatremia 4 (5.6) 4 (5.6) 
Alanine aminotransferase increased 4 (5.6) 1 (1.4) 
Pruritus 1 (2.0) 4 (5.6) 
Alopecia 4 (5.6) 
Dysphagia 2 (4.1) 2 (4.1) 4 (5.6) 

NOTE: Only AEs reported by >5% of the patients in the cetuximab or erlotinib treatment part are shown.

aFor one patient, the IRR was considered related to cetuximab and for 2 patients to lumretuzumab.

The severity of diarrhea was increased at 1,600 and 2,000 mg, particularly in combination with erlotinib. Although these diarrhea events did not qualify for DLTs as per protocol, a lower lumretuzumab dose of 800 mg every 2 weeks was selected as the recommended phase II dose for the combination with erlotinib, whereas a dose of 2,000 mg every 2 weeks was selected for the combination with cetuximab based on the observed safety profile.

Pharmacokinetics

Similarly as described for monotherapy with lumretuzumab (22), a linear pharmacokinetic profile was observed with doses of lumretuzumab of ≥400 mg every 2 weeks, indicating target-mediated drug disposition saturation (data not shown).

Biomarker analysis

Tumors of all enrolled patients were HER3 positive based on IHC analysis. In line with previously published data (22), membranous HER3 was downregulated in tumor and skin biopsy samples collected on day 14 of cycle 1 compared with baseline (Supplementary Tables S1 and S2).

In baseline tumor samples, HER3 protein and HER3 mRNA were generally more highly expressed in adenocarcinoma compared with those of squamous cell histology [HER3 IHC: median IRS 2.00 (n = 61) and 0.07 (n = 50), respectively, P < 0.0001; HER3 mRNA: median ΔCt 1.88 (n = 61) and −0.27 (n = 49), respectively, P < 0.0001, Supplementary Fig. S1]. Furthermore, HER3 protein expression was significantly correlated with HER3 mRNA expression (ρ = 0.61, P < 0.001, n = 112). Similarly, for the subset of patients with NSCLC, HER3 protein was more highly expressed in the nonsquamous compared with the squamous subtype [HER3 IHC: median IRS 2.25 (n = 14) and 0.06 (n = 36), respectively, P < 0.0001, Supplementary Fig. S2].

Conversely, analysis of baseline tumor biopsy samples using the research HRG qRT-PCR assay showed an overall higher expression of HRG mRNA in tumors of squamous cell histology compared with those of adenocarcinoma histology (P < 0.0001; Supplementary Fig. S3). Similarly, within NSCLC samples, median HRG mRNA log expression (ΔCt) was higher in sqNSCLC as compared with non-sqNSCLC (P < 0.0001, Supplementary Fig. S4).

In addition, HRG mRNA expression across tumor histologies was inversely correlated to HER3 protein (ρ = −0.24, P = 0.024, n = 89) and HER3 mRNA (ρ = −0.24, P = 0.022, n = 91).

Antitumor activity

In the cetuximab part, the ORR was 6.1% and the disease control rate [DCR, i.e., the percentage of patients with a best response of stable disease (SD), partial response (PR), or complete response (CR)] was 40.8% (Table 3).

Table 3.

Best overall response to treatment (RECIST)

Number of patients (%) withCetuximab partErlotinib part
respective assessmentn = 49n = 71
CR 1 (2.0) 
PR 2 (4.1) 3 (4.2) 
SD 17 (34.7) 28 (39.4) 
Progressive disease 18 (36.7) 30 (42.3) 
Not evaluablea 3 (6.1) 
Missingb 8 (16.3) 10 (14.1) 
ORR 6.1% 4.2% 
DCR 40.8% 43.7% 
Number of patients (%) withCetuximab partErlotinib part
respective assessmentn = 49n = 71
CR 1 (2.0) 
PR 2 (4.1) 3 (4.2) 
SD 17 (34.7) 28 (39.4) 
Progressive disease 18 (36.7) 30 (42.3) 
Not evaluablea 3 (6.1) 
Missingb 8 (16.3) 10 (14.1) 
ORR 6.1% 4.2% 
DCR 40.8% 43.7% 

aPatients had a postbaseline tumor assessment that was not evaluable. All 3 patients had clinical disease progression.

bPatients had a missing best overall response if no postbaseline tumor assessment was available. Of those, 4 patients in the cetuximab part and 5 patients in the erlotinib part had clinical disease progression. CR, complete response; DCR, disease control rate; ORR, objective response rate; PR, partial response; SD, stable disease.

Two patients with colorectal cancer showed a PR and one patient with SCCHN had a CR (Supplementary Fig. S5). The SCCHN patient had not been treated with an EGFR inhibitor previously. The duration of response in this patient was 16.9 months. The patient's tumor expressed relatively high levels of HRG mRNA (ΔCt: 0.09, median ΔCt in the cetuximab part: −2.44, n = 44) and EGFR protein (IRS: 3.0) and relatively low levels of HER3 mRNA (ΔCt: −1.06, median ΔCt in the cetuximab part: 1.43, n = 46) and HER3 protein (IRS: 0.0004) as determined on fresh baseline FFPE tumor biopsies. Overall, there was no detectable relationship between any of the exploratory biomarkers and clinical response defined by RECIST (data not shown).

In the erlotinib part, the ORR was 4.2% and the DCR was 43.7% (Table 3).

Three patients (4.2%) had a PR, one with ovarian cancer (Supplementary Fig. S6) and two with sqNSCLC (Fig. 2). The ovarian cancer patient had a duration of response of 19.8 months. This tumor showed high levels of HRG mRNA (ΔCt: −0.36, median ΔCt in the erlotinib part: −1.4, n = 47) and HER3 mRNA (ΔCt: 1.49, median ΔCt in the erlotinib part: 0.28, n = 68) and HER3 protein expression (IRS: 2.00) as determined on baseline biopsies. All partial responders in this cohort were EGFR inhibitor naïve, and neither of the two sqNSCLC patients nor the ovarian cancer patient harbored an EGFR-activating mutation.

Figure 2.

Best percentage change from baseline in sum of longest diameters of target lesions for the sqNSCLC patients in the extension phase of the erlotinib part (n = 28). Red bars, PD patients; blue bars, SD patients; and green bars, PR patients. *, Patients who were classified as HRG-high. Four patients were without target lesion assessment postbaseline (2 deaths, 1 clinical progression, 1 disease progression solely based on nontarget lesions).

Figure 2.

Best percentage change from baseline in sum of longest diameters of target lesions for the sqNSCLC patients in the extension phase of the erlotinib part (n = 28). Red bars, PD patients; blue bars, SD patients; and green bars, PR patients. *, Patients who were classified as HRG-high. Four patients were without target lesion assessment postbaseline (2 deaths, 1 clinical progression, 1 disease progression solely based on nontarget lesions).

Close modal

Overall, there was no detectable relationship between any of the analyzed exploratory biomarkers and clinical response according to RECIST (data not shown).

In both the cetuximab and the erlotinib part, phosphorylated HER3 and Akt were expressed at low levels in predose FFPE biopsy samples; expressions of both were reduced in on-treatment tumor biopsy samples in the cetuximab and erlotinib arm, reflecting the mechanism of action of lumretuzumab (Supplementary Table S2). There was no significant downregulation of total membranous EGFR nor a decrease in proliferative activity based on Ki67 staining, which is in line with findings from others (34). Other exploratory biomarkers were unchanged following treatment with lumretuzumab.

The 2 patients with a SLC3A2-NRG1 fusion gene were heavily pretreated with five therapy lines for metastatic disease, including prior erlotinib treatment. Both patients had SD as their best response.

Heregulin and HER3 mRNA levels and antitumor activity in the sqNSCLC extension cohort

Altogether, 32 patients with sqNSCLC were recruited into the extension cohort of the erlotinib part. HRG mRNA expression data were available for all patients, and RECIST response data were available for 29 patients (90.6%). Overall, the ORR was 6.3%, and the DCR was 56.3% (Fig. 2).

Twelve patients were considered HRG-high using the median ΔCt as the cutoff, determined from the analysis of an external cohort of 150 FFPE tumor samples from sqNSCLC patients. Among the 12 HRG-high patients, one had a PR as best response with a duration of 3.8 months. Hence, the ORR in the HRG-high population was 8.3%, while the DCR was 75.0% (9/12 patients) and the median PFS (range) was 3.7 (1.0–8.0) months, compared with an ORR of 5.0% (1/20 patients, with a response duration of 4.9 months), a DCR of 55.0% (11/20 patients), and a median PFS (range) of 1.8 (0.2–6.5) months for the HRG-low patients (Fig. 3). In the same cohort, low HER3 mRNA expression (using the median value of the cohort as a threshold to define high and low expression, corresponding to a ΔCt of −0.3) was associated with an ORR of 12.5% (2/16 patients) and a DCR of 43.8% (7/16 patients) and median PFS (range) of 1.8 (0.2–6.5) months as compared with an ORR of 0%, a DCR of 68.8% (11/16 patients), and a median PFS (range) of 2.9 (0.3–8) months for patients with high HER3 mRNA.

Figure 3.

Objective response and HRG mRNA log expression in sqNSCLC patients in the lumretuzumab + erlotinib extension cohort (n = 32).

Figure 3.

Objective response and HRG mRNA log expression in sqNSCLC patients in the lumretuzumab + erlotinib extension cohort (n = 32).

Close modal

HRG expression was also confirmed on FFPE tumor biopsies using in situ hybridization and IHC assays. Relative HRG expression was consistent across assays, and no differences were observed in the clinical response relationship (data not shown). In this cohort, there was no evidence that higher HRG or lower HER3 mRNA levels were correlated with improved response to lumretuzumab and erlotinib treatment.

This study describes the safety, pharmacokinetics, pharmacodynamics, and clinical activity of the combination of lumretuzumab, an anti-HER3 mAb, with cetuximab and erlotinib, respectively. The study aimed to detect signals of clinical activity (i) of anti-EGFR/HER3 combination therapy per se, and (ii) activity of this combination in a histologic entity enriched for higher HRG expression levels, that is, sqNSCLC.

The most common AEs in this study were gastrointestinal and skin toxicities, similar to the well-known side effects of cetuximab and erlotinib. The addition of lumretuzumab may have caused an increase in the incidence of diarrhea compared with the incidence for cetuximab monotherapy [77.6% (any grade) and 12.2% (grade ≥3) compared with 12.7% (any grade) and 1.2% (grade ≥3) for cetuximab monotherapy (35)] and erlotinib monotherapy [76.1% (any grade) and 15.5% (grade ≥3) compared with 55% (any grade) and 6% (grade ≥3) for erlotinib monotherapy (36)]. The severity of diarrhea seemed increased at the highest lumretuzumab doses tested, that is, at 1,600 and 2,000 mg. A similar diarrhea side effect profile as in the current study was seen for combination treatments of other HER3-targeting antibodies with cetuximab (37, 38) and erlotinib (38–40) in early clinical studies. Effects might be caused by a disinhibition of HER-modulated chloride channels in colonic epithelial cells leading to secretory diarrhea (41, 42).

Although not reaching a defined MTD for both combinations, diarrhea was the only AE for which a potential dose dependency was observed. On the basis of the pharmacokinetic profile and safety assessments, a dose of 2,000 mg in combination with cetuximab and a dose of 800 mg in combination with erlotinib were determined as the recommended phase II doses.

Overall, the efficacy seen in the cetuximab (ORR = 6.1%, DCR = 42.8%) and erlotinib (ORR = 4.2%, DCR = 43.6%) combination parts does not exceed what has previously been reported for cetuximab monotherapy (43, 44) or erlotinib monotherapy (45). Previous clinical studies on combination treatments of HER3-targeting antibodies in combination with erlotinib in NSCLC have demonstrated comparable response rates (40, 46, 47).

Subsequently we attempted to assess the clinical activity of lumretuzumab plus erlotinib in a patient population that was more likely to respond to HER3-targeting therapy. Preclinical studies (25, 26) as well as early clinical studies (22, 27, 28, 48, 49) reported an association between higher HRG mRNA expression levels and susceptibility to HER3-targeting therapies. Modest but statistically significant prolonged median PFS was reported for HRG-high–expressing patients, that is, a 0.2-month increase in a study reported by Sequist and colleagues (40) and a 1.6-month increase in a study reported by von Pawel and colleagues (46).

In the current study, we observed higher HRG mRNA expression levels in squamous carcinomas as compared with adenocarcinomas, which is in line with the described analysis from internal tumor bank samples and TCGA data. We therefore chose to enrich for higher HRG mRNA expression in the lumretuzumab plus erlotinib extension cohort by selectively enrolling sqNSCLC patients. However, lumretuzumab plus erlotinib did not demonstrate higher response rates, longer duration of responses, or more pronounced disease stabilization than what would have been expected with erlotinib alone, even when taking HRG and HER3 mRNA expression levels into account. Slightly higher DCR and PFS were observed in the HRG- and HER3-high–expressing sqNSCLC patients, suggesting some biological evidence for the hypothesis of HRG and HER3 expression levels and HER3 blockade. However, the short-lived responses and disease stabilization suggest that these tumors are not dependent on HER3 signaling and that alternative signaling pathways may be involved as escape mechanisms. In accordance to our findings, a phase II/III study in NSCLC patients treated with the HER3-targeting mAb patritumab and erlotinib was stopped recently due to a lack of efficacy (ORR of 2.2% for patritumab plus erlotinib compared with 6.3% for erlotinib alone and median PFS of 1.9 months for patritumab plus erlotinib compared with 2.7 months for erlotinib alone in patients considered to have high HRG levels; ref. 47). Similarly, no correlation was found between HER3 expression and response or clinical benefit in several phase I and II studies testing HER3-tageting therapies (27, 46, 50, 51). Recently, a dual-action antibody targeting EGFR and HER3 also failed to improve PFS and overall survival compared with cetuximab therapy in SCCHN patients in a randomized phase II trial, even in the HRG-high–expressing subpopulation (52).

In conclusion, combination treatment of lumretuzumab with cetuximab or erlotinib was manageable but demonstrated modest clinical activity. Apart from two exceptional responders, a patient with SCCHN achieving a prolonged CR and a patient with ovarian cancer achieving a prolonged PR, both showing higher expression levels of HRG mRNA, the current study was not able to identify a robust biomarker signal that could serve as response prediction biomarker for lumretuzumab in the future. Furthermore, in the presented sqNSCLC cohort, even higher HRG or HER3 mRNA expression levels were not associated with increased ORR or clinically meaningful durations of response, refuting our initial hypothesis. Thus, the response and resistance mechanisms to lumretuzumab-based therapy may be multifaceted or simply undetectable in our cohort of sqNSCLC. Together with the results from randomized trials in SCCHN (52) and NSCLC (47), however, our data provide further evidence that adding HER3 to EGFR-targeting therapy is not sufficient to derive clinically meaningful benefit.

W. Jacob, S. Vega-Harring, C. Adessi, and M. Weisser ownership interest (including patents) in Roche. A. Cervantes reports receiving speakers bureau honoraria from Roche. M. Lolkema reports receiving commercial research grants from Astellas, Johnson & Johnson, and Sanofi. E. Felip reports receiving speakers bureau honoraria from AstraZeneca, Novartis, and Pfizer and is a consultant/advisory board member for Boehringer Ingelheim, Eli Lilly, MSD, Pfizer, and Roche. No potential conflicts of interest were disclosed by the other authors.

Conception and design: D. Meulendijks, W. Jacob, E.E. Voest, J.H.M. Schellens, M. Thomas, G. Meneses-Lorente, I. James, M. Nguyen, F. Michielin, B. Bossenmaier, M. Weisser, U.N. Lassen

Development of methodology: W. Jacob, M.P. Lolkema, J.H.M. Schellens, M. Thomas, M. Ceppi, G. Meneses-Lorente, M. Weisser, U.N. Lassen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D. Meulendijks, E.E. Voest, M. Mau-Sorensen, M. Martinez-Garcia, A. Taus, T. Fleitas, A. Cervantes, M.P. Lolkema, M.H.G. Langenberg, M.J. De Jonge, S. Sleijfer, J.-Y. Han, A. Calles, E. Felip, S.-W. Kim, J.H.M. Schellens, I. James, R. Dua, L. Steiner, U.N. Lassen

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): W. Jacob, E.E. Voest, M.P. Lolkema, M.J. De Jonge, A. Calles, J.H.M. Schellens, S. Wilson, M. Thomas, M. Ceppi, G. Meneses-Lorente, I. James, S. Vega-Harring, M. Nguyen, C. Adessi, F. Michielin, M. Weisser, U.N. Lassen

Writing, review, and/or revision of the manuscript: D. Meulendijks, W. Jacob, E.E. Voest, M. Mau-Sorensen, M. Martinez-Garcia, A. Taus, T. Fleitas, A. Cervantes, M.H.G. Langenberg, M.J. De Jonge, S. Sleijfer, J.-Y. Han, A. Calles, E. Felip, S.-W. Kim, J.H.M. Schellens, M. Thomas, M. Ceppi, G. Meneses-Lorente, I. James, S. Vega-Harring, C. Adessi, F. Michielin, B. Bossenmaier, M. Weisser, U.N. Lassen

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D. Meulendijks, W. Jacob, F. Michielin, U.N. Lassen

Study supervision: D. Meulendijks, W. Jacob, M.J. De Jonge, M. Weisser, U.N. Lassen

Other (responsible for clinical development): M. Weisser

The authors thank the patients and their families for their participation in this study, and the staff at the study sites.

This study was funded by F. Hoffmann–La Roche Ltd.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Campbell
MR
,
Amin
D
,
Moasser
MM
. 
HER3 comes of age: new insights into its functions and role in signaling, tumor biology, and cancer therapy
.
Clin Cancer Res
2010
;
16
:
1373
83
.
2.
Sergina
NV
,
Rausch
M
,
Wang
DH
,
Blair
J
,
Hann
B
,
Shokat
KM
, et al
Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3
.
Nature
2007
;
445
:
437
41
.
3.
Jain
A
,
Penuel
E
,
Mink
S
,
Schmidt
J
,
Hodge
A
,
Favero
K
, et al
HER kinase axis receptor dimer partner switching occurs in response to EGFR tyrosine kinase inhibition despite failure to block cellular proliferation
.
Cancer Res
2010
;
70
:
1989
99
.
4.
Wang
S
,
Huang
X
,
Lee
CK
,
Liu
B
. 
Elevated expression of erbB3 confers paclitaxel resistance in erbB2-overexpressing breast cancer cells via upregulation of Survivin
.
Oncogene
2010
;
29
:
4225
36
.
5.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
6.
Wheeler
DL
,
Huang
S
,
Kruser
TJ
,
Nechrebecki
MM
,
Armstrong
EA
,
Benavente
S
, et al
Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members
.
Oncogene
2008
;
27
:
3944
56
.
7.
Bieche
I
,
Onody
P
,
Tozlu
S
,
Driouch
K
,
Vidaud
M
,
Lidereau
R
. 
Prognostic value of ERBB family mRNA expression in breast carcinomas
.
Int J Cancer
2003
;
106
:
758
65
.
8.
Muller-Tidow
C
,
Diederichs
S
,
Bulk
E
,
Pohle
T
,
Steffen
B
,
Schwable
J
, et al
Identification of metastasis-associated receptor tyrosine kinases in non-small cell lung cancer
.
Cancer Res
2005
;
65
:
1778
82
.
9.
Yi
ES
,
Harclerode
D
,
Gondo
M
,
Stephenson
M
,
Brown
RW
,
Younes
M
, et al
High c-erbB-3 protein expression is associated with shorter survival in advanced non-small cell lung carcinomas
.
Mod Pathol
1997
;
10
:
142
8
.
10.
Tanner
B
,
Hasenclever
D
,
Stern
K
,
Schormann
W
,
Bezler
M
,
Hermes
M
, et al
ErbB-3 predicts survival in ovarian cancer
.
J Clin Oncol
2006
;
24
:
4317
23
.
11.
Beji
A
,
Horst
D
,
Engel
J
,
Kirchner
T
,
Ullrich
A
. 
Toward the prognostic significance and therapeutic potential of HER3 receptor tyrosine kinase in human colon cancer
.
Clin Cancer Res
2012
;
18
:
956
68
.
12.
Yonesaka
K
,
Hirotani
K
,
Kawakami
H
,
Takeda
M
,
Kaneda
H
,
Sakai
K
, et al
Anti-HER3 monoclonal antibody patritumab sensitizes refractory non-small cell lung cancer to the epidermal growth factor receptor inhibitor erlotinib
.
Oncogene
2016
;
35
:
878
86
.
13.
Baselga
J
,
Cortes
J
,
Im
SA
,
Clark
E
,
Ross
G
,
Kiermaier
A
, et al
Biomarker analyses in CLEOPATRA: a phase III, placebo-controlled study of pertuzumab in human epidermal growth factor receptor 2-positive, first-line metastatic breast cancer
.
J Clin Oncol
2014
;
32
:
3753
61
.
14.
Makhija
S
,
Amler
LC
,
Glenn
D
,
Ueland
FR
,
Gold
MA
,
Dizon
DS
, et al
Clinical activity of gemcitabine plus pertuzumab in platinum-resistant ovarian cancer, fallopian tube cancer, or primary peritoneal cancer
.
J Clin Oncol
2010
;
28
:
1215
23
.
15.
Kurzeder
C
,
Bover
I
,
Marme
F
,
Rau
J
,
Pautier
P
,
Colombo
N
, et al
Double-blind, placebo-controlled, randomized phase III trial evaluating pertuzumab combined with chemotherapy for low tumor human epidermal growth factor receptor 3 mRNA-expressing platinum-resistant ovarian cancer (PENELOPE)
.
J Clin Oncol
2016
;
34
:
2516–25
.
16.
Sithanandam
G
,
Anderson
LM
. 
The ERBB3 receptor in cancer and cancer gene therapy
.
Cancer Gene Ther
2008
;
15
:
413
48
.
17.
Wilson
TR
,
Lee
DY
,
Berry
L
,
Shames
DS
,
Settleman
J
. 
Neuregulin-1-mediated autocrine signaling underlies sensitivity to HER2 kinase inhibitors in a subset of human cancers
.
Cancer Cell
2011
;
20
:
158
72
.
18.
Sheng
Q
,
Liu
XG
,
Fleming
E
,
Yuan
K
,
Piao
HY
,
Chen
JY
, et al
An activated ErbB3/NRG1 autocrine loop supports in vivo proliferation in ovarian cancer cells (vol 17, pg 298, 2010)
.
Cancer Cell
2010
;
17
:
412
.
19.
Zhou
BBS
,
Peyton
M
,
He
B
,
Liu
CN
,
Girard
L
,
Caudler
E
, et al
Targeting ADAM-mediated ligand cleavage to inhibit HER3 and EGFR pathways in non-small cell lung cancer
.
Cancer Cell
2006
;
10
:
39
50
.
20.
Shames
DS
,
Carbon
J
,
Walter
K
,
Jubb
AM
,
Kozlowski
C
,
Januario
T
, et al
High heregulin expression is associated with activated HER3 and may define an actionable biomarker in patients with squamous cell carcinomas of the head and neck
.
PLoS One
2013
;
8
:
e56765
.
21.
Mirschberger
C
,
Schiller
CB
,
Schraml
M
,
Dimoudis
N
,
Friess
T
,
Gerdes
CA
, et al
RG7116, a therapeutic antibody that binds the inactive HER3 receptor and is optimized for immune effector activation
.
Cancer Res
2013
;
73
:
5183
94
.
22.
Meulendijks
D
,
Jacob
W
,
Martinez-Garcia
M
,
Taus
A
,
Lolkema
MP
,
Voest
EE
, et al
First-in-human phase I study of lumretuzumab, a glycoengineered humanized anti-HER3 monoclonal antibody, in patients with metastatic or advanced HER3-positive solid tumors
.
Clin Cancer Res
2016
;
22
:
877
885
.
23.
Huang
S
,
Li
CR
,
Armstrong
EA
,
Peet
CR
,
Saker
J
,
Amler
LC
, et al
Dual targeting of EGFR and HER3 with MEHD7945A overcomes acquired resistance to EGFR inhibitors and radiation
.
Cancer Res
2013
;
73
:
824
33
.
24.
Kawakami
H
,
Okamoto
I
,
Yonesaka
K
,
Okamoto
K
,
Shibata
K
,
Shinkai
Y
, et al
The anti-HER3 antibody patritumab abrogates cetuximab resistance mediated by heregulin in colorectal cancer cells
.
Oncotarget
2014
;
5
:
11847
56
.
25.
Schoeberl
B
,
Faber
AC
,
Li
DN
,
Liang
MC
,
Crosby
K
,
Onsum
M
, et al
An ErbB3 antibody, MM-121, is active in cancers with ligand-dependent activation
.
Cancer Res
2010
;
70
:
2485
94
.
26.
Meetze
K
,
Vincent
S
,
Tyler
S
,
Mazsa
EK
,
Delpero
AR
,
Bottega
S
, et al
Neuregulin 1 expression is a predictive biomarker for response to AV-203, an ERBB3 inhibitory antibody, in human tumor models
.
Clin Cancer Res
2015
;
21
:
1106
14
.
27.
Mendell
J
,
Freeman
DJ
,
Feng
W
,
Hettmann
T
,
Schneider
M
,
Blum
S
, et al
Clinical translation and validation of a predictive biomarker for patritumab, an anti-human epidermal growth factor receptor 3 (HER3) monoclonal antibody, in patients with advanced non-small cell lung cancer
.
EBioMedicine
2015
;
2
:
264
71
.
28.
Juric
D
,
Dienstmann
R
,
Cervantes
A
,
Hidalgo
M
,
Messersmith
W
,
Blumenschein
GR
, et al
Safety and pharmacokinetics/pharmacodynamics of the first-in-class dual action HER3/EGFR antibody MEHD7945A in locally advanced or metastatic epithelial tumors
.
Clin Cancer Res
2015
;
21
:
2462
70
.
29.
NIH
. 
The Cancer Genome Atlas [TCGA]
.
Available from
: https://cancergenome.nih.gov/.
30.
Lee
ES
,
Son
DS
,
Kim
SH
,
Lee
J
,
Jo
J
,
Han
J
, et al
Prediction of recurrence-free survival in postoperative non-small cell lung cancer patients by using an integrated model of clinical information and gene expression
.
Clin Cancer Res
2008
;
14
:
7397
404
.
31.
Shin
DH
,
Lee
D
,
Hong
DW
,
Hong
SH
,
Hwang
JA
,
Lee
BI
, et al
Oncogenic function and clinical implications of SLC3A2-NRG1 fusion in invasive mucinous adenocarcinoma of the lung
.
Oncotarget
2016
;
7
:
69450
65
.
32.
Fernandez-Cuesta
L
,
Thomas
RK
. 
Molecular pathways: targeting NRG1 fusions in lung cancer
.
Clin Cancer Res
2015
;
21
:
1989
94
.
33.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
34.
Fracasso
PM
,
Burris
H
,
Arquette
MA
,
Govindan
R
,
Gao
F
,
Wright
LP
, et al
A phase 1 escalating single-dose and weekly fixed-dose study of cetuximab: Pharmacokinetic and pharmacodynamic rationale for dosing
.
Clin Cancer Res
2007
;
13
:
986
93
.
35.
Lenz
HJ
,
Van Cutsem
E
,
Khambata-Ford
S
,
Mayer
RJ
,
Gold
P
,
Stella
P
, et al
Multicenter phase II and translational study of cetuximab in metastatic colorectal carcinoma refractory to irinotecan, oxaliplatin, and fluoropyrimidines
.
J Clin Oncol
2006
;
24
:
4914
21
.
36.
Reck
M
,
Mok
T
,
Wolf
J
,
Heigener
D
,
Wu
YL
. 
Reviewing the safety of erlotinib in non-small cell lung cancer
.
Expert Opin Drug Saf
2011
;
10
:
147
57
.
37.
Cleary
JM
,
McRee
AJ
,
Shapiro
GI
,
Tolaney
SM
,
O'Neil
BH
,
Kearns
JD
, et al
A phase 1 study combining the HER3 antibody seribantumab (MM-121) and cetuximab with and without irinotecan
.
Invest New Drugs
2017
;
35
:
68
78
.
38.
Papadopoulos
KP
,
Adjei
AA
,
Rasco
DW
,
Liu
LM
,
Kao
RJ
,
Brownstein
CM
, et al
Phase 1 study of REGN1400 (anti-ErbB3) combined with erlotinib or cetuximab in patients (pts) with advanced non-small cell lung cancer (NSCLC), colorectal cancer (CRC), or head and neck cancer (SCCHN)
.
J Clin Oncol
32
:
5s
, 
2014
(
suppl; abstr 2516
).
39.
Sequist
LV
,
Modiano
MR
,
Rixe
O
,
Jackman
DM
,
Andreas
K
,
Pearlberg
J
, et al
Targeting EGFR and ERBB3 in lung cancer patients: clinical outcomes in a phase I trial of MM-121 in combination with erlotinib
.
J Clin Oncol
30:15s, 2012 (suppl; abstr 7556)
.
40.
Sequist
LV
,
Lopez-Chavez
A
,
Doebele
RC
,
Gray
JE
,
Harb
WA
,
Modiano
MR
, et al
A randomized phase 2 trial of MM-121, a fully human monoclonal antibody targeting ErbB3, in combination with erlotinib in EGFR wild-type NSCLC patients
.
J Clin Oncol
32:15s, 2014 (suppl; abstr 8051)
.
41.
Keely
SJ
,
Barrett
KE
. 
ErbB2 and ErbB3 receptors mediate inhibition of calcium-dependent chloride secretion in colonic epithelial cells
.
J Biol Chem
1999
;
274
:
33449
54
.
42.
Van Sebille
YZA
,
Gibson
RJ
,
Wardill
HR
,
Bowen
JM
. 
ErbB small molecule tyrosine kinase inhibitor (TKI) induced diarrhoea: Chloride secretion as a mechanistic hypothesis
.
Cancer Treat Rev
2015
;
41
:
646
52
.
43.
Cunningham
D
,
Humblet
Y
,
Siena
S
,
Khayat
D
,
Bleiberg
H
,
Santoro
A
, et al
Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer
.
N Engl J Med
2004
;
351
:
337
45
.
44.
Vermorken
JB
,
Trigo
J
,
Hitt
R
,
Koralewski
P
,
Diaz-Rubio
E
,
Rolland
F
, et al
Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy
.
J Clin Oncol
2007
;
25
:
2171
7
.
45.
Shepherd
FA
,
Pereira
JR
,
Ciuleanu
T
,
Tan
EH
,
Hirsh
V
,
Thongprasert
S
, et al
Erlotinib in previously treated non-small-cell lung cancer
.
N Engl J Med
2005
;
353
:
123
32
.
46.
Von Pawel
J
,
Tseng
J
,
Dediu
M
,
Schumann
C
,
Moritz
B
,
Mendell-Harary
J
, et al
Phase 2 HERALD study of patritumab (P) with erlotinib (E) in advanced NSCLC subjects (SBJs)
.
J Clin Oncol
32:5s, 2014 (suppl; abstr 8045)
.
47.
Paz-Arez
L
,
Serwatowski
P
,
Szczęsna
A
,
Von Pawel
J
,
Toschi
L
,
Tibor
C
, et al
P3.02b-045 patritumab plus erlotinib in EGFR wild-type advanced non–small cell lung cancer (NSCLC): part a results of HER3-lung study
.
J Thorac Oncol
2017
;
12
:
S1214
5
.
48.
Macbeath
G
,
Adiwijaya
B
,
Liu
J
,
Sequist
LV
,
Pujade-Lauraine
E
,
Higgins
M
, et al
246PA meta-analysis of biomarkers in three randomized, phase 2 studies of MM-121, a ligand-blocking anti-ERBB3 antibody, in patients with ovarian, lung, and breast cancers
.
Ann Oncol
2014
;
25
:
iv82
.
49.
Sarantopoulos
J
,
Gordon
MS
,
Harvey
RD
,
Sankhala
KK
,
Malik
L
,
Mahalingam
D
, et al
First-in-human phase 1 dose-escalation study of AV-203, a monoclonal antibody against ERBB3, in patients with metastatic or advanced solid tumors
.
J Clin Oncol
32:15s, 2014 (suppl; abstr 11113)
.
50.
Hill
AG
,
Findlay
M
,
Burge
M
,
Jackson
C
,
Alfonso
PG
,
Samuel
L
, et al
Randomized phase II study of duligotuzumab plus FOLFIRI versus cetuximab plus FOLFIRI in 2nd-line patients with KRAS wild-type (wt) metastatic colorectal cancer (mCRC)
.
Cancer Res
2015
;
75
Suppl 15
:
CT110
.
51.
Jimeno
A
,
Machiels
JP
,
Wirth
L
,
Specenier
P
,
Seiwert
TY
,
Mardjuadi
F
, et al
Phase Ib study of duligotuzumab (MEHD7945A) plus cisplatin/5-fluorouracil or carboplatin/paclitaxel for first-line treatment of recurrent/metastatic squamous cell carcinoma of the head and neck
.
Cancer
2016
;
122
:
3803
11
.
52.
Fayette
J
,
Wirth
L
,
Oprean
C
,
Udrea
A
,
Jimeno
A
,
Rischin
D
, et al
Randomized phase II study of duligotuzumab (MEHD7945A) vs. cetuximab in squamous cell carcinoma of the head and neck (MEHGAN Study)
.
Front Oncol
2016
;
6
:
232
.

Supplementary data