Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jul 15, 2018; 10(7): 159-171
Published online Jul 15, 2018. doi: 10.4251/wjgo.v10.i7.159
HER2 inhibition in gastro-oesophageal cancer: A review drawing on lessons learned from breast cancer
Hazel Lote, Nicola Valeri, Centre for Molecular Pathology, Institute of Cancer Research, Sutton SM2 5NG, United Kingdom
Hazel Lote, Nicola Valeri, Ian Chau, Department of Medicine, Royal Marsden Hospital, Sutton SM2 5PT, United Kingdom
ORCID number: Hazel Lote (0000-0003-1172-0372); Nicola Valeri (0000-0002-5426-5683); Ian Chau (0000-0003-0286-8703).
Author contributions: Lote H wrote the original manuscript and revised it following peer review comments; Valeri N reviewed the manuscript; Chau I reviewed and contributed to the content of the manuscript.
Supported by National Health Service funding to the National Institute for Health Research Biomedical Research Centre at the Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, No. A62, No. A100, No. A101 and No. A159; Cancer Research UK funding, No. CEA A18052.
Conflict-of-interest statement: No potential conflicts of interest relevant to this article were reported.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Ian Chau, FRCP (Hon), MD, MRCP, Doctor, Department of Medicine, Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, United Kingdom. ian.chau@rmh.nhs.uk
Telephone: +44-208-6613582 Fax: +44-208-6613890
Received: January 25, 2018
Peer-review started: January 26, 2018
First decision: March 7, 2018
Revised: May 25, 2018
Accepted: May 30, 2018
Article in press: May 30, 2018
Published online: July 15, 2018

Abstract

Human epidermal growth factor receptor 2 (HER2)-inhibition is an important therapeutic strategy in HER2-amplified gastro-oesophageal cancer (GOC). A significant proportion of GOC patients display HER2 amplification, yet HER2 inhibition in these patients has not displayed the success seen in HER2 amplified breast cancer. Much of the current evidence surrounding HER2 has been obtained from studies in breast cancer, and we are only recently beginning to improve our understanding of HER2-amplified GOC. Whilst there are numerous licensed HER2 inhibitors in breast cancer, trastuzumab remains the only licensed HER2 inhibitor for HER2-amplified GOC. Clinical trials investigating lapatinib, trastuzumab emtansine, pertuzumab and MM-111 in GOC have demonstrated disappointing results and have not yet changed the treatment paradigm. Trastuzumab deruxtecan may hold promise and is currently being investigated in phase II trials. HER2 amplified GOC differs from breast cancer due to inherent differences in the HER2 amino-truncation and mutation rate, loss of HER2 expression, alterations in HER2 signalling pathways and differences in insulin-like growth factor-1 receptor and MET expression. Epigenetic alterations involving different microRNA profiles in GOC as compared to breast cancer and intrinsic differences in the immune environment are likely to play a role. The key to effective treatment of HER2 amplified GOC lies in understanding these mechanisms and tailoring HER2 inhibition for GOC patients in order to improve clinical outcomes.

Key Words: Human epidermal growth factor receptor 2, Gastro-oesophageal cancer, Trastuzumab, Resistance, Biomarkers, Breast cancer

Core tip: Human epidermal growth factor receptor 2 (HER2)-inhibition is an important therapeutic strategy in HER2-amplified gastro-oesophageal cancer (GOC). A significant proportion of GOC patients display HER2 amplification, yet HER2 inhibition in these patients has not displayed the success seen in HER2 amplified breast cancer. We evaluate current clinical and laboratory evidence surrounding HER2 inhibition in GOC. Inherent differences in the HER2 receptor, signalling pathways, associated microRNA signature and immune environment may partly explain the disappointing clinical trial outcomes seen in GOC. Only with improved understanding of HER2 inhibition can effective treatment be provided in order to improve clinical outcomes for patients.



INTRODUCTION

Cancer therapy is becoming increasingly personalised and molecularly targeted, using biomarkers to identify patients most likely to respond to therapy. Human epidermal growth factor receptor 2 (HER2)-amplified cancer is defined as cancer with HER2 protein overexpression ± HER2 gene amplification[1]. It represents a molecularly-defined subgroup of malignancy and is known to exist in breast and gastro-oesophageal cancers (GOC), among others[1]. Whereas the treatment for HER2-amplified breast cancer patients has been extremely successful, the treatment for GOC has been less so. In this review, we explore the mechanisms by which HER2 amplification contributes to cancer progression and prognosis, methods of targeting HER2 amplification, mechanisms of resistance to HER2 therapy, strategies to overcome resistance, biomarkers and future directions.

HER2 RECEPTOR AND ITS INTERACTIONS

HER2, encoded by the ERBB2 oncogene on chromosome 17q21[2], is a member of the epidermal growth factor receptor (EGFR) family associated with tumour cell proliferation, apoptosis, adhesion, migration and differentiation[3]. All studies investigating HER2 receptor interactions have been conducted in breast cancer cells, and a literature search did not reveal any studies of HER2 receptor interactions conducted specifically in GOC. Given the relatively disappointing results seen in GOC, we suggest it may be worthwhile exploring HER2 receptor interactions specifically in GOC, to investigate whether there are any mechanistic differences in HER2 binding and signalling between breast and GOC.

HER2 RECEPTOR OVEREXPRESSION AND ONCOGENIC MECHANISMS IN BREAST AND GOC

In both breast cancer and GOC, HER2 overexpression occurs in approximately 20%[4,5]. The Gastric Cancer Genome Atlas [part of The Cancer Genome Atlas (TCGA)] recently classified gastric cancer into four subtypes and found that HER2 overexpression occurs only in Epstein-Barr virus (EBV)-positive tumours, genomically-stable (GS) tumours and tumours with chromosomal instability (CIN) but not in microsatellite unstable (MSI-high) tumours[6]. Mechanisms by which HER2 overexpression can be oncogenic are complex, with activation of RAS-MAPK, c-jun and Akt-mTOR pathways[3] (Figure 1). HER2 overexpression may lead to formation of HER2 homodimers and ligand-independent downstream signalling[3]. The majority of studies investigating HER2 overexpression oncogenicity have been conducted in breast cancer, and mechanisms may differ in GOC.

Figure 1
Figure 1 Simplified diagram showing signalling pathways related to human epidermal growth factor receptor 2 in gastric adenocarcinoma. VEGFR: Vascular endothelial growth factor receptor; FGFR: Fibroblast growth factor receptor; PKC: Protein kinase C; FAK: Focal adhesion kinase; HER2: Human epidermal growth factor receptor 2; MAPK: Mitogen-activated protein kinase; mTOR: Mammalian target of rapamycin.
INFLUENCE OF HER2 STATUS ON PROGNOSIS IN BREAST AND GASTRIC CANCER

In contrast to breast cancer, HER2 overexpression does not impact survival in GOC[2]. Large phase III prospective randomised controlled trials such as ToGA[5], LOGiC[7] and TYTAN[8] demonstrate that patients with HER2 amplified GOC who receive the control arm (chemotherapy alone) have an overall survival (OS) similar to all-comers (Table 1)[5,7-9]. In the first-line ToGA and LOGiC trials, OS was 11.1 mo and 10.5 mo, respectively, in the control arms[5,7], compared to OS in all-comers of 9.9 mo in the Phase III REAL2 trial[10]. In the 2nd-line TYTAN trial, OS was 8.9 mo in the control[8,9], which compared favourably to OS in all-comers treated with paclitaxel in the control arms of the RAINBOW (OS 7.4 mo)[11] and GOLD trials (OS 6.9 mo)[12]. This cross-trial comparison suggests that HER2 overexpression does not adversely affect GOC prognosis.

Table 1 Summary of selected randomized phase III HER2 trials in HER2-amplified gastro-oesophageal cancer and breast cancer.
Study titleSettingnTreatment armsPrimary endpointOSPFSHR and P value
Trastuzumab 1st line metastatic
ToGA[5]1st line metastatic GOC594Trastuzumab + chemotherapy vs chemotherapy aloneOSTrastuzumab + chemotherapy: 13.8 mo (95%CI: 12-16) Chemotherapy alone: OS 11.1 mo (10-13)Trastuzumab + chemotherapy: 6.7 mo (95%CI: 6-8) Chemotherapy alone: 5.5 mo (5-6)HR = 0.74; 95%CI: 0.60-0.91; P = 0.0046)
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2[19]1st line metastatic breast cancer469Trastuzumab + chemotherapy vs chemotherapy alonePFSTrastuzumab + chemotherapy: 25.1 mo Chemotherapy alone: 20.3 moTrastuzumab + chemotherapy: 7.4 mo Chemotherapy alone: 4.6 moP = 0.046
Lapatinib 1st line metastatic
LOGiC[7]1st line metastatic GOC545Lapatinib + CAPOX vs Placebo + CAPOXOSLapatinib + CAPOX: 12.2 mo (95%CI: 10.6-14.2) Placebo + CAPOX: 10.5 mo (9.0-11.3)Lapatinib + CAPOX: 6 mo (95%CI: 5.6-7.0) Placebo + CAPOX: 5.4 mo (4.4-5.7)HR = 0.91; 95%CI: 0.73-1.12 P value not significant (exact value not given)
Randomized trial of lapatinib vs placebo added to paclitaxel in the treatment of human epidermal growth factor receptor 2-overexpressing metastatic breast cancer[29]1st line metastatic breast cancer444Lapatinib + paclitaxel vs Placebo + paclitaxelOSLapatinib + paclitaxel: 27.8 mo (95%CI: 23.2-32.2 mo) Placebo + paclitaxel: 20.5 mo (17.9-24.3 mo)Lapatinib + paclitaxel: 9.7 mo (95%CI: 9.2-11.1 mo) Placebo + paclitaxel: 6.5 mo (5.5-7.3 mo)HR = 0.74; 95%CI: 0.58-0.94; P = 0.0124
Lapatinib 2nd line metastatic
Tytan[8]2nd line metastatic GOC261Lapatinib + Paclitaxel vs Paclitaxel aloneOSLapatinib + Paclitaxel: 11.0 mo Paclitaxel alone: 8.9 moLapatinib + Paclitaxel: 5.5 mo Paclitaxel alone: 4.4 moHR = 0.84; 95%CI: 0.64-1.11 P = 0.1044
Lapatinib plus capecitabine for HER2-Positive advanced breast Cancer[30]2nd line metastatic breast cancer324 included in preliminary analysisLapatinib + capecitabine vs capecitabine alonePFSNot reportedLapatinib + capecitabin: 8.4 mo Capecitabine alone: 4.4 moHR = 0.49; 95%CI: 0.34 to 0.71; P < 0.001
T-DM1 2nd line metastatic
GATSBY[35]2nd line metastatic GOC345T-DM1 vs taxaneOST-DM1: 7.9 mo Taxane: 8.6 moT-DM1: 2.7 mo Taxane: 2.9 moHR = 1.15, 95%CI: 0.87–1.51; P = 0·86
EMILIA[33]2nd line metastatic breast cancer991T-DM1 vs lapatinib + capecitabinePFST-DM1: 30.9 mo Lapatinib + capecitabine: 25.1 moT-DM1: 9.6 mo Lapatinib + capecitabine: 6.4 moHR = 0.65; 95%CI: 0.55 to 0.77; P < 0.001
HER2 SCORING CRITERIA, DISCORDANCE AND HETEROGENEITY IN GOC AND BREAST CANCER

The HER2 scoring system in breast cancer was developed prior to the scoring system for GOC and was standardised in 2007 following an expert panel forum[13]. The ToGA trial used a new immunohistochemistry (IHC) scoring criteria developed by Hofmann[14] for gastric cancer due to inherent biological differences compared to breast cancer, such as tumour heterogeneity and baso(lateral) membrane staining[5,14]. Some criteria were the same as breast cancer: HER2 positivity was defined as an IHC score of 3+ and/or erbB-2 amplification detected using fluorescent in-situ-hybridisation (FISH)[5,14]. Notably, GOC patients with highly amplified HER2 gene experience better response and survival than patients with lower HER2 gene amplification levels when treated with 1st-line trastuzumab plus chemotherapy for metastatic gastric cancer[15].

HER2 expression in primary and metastatic sites demonstrates heterogeneity more frequently in GOC than in breast cancer[16,17], and discordance between IHC and FISH results occur more frequently in GOC than in breast cancer[18]. This may explain the limited success of targeted anti-HER2 therapy in GOC. If only a small proportion of GOC cells shows HER2 overexpression and if our detection methods are unreliable, GOC cancer cells that do not overexpress HER2 will not be effectively targeted with anti-HER2 therapy, and we may be failing to treat adequately some patients with HER2 overexpression.

THERAPEUTIC AGENTS TARGETING THE HER2 SIGNALLING PATHWAY
Trastuzumab

The efficacy of trastuzumab (a monoclonal antibody against HER2) in breast cancer in combination with chemotherapy has been convincingly demonstrated in both metastatic (OS 25.1 mo in patients receiving trastuzumab + chemotherapy vs 20.3 mo in those receiving chemotherapy alone, Table 1)[19] and adjuvant settings[20]. Breast cancer OS is, however, influenced by the greater number of treatment options in the 2nd-line setting and beyond.

In GOC, trastuzumab is the only licensed anti-HER2 treatment, following positive results from the ToGA trial, an open-label, international, phase 3, randomised controlled trial evaluating trastuzumab plus platinum-fluoropyrimidine chemotherapy for 1st-line treatment of HER2 positive GOC (Table 1)[5]. Median OS was initially reported as 13.8 mo (95%CI: 12-16) in patients receiving trastuzumab plus chemotherapy vs 11.1 mo (10-13) in patients receiving chemotherapy alone (HR = 0.74; 95%CI: 0.60-0.91; P = 0.0046)[5]. This led to trastuzumab plus platinum-fluoropyrimidine chemotherapy followed by trastuzumab maintenance becoming the standard of care in 1st-line metastatic GOC patients[5]. Updated OS (after a further 1 year of follow-up) released by the United States Food and Drug Administration (FDA) in 2016 showed median OS of 13.1 mo (95%CI: 11.9-15.1) in the trastuzumab plus chemotherapy arm and 11.7 mo (95%CI: 10.3-13.0) in the control arm (HR = 0.8, 95%CI: 0.67-0.97)[21]. Subgroup analysis demonstrated that patients with IHC 3+ HER2 expression experienced the greatest benefit from trastuzumab (294 patients, HR = 0.66, 95%CI: 0.5-0.87). Patients with IHC 2+ HER2 expression gained less benefit from the addition of trastuzumab (160 patients, HR = 0.78, 95%CI: 0.55-1.10), and patients with IHC 1 or 1+ gained no benefit (133 patients, HR = 1.33, 95%CI: 0.92-1.92)[21]. Recent data on two different doses of trastuzumab in combination with chemotherapy in GOC found that a higher trastuzumab maintenance dose does not convey additional survival benefit (OS 12.5 mo in the 8 mg/kg + 6 mg/kg group vs 10.6 mo in the 8 mg/kg + 10 mg/kg group)[22].

It remains to be seen whether trastuzumab confers a survival benefit in the neo-adjuvant/perioperative/adjuvant setting in combination with chemotherapy + surgery ± radiotherapy, and several phase 2 trials are underway to address this question (UMIN 000016920, NCT01472029, NCT02250209, Table 2)[23,24]. Perioperative trastuzumab appears to be safe and well tolerated[25]. One Phase II trial evaluating capecitabine + oxaliplatin with trastuzumab three cycles pre-operatively and post-operatively followed by 12 mo adjuvant trastuzumab reported an 18 mo DFS of 71% (95%CI: 53%-83%), a 24 mo DFS of 60% and a median follow-up of 24.1 mo (median DFS and OS not reached)[26]. Although a phase III trial evaluating radiotherapy + chemotherapy ± trastuzumab is underway (NCT01196390, Table 2)[24], it is notable that trastuzumab is not being inve–stigated in phase III trials in the peri-operative GOC setting. This is likely due to the prohibitive number of patients (approximately 10000) that would require screening in order to recruit adequate numbers of patients for a sufficiently powered study, given that HER2 overexpression is around 20%[5], and a relatively small proportion of patients in Western countries are diagnosed with operable disease[27]. Only a proportion of these patients would have an adequate performance status to enter a clinical trial; therefore, trastuzumab will likely never be investigated in phase III trials in the peri-operative setting.

Table 2 Selected perioperative (neoadjuvant + adjuvant) clinical trials currently underway targeting HER2 in HER2-amplified localised gastro-oesophageal cancer.
Official study titleStage and study numberTreatment armsEstimated enrollmentPrimary endpoint
Trastuzumab
A randomized phase II trial of systemic chemotherapy with and without trastuzumab followed by surgery in HER2-positive advanced gastric or esophagogastric junction adenocarcinoma with extensive lymph node metastasis: Japan Clinical Oncology Group study JCOG1301 (Trigger Study)[23]Phase II UMIN 000016920Preoperative S-1 + cisplatin + trastuzumab vs S-1 + cisplatin Followed by adjuvant chemotherapy with S-1 for 1 yr130OS
Multicenter, explorative phase II study of perioperative 5-FU, leucovorin, docetaxel, and oxaliplatin (FLOT) in combination with trastuzumab in patients with HER2-positive, locally advanced, resectable adenocarcinoma of the gastroesophageal junction or stomach (HerFLOT)Phase II NCT01472029Pre-operative 5-FU + leucovorin + docetaxel + oxaliplatin (FLOT) + trastuzumab Post-operative trastuzumab monotherapy53pCR
Trastuzumab plus XELOX for HER2-positive stage III gastric cancer after D2 gastrectomy: prospective observational Study[77]Phase II NCT02250209Trastuzumab + capecitabine + oxaliplatin after D2 gastrectomy403-yr DFS
A phase III trial evaluating the addition of trastuzumab to trimodality treatment of HER2-overexpressing esophageal adenocarcinomaPhase III NCT01196390Radiotherapy + paclitaxel + carboplatin + trastuzumab vs Radiotherapy + paclitaxel + carboplatin591DFS
Lapatinib
A randomised phase II/III trial of peri-operative chemotherapy with or without bevacizumab in operable oesophagogastric adenocarcinoma and a feasibility study evaluating lapatinib in HER-2 positive oesophagogastric adenocarcinomas and (in selected centres) MRI and PET/CT sub-studies (STO3 trial)Phase II/III NCT00450203Epirubicin + cisplatin + capecitabine (ECX) + lapatinib vs ECX40 (within lapatinib sub-study)Safety
Pertuzumab + Trastuzumab
INtegratioN of trastuzumab, with or without pertuzumab, into perioperatiVe chemotherApy of HER-2 positive stomach cancer: the INNOVATION-TRIAL[77]Phase II NCT02205047Cisplatin/capecitabine or cisplatin/5-fluorouracil vs cisplatin/capecitabine + trastuzumab or cisplatin/5-fluorouracil + trastuzumab vs cisplatin/capecitabine + trastuzumab +pertuzumab or cisplatin/5-fluorouracil + trastuzumab + pertuzumab220Near complete pathological response rate
FLOT vs FLOT/Herceptin/Pertuzumab for perioperative therapy of adenocarcinoma of the stomach and gastroesophageal junction expressing HER-2 A phase II/III trial of the AIO. (PETRARCA study)Phase II/III NCT025814625-FU + leucovorin + docetaxel + oxaliplatin (FLOT) vs FLOT + trastuzumab + Pertuzumab404pCR OS
Feasibility study of chemoradiation, TRAstuzumab and pertuzumab in resectable HER2+esophageal carcinoma: the TRAP studyPhase I/II NCT02120911Pertuzumab + trastuzumab + standard chemoradiation with carboplatin and paclitaxel.40Safety

In advanced GOC, trastuzumab is being investigated in combination with bevacizumab (NCT01359397, Table 3), afatinib (NCT01522768, Table 3) and via intraperitoneal delivery (NCT01384253, Table 3)[24].

Table 3 Selected clinical trials currently underway targeting HER2 in advanced and metastatic HER2-amplified gastro-oesophageal cancer.
Official study titleStage and study numberTreatment armsEstimated enrollmentPrimary endpoint
Trastuzumab in combination with targeted therapies
Phase II study of docetaxel, oxaliplatin, capecitabine with bevacizumab and trastuzumab in case of HER2-positivity in patients with locally advanced or metastatic gastric cancer or adenocarcinoma of the gastro-oesophageal junction (B-DOCT study)Phase II NCT01359397Docetaxel, oxaliplatin, capecitabine, bevacizumab vs Docetaxel, oxaliplatin, capecitabine, bevacizumab, trastuzumabInformation not availablePFS
A phase II study of afatinib (BIBW 2992) and trastuzumab in patients with advanced HER2-positive trastuzumab-refractory advanced esophagogastric cancerPhase II NCT01522768Afatinib (BIBW 2992) + trastuzumab40ORR
Intraperitoneal trastuzumab
Phase I trial of intraperitoneal ²¹²Pb-TCMC-trastuzumab for HER-2 expressing malignancyPhase I NCT01384253²¹²Pb-TCMC-trastuzumab + trastuzumab36Safety
T-DM1
A combination study of kadcyla (trastuzumab emtansine, T-DM1) and capecitabine in patients with HER2-positive metastatic breast cancer and patients with HER2-positive locally advanced/ metastatic gastric cancer (TRAX-HER2 study)[78]Phase II NCT01702558Capecitabine + trastuzumab emtansine (T-DM1) vs T-DM1235Safety ORR
DS-8201
Phase 1, two-part, multicenter, non-randomized, open-label, multiple dose first-in-human study of DS-8201A, in subjects with advanced solid malignant tumors[36]Phase II NCT02564900Trastuzumab deruxtecan (DS-8201a)198Safety ORR
Lapatinib
Safety and clinical activity of lapatinib in patients with HER2-positive refractory advanced cancer: a phase II single arm prospective studyPhase II NCT02342587Lapatinib25ORR
New HER2 inhibitors
A phase I-II study to assess the safety, efficacy and pharmacokinetic profile of HM781-36B combined with paclitaxel and trastuzumab in patients with HER-2 positive advanced gastric cancerPhase I/II NCT01746771HM781-36B(Poziotinib) (Other Names: NOV120101) + paclitaxel + trastuzumab48Safety DLT
A phase 1, dose escalation study of MGAH22 in patients with refractory HER2 positive breast cancer and patients with other HER2 positive carcinomas for whom no standard therapy is availablePhase I NCT01148849MGAH22 (margetuximab)67Safety
A phase I multicenter, open-label, dose-escalation, and dose-expansion study to evaluate the safety, pharmacokinetics, immunogencity, and antitumor activity of MEDI4276 in subjects with select HER2-expressing advanced solid tumorsPhase I NCT02576548MEDI4276120Safety MTD
A phase I study of pyrotinib in combination with docetaxel in patients with HER2 positive advanced gastric cancerPhase I NCT02378389Pyrotinib + docetaxel28Safety
A two-part phase I, open label, dose escalation study to evaluate the safety, tolerability and pharmacokinetics of pyrotinib in patients whose disease progressed on prior HER2 targeted therapyPhase I NCT02500199Pyrotinib70Safety MTD
Neratinib
An open-label, multicenter, multinational, phase 2 study exploring the efficacy and safety of neratinib therapy in patients with solid tumors with activating HER2, HER3 or EGFR mutations or with EGFR gene amplificationPhase II NCT01953926Neratinib292ORR
HER2-targeted immunotherapy
A phase Ib/II study of pembrolizumab and monoclonal antibody therapy in patients with advanced cancer (PembroMab)[77]Phase I/II NCT02318901Pembrolizumab + trastuzumab vs pembrolizumab + ado-trastuzumab emtansine (T-DM1) vs pembrolizumab + cetuximab90Safety and dose-finding
A phase I study to evaluate the antitumor activity and safety of DUKE-002-VRP (HUHER2-ECD + TM), an alphaviral vector encoding the HER2 extracellular domain and transmembrane region, in patient with locally advanced or metastatic human epidermal growth factor receptor 2-positive (HER2+) cancers including breast cancerPhase I NCT01526473AVX90112Safety
HER2-peptide vaccination of patients with solid tumorsPhase I NCT02276300Cyclophosphamide sargramostim HER2-Peptid-Vakzine imiquimod12Safety
Lapatinib

Lapatinib is an oral tyrosine kinase inhibitor targeting EGFR and HER2[7,28]. In breast cancer, lapatinib demonstrated significant clinical benefit and is now a standard line of treatment[19,29,30]. In contrast, in GOC, although it showed promise in preclinical trials, lapatinib failed to translate into clinical benefit in both 1st-line (LOGiC)[7] and 2nd-line settings (TYTAN) (Table 1)[9]. The reasons for the disappointing results seen in GOC as compared to breast cancer may be related to lapatinib dosage, toxicities experienced, or different underlying HER2 signalling mechanisms in GOC and breast cancer. When lapatinib was combined with paclitaxel in a 1st-line breast cancer study and 2nd line GOC study (TYTAN), rates of AEs were broadly similar: 77% of patients in the lapatinib arm experienced diarrhoea in both the breast and TYTAN studies vs 29% of patients in the control arm in the breast study and 22% in the TYTAN study[8,29]. There was, however, a slightly higher rate of treatment discontinuation seen in GOC patients as compared to breast patients, with AEs resulting in treatment discontinuation in 16% in the lapatinib plus paclitaxel group vs 13% in the breast study)[8,29]. In the 1st-line GOC LOGiC trial (Table 1)[7], there were significantly higher toxicity rates in the lapatinib arm than the control arm, with 94% of patients experiencing adverse events (AEs) and 27% serious AEs (SAEs) in the lapatinib arm vs 88% AEs and 19% SAEs in the control arm. Diarrhoea occurred in 58% of patients receiving lapatinib vs 29% in the control arm, leading to lower relative drug exposure in the lapatinib arm[7]. Again lapatinib treatment resulted in higher rates of treatment discontinuation in GOC than breast cancer patients: 21% of patients in the lapatinib arm of LOGiC required treatment discontinuation vs 13% of breast cancer patients in the 2nd-line breast study receiving lapatinib plus capecitabine[30]. Overall, this suggests that the chemotherapy backbone with which to combine lapatinib is important, and chemotherapy drugs with overlapping toxicity may result in lower lapatinib dose-intensity and reduced efficacy in GOC. Additionally GOC patients frequently experience gastrointestinal side-effects prior to treatment and may be less able to tolerate lapatinib treatment. Another possible reason for the poor efficacy of lapatinib in GOC is that HER2 and EGFR signalling mechanisms may differ as compared to breast cancer.

Lapatinib is currently being investigated in Phase II/III trials in the peri-operative setting (STO3 trial, NCT00450203, Table 2)[24] and as monotherapy in the advanced setting (NCT02342587, Table 3)[24]. Safety data from the STO3 trial was presented at ESMO 2016 and suggested that administration of lapatinib at a dose of 1250 mg/d in combination with ECX chemotherapy (capecitabine 1000 mg/m2) was feasible, although there was increased diarrhoea (21% in ECX + lapatinib group vs 0% in ECX group) and neutropenia (42% in ECX + lapatinib group vs 21% in ECX group), which did not appear to compromise operative management[31].

T-DM1

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that combines the HER2-targeted properties of trastuzumab with the cytotoxic activity of emtansine, enabling selective delivery of chemotherapy to HER2-overexpressing cells[32]. Although T-DM1 demonstrated significant clinical benefit in the EMILIA breast cancer trial in the 2nd-line setting (Table 1)[33], a similar study (GATSBY, Table 1) in GOC failed to meet its primary endpoint or any of its secondary endpoints[34,35]. It is worth noting that nearly half of the patients in the GATSBY trial were from the Asia-Pacific region. These patients are generally fit with a good performance status; and, therefore, it is likely that a significant proportion will have received post-study treatment[35]. T-DM1 monotherapy vs T-DM1 + capecitabine is being investigated in combination with capecitabine chemotherapy in GOC in pretreated patients (NCT01702558, Table 3) and recruitment has been completed[24].

Trastuzumab deruxtecan

Trastuzumab deruxtecan (DS-8201) is an antibody-drug conjugate comprising a humanised antibody against HER2 and a topoisomerase I inhibitor “payload” bound together by an enzyme-cleavable linker[36]. A phase Iopen label dose escalation study recently presented at ASCO[37] demonstrated an overall response rate (ORR) of 46.7% in HER2+ breast cancer patients pretreated with T-DM1 and pertuzumab and an ORR of 44.4% in gastric cancer patients pretreated with trastuzumab[37]. This high response rate demonstrates that the “payload” bound to the anti-HER2 antibody can make a significant difference to treatment success. For the first time, similar response rates were seen in both breast and gastric cancers pretreated with HER2 inhibitors, and responses were seen even in low HER2-expressing tumours[36]. Results of the currently planned phase 2 trials are eagerly awaited (NCT02564900, Table 3)[24], and whether these response rates can translate into improved overall survival remains to be seen.

Pertuzumab

Pertuzumab is a humanised monoclonal antibody targeting a different HER2 epitope to trastuzumab[38], preventing formation of HER2-HER3 heterodimers[39]. It can be administered concurrently with trastuzumab[40]. In the CLEOPATRA breast cancer study, pertuzumab demonstrated significant clinical benefit when added to trastuzumab plus taxane chemotherapy[40]. Disappointingly, in advanced GOC, the phase III JACOB study of pertuzumab + trastuzumab failed to demonstrate a significant improvement in OS[41].

Pertuzumab is currently being explored in combination with trastuzumab and chemotherapy in the perioperative GOC setting in INNOVATION (NCT02205047) and PETRARCA trials (NCT02581462) and with the addition of radiotherapy in the TRAP trial (NCT02120911) (Table 2)[24].

Preclinical studies investigating pertuzumab in combination with T-DM1 in GOC cell lines and xenograft models found this combination caused growth inhibition but no tumour shrinkage[42]. A literature search did not reveal any clinical studies investigating this combination in GOC.

MM-111

MM-111 is a bispecific antibody fusion protein designed by Merrimack to inhibit HER3-ligand binding and signalling in HER2-amplified tumours by preventing formation of HER2-HER3 heterodimers[43,44]. Preclinical studies showed promise, leading to phase 1 and phase 2 studies in selected tumour types, including HER2-amplified breast and GOC[43,44]. However, the phase 2 study investigating MM-111 in HER2-amplified GOC patients was terminated early by the independent data monitoring committee when it was found that the addition of MM-111 to chemotherapy + trastuzumab resulted in a significantly poorer PFS and OS[43]. In light of the disappointing results seen in GOC[43], all further studies investigating MM-111 were withdrawn, and Merrimack announced that it does not plan to invest further in MM-111.

New HER2 inhibitors

Poziotinib is an oral pan-HER2 inhibitor whose role in combination with trastuzumab and paclitaxel is currently under investigation in advanced gastric cancer (NCT01746771, Table 3)[24]. Phase 1 studies in GOC are investigating MGAH22 (Margetuximab) (NCT01148849, Table 3)[24], a chimeric anti-HER2 monoclonal antibody similar to trastuzumab but engineered for increased binding[45]. Medimmune is investigating their HER2 inhibitor, MEDI4276, in a Phase 1 trial (NCT02576548, Table 3) in both breast and gastric cancers[24]. Pyrotinib is an oral tyrosine kinase inhibitor targeting both HER1 (EGFR) and HER2 and is currently being explored in phase 1 trials in GOC (NCT02378389, NCT02500199, Table 3)[24].

MECHANISMS THAT MAY AFFECT HER2 INHIBITION IN GOC

Resistance to HER2 therapy can be one of two types: primary (intrinsic) resistance occurs when there is no response to HER2 inhibitors and secondary (acquired) resistance occurs when there is an initial response followed by cessation of response[46]. Differentiating between these types of resistance is important, as it dictates the optimal timing of treatment strategies.

Alterations to the HER2 receptor

p95HER2: An aminotruncated form of HER2, known as p95HER2[46], lacks the region to which trastuzumab binds and is expressed in 20%-37% of breast cancer patients[47] and 60%-77% of GOC patients with HER2 amplified disease[48,49]. This may partly explain the poorer response to trastuzumab in GOC as compared to breast cancer.

HER2 mutation: Within the TCGA, 15 cases of ERBB2 mutation in GOC were detected using RNA evidence out of 215 non-hypermutated tumours[6]. Evaluation of HER2 mutation across an array of tumour types revealed HER2 mutations in around 5% of gastric cancer patients[50]. Neratinib, a pan-HER tyrosine kinase inhibitor, is being explored in HER2-mutated cancer (NCT01953926, Table 3)[24].

Loss of HER2 expression

A recent study presented at ASCO found that 35% of GOC treated with trastuzumab lost HER2 positivity[51]. Similarly, in breast cancer, loss of HER2 positivity has been reported in patients treated with neoadjuvant trastuzumab + chemotherapy or chemotherapy alone, and loss of HER2 positivity was associated with an increased risk of disease relapse[52].

Signalling pathways

PIK3CA/PTEN/PI3K/AKT/mTOR pathway: The antitumour activity of HER2 inhibitors requires downstream inhibition of PI3K/AKT[46,53]. BOLERO-3 was a randomised, double-blind, placebo-controlled phase 3 trial in HER2 positive, trastuzumab-resistant, advanced previously-treated breast cancer patients that explored whether the mTOR inhibitor everolimus might restore sensitivity to trastuzumab[54]. It demonstrated significant improvement in PFS with the addition of everolimus [7 mo (95%CI: 6.74-8.18) in the everolimus group vs 5.78 mo (5.49-6.9) in the placebo group][54]. The randomised phase 3 BOLERO-1 trial compared everolimus plus trastuzumab plus paclitaxel to placebo plus trastuzumab plus paclitaxel in order to assess whether addition of everolimus at treatment outset might prevent intrinsic resistance: primary endpoint (PFS) was not met[55].

Phase 3 clinical trials have not been conducted specifically in HER2 positive GOC patients[49]. The phase 3 GRANITE trial randomised 656 patients with advanced pretreated gastric cancer to either everolimus or matching placebo[56]. HER2 status was not an inclusion or exclusion criteria, and we do not know the percentage of HER2 positive patients within this trial. The primary endpoint (OS) was not met, and everolimus was associated with significant side-effects: 21.5% of patients receiving everolimus required drug discontinuation and 55.4% required dose adjustments/interruptions[56]. Such high rates of adverse events are concerning in the palliative setting, where quality of life is important.

IGF-1R expression

Insulin-like growth factor-1 receptor (IGF-1R) is involved in acquired resistance to HER2 blockade in breast cancer[46,57] and GOC[58] cells in vitro by forming heterodimers with HER2. Blockade of this heterodimer formation in vitro and in vivo restored sensitivity to HER2[57], and combination studies of HER2 blockade in combination with IGF-1R inhibitors were more effective than either agent alone[59]. Clinical studies exploring IGF-1R inhibitors in combination with HER2 inhibitors in breast cancer patients found no significant difference in PFS (NCT00684983)[49]; other studies evaluating this strategy were withdrawn, and there are no GOC studies[49].

MET overexpression

Clinical studies of MET inhibitors as monotherapy in HER2 negative breast cancer patients did not meet their primary endpoint[49,60]. In GOC, a randomized double-blind phase 3 clinical trial exploring MET inhibition in HER2 negative, MET positive GOC patients found no benefit from the addition of the MET inhibitor onartuzumab to chemotherapy[61]. Phase 2 results for an alternative MET inhibitor, tivatinib, similarly showed no survival advantage[62]. In light of these disappointing results, it is unlikely MET inhibition will be explored in the clinical setting in HER2-overexpressing breast or GOC patients.

HSP90

Combining HER2- and Heat shock protein (HSP90)-inhibition to overcome resistance to HER2 inhibitors showed promise preclinically in cell lines and mouse models in breast and GOC cell lines[63]. However, a phase 2 study in breast cancer has not yet released results[64], and a phase 2 study in gastric cancer was terminated (NCT01402401)[24].

MicroRNA

MicroRNAs (miRs) are small non-coding RNAs that control gene expression through messenger RNA degradation and post-transcriptional inhibition[65]. MiRs are tissue-specific, and different microRNA signatures may occur during resistance to HER2 inhibition in breast and GOC. In HER2 positive breast and gastric cancer cells, miR-21 overexpression leads to PTEN downregulation, suppression of trastuzumab-induced apoptosis and increased trastuzumab resistance[66,67]. MiRNA-542-3p downregulation promotes trastuzumab resistance in breast cancer via AKT activation[68]. MiR-7 functions as a suppressor of the oncogenic isoform of HER2, HER2Δ16, and reverses HER2Δ16-induced trastuzumab resistance in breast cancer[69]. The use of miRs not only as biomarkers but as targets for anticancer therapy may allow new therapeutic miR silencing in the future[70]. Inhibition of certain microRNAs may also enhance the effect of HER2 inhibition[71].

Immune response

Natural killer (NK) cells are required in order to exert trastuzumab’s therapeutic effect[46]. Mice deficient in NK cells show trastuzumab resistance[72] and when numbers of innate and adaptive immune cells in the tumour microenvironment increase, there is increased tumour eradication[73]. Trials studying the immune environment in GOC are underway (NCT02318901, NCT01526473, NCT02276300, Table 3)[24].

Biomarkers

Specific-uptake positron emission tomography (PET) scans: Targeted PET scans using radiolabelled trastuzumab (89Zr-Trastuzumab) to demonstrate HER2 uptake can give real-time information on HER2 expression levels, visually displaying the development of resistance with the advantage of being relatively non-invasive and, therefore, preferable for patients[74].

Circulating DNA: Circulating DNA may represent a clinically useful biomarker that reduces the need for invasive biopsies. Plasma DNA digital PCR can detect HER2 status in metastatic breast cancer patients[75]. A systematic review and meta-analysis has suggested that serum HER2 is a potential surrogate for tissue HER2 status in gastric cancer[76].

CONCLUSION

Despite numerous HER2 inhibitors being investigated in a number of settings, trastuzumab in advanced disease is still the only HER2 inhibitor licensed for clinical use in the treatment of GOC. Even within this setting, the overall survival benefit is far less than that seen in breast cancer. Other HER2 inhibitors that have demonstrated success in breast cancer have failed to reach statistically significant endpoints in GOC clinical trials, and it remains to be seen whether clinical trials currently underway will show improved results. HER2 heterogeneity, amino-truncation loss of HER2 expression and differences in signalling pathways may contribute to the disappointing clinical trial outcomes seen in GOC. Different microRNA signatures and immune environments are also likely to play a role. Development of new HER2 inhibition strategies in conjunction with further research into how the role of HER2 differs in GOC as compared to breast cancer is required. Clinical trials utilizing biomarkers such as specific uptake PET scans and circulating DNA may provide early insight into whether patients are responding to HER2 inhibition. Only with improved understanding of HER2 inhibition in GOC can effective treatment be provided in order to improve clinical outcomes for patients.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Oncology

Country of origin: United Kingdom

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Li W, Sugimura H S- Editor: Ji FF L- Editor: Filipodia E- Editor: Tan WW

References
1.  Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev. 2015;34:157-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 268]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
2.  Janjigian YY, Werner D, Pauligk C, Steinmetz K, Kelsen DP, Jäger E, Altmannsberger HM, Robinson E, Tafe LJ, Tang LH. Prognosis of metastatic gastric and gastroesophageal junction cancer by HER2 status: a European and USA International collaborative analysis. Ann Oncol. 2012;23:2656-2662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 237]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
3.  Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 2007;26:6469-6487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 628]  [Cited by in F6Publishing: 722]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
4.  Zhu X, Verma S. Targeted therapy in her2-positive metastatic breast cancer: a review of the literature. Curr Oncol. 2015;22:S19-S28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
5.  Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4615]  [Cited by in F6Publishing: 4831]  [Article Influence: 345.1]  [Reference Citation Analysis (1)]
6.  Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4230]  [Cited by in F6Publishing: 4294]  [Article Influence: 429.4]  [Reference Citation Analysis (2)]
7.  Hecht JR, Bang YJ, Qin SK, Chung HC, Xu JM, Park JO, Jeziorski K, Shparyk Y, Hoff PM, Sobrero A. Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC--A Randomized Phase III Trial. J Clin Oncol. 2016;34:443-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 397]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
8.  Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, Tsuji A, Omuro Y, Li J, Wang JW. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014;32:2039-2049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 437]  [Cited by in F6Publishing: 450]  [Article Influence: 45.0]  [Reference Citation Analysis (0)]
9.  Bang YJ, Satoh T, Yeh K-H, Katsura K, Yoshida P, Mukaiyama A, Kobayashi M, Ohtsu A. A randomized, open-label, phase III study of lapatinib in combination with weekly paclitaxel vs weekly paclitaxel alone in the second-line treatment of HER2 amplified advanced gastric cancer (AGC) in Asian population: Tytan study. J Clin Oncol. 2013;31:11-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
10.  Cunningham D, Starling N, Rao S, Iveson T, Nicolson M, Coxon F, Middleton G, Daniel F, Oates J, Norman AR; Upper Gastrointestinal Clinical Studies Group of the National Cancer Research Institute of the United Kingdom. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1579]  [Cited by in F6Publishing: 1611]  [Article Influence: 100.7]  [Reference Citation Analysis (0)]
11.  Wilke H, Muro K, Van Cutsem E, Oh SC, Bodoky G, Shimada Y, Hironaka S, Sugimoto N, Lipatov O, Kim TY. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224-1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1613]  [Cited by in F6Publishing: 1583]  [Article Influence: 158.3]  [Reference Citation Analysis (0)]
12.  Bang YJ. European Society for Medical Oncology Congress; 2016 Oct 7-11; Copenhagen, Denmark; Abstract LBA25. .  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol. 2007;25:118-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2665]  [Cited by in F6Publishing: 2539]  [Article Influence: 149.4]  [Reference Citation Analysis (0)]
14.  Hofmann M, Stoss O, Shi D, Büttner R, van de Vijver M, Kim W, Ochiai A, Rüschoff J, Henkel T. Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology. 2008;52:797-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 777]  [Cited by in F6Publishing: 868]  [Article Influence: 54.3]  [Reference Citation Analysis (0)]
15.  Gomez-Martin C, Plaza JC, Pazo-Cid R, Salud A, Pons F, Fonseca P, Leon A, Alsina M, Visa L, Rivera F. Level of HER2 gene amplification predicts response and overall survival in HER2-positive advanced gastric cancer treated with trastuzumab. J Clin Oncol. 2013;31:4445-4452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 149]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
16.  Tajiri R, Ooi A, Fujimura T, Dobashi Y, Oyama T, Nakamura R, Ikeda H. Intratumoral heterogeneous amplification of ERBB2 and subclonal genetic diversity in gastric cancers revealed by multiple ligation-dependent probe amplification and fluorescence in situ hybridization. Hum Pathol. 2014;45:725-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
17.  Seol H, Lee HJ, Choi Y, Lee HE, Kim YJ, Kim JH, Kang E, Kim SW, Park SY. Intratumoral heterogeneity of HER2 gene amplification in breast cancer: its clinicopathological significance. Mod Pathol. 2012;25:938-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 229]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
18.  Cho EY, Park K, Do I, Cho J, Kim J, Lee J, Kim S, Kim KM, Sohn TS, Kang WK. Heterogeneity of ERBB2 in gastric carcinomas: a study of tissue microarray and matched primary and metastatic carcinomas. Mod Pathol. 2013;26:677-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 70]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
19.  Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8204]  [Cited by in F6Publishing: 7828]  [Article Influence: 340.3]  [Reference Citation Analysis (0)]
20.  Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Láng I, Nitz U, Iwata H, Thomssen C, Lohrisch C, Suter TM, Rüschoff J, Suto T, Greatorex V, Ward C, Straehle C, McFadden E, Dolci MS, Gelber RD; Herceptin Adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-1672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3742]  [Cited by in F6Publishing: 3559]  [Article Influence: 187.3]  [Reference Citation Analysis (0)]
21.  Food and Drug Administration. Office of Medical Products and Tobacco.  Available from: http://www.fda.gov/AboutFDA/CentersOffices/OfficeofMedicalProductsandTobacco.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Shah MA, Xu RH, Bang YJ, Hoff PM, Liu T, Herráez-Baranda LA, Xia F, Garg A, Shing M, Tabernero J. HELOISE: Phase IIIb Randomized Multicenter Study Comparing Standard-of-Care and Higher-Dose Trastuzumab Regimens Combined With Chemotherapy as First-Line Therapy in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Gastric or Gastroesophageal Junction Adenocarcinoma. J Clin Oncol. 2017;35:2558-2567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 80]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
23.  Kataoka K, Tokunaga M, Mizusawa J, Machida N, Katayama H, Shitara K, Tomita T, Nakamura K, Boku N, Sano T. A randomized Phase II trial of systemic chemotherapy with and without trastuzumab followed by surgery in HER2-positive advanced gastric or esophagogastric junction adenocarcinoma with extensive lymph node metastasis: Japan Clinical Oncology Group study JCOG1301 (Trigger Study). Jpn J Clin Oncol. 2015;45:1082-1086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
24.  Chinese PLA General Hospital. T-XELOX in HER2-positive Stage III Gastric Cancer After D2 Gastrectomy.  Available from: https://clinicaltrials.gov/ct2/show/NCT02250209.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Abali H, Yalcin S, Onal HC, Dane F, Oksuzoglu B, Ozdemir N, Mertsoylu H, Artac M, Camci C, Karabulut B. A study of the combination of oxaliplatin, capecitabine, and herceptin (trastuzumab) and chemoradiotherapy in the adjuvant setting in operated patients with HER2+ gastric or gastro-esophageal junction cancer (TOXAG study). J Clin Oncol. 2016;34:182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
26.  Rivera F, Jimenez-Fonseca P, Alfonso PG, Gallego J, Limon ML, Alsina M, Lopez-Gomez L, Galan M, Falco E, Manzano JL. NEOHX study: Perioperative treatment with trastuzumab in combination with capecitabine and oxaliplatin (XELOX-T) in patients with HER-2 resectable stomach or esophagogastric junction (EGJ) adenocarcinoma-18 m DFS analysis. J Clin Oncol. 2015;33:107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
27.  Davidson M, Okines AF, Starling N. Current and Future Therapies for Advanced Gastric Cancer. Clin Colorectal Cancer. 2015;14:239-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
28.  Hecht JR, Bang JY, Qin SK, Chung HC, Xu JM, Park JO, Jeziorski K, Shparyk Y, Hoff PM, Sobrero AF. Lapatinib in combination with capecitabine plus oxaliplatin (CapeOx) in HER2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma (AC): The TRIO-013/LOGiC Trial. J Clin Oncol. 2013;31:4001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
29.  Guan Z, Xu B, DeSilvio ML, Shen Z, Arpornwirat W, Tong Z, Lorvidhaya V, Jiang Z, Yang J, Makhson A. Randomized trial of lapatinib versus placebo added to paclitaxel in the treatment of human epidermal growth factor receptor 2-overexpressing metastatic breast cancer. J Clin Oncol. 2013;31:1947-1953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
30.  Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733-2743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2515]  [Cited by in F6Publishing: 2380]  [Article Influence: 132.2]  [Reference Citation Analysis (0)]
31.  Smyth E, Rowley S, Allum W, Stenning S, Wotherspoon A, Robb C. A randomised phase II study of perioperative epirubicin, cisplatin and capecitabine (ECX) ± lapatinib for operable, HER-2 positive gastric, oesophagogastric junctional (OGJ) or lower oesophageal adenocarcinoma: Results from the UK MRC ST03 lapatinib feasibility study (ISRCTN 46020948). ESMO Cancer Congress 2016; 2016 Oct 7- 11; Copenhagen, Denmark; Abstract 870. .  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
32.  Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, Blättler WA, Lambert JM, Chari RV, Lutz RJ. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008;68:9280-9290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1119]  [Cited by in F6Publishing: 1138]  [Article Influence: 71.1]  [Reference Citation Analysis (0)]
33.  Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Diéras V, Guardino E. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783-1791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2411]  [Cited by in F6Publishing: 2448]  [Article Influence: 204.0]  [Reference Citation Analysis (0)]
34.  Landman A. 2016 ASCO Gastrointestinal Cancers Symposium. Lancet Oncol. 2016;17:282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
35.  Kang YK, Shah M, Ohtsu A, Van Cutsem E, Ajani J, van der Horst T. A randomized, open-label, multicenter, adaptive phase 2/3 study of trastuzumab emtansine (T-DM1) vs a taxane (TAX) in patients (pts) with previously treated HER2-positive locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma (LA/MGC/GEJC). J Clin Oncol. 2016;34:5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
36.  Doi T, Shitara K, Naito Y, Shimomura A, Fujiwara Y, Yonemori K, Shimizu C, Shimoi T, Kuboki Y, Matsubara N. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study. Lancet Oncol. 2017;18:1512-1522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 279]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
37.  Doi T, Iwata J, Tsurutani J, Takahashi S, Park H, Redfern CH, Shitara K, Shimizu C, Taniguchi H, Iwasa T. Single agent activity of DS-8201a, a HER2-targeting antibody-drug conjugate, in heavily pretreated HER2 expressing solid tumors. J Clin Oncol. 2017;35:108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
38.  Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski MX. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell. 2004;5:317-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 817]  [Cited by in F6Publishing: 797]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
39.  Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9:463-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 853]  [Cited by in F6Publishing: 857]  [Article Influence: 57.1]  [Reference Citation Analysis (0)]
40.  Baselga J, Cortés J, Kim SB, Im SA, Hegg R, Im YH, Roman L, Pedrini JL, Pienkowski T, Knott A. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1741]  [Cited by in F6Publishing: 1733]  [Article Influence: 144.4]  [Reference Citation Analysis (0)]
41.  Tabernero J, Hoff PM, Shen L, Ohtsu A, Shah MA, Cheng K, Song C, Wu H, Eng-Wong J, Kang Y. Pertuzumab (P) + trastuzumab (H) + chemotherapy (CT) for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (mGC/GEJC): Final analysis of a Phase III study (JACOB). Ann Oncol. 2017;28: v209-v268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
42.  Yamashita-Kashima Y, Shu S, Harada N, Fujimoto-Ouchi K. Enhanced antitumor activity of trastuzumab emtansine (T-DM1) in combination with pertuzumab in a HER2-positive gastric cancer model. Oncol Rep. 2013;30:1087-1093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
43.  Denlinger C, Maqueda M, Watkins D, Sym S, Bendell J, Park S. Randomized phase 2 study of paclitaxel (PTX), trastuzumab (T) with or without MM-111 in HER2 expressing gastroesophageal cancers (GEC). J Clin Oncol. 2016;34:4043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
44.  Denlinger C, Beeram M, Tolcher A, Goldstein L, Slichenmyer W, Murray J. A phase I/II and pharmacologic study of MM-111 in patients with advanced, refractory HER2-positive (HER2+) cancers. J Clin Oncol. 2010;28:169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
45.  Nordstrom JL, Gorlatov S, Zhang W, Yang Y, Huang L, Burke S, Li H, Ciccarone V, Zhang T, Stavenhagen J. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res. 2011;13:R123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 169]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
46.  Rexer BN, Arteaga CL. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications. Crit Rev Oncog. 2012;17:1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 242]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
47.  Molina MA, Sáez R, Ramsey EE, Garcia-Barchino MJ, Rojo F, Evans AJ, Albanell J, Keenan EJ, Lluch A, García-Conde J. NH(2)-terminal truncated HER-2 protein but not full-length receptor is associated with nodal metastasis in human breast cancer. Clin Cancer Res. 2002;8:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Lee J, Kim S, Kim P, Liu X, Lee T, Kim KM, Do IG, Park JO, Park SH, Jang J. A novel proteomics-based clinical diagnostics technology identifies heterogeneity in activated signaling pathways in gastric cancers. PLoS One. 2013;8:e54644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
49.  National Cancer Institute (NCI). Capecitabine and Lapatinib Ditosylate With or Without Cixutumumab in Treating Patients With Previously Treated HER2-Positive Stage IIIB-IV Breast Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT00684983.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Chmielecki J, Ross JS, Wang K, Frampton GM, Palmer GA, Ali SM, Palma N, Morosini D, Miller VA, Yelensky R. Oncogenic alterations in ERBB2/HER2 represent potential therapeutic targets across tumors from diverse anatomic sites of origin. Oncologist. 2015;20:7-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
51.  Janjigian Y, Riches J, Ku G, Imtiaz T, Capanu M, Chou J. Loss of human epidermal growth factor receptor 2 (HER2) expression in HER2-overexpressing esophagogastric (EG) tumors treated with trastuzumab. J Clin Oncol. 2015;33:63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
52.  Guarneri V, Dieci MV, Barbieri E, Piacentini F, Omarini C, Ficarra G, Bettelli S, Conte PF. Loss of HER2 positivity and prognosis after neoadjuvant therapy in HER2-positive breast cancer patients. Ann Oncol. 2013;24:2990-2994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
53.  Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K, Linn SC, Gonzalez-Angulo AM, Stemke-Hale K, Hauptmann M. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12:395-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1213]  [Cited by in F6Publishing: 1207]  [Article Influence: 71.0]  [Reference Citation Analysis (0)]
54.  André F, O’Regan R, Ozguroglu M, Toi M, Xu B, Jerusalem G, Masuda N, Wilks S, Arena F, Isaacs C. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014;15:580-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 358]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
55.  Hurvitz SA, Andre F, Jiang Z, Shao Z, Mano MS, Neciosup SP, Tseng LM, Zhang Q, Shen K, Liu D. Combination of everolimus with trastuzumab plus paclitaxel as first-line treatment for patients with HER2-positive advanced breast cancer (BOLERO-1): a phase 3, randomised, double-blind, multicentre trial. Lancet Oncol. 2015;16:816-829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 221]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
56.  Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, Sahmoud T, Shen L, Yeh KH, Chin K. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol. 2013;31:3935-3943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 357]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
57.  Nahta R, Yuan LX, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res. 2005;65:11118-11128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 539]  [Cited by in F6Publishing: 542]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
58.  Zhang Z, Wang J, Ji D, Wang C, Liu R, Wu Z, Liu L, Zhu D, Chang J, Geng R. Functional genetic approach identifies MET, HER3, IGF1R, INSR pathways as determinants of lapatinib unresponsiveness in HER2-positive gastric cancer. Clin Cancer Res. 2014;20:4559-4573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 55]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
59.  Chakraborty AK, Zerillo C, DiGiovanna MP. In vitro and in vivo studies of the combination of IGF1R inhibitor figitumumab (CP-751,871) with HER2 inhibitors trastuzumab and neratinib. Breast Cancer Res Treat. 2015;152:533-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
60.  Tolaney SM, Tan S, Guo H, Barry W, Van Allen E, Wagle N, Brock J, Larrabee K, Paweletz C, Ivanova E. Phase II study of tivantinib (ARQ 197) in patients with metastatic triple-negative breast cancer. Invest New Drugs. 2015;33:1108-1114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
61.  Shah M, Bang Y-J, Lordick F, Tabernero J, Chen M, Hack S. METGastric: A phase III study of onartuzumab plus mFOLFOX6 in patients with metastatic HER2-negative (HER2-) and MET-positive (MET+) adenocarcinoma of the stomach or gastroesophageal junction (GEC). J Clin Oncol. 2015;33:4012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
62.  Pant S, Patel M, Kurkjian C, Hemphill M, Flores M, Thompson D. A phase II study of the c-Met inhibitor tivantinib (tiv) in combination with FOLFOX for the treatment of patients (pts) with previously untreated metastatic adenocarcinoma of the distal esophagus, gastroesophageal (GE) junction, or stomach. J Clin Oncol. 2015;33:4065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
63.  Wainberg ZA, Anghel A, Rogers AM, Desai AJ, Kalous O, Conklin D, Ayala R, O’Brien NA, Quadt C, Akimov M. Inhibition of HSP90 with AUY922 induces synergy in HER2-amplified trastuzumab-resistant breast and gastric cancer. Mol Cancer Ther. 2013;12:509-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
64.  Kong A, Rea D, Ahmed S, Beck J, López López R, Biganzoli L. Phase IB/II study of the HSP90 inhibitor AUY922, in combination with trastuzumab, in patients with HER2+ advanced breast cancer. J Clin Oncol. 2012;30:530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
65.  Lovat F, Valeri N, Croce CM. MicroRNAs in the pathogenesis of cancer. Semin Oncol. 2011;38:724-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 161]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
66.  Eto K, Iwatsuki M, Watanabe M, Ida S, Ishimoto T, Iwagami S, Baba Y, Sakamoto Y, Miyamoto Y, Yoshida N. The microRNA-21/PTEN pathway regulates the sensitivity of HER2-positive gastric cancer cells to trastuzumab. Ann Surg Oncol. 2014;21:343-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
67.  De Mattos-Arruda L, Bottai G, Nuciforo PG, Di Tommaso L, Giovannetti E, Peg V, Losurdo A, Pérez-Garcia J, Masci G, Corsi F. MicroRNA-21 links epithelial-to-mesenchymal transition and inflammatory signals to confer resistance to neoadjuvant trastuzumab and chemotherapy in HER2-positive breast cancer patients. Oncotarget. 2015;6:37269-37280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 114]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
68.  Ma T, Yang L, Zhang J. MiRNA5423p downregulation promotes trastuzumab resistance in breast cancer cells via AKT activation. Oncol Rep. 2015;33:1215-1220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
69.  Huynh FC, Jones FE. MicroRNA-7 inhibits multiple oncogenic pathways to suppress HER2Δ16 mediated breast tumorigenesis and reverse trastuzumab resistance. PLoS One. 2014;9:e114419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
70.  Hayes J, Peruzzi PP, Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol Med. 2014;20:460-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1310]  [Cited by in F6Publishing: 1481]  [Article Influence: 148.1]  [Reference Citation Analysis (0)]
71.  Lote H, Zito D, Burke R, Smyth E, Braconi C, Cunningham D, Valeri N. MicroRNAs as biomarkers of resistance to HER2 inhibitors in combination with chemotherapy in gastro-oesophageal cancer cell lines. Ann Oncol. 2016;27:545-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
72.  Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med. 2000;6:443-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2060]  [Cited by in F6Publishing: 2021]  [Article Influence: 84.2]  [Reference Citation Analysis (0)]
73.  Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O, Yang X, Sattar H, Wang Y, Brown NK, Greene M. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18:160-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 419]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
74.  Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol. 2016;13:348-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 106]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
75.  Gevensleben H, Garcia-Murillas I, Graeser MK, Schiavon G, Osin P, Parton M, Smith IE, Ashworth A, Turner NC. Noninvasive detection of HER2 amplification with plasma DNA digital PCR. Clin Cancer Res. 2013;19:3276-3284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 137]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
76.  Zhang K, Cui J, Xi H, Bian S, Ma L, Shen W, Li J, Wang N, Wei B, Chen L. Serum HER2 Is a Potential Surrogate for Tissue HER2 Status in Gastric Cancer: A Systematic Review and Meta-Analysis. PLoS One. 2015;10:e0136322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
77.  European Organisation for Research and Treatment of Cancer - EORTC. Neoadjuvant Study Using Trastuzumab or Trastuzumab With Pertuzumab in Gastric or Gastroesophageal Junction Adenocarcinoma (INNOVATION). Available from: https://clinicaltrials.gov/ct2/show/NCT02205047. .  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Gelmon K, Vidal M, Sablin M-P, Serpanchy R, Soberino J, Cortes J. Trastuzumab emtansine (T-DM1) plus capecitabine (X) in patients with HER2-positive MBC: MO28230 TRAX-HER2 phase 1 results. J Clin Oncol. 2014;32:606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]