Dual-targeting strategy using trastuzumab and lapatinib in a patient with HER2 gene amplification in recurrent metachronous metastatic gallbladder carcinoma.
Journal: 2019/May - Journal of International Medical Research
ISSN: 1473-2300
Abstract:
Gallbladder carcinoma (GBC) is a rare and highly aggressive tumor. Early diagnosis is challenging, which results in a poor prognosis using systemic therapy. Recent studies have identified a subset of GBC patients with HER2 gene (ERBB2) amplification that could benefit from HER2-targeted therapy. Here, we report one patient with recurrent metachronous GBC with metastasis, who received the combination of trastuzumab and lapatinib. This approach achieved a partial response for both the brain and the lung metastases. This study demonstrated that HER2 inhibition is a promising therapeutic strategy for GBC with HER2 amplification and, combined with lapatinib, it can effectively target brain metastasis.
Relations:
Content
References
(17)
Diseases
(1)
Drugs
(2)
Genes
(2)
Processes
(1)
Anatomy
(3)
Similar articles
Articles by the same authors
Discussion board
The Journal of International Medical Research. May/31/2019; 47(6): 2768-2777
Published online May/19/2019

Dual-targeting strategy using trastuzumab and lapatinib in a patient with HER2 gene amplification in recurrent metachronous metastatic gallbladder carcinoma

+3 authors

Abstract

Introduction

Anti-HER2 molecular targeted agents are established for treating breast cancer,13 gastric cancer,4 and metastatic colorectal cancer5 in patients with HER2 gene amplification (an increase in the copy number). A combination of two HER2-targeted therapies, such as trastuzumab and lapatinib, has also been demonstrated to have clinical efficacy in advanced HER2-positive metastatic breast cancers2,3 and colorectal cancers.5

Lapatinib (GW572016) is a selective inhibitor of epidermal growth factor receptor (EGFR) and HER-2 tyrosine kinase,6 and trastuzumab is a humanized monoclonal antibody that targets the HER-2 extracellular domain.7 The dual targeting of HER2-positive tumors using trastuzumab and lapatinib results from the primary and acquired resistance of these two drugs, their partly non-overlapping mechanisms of action, and the well characterized synergistic interaction between them in HER2 breast-cancer models. Clinically, trastuzumab primarily induces pro-apoptotic effects, while lapatinib inhibits proliferation.8,9 Clinical evidence shows indirect support for dual HER2 blockade. In patients with refractory breast cancer who are treated with trastuzumab, lapatinib plus trastuzumab prolonged the progression-free survival compared with lapatinib alone. Konecny et al.10 analyzed the combination of trastuzumab plus lapatinib, and they observed synergistic drug interactions in four HER-2-overexpressing breast cancer cell lines. Additionally, similar results were obtained in the study reported by Baselga et al.,11 which suggested complementary mechanisms of action and synergistic anti-tumor activity for the monoclonal antibody in a model of breast cancer that overexpressed HER2. Xu et al.3 conducted a systematic review and meta-analysis of randomized controlled trials (RCTs), and the results showed that lapatinib plus trastuzumab significantly prolonged the pathological complete response, overall survival, and event-free survival with tolerance in HER2-positive breast cancer patients. Moreover, the combination of trastuzumab and lapatinib was also effective and well tolerated in patients with HER2-positive metastatic colorectal cancer refractory disease.5

Gallbladder carcinoma (GBC) is a rare and highly aggressive tumor. Early diagnosis is challenging, which results in a poor prognosis in patients who receive systemic therapy. However, combination therapy with trastuzumab and lapatinib has not been reported in patients with GBC. Here, we present the case of a patient with dual-targeted HER2 therapy including lapatinib that was used to treat advanced GBC with brain metastases.

Case presentation

On 4 January 2016, a 48-year-old Chinese female patient was admitted to the Shaoxing People’s Hospital because of nausea, vomiting, dizziness, headache, crooked mouth, and slurred speech. Magnetic resonance imaging (MRI) showed multi-site metastatic brain tumors (Figure 1a) and bilateral adrenal gland metastasis (data not shown). Chest computed tomography (CT) scans revealed multi-site metastatic lung tumors (Figure 1b). This patient was diagnosed with GBC via cholecystectomy that was performed because of cholecystitis and gallstones in 2010 (pT3N0M0; TNM staging: IIB; Nevin staging: III) (Figure 1e). Lung metastasis was found in 2014, and brain metastasis was detected in 2015. The patient’s medical history is shown in Figure 2.

Figure 1.

Dual-targeted therapy with trastuzumab and lapatinib were effective in advanced gallbladder carcinoma patients with multi-site brain and lung metastases.

Magnetic resonance imaging showed the status of brain metastases before (a) and after (c) three cycles of dual-HER2 targeted therapy with trastuzumab and lapatinib. Chest computed tomography scans showed the status of lung metastasis before (b) and after (d) three cycles of dual HER2-targeted therapy with trastuzumab and lapatinib. (e) A biopsy identified that the gallbladder tumor was moderate-differentiated adenocarcinoma (hematoxylin and eosin staining; original magnification ×100). (f) A biopsy showed that the metastatic brain tumors were adenocarcinoma (hematoxylin and eosin staining; original magnification ×100).

Figure 2.
Medical history, clinical interventions, and metastasis development.

One month after admission, the patient underwent surgery to remove one of the largest metastatic tumors (longest diameter in the transverse plane, 38.96 mm) in the left frontal lobe. The biopsy showed that the metastatic tumor was adenocarcinoma (Figure 1f). The patient was diagnosed with the GBC brain metastasis after a comprehensive review. Genetic profiling of 416 cancer-relevant genes was performed using targeted next generation sequencing (NGS; see Materials and Methods) to develop potential molecular therapeutic approaches. GBC primary tumor and brain metastatic tissues, as well as the circulating tumor DNA (ctDNA) in the blood were collected from the patient for genetic profiling. The sequencing results showed a significant increase in the HER2 copy number in the GBC primary tumor (8.6×), brain metastasis (14.6×), and ctDNA (2.5×) (Table 1). Thus, dual-targeted therapy was initiated on 6 March, 2016 using trastuzumab (administered intravenously at an initial dose of 8 mg/kg, followed by subsequent doses of 6 mg/kg once per week for 3 weeks) and lapatinib (administered orally each day at a dose of 1000 mg per day in 21-day treatment cycles), based on previously published work in breast cancer2,3 (i.e., one dose of trastuzumab each week and one dose of oral lapatinib each day). No concurrent chemotherapy was administrated.

Table 1.
Genetic alterations identified in samples before/after HER2-targeted therapy.
Before HER2-targeted therapyAfter three cycles of anti-HER2 targeted therapy
Sample typeSample IDGenetic alterations identifiedMAF (%)/Fold change (×)
ctDNAP16020611144TP53: p.K291X (c.A871T)4%
CDK12: amplification2.87×
ERBB2: amplification2.48×
FFPE sample ofF16020611145TP53: p.K291X (c.A871T)31%
gallbladder tumorCDK12: amplification9.03×
ERBB2: amplification8.40×
FFPE sample ofF16020611146TP53: p.K291X (c.A871T)72%
brain tumorCDK12: amplification11.28×
ERBB2: amplification14.60×
RB1: Single-copy loss0.53×
ctDNAP16051918361TP53: p.K291X (c.A871T)0.5%
CDK12: amplificationNot detected
ERBB2: amplificationNot detected
RB1: Single-copy lossNot detected
MAF, mutant allele frequency; ctDNA, circulating tumor DNA; FFPE, formalin-fixed paraffin-embedded.

Three weeks after dual HER2 targeted therapy with trastuzumab and lapatinib, the follow-up chest and brain MRI revealed that the patient achieved a partial response in brain metastatic tumors (Figure 1c) and lung metastatic tumors (Figure 1d), after three cycles of treatment based on the Response Criteria Evaluation in Solid Tumors (RECIST) version 1.1. The MRI results showed that the metastatic tumors had shrunk. A concurrent blood sample was collected for genetic testing of ctDNA using targeted NGS. The HER2 copy numbers did not increase at this time (Table 1), suggesting that the tumors were cleared after trastuzumab and lapatinib dual therapy. It was also accompanied by a carcinoembryonic antigen (CEA) response (CEA levels decreased from 4030 ng/mL to 660 ng/mL). The patient developed grade 3 adverse reactions including fatigue, skin rash, and increased bilirubin concentration, but no grade 4 or 5 adverse reactions were observed. In summary, dual-targeted therapy with trastuzumab and lapatinib offered a chemotherapy-free option and achieved tumor suppression with an acceptable safety profile for this patient with HER2 amplification in GBC.

Materials and methods

Patient information

The patient provided written informed consent. The study was approved by the ethics committee at the institute and was performed in strict accordance with the principles of the Declaration of Helsinki and the International Conference on Harmonization and Good Clinical Practice guidelines.

Tumor assessment

Tumor measurements were performed by a radiologist (ZXL) before treatment and were assessed every 2 months in accordance with RECIST (version 1.1). The patient was examined using a CT of the chest, abdomen, or pelvis, and MRI brain scans when clinically indicated in accordance with the protocol. All tumor measurements were reviewed by a radiologist (ZXL) who read the MRI or CT scans, and collected, stored, and interpreted the imaging results.

Adverse reaction assessment

The patient’s health was continuously monitored and graded in accordance with the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Laboratory test assessments (hematology, serum chemistry, urine analysis, and tumor markers) were performed at baseline (day 1 of every cycle), and at the end of treatment. The left ventricular ejection fraction was examined at baseline (day 1 of every cycle) and at the end of treatment.

DNA extraction

Formalin-fixed paraffin-embedded (FFPE) tissues were sectioned, and hematoxylin and eosin (H&E)-stained slides were reviewed by surgical pathologists (FL and AJS). Tumor tissue areas were dissected from ten serial sections (4 μm) guided by H&E slides as templates. The FFPE tissues were deparaffinized with xylene and genomic DNA was extracted using a QIAamp DNA FFPE tissue kit (Qiagen, Valencia, CA, USA). The concentration of DNA was determined using a Qubit dsDNA HS assay kit on the Qubit 3.0 (Life Technologies, Carlsbad, CA, USA), in strict accordance with the manufacturer’s protocol. DNA (A260/280 and A260/230) was measured using the Nanodrop-2000 (Thermo Fisher Scientific, Waltham, MA, USA).

Whole blood (8 mL) was collected using EDTA blood collection tubes (BD Biosciences, San Jose, CA, USA) that were centrifuged within 2 hours at 1800 ×g for 10 minutes at ≥4°C to collect the plasma. ctDNA was extracted from plasma using a QIAamp circulating nucleic acid kit (Qiagen). The purified ctDNA was quantified using a Qubit dsDNA HS assay kit on the Qubit 3.0 Fluorometer (Life Technologies).

Library construction

The sequencing library was prepared using a KAPA Hyper Prep Kit (Kapa Biosystems, Boston, MA, USA), in accordance with the manufacturing protocol. Briefly, 1 μg genomic DNA sample was fragmented into 350 bp in an Adaptive Focused Acoustics (AFA) fiber snap-cap microTUBE using Covaris M220 (Covaris, Woburn, MA, USA) or 10–50 ng ctDNA underwent end repairing, A-tailing, adapter ligation, size selection, and finally PCR amplification. Library concentration was determined using a Qubit dsDNA HS assay kit on the Qubit 3.0 Fluorometer (Life Technologies).

Hybrid capture and ultra-deep next generation sequencing

The 5ʹ-biotinylated probe solution was used as capture probes, which targeted 416 cancer-related genes (Table 2). The capture reaction was performed using the NimbleGen SeqCap EZ hybridization and Wash Kit (Roche) with 1 μg of pooled libraries, 5 μg of human Cot-1 DNA, 1 unit of adapter-specific blocker DNA, and the capture probes. The solution hybridization was performed for 16–18 hours at 65°C. The captured targets were selected by pulling down the biotinylated probe/target hybrids using streptavidin-coated magnetic beads, and the off-target library was removed by washing with wash buffer, followed by PCR amplification of the target-enriched libraries. Sequencing libraries were quantified by qPCR using the KAPA Library Quantification kit (KAPA Biosystems, Boston, MA, USA), sized on a Bioanalyzer 2100 (Agilent Technologies, Santa Clara, CA, USA), and then deep-sequenced on an Illumina HiSeq 4000 using the PE150 kit (Illumina Inc., San Diego, CA, USA).

Table 2.
Gene targeted in hybridization capture.
ABCB1 (MDR1)CDK4ERBB4HGFMSH2PTENSTK11
ABCC2 (MRP2)CDK6ERCC1HNF1AMSH3PTPN11STMN1
ACTBCDK8ERCC2HNF1BMSH6PTPN2STX11
ADH1BCDKN1BERCC3HRASMTHFRPTPN6STXBP2
AIPCDKN1CERCC4ID3MTORPTPROSUFU
AKT1CDKN2AERCC5IDH1MUTYHQKISUZ12
AKT2CDKN2BESR1IDH2MYCRAC1SYN3
AKT3CDKN2CETV1IGF1RMYCL1RAD21TCN2
ALDH2CEBPAETV4IGF2MYCNRAD50TEK
ALKCEP57EWSR1IKBKEMYD88RAD51TEKT4
AMER1CHD4EXT1IKZF1MYNNRAD51CTERC
AP3B1CHEK1EXT2IKZF2NBNRAD51DTERT
APCCHEK2EZH2IKZF3NCSTNRAF1TET2
ARCKS1BFANCAIL13NF1RARATGFBR2
ARAFCREBBPFANCBIL7RNF2RASGEF1ATLE1
ARID1ACRKLFANCCINPP4BNFKBIARB1TLE4
ARID2CROTFANCD2INPP5DNKX2-1RECQL4TMEM127
ARID5BCSF1RFANCEIRF1NOTCH1RELNTMPRSS2
ASXL1CSF3RFANCFIRF2NOTCH2RETTNFAIP3
ATMCTCFFANCGIRF4NPM1RHBDF2TNFRSF14
ATRCTLA4FANCIIRF8NQO1RHOATNFRSF17
ATRXCTNNB1FANCLJAK1NRASRICTORTNFRSF19
AURKACUX1FANCMJAK2NRG1RNF146TOP1
AURKBCXCR4FAT1JAK3NSD1RNF43TOP2A
AXIN1CYLDFBXO11JARID2NT5C2ROS1TP53
AXLCYP19A1FCGR2BJUNNTRK1RPTORTP63
B2MCYP2B6*6FGF19KDM2BPAG1RRM1TPMT
BAP1CYP2C19*2FGFR1KDM5APAK3RTEL1TRAF2
BARD1CYP2C9*3FGFR2KDR (VEGFR2)PALB2RUNX1TRAF3
BCL2CYP2D6*3FGFR3KIF1BPARK2SBDSTRAF5
BCL2L1CYP2D6*4FGFR4KITPAX5SDHATSC1
BCL2L2CYP2D6*5FHKMT2BPBRM1SDHAF2TSC2
BCORL1CYP2D6*6FIP1L1KMT2CPCSDHBTSHR
BCL2L11 (BIM)CYP3A4*4FLCNKRASPDCD1SDHCTTF1
BLMCYP3A5*1FLT1 (VEGFR1)LEF1PDCD1LG2 (PD-L2)SDHDTUBB3
BMPR1ACYP3A5*3FLT3LMO1PDGFRASERP2TYMS
BRAFDAB2FLT4 (VEGFR4)LSP1PDGFRBSETBP1TYR
BRCA1DAXXGADD45BLYNPDK1SETD2U2AF1
BRCA2DDR2GATA1LYSTPHF6SF3B1UGT1A1
BRD4DDX1GATA2LZTR1PHOX2BSGK1UNC13D
BRIP1DHFRGATA3MAP2K1 (MEK1)PICK3R1SH2D1AVEGFA
BTG2DICER1GATA4MAP2K2 (MEK2)PIK3C3SLX4VHL
BTKDIS3L2GATA6MAP2K4PIK3CASMAD2WISP3
BTLADLG2GNA11MAP3K1PIK3CDSMAD3WRN
BUB1BDMNT3AGNA13MCL1PIK3R1SMAD4WT1
c11orf30DNM2GNAQMDM2PIK3R2SMAD7XIAP
CALRDOCK1GNASMDM4PLCE1SMARCA4XPA
CBLDOT1LGPC3MECOMPLK1SMARCB1XPC
CCND1DPYDGRIN2AMED12PMS1SMC1AXPO1
CCNE1DUSP2GRM3MEF2BPMS2SMC3XRCC1
CCT6BEBF1GSTM1MEN1POLD1SMOYAP1
CD22ECT2LGSTP1METPOLD3SOX2ZAP70
CD274 (PD-L1)EEDGSTT1MGMTPOLESPOPZBTB20
CD58EGFRHBA1MITFPOT1SRCZNF217
CD70EGR1HBA2MLH1PPP2R1ASRSF2ZNF703
CDAEP300HBBMLLPRDM1STAG2ZRSR2
CDC73EPCAMHDAC1MLLT10PRF1STAT3
CDH1EPHA3HDAC2MLPHPRKAR1ASTAT5A
CDK10ERBB2 (HER2)HDAC4MPLPRKCISTAT5B
CDK12ERBB3HDAC7MRE11APTCH1STIL

Sequence alignment and data processing

Quality control was applied using Trimmomatic.12 High quality reads were mapped to the human genome (hg19, GRCh37 Genome Reference Consortium Human Reference 37) using a modified Burrows-Wheeler Aligner (BWA) version 0.7.1213 with BWA-MEM algorithm and default parameters. The Genome Analysis Toolkit14 (GATK, version 3.4-0) was modified and used to locally realign the BAM files at intervals with insertion/deletion (indel) mismatches and to recalibrate the BAM file read base quality scores. Single nucleotide variants (SNVs) and short indels were identified using VarScan2 2.3.915 with a minimum variant allele frequency threshold set at 0.01 and a p-value threshold for calling variants set at 0.05 to generate variant call format (VCF) files. All SNVs/indels were annotated using ANNOVAR, and each SNV/indel was manually checked using the integrative genomics viewer (IGV).16 Copy number variations (CNVs) were identified using ADTEx 1.0.4.17 The analysis and data are presented as copy number changes in all Tables.

Discussion

Recent studies have identified a subset of patients who are undergoing treatment for advanced GBC with HER2 overexpression or amplification that could potentially benefit from HER2-targeted therapy.6,7 Some studies previously reported the use of HER2-targeted treatment to treat HER2-positive gallbladder cancers. One case included a significant reduction in lung metastasis in patients with HER2-amplified metastatic cholangiocarcinoma that was treated with trastuzumab and paclitaxel.8 Another study retrospectively reviewed GBC patients who received HER2-directed therapy with trastuzumab, lapatinib, or pertuzumab, and showed that HER2 blockage was a promising treatment strategy in HER2-positive GBC patients.7 Most of those patients received a single HER2-targeted reagent combined with chemotherapy.

Clinical trials have demonstrated that lapatinib can be used as a therapeutic option for advanced HER2 alterations in cancers with brain metastasis.9 Considering the multi-site brain and lung metastases of the GBC patients, we administrated dual-targeted HER2 therapy with trastuzumab and lapatinib and achieved PR after 4 months of treatment without concurrent chemotherapy. In summary, this is the first study to administer dual-targeted HER2 therapy including lapatinib to treat patients with advanced GBC with brain metastases. Patient follow-up and further multi-center clinical trials are necessary to investigate the long-term efficacy.

Conclusion

HER2 inhibition is a promising therapeutic strategy for GBC with HER2 amplification and used in combination with lapatinib, it can effectively target brain metastasis.

Declaration of conflicting interest

The authors declare that there is no conflict of interest.

Disclosure

Xue Wu, Yichuan Liu, and Yang W. Shao are the shareholders or employees of Geneseeq Technology Inc.

Funding

This work was supported by National Natural Science Foundation of China (Grants No. 81374014 and 81472210) and Zhejiang Provincial Medical and Healthy Science and Technology Projects (Grant No. 2013KYA228).

References

  • 1. SlamonDJLeyland-JonesBShakSet alUse of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.N Engl J Med2001; 344: 783792.[PubMed][Google Scholar]
  • 2. BlackwellKLBursteinHJStornioloAMet alRandomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer.J Clin Oncol2010; 28: 11241130.[PubMed][Google Scholar]
  • 3. XuZQZhangYLiNet alEfficacy and safety of lapatinib and trastuzumab for HER2-positive breast cancer: a systematic review and meta-analysis of randomised controlled trials.BMJ Open2017; 7: e013053.[Google Scholar]
  • 4. BangYJVan CutsemEFeyereislovaAet alTrastuzumab 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.Lancet2010; 376: 687697.[PubMed][Google Scholar]
  • 5. Sartore-BianchiATrusolinoLMartinoCet alDual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol2016; 17: 738746.[PubMed][Google Scholar]
  • 6. WoodERTruesdaleATMcDonaldOBet alA unique structure for epidermal growth factor receptor bound to GW572016 (lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells.Cancer Res2004; 64: 66526659.[PubMed][Google Scholar]
  • 7. ClaudiaBPaoloPLuciaDM.Trastuzumab emtansine in the treatment of HER-2-positive metastatic breast cancer patients. Future Oncol2013; 9: 955957.[PubMed][Google Scholar]
  • 8. DaveBMigliaccioIGutierrezMCet alLoss of phosphatase and tensin homolog or phosphoinositol-3 kinase activation and response to trastuzumab or lapatinib in human epidermal growth factor receptor 2–overexpressing locally advanced breast cancers.J Clin Oncol2011; 29: 166.[PubMed][Google Scholar]
  • 9. MohsinSKWeissHLGutierrezMCet alNeoadjuvant trastuzumab induces apoptosis in primary breast cancers.J Clin Oncol2005; 23: 24602468.[PubMed][Google Scholar]
  • 10. KonecnyGEPegramMDNatarajanVet alActivity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells.Cancer Res2006; 66: 16301639.[PubMed][Google Scholar]
  • 11. EvandroDAHolmesAPMartinePGet alLapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response.Lancet Oncol2014; 15: 11371146.[PubMed][Google Scholar]
  • 12. BolgerAMLohseMUsadelB.Trimmomatic: a flexible trimmer for Illumina sequence data.Bioinformatics2014; 30: 21142120.[PubMed][Google Scholar]
  • 13. LiHDurbinR.Fast and accurate short read alignment with Burrows-Wheeler transform.Bioinformatics2009; 25: 17541760.[PubMed][Google Scholar]
  • 14. McKennaAHannaMBanksEet alThe genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data.Genome Res2010; 20: 12971303.[PubMed][Google Scholar]
  • 15. KoboldtDCZhangQLarsonDEet alVarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing.Genome Res2012; 22: 568576.[PubMed][Google Scholar]
  • 16. RobinsonJTThorvaldsdottirHWincklerWet alIntegrative genomics viewer.Nat Biotechnol2011; 29: 2426.[PubMed][Google Scholar]
  • 17. AmarasingheKCLiJHunterSMet alInferring copy number and genotype in tumour exome data.BMC Genomics2014; 15: 732.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.