1
|
Human Epidermal Growth Factor Receptor-2 Promotes Invasion and Metastasis in Gastric Cancer by Activating Mitogen-activated Protein Kinase Signaling. Appl Immunohistochem Mol Morphol 2020; 27:529-534. [PMID: 29734245 DOI: 10.1097/pai.0000000000000672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing evidence supports an important role for the human epidermal growth factor receptor-2 (HER2) gene and mitogen-activated protein kinase (MAPK) signaling pathways in the progression of human cancers by enhancing cancer cell metastasis and proliferation. However, the relationship between HER2 and MAPK signaling pathways in gastric cancer (GC) remains unclear. In the present study, dual in situ hybridization was performed to detect HER2 gene amplification and reverse transcription-quantitative polymerase chain reaction was used to investigate the mRNA expression of members of the MAPK signaling pathway, including rapidly accelerated fibrosarcoma (RAF), extracellular regulated signal-activated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK), in 112 primary GC tissue samples. The results revealed that 19/112 (17%) of tissue samples showed positive amplification of HER2, which was correlated with tumor invasion and metastasis. Upregulation of RAF, ERK, p38, and JNK was also observed in samples associated with metastasis. Moreover, the expression levels of RAF and ERK in samples with HER2 gene amplification were significantly increased compared with those without HER2 amplification. However, the expression levels of both p38 and JNK were not significantly correlated with HER2 gene amplification. Our results simultaneously showed the association between HER2 gene amplification and the expression levels of MAPK signaling pathway proteins and clinicopathologic characteristics in GC. These findings provide the basis for investigating the regulation of MAPK signaling pathways by HER2 and potential therapeutic targets for inhibiting metastasis and invasion in GC.
Collapse
|
2
|
Validating a targeted next-generation sequencing assay and profiling somatic variants in Chinese non-small cell lung cancer patients. Sci Rep 2020; 10:2070. [PMID: 32034196 PMCID: PMC7005734 DOI: 10.1038/s41598-020-58819-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/29/2019] [Indexed: 02/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is featured with complex genomic alterations. Molecular profiling of large cohort of NSCLC patients is thus a prerequisite for precision medicine. We first validated the detection performance of a next-generation sequencing (NGS) cancer hotspot panel, OncoAim, on formalin-fixed paraffin-embedded (FFPE) samples. We then utilized OncoAim to delineate the genomic aberrations in Chinese NSCLC patients. Overall detection performance was powerful for mutations with allele frequency (MAF) ≥ 5% at >500 × coverage depth, with >99% sensitivity, high specificity (positive predictive value > 99%), 94% accuracy and 96% repeatability. Profiling 422 NSCLC FFPE samples revealed that patient characteristics, including gender, age, lymphatic spread, histologic grade and histologic subtype were significantly associated with the mutation incidence of EGFR and TP53. Moreover, RTK signaling pathway activation was enriched in adenocarcinoma, while PI(3)K pathway activation, oxidative stress pathway activation, and TP53 pathway inhibition were more prevalent in squamous cell carcinoma. Additionally, novel co-existence (e.g., variants in BRAF and PTEN) and mutual-exclusiveness (e.g., alterations in EGFR and NFE2L2) were found. Finally, we revealed distinct mutation spectrum in TP53, as well as a previously undervalued PTEN aberration. Our findings could aid in improving diagnosis, prognosis and personalized therapeutic decisions of Chinese NSCLC patients.
Collapse
|
3
|
Abstract
Metastasis, the dissemination of cancer cells from primary tumors, represents a major hurdle in the treatment of cancer. The epithelial-mesenchymal transition (EMT) has been studied in normal mammalian development for decades, and it has been proposed as a critical mechanism during cancer progression and metastasis. EMT is tightly regulated by several internal and external cues that orchestrate the shifting from an epithelial-like phenotype into a mesenchymal phenotype, relying on a delicate balance between these two stages to promote metastatic development. EMT is thought to be induced in a subset of metastatic cancer stem cells (MCSCs), bestowing this population with the ability to spread throughout the body and contributing to therapy resistance. The EMT pathway is of increasing interest as a novel therapeutic avenue in the treatment of cancer, and could be targeted to prevent tumor cell dissemination in early stage patients or to eradicate existing metastatic cells in advanced stages. In this review, we describe the sequence of events and defining mechanisms that take place during EMT, and how these interactions drive cancer cell progression into metastasis. We summarize clinical interventions focused on targeting various aspects of EMT and their contribution to preventing cancer dissemination.
Collapse
Affiliation(s)
- Mohini Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Nicolas Yelle
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Chitra Venugopal
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Sheila K Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
4
|
Qi L, Zhou L, Lu M, Yuan K, Li Z, Wu G, Huang X, Shen Y, Zhao M, Fu W, Chu B, Wang G, Ren F, Ma D, Chen J. Development of a highly specific HER2 monoclonal antibody for immunohistochemistry using protein microarray chips. Biochem Biophys Res Commun 2017; 484:248-254. [PMID: 28111342 DOI: 10.1016/j.bbrc.2017.01.086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
HER2 is an orphan receptor tyrosine kinase of the EGFR families and is considered to be a key tumor driver gene [1]. Breast cancer and gastric cancer with HER2 amplification can be effectively treated by its neutralizing antibody, Herceptin. In clinic, Immunohistochemistry (IHC) was used as the primary screening method to diagnose HER2 amplification [2]. However, recent evidence suggested that the frequently used rabbit HER2 antibody 4B5 cross reacted with another family member HER4 [3]. IHC staining with 4B5 also indicated that there was strong non-specific cytoplasmic and nuclear signals in normal gastric mucosal cells and some gastric cancer samples. Using a protein lysate array which covers 85% of the human proteome, we have confirmed that the 4B5 bound to HER4 and a nuclear protein ZSCAN18 besides HER2. The non-specific binding accounts for the unexpected cytoplasmic and nuclear staining of 4B5 of normal gastric epithelium. Finally, we have developed a novel mouse HER2 monoclonal antibody UMAB36 with similar sensitivity to 4B5 but only reacted to HER2 across the 17,000 proteins on the protein chip. In 129 breast cancer and 158 gastric cancer samples, UMAB36 showed 100% sensitivity and specificity comparing to the HER2 FISH reference results with no unspecific staining in the gastric mucosa layer. Therefore, UMAB36 could provide as an alternative highly specific IHC reagent for testing HER2 amplification in gastric cancer populations.
Collapse
Affiliation(s)
- Lili Qi
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Lixin Zhou
- Department of Pathology, Beijing Cancer Hospital, No. 52 Fu-Cheng Road, Haidian District, Beijing, 100142, PR China
| | - Mingmin Lu
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, PR China
| | - Kehu Yuan
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Zhongwu Li
- Department of Pathology, Beijing Cancer Hospital, No. 52 Fu-Cheng Road, Haidian District, Beijing, 100142, PR China
| | - Guiyin Wu
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Xiaozheng Huang
- Department of Pathology, Beijing Cancer Hospital, No. 52 Fu-Cheng Road, Haidian District, Beijing, 100142, PR China
| | - Yi Shen
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Min Zhao
- Department of Pathology, Beijing Cancer Hospital, No. 52 Fu-Cheng Road, Haidian District, Beijing, 100142, PR China
| | - Wei Fu
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Boyang Chu
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Guangli Wang
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA
| | - Fangfang Ren
- Department of Biochemistry and Molecular Biology, Medical College of Soochow University, Suzhou, 215123, PR China.
| | - Donghui Ma
- OriGene Technologies, 9620 Medical Center Dr., Suite 200, Rockville, MD, 20850, USA.
| | - Jian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, PR China.
| |
Collapse
|
5
|
Afonso SL, Cataneo AJM, de Oliveira Carvalho PE. Lapatinib for advanced breast cancer overexpressing HER2. Hippokratia 2016. [DOI: 10.1002/14651858.cd009713.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Sergio L Afonso
- Marilia Medical School; Evidence Based Health Actions and Oncology; Sperendio Cabrini 295 Marilia Sao Paulo Brazil 17516-300
| | - Antonio José Maria Cataneo
- São Paulo State University; Department of Surgery & Orthopedics; Distrito de Rubião Júnior, s/n São Paulo Brazil 18618-970
| | - Paulo Eduardo de Oliveira Carvalho
- Marilia Medical School; Evidence Based Health Actions Department and Thoracic Surgery Department; Avenida Monte Carmelo, 800 Bairro Fragata Marilia Sao Paulo Brazil 17519-030
| |
Collapse
|
6
|
Wang SR, Malik S, Tan IB, Chan YS, Hoi Q, Ow JL, He CZ, Ching CE, Poh DYS, Seah HM, Cheung KHT, Perumal D, Devasia AG, Pan L, Ang S, Lee SE, Ten R, Chua C, Tan DSW, Qu JZZ, Bylstra YM, Lim L, Lezhava A, Ng PC, Wong CW, Lim T, Tan P. Technical Validation of a Next-Generation Sequencing Assay for Detecting Actionable Mutations in Patients with Gastrointestinal Cancer. J Mol Diagn 2016; 18:416-424. [PMID: 26970585 DOI: 10.1016/j.jmoldx.2016.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/02/2015] [Accepted: 01/08/2016] [Indexed: 02/06/2023] Open
Abstract
Targeted next-generation sequencing is becoming increasingly common as a clinical diagnostic and prognostic test for patient- and tumor-specific genetic profiles as well as to optimally select targeted therapies. Here, we describe a custom-developed, next-generation sequencing test for detecting single-nucleotide variants (SNVs) and short insertions and deletions (indels) in 93 genes related to gastrointestinal cancer from routine formalin-fixed, paraffin-embedded clinical specimens. We implemented a validation strategy, based on the College of American Pathologists requirements, using reference DNA mixtures from cell lines with known genetic variants, which model a broad range of allele frequencies. Test sensitivity achieved >99% for both SNVs and indels, with allele frequencies >10%, with high specificity (97.4% for SNVs and 93.6% for indels). We further confirmed test accuracies using primary formalin-fixed, paraffin-embedded colorectal cancer specimens characterized by alternative and conventional clinical diagnostic technologies. Robust performance was observed on the formalin-fixed, paraffin-embedded specimens: sensitivity was 97.2% and specificity was 99.2%. We also observed high intrarun and inter-run reproducibility, as well as a low cross-contamination rate. Overall assessment using cell line samples and formalin-fixed, paraffin-embedded samples showed that our custom next-generation sequencing assay has consistent detection sensitivity down to 10% variant frequency.
Collapse
Affiliation(s)
| | - Simeen Malik
- Cancer & Stem Cell Biology Program, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Iain B Tan
- Genome Institute of Singapore, Singapore; Cancer & Stem Cell Biology Program, Duke-National University of Singapore Graduate Medical School, Singapore; Department of Medical Oncology, National Cancer Centre Singapore, Singapore; Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| | | | | | - Jack L Ow
- Genome Institute of Singapore, Singapore
| | | | | | | | | | | | | | | | - Lu Pan
- Genome Institute of Singapore, Singapore
| | - Shimin Ang
- Genome Institute of Singapore, Singapore
| | | | - Rachel Ten
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Clarinda Chua
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Daniel S W Tan
- Genome Institute of Singapore, Singapore; Department of Medical Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore
| | | | - Yasmin M Bylstra
- Genome Institute of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Lionel Lim
- Genome Institute of Singapore, Singapore
| | | | | | | | - Tony Lim
- Genome Institute of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Patrick Tan
- Genome Institute of Singapore, Singapore; Cancer & Stem Cell Biology Program, Duke-National University of Singapore Graduate Medical School, Singapore; Cancer Science Institute Singapore, National University of Singapore, Singapore; Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
7
|
Xie J, Lu X, Wu X, Lin X, Zhang C, Huang X, Chang Z, Wang X, Wen C, Tang X, Shi M, Zhan Q, Chen H, Deng X, Peng C, Li H, Fang Y, Shao Y, Shen B. Capture-based next-generation sequencing reveals multiple actionable mutations in cancer patients failed in traditional testing. Mol Genet Genomic Med 2016; 4:262-72. [PMID: 27247954 PMCID: PMC4867560 DOI: 10.1002/mgg3.201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Targeted therapies including monoclonal antibodies and small molecule inhibitors have dramatically changed the treatment of cancer over past 10 years. Their therapeutic advantages are more tumor specific and with less side effects. For precisely tailoring available targeted therapies to each individual or a subset of cancer patients, next-generation sequencing (NGS) has been utilized as a promising diagnosis tool with its advantages of accuracy, sensitivity, and high throughput. METHODS We developed and validated a NGS-based cancer genomic diagnosis targeting 115 prognosis and therapeutics relevant genes on multiple specimen including blood, tumor tissue, and body fluid from 10 patients with different cancer types. The sequencing data was then analyzed by the clinical-applicable analytical pipelines developed in house. RESULTS We have assessed analytical sensitivity, specificity, and accuracy of the NGS-based molecular diagnosis. Also, our developed analytical pipelines were capable of detecting base substitutions, indels, and gene copy number variations (CNVs). For instance, several actionable mutations of EGFR,PIK3CA,TP53, and KRAS have been detected for indicating drug susceptibility and resistance in the cases of lung cancer. CONCLUSION Our study has shown that NGS-based molecular diagnosis is more sensitive and comprehensive to detect genomic alterations in cancer, and supports a direct clinical use for guiding targeted therapy.
Collapse
Affiliation(s)
- Jing Xie
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Department of PathologyRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiongxiong Lu
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xue Wu
- Department of Research and Development Geneseeq Technology Inc. Toronto Ontario Canada
| | - Xiaoyi Lin
- Department of Laboratory Medicine Ruijin Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Chao Zhang
- Department of Research and Development Geneseeq Technology Inc. Toronto Ontario Canada
| | - Xiaofang Huang
- Department of Research and Development Geneseeq Technology Inc. Toronto Ontario Canada
| | - Zhili Chang
- Department of Research and Development Geneseeq Technology Inc. Toronto Ontario Canada
| | - Xinjing Wang
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Chenlei Wen
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaomei Tang
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Minmin Shi
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qian Zhan
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hao Chen
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaxing Deng
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Chenghong Peng
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hongwei Li
- Pancreatic Disease Centre Ruijin Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Yuan Fang
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yang Shao
- Department of Research and DevelopmentGeneseeq Technology Inc.TorontoOntarioCanada; Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
| | - Baiyong Shen
- Research Institute of Pancreatic DiseaseRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Pancreatic Disease CentreRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina; Shanghai Institute of Digestive SurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
8
|
Kang DY, Darvin P, Yoo YB, Joung YH, Sp N, Byun HJ, Yang YM. Methylsulfonylmethane inhibits HER2 expression through STAT5b in breast cancer cells. Int J Oncol 2015; 48:836-42. [PMID: 26648017 DOI: 10.3892/ijo.2015.3277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/22/2015] [Indexed: 11/06/2022] Open
Abstract
Breast cancer is the most common cancer in women globally. The factors that increase risk include: late age at first birth, alcohol, radiation exposure, family history of breast cancer, and postmenopausal hormone therapy. Numerous drugs are being developed to treat breast cancer. Among them, Herceptin is used for the treatment of human epidermal growth factor receptor 2 (HER2)-positive cases and targets HER2 effectively and efficiently, but it is very expensive. Methylsulfonylmethane (MSM) is an organic sulfur-containing natural compound having no reported toxicity. We examined MSM in breast cancer cell lines and found it inhibited the proliferation of estrogen receptor-positive and HER2-positive breast cancer cells in a dose-dependent manner. It also suppressed the activation of STAT5b and expression of HER2 in breast cancer cells. We determined the STAT5b binding site (GAS element) in the HER2 gene. Detailed analysis showed that MSM decreased the ability of STAT5b to bind the promoter of the HER2 gene and a luciferase assay demonstrated reduced activity. We confirmed that MSM can effectively regulate STAT5b, and thereby decrease HER2 expression. Therefore, we recommend the use of MSM as an inhibitor for the management of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Dong Young Kang
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Pramod Darvin
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Young Beom Yoo
- Department of Surgery, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Youn Hee Joung
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Nipin Sp
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Hyo Joo Byun
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Young Mok Yang
- Department of Pathology, School of Medicine, and Institute of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Peng Z, Liu Y, Li Y, Zhang X, Zhou J, Lu M, Li Q, Shen L. Serum HER2 extracellular domain as a potential alternative for tissue HER2 status in metastatic gastric cancer patients. Biomark Med 2015; 8:663-70. [PMID: 25123035 DOI: 10.2217/bmm.14.10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AIM We investigated whether serum concentrations of the HER2 extracellular domain (ECD) can be used as an alternative to test tissue HER2 status in metastatic gastric cancer. MATERIALS & METHODS A total of 133 cases of metastatic gastric cancer were included in present study. Serum HER2 ECD was measured by chemiluminescence immunoassay (CLIA). Receiver operating characteristic curve analysis was used to determine optimal serum HER2 ECD concentrations for differentiation between positive and negative HER2 status. RESULTS The median level of serum HER2 ECD was 9.6 ng/ml in metastatic gastric cancer patients. There was a significant relationship between serum and tissue levels of HER2 protein (p < 0.001). Area under the curve for serum HER2 ECD was 0.771 (95% CI: 0.682-0.860). CONCLUSION Levels of serum HER2 ECD are highly correlated with tissue HER2 status in metastatic gastric cancer. Serum HER2 ECD assay can be considered as a potential alternative for tissue HER2 status.
Collapse
Affiliation(s)
- Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
10
|
KRAS and BRAF Mutation Detection: Is Immunohistochemistry a Possible Alternative to Molecular Biology in Colorectal Cancer? Gastroenterol Res Pract 2015; 2015:753903. [PMID: 25983749 PMCID: PMC4422999 DOI: 10.1155/2015/753903] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/07/2015] [Indexed: 01/05/2023] Open
Abstract
KRAS genotyping is mandatory in metastatic colorectal cancer treatment prior to undertaking antiepidermal growth factor receptor (EGFR) monoclonal antibody therapy. BRAF V600E mutation is often present in colorectal carcinoma with CpG island methylator phenotype and microsatellite instability. Currently, KRAS and BRAF evaluation is based on molecular biology techniques such as SNaPshot or Sanger sequencing. As molecular testing is performed on formalin-fixed paraffin-embedded (FFPE) samples, immunodetection would appear to be an attractive alternative for detecting mutations. Thus, our objective was to assess the validity of KRAS and BRAF immunodetection of mutations compared with the genotyping reference method in colorectal adenocarcinoma. KRAS and BRAF genotyping was assessed by SNaPshot. A rabbit anti-human KRAS polyclonal antibody was tested on 33 FFPE colorectal tumor samples with known KRAS status. Additionally, a mouse anti-human BRAF monoclonal antibody was tested on 30 FFPE tumor samples with known BRAF status. KRAS immunostaining demonstrated both poor sensitivity (27%) and specificity (64%) in detecting KRAS mutation. Conversely, BRAF immunohistochemistry showed perfect sensitivity (100%) and specificity (100%) in detecting V600E mutation. Although molecular biology remains the reference method for detecting KRAS mutation, immunohistochemistry could be an attractive method for detecting BRAF V600E mutation in colorectal cancer.
Collapse
|
11
|
Ali SM, Sanford EM, Klempner SJ, Rubinson DA, Wang K, Palma NA, Chmielecki J, Yelensky R, Palmer GA, Morosini D, Lipson D, Catenacci DV, Braiteh F, Erlich R, Stephens PJ, Ross JS, Ou SHI, Miller VA. Prospective comprehensive genomic profiling of advanced gastric carcinoma cases reveals frequent clinically relevant genomic alterations and new routes for targeted therapies. Oncologist 2015; 20:499-507. [PMID: 25882375 DOI: 10.1634/theoncologist.2014-0378] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 02/04/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major global cancer burden and the second most common cause of global cancer-related deaths. The addition of anti-ERBB2 (HER2) targeted therapy to chemotherapy improves survival for ERBB2-amplified advanced GC patients; however, the majority of GC patients do not harbor this alteration and thus cannot benefit from targeted therapy under current practice paradigms. MATERIALS AND METHODS Prospective comprehensive genomic profiling of 116 predominantly locally advanced or metastatic (90.0%) gastric cancer cases was performed to identify genomic alterations (GAs) associated with a potential response to targeted therapies approved by the U.S. Food and Drug Administration or targeted therapy-based clinical trials. RESULTS Overall, 78% of GC cases harbored one clinically relevant GA or more, with the most frequent alterations being found in TP53 (50%), ARID1A (24%), KRAS (16%), CDH1 (15%), CDKN2A (14%), CCND1 (9.5%), ERBB2 (8.5%), PIK3CA (8.6%), MLL2 (6.9%), FGFR2 (6.0%), and MET (6.0%). Receptor tyrosine kinase genomic alterations were detected in 20.6% of cases, primarily ERBB2, FGFR2, and MET amplification, with ERBB2 alterations evenly split between amplifications and base substitutions. Rare BRAF mutations (2.6%) were also observed. One MET-amplified GC patient responded for 5 months to crizotinib, a multitargeted ALK/ROS1/MET inhibitor. CONCLUSION Comprehensive genomic profiling of GC identifies clinically relevant GAs that suggest benefit from targeted therapy including MET-amplified GC and ERBB2 base substitutions.
Collapse
Affiliation(s)
- Siraj M Ali
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Eric M Sanford
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Samuel J Klempner
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Douglas A Rubinson
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Kai Wang
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Norma A Palma
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Juliann Chmielecki
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Roman Yelensky
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Gary A Palmer
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Deborah Morosini
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Doron Lipson
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Daniel V Catenacci
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Fadi Braiteh
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Rachel Erlich
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Philip J Stephens
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Sai-Hong Ignatius Ou
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | - Vincent A Miller
- Foundation Medicine Inc., Cambridge, Massachusetts, USA; Chao Family Comprehensive Cancer Center, Division of Hematology-Oncology, Department of Medicine, University of California Irvine School of Medicine, Orange, California, USA; Dana-Farber Cancer Institute, Boston, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| |
Collapse
|
12
|
Schink JC, Trosman JR, Weldon CB, Siziopikou KP, Tsongalis GJ, Rademaker AW, Patel JD, Benson AB, Perez EA, Gradishar WJ. Biomarker testing for breast, lung, and gastroesophageal cancers at NCI designated cancer centers. J Natl Cancer Inst 2014; 106:dju256. [PMID: 25217578 PMCID: PMC4176043 DOI: 10.1093/jnci/dju256] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/03/2014] [Accepted: 07/17/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Molecular biomarkers, a cornerstone of precision oncology, are critical in breast, gastroesophageal, and non-small cell lung cancer management (BC, GEC, NSCLC). Testing practices are intensely debated, impacting diagnostic quality and affecting pathologists, oncologists and patients. However, little is known about testing approaches used in practice. Our study described biomarker practices in BC, GEC, and NSCLC at the leading US cancer centers. METHODS We conducted a survey of the National Cancer Institute (NCI) designated centers on BC, GEC, and NSCLC biomarker testing. We used simple frequencies to describe practices, two-sided Fisher's exact test and two-sided McNemar's test for cross-cancer comparison. All statistical tests were two-sided. RESULTS For BC human epidermal growth factor receptor 2 (HER2), 39% of centers combine guidelines by using in situ hybridization (ISH) and immunohistochemistry (IHC) concurrently, and 21% reflex-test beyond guideline-recommended IHC2+. For GEC HER2, 44% use ISH and IHC concurrently, and 28% reflex-test beyond IHC2+. In NSCLC, the use of IHC is limited to 4% for epidermal growth factor receptor (EGFR) and 7% for anaplastic lymphoma kinase (ALK). 43.5% test NSCLC biomarkers on oncologist order; 34.5% run all biomarkers upfront, and 22% use a sequential protocol. NSCLC external testing is statistically significantly higher than BC (P < .0001) and GEC (P < .0001). NSCLC internally developed tests are statistically significantly more common than BC (P < .0001) and GEC (P < .0001). CONCLUSIONS At the NCI cancer centers, biomarker testing practices vary, but exceeding guidelines is a common practice for established biomarkers and emerging practice for newer biomarkers. Use of internally developed tests declines as biomarkers mature. Implementation of multibiomarker protocols is lagging. Our study represents a step toward developing a biomarker testing practice landscape.
Collapse
Affiliation(s)
- Julian C Schink
- * Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Julia R Trosman
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Christine B Weldon
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Kalliopi P Siziopikou
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Gregory J Tsongalis
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Alfred W Rademaker
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Jyoti D Patel
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Al B Benson
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Edith A Perez
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - William J Gradishar
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI.
| |
Collapse
|
13
|
Ding R, Yu YH. New advances in HER2 testing in gastric cancer. Shijie Huaren Xiaohua Zazhi 2013; 21:4098-4103. [DOI: 10.11569/wcjd.v21.i36.4098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common malignancies and represents the second leading cause of cancer death worldwide. The ToGA trial has shown that the humanized anti-HER2 (human epidermal growth factor receptor-2) monoclonal antibody Trastuzumab is effective in prolonging survival in patients with HER2-positive carcinoma of the stomach and the gastroesophageal junction (GEJ). Trastuzumab in combination with chemotherapy can be considered as a new standard option for patients with HER2-positive advanced gastric or gastro-oesophageal junction cancer. Therefore, it is crucial to accurately screen patients with HER2-positive gastric cancer. This article mainly discusses the recent advances in HER2 testing in gastric cancer.
Collapse
|
14
|
Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, Schnall-Levin M, White J, Sanford EM, An P, Sun J, Juhn F, Brennan K, Iwanik K, Maillet A, Buell J, White E, Zhao M, Balasubramanian S, Terzic S, Richards T, Banning V, Garcia L, Mahoney K, Zwirko Z, Donahue A, Beltran H, Mosquera JM, Rubin MA, Dogan S, Hedvat CV, Berger MF, Pusztai L, Lechner M, Boshoff C, Jarosz M, Vietz C, Parker A, Miller VA, Ross JS, Curran J, Cronin MT, Stephens PJ, Lipson D, Yelensky R. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 2013; 31:1023-31. [PMID: 24142049 PMCID: PMC5710001 DOI: 10.1038/nbt.2696] [Citation(s) in RCA: 1723] [Impact Index Per Article: 143.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/19/2013] [Indexed: 02/07/2023]
Abstract
As more clinically relevant cancer genes are identified, comprehensive diagnostic approaches are needed to match patients to therapies, raising the challenge of optimization and analytical validation of assays that interrogate millions of bases of cancer genomes altered by multiple mechanisms. Here we describe a test based on massively parallel DNA sequencing to characterize base substitutions, short insertions and deletions (indels), copy number alterations and selected fusions across 287 cancer-related genes from routine formalin-fixed and paraffin-embedded (FFPE) clinical specimens. We implemented a practical validation strategy with reference samples of pooled cell lines that model key determinants of accuracy, including mutant allele frequency, indel length and amplitude of copy change. Test sensitivity achieved was 95-99% across alteration types, with high specificity (positive predictive value >99%). We confirmed accuracy using 249 FFPE cancer specimens characterized by established assays. Application of the test to 2,221 clinical cases revealed clinically actionable alterations in 76% of tumors, three times the number of actionable alterations detected by current diagnostic tests.
Collapse
Affiliation(s)
| | | | - Geoff A Otto
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Kai Wang
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Jie He
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Jared White
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Peter An
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - James Sun
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Frank Juhn
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Kiel Iwanik
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Jamie Buell
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Emily White
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Mandy Zhao
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | | | | | - Vera Banning
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | | | - Zac Zwirko
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Amy Donahue
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Himisha Beltran
- Department of Medicine, Division of Hematology and Medical Oncology,
Weill Medical College of Cornell University, New York, New York, USA
- Institute for Precision Medicine, Weill Cornell Medical College and
New York-Presbyterian Hospital
| | - Juan Miguel Mosquera
- Institute for Precision Medicine, Weill Cornell Medical College and
New York-Presbyterian Hospital
- Department of Pathology and Laboratory Medicine, Weill Medical
College of Cornell University, New York, New York, USA
| | - Mark A Rubin
- Institute for Precision Medicine, Weill Cornell Medical College and
New York-Presbyterian Hospital
- Department of Pathology and Laboratory Medicine, Weill Medical
College of Cornell University, New York, New York, USA
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New
York, New York, USA
| | - Cyrus V Hedvat
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New
York, New York, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New
York, New York, USA
| | - Lajos Pusztai
- Yale Cancer Center Genetics and Genomics Program, Yale School of
Medicine, New Haven, Connecticut, USA
| | | | - Chris Boshoff
- UCL Cancer Institute, University College London, London, UK
| | - Mirna Jarosz
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Alex Parker
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Jeffrey S Ross
- Foundation Medicine, Cambridge, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Albany Medical
College, Albany, New York, USA
| | - John Curran
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | | | - Doron Lipson
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | |
Collapse
|
15
|
Magee MS, Snook AE, Marszalowicz GP, Waldman SA. Immunotherapeutic strategies to target prognostic and predictive markers of cancer. Biomark Med 2013; 7:23-35. [PMID: 23387482 DOI: 10.2217/bmm.12.110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Throughout the last century medical advances in cancer treatment in the fields of surgery, radiation therapy and chemotherapy have greatly impacted patients' survival rates. Nevertheless, cancer remains a significant cause of mortality, with an estimated 7.6 million deaths worldwide in 2008, reflecting the inability of existing therapies to effectively cure disease. The emergence of vaccines and their successes in preventing the spread of infectious diseases has demonstrated the unique specificity and therapeutic potential of the immune system. This potential has driven the development of novel cancer immunotherapeutics. This review focuses on the current status of the use of immunologic effectors to target known biomarkers in cancer.
Collapse
Affiliation(s)
- Michael S Magee
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, 132 South 10th Street, 1170 Main, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
16
|
Toomey PG, Vohra NA, Ghansah T, Sarnaik AA, Pilon-Thomas SA. Immunotherapy for gastrointestinal malignancies. Cancer Control 2013; 20:32-42. [PMID: 23302905 DOI: 10.1177/107327481302000106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers are the most common human tumors encountered worldwide. The majority of GI cancers are unresectable at the time of diagnosis, and in the subset of patients undergoing resection, few are cured. There is only a modest improvement in survival with the addition of modalities such as chemotherapy and radiation therapy. Due to an increasing global cancer burden, it is imperative to integrate alternative strategies to improve outcomes. It is well known that cancers possess diverse strategies to evade immune detection and destruction. This has led to the incorporation of various immunotherapeutic strategies, which enable reprogramming of the immune system to allow effective recognition and killing of GI tumors. METHODS A review was conducted of the results of published clinical trials employing immunotherapy for esophageal, gastroesophageal, gastric, hepatocellular, pancreatic, and colorectal cancers. RESULTS Monoclonal antibody therapy has come to the forefront in the past decade for the treatment of colorectal cancer. Immunotherapeutic successes in solid cancers such as melanoma and prostate cancer have led to the active investigation of immunotherapy for GI malignancies, with some promising results. CONCLUSIONS To date, monoclonal antibody therapy is the only immunotherapy approved by the US Food and Drug Administration for GI cancers. Initial trials validating new immunotherapeutic approaches, including vaccination-based and adoptive cell therapy strategies, for GI malignancies have demonstrated safety and the induction of antitumor immune responses. Therefore, immunotherapy is at the forefront of neoadjuvant as well as adjuvant therapies for the treatment and eradication of GI malignancies.
Collapse
Affiliation(s)
- Paul G Toomey
- Department of Surgery, USF Health Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
17
|
Bayrak M, Olmez OF, Kurt E, Cubukcu E, Evrensel T, Kanat O, Manavoglu O. Prognostic significance of c-erbB2 overexpression in patients with metastatic gastric cancer. Clin Transl Oncol 2013; 15:307-312. [PMID: 22911549 DOI: 10.1007/s12094-012-0921-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/19/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Overexpression of the gene c-erbB2, which encodes a receptor tyrosine kinase, has been associated with prognosis and response to therapy in several solid tumors. This study was designed to test whether c-erb-B2 overexpression can be related to prognosis of patients with metastatic gastric cancer. METHODS Between 2005 and 2010, 46 cases of metastatic gastric cancer were evaluated immunohistochemically for c-erb-B2 overexpression. Overall survival (OS) and time-to-progression (TTP) served as the main outcome measures. RESULTS c-erbB2 was overexpressed in 19 (41.3 %) cases and 8 patients (17.4 %) had a c-erbB2 score of 3+ (a strong complete membrane staining observed in >10 % of the tumor cells). c-erbB2 expression was not associated with the clinicohistological characteristics of the study participants. The mean OS was 11.48 ± 1.03 months, whereas the mean TTP was 8.28 ± 0.8 months. Compared with patients with a score of 2+ or less (n = 38), those with a c-erbB2 score of 3+ (n = 8) had both a significantly lower OS (15.55 ± 1.63 vs. 8.22 ± 0.88 months, respectively, p < 0.05) and TTP (10.72 ± 1.81 vs. 6.11 ± 0.61 months, respectively, p < 0.05). After allowance for potential confounders, Cox regression analysis identified a c-erbB2 score of 3+ as an independent predictor of both OS (hazard ratio = 1.9; 95 % confidence interval = 1.1-3.7, p < 0.05) and TTP (hazard ratio = 1.8; 95 % confidence interval = 1.1-4.1, p < 0.05). CONCLUSION Our results suggest that c-erbB-2 overexpression may have a prognostic significance in patients with metastatic gastric cancer.
Collapse
Affiliation(s)
- Muharrem Bayrak
- Department of Medical Oncology, Uludag University Medical School, Bursa 16059, Turkey
| | | | | | | | | | | | | |
Collapse
|
18
|
Mazouni C, Rimareix F, Mathieu MC, Uzan C, Bourgier C, André F, Delaloge S, Garbay JR. Outcome in breast molecular subtypes according to nodal status and surgical procedures. Am J Surg 2013; 205:662-7. [PMID: 23312273 DOI: 10.1016/j.amjsurg.2012.06.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 06/01/2012] [Accepted: 06/08/2012] [Indexed: 01/16/2023]
Abstract
BACKGROUND The purpose of our study was to evaluate the surgical treatment and outcome of breast cancer according to molecular subtypes. METHODS We identified 1,194 patients consecutively treated for primary breast cancer from 2004 to 2010. The type of surgery, pathological findings, local recurrence, and distant metastasis were evaluated for 5 molecular subtypes: luminal A and B, luminal HER2 (Human Epidermal Growth Factor Receptor 2), HER2 , and triple negative. RESULTS Breast-conserving surgery (BCS) was performed more frequently in luminal A (70.6%), triple-negative (66.2%), and luminal HER2 tumors (60.9%) (P < .001). A sentinel node biopsy was performed more frequently in luminal A (60%), and luminal HER2 (29.3%) types (P < .001). Among the 791 BCS, positive nodes were observed more often in HER2 (50%) and luminal B (44.9%) types (P = .0003). The number of local recurrences was higher in the node-negative luminal B subtype (3.4%). CONCLUSIONS Molecular subtypes exert an impact on BCS and nodal surgery rates. The local relapse rates are influenced by the molecular subtypes according to the nodal status.
Collapse
Affiliation(s)
- Chafika Mazouni
- Department of Breast Surgery, Institut Gustave Roussy, Villejuif, France.
| | | | | | | | | | | | | | | |
Collapse
|