1
|
Kaifi J, Joshi K, Suvilesh K, Natesh N, Manjunath Y, Coberly J, Schlink S, Kunin J, Prather R, Whitworth K, Nelson B, Bryan J, Hoffman T, Golzy M, Raju M, Teixeiro E, Telugu B, Rachagani S. Characterization of A Bronchoscopically Induced Transgenic Lung Cancer Pig Model for Human Translatability. RESEARCH SQUARE 2025:rs.3.rs-5054204. [PMID: 39975891 PMCID: PMC11838710 DOI: 10.21203/rs.3.rs-5054204/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background There remains a need for animal models with human translatability in lung cancer (LC) research. Findings in pigs have high impact on humans due to similar anatomy and physiology. We present the characterization of a bronchoscopically-induced LC model in Oncopigs carrying inducible KRASG12D and TP53R167H mutations. Methods Twelve Oncopigs underwent 29 injections via flexible bronchoscopy. Eighteen Adenovirus-Cre recombinase gene (AdCre) inductions were performed endobronchially (n = 6) and transbronchially with a needle (n = 12). Eleven control injections were performed without AdCre. Oncopigs underwent serial contrast-enhanced chest CT with clinical follow-up for 29 weeks. Following autopsy, lung and organ tissues underwent histopathology, immunohistochemistry, and RNA-sequencing with comparative analysis with The Cancer Genome Atlas (TCGA) human LC data. Results All 18 sites of AdCre injections had lung consolidations on CT imaging. Transbronchial injections led to histopathologic invasive cancer and/or carcinoma in situ (CIS) in 11/12 (91.7%), and invasive cancer (excluding CIS) in 8/12 (66.6%). Endobronchial inductions led to invasive cancer in 3/6 (50%). A soft tissue metastasis was observed in one Oncopig. Immunohistochemistry confirmed expression of Pan-CK+/epithelial cancer cells, with macrophages and T cells infiltration in the tumor microenvironment. Transcriptome comparison showed 54.3% overlap with human LC (TCGA), in contrast to 29.88% overlap of KRAS-mutant mouse LC with human LC. Conclusions The transgenic and immunocompetent Oncopig model has a high rate of LC following bronchoscopic transbronchial induction. Overlap of the Oncopig LC transcriptome with human LC transcriptome was noted. This pig model is expected to have high clinical translatability to the human LC patient.
Collapse
|
2
|
Bhat N, Al-Mathkour M, Maacha S, Lu H, El-Rifai W, Ballout F. Esophageal adenocarcinoma models: a closer look. Front Mol Biosci 2024; 11:1440670. [PMID: 39600303 PMCID: PMC11589788 DOI: 10.3389/fmolb.2024.1440670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Esophageal adenocarcinoma (EAC) is a subtype of esophageal cancer with significant morbidity and mortality rates worldwide. Despite advancements in tumor models, the underlying cellular and molecular mechanisms driving EAC pathogenesis are still poorly understood. Therefore, gaining insights into these mechanisms is crucial for improving patient outcomes. Researchers have developed various models to better understand EAC and evaluate clinical management strategies. However, no single model fully recapitulates the complexity of EAC. Emerging technologies, such as patient-derived organoids and immune-competent mouse models, hold promise for personalized EAC research and drug development. In this review, we shed light on the various models for studying EAC and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Nadeem Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Marwah Al-Mathkour
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Selma Maacha
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Farah Ballout
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
3
|
Joshi K, Suvilesh KN, Natesh NS, Manjunath Y, Coberly J, Schlink S, Kunin JR, Prather RS, Whitworth K, Nelson B, Bryan JN, Hoffman T, Golzy M, Raju M, Teixeiro E, Telugu BP, Kaifi JT, Rachagani S. Characterization of A Bronchoscopically Induced Transgenic Lung Cancer Pig Model for Human Translatability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621940. [PMID: 39569144 PMCID: PMC11577786 DOI: 10.1101/2024.11.04.621940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Background There remains a need for animal models with human translatability in lung cancer (LC) research. Findings in pigs have high impact on humans due to similar anatomy and physiology. We present the characterization of a bronchoscopically-induced LC model in Oncopigs carrying inducible KRASG12D and TP53R167H mutations. Methods Twelve Oncopigs underwent 29 injections via flexible bronchoscopy. Eighteen Adenovirus-Cre recombinase gene (AdCre) inductions were performed endobronchially (n=6) and transbronchially with a needle (n=12). Eleven control injections were performed without AdCre. Oncopigs underwent serial contrast-enhanced chest CT with clinical follow-up for 29 weeks. Following autopsy, lung and organ tissues underwent histopathology, immunohistochemistry, and RNA-sequencing with comparative analysis with The Cancer Genome Atlas (TCGA) human LC data. Results All 18 sites of AdCre injections had lung consolidations on CT imaging. Transbronchial injections led to histopathologic invasive cancer and/or carcinoma in situ (CIS) in 11/12 (91.7%), and invasive cancer (excluding CIS) in 8/12 (66.6%). Endobronchial inductions led to invasive cancer in 3/6 (50%). A soft tissue metastasis was observed in one Oncopig. Immunohistochemistry confirmed expression of Pan-CK+/epithelial cancer cells, with macrophages and T cells infiltration in the tumor microenvironment. Transcriptome comparison showed 54.3% overlap with human LC (TCGA), in contrast to 29.88% overlap of KRAS-mutant mouse LC with human LC. Conclusions The transgenic and immunocompetent Oncopig model has a high rate of LC following bronchoscopic transbronchial induction. Overlap of the Oncopig LC transcriptome with human LC transcriptome was noted. This pig model is expected to have high clinical translatability to the human LC patient.
Collapse
Affiliation(s)
- Kirtan Joshi
- Department of Medicine/Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Surgery, University of Missouri, Columbia, Missouri, United States of America
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Kanve N. Suvilesh
- Department of Surgery, University of Missouri, Columbia, Missouri, United States of America
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States of America
| | - Nagabhishek Sirpu Natesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Yariswamy Manjunath
- Department of Surgery, University of Missouri, Columbia, Missouri, United States of America
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States of America
| | - Jared Coberly
- Department of Clinical Pathology, University of Missouri, Columbia, Missouri, United States of America
| | - Sarah Schlink
- Office of Animal Resources, University of Missouri, Columbia, Missouri, United States of America
| | - Jeffrey R. Kunin
- Department Of Radiology, University of Missouri, Columbia, Missouri, United States of America
| | - Randall S. Prather
- Animal Science Research Center of the National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Kristin Whitworth
- Animal Science Research Center of the National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Benjamin Nelson
- Animal Science Research Center of the National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jeffrey N. Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| | - Timothy Hoffman
- Department of Medicine/Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri, United States of America
| | - Mojgan Golzy
- Biomedical Informatics, Biostatistics and Medical Epidemiology, University of Missouri, Columbia, Missouri, United States of America
| | - Murugesan Raju
- Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, United States of America
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States of America
| | - Bhanu P. Telugu
- Animal Science Research Center of the National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jussuf T. Kaifi
- Department of Surgery, University of Missouri, Columbia, Missouri, United States of America
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
- Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, United States of America
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States of America
- Alvin J. Siteman Cancer Center, Washington University, Saint Louis, Missouri, United States of America
| | - Satyanarayana Rachagani
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, Missouri, United States of America
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
4
|
Mahmoudian RA, Farshchian M, Golyan FF, Mahmoudian P, Alasti A, Moghimi V, Maftooh M, Khazaei M, Hassanian SM, Ferns GA, Mahaki H, Shahidsales S, Avan A. Preclinical tumor mouse models for studying esophageal cancer. Crit Rev Oncol Hematol 2023; 189:104068. [PMID: 37468084 DOI: 10.1016/j.critrevonc.2023.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Preclinical models are extensively employed in cancer research because they can be manipulated in terms of their environment, genome, molecular biology, organ systems, and physical activity to mimic human behavior and conditions. The progress made in in vivo cancer research has resulted in significant advancements, enabling the creation of spontaneous, metastatic, and humanized mouse models. Most recently, the remarkable and extensive developments in genetic engineering, particularly the utilization of CRISPR/Cas9, transposable elements, epigenome modifications, and liquid biopsies, have further facilitated the design and development of numerous mouse models for studying cancer. In this review, we have elucidated the production and usage of current mouse models, such as xenografts, chemical-induced models, and genetically engineered mouse models (GEMMs), for studying esophageal cancer. Additionally, we have briefly discussed various gene-editing tools that could potentially be employed in the future to create mouse models specifically for esophageal cancer research.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Farshchian
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Alasti
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Moghimi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
5
|
Huo Z, Bilang R, Supuran CT, von der Weid N, Bruder E, Holland-Cunz S, Martin I, Muraro MG, Gros SJ. Perfusion-Based Bioreactor Culture and Isothermal Microcalorimetry for Preclinical Drug Testing with the Carbonic Anhydrase Inhibitor SLC-0111 in Patient-Derived Neuroblastoma. Int J Mol Sci 2022; 23:ijms23063128. [PMID: 35328549 PMCID: PMC8955558 DOI: 10.3390/ijms23063128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/23/2022] Open
Abstract
Neuroblastoma is a rare disease. Rare are also the possibilities to test new therapeutic options for neuroblastoma in clinical trials. Despite the constant need to improve therapy and outcomes for patients with advanced neuroblastoma, clinical trials currently only allow for testing few substances in even fewer patients. This increases the need to improve and advance preclinical models for neuroblastoma to preselect favorable candidates for novel therapeutics. Here we propose the use of a new patient-derived 3D slice-culture perfusion-based 3D model in combination with rapid treatment evaluation using isothermal microcalorimetry exemplified with treatment with the novel carbonic anhydrase IX and XII (CAIX/CAXII) inhibitor SLC-0111. Patient samples showed a CAIX expression of 18% and a CAXII expression of 30%. Corresponding with their respective CAIX expression patterns, the viability of SH-EP cells was significantly reduced upon treatment with SLC-0111, while LAN1 cells were not affected. The inhibitory effect on SH-SY5Y cells was dependent on the induction of CAIX expression under hypoxia. These findings corresponded to thermogenesis of the cells. Patient-derived organotypic slice cultures were treated with SLC-0111, which was highly effective despite heterogeneity of CAIX/CAXII expression. Thermogenesis, in congruence with the findings of the histological observations, was significantly reduced in SLC-0111-treated samples. In order to extend the evaluation time, we established a perfusion-based approach for neuroblastoma tissue in a 3D perfusion-based bioreactor system. Using this system, excellent tissue quality with intact tumor cells and stromal structure in neuroblastoma tumors can be maintained for 7 days. The system was successfully used for consecutive drug response monitoring with isothermal microcalorimetry. The described approach for drug testing, relying on an advanced 3D culture system combined with a rapid and highly sensitive metabolic assessment, can facilitate development of personalized treatment strategies for neuroblastoma.
Collapse
Affiliation(s)
- Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Remo Bilang
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Claudiu T. Supuran
- Department Neurofarba, Sezione di Scienze Farmaceutiche, University of Florence, 50121 Florence, Italy;
| | - Nicolas von der Weid
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
- Department of Hematology and Oncology, University Children’s Hospital Basel, 4031 Basel, Switzerland
| | - Elisabeth Bruder
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland;
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland; (I.M.); (M.G.M.)
| | - Manuele G. Muraro
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland; (I.M.); (M.G.M.)
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
- Correspondence:
| |
Collapse
|
6
|
Weidle UH, Nopora A. MicroRNAs and Corresponding Targets in Esophageal Cancer as Shown In Vitro and In Vivo in Preclinical Models. Cancer Genomics Proteomics 2022; 19:113-129. [PMID: 35181582 DOI: 10.21873/cgp.20308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/08/2023] Open
Abstract
Squamous cell carcinoma of the esophagus is associated with a dismal prognosis. Therefore, identification of new targets and implementation of new treatment modalities are issues of paramount importance. Based on a survey of the literature, we identified microRNAs conferring antitumoral activity in preclinical in vivo experiments. In the category of miRs targeting secreted factors and transmembrane receptors, four miRs were up-regulated and 10 were down-regulated compared with five out of nine in the category transcription factors, and six miRs were down-regulated in the category enzymes, including metabolic enzymes. The down-regulated miRs have targets which can be inhibited by small molecules or antibody-related entities, or re-expressed by reconstitution therapy. Up-regulated miRs have targets which can be reconstituted with small molecules or inhibited with antagomirs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
7
|
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021; 11:3379-3392. [PMID: 34900524 PMCID: PMC8642427 DOI: 10.1016/j.apsb.2021.03.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers with high morbidity and mortality rates. EC includes two histological subtypes, namely esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC primarily occurs in East Asia, whereas EAC occurs in Western countries. The currently available treatment strategies for EC include surgery, chemotherapy, radiation therapy, molecular targeted therapy, and combinations thereof. However, the prognosis remains poor, and the overall five-year survival rate is very low. Therefore, achieving the goal of effective treatment remains challenging. In this review, we discuss the latest developments in chemotherapy and molecular targeted therapy for EC, and comprehensively analyze the application prospects and existing problems of immunotherapy. Collectively, this review aims to provide a better understanding of the currently available drugs through in-depth analysis, promote the development of new therapeutic agents, and eventually improve the treatment outcomes of patients with EC.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jian Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
8
|
Metastatic Esophageal Carcinoma Cells Exhibit Reduced Adhesion Strength and Enhanced Thermogenesis. Cells 2021; 10:cells10051213. [PMID: 34065626 PMCID: PMC8156073 DOI: 10.3390/cells10051213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 12/27/2022] Open
Abstract
Despite continuous improvements in multimodal therapeutic strategies, esophageal carcinoma maintains a high mortality rate. Metastases are a major life-limiting component; however, very little is known about why some tumors have high metastatic potential and others not. In this study, we investigated thermogenic activity and adhesion strength of primary tumor cells and corresponding metastatic cell lines derived from two patients with metastatic adenocarcinoma of the esophagus. We hypothesized that the increased metastatic potential of the metastatic cell lines correlates with higher thermogenic activity and decreased adhesion strength. Our data show that patient-derived metastatic esophageal tumor cells have a higher thermogenic profile as well as a decreased adhesion strength compared to their corresponding primary tumor cells. Using two paired esophageal carcinoma cell lines of primary tumor and lymph nodes makes the data unique. Both higher specific thermogenesis profile and decreased adhesion strength are associated with a higher metastatic potential. They are in congruence with the clinical patient presentation. Understanding these functional, biophysical properties of patient derived esophageal carcinoma cell lines will enable us to gain further insight into the mechanisms of metastatic potential of primary tumors and metastases. Microcalorimetric evaluation will furthermore allow for rapid assessment of new treatment options for primary tumor and metastases aimed at decreasing the metastatic potential.
Collapse
|
9
|
Gao P, Liu H, Yang Z, Hui Y, Shi Z, Yang Z, Song M, Yao M, Fan W, Yang J, Hao Y, Fan T. Development of a Novel Highly Spontaneous Metastatic Model of Esophageal Squamous Cell Carcinoma Using Renal Capsule Technology. Onco Targets Ther 2021; 14:785-793. [PMID: 33574674 PMCID: PMC7872218 DOI: 10.2147/ott.s290564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/13/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Increasing evidence has demonstrated that animal models are imperative to investigate the potential molecular mechanism of metastasis and discover anti-metastasis drugs; however, efficient animal models to unveil the underlying mechanisms of metastasis in esophageal squamous cell carcinoma (ESCC) are limited. METHODS ESCC cell EC9706 with high invasiveness was screened by repeated Transwell assays. Its biological characteristics were identified by flow cytometry as well as by the wound healing and CCK-8 assays. Besides, the levels of epithelial-mesenchymal transition-related markers were examined using Western blotting. Parental (EC9706-I0) and subpopulation (EC9706-I3) cells were employed to establish the renal capsule model. Next, the tumor growth was detected by a live animal imaging system, and hematoxylin and eosin staining was applied to evaluate the metastatic status in ESCC. RESULTS EC9706-I3 cells showed rapid proliferation ability, S phase abundance, and high invasive ability; obvious upregulation in N-cadherin, Snail, Vimentin, and Bit1; and downregulation in E-cadherin. EC9706-I3 cells were less sensitive to the chemotherapy drug 5-fluorouracil than EC9706-I0 cells; however, both cell lines reached a tumorigenesis rate of 100% in the renal capsule model. The live animal imaging system revealed that the tumors derived from EC9706-I0 cells grew more slowly than those from EC9706-I3 cells at weeks 3-14. The EC9706-I3 xenograft model displayed a spontaneous metastatic site, including kidney, heart, liver, lung, pancreas, and spleen, with a distant metastatic rate of 80%. CONCLUSION Our data suggested that the metastatic model was successfully established, providing a novel platform for further exploring the molecular mechanisms of metastasis in ESCC patients.
Collapse
Affiliation(s)
- Pan Gao
- People’s Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People’s Republic of China
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Hongtao Liu
- Laboratory for Cell Biology, College of Life Sciences of Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Zhenzhen Yang
- People’s Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People’s Republic of China
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Yiran Hui
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
- University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, Guangdong, 518106, People’s Republic of China
| | - Zhuangzhuang Shi
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Zhen Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Min Song
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Menghui Yao
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Wenfei Fan
- People’s Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Jinhua Yang
- People’s Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Yibin Hao
- People’s Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Tianli Fan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| |
Collapse
|
10
|
Zhang W, Williams TA, Bhagwath AS, Hiermann JS, Peacock CD, Watkins DN, Ding P, Park JY, Montgomery EA, Forastiere AA, Jie C, Cantarel BL, Pham TH, Wang DH. GEAMP, a novel gastroesophageal junction carcinoma cell line derived from a malignant pleural effusion. J Transl Med 2020; 100:16-26. [PMID: 31292541 PMCID: PMC6920545 DOI: 10.1038/s41374-019-0278-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
Abstract
Gastroesophageal junction (GEJ) cancer remains a clinically significant disease in Western countries due to its increasing incidence, which mirrors that of esophageal cancer, and poor prognosis. To develop novel and effective approaches for prevention, early detection, and treatment of patients with GEJ cancer, a better understanding of the mechanisms driving pathogenesis and malignant progression of this disease is required. These efforts have been limited by the small number of available cell lines and appropriate preclinical animal models for in vitro and in vivo studies. We have established and characterized a novel GEJ cancer cell line, GEAMP, derived from the malignant pleural effusion of a previously treated GEJ cancer patient. Comprehensive genetic analyses confirmed a clonal relationship between GEAMP cells and the primary tumor. Targeted next-generation sequencing identified 56 nonsynonymous alterations in 51 genes including TP53 and APC, which are commonly altered in GEJ cancer. In addition, multiple copy-number alterations were found including EGFR and K-RAS gene amplifications and loss of CDKN2A and CDKN2B. Histological examination of subcutaneous flank xenografts in nude and NOD-SCID mice showed a carcinoma with mixed squamous and glandular differentiation, suggesting GEAMP cells contain a subpopulation with multipotent potential. Finally, pharmacologic inhibition of the EGFR signaling pathway led to downregulation of key downstream kinases and inhibition of cell proliferation in vitro. Thus, GEAMP represents a valuable addition to the limited number of bona fide GEJ cancer cell lines.
Collapse
Affiliation(s)
- Wei Zhang
- Esophageal Diseases Center and Division of Hematology-Oncology, Department of Internal Medicine and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Taylor A. Williams
- Esophageal Diseases Center and Division of Hematology-Oncology, Department of Internal Medicine and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ankur S. Bhagwath
- Esophageal Diseases Center and Division of Hematology-Oncology, Department of Internal Medicine and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jared S. Hiermann
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Craig D. Peacock
- Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - D. Neil Watkins
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Peiguo Ding
- Esophageal Diseases Center and Division of Hematology-Oncology, Department of Internal Medicine and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason Y. Park
- Department of Pathology and the Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth A. Montgomery
- Division of Gastrointestinal and Liver Pathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Arlene A. Forastiere
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chunfa Jie
- Department of Biochemistry and Nutrition, Des Moines University, Des Moines, IA, USA
| | - Brandi L. Cantarel
- Bioinformatics Core Facility, Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thai H. Pham
- Esophageal Diseases Center and Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA,VA North Texas Health Care System, Dallas, TX, USA
| | - David H. Wang
- Esophageal Diseases Center and Division of Hematology-Oncology, Department of Internal Medicine and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,VA North Texas Health Care System, Dallas, TX, USA
| |
Collapse
|
11
|
Kashima H, Noma K, Ohara T, Kato T, Katsura Y, Komoto S, Sato H, Katsube R, Ninomiya T, Tazawa H, Shirakawa Y, Fujiwara T. Cancer-associated fibroblasts (CAFs) promote the lymph node metastasis of esophageal squamous cell carcinoma. Int J Cancer 2018; 144:828-840. [PMID: 30367467 DOI: 10.1002/ijc.31953] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/29/2018] [Accepted: 10/11/2018] [Indexed: 12/11/2022]
Abstract
Lymph node metastasis is a pathognomonic feature of spreading tumors, and overcoming metastasis is a challenge in attaining more favorable clinical outcomes. Esophageal cancer is an aggressive tumor for which lymph node metastasis is a strong poor prognostic factor, and the tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in particular, has been implicated in esophageal cancer progression. CAFs play a central role in the TME and have been reported to provide suitable conditions for the progression of esophageal cancer, similar to their role in other malignancies. However, little is known concerning the relevance of CAFs to the lymph node metastasis of esophageal cancer. Here, we used clinical samples of esophageal cancer to reveal that CAFs promote lymph node metastasis and subsequently verified the intercellular relationships in vitro and in vivo using an orthotopic metastatic mouse model. In the analysis of clinical samples, FAP+ CAFs were strongly associated with lymph node metastasis rather than with other prognostic factors. Furthermore, CAFs affected the ability of esophageal cancer cells to acquire metastatic phenotypes in vitro; this finding was confirmed by data from an in vivo orthotopic metastatic mouse model showing that the number of lymph node metastases increased upon injection of cocultured cancer cells and CAFs. In summary, we verified in vitro and in vivo that the accumulation of CAFs enhances the lymph node metastasis of ESCC. Our data suggest that CAF targeted therapy can reduce lymph node metastasis and improve the prognosis of patients with esophageal cancer in the future.
Collapse
Affiliation(s)
- Hajime Kashima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiaki Ohara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Pathology & Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Kato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuki Katsura
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Komoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Sato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryoichi Katsube
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Ninomiya
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasuhiro Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
12
|
Drenckhan A, Freytag M, Supuran CT, Sauter G, Izbicki JR, Gros SJ. CAIX furthers tumour progression in the hypoxic tumour microenvironment of esophageal carcinoma and is a possible therapeutic target. J Enzyme Inhib Med Chem 2018; 33:1024-1033. [PMID: 29865880 PMCID: PMC6010094 DOI: 10.1080/14756366.2018.1475369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hypoxic tumour microenvironment of solid tumours represents an important starting point for modulating progression and metastatic spread. Carbonic anhydrase IX (CAIX) is a known HIF-1α-dependent key player in maintaining cell pH conditions under hypoxia. We show that CAIX is strongly expressed in esophageal carcinoma tissues. We hypothesize that a moderate CAIX expression facilitates metastases and thereby worsens prognosis. Selective inhibition of CAIX by specific CAIX inhibitors and a CAIX knockdown effectively inhibit proliferation and migration in vitro. In the orthotopic esophageal carcinoma model, the humanized HER2 antibody trastuzumab down-regulates CAIX, possibly through CAIX’s linkage with HER2 in the hypoxic microenvironment. Our results show CAIX to be an essential part of the tumour microenvironment and a possible master regulator of tumour progression. This makes CAIX a highly effective and feasible therapeutic target for selective cancer treatment.
Collapse
Affiliation(s)
- Astrid Drenckhan
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Morton Freytag
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Claudiu T Supuran
- b Department Neurofarba , Section of Pharmaceutical Sciences, University of Florence , Florence , Italy
| | - Guido Sauter
- c Department of Pathology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Jakob R Izbicki
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Stephanie J Gros
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,d Department of Pediatric Surgery , Ûniversity Children's Hospital Basel , Basel , Switzerland
| |
Collapse
|
13
|
Liu DS, Hoefnagel SJM, Fisher OM, Krishnadath KK, Montgomery KG, Busuttil RA, Colebatch AJ, Read M, Duong CP, Phillips WA, Clemons NJ. Novel metastatic models of esophageal adenocarcinoma derived from FLO-1 cells highlight the importance of E-cadherin in cancer metastasis. Oncotarget 2018; 7:83342-83358. [PMID: 27863424 PMCID: PMC5347774 DOI: 10.18632/oncotarget.13391] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
There is currently a paucity of preclinical models available to study the metastatic process in esophageal cancer. Here we report FLO-1, and its isogenic derivative FLO-1LM, as two spontaneously metastatic cell line models of human esophageal adenocarcinoma. We show that FLO-1 has undergone epithelial-mesenchymal transition and metastasizes following subcutaneous injection in mice. FLO-1LM, derived from a FLO-1 liver metastasis, has markedly enhanced proliferative, clonogenic, anti-apoptotic, invasive, immune-tolerant and metastatic potential. Genome-wide RNAseq profiling revealed a significant enrichment of metastasis-related pathways in FLO-1LM cells. Moreover, CDH1, which encodes the adhesion molecule E-cadherin, was the most significantly downregulated gene in FLO-1LM compared to FLO-1. Consistent with this, repression of E-cadherin expression in FLO-1 cells resulted in increased metastatic activity. Importantly, reduced E-cadherin expression is commonly reported in esophageal adenocarcinoma and independently predicts poor patient survival. Collectively, these findings highlight the biological importance of E-cadherin activity in the pathogenesis of metastatic esophageal adenocarcinoma and validate the utility of FLO-1 parental and FLO-1LM cells as preclinical models of metastasis in this disease.
Collapse
Affiliation(s)
- David S Liu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sanne J M Hoefnagel
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Oliver M Fisher
- Gastroesophageal Cancer Program, St Vincent's Centre for Applied Medical Research, Darlinghurst, New South Wales, 2010, Australia
| | - Kausilia K Krishnadath
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands.,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Karen G Montgomery
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Rita A Busuttil
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia.,The University of Melbourne Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, 3010, Australia
| | - Andrew J Colebatch
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Matthew Read
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Cuong P Duong
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Wayne A Phillips
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia.,University of Melbourne Department of Surgery, St Vincent's Hospital, Fitzroy, Victoria, 3065, Australia
| | - Nicholas J Clemons
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
14
|
Superior Therapeutic Efficacy of Nanoparticle Albumin Bound Paclitaxel Over Cremophor-Bound Paclitaxel in Experimental Esophageal Adenocarcinoma. Transl Oncol 2018; 11:426-435. [PMID: 29475139 PMCID: PMC5884213 DOI: 10.1016/j.tranon.2018.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is the fastest growing cancer in the western world and the overall 5 year survival rate of EAC is below 20%. Most patients with EAC present with locally advanced or widespread metastatic disease, where current treatment is largely ineffective. Therefore, new therapeutic approaches are urgently needed. Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a novel albumin-stabilized, cremophor-free and water soluble nanoparticle formulation of paclitaxel, and the potential role of nab-paclitaxel has not been tested yet in experimental EAC. Here we tested the antiproliferative and antitumor efficacy with survival advantage of nab-paclitaxel as monotherapy and in combinations in in-vitro, and in murine subcutaneous xenograft and peritoneal metastatic survival models of human EAC. Nab-paclitaxel significantly inhibited in-vitro cell proliferation with higher in-vivo antitumour efficacy and survival benefit compared to paclitaxel or carboplatin treatments both in mono- and combination therapies. Nab-paclitaxel treatment increased expression of mitotic-spindle associated phospho-stathmin, decreased expression of proliferative markers and enhanced apoptosis. This study demonstrates that nab-paclitaxel had stronger antiproliferative and antitumor activity in experimental EAC than the current standard chemotherapeutic agents which supports the rationale for its clinical use in EAC.
Collapse
|
15
|
Zhang XF, Pan K, Weng DS, Chen CL, Wang QJ, Zhao JJ, Pan QZ, Liu Q, Jiang SS, Li YQ, Zhang HX, Xia JC. Cytotoxic T lymphocyte antigen-4 expression in esophageal carcinoma: implications for prognosis. Oncotarget 2018; 7:26670-9. [PMID: 27050369 PMCID: PMC5042006 DOI: 10.18632/oncotarget.8476] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/02/2016] [Indexed: 02/07/2023] Open
Abstract
To examine the relationship between cytotoxic T lymphocyte antigen-4 (CTLA-4) expression and esophageal carcinoma prognosis, CTLA-4 expression was immunohistochemically detected in paraffin-embedded primary tumor specimens from 158 patients with esophageal cancer. CTLA-4 was detected in the cytoplasm and cell membranes of esophageal cancer cells and in interstitial lymphocytes. In univariate analyses (log-rank), higher interstitial CTLA-4+ lymphocyte density and higher tumor CTLA-4 expression were associated with shorter overall survival (OS). After controlling for age and clinical stage, multivariate analysis (Cox) found that tumor CTLA-4 expression was an independent predictor of shorter OS (HR 2.016, P = 0.004). These results indicate that CTLA-4 expression in the tumor environment (both lymphocytes and tumor cells) is associated with poorer prognosis. In addition, CTLA-4 profiles may be useful for predicting the benefits and toxicity of CTLA-4 blockade in patients with esophageal carcinoma.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Ke Pan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Chang-Long Chen
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qi-Jing Wang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qiu-Zhong Pan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qing Liu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Shan-Shan Jiang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Yong-Qiang Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Hong-Xia Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| |
Collapse
|
16
|
Hoffmann M, Pasch S, Schamberger T, Maneck M, Möhlendick B, Schumacher S, Brockhoff G, Knoefel WT, Izbicki J, Polzer B, Stoecklein NH, Klein CA. Diagnostic pathology of early systemic cancer: ERBB2 gene amplification in single disseminated cancer cells determines patient survival in operable esophageal cancer. Int J Cancer 2017; 142:833-843. [PMID: 29044505 DOI: 10.1002/ijc.31108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/07/2017] [Accepted: 09/28/2017] [Indexed: 11/05/2022]
Abstract
Early metastatic dissemination and evolution of disseminated cancer cells (DCCs) outside the primary tumor is one reason for the failure of adjuvant therapies because it generates molecular genotypes and phenotypes different from primary tumors, which still underlie therapy decisions. Since ERBB2 amplification in esophageal DCCs but not in primary tumor cells predict outcome, we aimed to establish an assay with diagnostic reliability for single DCCs or circulating tumor cells. For this, we evaluated copy number alterations of more than 600 single DCCs from multiple cancer types to define reference regions suitable for quantification of target regions, such as ERBB2. We then compared ERBB2 quantitative PCR (qPCR) measurements with fluorescent in situ hybridization (FISH) data of various breast cancer cell lines and identified the aberration-calling threshold. The method was applied to two independent cohorts of esophageal cancer patients from Hamburg (n = 59) and Düsseldorf (n = 53). We found a high correlation between the single cell qPCR assay and the standard FISH assay (R = 0.98) and significant associations between amplification and survival for both patient cohorts (Hamburg (HH), p = 0.033; Düsseldorf (D), p = 0.052; pooled HH + D, p = 0.002) when applied to DCCs of esophageal cancer patients. Detection of a single ERBB2-amplified DCC was the most important risk factor for death from esophageal cancer (relative risk = 4.22; 95% CI = 1.91-9.32; p < 0.001). In our study, we detected ERBB2-amplified cells in 7% of patients. These patients could benefit from anti-ERBB2 targeting therapies.
Collapse
Affiliation(s)
- Martin Hoffmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Regensburg, Germany
| | - Sophie Pasch
- Chair of Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Thomas Schamberger
- Chair of Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Matthias Maneck
- Chair of Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Birte Möhlendick
- Department of General, Visceral and Pediatric Surgery, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Sarah Schumacher
- Department of General, Visceral and Pediatric Surgery, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Gero Brockhoff
- Department of Gynecology, Caritas-Hospital St. Josef, University of Regensburg, Germany
| | - Wolfram Trudo Knoefel
- Department of General, Visceral and Pediatric Surgery, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Jakob Izbicki
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Polzer
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Regensburg, Germany
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Christoph A Klein
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Regensburg, Germany.,Chair of Experimental Medicine and Therapy Research, University of Regensburg, Germany
| |
Collapse
|
17
|
Melsens E, De Vlieghere E, Descamps B, Vanhove C, De Wever O, Ceelen W, Pattyn P. Improved xenograft efficiency of esophageal adenocarcinoma cell lines through in vivo selection. Oncol Rep 2017; 38:71-81. [PMID: 28504813 PMCID: PMC5492849 DOI: 10.3892/or.2017.5640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/18/2016] [Indexed: 01/29/2023] Open
Abstract
The present study aimed to investigate the orthotopic growth potential of two generally available esophageal adenocarcinoma cell lines, OE33 and OACM5 1.C, and a third in vivo selected subpopulation, OACM5 1.C SC1. One group of mice was subcutaneously injected in the hind legs. Tumor growth was measured with calipers. Another group was injected orthotopically in the distal esophageal wall through median laparotomy. Tumor development was evaluated macroscopically and confirmed microscopically. A subset of mice was evaluated with magnetic resonance imaging (MRI) to follow tumor progression. Additionally, functional cell line characteristics were evaluated in vitro (clonogenic, collagen invasion and sphere formation assays, and protein analysis of cell-cell adhesion and cytoskeletal proteins) to better understand xenograft behavior. OE33 cells were shown to be epithelial-like, whereas OACM5 1.C and OACM5 1.C SC1 were more mesenchymal-like. The three cell lines were non-invasive into native type I collagen gels. In vivo, OE33 cells led to 63.6 and 100% tumor nodules after orthotopic (n=12) and subcutaneous (n=8) injection, respectively. Adversely, OACM5 1.C cells did not lead to tumor formation after orthotopic injection (n=6) and only 50% of subcutaneous injections led to tumor nodules (n=8). However, the newly established cell line OACM5 1.C SC1 resulted in 33% tumor formation when orthotopically injected (n=6) and in 100% tumors when injected subcutaneously (n=8). The higher xenograft rate of OACM5 1.C SC1 (P<0.05) corresponded with a higher clonogenic potential compared to its parental cell line (P<0.0001). All models showed local tumor growth without metastasis formation. In conclusion, OACM5 1.C has a poor tumor take rate at an orthotopic and ectopic site. A subpopulation obtained through in vivo selection, OACM5 1.C SC1, gives a significant higher take rate, ectopically. Furthermore, OE33 establishes orthotopic (and subcutaneous) xenografts in mice. These models can be of interest for future studies, and their slow growth rates are a challenge for therapeutic intervention.
Collapse
Affiliation(s)
- Elodie Melsens
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Piet Pattyn
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
18
|
A novel intraperitoneal metastatic xenograft mouse model for survival outcome assessment of esophageal adenocarcinoma. PLoS One 2017; 12:e0171824. [PMID: 28225784 PMCID: PMC5321464 DOI: 10.1371/journal.pone.0171824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) has become the dominant type of esophageal cancer in United States. The 5-year survival rate of EAC is below 20% and most patients present with locally advanced or widespread metastatic disease, where current treatment is largely ineffective. Therefore, new therapeutic approaches are urgently needed. Improvement of EAC patient outcome requires well-characterized animal models in which to evaluate novel therapeutics. In this study we aimed to establish a peritoneal dissemination xenograft mouse model of EAC that would support survival outcome analyses. To find the best candidate cell line from 7 human EAC cell lines of different origin named ESO26, OE33, ESO51, SK-GT-2, OE19, OACM5.1C and Flo-1 were injected intraperitoneally/subcutaneously into SCID mice. The peritoneal/xenograft tumor formation and mouse survival were compared among different groups. All cell lines injected subcutaneously formed tumors within 3 months at variable rates. All cell lines except OACM5.1C formed intraperitoneal tumors within 3 months at variable rates. Median animal survival with peritoneal dissemination was 108 days for ESO26 cells (5X106), 65 days for OE33 cells (5X106), 88 days for ESO51 cells (5X106), 76 days for SK-GT-2 cells (5X106), 55 days for OE19 cells (5X106), 45 days for OE19 cells (10X106) and 82 days for Flo-1 cells (5X106). Interestingly, only in the OE19 model all mice (7/7 for 5X106 and 5/5 for10X106) developed bloody ascites with liver metastasis after intraperitoneal injection. The median survival time of these animals was the shortest (45 days for 10X106 cells). In addition, median survival was significantly increased after paclitaxel treatment compared with the control group (57 days versus 45 days, p = 0.0034) along with a significant decrease of the relative subcutaneous tumor volume (p = 0.00011). Thus peritoneal dissemination mouse xenograft model for survival outcome assessment after intraperitoneal injection of OE19 cells will be very useful for the evaluation of cancer therapeutics.
Collapse
|
19
|
Tétreault MP. Esophageal Cancer: Insights From Mouse Models. CANCER GROWTH AND METASTASIS 2015; 8:37-46. [PMID: 26380556 PMCID: PMC4558891 DOI: 10.4137/cgm.s21218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 12/30/2022]
Abstract
Esophageal cancer is the eighth leading cause of cancer and the sixth most common cause of cancer-related death worldwide. Despite recent advances in the development of surgical techniques in combination with the use of radiotherapy and chemotherapy, the prognosis for esophageal cancer remains poor. The cellular and molecular mechanisms that drive the pathogenesis of esophageal cancer are still poorly understood. Hence, understanding these mechanisms is crucial to improving outcomes for patients with esophageal cancer. Mouse models constitute valuable tools for modeling human cancers and for the preclinical testing of therapeutic strategies in a manner not possible in human subjects. Mice are excellent models for studying human cancers because they are similar to humans at the physiological and molecular levels and because they have a shorter gestation time and life cycle. Moreover, a wide range of well-developed technologies for introducing genetic modifications into mice are currently available. In this review, we describe how different mouse models are used to study esophageal cancer.
Collapse
Affiliation(s)
- Marie-Pier Tétreault
- Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
20
|
Overholser J, Ambegaokar KH, Eze SM, Sanabria-Figueroa E, Nahta R, Bekaii-Saab T, Kaumaya PTP. Anti-Tumor Effects of Peptide Therapeutic and Peptide Vaccine Antibody Co-targeting HER-1 and HER-2 in Esophageal Cancer (EC) and HER-1 and IGF-1R in Triple-Negative Breast Cancer (TNBC). Vaccines (Basel) 2015; 3:519-43. [PMID: 26350593 PMCID: PMC4586465 DOI: 10.3390/vaccines3030519] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/29/2015] [Accepted: 06/29/2015] [Indexed: 12/19/2022] Open
Abstract
Despite the promise of targeted therapies, there remains an urgent need for effective treatment for esophageal cancer (EC) and triple-negative breast cancer (TNBC). Current FDA-approved drugs have significant problems of toxicity, safety, selectivity, efficacy and development of resistance. In this manuscript, we demonstrate that rationally designed peptide vaccines/mimics are a viable therapeutic strategy for blocking aberrant molecular signaling pathways with high affinity, specificity, potency and safety. Specifically, we postulate that novel combination treatments targeting members of the EGFR family and IGF-1R will yield significant anti-tumor effects in in vitro models of EC and TNBC possibly overcoming mechanisms of resistance. We show that the combination of HER-1 and HER-2 or HER-1 and IGF-1R peptide mimics/vaccine antibodies exhibited enhanced antitumor properties with significant inhibition of tumorigenesis in OE19 EC and MDA-MB-231 TNBC cell lines. Our work elucidates the mechanisms of HER-1/IGF-1R and HER-1/HER-2 signaling in these cancer cell lines, and the promising results support the rationale for dual targeting with HER-1 and HER-2 or IGF-1R as an improved treatment regimen for advanced therapy tailored to difference types of cancer.
Collapse
Affiliation(s)
- Jay Overholser
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Kristen Henkins Ambegaokar
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Siobhan M Eze
- Department of Pharmacology, Emory University and Winship Cancer Institute, Atlanta, GA 30322, USA.
| | - Eduardo Sanabria-Figueroa
- Molecular and Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Rita Nahta
- Department of Pharmacology, Emory University and Winship Cancer Institute, Atlanta, GA 30322, USA.
- Molecular and Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Tanios Bekaii-Saab
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Pravin T P Kaumaya
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
21
|
Hiroshima Y, Zhang Y, Zhao M, Zhang N, Murakami T, Maawy A, Mii S, Uehara F, Yamamoto M, Miwa S, Yano S, Momiyama M, Mori R, Matsuyama R, Chishima T, Tanaka K, Ichikawa Y, Bouvet M, Endo I, Hoffman RM. Tumor-Targeting Salmonella typhimurium A1-R in Combination with Trastuzumab Eradicates HER-2-Positive Cervical Cancer Cells in Patient-Derived Mouse Models. PLoS One 2015; 10:e0120358. [PMID: 26047477 PMCID: PMC4457918 DOI: 10.1371/journal.pone.0120358] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/20/2015] [Indexed: 01/26/2023] Open
Abstract
We have previously developed mouse models of HER-2-positive cervical cancer. Tumors in nude mice had histological structures similar to the original tumor and were stained by anti-HER-2 antibody in the same pattern as the patient’s cancer. We have also previously developed tumor-targeting Salmonella typhimurium A1-R and have demonstrated its efficacy against patient-derived tumor mouse models, both alone and in combination. In the current study, we determined the efficacy of S. typhimurium A1-R in combination with trastuzumab on a patient-cancer nude-mouse model of HER-2 positive cervical cancer. Mice were randomized to 5 groups and treated as follows: (1) no treatment; (2) carboplatinum (30 mg/kg, ip, weekly, 5 weeks); (3) trastuzumab (20 mg/kg, ip, weekly, 5 weeks); (4) S. typhimurium A1-R (5 × 107 CFU/body, ip, weekly, 5 weeks); (5) S. typhimurium A1-R (5 × 107 CFU/body, ip, weekly, 5 weeks) + trastuzumab (20 mg/kg, ip, weekly, 5 weeks). All regimens had significant efficacy compared to the untreated mice. The relative tumor volume of S. typhimurium A1-R + trastuzumab-treated mice was smaller compared to trastuzumab alone (p = 0.007) and S. typhimurium A1-R alone (p = 0.039). No significant body weight loss was found compared to the no treatment group except for carboplatinum-treated mice (p = 0.021). Upon histological examination, viable tumor cells were not detected, and replaced by stromal cells in the tumors treated with S. typhimurium A1-R + trastuzumab. The results of the present study suggest that S. typhimurium A1-R and trastuzumab in combination are highly effective against HER-2-expressing cervical cancer.
Collapse
Affiliation(s)
- Yukihiko Hiroshima
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yong Zhang
- AntiCancer, Inc., San Diego, California, United States of America
| | - Ming Zhao
- AntiCancer, Inc., San Diego, California, United States of America
| | - Nan Zhang
- AntiCancer, Inc., San Diego, California, United States of America
| | - Takashi Murakami
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ali Maawy
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Sumiyuki Mii
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Fuminari Uehara
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Mako Yamamoto
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Shinji Miwa
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Shuya Yano
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Masashi Momiyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryutaro Mori
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Chishima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kuniya Tanaka
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yasushi Ichikawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Robert M. Hoffman
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
22
|
Boonstra JJ, Tilanus HW, Dinjens WNM. Translational research on esophageal adenocarcinoma: from cell line to clinic. Dis Esophagus 2015; 28:90-6. [PMID: 23795680 DOI: 10.1111/dote.12095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human esophageal adenocarcinoma (EAC) cell lines have made a substantial contribution to elucidating mechanisms of carcinogenesis and drug discovery. Model research on EAC relies almost entirely on a relatively small set of established tumor cell lines because appropriate animal models are lacking. Nowadays, more than 20% of all fundamental translational research studies regarding EAC are partially or entirely based on these cell lines. The ready availability of these cell lines to investigators worldwide have resulted in more than 250 publications, including many examples of important biomedical discoveries. The high genomic similarities (but certainly not completely identical) between the EAC cell lines and their original tumors provide rational for their use. Recently, in a collaborative effort all available EAC cell lines have been verified resulting in the establishment of a reliable panel of 10 EAC cell lines. It could be expected that the value of these cell lines increases as unlimited source of tumor material because new biomedical techniques require more tumor cells and the supply of viable tumor cells is diminishing because of neoadjuvant chemo(radio)therapy of patients with EAC. Here, we review the history of the EAC cell lines and their utility in translational research and biomedical discovery.
Collapse
Affiliation(s)
- J J Boonstra
- Department of Pathology, Josephine Nefkens Institute, University Medical Center, Rotterdam, The Netherlands; Department of Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
23
|
Establishment of a Non-Invasive Semi-Quantitative Bioluminescent Imaging Method for Monitoring of an Orthotopic Esophageal Cancer Mouse Model. PLoS One 2014; 9:e114562. [PMID: 25493557 PMCID: PMC4262421 DOI: 10.1371/journal.pone.0114562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/11/2014] [Indexed: 11/19/2022] Open
Abstract
Orthotopic models of various types of tumors are widely used in anti-tumor therapeutic experiments in preclinical studies. However, there are few ways to appropriately monitor therapeutic effect in orthotopic tumor models, especially for tumors invisible from the outside. In this study we aimed to establish a non-invasive semi-quantitative bioluminescent imaging method of monitoring an orthotopic esophageal cancer mouse model. We confirmed that the TE8 esophageal cancer cell line implanted orthotopically into the abdominal esophagus of nu/nu mice (n = 5) developed not only a main tumor at the implanted site, but also local lymph node metastases and peritoneal disseminations within 6 weeks after inoculation. We established a TE8 cell line that stably expressed the firefly luciferase gene (TE8-Luc). We showed that TE8-Luc cells implanted subcutaneously into nu/nu mice (n = 5) grew over time until 5 weeks after inoculation. Tumor volume was strongly correlated with luminescent intensity emitted from the tumor, which was quantified using the IVIS imaging system. We then showed that TE8-Luc cells implanted orthotopically into the mouse abdominal esophagus (n = 8) also formed a tumor and that the luminescent intensity of such a tumor, as detected by IVIS, increased over time until 7 weeks after inoculation and was therefore likely to reflect tumor progression. We therefore propose that this orthotopic esophageal cancer model, monitored using the non-invasive semi-quantitative IVIS imaging system, will be useful for in vivo therapeutic experiments against esophageal cancer. This experimental setting is expected to contribute to the development of novel therapeutic technologies for esophageal cancer in preclinical studies.
Collapse
|
24
|
Uzunoglu FG, Koenig AM, Izbicki JR. The potential for targeting HER2 therapeutically in esophageal cancer – a grasp at straws? Expert Opin Ther Targets 2014; 18:1421-6. [DOI: 10.1517/14728222.2014.956086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
25
|
Zhang J, Jiang D, Li X, Lv J, Xie L, Zheng L, Gavine PR, Hu Q, Shi Y, Tan L, Ge D, Xu S, Li L, Zhu L, Hou Y, Wang Q. Establishment and characterization of esophageal squamous cell carcinoma patient-derived xenograft mouse models for preclinical drug discovery. J Transl Med 2014; 94:917-26. [PMID: 24999713 DOI: 10.1038/labinvest.2014.77] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 03/27/2014] [Accepted: 04/08/2014] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to establish and characterize patient-derived esophageal squamous cell carcinoma xenograft (PDECX) mice for utilization in antitumor drug discovery. A total of 96 esophageal squamous cell carcinoma (ESCC) tissues from Chinese patients were transplanted subcutaneously into immunodeficient mice. Histology, EGFR, K-ras, B-raf, and PIK3CA mutations, and HER2 gene amplifications were analyzed in both patient tumors and mouse xenograft tissues using immunohistochemistry, mutant-enriched liquid chip sequencing and fluorescence in situ hybridization assays, respectively. Furthermore, in vivo efficacy studies using five PDECX mice harboring a variety of genetic aberrations were performed using the chemotherapy agents 5-fluorouracil (5-FU) and cisplatin. Thirty-seven PDECX mouse models were successfully established in immunodeficient mice. Pathological analysis revealed similar histological architecture and degrees of differentiation between patient ESCC and xenografted tumors. No mutations were identified in EGFR, K-ras, and B-raf genes in either xenograft models or patient ESCC tissues. In contrast, PIK3CA gene mutations were detected in 12.5% (12/96) ESCC patients and 18.9% (7/37) PDECX models. Interestingly, patient ESCC tissues exhibiting HER2 overexpression or gene amplification were unable to survive in immunodeficient mice. Further analysis showed that PDECX models carrying HER2 2+ expression had no response to 5-FU/cisplatin, compared with HER2-negative models. In conclusion, a panel of PDECX mouse models, which include PIK3CA mutant and HER2-positive models, was established and characterized thus mimicking the current clinical genetic setting of esophageal carcinoma. The sensitivity of HER2-negative ESCC models to chemotherapy supports stratification approaches in the treatment of esophageal carcinoma patients and warrants further investigation of the impact of PI3KCA on treatment response.
Collapse
Affiliation(s)
- Jingchuan Zhang
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaojing Li
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jing Lv
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Liang Xie
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Li Zheng
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Paul R Gavine
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Qin Hu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yuan Shi
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lijie Tan
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Di Ge
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Songtao Xu
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Leon Li
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Lifang Zhu
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qun Wang
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Drenckhan A, Grob T, Dupree A, Dohrmann T, Mann O, Izbicki JR, Gros SJ. Esophageal carcinoma cell line with high EGFR polysomy is responsive to gefitinib. Langenbecks Arch Surg 2014; 399:879-88. [DOI: 10.1007/s00423-014-1235-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 07/16/2014] [Indexed: 10/25/2022]
|
27
|
Shiraishi K, Mimura K, Izawa S, Inoue A, Shiba S, Maruyama T, Watanabe M, Kawaguchi Y, Inoue M, Fujii H, Kono K. Lapatinib acts on gastric cancer through both antiproliferative function and augmentation of trastuzumab-mediated antibody-dependent cellular cytotoxicity. Gastric Cancer 2013. [PMID: 23187882 DOI: 10.1007/s10120-012-0219-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Trastuzumab has been recently approved for clinical use to treat HER2-expressing advanced gastric cancer, and anti-HER2-targeting therapy has become a promising option for gastric cancer. Lapatinib is a dual tyrosine kinase inhibitor targeting EGFR and HER2. The aim of the present study was to explore the utility of lapatinib for gastric cancer, with a particular focus on trastuzumab-mediated antibody-dependent cellular cytotoxicity (ADCC). METHODS Nine gastric cancer cell lines were evaluated for the effects of lapatinib on the cell-surface accumulation of HER2 and analyzed for their additional effects on trastuzumab-mediated ADCC. Also, HER2 signaling with Western blot, proliferative function with the MTT assay, and apoptosis-inducing activity with 7ADD/Annexin-V were investigated when a panel of gastric cancer cell lines was treated with lapatinib. RESULTS Lapatinib inhibited HER2 signaling and cell proliferation in the panel of gastric cancer cell lines. Lapatinib also induced the accumulation of HER2 on the cell surface, resulting in the enhancement of trastuzumab-mediated ADCC of gastric cancer. CONCLUSIONS Lapatinib exhibits inhibitory activity in gastric cancer cells, and the combination of lapatinib with trastuzumab may be a promising treatment strategy for gastric cancer patients.
Collapse
Affiliation(s)
- Kensuke Shiraishi
- First Department of Surgery, University of Yamanashi, Yamanashi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Involvement of CXCR4 chemokine receptor in metastastic HER2-positive esophageal cancer. PLoS One 2012; 7:e47287. [PMID: 23082154 PMCID: PMC3474831 DOI: 10.1371/journal.pone.0047287] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/14/2012] [Indexed: 12/20/2022] Open
Abstract
A functional linkage of the structurally unrelated receptors HER2 and CXCR4 has been suggested for breast cancer but has not been evaluated for esophageal carcinoma. The inhibition of HER2 leads to a reduction of primary tumor growth and metastases in an orthotopic model of esophageal carcinoma. The chemokine receptor CXCR4 has been implicated in metastatic dissemination of various tumors and correlates with poor survival in esophageal carcinoma. The aim of this study was to investigate a correlation between the expression levels of HER2 and CXCR4 and to evaluate the involvement of CXCR4-expression in HER2-positive esophageal carcinoma. The effects of HER2-inhibition with trastuzumab and of CXCR4-inhibition with AMD3100 on primary tumor growth, metastatic homing, and receptor expression were evaluated in vitro and in an orthotopic model of metastatic esophageal carcinoma using MRI for imaging. The clinical relevance of HER2- and CXCR4-expression was examined in esophageal carcinoma patients. A significant correlation of HER2- and CXCR4-expression in primary tumor and metastases exists in the orthotopic model. Trastuzumab and AMD3100 treatment led to a significant reduction of primary tumor growth, metastases and micrometastases. HER2-expression was significantly elevated under AMD3100 treatment in the primary tumor and particularly in the metastases. The positive correlation between HER2- and CXCR4-expression was validated in esophageal cancer patients. The correlation of CXCR4- and HER2-expression and the elevation of HER2-expression and reduction of metastases through CXCR4-inhibition suggest a possible functional linkage and a role in tumor dissemination in HER2-positive esophageal carcinoma.
Collapse
|
29
|
Drenckhan A, Kurschat N, Dohrmann T, Raabe N, Koenig AM, Reichelt U, Kaifi JT, Izbicki JR, Gros SJ. Effective inhibition of metastases and primary tumor growth with CTCE-9908 in esophageal cancer. J Surg Res 2012; 182:250-6. [PMID: 23117118 DOI: 10.1016/j.jss.2012.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/18/2012] [Accepted: 09/26/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND In spite of multimodular treatment, the therapeutic options for esophageal carcinoma are limited, and metastases remain the leading cause of tumor-related mortality. Expression of the chemokine receptor CXCR4 significantly correlates with poor survival rates in patients with esophageal carcinoma and is associated with lymph node and bone marrow metastases. The aim of this study was to evaluate the effect of the CXCR4 antagonist CTCE-9908 on metastatic homing and primary tumor growth in vitro and in vivo in an orthotopic xenograft model of esophageal cancer. MATERIALS AND METHODS OE19 cells were examined for stromal cell-derived factor 1 alpha-mediated migration under CTCE-9908 treatment. The CTCE-9908 treatment was further evaluated in an in vitro proliferation assay and orthotopic esophageal model, accompanied by magnetic resonance imaging. Tumor and metastases were immunohistochemically examined for CXCR4 expression. RESULTS CTCE-9908 has an inhibitory effect on stromal cell-derived factor 1 alpha-mediated migration and proliferation of OE19 cells. Treatment with CTCE-9908 in the orthotopic esophageal model leads to a reduction of metastatic spread and primary tumor growth. This was confirmed by magnetic resonsance imaging. Treatment with CTCE-9908 results in altered CXCR4 expression pattern exhibiting a high degree of variability. CONCLUSION CTCE-9908 effectively inhibits OE19 cell migration and proliferation in vitro, reduces metastases to lung, liver, and lymph nodes in vivo, and moreover leads to tumor growth reduction in an orthotopic model of esophageal carcinoma.
Collapse
Affiliation(s)
- Astrid Drenckhan
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wu X, Zhang J, Zhen R, Lv J, Zheng L, Su X, Zhu G, Gavine PR, Xu S, Lu S, Hou J, Liu Y, Xu C, Tan Y, Xie L, Yin X, He D, Ji Q, Hou Y, Ge D. Trastuzumab anti-tumor efficacy in patient-derived esophageal squamous cell carcinoma xenograft (PDECX) mouse models. J Transl Med 2012; 10:180. [PMID: 22935382 PMCID: PMC3485623 DOI: 10.1186/1479-5876-10-180] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/28/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Trastuzumab is currently approved for the clinical treatment of breast and gastric cancer patients with HER-2 positive tumors, but not yet for the treatment of esophageal carcinoma patients, whose tumors typically show 5 ~ 35% HER-2 gene amplification and 0 ~ 56% HER-2 protein expression. This study aimed to investigate the therapeutic efficacy of Trastuzumab in patient-derived esophageal squamous cell carcinoma xenograft (PDECX) mouse models. METHODS PDECX models were established by implanting patient esophageal squamous cell carcinoma (ESCC) tissues into immunodeficient (SCID/nude) mice. HER-2 gene copy number (GCN) and protein expression were determined in xenograft tissues and corresponding patient EC samples by FISH and IHC analysis. Trastuzumab anti-tumor efficacy was evaluated within these PDECX models (n = 8 animals/group). Furthermore, hotspot mutations of EGFR, K-ras, B-raf and PIK3CA genes were screened for in the PDECX models and their corresponding patient's ESCC tissues. Similarity between the PDECX models and their corresponding patient's ESCC tissue was confirmed by histology, morphology, HER-2 GCN and mutation. RESULTS None of the PDECX models (or their corresponding patient's ESCC tissues) harbored HER-2 gene amplification. IHC staining showed HER-2 positivity (IHC 2+) in 2 PDECX models and negativity in 3 PDECX models. Significant tumor regression was observed in the Trastuzumab-treated EC044 HER-2 positive model (IHC 2+). A second HER-2 positive (IHC 2+) model, EC039, harbored a known PIK3CA mutation and showed strong activation of the AKT signaling pathway and was insensitive to Trastuzumab treatment, but could be resensitised using a combination of Trastuzumab and AKT inhibitor AZD5363. In summary, we established 5 PDECX mouse models and demonstrated tumor regression in response to Trastuzumab treatment in a HER-2 IHC 2+ model, but resistance in a HER-2 IHC 2+/PIK3CA mutated model. CONCLUSIONS This study demonstrates Trastuzumab-induced tumor regressions in HER-2 positive tumors, and highlights PIK3CA mutation as a potential resistance mechanism to Trastuzumab treatment in pre-clinical patient-derived EC xenograft models.
Collapse
Affiliation(s)
- Xianhua Wu
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Enomoto K, Sho M, Wakatsuki K, Takayama T, Matsumoto S, Nakamura S, Akahori T, Tanaka T, Migita K, Ito M, Nakajima Y. Prognostic importance of tumour-infiltrating memory T cells in oesophageal squamous cell carcinoma. Clin Exp Immunol 2012; 168:186-91. [PMID: 22471279 DOI: 10.1111/j.1365-2249.2012.04565.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Memory T cells survive for many months and years and are critically important for host defence in humans. In tumour immunity, they have been also suggested to play a significant role in tumour progression and metastasis. However, the role of memory T cells in actual human cancer remains largely unknown. In this study, the clinical importance of tumour-infiltrating CD45RO(+) memory T cells was investigated in human oesophageal squamous cell carcinoma (OSCC). CD45RO(+) T cells were evaluated by immunohistochemistry in primary OSCC tumours from 105 patients. Patients were classified into two groups as CD45RO(+hi) or CD45RO(+lo) based on the number of cells stained positively for CD45RO. No significant difference was observed between CD45RO status and several clinicopathological prognostic factors. However, the postoperative overall and disease-free survival for CD45RO(+hi) patients was significantly better than for CD45RO(+lo) patients. Furthermore, there were significant correlations of CD45RO status in the primary tumour with postoperative lymph node and pulmonary recurrence, suggesting that memory T cells may control postoperative metastatic recurrence. Most importantly, CD45RO(+) memory T cell status has a significant prognostic value for OSCC independently of conventional tumour-node-metastasis (TNM) classification. Our study may provide a rationale for developing a novel immunotherapy in intentional induction of memory T cells for the treatment of oesophageal cancer.
Collapse
Affiliation(s)
- K Enomoto
- Department of Surgery, Nara Medical University, Nara, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gros SJ. Orthotopic models of esophageal carcinoma and their use in drug discovery. ACTA ACUST UNITED AC 2012; Chapter 14:Unit14.20. [PMID: 21898333 DOI: 10.1002/0471141755.ph1420s54] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The protocol detailed in this unit is for the establishment of an orthotopic model of human esophageal adenocarcinoma in NMRI/nu mice. The resultant tumor has high metastatic potential, spreading readily to liver, lungs, and lymph nodes. This model is useful for studying primary esophageal carcinoma, tumor biology, pathogenesis, tumor progression, metastatic homing, and the efficacy of therapeutic approaches for treating this condition. The practical use of this preclinical model for drug discovery is illustrated with data from a study on the chemotherapeutic effects of HER2-targeted therapy.
Collapse
Affiliation(s)
- Stephanie J Gros
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Trastuzumab has anti-metastatic and anti-angiogenic activity in a spontaneous metastasis xenograft model of esophageal adenocarcinoma. Cancer Lett 2011; 308:54-61. [PMID: 21570176 DOI: 10.1016/j.canlet.2011.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/15/2011] [Accepted: 04/18/2011] [Indexed: 01/02/2023]
Abstract
HER-2/neu over-expression occurs in 10-40% of patients with esophageal adenocarcinoma. Therefore, inhibitory effects of trastuzumab on proliferation, neoangiogenesis and metastatic spread of the esophageal adenocarcinoma cell line PT1590 were investigated (subcutaneous xenograft model). PT1590 revealed an amplified copy number of c-erbB2 and HER-2/neu over-expression occured in xenograft tumors and spontaneous lung metastases. PT1590 proliferation was significantly inhibited by trastuzumab in vitro. In vivo, tumor weight, volume, microvessel density and number of lung metastases decreased significantly after three weeks of treatment. These data suggest the importance of HER-2/neu for metastatic spread in esophageal adenocarcinoma and encourages clinical trials.
Collapse
|
34
|
Analysis of HER2 gene amplification and protein expression in esophageal squamous cell carcinoma. Med Oncol 2011; 29:933-40. [PMID: 21318736 DOI: 10.1007/s12032-011-9850-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Accepted: 01/27/2011] [Indexed: 12/20/2022]
Abstract
The HER2 gene, which is located on chromosomes 17, is a therapeutic target for cancer. Amplification of HER2 has been described in several tumor types. However, few studies of HER2 gene amplification and protein expression in esophageal carcinoma have been conducted. This study was to investigate the relationship between the expression of HER2/neu and the clinical characteristics, including survival rate, of esophageal squamous carcinoma. The clinical data of 145 patients admitted in Renmin Hospital of Wuhan University, from 2000 to 2005, were reviewed. The HER2 protein expression and gene status in 145 esophageal carcinomas were evaluated using immunohistochemistry and fluorescence in situ hybridization. The survival rate was calculated by the Kaplan-Meier method and the log-rank test using SPSS13.0 software. Compared to normal esophageal epithelium (23/95, 24.2%), HER2 protein was overexpressed in most esophageal squamous carcinoma tissues (60/145, 41.4%), of which 45 (31.0%) were 2+ and 15 (10.4%) were 3+, HER2 overexpression associated significantly with HER2 gene amplification. There is a correlation between the overexpression of HER2 and the differentiation of the carcinoma, the HER2 gene amplification and the differentiation of the carcinoma and the tumor stage. According to univariate analysis, there was a significant difference in survival rates when cases with and without HER-2/neu overexpression or amplification were compared. HER-2/neu amplification/overexpression may be used as an independent prognostic factor in patients with esophageal squamous cancer, and patients with HER-2/neu amplification/overexpression might be potential candidates for new adjuvant therapies that involve the use of humanized monoclonal antibodies.
Collapse
|