1
|
Cox KE, Amirfakhri S, Lwin TM, Hosseini M, Ghosh P, Obonyo M, Hoffman RM, Yazaki PJ, Bouvet M. A new locoregional mouse model of gastric cancer for identifying probes for fluorescence guided surgery. Surgery 2025; 181:109270. [PMID: 40043513 DOI: 10.1016/j.surg.2025.109270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/12/2024] [Accepted: 02/01/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND In gastric cancer, the only opportunity for a cure is with surgical resection. Fluorescence-guided surgery is an emerging field that has the potential to improve rates of R0 resections. Mouse models with patterns of disease spread that would be deemed operable in patients are required for the testing of potential fluorescence-guided surgery probes. METHODS One million cells of the human gastric cancer cell line MKN45 were suspended in 50 μL of phosphate-buffered saline and Matrigel and injected into the mouse stomach with a 29-gauge needle. After 8 to 12 weeks of tumor growth, mice were killed and laparotomy was performed to determine rates of tumor engraftment, local or distant spread, and involvement of celiac lymph nodes. For tumor labeling, mice were randomized to receive intravenous injection of an anti-CEA antibody (M5A), or IgG as a control, conjugated with the near-infrared dye IRDye800CW. Fluorescence imaging was performed using the LI-COR Pearl Imaging System 72 hours later. RESULTS Infiltrative tumors were identified in 76.5% (n = 34) of mice. Intra-abdominal or peritoneal metastases were seen in 23.5% and carcinomatosis was seen in 5.9% of mice. Celiac lymph node metastases were seen in 55.5% of mice. M5A-IR800 administration resulted in bright labeling of primary tumors and metastatic celiac lymph nodes. Hematoxylin and eosin staining demonstrated incorporation of the gastric cancer cells throughout the layers of the mouse stomach and the presence of metastatic gastric cancer cells in celiac lymph nodes. CONCLUSION This new locoregional mouse model can be used to validate additional agents for their use in fluorescence guided surgical resection of gastric cancer.
Collapse
Affiliation(s)
- Kristin E Cox
- Department of Surgery, University of California San Diego, La Jolla, CA; VA San Diego Healthcare System, La Jolla, CA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, La Jolla, CA; VA San Diego Healthcare System, La Jolla, CA
| | - Thinzar M Lwin
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego, La Jolla, CA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Marygorret Obonyo
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA; VA San Diego Healthcare System, La Jolla, CA; AntiCancer Inc, San Diego, CA
| | - Paul J Yazaki
- Department of Immunology & Theranostics, Beckman Research Institute of the City of Hope, Duarte, CA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA; VA San Diego Healthcare System, La Jolla, CA.
| |
Collapse
|
2
|
De Vleeschauwer SI, van de Ven M, Oudin A, Debusschere K, Connor K, Byrne AT, Ram D, Rhebergen AM, Raeves YD, Dahlhoff M, Dangles-Marie V, Hermans ER. OBSERVE: guidelines for the refinement of rodent cancer models. Nat Protoc 2024; 19:2571-2596. [PMID: 38992214 DOI: 10.1038/s41596-024-00998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 02/23/2024] [Indexed: 07/13/2024]
Abstract
Existing guidelines on the preparation (Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE)) and reporting (Animal Research: Reporting of In Vivo Experiments (ARRIVE)) of animal experiments do not provide a clear and standardized approach for refinement during in vivo cancer studies, resulting in the publication of generic methodological sections that poorly reflect the attempts made at accurately monitoring different pathologies. Compliance with the 3Rs guidelines has mainly focused on reduction and replacement; however, refinement has been harder to implement. The Oncology Best-practices: Signs, Endpoints and Refinements for in Vivo Experiments (OBSERVE) guidelines are the result of a European initiative supported by EurOPDX and INFRAFRONTIER, and aim to facilitate the refinement of studies using in vivo cancer models by offering robust and practical recommendations on approaches to research scientists and animal care staff. We listed cancer-specific clinical signs as a reference point and from there developed sets of guidelines for a wide variety of rodent models, including genetically engineered models and patient derived xenografts. In this Consensus Statement, we systematically and comprehensively address refinement and monitoring approaches during the design and execution of murine cancer studies. We elaborate on the appropriate preparation of tumor-initiating biologicals and the refinement of tumor-implantation methods. We describe the clinical signs to monitor associated with tumor growth, the appropriate follow-up of animals tailored to varying clinical signs and humane endpoints, and an overview of severity assessment in relation to clinical signs, implantation method and tumor characteristics. The guidelines provide oncology researchers clear and robust guidance for the refinement of in vivo cancer models.
Collapse
Affiliation(s)
| | - Marieke van de Ven
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Karlijn Debusschere
- Animal Core Facility VUB, Brussels, Belgium
- Core ARTH Animal Facilities, Medicine and Health Sciences Ghent University, Ghent, Belgium
| | - Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Doreen Ram
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Els R Hermans
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Wang C, Xie GM, Zhang LP, Yan S, Xu JL, Han YL, Luo MJ, Gong JN. High Engraftment and Metastatic Rates in Orthotopic Xenograft Models of Gastric Cancer via Direct Implantation of Tumor Cell Suspensions. Cancers (Basel) 2024; 16:759. [PMID: 38398149 PMCID: PMC10886682 DOI: 10.3390/cancers16040759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Although the implantation of intact tumor fragments is a common practice to generate orthotopic xenografts to study tumor invasion and metastasis, the direct implantation of tumor cell suspensions is necessary when prior manipulations of tumor cells are required. However, the establishment of orthotopic xenografts using tumor cell suspensions is not mature, and a comparative study directly comparing their engraftment and metastatic capabilities is lacking. It is unclear whether tumor fragments are superior to cell suspensions for successful engraftment and metastasis. In this study, we employed three GC cell lines with varying metastatic capacities to stably express firefly luciferase for monitoring tumor progression in real time. We successfully minimized the risk of cell leakage during the orthotopic injection of tumor cell suspensions without Corning Matrigel by systematically optimizing the surgical procedure, injection volume, and needle size options. Comparable high engraftment and metastatic rates between these two methods were demonstrated using MKN-45 cells with a strong metastatic ability. Importantly, our approach can adjust the rate of tumor progression flexibly and cuts the experimental timeline from 10-12 weeks (for tumor fragments) to 4-5 weeks. Collectively, we provided a highly reproducible procedure with a shortened experimental timeline and low cost for establishing orthotopic GC xenografts via the direct implantation of tumor cell suspensions.
Collapse
Affiliation(s)
- Chao Wang
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Guo-Min Xie
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Li-Ping Zhang
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Shuo Yan
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Jia-Li Xu
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Yun-Lin Han
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| | - Ming-Jie Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510080, China;
| | - Jia-Nan Gong
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; (C.W.); (G.-M.X.); (L.-P.Z.); (S.Y.); (J.-L.X.); (Y.-L.H.)
| |
Collapse
|
4
|
Zhao C, Rong Z, Ding J, Wang L, Wang B, Ding L, Meng L, Meng X, Wang F, Yang Z, Shou C, Zhu H. Targeting Claudin 18.2 Using a Highly Specific Antibody Enables Cancer Diagnosis and Guided Surgery. Mol Pharm 2022; 19:3530-3541. [PMID: 35344359 DOI: 10.1021/acs.molpharmaceut.1c00947] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Claudin 18.2 (CLDN18.2) is a new potential target for cancer therapy, especially for advanced gastric cancer (AGC). A molecular targeting probe is of importance for patient stratification and therapeutic guidance. Here, we explored an antibody-dependent molecular imaging strategy for specific detection and surgery guidance based on a CLDN18.2-specific antibody, 5C9. Two imaging probes, 124I-5C9 and Cy5.5-5C9, were synthesized. The specificity to CLDN18.2 being evidenced in the cellular experiments with control, the diagnostic utility was assessed by immunopositron emission tomography (immuno-PET) and fluorescence imaging using xenograft models. A near-infrared fluorescent II imaging probe FD1080-5C9 was designed to facilitate the comprehensive surgical removal of lesions. 124I-5C9 immuno-PET imaging clearly delineated subcutaneous CLDN18.2-positive tumors, with a peak uptake (maximum standardized uptake value; SUVmax) of 2.25 ± 0.30, whereas the highest values for the 124I-IgG and blocking groups were 0.70 ± 0.13 and 0.66 ± 0.12, respectively. Cy5.5-5C9 fluorescence imaging showed similar results. As proof of the diagnosis and guided surgery (DGS) concept, 124I-5C9 and FD1080-5C9 were simultaneously administered in orthotopic CLDN18.2-positive tumor models, facilitating the comprehensive resection of tumor tissue. Combined, 124I-5C9 and FD1080-5C9 are both promising DGS tools: the former reveals CLDN18.2 in lesions as a PET probe, and the latter can guide surgery. These results provide a utility molecular imaging strategy for specific detection and surgery guidance based on a CLDN18.2-specific antibody both in AGC and other cancers.
Collapse
Affiliation(s)
- Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Zhuona Rong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jin Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Lixin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Bing Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Lei Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Feng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Chengchao Shou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
5
|
Kiblitskaya AA, Goncharova AS, Anisimov AE, Snezhko AV, Dimitriadi SN, Maslov AA, Gevorkyan YA, Kolesnikov EN. Antitumor effect of radiation therapy on orthotopic PDX models of human esophageal adenocarcinoma. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2021. [DOI: 10.24075/brsmu.2021.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As a rule, esophageal adenocarcinoma develops in the lower esophagus. Life expectancy and survival rates depend on the cancer stage and the general health of the patient. Chemoradiotherapy is the most successful treatment approach to this type of cancer. The choice of optimal radiation doses for achieving the best possible therapeutic effect is still a challenge. The aim of this paper was to study effective radiation doses and assess response of human esophageal adenocarcinoma to radiation using a PDX model. The study was conducted in female Balb/c nude mice (n = 25). Fragments of the donor tumor were implanted into the cervical esophagus of immunodeficient mice. Effects of radiation on the obtained orthotopic xenografts were studied after each of 3 irradiation sessions (4, 6, 8, and 10 Gy in each of the experimental groups, respectively). First-passage xenografts reproduced the morphology of the donor tumor. The mean tumor volume differed significantly between the control group and the experimental groups exposed to 6, 8 or 10 Gy (р ≤ 0.01) after each irradiation session. Tumor growth delay was significant after exposure to the total dose of 18 Gy. The further radiation dose increase was ineffective. The reduction of tumor volume in the xenografts was correlated to the increase in the one-time radiation dose. The total dose over 18 Gy produced a detrimental effect on the hematopoietic system and blood biochemistry of the experimental mice.
Collapse
Affiliation(s)
- AA Kiblitskaya
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - AS Goncharova
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - AE Anisimov
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - AV Snezhko
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - SN Dimitriadi
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - AA Maslov
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - YA Gevorkyan
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| | - EN Kolesnikov
- National Medical Research Center for Oncology, Rostov-on-Don, Russia
| |
Collapse
|
6
|
Song S, Xu Y, Huo L, Zhao S, Wang R, Li Y, Scott AW, Pizzi MP, Wang Y, Fan Y, Harada K, Jin J, Ma L, Yao X, Shanbhag ND, Gan Q, Roy-Chowdhuri S, Badgwell BD, Wang Z, Wang L, Ajani JA. Patient-derived cell lines and orthotopic mouse model of peritoneal carcinomatosis recapitulate molecular and phenotypic features of human gastric adenocarcinoma. J Exp Clin Cancer Res 2021; 40:207. [PMID: 34162421 PMCID: PMC8223395 DOI: 10.1186/s13046-021-02003-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric adenocarcinoma with peritoneal carcinomatosis (PC) is therapy resistant and leads to poor survival. To study PC in depth, there is an urgent need to develop representative PC-derived cell lines and metastatic models to study molecular mechanisms of PC and for preclinical screening of new therapies. METHODS PC cell lines were developed from patient-derived PC cells. The tumorigenicity and metastatic potential were investigated by subcutaneously (PDXs) and orthotopically. Karyotyping, whole-exome sequencing, RNA-sequencing, and functional studies were performed to molecularly define the cell lines and compare genomic and phenotypic features of PDX and donor PC cells. RESULTS We established three PC cell lines (GA0518, GA0804, and GA0825) and characterized them in vitro. The doubling times were 22, 39, and 37 h for GA0518, GA0804, and GA0825, respectively. Expression of cancer stem cell markers (CD44, ALDH1, CD133 and YAP1) and activation of oncogenes varied among the cell lines. All three PC cell lines formed PDXs. Interestingly, all three PC cell lines formed tumors in the patient derived orthotopic (PDO) model and GA0518 cell line consistently produced PC in mice. Moreover, PDXs recapitulated transcriptomic and phenotypic features of the donor PC cells. Finally, these cell lines were suitable for preclinical testing of chemotherapy and target agents in vitro and in vivo. CONCLUSION We successfully established three patient-derived PC cell lines and an improved PDO model with high incidence of PC associated with malignant ascites. Thus, these cell lines and metastatic PDO model represent excellent resources for exploring metastatic mechanisms of PC in depth and for target drug screening and validation by interrogating GAC for translational studies.
Collapse
Affiliation(s)
- Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Yan Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P. R. China
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Shuangtao Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ruiping Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P. R. China
| | - Ailing W Scott
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Yibo Fan
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Namita D Shanbhag
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Qiong Gan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P. R. China
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Kang W, Maher L, Michaud M, Bae SW, Kim S, Lee HS, Im SA, Yang HK, Lee C. Development of a Novel Orthotopic Gastric Cancer Mouse Model. Biol Proced Online 2021; 23:1. [PMID: 33390162 PMCID: PMC7780388 DOI: 10.1186/s12575-020-00137-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/30/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Gastric cancer metastasis is a highly fatal disease with a five-year survival rate of less than 5%. One major obstacle in studying gastric cancer metastasis is the lack of faithful models available. The cancer xenograft mouse models are widely used to elucidate the mechanisms of cancer development and progression. Current procedures for creating cancer xenografts include both heterotopic (i.e., subcutaneous) and orthotopic transplantation methods. Compared to the heterotopic model, the orthotopic model has been shown to be the more clinically relevant design as it enables the development of cancer metastasis. Although there are several methods in use to develop the orthotopic gastric cancer model, there is not a model which uses various types of tumor materials, such as soft tissues, semi-liquid tissues, or culture derivatives, due to the technical challenges. Thus, developing the applicable orthotopic model which can utilize various tumor materials is essential. RESULTS To overcome the known limitations of the current orthotopic gastric cancer models, such as exposure of tumor fragments to the neighboring organs or only using firm tissues for the orthotopic implantation, we have developed a new method allowing for the complete insertion of soft tissue fragments or homogeneously minced tissues into the stomach submucosa layer of the immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mouse. With this completely-closed transplantation method, tumors with various types of tissue may be used to establish orthotopic gastric cancer models without the risks of exposure to nearby organs or cell leakage. This surgical procedure was highly reproducible in generating forty-eight mouse models with a surgery success rate of 96% and tumor formation of 93%. Among four orthotopic patient-derived xenograft (PDX) models that we generated in this study, we verified that the occurrence of organotropic metastasis in either the liver or peritoneal cavity was the same as that of the donor patients. CONCLUSION Here we describe a new protocol, step by step, for the establishment of orthotopic xenograft of gastric cancer. This novel technique will be able to increase the use of orthotopic models in broader applications for not only gastric cancer research but also any research related to the stomach microenvironment.
Collapse
Affiliation(s)
- Wonyoung Kang
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Leigh Maher
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Michael Michaud
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Seong-Woo Bae
- Cancer Research Institute, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Seongyeong Kim
- Cancer Research Institute, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, 03080, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, 103 Daehang-Ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA.
| |
Collapse
|
8
|
Reddavid R, Corso S, Moya-Rull D, Giordano S, Degiuli M. Patient-Derived Orthotopic Xenograft models in gastric cancer: a systematic review. Updates Surg 2020; 72:951-966. [PMID: 32253687 DOI: 10.1007/s13304-020-00751-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 03/19/2020] [Indexed: 01/23/2023]
Abstract
Patient-Derived Xenografts (PDXs) are, so far, the best preclinical model to validate targets and predictors of response to therapy. While subcutaneous implantation very rarely allows metastatic dissemination, orthotopic implantation (Patient-Derived Orthotopic Xenograft-PDOX) increases metastatic capability. Using a modified tool to analyze model validity, we performed a systematic review of Embase, PubMed, and Web of Science up to December 2018 to identify all original publications describing gastric cancer (GC) PDOXs. We identified ten studies of PDOX model validation from January 1981 to December 2018 that fulfilled the inclusion and exclusion criteria. Most models (70%) were derived from human GC cell lines rather than tissue fragments. In 90% of studies, the implantation was performed in the subserosal layer. Tumour engraftment rate ranged from 0 to 100%, despite the technique. Metastases were observed in 40% of PDOX models implanted into the subserosal layer, employing either cell suspension or cell line-derived tumour fragments. According to our modified model validity tool, half of the studies were defined as unclear because one or more validation criteria were not reported. Available GC PDOX models are not adequate according to our model validity tool. There is no demonstration that the submucosal site is more effective than the subserosal layer, and that tissue fragments are better than cell suspensions for successful engraftment and metastatic spread. Further studies should strictly employ model validity tools and large samples with orthotopic implant sites mirroring as much as possible the donor tumour characteristics.
Collapse
Affiliation(s)
- Rossella Reddavid
- Department of Oncology, Division of Surgical Oncology and Digestive Surgery, San Luigi University Hospital, University of Turin, Regione Gonzole 10, 10043, Orbassano, TO, Italy.
| | - Simona Corso
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Maurizio Degiuli
- Department of Oncology, Division of Surgical Oncology and Digestive Surgery, San Luigi University Hospital, University of Turin, Regione Gonzole 10, 10043, Orbassano, TO, Italy.
| |
Collapse
|
9
|
Orthotopic Patient-Derived Xenografts of Gastric Cancer to Decipher Drugs Effects on Cancer Stem Cells and Metastatic Dissemination. Cancers (Basel) 2019; 11:cancers11040560. [PMID: 31010193 PMCID: PMC6520896 DOI: 10.3390/cancers11040560] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer is the third leading cause of cancer mortality worldwide. Cancer stem cells (CSC) are at the origin of tumor initiation, chemoresistance, and the formation of metastases. However, there is a lack of mouse models enabling the study of the metastatic process in gastric adenocarcinoma (GC). The aims of this study were to develop original mouse models of patient-derived primary GC orthotopic xenografts (PDOX) allowing the development of distant metastases as preclinical models to study the anti-metastatic efficiency of drugs such as the phosphatidylinositol 3-kinase (PI3K) inhibitor Buparlisib (BKM120). Luciferase-encoding cells generated from primary GC were injected into the stomach wall of immunocompromised mice; gastric tumor and metastases development were followed by bioluminescence imaging. The anti-CSC properties of BKM120 were evaluated on the GC cells’ phenotype (CD44 expression) and tumorigenic properties in vitro and in vivo on BKM120-treated mice. After eight weeks, PDOX mice formed tumors in the stomach as well as distant metastases, that were enriched in CSC, in the liver, the lung, and the peritoneal cavity. BKM120 treatment significantly inhibited the CSC properties in vitro and reduced the number of distant metastases in mice. These new preclinical models offer the opportunity to study the anti-metastatic efficiency of new CSC-based therapeutic strategies.
Collapse
|
10
|
Feng HY, Zhang Y, Liu HJ, Dong X, Yang SC, Lu Q, Meng F, Chen HZ, Sun P, Fang C. Characterization of an orthotopic gastric cancer mouse model with lymph node and organ metastases using bioluminescence imaging. Oncol Lett 2018; 16:5179-5185. [PMID: 30250585 PMCID: PMC6144215 DOI: 10.3892/ol.2018.9313] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 07/16/2018] [Indexed: 12/19/2022] Open
Abstract
Lymph node (LN) metastasis of gastric cancer (GC) is the strongest prognostic indicator for this disease; however, the majority of the LN metastasis profiles of GC remain unknown, which notably hinders the therapeutic efficacy in clinic. In the present study, an orthotopic model of human GC was established for investigation of time-dependent LN metastasis patterns in mice. Luciferase-expressing NCI-N87 human GC cells were injected into the subserosa of the gastric body, resulting in a tumor formation rate of 100%. LN metastasis at four different anatomical positions in the abdomen were characterized until week 10 after tumor cell injection using sensitive bioluminescence imaging and histopathological analyses. Skip LN metastases were observed at later stages (weeks 8-10) of the experiment. Metastases in other major organs, including liver, spleen and lung, were also examined. Characterization of this orthotopic GC model and metastasis patterns in LNs and major organs should aid in the preclinical GC research regarding the metastatic mechanism and drug development.
Collapse
Affiliation(s)
- Hai-Yi Feng
- Department of General Surgery, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China.,College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Yunpeng Zhang
- Department of General Surgery, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Hai-Jun Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Xiao Dong
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Si-Cong Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Fanping Meng
- College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Peng Sun
- Department of General Surgery, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China.,Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
11
|
Busuttil RA, Liu DS, Di Costanzo N, Schröder J, Mitchell C, Boussioutas A. An orthotopic mouse model of gastric cancer invasion and metastasis. Sci Rep 2018; 8:825. [PMID: 29339747 PMCID: PMC5770387 DOI: 10.1038/s41598-017-19025-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/11/2017] [Indexed: 12/31/2022] Open
Abstract
Gastric cancer is a leading cause of cancer death worldwide, with advanced stage being correlated to the level of tumour invasion and metastasis. Current research is heavily focused on the identification and development of efficacious therapeutics targeting these fundamental hallmarks of cancer, however there are currently no animal models that mimic the invasive phenotypes observed in humans. To address this we have developed an orthotopic mouse model whereby gastric cancer cell lines are tagged with luciferase and injected into the subserosal layer of the stomach. This allows for the monitoring of primary tumour growth and metastasis in real-time as well as quantitation of the degree of tumour invasion through the stomach wall by immunohistochemistry. We have three models based on the degree of invasion and metastasis that are cell line specific: The AGS cells develop into invasive tumours by 4-weeks with no evidence of metastases, MKN45 cells are moderately metastatic with minimal invasion till week 2 and MKN28 cells are highly metastatic and fully invasive by week 1. These models have utility as a tool for testing the efficacy of anti-tumour, anti-invasive and anti-metastatic therapies in the setting of gastric cancer, which currently has poor treatment options.
Collapse
Affiliation(s)
- Rita A Busuttil
- Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - David S Liu
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Cancer Biology and Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,Department of Surgery, Austin Hospital, Heidelberg, VIC, Australia
| | - Natasha Di Costanzo
- Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Jan Schröder
- Bioinformatics Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Catherine Mitchell
- School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia.,Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Alex Boussioutas
- Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Parkville, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
12
|
Zipper-interacting protein kinase promotes epithelial-mesenchymal transition, invasion and metastasis through AKT and NF-kB signaling and is associated with metastasis and poor prognosis in gastric cancer patients. Oncotarget 2016; 6:8323-38. [PMID: 25831050 PMCID: PMC4480755 DOI: 10.18632/oncotarget.3200] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/23/2015] [Indexed: 12/21/2022] Open
Abstract
Zipper-interacting Protein Kinase (ZIPK) belongs to the death-associated protein kinase family. ZIPK has been characterized as a tumor suppressor in various tumors, including gastric cancer. On the other hand, ZIPK also promotes cell survival. In this study, both in vitro and in vivo assays indicated that ZIPK promoted cell growth, proliferation, migration, invasion, tumor formation and metastasis in nude mice. ZIPK induced epithelial-mesenchymal transition (EMT) with increasing expression of β-catenin, mesenchymal markers, Snail and Slug, and with decreasing expression of E-cadherin. Furthermore, ZIPK activated the AKT/IκB/NF-κB pathway, which can promote EMT and metastasis. Additionally, ZIPK expression was detected in human primary gastric cancer and their matched metastatic lymph node samples by immunohistochemistry. Increased expression of ZIPK in lymph node metastases was significantly associated with stage VI and abdominal organ invasion. Survival analysis revealed that patients with increased ZIPK expression in metastatic lymph nodes had poor disease-specific survival. Taken together, our study reveals that ZIPK is a pro-oncogenic factor, which promotes cancer metastasis.
Collapse
|
13
|
Hotz B, Keilholz U, Fusi A, Buhr HJ, Hotz HG. In vitro and in vivo antitumor activity of cetuximab in human gastric cancer cell lines in relation to epidermal growth factor receptor (EGFR) expression and mutational phenotype. Gastric Cancer 2012; 15:252-64. [PMID: 22011788 DOI: 10.1007/s10120-011-0102-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 09/14/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Targeting the epidermal growth factor receptor (EGFR) pathway is an important approach for a variety of tumors. This study assessed the effect of cetuximab, an anti-EGFR monoclonal antibody, on three gastric cancer cell lines with different phenotypes in vitro and in a therapeutic orthotopic murine gastric cancer model. METHODS Three human gastric cancer cell lines (AGS, MKN-45, NCI-N87) were evaluated for cell surface EGFR expression, and K-ras and BRAF mutations. In vitro, the effects of cetuximab, carboplatin, irinotecan, and docetaxel were investigated. Orthotopic tumors derived from MKN-45 and NCI-N87 were established in nude mice. After 4 weeks, the animals received cetuximab (1 mg/kg, weekly i.p.) or carboplatin (20 mg/kg, weekly i.p.), or both agents. The volume of the primary tumor and local and systemic tumor spread were determined at autopsy at 14 weeks. Tumor sections were immunostained for EGFR, as well as stained for CD31 to analyze microvessel density. RESULTS Cell surface expression of EGFR was found only in AGS and NCI-N87 cells. AGS cells displayed a codon 12 K-ras mutation, and all three cell lines were BRAF wild-type. In vitro, cetuximab significantly reduced cell viability and proliferation only in EGFR-positive/K-ras wild-type NCI-N87 cells (-48%). In vivo, cetuximab in combination with carboplatin synergistically reduced tumor volume (-75%), dissemination (-63%), and vascularization (-47%) in NCI-N87 xenografts. Tumors derived from EGFR-negative MKN-45 cells were unaffected by cetuximab. CONCLUSIONS Cetuximab is effective in K-ras wild-type, EGFR-expressing gastric cancer cell lines and xenografts. In vivo, the combination of cetuximab with carboplatin displayed synergistic antitumor activity.
Collapse
Affiliation(s)
- Birgit Hotz
- Department of Surgery, Charité-School of Medicine, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | | | | | | | | |
Collapse
|
14
|
Zheng MJ, Wang J, Chen YW, Xu L, Xue DD, Fu W, Zhang YF, Du Q, Zhao Y, Ling LJ, Ding Q, Liu XA, Zha XM, Zheng W, Xia TS, Wang S. A novel mouse model of gastric cancer with human gastric microenvironment. Cancer Lett 2012; 325:108-15. [PMID: 22750094 DOI: 10.1016/j.canlet.2012.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/21/2012] [Accepted: 06/24/2012] [Indexed: 01/14/2023]
Abstract
Mouse models play an irreplaceable role in the in vivo research of human gastric cancer. In this study, we developed a novel human Gastric tissue-derived Orthotopic and Metastatic (GOM) mouse model of human gastric cancer, in which the human normal gastric tissues were implanted subcutaneously into immunodeficient mice to create a human gastric microenvironment. Then, human gastric cancer cells were injected into the implants. GOM model could mimic the interactions between human gastric microenvironment and human gastric cancer cells, which help exhibit the real characteristics of tumor cells, and finally mimic the clinical-like tumor proliferation and metastases of human beings.
Collapse
Affiliation(s)
- Ming-Jie Zheng
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Li Y, Li B, Xiang CP, Zhang Y, Li YY, Wu XL. Characterization of gastric cancer models from different cell lines orthotopically constructed using improved implantation techniques. World J Gastroenterol 2012; 18:136-43. [PMID: 22253519 PMCID: PMC3257440 DOI: 10.3748/wjg.v18.i2.136] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 06/21/2011] [Accepted: 06/28/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop orthotopic gastric cancer mouse models from different cell lines and characterize the tumor features to assist further in preclinical trials and clinical treatment strategies.
METHODS: Human gastric cancer SGC-7901 and BGC-823 cell suspensions were injected subcutaneously into nude mice to develop solid tumors, and tumor tissue pieces were then implanted under the serous coat of the stomach. An autopsy was performed on all animals of the SGC-7901 and BGC-823 models to observe the primary tumor growth and metastases using pathological and immunohistochemical methods.
RESULTS: Both models showed large tumors in situ resulting in pressure and infiltration of the adjacent organs. The gastric cavity became smaller, along with stenosis of the cardia or pylorus. There were biological and statistical differences between the two models. The metastasis rate in involved organs (lymph nodes, kidney, spleen, testis) was significantly higher in the BGC-823 model compared to the SGC-7901 model (P < 0.05 or P < 0.01). The median survival of the BGC-823 model was shorter than that of SGC-7901 (23 d vs 84 d, P < 0.05). Histopathologically, the primary tumor and metastatic lesions of the two models showed obvious atypia and mucus in the cytoplasm. Compared with the SGC-7901 model, BGC-823 appeared more poorly differentiated (absence of adenoid structure), had a smaller volume, and richer capillary structure. Immunohistochemical staining revealed cytokeratin 20 and epithelial membrane antigen expression was positive in the SGC-7901 tumors, while negative in BGC-823 ones.
CONCLUSION: Models using the SGC-7901 and BGC-823 cell lines were established which could function in gastric cancer research on carcinogenesis mechanism and drug discovery. The two models showed different tumor behavior and the latter was more malignant than the former.
Collapse
|
16
|
Abstract
Gastric cancer is the second most common cause of cancer-related mortality (9.7% of the total) worldwide. Gastric carcinogenesis is a multiple-step process that involves multiple factors, such as bacteria, immune response and host factors. Animal models play a crucial role in the research of the biological behavior, diagnosis and treatment of gastric cancer. In this article we will review current advances in the development of animal models of gastric cancer in terms of microorganism-induced models, chemical carcinogen-induced models, tumor cell implantation, genetically modified models and in silico models.
Collapse
|
17
|
Li Y, Li B, Zhang Y, Xiang CP, Li YY, Wu XL. Serial observations on an orthotopic gastric cancer model constructed using improved implantation technique. World J Gastroenterol 2011; 17:1442-7. [PMID: 21472102 PMCID: PMC3070017 DOI: 10.3748/wjg.v17.i11.1442] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/30/2010] [Accepted: 01/06/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a gastric cancer nude-mouse model with improved orthotopic implantation and investigate its biological characteristics at different time points.
METHODS: Human gastric cancer SGC-7901 cell suspensions were injected subcutaneously into a nude mouse to develop solid tumors, and the tumor tissue pieces were implanted under the serous coat. The nude mice were then euthanized in group every two weeks to observe the primary tumor growth and metastases.
RESULTS: Within 2-4 wk, there were no obvious changes about the primary tumor in stomach. At the sixth week, the primary tumor began to grow fast, resulting in incrassation of the gastric wall and stenosis of the gastric cavity, and metastases into the liver and lymph nodes were detected. The tumor, which compressed the adjacent organs, gradually became bigger and bigger followed by stenosis or vanishment of the gastric cavity from 8 to 12 wk. There were massive metastases, and the rate of metastasis was 58% in lymph nodes, 78% in liver, 39% in kidney, and 81% in peritoneum or septum.
CONCLUSION: A gastric cancer model is established, which can simulate the clinical tumor behavior and provide experimental carrier for clinical trials of gastric cancer treatment.
Collapse
|
18
|
Kim KB, Yang JY, Kwack SJ, Park KL, Kim HS, Ryu DH, Kim YJ, Hwang GS, Lee BM. Toxicometabolomics of urinary biomarkers for human gastric cancer in a mouse model. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2010; 73:1420-1430. [PMID: 20954069 DOI: 10.1080/15287394.2010.511545] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Toxicometabolomics of urinary biomarkers for human gastric cancer in a mouse model was investigated using (1)H-nuclear magnetic resonance (NMR) spectroscopy. A human gastric adenocarcinoma cell line (1 × 10(7) cells/ml) was grafted onto the skin of the back of intact male BALB/c-nu/nu mice. After the xenografted tumors developed, urine was collected and analyzed for endogenous metabolites. Global profiling combined with principal components analysis (PCA), partial least squares-discriminant analysis (PLS-DA), and orthogonal projections to latent squares-discriminant analysis (OPLS-DA) showed distinct separation of clusters between control and tumor-bearing mice. Targeted profiling revealed significant changes in trimethylamine oxide (TMAO), 3-indoxylsulfate, hippurate, and citrate levels in mice carrying human gastric cancer cells compared to normal mice. The levels of TMAO (0.41-fold) and hippurate (0.26-fold) in tumor-bearing mice were significantly decreased, whereas the levels of 3-indoxylsulfate (3.39-fold), 2-oxoglutarate (2.32-fold), and citrate (1.9-fold) were significantly increased in urine samples of tumor-bearing mice. Data suggest that TMAO, hippurate, 3-indoxylsulfate, 2-oxoglutarate, and citrate may serve as useful urinary biomarkers for gastric tumorigenesis in a mouse model.
Collapse
Affiliation(s)
- Kyu-Bong Kim
- Department of Pharmaceutical Engineering, Inje University, Obang-dong, Gimhae, Gyungnam, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|