1
|
Wang X, Fang Y, Liang W, Wong CC, Qin H, Gao Y, Liang M, Song L, Zhang Y, Fan M, Liu C, Lau HCH, Xu L, Li X, Song W, Wang J, Wang N, Yang T, Mo M, Zhang X, Fang J, Liao B, Sung JJY, Yu J. Fusobacterium nucleatum facilitates anti-PD-1 therapy in microsatellite stable colorectal cancer. Cancer Cell 2024; 42:1729-1746.e8. [PMID: 39303724 DOI: 10.1016/j.ccell.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/23/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Microsatellite stable (MSS) colorectal cancers (CRCs) are often resistant to anti-programmed death-1 (PD-1) therapy. Here, we show that a CRC pathogen, Fusobacterium nucleatum (Fn), paradoxically sensitizes MSS CRC to anti-PD-1. Fecal microbiota transplantation (FMT) from patients with Fn-high MSS CRC to germ-free mice bearing MSS CRC confers sensitivity to anti-PD-1 compared to FMT from Fn-low counterparts. Single Fn administration also potentiates anti-PD-1 efficacy in murine allografts and CD34+-humanized mice bearing MSS CRC. Mechanistically, we demonstrate that intratumoral Fn generates abundant butyric acid, which inhibits histone deacetylase (HDAC) 3/8 in CD8+ T cells, inducing Tbx21 promoter H3K27 acetylation and expression. TBX21 transcriptionally represses PD-1, alleviating CD8+ T cell exhaustion and promoting effector function. Supporting this notion, knockout of a butyric acid-producing gene in Fn abolishes its anti-PD-1 boosting effect. In patients with MSS CRC, high intratumoral Fn predicts favorable response to anti-PD-1 therapy, indicating Fn as a potential biomarker of immunotherapy response in MSS CRC.
Collapse
Affiliation(s)
- Xueliang Wang
- Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Fang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Liang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chi Chun Wong
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Huanlong Qin
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yaohui Gao
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Meinong Liang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Song
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongxin Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Miao Fan
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuanfa Liu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Lixia Xu
- Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxing Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wu Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junlin Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Na Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Yang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengmiao Mo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Zhang
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Liao
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Joseph J Y Sung
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jun Yu
- Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
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
|
Deng F, Li Y, Yang B, Sang R, Deng W, Kansara M, Lin F, Thavaneswaran S, Thomas DM, Goldys EM. Topological barrier to Cas12a activation by circular DNA nanostructures facilitates autocatalysis and transforms DNA/RNA sensing. Nat Commun 2024; 15:1818. [PMID: 38443394 PMCID: PMC10914725 DOI: 10.1038/s41467-024-46001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
Control of CRISPR/Cas12a trans-cleavage is crucial for biosensor development. Here, we show that small circular DNA nanostructures which partially match guide RNA sequences only minimally activate Cas12a ribonucleoproteins. However, linearizing these structures restores activation. Building on this finding, an Autocatalytic Cas12a Circular DNA Amplification Reaction (AutoCAR) system is established which allows a single nucleic acid target to activate multiple ribonucleoproteins, and greatly increases the achievable reporter cleavage rates per target. A rate-equation-based model explains the observed near-exponential rate trends. Autocatalysis is also sustained with DNA nanostructures modified with fluorophore-quencher pairs achieving 1 aM level (<1 copy/μL) DNA detection (106 times improvement), without additional amplification, within 15 min, at room temperature. The detection range is tuneable, spanning 3 to 11 orders of magnitude. We demonstrate 1 aM level detection of SNP mutations in circulating tumor DNA from blood plasma, genomic DNA (H. Pylori) and RNA (SARS-CoV-2) without reverse transcription as well as colorimetric lateral flow tests of cancer mutations with ~100 aM sensitivity.
Collapse
Affiliation(s)
- Fei Deng
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Yi Li
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Biyao Yang
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Rui Sang
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Maya Kansara
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2011, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2011, Australia
- Omico, Australian Genomic Cancer Medicine Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Frank Lin
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2011, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Subotheni Thavaneswaran
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2011, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2011, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2011, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2011, Australia
- Omico, Australian Genomic Cancer Medicine Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
4
|
Uneyama M, Chambers JK, Fujii T, Nakashima K, Uchida K. Establishment and characterization of a novel cell line and xenotransplant mouse model derived from feline colorectal adenocarcinoma. Vet Pathol 2024; 61:190-200. [PMID: 37515543 DOI: 10.1177/03009858231189858] [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] [Indexed: 07/31/2023]
Abstract
Colorectal adenocarcinoma is an aggressive malignant tumor in cats that frequently metastasizes to the lymph nodes and/or distant organs. However, research on feline colorectal adenocarcinoma is limited, and experimental models have not been established. A novel cell line, FeLeco-G7, was established from the lymph node of a 12-year-old spayed female Maine Coon cat with metastatic colorectal adenocarcinoma. FeLeco-G7 cells were polygonal with abundant cytoplasm and adherent growth. The population-doubling time was approximately 28.3 hours, and the mean number of chromosomes was 37.6±0.1 per cell (ranging between 32 and 41). Consistent with the original tumor, FeLeco-G7 cells were immunopositive for cytokeratin (CK) 20 and CDX2, and immunonegative for CD10 and CK7. Nuclear accumulation of β-catenin was rarely observed. Mutation analysis suggested TP53 gene alterations. A subcutaneous injection of FeLeco-G7 cells into immunodeficient mice resulted in the formation of a mass at the injection site without the development of metastatic lesions. An orthotopic (intrarectal) transplantation of FeLeco-G7 cells caused cachexia and diffuse involvement of the rectal mucosa in one of the 3 mice and the formation of masses around the rectum in the other 2 mice. Metastases to the regional lymph nodes and lungs were detected in three of the 3 and one of the 3 mice, respectively. The histological findings and immunohistochemical features of these masses were similar to those of the original tumor. These results suggest that FeLeco-G7 cells and the orthotopically transplanted mouse model are valuable tools for further molecular and therapeutic research on feline colorectal adenocarcinoma.
Collapse
Affiliation(s)
| | | | | | - Ko Nakashima
- Japan Small Animal Medical Center, Tokorozawa, Japan
| | | |
Collapse
|
5
|
Zhong Z, Zhang H, Nan K, Zhong J, Wu Q, Lu L, Yue Y, Zhang Z, Guo M, Wang Z, Xia J, Xing Y, Fu Y, Yu B, Zhou W, Sun X, Shen Y, Chen W, Zhang J, Zhang J, Ma D, Chu Y, Liu R, Miao C. Fasting-Mimicking Diet Drives Antitumor Immunity against Colorectal Cancer by Reducing IgA-Producing Cells. Cancer Res 2023; 83:3529-3543. [PMID: 37602826 PMCID: PMC10618736 DOI: 10.1158/0008-5472.can-23-0323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/05/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
As a safe, feasible, and inexpensive dietary intervention, fasting-mimicking diet (FMD) exhibits excellent antitumor efficacy by regulating metabolism and boosting antitumor immunity. A better understanding of the specific mechanisms underlying the immunoregulatory functions of FMD could help improve and expand the clinical application of FMD-mediated immunotherapeutic strategies. In this study, we aimed to elucidate the role of metabolic reprogramming induced by FMD in activation of antitumor immunity against colorectal cancer. Single-cell RNA sequencing analysis of intratumoral immune cells revealed that tumor-infiltrating IgA+ B cells were significantly reduced by FMD treatment, leading to the activation of antitumor immunity and tumor regression in murine colorectal cancer models. Mechanistically, FMD delayed tumor growth by repressing B-cell class switching to IgA. Therefore, FMD-induced reduction of IgA+ B cells overcame the suppression of CD8+ T cells. The immunoregulatory and antitumor effects of FMD intervention were reversed by IgA+ B-cell transfer. Moreover, FMD boosted fatty acid oxidation (FAO) to trigger RUNX3 acetylation, thus inactivating Cα gene transcription and IgA class switching. IgA+ B-cell expansion was also impeded in patients placed on FMD, while B-cell expression of carnitine palmitoyl transferase 1A (CPT1A), the rate-limiting enzyme of FAO, was increased. Furthermore, CPT1A expression was negatively correlated with both IgA+ B cells and IgA secretion within colorectal cancer. Together, these results highlight that FMD holds great promise for treating colorectal cancer. Furthermore, the degree of IgA+ B cell infiltration and FAO-associated metabolic status are potential biomarkers for evaluating FMD efficacy. SIGNIFICANCE Metabolic reprogramming of B cells induced by fasting-mimicking diet suppresses IgA class switching and production to activate antitumor immunity and inhibit tumor growth. See related commentary by Bush and Perry, p. 3493.
Collapse
Affiliation(s)
- Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Qichao Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Lihong Lu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhenyu Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Miaomiao Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhiqiang Wang
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jie Xia
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun Xing
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ying Fu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xingfeng Sun
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| |
Collapse
|
6
|
Kim JK, Wu C, Del Latto M, Gao Y, Choi SH, Kierstead M, Gabriel Sauvé CE, Firat C, Perez AC, Sillanpaa J, Chen CT, Lawrence KE, Paty PB, Barriga FM, Wilkinson JE, Shia J, Sawyers CL, Lowe SW, García-Aguilar J, Romesser PB, Smith JJ. An immunocompetent rectal cancer model to study radiation therapy. CELL REPORTS METHODS 2022; 2:100353. [PMID: 36590695 PMCID: PMC9795330 DOI: 10.1016/j.crmeth.2022.100353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/18/2022] [Accepted: 08/31/2022] [Indexed: 11/24/2022]
Abstract
We describe a mouse model of rectal cancer (RC) involving rapid tumor organoid engraftment via orthotopic transplantation in an immunocompetent setting. This approach uses simple mechanical disruption to allow engraftment, avoiding the use of dextran sulfate sodium. The resulting RC tumors invaded from the mucosal surface and metastasized to distant organs. Histologically, the tumors closely resemble human RC and mirror remodeling of the tumor microenvironment in response to radiation. This murine RC model thus recapitulates key aspects of human RC pathogenesis and presents an accessible approach for more physiologically accurate, preclinical efficacy studies.
Collapse
Affiliation(s)
- Jin K. Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Del Latto
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yajing Gao
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Seo-Hyun Choi
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Kierstead
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Canan Firat
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Almudena Chaves Perez
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jussi Sillanpaa
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chin-Tung Chen
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kayla E. Lawrence
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philip B. Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Francisco M. Barriga
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John E. Wilkinson
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles L. Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julio García-Aguilar
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul B. Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - J. Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
7
|
Small Animal In Situ Drug Delivery Effects via Transdermal Microneedles Array versus Intravenous Injection: A Pilot Observation Based on Photoacoustic Tomography. Pharmaceutics 2022; 14:pharmaceutics14122689. [PMID: 36559183 PMCID: PMC9788625 DOI: 10.3390/pharmaceutics14122689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Intravenous injection is a rapid, low-cost, and direct method that is commonly used to deliver multifarious biotherapeutics and vaccines. However, intravenous injection often causes trauma or tissue injury that requires professional operation. Transdermal drug delivery overcomes the aforementioned defects, and the microneedles (MNs) array is one of the most promising transdermal drug delivery platforms. Timely, precise, and non-invasive monitoring and evaluation of the effects of MNs in transdermal administration is significant to the research of drug efficiency response to specific diseases. In this sense, photoacoustic computed tomography (PACT), which provides wavelength-selective and deep-penetrating optical contrast, could be a promising imaging tool for in situ evaluation of the treatment effects. In this work, we propose the use of PACT to non-invasively assess the effects of real-time drug delivery in glioma tumors through transdermal administration with degradable indocyanine green-loaded hyaluronic acid MNs (ICG-HA-MNs). The outcome is systematically and quantitatively compared with that via intravenous injection. It is found that the photoacoustic signals of ICG in the tumor site express a faster elevation and shorter duration time in the intravenous injection group; by contrast, the photoacoustic signals demonstrate a lower intensity but prolonged duration time in the MNs group. The observed phenomenon indicates faster response but shorter drug duration for intravenous injection, which is in contrast with the lower loading but prolonged performance for transdermal drug delivery with MNs. These results exhibit good consistency with the earlier, common-sense findings reported from other aspects, confirming that PACT can serve as a potential imaging tool to precisely, non-invasively, and quickly evaluate in situ drug delivery effects and provide constructive guidance for the design and fabrication of microneedles.
Collapse
|
8
|
Recent advances in the development of transplanted colorectal cancer mouse models. Transl Res 2022; 249:128-143. [PMID: 35850446 DOI: 10.1016/j.trsl.2022.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/13/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022]
Abstract
Despite progress in prevention and treatment, colorectal cancer (CRC) remains the third most common malignancy worldwide and the second most common cause of cancer death in 2020. To evaluate various characteristics of human CRC, a variety of mouse models have been established. Transplant mouse models have distinct advantages in studying the clinical behavior and therapeutic progress of CRC. Host, xenograft, and transplantation routes are the basis of transplant mouse models. As the effects of the tumor microenvironment and the systemic environment on cancer cells are gradually revealed, 3 key elements of transplanted CRC mouse models have been revolutionized. This has led to the development of humanized mice, patient-derived xenografts, and orthotopic transplants that reflect the human systemic environment, patient's tumor of origin, and tumor growth microenvironments in immunodeficient mice, respectively. These milestone events have allowed for great progress in tumor biology and the treatment of CRC. This article reviews the evolution of these events and points out their strengths and weaknesses as innovative and useful preclinical tools to study CRC progression and metastasis and to exploit novel treatment schedules by establishing a testing platform. This review article depicts the optimal transplanted CRC mouse models and emphasizes the significance of surgical models in the study of CRC behavior and treatment response.
Collapse
|
9
|
Li C, Li T, Niu K, Xiao Z, Huang J, Pan X, Sun Y, Wang Y, Ma D, Xie P, Shuai X, Meng X. Mild phototherapy mediated by manganese dioxide-loaded mesoporous polydopamine enhances immunotherapy against colorectal cancer. Biomater Sci 2022; 10:3647-3656. [PMID: 35670464 DOI: 10.1039/d2bm00505k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the main challenges in applying the immune checkpoint blockade to treat colorectal cancer (CRC) is the immunosuppressive tumor microenvironment. Owing to its excellent cancer cell killing ability and immune activation, mild photothermal therapy (PTT) has shown bright promise to sensitize tumors to immune checkpoint inhibition through turning the immunologically "cold" tumors into "hot" ones. Herein, a mild photothermal effect-assisted theragnostic nanodrug (MnO2@MPDA-PEG NPs) is developed by incorporating MnO2 into PEGylated-mesoporous polydopamine nanoparticles (MPDA-PEG NPs). The presence of PEG endows the theragnostic nanodrug with high biostability. After accumulation in colorectal tumor, the theragnostic nanodrug responds to the tumor microenvironment, leading to the simultaneous release of Mn2+ which serves as a magnetic resonance imaging (MRI) contrast agent for tumor imaging. The released Mn2+ could also promote mild photothermal treatment-induced immune response, including the maturation of BMDC cells. In vivo antitumor studies on a CT26 model demonstrate that MnO2@MPDA-PEG NPs could be a promising dual-imaging theragnostic nanodrug to potentiate the systemic antitumor immunities.
Collapse
Affiliation(s)
- Caiying Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Tan Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Kexin Niu
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Zecong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jing Huang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Ximin Pan
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Yi Sun
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Yongchen Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Decai Ma
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| |
Collapse
|
10
|
Yi B, Cheng H, Wyczechowska D, Yu Q, Li L, Ochoa AC, Riker AI, Xi Y. Sulindac Modulates the Response of Proficient MMR Colorectal Cancer to Anti-PD-L1 Immunotherapy. Mol Cancer Ther 2021; 20:1295-1304. [PMID: 33879557 PMCID: PMC8295201 DOI: 10.1158/1535-7163.mct-20-0934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/26/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has been widely used to treat different human cancers, particularly advanced solid tumors. However, clinical studies have reported that ICI immunotherapy benefits only ∼15% of patients with colorectal cancer, specifically those with tumors characterized by microsatellite instability (MSI), a molecular marker of defective DNA mismatch repair (dMMR). For the majority of patients with colorectal cancer who carry proficient MMR (pMMR), ICIs have shown little clinical benefit. In this study, we examined the efficacy of sulindac to enhance the response of pMMR colorectal cancer to anti-PD-L1 immunotherapy. We utilized a CT26 syngeneic mouse tumor model to compare the inhibitory effects of PD-L1 antibody (Ab), sulindac, and their combination on pMMR colorectal cancer tumor growth. We found that mice treated with combination therapy showed a significant reduction in tumor volume, along with increased infiltration of CD8+ T lymphocytes in the tumor tissues. We also demonstrated that sulindac could downregulate PD-L1 by blocking NF-κB signaling, which in turn led to a decrease in exosomal PD-L1. Notably, PD-L1 Ab can be bound and consumed by exosomal PD-L1 in the blood circulation. Therefore, in combination therapy, sulindac downregulating PD-L1 leads to increased availability of PD-L1 Ab, which potentially improves the overall efficacy of anti-PD-L1 therapy. We also show that low-dose sulindac does not appear to have a systemic inhibitory effect on prostaglandin E2 (PGE2). In conclusion, our findings provide unique insights into the mechanism of action and efficacy for sulindac as an immunomodulatory agent in combination with anti-PD-L1 therapy for the treatment of pMMR colorectal cancer.
Collapse
Affiliation(s)
- Bin Yi
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Hao Cheng
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Dorota Wyczechowska
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Qingzhao Yu
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Li Li
- Ochsner Clinical School, University of Queensland, and Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, Louisiana
| | - Augusto C Ochoa
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Adam I Riker
- Geaton and JoAnn DeCesaris Cancer Institute, Anne Arundel Medical Center, Luminis Health, Annapolis, Maryland
| | - Yaguang Xi
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
11
|
Falvo E, Damiani V, Conti G, Boschi F, Messana K, Giacomini P, Milella M, De Laurenzi V, Morea V, Sala G, Fracasso G, Ceci P. High activity and low toxicity of a novel CD71-targeting nanotherapeutic named The-0504 on preclinical models of several human aggressive tumors. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:63. [PMID: 33568214 PMCID: PMC7877078 DOI: 10.1186/s13046-021-01851-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Background Ferritin receptor (CD71) is an example of a very attractive cancer target, since it is highly expressed in virtually all tumor types, including metastatic loci. However, this target can be considered to be inaccessible to conventional target therapies, due to its presence in many healthy tissues. Here, we describe the preclinical evaluation of a tumor proteases-activatable human ferritin (HFt)-based drug carrier (The-0504) that is able to selectively deliver the wide-spectrum topoisomerase I inhibitor Genz-644282 to CD71-expressing tumors, preventing the limiting toxic effects associated with CD71-targeting therapies. Methods CD71 expression was evaluated using flow cytometry and immunohistochemistry techniques. The-0504 antiproliferative activity towards several cancer cell lines was assessed in vitro. The-0504 antitumor efficacy and survival benefit were evaluated in different human tumors, which had been grown either as xenografts or patient-derived xenografts in mice. The-0504 toxicology profile was investigated in multiple-cycle repeat-dose study in rodents. Results In vitro studies indicate that The-0504 is highly specific for CD71 expressing cells, and that there is a relationship between CD71 levels and The-0504 anticancer activity. In vivo treatments with The-0504 showed a remarkable efficacy, eradicating several human tumors of very diverse and aggressive histotypes, such as pancreas, liver and colorectal carcinomas, and triple-negative breast cancer. Conclusions Durable disease-free survival, persistent antitumor responses after discontinuation of treatment and favorable toxicology profile make The-0504 an ideal candidate for clinical development as a novel, CD71-targeted, low-toxicity alternative to chemotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01851-8.
Collapse
Affiliation(s)
- Elisabetta Falvo
- CNR - National Research Council of Italy, Institute of Molecular Biology and Pathology, Rome, Italy.
| | - Verena Damiani
- Center for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Giamaica Conti
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Katia Messana
- IRCCS Regina Elena National Cancer Institute, Oncogenomics and Epigenetics, Rome, Italy
| | - Patrizio Giacomini
- IRCCS Regina Elena National Cancer Institute, Oncogenomics and Epigenetics, Rome, Italy
| | - Michele Milella
- Oncologia Medica, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
| | - Vincenzo De Laurenzi
- Center for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Veronica Morea
- CNR - National Research Council of Italy, Institute of Molecular Biology and Pathology, Rome, Italy
| | - Gianluca Sala
- Center for Advanced Studies and Technology (CAST), Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona, Verona, Italy.
| | - Pierpaolo Ceci
- CNR - National Research Council of Italy, Institute of Molecular Biology and Pathology, Rome, Italy.,Thena Biotech, Latina, Italy
| |
Collapse
|
12
|
Lu W, Yu W, He J, Liu W, Yang J, Lin X, Zhang Y, Wang X, Jiang W, Luo J, Zhang Q, Yang H, Peng S, Yi Z, Ren S, Chen J, Siwko S, Nussinov R, Cheng F, Zhang H, Liu M. Reprogramming immunosuppressive myeloid cells facilitates immunotherapy for colorectal cancer. EMBO Mol Med 2021; 13:e12798. [PMID: 33283987 PMCID: PMC7799360 DOI: 10.15252/emmm.202012798] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint blockade (ICB) has a limited effect on colorectal cancer, underlining the requirement of co-targeting the complementary mechanisms. Here, we identified prostaglandin E2 (PGE2 ) receptor 4 (EP4) as the master regulator of immunosuppressive myeloid cells (IMCs), which are the major driver of resistance to ICB therapy. PGE2 -bound EP4 promotes the differentiation of immunosuppressive M2 macrophages and myeloid-derived suppressor cells (MDSCs) and reduces the expansion of immunostimulated M1 macrophages. To explore the immunotherapeutic role of EP4 signaling, we developed a novel and selective EP4 antagonist TP-16. TP-16 effectively blocked the function of IMCs and enhanced cytotoxic T-cell-mediated tumor elimination in vivo. Cell co-culture experiments revealed that TP-16 promoted T-cell proliferation, which was impaired by tumor-derived CD11b+ myeloid cells. Notably, TP-16 and anti-PD-1 combination therapy significantly impeded tumor progression and prolonged mice survival. We further demonstrated that TP-16 increased responsiveness to anti-PD-1 therapy in an IMC-related spontaneous colorectal cancer mouse model. In summary, this study demonstrates that inhibition of EP4-expressing IMCs may offer a potential strategy for enhancing the efficacy of immunotherapy for colorectal cancer.
Collapse
Affiliation(s)
- Weiqiang Lu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Weiwei Yu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Jiacheng He
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Wenjuan Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Junjie Yang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Xianhua Lin
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Yuanjin Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Wenhao Jiang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Jian Luo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Shihong Peng
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Shancheng Ren
- Department of UrologyChanghai HospitalSecond Military UniversityShanghaiChina
| | - Jing Chen
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Stefan Siwko
- Department of Molecular and Cellular MedicineInstitute of Biosciences and TechnologyTexas A&M University Health Science CenterHoustonTXUSA
| | - Ruth Nussinov
- Cancer and Inflammation ProgramLeidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research Sponsored by the National Cancer InstituteFrederickMDUSA
- Department of Human Molecular Genetics and BiochemistrySackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research InstituteCleveland ClinicClevelandOHUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOHUSA
- Case Comprehensive Cancer CenterCase Western Reserve University School of MedicineClevelandOHUSA
| | - Hankun Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| |
Collapse
|
13
|
Jelski W, Mroczko B. Biochemical Markers of Colorectal Cancer - Present and Future. Cancer Manag Res 2020; 12:4789-4797. [PMID: 32606968 PMCID: PMC7319530 DOI: 10.2147/cmar.s253369] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
According to a report by the National Cancer Institute, colorectal cancer (CRC) is one of the most common types of cancer worldwide. CRC is often recognized too late for successful therapy. Tumor markers have been sought for a number of years to detect the transformation of malignant cells at the earliest possible stage. They are usually proteins associated with a malignancy and might be clinically useful in patients with cancer. Several classical markers have been used to recognize colorectal cancer, including carcinoembryonic antigen (CEA), carbohydrate antigen (CA 19.9), tissue polypeptide specific antigen (TPS) and tumor-associated glycoprotein-72 (TAG-72). None of these tests, however, have excellent diagnostic accuracy. Recent studies have been conducted on the use of hematopoietic growth factors (HGFs) and various enzymes in the diagnosis and prognosis of colorectal cancer. These include macrophage-colony stimulating factor (M-CSF) and granulocyte-macrophage-colony stimulating factor (GM-CSF), interleukin-3, interleukin-6 and enzymes (alcohol dehydrogenase and lysosomal exoglycosidases). Significantly, most cancer deaths are not caused by the primary tumor itself but by its spread. Analysis of circulating cancer cells (CTCs), ie, factors responsible for metastasis, may be a source of information useful in the treatment of patients with colorectal cancer. Currently available markers have significant limitations.
Collapse
Affiliation(s)
- Wojciech Jelski
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland.,Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
14
|
Bürtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: Past, present and future perspectives. World J Gastroenterol 2020; 26:1394-1426. [PMID: 32308343 PMCID: PMC7152519 DOI: 10.3748/wjg.v26.i13.1394] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common diagnosed malignancy among both sexes in the United States as well as in the European Union. While the incidence and mortality rates in western, high developed countries are declining, reflecting the success of screening programs and improved treatment regimen, a rise of the overall global CRC burden can be observed due to lifestyle changes paralleling an increasing human development index. Despite a growing insight into the biology of CRC and many therapeutic improvements in the recent decades, preclinical in vivo models are still indispensable for the development of new treatment approaches. Since the development of carcinogen-induced rodent models for CRC more than 80 years ago, a plethora of animal models has been established to study colon cancer biology. Despite tenuous invasiveness and metastatic behavior, these models are useful for chemoprevention studies and to evaluate colitis-related carcinogenesis. Genetically engineered mouse models (GEMM) mirror the pathogenesis of sporadic as well as inherited CRC depending on the specific molecular pathways activated or inhibited. Although the vast majority of CRC GEMM lack invasiveness, metastasis and tumor heterogeneity, they still have proven useful for examination of the tumor microenvironment as well as systemic immune responses; thus, supporting development of new therapeutic avenues. Induction of metastatic disease by orthotopic injection of CRC cell lines is possible, but the so generated models lack genetic diversity and the number of suited cell lines is very limited. Patient-derived xenografts, in contrast, maintain the pathological and molecular characteristics of the individual patient’s CRC after subcutaneous implantation into immunodeficient mice and are therefore most reliable for preclinical drug development – even in comparison to GEMM or cell line-based analyses. However, subcutaneous patient-derived xenograft models are less suitable for studying most aspects of the tumor microenvironment and anti-tumoral immune responses. The authors review the distinct mouse models of CRC with an emphasis on their clinical relevance and shed light on the latest developments in the field of preclinical CRC models.
Collapse
Affiliation(s)
- Florian Bürtin
- Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Christina S Mullins
- Department of Thoracic Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, Rostock 18057, Germany
| |
Collapse
|
15
|
Xu Y, Zhang L, Wang Q, Zheng M. Comparison of Different Colorectal Cancer With Liver Metastases Models Using Six Colorectal Cancer Cell Lines. Pathol Oncol Res 2020; 26:2177-2183. [PMID: 32172478 DOI: 10.1007/s12253-020-00805-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
At present, modeling methods of colorectal cancer with liver metastases have significant limitations. Here, we established orthotopic and ectopic hepatic metastases models using six colorectal cancer cell lines to choose an ideal animal model for studying colorectal cancer growth and liver metastases. Luciferin-expressing six colorectal cancer cell lines were used to induce animal models of colorectal cancer with liver metastases by intra-splenic injection or implantation of tumor tissue in the caecum. Tumors growth and metastatic events were observed by bioluminescence imaging. In orthotopic transplantation group, six cell lines all had taken rates of 100% for orthotopic tumors but showed variations in rates of growth. HCT-116 cell developed the 50% liver metastases. However, the ectopic transplantation group achieved higher liver metastatic rate, with the highest frequencies for HCT116 cell (90%) and SW620 cell (77.8%). Furthermore, the time to develop liver metastases and survival rates of bearing-tumor mice were shorter than orthotopic transplantation group. Additionally, six colorectal cancer cell lines resulted in more lymph node metastases in orthotopic transplantation group, whereas produced widespread peritoneal seeding in ectopic transplantation group. Bioluminescence imaging and pathological findings confirmed the growth and metastatic characteristics of tumors. Two animal models of colorectal cancer using six cell lines showed highly variations in rates of growth, survival rates of bearing-tumor mice and frequencies of metastases. The study provides useful information for the establishment of clinically relevant colorectal cancer with liver metastases animal models.
Collapse
Affiliation(s)
- Yuting Xu
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China.
| | - Lin Zhang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Maojin Zheng
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| |
Collapse
|
16
|
Oliveira RC, Abrantes AM, Tralhão JG, Botelho MF. The role of mouse models in colorectal cancer research-The need and the importance of the orthotopic models. Animal Model Exp Med 2020; 3:1-8. [PMID: 32318654 PMCID: PMC7167241 DOI: 10.1002/ame2.12102] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/06/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer is a worldwide health burden, with high incidence and mortality, especially in the advanced stages of the disease. Preclinical models are very important and valuable to discover and validate early and specific biomarkers as well as new therapeutic targets. In order to accomplish that, the animal models must replicate the clinical evolution of the disease in all of its phases. In this article, we review the existent mouse models, with their strengths and weaknesses in the replication of human cancer disease progression, with major focus on orthotopic models.
Collapse
Affiliation(s)
- Rui C. Oliveira
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Pathology DepartmentUniversity Hospital (CHUC)CoimbraPortugal
| | - Ana Margarida Abrantes
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
| | - José Guilherme Tralhão
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
- Surgery A DepartmentFaculty of MedicineUniversity Hospital (CHUC)CoimbraPortugal
| | - Maria Filomena Botelho
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
| |
Collapse
|
17
|
An In Vivo Mouse Model of Pelvic Recurrence of Human Colorectal Cancer. Sci Rep 2019; 9:19630. [PMID: 31873140 PMCID: PMC6928073 DOI: 10.1038/s41598-019-56152-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/05/2019] [Indexed: 11/10/2022] Open
Abstract
Pelvic recurrence of colorectal cancer is a crucial problem because radical surgery can lead to excessive invasion. Novel therapeutic strategies are required instead of surgery. However, there are few suitable models because of the difficulty in transplanting and observing tumors in the pelvis. We have established an appropriate injection site suitable for the establishment of colorectal cancer pelvic recurrence that allows for the observation of tumor growth. DLD-1 cells stably expressing luciferase (DLD-1 clone#1-Luc) were inoculated into various points of female BALB/c nude mice and the engrafted cells were analyzed with an imaging system employing bioluminescent signals and computed tomography. Weekly analysis with the imaging system showed that a triangular area defined by the vagina, the anus, and the ischial spine was suitable for the engraftment of pelvic tumors. The imaging system was able to detect the engrafted tumor 7 days after the inoculation of cells. Weight loss was observed in our model, and overall survival was 21–42 days. Tumor involvement of adjacent organs was detected histopathologically, as is the case in the clinical situation. These findings suggest that this model is valid for evaluations of the therapeutic effects of novel treatments under development. It is hoped that this model will be used in preclinical research.
Collapse
|
18
|
Hunter P. Illuminating human disease: The potential of in vivo imaging for preclinical research and diagnostics. EMBO Rep 2019; 20:e49195. [PMID: 31523923 PMCID: PMC6776895 DOI: 10.15252/embr.201949195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
New in vivo imaging technologies, including optical methods to observe biological processes in real time, show great promise for preclinical research and diagnostics of human diseases.
Collapse
|
19
|
Plummer R, Papageorge M, Ciomek N, Liu T, Yoo J. Myofibroblasts Enhance Tumor Growth in a Novel Mouse Model of Colorectal Cancer. J Surg Res 2019; 244:374-381. [PMID: 31325658 DOI: 10.1016/j.jss.2019.06.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/21/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Communication between colorectal cancer and stromal cells alters the tumor microenvironment to regulate locoregional disease and cancer progression. However, colon cancer-stromal cell interactions are difficult to study in vivo. Limitations of existing animal models include the use of immunocompromised mice, the inability to genetically modify a cell population in a single organ system, or a lack of anatomic context. Our goal was to develop a novel mouse model of colorectal cancer that is capable of studying tumor-stromal cell interactions in the native colon of immune-competent mice. METHODS Primary mouse myofibroblasts were isolated from the colon of C57BL/6 mice and were grown in cell culture. Genetically defined (ApcΔ/Δ; Kras G12D/+; Trp53Δ/Δ) primary mouse colon cancer cells were suspended in serum-free media (20 μL) at varying concentrations (5 × 103 to 4 × 104 cells) either alone or in combination with syngeneic myofibroblasts (2 × 105 cells). After isoflurane anesthesia, a colonoscopy was performed on immune-competent 8- to 10-week-old C57BL/6 mice with endoscopic microinjection of the cell suspension into the submucosal space of the colon wall utilizing a small animal colonoscope. Surveillance endoscopy was used to assess for tumor growth, along with histologic analysis. Tumor size is presented on a grading system based on tumor diameter relative to colon circumference. RESULTS A total of 33 mice were injected with a survival rate of 88% (29/33). Endoscopic microinjection of colorectal cancer cells resulted in dose-dependent tumor growth in the distal mouse colon that could be assessed endoscopically without animal sacrifice. Growth curves varied depending on the concentration of injected colorectal cancer cells, with no growth at the lowest concentration of injected cells (5 × 103 cells), progressive growth over 4 wk using 1-2 × 104 cells, while the highest colorectal cancer cell concentration (4 × 104 cells) led to larger tumors at week 1 followed by a steady decline in tumor growth over the 4-wk time period. Combined microinjection of 2 × 104 colorectal cancer cells with 2 × 105 myofibroblasts resulted in much larger tumors that persisted over the 4-wk time period and which were composed primarily of colorectal cancer cells. Immunofluorescence microscopy after coinjection of colorectal cancer cells with green fluorescent protein positive myofibroblasts confirmed that the injected myofibroblasts are present and remain viable over the 4-wk time period. CONCLUSIONS Endoscopic submucosal microinjection of primary mouse colorectal cancer cells is feasible and leads to reliable and reproducible short-term growth of colon tumors in immune-competent mice. Coinjection of primary mouse colorectal cancer cells with syngeneic myofibroblasts leads to enhanced tumor growth. Coimplantation of colorectal cancer cells with syngeneic myofibroblasts provides a novel platform to study tumor-stromal interactions in the native colon of immune-competent mice.
Collapse
Affiliation(s)
- Robert Plummer
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Marianna Papageorge
- Department of Surgery, Yale University School of Medicine, Yale New Haven Hospital, New Haven, Connecticut
| | - Natalie Ciomek
- Department of Pathology, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Tiegang Liu
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - James Yoo
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
20
|
Paulson B, Lee S, Kim Y, Moon Y, Kim JK. Miniaturized omnidirectional flexible side-view endoscope for rapid monitoring of thin tubular biostructures. BIOMEDICAL OPTICS EXPRESS 2019; 10:2264-2274. [PMID: 31149372 PMCID: PMC6524575 DOI: 10.1364/boe.10.002264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 06/09/2023]
Abstract
Endoscopic imaging allows longitudinal observation of epithelial pathologies in tubular organs throughout the body. However, the imaging and optical diagnosis of tubular biostructures such as small animal models and small pediatric organs require appropriately miniaturized devices. A miniaturized catadioptric flexible side-view endoscope is proposed with omnidirectional field of view (FOV) in the transverse direction and sub-mm-scale feature resolution. The FOV in the longitudinal direction is 50°. Images are unwrapped and stitched together to form composite images of the target by two different algorithms, revealing a composite FOV of more than 3.5 cm × 360°. The endoscope is well suited for minimally invasive rapid monitoring of thin tubular organs in pediatric patients, as well as for imaging of small animal disease models at near-cellular resolution.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - SangHwa Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Youngkyu Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Youngjin Moon
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| |
Collapse
|
21
|
Paulson B, Kim IH, Namgoong JM, Kim YG, Lee S, Moon Y, Shin DM, Choo MS, Kim JK. Longitudinal micro-endoscopic monitoring of high-success intramucosal xenografts for mouse models of colorectal cancer. Int J Med Sci 2019; 16:1453-1460. [PMID: 31673236 PMCID: PMC6818213 DOI: 10.7150/ijms.35666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/02/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently lethal forms of cancer. Intramucosal injection allows development of better mouse models of CRC, as orthotopic xenografts allow development of adenocarcinoma in the submucosa of the mouse colon wall. In this paper, a method of orthotopic injection is monitored longitudinally using cellular-resolution real-time in vivo fluorescence microendoscopy, following the injection of three different cell lines: 3T3-GFP to confirm immunosuppression and HCT116-RFP cells to model CRC. Adenoma formation is first observable after 7 to 10 days, and by use of 33 G needles a tumor induction rate of greater than 85% is documented. An additional experiment on the injection of rapamycin reveals drug efficacy and localization between 24 and 48 hours, and suggests the promise of real-time cellular-resolution fluorescence micro-endoscopy for developing longitudinal therapy regimes in mural models of CRC.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Ick Hee Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27101, USA
| | - Jung-Man Namgoong
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Young Gyu Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Sanghwa Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Youngjin Moon
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Dong-Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Myung-Soo Choo
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| |
Collapse
|
22
|
Felton J, Cheng K, Shang AC, Hu S, Larabee SM, Drachenberg CB, Raufman JP. Two sides to colon cancer: mice mimic human anatomical region disparity in colon cancer development and progression. ACTA ACUST UNITED AC 2018; 4. [PMID: 31742233 PMCID: PMC6860924 DOI: 10.20517/2394-4722.2018.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aim: Strong evidence reveals important differences between cancers in the proximal vs. distal colon. Animal models of metastatic colon cancer are available but with varying degrees of reproducibility and several important limitations. We explored whether there were regional differences in the location of murine colon cancers and assessed the utility of murine models to explore the biological basis for such differences. Methods: We re-analyzed data from our previous studies to assess the regional distribution of murine colon cancer. In survival surgery experiments, we injected HT-29 human colon cancer cells into the wall of the cecum or distal colon of Nu(NCr)-Foxn1nu or NOD.Cg-PrkdcscidIl2rgTim1Wji/SzJ mice and compared the development of primary tumors and metastases. Results: Within 7–17 weeks after intramural cecal injection of HT-29 cells, eight mice failed to develop solid primary tumors or metastases. In contrast, within four weeks after cell injection into the distal colon, 13 mice developed metastases - 12 mice developed subcutaneous metastases; of these, four developed liver metastases and one developed both liver and lung metastases. One mouse developed liver metastases only. Histological examination confirmed these lesions were adenocarcinomas. Conclusion: Our findings reveal the preferential growth of murine colon neoplasia and invasive human orthotopic xenografts in the distal mouse colon. The new approach of injecting cells into the distal colon wall results in a pattern of colon cancer development that closely mimics the progression of metastatic colon cancer in humans. This novel model of colon neoplasia has great potential for exploring anatomical differences in colon cancer and testing novel therapeutics.
Collapse
Affiliation(s)
- Jessica Felton
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kunrong Cheng
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aaron C Shang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shien Hu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shannon M Larabee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jean-Pierre Raufman
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|