8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      AAV‐mediated gene transfer of DNase I in the liver of mice with colorectal cancer reduces liver metastasis and restores local innate and adaptive immune response

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Liver metastasis is a major cause of colorectal cancer‐related death. Although DNase I displays antimetastatic activity by inhibition of NETs, its clinical use is limited due to its short biologic half‐life. AAV‐mediated gene expression of DNase I is a novel therapeutic strategy that could reduce the development of liver metastases through modulation of innate and adaptive tumor immunity.

          Abstract

          Liver metastasis is the main cause of colorectal cancer (CRC)‐related death. Neutrophil extracellular traps (NETs) play important roles in CRC progression. Deoxyribonuclease I (DNase I) has been shown to alter NET function by cleaving DNA strands comprising the NET backbone. Moreover, DNase I displays high antimetastatic activity in multiple tumor models. To circumvent long‐term daily administrations of recombinant DNase I, we have developed an adeno‐associated virus (AAV) gene therapy vector to specifically express DNase I in the liver. In this study, we demonstrate AAV‐mediated DNase I liver gene transfer following a single intravenous injection suppresses the development of liver metastases in a mouse model of CRC liver metastasis. Increased levels of neutrophils and NET formation in tumors are associated with poor prognosis in many patients with advanced cancers. Neutrophil infiltration and NET formation were inhibited in tumor tissues with AAV‐DNase I treatment. This approach restored local immune responses at the tumor site by increasing the percentage of CD8 + T cells while keeping CD4 + T cells similar between AAV‐DNase I and AAV‐null treatments. Our data suggest that AAV‐mediated DNase I liver gene transfer is a safe and effective modality to inhibit metastasis and represents a novel therapeutic strategy for CRC.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: not found

          Cancer statistics, 2019

          Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data, available through 2015, were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data, available through 2016, were collected by the National Center for Health Statistics. In 2019, 1,762,450 new cancer cases and 606,880 cancer deaths are projected to occur in the United States. Over the past decade of data, the cancer incidence rate (2006-2015) was stable in women and declined by approximately 2% per year in men, whereas the cancer death rate (2007-2016) declined annually by 1.4% and 1.8%, respectively. The overall cancer death rate dropped continuously from 1991 to 2016 by a total of 27%, translating into approximately 2,629,200 fewer cancer deaths than would have been expected if death rates had remained at their peak. Although the racial gap in cancer mortality is slowly narrowing, socioeconomic inequalities are widening, with the most notable gaps for the most preventable cancers. For example, compared with the most affluent counties, mortality rates in the poorest counties were 2-fold higher for cervical cancer and 40% higher for male lung and liver cancers during 2012-2016. Some states are home to both the wealthiest and the poorest counties, suggesting the opportunity for more equitable dissemination of effective cancer prevention, early detection, and treatment strategies. A broader application of existing cancer control knowledge with an emphasis on disadvantaged groups would undoubtedly accelerate progress against cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Neutrophil extracellular traps kill bacteria.

            Neutrophils engulf and kill bacteria when their antimicrobial granules fuse with the phagosome. Here, we describe that, upon activation, neutrophils release granule proteins and chromatin that together form extracellular fibers that bind Gram-positive and -negative bacteria. These neutrophil extracellular traps (NETs) degrade virulence factors and kill bacteria. NETs are abundant in vivo in experimental dysentery and spontaneous human appendicitis, two examples of acute inflammation. NETs appear to be a form of innate response that binds microorganisms, prevents them from spreading, and ensures a high local concentration of antimicrobial agents to degrade virulence factors and kill bacteria.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Accessories to the crime: functions of cells recruited to the tumor microenvironment.

              Mutationally corrupted cancer (stem) cells are the driving force of tumor development and progression. Yet, these transformed cells cannot do it alone. Assemblages of ostensibly normal tissue and bone marrow-derived (stromal) cells are recruited to constitute tumorigenic microenvironments. Most of the hallmarks of cancer are enabled and sustained to varying degrees through contributions from repertoires of stromal cell types and distinctive subcell types. Their contributory functions to hallmark capabilities are increasingly well understood, as are the reciprocal communications with neoplastic cancer cells that mediate their recruitment, activation, programming, and persistence. This enhanced understanding presents interesting new targets for anticancer therapy. Copyright © 2012 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Allan.Tsung@osumc.edu
                Journal
                Mol Oncol
                Mol Oncol
                10.1002/(ISSN)1878-0261
                MOL2
                Molecular Oncology
                John Wiley and Sons Inc. (Hoboken )
                1574-7891
                1878-0261
                05 September 2020
                November 2020
                : 14
                : 11 ( doiID: 10.1002/mol2.v14.11 )
                : 2920-2935
                Affiliations
                [ 1 ] Division of Surgical Oncology Department of Surgery The Ohio State University Wexner Medical Center Columbus OH USA
                [ 2 ] Department of Gastroenterology Tongji Medical College Tongji Hospital Huazhong University of Science and Technology Wuhan China
                [ 3 ] Department of Pediatrics Tongji Medical College Tongji Hospital Huazhong University of Science and Technology Wuhan China
                [ 4 ] Human Microbiology Institute New York NY USA
                [ 5 ] CLS‐Therapeutics New York NY USA
                [ 6 ] Molecular Neurogenetics Unit Harvard Medical School Massachusetts General Hospital Charlestown MA USA
                [ 7 ] M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences Moscow Russia
                Author notes
                [*] [* ] Correspondence

                A. Tsung, Division of Surgical Oncology, Department of Surgery, N924 Doan Hall, 410 West 10th Ave, Columbus, OH 43210, USA

                Fax: +1 614‐293‐3465

                Tel: +1 614‐293‐8304

                E‐mail: Allan.Tsung@ 123456osumc.edu

                Author information
                https://orcid.org/0000-0002-5674-3554
                Article
                MOL212787
                10.1002/1878-0261.12787
                7607180
                32813937
                4de8bfdc-2d06-4671-bfef-74523de57727
                © 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 04 May 2020
                : 20 July 2020
                : 18 August 2020
                Page count
                Figures: 5, Tables: 0, Pages: 16, Words: 8591
                Funding
                Funded by: National Natural Science Foundation of China , open-funder-registry 10.13039/501100001809;
                Award ID: 81700515
                Funded by: Foundation for the National Institutes of Health , open-funder-registry 10.13039/100000009;
                Award ID: R01‐CA214865‐02
                Award ID: R01‐GM95566
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                November 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.9.3 mode:remove_FC converted:03.11.2020

                Oncology & Radiotherapy
                adeno‐associated virus,deoxyribonuclease i,metastatic colorectal cancer,neutrophil extracellular traps

                Comments

                Comment on this article