Your browser doesn't support javascript.
loading
Group A Streptococcal Collagen-like Protein 1 Restricts Tumor Growth in Murine Pancreatic Adenocarcinoma and Inhibits Cancer-Promoting Neutrophil Extracellular Traps.
Henderson, Emily A; Ivey, Abby; Choi, Soo; Santiago, Stell; McNitt, Dudley; Liu, Tracy W; Lukomski, Slawomir; Boone, Brian A.
Affiliation
  • Henderson EA; Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV.
  • Ivey A; West Virginia University Cancer Institute, West Virginia University, Morgantown, WV.
  • Choi S; Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV.
  • Santiago S; Department of Pathology, West Virginia University, Morgantown, WV.
  • McNitt D; Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV.
  • Liu TW; Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV.
  • Lukomski S; West Virginia University Cancer Institute, West Virginia University, Morgantown, WV.
  • Boone BA; Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV.
bioRxiv ; 2024 Jan 19.
Article de En | MEDLINE | ID: mdl-38293049
ABSTRACT
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer associated with an immunosuppressive environment. Neutrophil extracellular traps (NETs) were initially described in the context of infection but have more recently been implicated in contributing to the tolerogenic immune response in PDAC. Thus, NETs are an attractive target for new therapeutic strategies. Group A Streptococcus (GAS) has developed defensive strategies to inhibit NETs. In the present work, we propose utilizing intra-tumoral GAS injection to stimulate anti-tumor activity by inhibiting cancer-promoting NETs. Injection of three different M-type GAS strains reduced subcutaneous pancreatic tumor volume compared to control in two different murine PDAC models. Limitation of tumor growth was dependent on streptococcal collagen-like protein 1 (Scl1), as isogenic mutant strain devoid of Scl1 did not reduce tumor size. We further show that Scl1 plays a role in localizing GAS to the tumor site, thereby limiting the systemic spread of bacteria and off-target effects. While mice did elicit a humoral immune response to GAS antigens, tested sera were negative toward Scl1 antigen following intra-tumoral treatment with Scl1-expressing GAS. M1 GAS inhibited NET formation when co-cultured with neutrophils while Scl1-devoid mutant strain did not. Recombinant Scl1 protein inhibited NETs ex vivo in a dose-dependent manner by suppressing myeloperoxidase activity. Altogether, we demonstrate that intra-tumoral GAS injections reduce PDAC growth, which is facilitated by Scl1, in part through inhibition of cancer promoting NETs. This work offers a novel strategy by which NETs can be targeted through Scl1 protein and potentiates its use as a cancer therapeutic.
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Langue: En Journal: BioRxiv Année: 2024 Type de document: Article Pays de publication: EEUU / ESTADOS UNIDOS / ESTADOS UNIDOS DA AMERICA / EUA / UNITED STATES / UNITED STATES OF AMERICA / US / USA

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Langue: En Journal: BioRxiv Année: 2024 Type de document: Article Pays de publication: EEUU / ESTADOS UNIDOS / ESTADOS UNIDOS DA AMERICA / EUA / UNITED STATES / UNITED STATES OF AMERICA / US / USA