Your browser doesn't support javascript.
loading
Senescent cancer cell vaccines induce cytotoxic T cell responses targeting primary tumors and disseminated tumor cells.
Liu, Yue; Pagacz, Joanna; Wolfgeher, Donald J; Bromerg, Kenneth D; Gorman, Jacob V; Kron, Stephen J.
Afiliação
  • Liu Y; Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA.
  • Pagacz J; Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA.
  • Wolfgeher DJ; Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA.
  • Bromerg KD; Oncology Discovery, AbbVie, North Chicago, Illinois, USA.
  • Gorman JV; Oncology Discovery, AbbVie, North Chicago, Illinois, USA.
  • Kron SJ; Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA skron@uchicago.edu.
J Immunother Cancer ; 11(2)2023 02.
Article em En | MEDLINE | ID: mdl-36792123
ABSTRACT

BACKGROUND:

Immune tolerance contributes to resistance to conventional cancer therapies such as radiation. Radiotherapy induces immunogenic cell death, releasing a burst of tumor antigens, but this appears insufficient to stimulate an effective antitumor immune response. Radiation also increases infiltration of cytotoxic T lymphocytes (CTLs), but their effector function is short lived. Although CTL exhaustion may be at fault, combining immune checkpoint blockade with radiation is insufficient to restore CTL function in most patients. An alternative model is that antigen presentation is the limiting factor, suggesting a defect in dendritic cell (DC) function.

METHODS:

Building on our prior work showing that cancer cells treated with radiation in the presence of the poly(ADP-ribose) polymerase-1 inhibitor veliparib undergo immunogenic senescence, we reexamined senescent cells (SnCs) as preventative or therapeutic cancer vaccines. SnCs formed in vitro were cocultured with splenocytes and evaluated by scRNA-seq to examine immunogenicity. Immature bone-marrow-derived DCs cocultured with SnCs were examined for maturation and activation by flow cytometry and T cell proliferation assays. Viable SnCs or SnC-activated DCs were injected subcutaneously, and vaccine effects were evaluated by analysis of immune response, prevention of tumor engraftment, regression of established tumors and/or potentiation of immunotherapy or radiotherapy.

RESULTS:

Murine CT26 colon carcinoma or 4T1 mammary carcinoma cells treated with radiation and veliparib form SnCs that promote DC maturation and activation in vitro, leading to efficient, STING-dependent CTL priming. Injecting mice with SnCs induces antigen-specific CTLs and confers protection from tumor engraftment. Injecting immunogenic SnCs into tumor-bearing mice increases inflammation with activated CTLs, suppresses tumor growth, potentiates checkpoint blockade, enhances radiotherapy and blocks colonization by disseminated tumor cells. Addressing the concern that reinjecting tumor cells into patients may be impractical, DCs activated with SnCs in vitro were similarly effective to SnCs in suppressing established tumors and blocking metastases.

CONCLUSIONS:

Therapeutic vaccines based on senescent tumor cells and/or SnC-activated DCs have the potential to improve genotoxic and immune therapies and limit recurrence or metastasis.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Carcinoma / Neoplasias do Colo / Vacinas Anticâncer Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Carcinoma / Neoplasias do Colo / Vacinas Anticâncer Idioma: En Ano de publicação: 2023 Tipo de documento: Article