Your browser doesn't support javascript.
loading
Impaired circulating myeloid CD1c+ dendritic cell function in human glioblastoma is restored by p38 inhibition - implications for the next generation of DC vaccines.
Adhikaree, Jason; Franks, Hester Ann; Televantos, Constantinos; Vaghela, Poonam; Kaur, Aanchal Preet; Walker, David; Schmitz, Marc; Jackson, Andrew Mark; Patel, Poulam Manubhai.
Afiliación
  • Adhikaree J; Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK.
  • Franks HA; Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK.
  • Televantos C; Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK.
  • Vaghela P; Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK.
  • Kaur AP; Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK.
  • Walker D; Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK.
  • Schmitz M; Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany.
  • Jackson AM; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
  • Patel PM; German Cancer Consortium (DKTK), Dresden, Germany.
Oncoimmunology ; 8(7): 1593803, 2019.
Article en En | MEDLINE | ID: mdl-31143512
ABSTRACT
Current treatments for glioblastoma (GBM) have limited efficacy and significant morbidity and therefore new strategies are urgently needed. Dendritic cells have the power to create anti-tumor immune responses. The greater potency of circulating dendritic cells (DC) over laboratory-generated monocyte-derived DC makes them exciting new immunotherapeutic candidates. To determine the immune status of GBM patients we initially investigated the frequency and function of circulating DC subsets. Furthermore, we tested the therapeutic potential of inhibiting the p38 mitogen-activated protein kinase pathway (p38i) in circulating DC to overcome DC dysfunction. GBM patients (n = 16) had significantly reduced numbers of the major myeloid circulating dendritic cell (cDC2) and plasmacytoid DC vs healthy controls; 1736 vs 4975 (p = 0.028) and 893 vs 2287 cells/mL (P = <0.001) respectively. This inversely correlated with dexamethasone (Dex) dose in a log-linear model, and disease status. Patients' cDC2 were immature with impaired interleukin (IL)-12 secretion, reduced IL-12IL-10 ratio, and low HLA-DR and CD86 expression. Exposure of healthy donor cDC2 to Dex or GBM cell lysate resulted in a similar low IL-12IL-10 ratio. Inhibition of p38 restored the IL-12IL-10 balance in Dex or tumor lysate-conditioned healthy cDC2 and enhanced T-cell proliferation and interferon-gamma (IFNγ) production. Importantly, patient-derived cDC2 showed a similar reversal of DC dysfunction with p38i. This study demonstrates the therapeutic potential of developing the next generation of DC vaccines using enhanced p38i-conditioned cDC2. We will therefore shortly embark on a clinical trial of adoptively transferred, p38 MAPK-inhibited cDC2 in adults with GBM.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Clinical_trials Idioma: En Revista: Oncoimmunology Año: 2019 Tipo del documento: Article País de afiliación: Reino Unido

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Clinical_trials Idioma: En Revista: Oncoimmunology Año: 2019 Tipo del documento: Article País de afiliación: Reino Unido