Your browser doesn't support javascript.
loading
Decitabine enhances targeting of AML cells by CD34+ progenitor-derived NK cells in NOD/SCID/IL2Rgnull mice.
Cany, Jeannette; Roeven, Mieke W H; Hoogstad-van Evert, Janneke S; Hobo, Willemijn; Maas, Frans; Franco Fernandez, Rosalia; Blijlevens, Nicole M A; van der Velden, Walter J; Huls, Gerwin; Jansen, Joop H; Schaap, Nicolaas P M; Dolstra, Harry.
Afiliação
  • Cany J; Department of Laboratory Medicine, Laboratory of Hematology.
  • Roeven MWH; Department of Laboratory Medicine, Laboratory of Hematology.
  • Hoogstad-van Evert JS; Department of Hematology, and.
  • Hobo W; Department of Laboratory Medicine, Laboratory of Hematology.
  • Maas F; Department of Gynecology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands; and.
  • Franco Fernandez R; Department of Laboratory Medicine, Laboratory of Hematology.
  • Blijlevens NMA; Department of Laboratory Medicine, Laboratory of Hematology.
  • van der Velden WJ; Department of Laboratory Medicine, Laboratory of Hematology.
  • Huls G; Department of Hematology, and.
  • Jansen JH; Department of Hematology, and.
  • Schaap NPM; Department of Hematology, University Medical Center Groningen, Groningen, The Netherlands.
  • Dolstra H; Department of Laboratory Medicine, Laboratory of Hematology.
Blood ; 131(2): 202-214, 2018 01 11.
Article em En | MEDLINE | ID: mdl-29138222
ABSTRACT
Combining natural killer (NK) cell adoptive transfer with hypomethylating agents (HMAs) is an attractive therapeutic approach for patients with acute myeloid leukemia (AML). However, data regarding the impact of HMAs on NK cell functionality are mostly derived from in vitro studies with high nonclinical relevant drug concentrations. In the present study, we report a comparative study of azacitidine (AZA) and decitabine (DAC) in combination with allogeneic NK cells generated from CD34+ hematopoietic stem and progenitor cells (HSPC-NK cells) in in vitro and in vivo AML models. In vitro, low-dose HMAs did not impair viability of HSPC-NK cells. Furthermore, low-dose DAC preserved HSPC-NK killing, proliferation, and interferon gamma production capacity, whereas AZA diminished their proliferation and reactivity. Importantly, we showed HMAs and HSPC-NK cells could potently work together to target AML cell lines and patient AML blasts. In vivo, both agents exerted a significant delay in AML progression in NOD/SCID/IL2Rgnull mice, but the persistence of adoptively transferred HSPC-NK cells was not affected. Infused NK cells showed sustained expression of most activating receptors, upregulated NKp44 expression, and remarkable killer cell immunoglobulin-like receptor acquisition. Most importantly, only DAC potentiated HSPC-NK cell anti-leukemic activity in vivo. Besides upregulation of NKG2D- and DNAM-1-activating ligands on AML cells, DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells. In addition, treatment resulted in increased numbers of HSPC-NK cells in the bone marrow compartment, suggesting that DAC could positively modulate NK cell activity, trafficking, and tumor targeting. These data provide a rationale to explore combination therapy of adoptive HSPC-NK cells and DAC in patients with AML.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Azacitidina / Células Matadoras Naturais / Leucemia Mieloide Aguda / Imunoterapia Adotiva / Decitabina / Antimetabólitos Antineoplásicos Limite: Animals / Humans Idioma: En Revista: Blood Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Azacitidina / Células Matadoras Naturais / Leucemia Mieloide Aguda / Imunoterapia Adotiva / Decitabina / Antimetabólitos Antineoplásicos Limite: Animals / Humans Idioma: En Revista: Blood Ano de publicação: 2018 Tipo de documento: Article