Your browser doesn't support javascript.
loading
RIG-I-based immunotherapy enhances survival in preclinical AML models and sensitizes AML cells to checkpoint blockade.
Ruzicka, Michael; Koenig, Lars M; Formisano, Simone; Boehmer, Daniel F R; Vick, Binje; Heuer, Eva-M; Meinl, Hanna; Kocheise, Lorenz; Zeitlhöfler, Marcus; Ahlfeld, Julia; Kobold, Sebastian; Endres, Stefan; Subklewe, Marion; Duewell, Peter; Schnurr, Max; Jeremias, Irmela; Lichtenegger, Felix S; Rothenfusser, Simon.
Afiliação
  • Ruzicka M; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Koenig LM; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Formisano S; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
  • Boehmer DFR; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Vick B; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Heuer EM; Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
  • Meinl H; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Kocheise L; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Zeitlhöfler M; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Ahlfeld J; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Kobold S; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Endres S; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
  • Subklewe M; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Duewell P; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany.
  • Schnurr M; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
  • Jeremias I; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.
  • Lichtenegger FS; Laboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany.
  • Rothenfusser S; German Cancer Consortium (DKTK), partner site Munich, Munich, Germany.
Leukemia ; 34(4): 1017-1026, 2020 04.
Article em En | MEDLINE | ID: mdl-31740809
ABSTRACT
Retinoic acid-inducible gene-I (RIG-I) is a cytoplasmic immune receptor sensing viral RNA. It triggers the release of type I interferons (IFN) and proinflammatory cytokines inducing an adaptive cellular immune response. We investigated the therapeutic potential of systemic RIG-I activation by short 5'-triphosphate-modified RNA (ppp-RNA) for the treatment of acute myeloid leukemia (AML) in the syngeneic murine C1498 AML tumor model. ppp-RNA treatment significantly reduced tumor burden, delayed disease onset and led to complete remission including immunological memory formation in a substantial proportion of animals. Therapy-induced tumor rejection was dependent on CD4+ and CD8+ T cells, but not on NK or B cells, and relied on intact IFN and mitochondrial antiviral signaling protein (MAVS) signaling in the host. Interestingly, ppp-RNA treatment induced programmed death ligand 1 (PD-L1) expression on AML cells and established therapeutic sensitivity to anti-PD-1 checkpoint blockade in vivo. In immune-reconstituted humanized mice, ppp-RNA treatment reduced the number of patient-derived xenografted (PDX) AML cells in blood and bone marrow while concomitantly enhancing CD3+ T cell counts in the respective tissues. Due to its ability to establish a state of full remission and immunological memory, our findings show that ppp-RNA treatment is a promising strategy for the immunotherapy of AML.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Receptores Virais / RNA de Cadeia Dupla / Leucemia Mieloide Aguda / Anticorpos Neutralizantes / Proteína DEAD-box 58 / Imunoterapia Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Leukemia Assunto da revista: HEMATOLOGIA / NEOPLASIAS Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Alemanha

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Receptores Virais / RNA de Cadeia Dupla / Leucemia Mieloide Aguda / Anticorpos Neutralizantes / Proteína DEAD-box 58 / Imunoterapia Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Leukemia Assunto da revista: HEMATOLOGIA / NEOPLASIAS Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Alemanha