Your browser doesn't support javascript.
loading
Myeloid checkpoint blockade improves killing of T-acute lymphoblastic leukemia cells by an IgA2 variant of daratumumab.
Baumann, Niklas; Arndt, Christian; Petersen, Judith; Lustig, Marta; Rösner, Thies; Klausz, Katja; Kellner, Christian; Bultmann, Miriam; Bastian, Lorenz; Vogiatzi, Fotini; Leusen, Jeanette H W; Burger, Renate; Schewe, Denis M; Peipp, Matthias; Valerius, Thomas.
Afiliação
  • Baumann N; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Arndt C; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Petersen J; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Lustig M; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Rösner T; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Klausz K; Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian- Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Kellner C; Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany.
  • Bultmann M; Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Bastian L; Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Vogiatzi F; Pediatric Hematology/Oncology, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Leusen JHW; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.
  • Burger R; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Schewe DM; Children's Hospital, University Medical Center Magdeburg, Magdeburg, Germany.
  • Peipp M; Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian- Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
  • Valerius T; Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
Front Immunol ; 13: 949140, 2022.
Article em En | MEDLINE | ID: mdl-36052078
ABSTRACT
Antibody-based immunotherapy is increasingly employed to treat acute lymphoblastic leukemia (ALL) patients. Many T-ALL cells express CD38 on their surface, which can be targeted by the CD38 antibody daratumumab (DARA), approved for the treatment of multiple myeloma. Tumor cell killing by myeloid cells is relevant for the efficacy of many therapeutic antibodies and can be more efficacious with human IgA than with IgG antibodies. This is demonstrated here by investigating antibody-dependent cellular phagocytosis (ADCP) by macrophages and antibody-dependent cell-mediated cytotoxicity (ADCC) by polymorphonuclear (PMN) cells using DARA (human IgG1) and an IgA2 isotype switch variant (DARA-IgA2) against T-ALL cell lines and primary patient-derived tumor cells. ADCP and ADCC are negatively regulated by interactions between CD47 on tumor cells and signal regulatory protein alpha (SIRPα) on effector cells. In order to investigate the impact of this myeloid checkpoint on T-ALL cell killing, CD47 and glutaminyl-peptide cyclotransferase like (QPCTL) knock-out T-ALL cells were employed. QPTCL is an enzymatic posttranslational modifier of CD47 activity, which can be targeted by small molecule inhibitors. Additionally, we used an IgG2σ variant of the CD47 blocking antibody magrolimab, which is in advanced clinical development. Moreover, treatment of T-ALL cells with all-trans retinoic acid (ATRA) increased CD38 expression leading to further enhanced ADCP and ADCC, particularly when DARA-IgA2 was applied. These studies demonstrate that myeloid checkpoint blockade in combination with IgA2 variants of CD38 antibodies deserves further evaluation for T-ALL immunotherapy.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Antígeno CD47 / Leucemia-Linfoma Linfoblástico de Células T Precursoras Limite: Humans Idioma: En Revista: Front Immunol Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Antígeno CD47 / Leucemia-Linfoma Linfoblástico de Células T Precursoras Limite: Humans Idioma: En Revista: Front Immunol Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha