4.7 Article

Bifunctional PD-1 x αCD3 x αCD33 fusion protein reverses adaptive immune escape in acute myeloid leukemia

期刊

BLOOD
卷 132, 期 23, 页码 2484-2494

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood-2018-05-849802

关键词

-

资金

  1. Deutsche Forschungsgemeinschaft (DFG) [CRC1243]
  2. Bavarian Elite Graduate Training Network i-Target
  3. Bavarian Ministry of Economic Affairs
  4. Marie-Sklodowska-Curie Training Network Immutrain - H2020 Program of the European Union
  5. DFG fellowship through the Research Training Group 1202 Oligonucleotides in cell biology and therapy
  6. Else-Kroner Fresenius-Stiftung
  7. German Cancer Aid
  8. European Research Council

向作者/读者索取更多资源

The CD33-targeting bispecific T-cell engager (BiTE) AMG 330 proved to be highly efficient in mediating cytolysis of acute myeloid leukemia (AML) cells in vitro and in mouse models. Yet, T-cell activation is correlated with upregulation of programmed cell death-ligand 1 (PD-L1) and other inhibitory checkpoints on AML cells that confer adaptive immune resistance. PD-1 and PD-L1 blocking agents may counteract T-cell dysfunction, however, at the expense of broadly distributed immune-related adverse events (irAEs). We developed a bifunctional checkpoint inhibitory T cell-engaging (CiTE) antibody that combines T-cell redirection to CD33 on AML cells with locally restricted immune checkpoint blockade. This is accomplished by fusing the extracellular domain of PD-1 (PD-1(ex)), which naturally holds a low affinity to PD-L1, to an alpha CD3. alpha CD33 BiTE-like scaffold. By a synergistic effect of checkpoint blockade and avidity-dependent binding, the PD-1(ex) attachment increases T-cell activation (3.3-fold elevation of interferon-g) and leads to efficient and highly selective cytotoxicity against CD33(+)PD-L1(+) cell lines (50% effective concentration 5 2.3-26.9 pM) as well as patient-derived AML cells (n = 8). In a murine xenograft model, the CiTE induces complete AML eradication without initial signs of irAEs as measured by body weight loss. We conclude that our molecule preferentially targets AML cells, whereas high-affinity blockers, such as clinically approved anticancer agents, also address PD-L1(+) non-AML cells. By combining the high efficacy of T-cell engagers with immune checkpoint blockade in a single molecule, we expect to minimize irAEs associated with the systemic application of immune checkpoint inhibitors and suggest high therapeutic potential, particularly for patients with relapsed/refractory AML.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据