4.6 Article

Antibody-Drug Conjugate Efficacy in Neuroblastoma: Role of Payload, Resistance Mechanisms, Target Density, and Antibody Internalization

期刊

MOLECULAR CANCER THERAPEUTICS
卷 20, 期 11, 页码 2228-2239

出版社

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1535-7163.MCT-20-1034

关键词

-

类别

资金

  1. Damon Runyon Cancer Research Foundation [PST-07-16]
  2. St. Baldrick's Foundation-Stand Up to Cancer Dream Team Translational Research Grant [SU2C-AACR-DT27-17]
  3. Hyundai Hope on Wheels Young Investigator Award
  4. NCI [K08 CA230223, U54 CA232568]
  5. EVAN Foundation
  6. Alex's Lemonade Stand Foundation

向作者/读者索取更多资源

In this study, it was found that DNA binding drugs exhibit higher cytotoxicity to neuroblastomas compared to tubulin-binding or DNA topoisomerase 1 inhibiting drugs. The internalization kinetics of antibodies in neuroblastomas correlated significantly with GPC2 cell surface density, while sensitivity to PBD dimers primarily determined the efficacy of corresponding ADC, outweighing the influence of GPC2 cell surface density and antibody internalization.
Antibody-drug conjugates (ADC) are a targeted cancer therapy that utilize the specificity of antibodies to deliver potent drugs selectively to tumors. Here we define the complex interaction among factors that dictate ADC efficacy in neuroblastoma by testing both a comprehensive panel of ADC payloads in a diverse set of neuroblastoma cell lines and utilizing the glypican 2 (GPC2)-targeting D3-GPC2-PBD ADC to study the role of target antigen density and antibody internalization in ADC efficacy in neuroblastoma. We first find that DNA binding drugs are significantly more cytotoxic to neuroblastomas than payloads that bind tubulin or inhibit DNA topoisomerase 1. We additionally show that neuroblastomas with high expression of the ABCB1 drug transporter or that harbor a TP53 mutation are significantly more resistant to tubulin and DNA/DNA topoisomerase 1 binding payloads, respectively. Next, we utilized the GPC2-specific D3-GPC2-IgG1 antibody to show that neuroblastomas internalize this antibody/GPC2 complex at significantly different rates and that these antibody internalization kinetics correlate significantly with GPC2 cell surface density. However, sensitivity to pyrrolobenzodiazepine (PBD) dimers primarily dictated sensitivity to the corresponding D3-GPC2-PBD ADC, overall having a larger influence on ADC efficacy than GPC2 cell surface density or antibody internalization. Finally, we utilized GPC2 isogenic Kelly neuroblastoma cells with different levels of cell surface GPC2 expression to define the threshold of target density required for ADC efficacy. Taken together, DNA binding ADC payloads should be prioritized for development for neuroblastoma given their superior efficacy and considering that ADC payload sensitivity is a major determinant of ADC efficacy.

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

4.6
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