Abstract 2789: Tumor suppressor genotype dramatically impacts lung cancer response to KRAS G12C inhibitors in vivo

Paul J. McMurdie,Ian P. Winters,Lily M. Blair, Lafia Sebastian,Vy B. Tran, Gabriel Grenot, Edwin A. Apilado, Edwin A. Apilado,Ian Lai, Gregory D. Wall,Dmitri A. Petrov,Monte M. Winslow,Michael J. Rosen, Joseph Juan

Cancer Research(2023)

引用 0|浏览0
暂无评分
摘要
Abstract Cancer is characterized by diverse genomic alterations that change cell state and can modify therapy responses. In some cases, the relationships between genotype and drug responses are obvious, such as the response of tumors with defined oncogenic mutations to inhibitors of those mutant proteins. However, covalent KRAS G12C inhibitors (G12Ci) induce clinical responses in less than half of patients with oncogenic KRAS G12C-driven lung cancer, illustrating that genomic alterations not obviously related to the drug mechanism can have a dramatic impact. We have developed and optimized an in vivo platform to identify pharmacogenomic interactions that dictate lung cancer responses to therapies. By coupling somatic genome editing, tumor barcoding, ultra-deep barcode sequencing, and robust statistical methods in genetically engineered mouse models of human lung cancer, we can quantify and compare candidate therapies across thousands of clonal tumors of diverse tumor suppressor genotypes. This platform provides a uniquely high-throughput and quantitative assessment of tumors that are initiated de novo within the natural immunocompetent tissue environment. To uncover genotypes that are particularly important in controlling the response of tumors to oncogenic KRAS inhibition, we applied this platform to quantify the impact of 59 tumor suppressor genes on KRASG12C-driven lung tumor responses to G12Ci. Treatment resulted in approximately three-quarters reduction in both average tumor sizes and overall tumor burden, with a clear drug and dose dependence. Approximately one-quarter of inactivated genes significantly and consistently altered tumor responses across numerous G12Ci and replicate studies. Interestingly, most of these genes are thought to be in pathways not directly related to RAS signaling, and they exhibit a striking pattern in which treatment sensitivity is positively correlated with strength of the tumor suppressor effect. This pharmacogenomic profile for G12Ci was categorically different than the profiles we have observed for chemotherapy and other therapies. Overall, these results provide direct causal evidence that certain tumor suppressor genotypes dramatically shift the effectiveness of G12Ci in vivo and generate hypotheses about patients likely to benefit from G12Ci relative to alternatives. Analysis of available human data provides early clinical support for some of these hypotheses. Ongoing efforts to define pharmacogenomic profiles of combination therapies could be of even greater importance. Platforms that can accurately predict how tumor genotype drives responses have the potential to transform precision cancer therapy, enabling more effective patient stratification and therapy combinations. Citation Format: Paul J. McMurdie, Ian P. Winters, Lily M. Blair, Lafia Sebastian, Vy Tran, Gabriel Grenot, Edwin A. Apilado, Edwin A. Apilado, Ian K. Lai, Gregory D. Wall, Dmitri A. Petrov, Monte M. Winslow, Michael J. Rosen, Joseph Juan. Tumor suppressor genotype dramatically impacts lung cancer response to KRAS G12C inhibitors in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2789.
更多
查看译文
关键词
kras g12c inhibitors,tumor suppressor genotype,lung cancer response,lung cancer
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要