4.8 Article

3D Functional Genomics Screens Identify CREBBP as a Targetable Driver in Aggressive Triple-Negative Breast Cancer

期刊

CANCER RESEARCH
卷 81, 期 4, 页码 847-859

出版社

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/0008-5472.CAN-20-1822

关键词

-

类别

资金

  1. Breast Cancer Now
  2. Breast Cancer Now's Catalyst Programme [2018NovPR100]
  3. Pfizer
  4. Institute of Cancer Research
  5. MRC
  6. Breast Cancer Now [2013NovPhD185, NC3Rs NC/P001262/1, 2014NovPR36, CTR-Q4]
  7. National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London
  8. Cancer Research UK
  9. British Columbia Cancer Agency Branch
  10. NHS
  11. ICR
  12. MRC [MC_PC_18051] Funding Source: UKRI

向作者/读者索取更多资源

This study highlights the identification of CREBBP as a novel driver in aggressive TNBC, with associated genetic vulnerability in tumor cells with alterations in CREBBP, and provides a preclinical rationale for assessing CREBBP alterations as a biomarker of CDK4/6 inhibitor response in a new patient population. Targeting CREBBP alterations with clinical CDK4/6 inhibitors selectively impairs growth in spheroids, cell line xenografts, and patient-derived models from multiple tumor types.
Triple-negative breast cancers (TNBC) are resistant to standard-of-care chemotherapy and lack known targetable driver gene alterations. Identification of novel drivers could aid the discovery of new treatment strategies for this hard-to-treat patient population, yet studies using high-throughput and accurate models to define the functions of driver genes in TNBC to date have been limited. Here, we employed unbiased functional genomics screening of the 200 most frequently mutated genes in breast cancer, using spheroid cultures to model in vivo-like conditions, and identified the histone acetyltransferase CREBBP as a novel tumor suppressor in TNBC. CREBBP protein expression in patient tumor samples was absent in 8% of TNBCs and at a high frequency in other tumors, including squamous lung cancer, where CREBBP-inactivating mutations are common. In TNBC, CREBBP alterations were associated with higher genomic heterogeneity and poorer patient survival and resulted in upregulation and dependency on a FOXM1 proliferative program. Targeting FOXM1-driven proliferation indirectly with clinical CDK4/6 inhibitors (CDK4/6i) selectively impaired growth in spheroids, cell line xenografts, and patient-derived models from multiple tumor types with CREBBP mutations or loss of protein expression. In conclusion, we have identified CREBBP as a novel driver in aggressive TNBC and identified an associated genetic vulnerability in tumor cells with alterations in CREBBP and provide a preclinical rationale for assessing CREBBP alterations as a biomarker of CDK4/6i response in a new patient population. Significance: This study demonstrates that CREBBP genomic alterations drive aggressive TNBC, lung cancer, and lymphomas and may be selectively treated with clinical CDK4/6 inhibitors.

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

4.8
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