Title Clonal tracing reveals diverse patterns of response to immune checkpoint blockade
Authors Gu, Shengqing Stan
Wang, Xiaoqing
Hu, Xihao
Jiang, Peng
Li, Ziyi
Traugh, Nicole
Bu, Xia
Tang, Qin
Wang, Chenfei
Zeng, Zexian
Fu, Jingxin
Meyer, Cliff
Zhang, Yi
Cejas, Paloma
Lim, Klothilda
Wang, Jin
Zhang, Wubing
Tokheim, Collin
Sahu, Avinash Das
Xing, Xiaofang
Kroger, Benjamin
Ouyang, Zhangyi
Long, Henry
Freeman, Gordon J.
Brown, Myles
Liu, X. Shirley
Affiliation Dana Farber Canc Inst, Dept Data Sci, Boston, MA 02215 USA
Harvard TH Chan Sch Publ Hlth, Dept Biostat, Boston, MA 02215 USA
Dana Farber Canc Inst, Ctr Funct Canc Epigenet, Boston, MA 02215 USA
Dana Farber Canc Inst, Dept Med Oncol, Boston, MA 02215 USA
Harvard Med Sch, Boston, MA 02115 USA
Tongji Univ, Shanghai Pulm Hosp, Sch Life Sci & Technol, Clin Translat Res Ctr, Shanghai 200433, Peoples R China
Peking Univ, Canc Hosp & Inst, Canc Translat Res Lab, Div Gastrointestinal, Beijing 100142, Peoples R China
Univ Texas Southwestern Med Sch, Dallas, TX 75390 USA
Keywords CD8(+) T-CELLS
PD-1 BLOCKADE
ACQUIRED-RESISTANCE
ANTITUMOR IMMUNITY
CLINICAL ACTIVITY
CTLA-4 BLOCKADE
DENDRITIC CELLS
TUMOR-CELLS
CHIP-SEQ
B-CELLS
Issue Date 15-Oct-2020
Publisher GENOME BIOLOGY
Abstract Background Immune checkpoint blockade (ICB) therapy has improved patient survival in a variety of cancers, but only a minority of cancer patients respond. Multiple studies have sought to identify general biomarkers of ICB response, but elucidating the molecular and cellular drivers of resistance for individual tumors remains challenging. We sought to determine whether a tumor with defined genetic background exhibits a stereotypic or heterogeneous response to ICB treatment. Results We establish a unique mouse system that utilizes clonal tracing and mathematical modeling to monitor the growth of each cancer clone, as well as the bulk tumor, in response to ICB. We find that tumors derived from the same clonal populations showed heterogeneous ICB response and diverse response patterns. Primary response is associated with higher immune infiltration and leads to enrichment of pre-existing ICB-resistant cancer clones. We further identify several cancer cell-intrinsic gene expression signatures associated with ICB resistance, including increased interferon response genes and glucocorticoid response genes. These findings are supported by clinical data from ICB treatment cohorts. Conclusions Our study demonstrates diverse response patterns from the same ancestor cancer cells in response to ICB. This suggests the value of monitoring clonal constitution and tumor microenvironment over time to optimize ICB response and to design new combination therapies. Furthermore, as ICB response may enrich for cancer cell-intrinsic resistance signatures, this can affect interpretations of tumor RNA-seq data for response-signature association studies.
URI http://hdl.handle.net/20.500.11897/593188
ISSN 1474-760X
DOI 10.1186/s13059-020-02166-1
Indexed SCI(E)
Appears in Collections: 北京肿瘤医院

Files in This Work
There are no files associated with this item.

Web of Science®


0

Checked on Last Week

Scopus®



Checked on Current Time

百度学术™


0

Checked on Current Time

Google Scholar™





License: See PKU IR operational policies.