4.8 Article

Ovarian cancer mutational processes drive site-specific immune evasion

期刊

NATURE
卷 612, 期 7941, 页码 778-+

出版社

NATURE PORTFOLIO
DOI: 10.1038/s41586-022-05496-1

关键词

-

资金

  1. Cycle for Survival supporting Memorial Sloan Kettering Cancer Center
  2. Nicholls Biondi Chair in Computational Oncology
  3. Ovarian Cancer Research Alliance (OCRA) Collaborative Research Development Grant [648007]
  4. OCRA Liz Tilberis Award [657721]
  5. OCRA Ann Schreiber Mentored Investigator Award [650687]
  6. Department of Defense Congressionally Directed Medical Research Programs (CDMRP) award [W81XWH-20-1-0565]
  7. CDMRP Ovarian Cancer Research Academy award [OC150111]
  8. CDMRP award [W81XWH-21-1-0561]
  9. LesLois Shaw Foundation [C42358/A27460]
  10. Cancer Research UK Grand Challenge Program [C42358/A27460]
  11. Breast Cancer Research Foundation grant
  12. National Cancer Institute (NCI) grant [P50-CA247749]
  13. Marie-Josee and Henry R. Kravis Center for Molecular Oncology
  14. NCI Cancer Center Core grant [P30-CA008748]
  15. NCI [F30-CA257414, K12-CA184746]
  16. NIH T32 MD-PhD training program [GM007739]
  17. [P50-CA247749-01]

向作者/读者索取更多资源

This study provides insights into the relationship between mutational processes, anatomical sites, and immune resistance mechanisms in HGSOC, highlighting the importance of personalized immunotherapeutic approaches and early detection research.
High-grade serous ovarian cancer (HGSOC) is an archetypal cancer of genomic instability(1-4) patterned by distinct mutational processes(5,6), tumour heterogeneity(7-9) and intraperitoneal spread(7,8,10). Immunotherapies have had limited efficacy in HGSOC(11-13), highlighting an unmet need to assess how mutational processes and the anatomical sites of tumour foci determine the immunological states of the tumour microenvironment. Here we carried out an integrative analysis of whole-genome sequencing, single-cell RNA sequencing, digital histopathology and multiplexed immunofluorescence of 160 tumour sites from 42 treatment-naive patients with HGSOC. Homologous recombination-deficient HRD-Dup (BRCA1 mutant-like) and HRD-Del (BRCA2 mutant-like) tumours harboured inflammatory signalling and ongoing immunoediting, reflected in loss of HLA diversity and tumour infiltration with highly differentiated dysfunctional CD8(+) T cells. By contrast, foldback-inversion-bearing tumours exhibited elevated immunosuppressive TGF beta signalling and immune exclusion, with predominantly naive/stem-like and memory T cells. Phenotypic state associations were specific to anatomical sites, highlighting compositional, topological and functional differences between adnexal tumours and distal peritoneal foci. Our findings implicate anatomical sites and mutational processes as determinants of evolutionary phenotypic divergence and immune resistance mechanisms in HGSOC. Our study provides a multi-omic cellular phenotype data substrate from which to develop and interpret future personalized immunotherapeutic approaches and early detection research.

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