Your browser doesn't support javascript.
loading
Epigenetic promoter alterations in GI tumour immune-editing and resistance to immune checkpoint inhibition.
Sundar, Raghav; Huang, Kie-Kyon; Kumar, Vikrant; Ramnarayanan, Kalpana; Demircioglu, Deniz; Her, Zhisheng; Ong, Xuewen; Bin Adam Isa, Zul Fazreen; Xing, Manjie; Tan, Angie Lay-Keng; Tai, David Wai Meng; Choo, Su Pin; Zhai, Weiwei; Lim, Jia Qi; Das Thakur, Meghna; Molinero, Luciana; Cha, Edward; Fasso, Marcella; Niger, Monica; Pietrantonio, Filippo; Lee, Jeeyun; Jeyasekharan, Anand D; Qamra, Aditi; Patnala, Radhika; Fabritius, Arne; De Simone, Mark; Yeong, Joe; Ng, Cedric Chuan Young; Rha, Sun Young; Narita, Yukiya; Muro, Kei; Guo, Yu Amanda; Skanderup, Anders Jacobsen; So, Jimmy Bok Yan; Yong, Wei Peng; Chen, Qingfeng; Göke, Jonathan; Tan, Patrick.
Afiliação
  • Sundar R; Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore gmstanp@duke-nus.edu.sg raghav_sundar@nuhs.edu.sg.
  • Huang KK; Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
  • Kumar V; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Ramnarayanan K; The N.1 Institute for Health, National University of Singapore, Singapore.
  • Demircioglu D; Singapore Gastric Cancer Consortium, Singapore.
  • Her Z; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Ong X; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Bin Adam Isa ZF; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Xing M; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
  • Tan AL; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.
  • Tai DWM; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Choo SP; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Zhai W; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
  • Lim JQ; Diagnostic Development Hub (DxD), Agency for Science, Technology and Research, Singapore.
  • Das Thakur M; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Molinero L; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
  • Cha E; Diagnostic Development Hub (DxD), Agency for Science, Technology and Research, Singapore.
  • Fasso M; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.
  • Niger M; Division of Medical Oncology, National Cancer Centre, Singapore.
  • Pietrantonio F; Division of Medical Oncology, National Cancer Centre, Singapore.
  • Lee J; Curie Oncology, Singapore.
  • Jeyasekharan AD; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
  • Qamra A; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
  • Patnala R; Department of Development Sciences, Genentech, San Francisco, California, USA.
  • Fabritius A; Department of Development Sciences, Genentech, San Francisco, California, USA.
  • De Simone M; Department of Development Sciences, Genentech, San Francisco, California, USA.
  • Yeong J; Department of Development Sciences, Genentech, San Francisco, California, USA.
  • Ng CCY; Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
  • Rha SY; Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
  • Narita Y; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
  • Muro K; Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore.
  • Guo YA; Cancer Science Institute of Singapore, National University of Singapore, Singapore.
  • Skanderup AJ; Statistical Programming and Analytics, Roche Canada, Mississauga, Ontario, Canada.
  • So JBY; University Health Network, Toronto, Ontario, Canada.
  • Yong WP; Sci-illustrate, Munich, Germany.
  • Chen Q; Sci-illustrate, Munich, Germany.
  • Göke J; InSilico Genomics, Phoenix, Arizona, USA.
  • Tan P; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.
Gut ; 71(7): 1277-1288, 2022 07.
Article em En | MEDLINE | ID: mdl-34433583
ABSTRACT

OBJECTIVES:

Epigenomic alterations in cancer interact with the immune microenvironment to dictate tumour evolution and therapeutic response. We aimed to study the regulation of the tumour immune microenvironment through epigenetic alternate promoter use in gastric cancer and to expand our findings to other gastrointestinal tumours.

DESIGN:

Alternate promoter burden (APB) was quantified using a novel bioinformatic algorithm (proActiv) to infer promoter activity from short-read RNA sequencing and samples categorised into APBhigh, APBint and APBlow. Single-cell RNA sequencing was performed to analyse the intratumour immune microenvironment. A humanised mouse cancer in vivo model was used to explore dynamic temporal interactions between tumour kinetics, alternate promoter usage and the human immune system. Multiple cohorts of gastrointestinal tumours treated with immunotherapy were assessed for correlation between APB and treatment outcomes.

RESULTS:

APBhigh gastric cancer tumours expressed decreased levels of T-cell cytolytic activity and exhibited signatures of immune depletion. Single-cell RNAsequencing analysis confirmed distinct immunological populations and lower T-cell proportions in APBhigh tumours. Functional in vivo studies using 'humanised mice' harbouring an active human immune system revealed distinct temporal relationships between APB and tumour growth, with APBhigh tumours having almost no human T-cell infiltration. Analysis of immunotherapy-treated patients with GI cancer confirmed resistance of APBhigh tumours to immune checkpoint inhibition. APBhigh gastric cancer exhibited significantly poorer progression-free survival compared with APBlow (median 55 days vs 121 days, HR 0.40, 95% CI 0.18 to 0.93, p=0.032).

CONCLUSION:

These findings demonstrate an association between alternate promoter use and the tumour microenvironment, leading to immune evasion and immunotherapy resistance.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Gástricas / Neoplasias Gastrointestinais Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Gut Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Gástricas / Neoplasias Gastrointestinais Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Gut Ano de publicação: 2022 Tipo de documento: Article