Patient‐derived acellular ascites fluid affects drug responses in ovarian cancer cell lines through the activation of key signalling pathways

Author:

Bischof Katharina12ORCID,Cremaschi Andrea3456,Eroukhmanoff Lena3,Landskron Johannes3,Flage‐Larsen Lise‐Lotte3,Gade Alexandra3,Bjørge Line78,Urbanucci Alfonso910ORCID,Taskén Kjetil111ORCID

Affiliation:

1. Department of Cancer Immunology, Institute for Cancer Research University of Oslo Norway

2. Division of Cancer Medicine, Department of Gynecological Oncology Oslo University Hospital Norway

3. Centre for Molecular Medicine Norway (NCMM) Nordic EMBL Partnership, University of Oslo Norway

4. Oslo Centre for Biostatistics and Epidemiology University of Oslo Norway

5. Singapore Institute for Clinical Sciences, A*STAR Singapore

6. Yong Loo Lin School of Medicine National University of Singapore Singapore

7. Department of Obstetrics and Gynaecology Haukeland University Hospital Bergen Norway

8. Department of Clinical Science, Centre for Cancer Biomarkers CCBIO University of Bergen Norway

9. Faculty of Medicine and Health Technology TAYS Cancer Centre and FICAN Mid, Tampere University Finland

10. Department of Tumor Biology, Institute for Cancer Research University of Oslo Norway

11. Institute of Clinical Medicine University of Oslo Norway

Abstract

Malignant ascites is commonly produced in advanced epithelial ovarian cancer (EOC) and serves as unique microenvironment for tumour cells. Acellular ascites fluid (AAF) is rich in signalling molecules and has been proposed to play a role in the induction of chemoresistance. Through in vitro testing of drug sensitivity and by assessing intracellular phosphorylation status in response to mono‐ and combination treatment of five EOC cell lines after incubation with AAFs derived from 20 different patients, we investigated the chemoresistance‐inducing potential of ascites. We show that the addition of AAFs to the culture media of EOC cell lines has the potential to induce resistance to standard‐of‐care drugs (SCDs). We also show that AAFs induce time‐ and concentration‐dependent activation of downstream signalling to signal transducer and activator of transcription 3 (STAT3), and concomitantly altered phosphorylation of mitogen‐activated protein kinase kinase (MEK), phosphoinositide 3‐kinase (PI3K)–protein kinase B (AKT) and nuclear factor NF‐kappa‐B (NFκB). Antibodies targeting the interleukin‐6 receptor (IL6R) effectively blocked phosphorylation of STAT3 and STAT1. Treatments with SCDs were effective in reducing cell viability in only a third of 30 clinically relevant conditions examined, defined as combinations of drugs, different cell lines and AAFs. Combinations of SCDs and novel therapeutics such as trametinib, fludarabine or rapamycin were superior in another third. Notably, we could nominate effective treatment combinations in almost all conditions except in 4 out of 30 conditions, in which trametinib or fludarabine showed higher efficacy alone. Taken together, our study underscores the importance of the molecular characterisation of individual patients' AAFs and the impact on treatment resistance as providing clinically meaningful information for future precision treatment approaches in EOC.

Funder

Kreftforeningen

Research Council of Finland

Publisher

Wiley

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3