PT - JOURNAL ARTICLE AU - Au, Lewis AU - Hatipoglu, Emine AU - de Massy, Marc Robert AU - Litchfield, Kevin AU - Rowan, Andrew AU - Thompson, Rachael AU - Schnidrig, Desiree AU - Byrne, Fiona AU - Beattie, Gordon AU - Horswell, Stuart AU - Fotiadis, Nicos AU - Hazell, Steve AU - Nicol, David AU - Shepherd, Scott Thomas Colville AU - Fendler, Annika AU - Mason, Robert AU - Attig, Jan AU - Joshi, Kroopa AU - Uddin, Imran AU - Becker, Pablo AU - Sunderland, Mariana Werner AU - Akarca, Ayse AU - Puccio, Ignazio AU - Yang, William AU - Lund, Tom AU - Dhillon, Kim AU - Vasquez, Marcos Duran AU - Ghorani, Ehsan AU - Xu, Hang AU - López, José Ignacio AU - Green, Anna AU - Mahadeva, Ula AU - Borg, Elaine AU - Mitchison, Miriam AU - Moore, David AU - Proctor, Ian AU - Falzon, Mary AU - Furness, Andrew AU - Pickering, Lisa AU - Reading, James L. AU - Salgado, Roberto AU - Marafioti, Teresa AU - Jamal-Hanjani, Mariam AU - , AU - Kassiotis, George AU - Chain, Benny AU - Larkin, James AU - Swanton, Charles AU - Quezada, Sergio A AU - Turajlic, Samra AU - , TI - Determinants of anti-PD1 response and resistance in clear cell renal cell carcinoma AID - 10.1101/2021.03.19.21253661 DP - 2021 Jan 01 TA - medRxiv PG - 2021.03.19.21253661 4099 - http://medrxiv.org/content/early/2021/03/28/2021.03.19.21253661.short 4100 - http://medrxiv.org/content/early/2021/03/28/2021.03.19.21253661.full AB - Antigen recognition and T-cell mediated cytotoxicity in clear-cell renal cell carcinoma (ccRCC) remains incompletely understood. To address this knowledge gap, we analysed 115 multiregion tumour samples collected from 15 treatment-naïve patients pre- and post-nivolumab therapy, and at autopsy in three patients. We performed whole-exome sequencing, RNAseq, TCRseq, multiplex immunofluorescence and flow cytometry analyses and correlated with clinical response. We observed pre-treatment intratumoural TCR clonal expansions suggesting pre-existing immunity. Nivolumab maintained pre-treatment expanded, clustered TCR clones in responders, suggesting ongoing antigen-driven stimulation of T-cells. T-cells in responders were enriched for expanded TCF7+CD8+ T-cells and upregulated GZMK/B upon nivolumab-binding. By contrast, nivolumab promoted accumulation of new TCR clones in non-responders, replacing pre-treatment expanded clonotypes. In this dataset, mutational features did not correlate with response to nivolumab and human endogenous retrovirus expression correlated indirectly. Our data suggests that nivolumab potentiates clinical responses in ccRCC by binding pre-existing expanded CD8+ T-cells to enhance cytotoxicity.Competing Interest StatementL.A. is funded by the Royal Marsden Cancer Charity. J.A. is a full-time employee of Hoffmann-La Roche AG (Basel, Switzerland). L.P. has received research funding from Pierre Fabre, and honoria from Pfizer, Ipsen, Bristol-Myers Squibb, and EUSA Pharma. S.T. is funded by Cancer Research UK (C50947/A18176), the National Institute for Health Research (NIHR) Biomedical Research Centre at the Royal Marsden Hospital and Institute of Cancer Research (A109), the Kidney and Melanoma Cancer Fund of The Royal Marsden Cancer Charity, The Rosetrees Trust (A2204), Ventana Medical Systems Inc (10467/10530), the National Institute of Health (US) and the Melanoma Research Alliance. R.S. has received non-financial support from Merck and Bristol Myers Squibb, research support from Merck, Puma Biotechnology, and Roche; and advisory board fees for Bristol Myers Squibb; and personal fees from Roche for an advisory board related to a trial-research project; all related to breast cancer research projects. R.S. reports no conflict of interests related to this project. G.K. receives core funding from the Francis Crick Institute (FC0010099). M.J.H. is a Cancer Research UK (CRUK) Clinician Scientist (RCCFEL\100099) and has received funding from CRUK, National Institute for Health Research, Rosetrees Trust, UKI NETs and NIHR University College London Hospitals Biomedical Research Centre. M.J.H. is a member of the Scientific Advisory Board and Steering Committee for Achilles Therapeutics. J.L. has received research funding from Bristol-Myers Squibb, Merck, Novartis, Pfizer, Achilles Therapeutics, Roche, Nektar Therapeutics, Covance, Immunocore, Pharmacyclics, and Aveo, and served as a consultant to Achilles, AstraZeneca, Boston Biomedical, Bristol-Myers Squibb, Eisai, EUSA Pharma, GlaxoSmithKline, Ipsen, Imugene, Incyte, iOnctura, Kymab, Merck Serono, Nektar, Novartis, Pierre Fabre, Pfizer, Roche Genentech, Secarna, and Vitaccess. C.S. acknowledges grant support from Pfizer, AstraZeneca, Bristol-Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Archer Dx Inc (collaboration in minimal residual disease sequencing technologies) and Ono Pharmaceutical, is an AstraZeneca Advisory Board member and Chief Investigator for the MeRmaiD1 clinical trial, has consulted for Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol-Myers Squibb, Celgene, AstraZeneca, Illumina, Genentech, Roche-Ventana, GRAIL, Medicxi, Bicycle Therapeutics, and the Sarah Cannon Research Institute, has stock options in Apogen Biotechnologies, Epic Bioscience, GRAIL, and has stock options and is co-founder of Achilles Therapeutics. Patents: C.S. holds European patents relating to assay technology to detect tumour recurrence (PCT/GB2017/053289); to targeting neoantigens (PCT/EP2016/059401), identifying patent response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), identifying patients who respond to cancer treatment (PCT/GB2018/051912), a US patent relating to detecting tumour mutations (PCT/US2017/28013) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892). C.S. is Royal Society Napier Research Professor (RP150154). His work is supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001169), the UK Medical Research Council (FC001169), and the Wellcome Trust (FC001169). C.S. is funded by Cancer Research UK (TRACERx, PEACE and CRUK Cancer Immunotherapy Catalyst Network), Cancer Research UK Lung Cancer Centre of Excellence, the Rosetrees Trust, Butterfield and Stoneygate Trusts, NovoNordisk Foundation (ID16584), Royal Society Research Professorship Enhancement Award (RP/EA/180007), the NIHR BRC at University College London Hospitals, the CRUK-UCL Centre, Experimental Cancer Medicine Centre and the Breast Cancer Research Foundation, USA (BCRF). His research is supported by a Stand Up To Cancer-LUNGevity-American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (SU2C-AACR-DT23-17). Stand Up To Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the Scientific Partner of SU2C. C.S. also receives funding from the European Research Council (ERC) under the European Unions Seventh Framework Programme (FP7/2007-2013) Consolidator Grant (FP7-THESEUS-617844), European Commission ITN (FP7-PloidyNet 607722), an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Unions Horizon 2020 research and innovation programme (835297) and Chromavision from the European Unions Horizon 2020 research and innovation programme (665233). ST has received speaking fees from Roche, Astra Zeneca, Novartis and Ipsen. ST has the following patents filed: Indel mutations as a therapeutic target and predictive biomarker PCTGB2018/051892 and PCTGB2018/051893 and Clear Cell Renal Cell Carcinoma Biomarkers P113326GB.Clinical TrialNCT02446860Funding StatementThe ADAPTeR study (CA209-129) is sponsored by The Royal Marsden NHS Foundation Trust, and partly funded by National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) at the Royal Marsden Hospital and Institute of Cancer Research (ICR) (A80), and Cancer Research UK (CRUK) (17767). Bristol-Myers Squibb was the drug provider. The Francis Crick Institute, which receives its core funding from CRUK (FC010110), the UK Medical Research Council (FC010110), the Wellcome Trust (FC010110). For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission. TRACERx Renal is funded by NIHR BRC at the Royal Marsden Hospital and Institute of Cancer Research (A109).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:ADAPTeR (NCT02446860) was initially approved by NRES Committee London Fulham on 01/12/2014. ADAPTeR is performed in accordance with the ethical principles in the Declaration of Helsinki, Good Clinical Practice and applicable regulatory requirements.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesFurther information and requests for resources, data and reagents should be directed to and will be fulfilled by the Lead Contact, Samra Turajlic (samra.turajlic@crick.ac.uk).