PT - JOURNAL ARTICLE AU - Seyedeh M. Zekavat AU - Vanesa Viana-Huete AU - María A. Zuriaga AU - Md Mesbah Uddin AU - Mark Trinder AU - Kaavya Paruchuri AU - Nuria Matesanz AU - Virginia Zorita AU - Alba Ferrer-Pérez AU - Marta Amorós-Pérez AU - Scott M. Damrauer AU - Christie M. Ballantyne AU - Abhishek Niroula AU - Christopher J. Gibson AU - James Pirruccello AU - Gabriel Griffin AU - Benjamin L. Ebert AU - Peter Libby AU - Valentín Fuster AU - Hongyu Zhao AU - Pradeep Natarajan AU - Alexander G. Bick AU - José J Fuster AU - Derek Klarin TI - <em>TP53-</em>mediated clonal hematopoiesis confers increased risk for incident peripheral artery disease AID - 10.1101/2021.08.22.21262430 DP - 2021 Jan 01 TA - medRxiv PG - 2021.08.22.21262430 4099 - http://medrxiv.org/content/early/2021/08/23/2021.08.22.21262430.short 4100 - http://medrxiv.org/content/early/2021/08/23/2021.08.22.21262430.full AB - Background Somatic mutations in blood indicative of clonal hematopoiesis of indeterminate potential (CHIP), particularly in DNMT3A, TET2, and JAK2, are associated with an increased risk of hematologic malignancy, coronary artery disease, and all-cause mortality. However, whether CHIP is associated with increased risk of peripheral artery disease (PAD) remains unknown. In addition, chemotherapy frequently causes mutations in DNA Damage Repair (DDR) genes TP53 and PPM1D, and whether CHIP caused by somatic mutations in DDR genes results in increased risk of atherosclerosis is unclear. We sought to test whether CHIP, and CHIP caused by DDR genes, associates with incident peripheral artery disease (PAD) and atherosclerosis.Methods We identified CHIP among 50,122 exome sequences in individuals from UK and Mass General Brigham Biobanks and tested CHIP status (N=2,851) with incident PAD and atherosclerosis across multiple arterial beds. To mimic the human scenario of clonal hematopoiesis and test whether the expansion of p53-deficient hematopoietic cells contributes to atherosclerosis, a competitive bone marrow transplantation (BMT) strategy was used to generate atherosclerosis-prone Ldlr-/- chimeric mice carrying 20% Trp53-/- hematopoietic cells (20% KO-BMT mice). We then evaluated aortic plaque burden and plaque macrophage accumulation 12 weeks after grafting.Results CHIP associated with incident PAD (HR 1.7; P=2.2x10-5) and atherosclerosis in multiple beds (HR 1.3; P=9.7x10-5), with increased risk among individuals with DDR CHIP (HR 2.0; P=0.0084). Among atherosclerosis-prone Ldlr null mice, the p53 -/- 20% KO-BMT mice demonstrated increased aortic plaque size (p=0.013) and accumulation of p53-/- plaque macrophages (P&lt;0.001), driven by an abundance of p53-deficient plaque macrophages. The expansion of p53-deficient cells did not affect the expression of the pro-inflammatory cytokines IL-6 and IL-1β in the atherosclerotic aortic wall.Conclusions Our findings highlight the role of CHIP as a broad driver of atherosclerosis across the entire arterial system, with evidence of increased plaque among p53 -/- 20% KO-BMT mice via expansion of plaque macrophages. These observations provide new insight into the link between CHIP and cardiovascular disease, and lend human genetic support to the concept that post-cytotoxic chemotherapy patients may benefit from surveillance for atherosclerotic conditions in addition to therapy-related myeloid neoplasms.Competing Interest StatementP.N. reported grants from Amgen during the conduct of the study and grants from Boston Scientific; grants and personal fees from Apple; personal fees from Novartis and Blackstone Life Sciences; and spousal employment at Vertex all outside the submitted work. P.L. is an unpaid consultant to, or involved in clinical trials for Amgen, AstraZeneca, Baim Institute, Beren Therapeutics, Esperion, Therapeutics, Genentech, Kancera, Kowa Pharmaceuticals, Medimmune, Merck, Norvo Nordisk, Merck, Novartis, Pfizer, Sanofi-Regeneron. Dr. Libby is a member of scientific advisory board for Amgen, Corvidia Therapeutics, DalCor Pharmaceuticals, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Novartis, and XBiotech, Inc. P.L.'s laboratory has received research funding in the last 2 years from Novartis, he is on the Board of Directors of XBiotech, Inc, and has a financial interest in Xbiotech, a company developing therapeutic human antibodies. These interests were reviewed and are managed by Brigham and Women's Hospital and Partners HealthCare in accordance with their conflict of interest policies. The other authors do not report any disclosures.Funding StatementP.N. is supported by a Hassenfeld Scholar Award from the Massachusetts General Hospital, grants from the National Heart, Lung, and Blood Institute (R01HL1427, R01HL148565, and R01HL148050), and from Fondation Leducq (TNE-18CVD04). S.M.Z is supported by the NIH National Heart, Lung, and Blood Institute (1F30HL149180-01) and the NIH Medical Scientist Training Program Training Grant (T32GM136651). A.G.B. is supported by a Burroughs Wellcome Fund Career Award for Medical Scientists, a NIH Director Early Independence Award (DP5-OD029586) and a NHLBI BioData Catalyst Fellowship (OT3 HL147154-01). J.P.P is supported by a John S LaDue Memorial Fellowship. K.P. is supported by NIH grant 5-T32HL007208-43. J.J.F. is supported by grants RYC-2016-20026 and RTI2018-093554-A-I00) from the Spanish 'Ministerio de Ciencia e Innovacion', a 2019 Leonardo Grant for Researchers and Cultural Creators from the BBVA Foundation, the European Research Area Network on Cardiovascular Diseases CHEMICAL (grant AC19/00133 from the 'Spanish Instituto de Salud Carlos III') and the Leducq Foundation (TNE-18CVD04). The project leading to these results received funding from 'la Caixa' Foundation (ID 100010434), under agreement HR17-00267. The Centro Nacional de Investigaciones Cardiovasculares (CNIC) is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovacion and the Pro CNIC Foundation. P.L. receives funding support from the National Heart, Lung, and Blood Institute (1R01HL134892), the American Heart Association (18CSA34080399), the RRM Charitable Fund, and the Simard Fund.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:UK Biobank analyses were conducted using Application 7089.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.YesUKB individual-level data are available for request by application (https://www.ukbiobank.ac.uk). Individual-level MGBB data are available from https://personalizedmedicine.partners.org/Biobank/Default.aspx, but restrictions apply to the availability of these data, which were used under IRB approval for the current study, and so are not publicly available. The present article includes all other data generated or analyzed during this study.CHIPclonal hematopoiesis of indeterminate potentialPADperipheral artery disease