Blokzijl F, de Ligt J, Jager M, Sasselli V, Roerink S, Sasaki N, et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature. 2016;538(7624):260–4.
Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells. Science. 2015;349(6255):1483–9.
Robertson NA, Latorre-Crespo E, Terradas-Terradas M, Lemos-Portela J, Purcell AC, Livesey BJ, et al. Longitudinal dynamics of clonal hematopoiesis identifies gene-specific fitness effects. Nat Med. 2022;28(7):1439–46.
Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.
Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126(1):9–16.
Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–98.
Jaiswal S, Ebert BL. Clonal hematopoiesis in human aging and disease. Science. 2019;366(6465):eaan4673.
Hoermann G, Greiner G, Griesmacher A, Valent P. Clonal hematopoiesis of indeterminate potential: a multidisciplinary challenge in personalized hematology. J Pers Med. 2020;10(3):94.
Dorsheimer L, Assmus B, Rasper T, Ortmann CA, Ecke A, Abou-El-Ardat K, et al. Association of mutations contributing to clonal hematopoiesis with prognosis in chronic ischemic heart failure. JAMA Cardiol. 2019;4(1):25–33.
Li J, Wang C, Liu J, Yu Y, Liu Y, Peng Q, et al. A feedback loop: interactions between inflammatory signals and clonal hematopoiesis in cardiovascular disease. Mol Biol Rep. 2021;48(4):3785–98.
Lin AE, Bapat AC, Xiao L, Niroula A, Ye J, Wong WJ, et al. Clonal hematopoiesis of indeterminate potential with loss of TET2 enhances risk for atrial fibrillation through NLRP3 inflammasome activation. Circulation. 2024;149(18):1419–34.
Dawoud AAZ, Tapper WJ, Cross NCP. Clonal myelopoiesis in the UK biobank cohort: ASXL1 mutations are strongly associated with smoking. Leukemia. 2020;34(10):2660–72.
Bick AG, Weinstock JS, Nandakumar SK, Fulco CP, Bao EL, Zekavat SM, et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature. 2020;586(7831):763–8.
Miller PG, Qiao D, Rojas-Quintero J, Honigberg MC, Sperling AS, Gibson CJ, et al. Association of clonal hematopoiesis with chronic obstructive pulmonary disease. Blood. 2022;139(3):357–68.
Schleicher WE, Hoag B, De Dominici M, DeGregori J, Pietras EM. CHIP: a clonal odyssey of the bone marrow niche. J Clin Invest. 2024;134(15):e180068.
Bianchini M, Möller-Ramon Z, Weber C, Megens RTA, Duchêne J. Short-term western diet causes rapid and lasting alterations of bone marrow physiology. Thromb Haemost. 2023;123(11):1100–4.
Bolton KL, Ptashkin RN, Gao T, Braunstein L, Devlin SM, Kelly D, et al. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat Genet. 2020;52(11):1219–26.
Coombs CC, Zehir A, Devlin SM, Kishtagari A, Syed A, Jonsson P, et al. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell. 2017;21(3):374-82.e4.
Wong TN, Miller CA, Jotte MRM, Bagegni N, Baty JD, Schmidt AP, et al. Cellular stressors contribute to the expansion of hematopoietic clones of varying leukemic potential. Nat Commun. 2018;9(1):455.
Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017;377(2):111–21.
Xie X, Su M, Ren K, Ma X, Lv Z, Li Z, et al. Clonal hematopoiesis and bone marrow inflammation. Transl Res. 2023;255:159–70.
Hemmati S, Haque T, Gritsman K. Inflammatory signaling pathways in preleukemic and leukemic stem cells. Front Oncol. 2017;7:265.
Greenfield G, McPherson S, Smith J, Mead A, Harrison C, Mills K, et al. Modification of the histone landscape with Jak inhibition in myeloproliferative neoplasms. Cancers. 2020;12(9):2669.
Ogura Y, Mimura I. Epigenetic roles in clonal hematopoiesis and aging kidney-related chronic kidney disease. Front Cell Dev Biol. 2023;11:1281850.
Gumuser ED, Schuermans A, Cho SMJ, Sporn ZA, Uddin MM, Paruchuri K, et al. Clonal hematopoiesis of indeterminate potential predicts adverse outcomes in patients with atherosclerotic cardiovascular disease. J Am Coll Cardiol. 2023;81(20):1996–2009.
Agrawal M, Niroula A, Cunin P, McConkey M, Shkolnik V, Kim PG, et al. TET2-mutant clonal hematopoiesis and risk of gout. Blood. 2022;140(10):1094–103.
Tobias DK, Manning AK, Wessel J, Raghavan S, Westerman KE, Bick AG, et al. Clonal hematopoiesis of indeterminate potential (CHIP) and incident type 2 diabetes risk. Diabetes Care. 2023;46(11):1978–85.
Hu Z, Yang J, Zhang S, Li M, Zuo C, Mao C, et al. AAV-mediated carboxyl terminus of Hsp70 interacting protein overexpression mitigates the cognitive and pathological phenotypes of APP/PS1 mice. Neural Regen Res. 2025;20(1):253–64.
Vlasschaert C, Robinson-Cohen C, Chen J, Akwo E, Parker AC, Silver SA, et al. Clonal hematopoiesis of indeterminate potential is associated with acute kidney injury. Nat Med. 2024;30(3):810–7.
Wong WJ, Emdin C, Bick AG, Zekavat SM, Niroula A, Pirruccello JP, et al. Clonal haematopoiesis and risk of chronic liver disease. Nature. 2023;616(7958):747–54.
Bick AG, Popadin K, Thorball CW, Uddin MM, Zanni MV, Yu B, et al. Increased prevalence of clonal hematopoiesis of indeterminate potential amongst people living with HIV. Sci Rep. 2022;12(1):577.
Bernstein N, Spencer Chapman M, Nyamondo K, Chen Z, Williams N, Mitchell E, et al. Analysis of somatic mutations in whole blood from 200,618 individuals identifies pervasive positive selection and novel drivers of clonal hematopoiesis. Nat Genet. 2024;56(6):1147–55.
Miles LA, Bowman RL, Merlinsky TR, Csete IS, Ooi AT, Durruthy-Durruthy R, et al. Single-cell mutation analysis of clonal evolution in myeloid malignancies. Nature. 2020;587(7834):477–82.
Yang Y, Dai Y, Yang X, Wu S, Wang Y. DNMT3A mutation-induced CDK1 overexpression promotes leukemogenesis by modulating the interaction between EZH2 and DNMT3A. Biomolecules. 2021;11(6):781.
Koya J, Kataoka K, Sato T, Bando M, Kato Y, Tsuruta-Kishino T, et al. DNMT3A R882 mutants interact with polycomb proteins to block haematopoietic stem and leukaemic cell differentiation. Nat Commun. 2016;7:10924.
Anteneh H, Fang J, Song J. Structural basis for impairment of DNA methylation by the DNMT3A R882H mutation. Nat Commun. 2020;11(1):2294.
Challen GA, Sun D, Jeong M, Luo M, Jelinek J, Berg JS, et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet. 2011;44(1):23–31.
Mack TM, Raddatz MA, Pershad Y, Nachun DC, Taylor KD, Guo X, et al. Epigenetic and proteomic signatures associate with clonal hematopoiesis expansion rate. Nat Aging. 2024;4(8):1043–52.
Koh KP, Yabuuchi A, Rao S, Huang Y, Cunniff K, Nardone J, et al. Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell. 2011;8(2):200–13.
Rasmussen KD, Jia G, Johansen JV, Pedersen MT, Rapin N, Bagger FO, et al. Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes Dev. 2015;29(9):910–22.
Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355(6327):842–7.
Li S, Feng J, Wu F, Cai J, Zhang X, Wang H, et al. TET2 promotes anti-tumor immunity by governing G-MDSCs and CD8(+) T-cell numbers. EMBO Rep. 2020;21(10):e49425.
Pan W, Zhu S, Qu K, Meeth K, Cheng J, He K, et al. The DNA methylcytosine dioxygenase tet2 sustains immunosuppressive function of tumor-infiltrating myeloid cells to promote melanoma progression. Immunity. 2017;47(2):284-97.e5.
Yue X, Lio CJ, Samaniego-Castruita D, Li X, Rao A. Loss of TET2 and TET3 in regulatory T cells unleashes effector function. Nat Commun. 2019;10(1):2011.
Gelsi-Boyer V, Trouplin V, Adélaïde J, Bonansea J, Cervera N, Carbuccia N, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800.
Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20(7):1126–67.
Fujino T, Goyama S, Sugiura Y, Inoue D, Asada S, Yamasaki S, et al. Mutant ASXL1 induces age-related expansion of phenotypic hematopoietic stem cells through activation of Akt/mTOR pathway. Nat Commun. 2021;12(1):1826.
Nagase R, Inoue D, Pastore A, Fujino T, Hou HA, Yamasaki N, et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J Exp Med. 2018;215(6):1729–47.
Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011;364(26):2496–506.
Metzeler KH, Becker H, Maharry K, Radmacher MD, Kohlschmidt J, Mrózek K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category. Blood. 2011;118(26):6920–9.
Chou WC, Huang HH, Hou HA, Chen CY, Tang JL, Yao M, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94.
Ni Y, Low JT, Silke J, O’Reilly LA. Digesting the role of JAK-STAT and cytokine signaling in oral and gastric cancers. Front Immunol. 2022;13:835997.
Shi Y, Zhang Z, Qu X, Zhu X, Zhao L, Wei R, et al. Roles of STAT3 in leukemia (review). Int J Oncol. 2018;53(1):7–20.
Libby P, Molinaro R, Sellar RS, Ebert BL. Jak-ing up the plaque’s lipid core…and even more. Circ Res. 2018;123(11):1180–2.
Diz-Küçükkaya R, İyigün T, Albayrak Ö, Eker C, Günel T. JAK2V617F mutation in endothelial cells of patients with atherosclerotic carotid disease. Turk J Haematol. 2024;41(3):167–74.
Leiva O, Hobbs G, Ravid K, Libby P. Cardiovascular disease in myeloproliferative neoplasms: JACC: cardiooncology state-of-the-art review. JACC CardioOncol. 2022;4(2):166–82.
Verstovsek S, Krečak I, Heidel FH, De Stefano V, Bryan K, Zuurman MW, et al. Identifying patients with polycythemia vera at risk of thrombosis after hydroxyurea initiation: the polycythemia vera-advanced integrated models (PV-AIM) project. Biomedicines. 2023. https://doi.org/10.3390/biomedicines11071925.
Wolach O, Sellar RS, Martinod K, Cherpokova D, McConkey M, Chappell RJ, et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med. 2018. https://doi.org/10.1126/scitranslmed.aan8292.
Gillis NK, Ball M, Zhang Q, Ma Z, Zhao Y, Yoder SJ, et al. Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, case-control study. Lancet Oncol. 2017;18(1):112–21.
Severson EA, Riedlinger GM, Connelly CF, Vergilio JA, Goldfinger M, Ramkissoon S, et al. Detection of clonal hematopoiesis of indeterminate potential in clinical sequencing of solid tumor specimens. Blood. 2018;131(22):2501–5.
Buttigieg MM, Vlasschaert C, Bick AG, Vanner RJ, Rauh MJ. Inflammatory reprogramming of the solid tumor microenvironment by infiltrating clonal hematopoiesis is associated with adverse outcomes. Cell Rep Med. 2025;6(3):101989.
Kessler MD, Damask A, O’Keeffe S, Banerjee N, Li D, Watanabe K, et al. Common and rare variant associations with clonal haematopoiesis phenotypes. Nature. 2022;612(7939):301–9.
Wang A, Xu Y, Yu Y, Nead KT, Kim T, Xu K, et al. Clonal hematopoiesis and risk of prostate cancer in large samples of European ancestry men. Hum Mol Genet. 2023;32(3):489–95.
Pich O, Bernard E, Zagorulya M, Rowan A, Pospori C, Slama R, et al. Tumor-infiltrating clonal hematopoiesis. N Engl J Med. 2025;392(16):1594–608.
Desai P, Zhou Y, Grenet J, Handelman SK, Crispino CM, Tarbay LN, et al. Association of clonal hematopoiesis and mosaic chromosomal alterations with solid malignancy incidence and mortality. Cancer. 2024;130(22):3879–87.
Morganti S, Gibson CJ, Jin Q, Santos K, Patel A, Wilson A, et al. Prevalence, dynamics, and prognostic role of clonal hematopoiesis of indeterminate potential in patients with breast cancer. J Clin Oncol. 2024;42(31):3666–79.
Sudlow C, Gallacher J, Allen N, Beral V, Burton P, Danesh J, et al. UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015;12(3):e1001779.
Feng Y, Yuan Q, Newsome RC, Robinson T, Bowman RL, Zuniga AN, et al. Hematopoietic-specific heterozygous loss of Dnmt3a exacerbates colitis-associated colon cancer. J Exp Med. 2023. https://doi.org/10.1084/jem.20230011.
Singh J, Li N, Ashrafi E, Thao LTP, Curtis DJ, Wood EM, et al. Clonal hematopoiesis of indeterminate potential as a prognostic factor: a systematic review and meta-analysis. Blood Adv. 2024;8(14):3771–84.
Liang Y, Chiu PK, Zhu Y, Wong CY, Xiong Q, Wang L, et al. Whole-exome sequencing reveals a comprehensive germline mutation landscape and identifies twelve novel predisposition genes in Chinese prostate cancer patients. PLoS Genet. 2022;18(9):e1010373.
Akhiyat N, Lasho T, Ganji M, Toya T, Shi CX, Chen X, et al. Clonal hematopoiesis of indeterminate potential is associated with coronary microvascular dysfunction in early non-obstructive coronary artery disease. Arterioscler Thromb Vasc Biol. 2023;43(5):774–83.
Schuermans A, Vlasschaert C, Nauffal V, Cho SMJ, Uddin MM, Nakao T, et al. Clonal haematopoiesis of indeterminate potential predicts incident cardiac arrhythmias. Eur Heart J. 2024;45(10):791–805.
Zekavat SM, Viana-Huete V, Matesanz N, Jorshery SD, Zuriaga MA, Uddin MM, et al. TP53-mediated clonal hematopoiesis confers increased risk for incident atherosclerotic disease. Nat Cardiovasc Res. 2023;2:144–58.
Büttner P, Böttner J, Krohn K, Baber R, Platzbecker U, Cross M, et al. Clonal hematopoiesis mutations are present in atherosclerotic lesions in peripheral artery disease. Int J Mol Sci. 2023. https://doi.org/10.3390/ijms24043962.
Vlasschaert C, McNaughton AJM, Chong M, Cook EK, Hopman W, Kestenbaum B, et al. Association of clonal hematopoiesis of indeterminate potential with worse kidney function and anemia in two cohorts of patients with advanced chronic kidney disease. J Am Soc Nephrol. 2022;33(5):985–95.
Fuster JJ, Zuriaga MA, Zorita V, MacLauchlan S, Polackal MN, Viana-Huete V, et al. TET2-loss-of-function-driven clonal hematopoiesis exacerbates experimental insulin resistance in aging and obesity. Cell Rep. 2020;33(4):108326.
Kim PG, Niroula A, Shkolnik V, McConkey M, Lin AE, Słabicki M, et al. Dnmt3a-mutated clonal hematopoiesis promotes osteoporosis. J Exp Med. 2021;218(12):e20211872.
Kuhnert S, Mansouri S, Rieger MA, Savai R, Avci E, Díaz-Piña G, et al. Association of clonal hematopoiesis of indeterminate potential with inflammatory gene expression in patients with COPD. Cells. 2022;11(13):2121.
Rocco JM, Zhou Y, Liu NS, Laidlaw E, Galindo F, Anderson MV, et al. Clonal hematopoiesis in people with advanced HIV and associated inflammatory syndromes. JCI Insight. 2024;9(9):e174783.
Schenz J, Rump K, Siegler BH, Hemmerling I, Rahmel T, Thon JN, et al. Increased prevalence of clonal hematopoiesis of indeterminate potential in hospitalized patients with COVID-19. Front Immunol. 2022;13:968778.
Bolton KL, Koh Y, Foote MB, Im H, Jee J, Sun CH, et al. Clonal hematopoiesis is associated with risk of severe COVID-19. Nat Commun. 2021;12(1):5975.
Liu X, Xue H, Wirdefeldt K, Song H, Smedby K, Fang F, et al. Clonal hematopoiesis of indeterminate potential and risk of neurodegenerative diseases. J Intern Med. 2024;296(4):327–35.
Wang H, Divaris K, Pan B, Li X, Lim JH, Saha G, et al. Clonal hematopoiesis driven by mutated DNMT3A promotes inflammatory bone loss. Cell. 2024;187(14):3690-711.e19.
Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021;12(1):11.
Boucai L, Falcone J, Ukena J, Coombs CC, Zehir A, Ptashkin R, et al. Radioactive iodine-related clonal hematopoiesis in thyroid cancer is common and associated with decreased survival. J Clin Endocrinol Metab. 2018;103(11):4216–23.
Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka HM, et al. International consensus classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data. Blood. 2022;140(11):1200–28.
Guilatco AJ, Shah MV, Weivoda MM. Senescence in the bone marrow microenvironment: a driver in development of therapy-related myeloid neoplasms. J Bone Oncol. 2024;47:100620.
Guarnera L, Pascale MR, Hajrullaj H, Cristiano A, Mallegni F, Onorato A, et al. The role of clonal progression leading to the development of therapy-related myeloid neoplasms. Ann Hematol. 2024;103(9):3507–17.
Malcikova J, Stano-Kozubik K, Tichy B, Kantorova B, Pavlova S, Tom N, et al. Detailed analysis of therapy-driven clonal evolution of TP53 mutations in chronic lymphocytic leukemia. Leukemia. 2015;29(4):877–85.
Lazarian G, Cymbalista F, Baran-Marszak F. Impact of low-burden TP53 mutations in the management of CLL. Front Oncol. 2022;12:841630.
Arisi MF, Dotan E, Fernandez SV. Circulating tumor DNA in precision oncology and its applications in colorectal cancer. Int J Mol Sci. 2022. https://doi.org/10.3390/ijms23084441.
Hallermayr A, Keßler T, Steinke-Lange V, Heitzer E, Holinski-Feder E, Speicher M. The utility of liquid biopsy in clinical genetic diagnosis of cancer and monogenic mosaic disorders. Med Genet. 2023;35(4):275–84.
Jensen K, Konnick EQ, Schweizer MT, Sokolova AO, Grivas P, Cheng HH, et al. Association of clonal hematopoiesis in DNA repair genes with prostate cancer plasma cell-free DNA testing interference. JAMA Oncol. 2021;7(1):107–10.
Ococks E, Frankell AM, Masque Soler N, Grehan N, Northrop A, Coles H, et al. Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann Oncol. 2021;32(4):522–32.
Chan HT, Nagayama S, Chin YM, Otaki M, Hayashi R, Kiyotani K, et al. Clinical significance of clonal hematopoiesis in the interpretation of blood liquid biopsy. Mol Oncol. 2020;14(8):1719–30.
Marshall CH, Gondek LP, Luo J, Antonarakis ES. Clonal hematopoiesis of indeterminate potential in patients with solid tumor malignancies. Cancer Res. 2022;82(22):4107–13.
Semenkovich NP, Szymanski JJ, Earland N, Chauhan PS, Pellini B, Chaudhuri AA. Genomic approaches to cancer and minimal residual disease detection using circulating tumor DNA. J Immunother Cancer. 2023. https://doi.org/10.1136/jitc-2022-006284.
Gurnari C, Fabiani E, Falconi G, Travaglini S, Ottone T, Cristiano A, et al. From clonal hematopoiesis to therapy-related myeloid neoplasms: the silent way of cancer progression. Biology. 2021;10(2):128.
Weitzel JN, Chao EC, Nehoray B, Van Tongeren LR, LaDuca H, Blazer KR, et al. Somatic TP53 variants frequently confound germ-line testing results. Genet Med. 2018;20(8):809–16.
Schaeffer EM, Srinivas S, Adra N, An Y, Bitting R, Chapin B, et al. NCCN guidelines® insights: prostate cancer, version 3.2024. J Natl Compr Canc Netw. 2024;22(3):140–50.
Shi C, Aboumsallem JP, Suthahar N, de Graaf AO, Jansen JH, van Zeventer IA, et al. Clonal haematopoiesis of indeterminate potential: associations with heart failure incidence, clinical parameters and biomarkers. Eur J Heart Fail. 2023;25(1):4–13.
Libby P, Sidlow R, Lin AE, Gupta D, Jones LW, Moslehi J, et al. Clonal hematopoiesis: crossroads of aging, cardiovascular disease, and cancer: JACC review topic of the week. J Am Coll Cardiol. 2019;74(4):567–77.
Reiner AP, Roberts MB, Honigberg MC, Kooperberg C, Desai P, Bick AG, et al. Association of clonal hematopoiesis of indeterminate potential with incident heart failure with preserved ejection fraction. medRxiv. 2023;06.07.23291038.
Böhme M, Desch S, Rosolowski M, Scholz M, Krohn K, Büttner P, et al. Impact of clonal hematopoiesis in patients with cardiogenic shock complicating acute myocardial infarction. J Am Coll Cardiol. 2022;80(16):1545–56.
Scolari FL, Abelson S, Brahmbhatt DH, Medeiros JJF, Fan CS, Fung NL, et al. Clonal haematopoiesis is associated with higher mortality in patients with cardiogenic shock. Eur J Heart Fail. 2022;24(9):1573–82.
de Boer RA, Meijers WC, van der Meer P, van Veldhuisen DJ. Cancer and heart disease: associations and relations. Eur J Heart Fail. 2019;21(12):1515–25.
Marnell CS, Bick A, Natarajan P. Clonal hematopoiesis of indeterminate potential (CHIP): linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J Mol Cell Cardiol. 2021;161:98–105.
Papa V, Marracino L, Fortini F, Rizzo P, Campo G, Vaccarezza M, et al. Translating evidence from clonal hematopoiesis to cardiovascular disease: a systematic review. J Clin Med. 2020;9(8):2480.
Kumar P, Kopecky SL, Yang EH, Oren O. Clonal hematopoiesis of indeterminate potential and cardiovascular disease. Curr Oncol Rep. 2020;22(9):87.
Gerecke C, Egea Rodrigues C, Homann T, Kleuser B. The role of ten-eleven translocation proteins in inflammation. Front Immunol. 2022;13:861351.
Sano S, Oshima K, Wang Y, MacLauchlan S, Katanasaka Y, Sano M, et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1β/NLRP3 inflammasome. J Am Coll Cardiol. 2018;71(8):875–86.
Dhimolea E. Canakinumab. MAbs. 2010;2(1):3–13.
Svensson EC, Madar A, Campbell CD, He Y, Sultan M, Healey ML, et al. TET2-driven clonal hematopoiesis and response to canakinumab: an exploratory analysis of the cantos randomized clinical trial. JAMA Cardiol. 2022;7(5):521–8.
Belizaire R, Wong WJ, Robinette ML, Ebert BL. Clonal haematopoiesis and dysregulation of the immune system. Nat Rev Immunol. 2023;23(9):595–610.
Abplanalp WT, Mas-Peiro S, Cremer S, John D, Dimmeler S, Zeiher AM. Association of clonal hematopoiesis of indeterminate potential with inflammatory gene expression in patients with severe degenerative aortic valve stenosis or chronic postischemic heart failure. JAMA Cardiol. 2020;5(10):1170–5.
Huang J, Xie J, Wang Y, Sheng M, Sun Y, Chen P, et al. STING mediates increased self-renewal and lineage skewing in DNMT3A-mutated hematopoietic stem/progenitor cells. Leukemia. 2025;39(4):929–41.
Steyers CM 3rd, Miller FJ Jr. Endothelial dysfunction in chronic inflammatory diseases. Int J Mol Sci. 2014;15(7):11324–49.
Dijkshoorn B, Raadsen R, Nurmohamed MT. Cardiovascular disease risk in rheumatoid arthritis anno 2022. J Clin Med. 2022;11(10):2704.
Shi Y, Zhang H, Huang S, Yin L, Wang F, Luo P, et al. Epigenetic regulation in cardiovascular disease: mechanisms and advances in clinical trials. Signal Transduct Target Ther. 2022;7(1):200.
Shumliakivska M, Luxán G, Hemmerling I, Scheller M, Li X, Müller-Tidow C, et al. DNMT3A clonal hematopoiesis-driver mutations induce cardiac fibrosis by paracrine activation of fibroblasts. Nat Commun. 2024;15(1):606.
Zhang X, Su J, Jeong M, Ko M, Huang Y, Park HJ, et al. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat Genet. 2016;48(9):1014–23.
Sano S, Oshima K, Wang Y, Katanasaka Y, Sano M, Walsh K. CRISPR-mediated gene editing to assess the roles of Tet2 and Dnmt3a in clonal hematopoiesis and cardiovascular disease. Circ Res. 2018;123(3):335–41.
Rauch PJ, Gopakumar J, Silver AJ, Nachun D, Ahmad H, McConkey M, et al. Loss-of-function mutations in Dnmt3a and Tet2 lead to accelerated atherosclerosis and concordant macrophage phenotypes. Nat Cardiovasc Res. 2023;2(9):805–18.
Castillo JJ, Mull N, Reagan JL, Nemr S, Mitri J. Increased incidence of non-Hodgkin lymphoma, leukemia, and myeloma in patients with diabetes mellitus type 2: a meta-analysis of observational studies. Blood. 2012;119(21):4845–50.
Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–6.
Pasupuleti SK, Ramdas B, Burns SS, Palam LR, Kanumuri R, Kumar R, et al. Obesity-induced inflammation exacerbates clonal hematopoiesis. J Clin Invest. 2023;133(11):e163968.
Andersson-Assarsson JC, van Deuren RC, Kristensson FM, Steehouwer M, Sjöholm K, Svensson PA, et al. Evolution of age-related mutation-driven clonal haematopoiesis over 20 years is associated with metabolic dysfunction in obesity. EBioMedicine. 2023;92:104621.
Khoo CM, Sairazi S, Taslim S, Gardner D, Wu Y, Lee J, et al. Ethnicity modifies the relationships of insulin resistance, inflammation, and adiponectin with obesity in a multiethnic Asian population. Diabetes Care. 2011;34(5):1120–6.
Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.
Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol. 2020;17(3):137–44.
Abplanalp WT, Cremer S, John D, Hoffmann J, Schuhmacher B, Merten M, et al. Clonal hematopoiesis-driver DNMT3A mutations alter immune cells in heart failure. Circ Res. 2021;128(2):216–28.
Kim MJ, Song H, Koh Y, Lee H, Park HE, Choi SH, et al. Clonal hematopoiesis as a novel risk factor for type 2 diabetes mellitus in patients with hypercholesterolemia. Front Public Health. 2023;11:1181879.
Bonnefond A, Skrobek B, Lobbens S, Eury E, Thuillier D, Cauchi S, et al. Association between large detectable clonal mosaicism and type 2 diabetes with vascular complications. Nat Genet. 2013;45(9):1040–3.
Denicolò S, Vogi V, Keller F, Thöni S, Eder S, Heerspink HJL, et al. Clonal hematopoiesis of indeterminate potential and diabetic kidney disease: a nested case-control study. Kidney Int Rep. 2022;7(4):876–88.
Oh TJ, Song H, Koh Y, Choi SH. The presence of clonal hematopoiesis is negatively associated with diabetic peripheral neuropathy in type 2 diabetes. Endocrinol Metab. 2022;37(2):243–8.
Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017;23(9):1072–9.
Cook EK, Izukawa T, Young S, Rosen G, Jamali M, Zhang L, et al. Comorbid and inflammatory characteristics of genetic subtypes of clonal hematopoiesis. Blood Adv. 2019;3(16):2482–6.
Bick AG, Pirruccello JP, Griffin GK, Gupta N, Gabriel S, Saleheen D, et al. Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation. 2020;141(2):124–31.
Ponzetti M, Rucci N. Updates on osteoimmunology: what’s new on the cross-talk between bone and immune system. Front Endocrinol. 2019;10:236.
Winter S, Götze KS, Hecker JS, Metzeler KH, Guezguez B, Woods K, et al. Clonal hematopoiesis and its impact on the aging osteo-hematopoietic niche. Leukemia. 2024;38(5):936–46.
Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–41.
Major TJ, Takei R, Matsuo H, Leask MP, Sumpter NA, Topless RK, et al. A genome-wide association analysis reveals new pathogenic pathways in gout. Nat Genet. 2024;56(11):2392–406.
Kestenbaum B, Bick AG, Vlasschaert C, Rauh MJ, Lanktree MB, Franceschini N, et al. Clonal hematopoiesis of indeterminate potential and kidney function decline in the general population. Am J Kidney Dis. 2023;81(3):329–35.
Castiglione M, Jiang YP, Mazzeo C, Lee S, Chen JS, Kaushansky K, et al. Endothelial JAK2V617F mutation leads to thrombosis, vasculopathy, and cardiomyopathy in a murine model of myeloproliferative neoplasm. J Thromb Haemost. 2020;18(12):3359–70.
Leung N, Bridoux F, Nasr SH. Monoclonal gammopathy of renal significance. N Engl J Med. 2021;384(20):1931–41.
Etgen T, Sander D, Chonchol M, Briesenick C, Poppert H, Förstl H, et al. Chronic kidney disease is associated with incident cognitive impairment in the elderly: the INVADE study. Nephrol Dial Transplant. 2009;24(10):3144–50.
Xiao C, Tamura MK, Pan Y, Rao V, Missikpode C, Vlasschaert C, et al. Clonal hematopoiesis of indeterminate potential is associated with reduced risk of cognitive impairment in patients with chronic kidney disease. Alzheimers Dement. 2024;20(10):6960–71.
Bouzid H, Belk JA, Jan M, Qi Y, Sarnowski C, Wirth S, et al. Clonal hematopoiesis is associated with protection from Alzheimer’s disease. Nat Med. 2023;29(7):1662–70.
Weiner DE, Scott TM, Giang LM, Agganis BT, Sorensen EP, Tighiouart H, et al. Cardiovascular disease and cognitive function in maintenance hemodialysis patients. Am J Kidney Dis. 2011;58(5):773–81.
Kislikova M, Lopez MAB, Salinas FJF, Blanco JAP, Molina MPG, Fernandez AA, et al. Clonal hematopoiesis of indeterminate potential and cardiovascular risk in patients with chronic kidney disease without previous cardiac pathology. Life. 2023;13(9):1801.
Nistala R, Whaley-Connell A, Sowers JR. Redox control of renal function and hypertension. Antioxid Redox Signal. 2008;10(12):2047–89.
Silver AJ, Vlasschaert C, Mack T, Sharber B, Xu Y, Bick AG, et al. Solid organ transplant recipients exhibit more TET2-mutant clonal hematopoiesis of indeterminate potential not driven by increased transplantation risk. Clin Cancer Res. 2024;30(11):2475–85.
Huang Z, Vlasschaert C, Robinson-Cohen C, Pan Y, Sun X, Lash JP, et al. Emerging evidence on the role of clonal hematopoiesis of indeterminate potential in chronic kidney disease. Transl Res. 2023;256:87–94.
Vlasschaert C, Rauh MJ, Lanktree MB. Response to: “clonal hematopoiesis of indeterminate potential and diabetic kidney disease: a nested case-control study.” Kidney Int Rep. 2022;7(11):2543.
Cantero-Navarro E, Fernández-Fernández B, Ramos AM, Rayego-Mateos S, Rodrigues-Diez RR, Sánchez-Niño MD, et al. Renin-angiotensin system and inflammation update. Mol Cell Endocrinol. 2021;529:111254.
Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–46.
Jia Y, Zheng Z, Xue M, Zhang S, Hu F, Li Y, et al. Extracellular vesicles from albumin-induced tubular epithelial cells promote the M1 macrophage phenotype by targeting klotho. Mol Ther. 2019;27(8):1452–66.
Hormaechea-Agulla D, Matatall KA, Le DT, Kain B, Long X, Kus P, et al. Chronic infection drives Dnmt3a-loss-of-function clonal hematopoiesis via IFNγ signaling. Cell Stem Cell. 2021;28(8):1428-42.e6.
Miller PG, Fell GG, Foy BH, Scherer AK, Gibson CJ, Sperling AS, et al. Clonal hematopoiesis of indeterminate potential and risk of death from COVID-19. Blood. 2022;140(18):1993–7.
Kang CK, Choi B, Kim S, Sun CH, Yoon SH, Kim K, et al. Clinical impact of clonal hematopoiesis on severe COVID-19 patients without canonical risk factors. Haematologica. 2023;108(1):257–60.
Dharan NJ, Yeh P, Bloch M, Yeung MM, Baker D, Guinto J, et al. HIV is associated with an increased risk of age-related clonal hematopoiesis among older adults. Nat Med. 2021;27(6):1006–11.
Knudsen AD, Eskelund CW, Benfield T, Zhao Y, Gelpi M, Køber L, et al. Clonal hematopoiesis of indeterminate potential in persons with HIV. AIDS. 2024;38(4):487–95.
Bhattacharya R, Uddin MM, Patel AP, Niroula A, Finneran P, Bernardo R, et al. Risk factors for clonal hematopoiesis of indeterminate potential in people with HIV: a report from the REPRIEVE trial. Blood Adv. 2024;8(4):959–67.
Duployez N, Demonchy J, Berthon C, Goutay J, Caplan M, Moreau AS, et al. Clinico-biological features and clonal hematopoiesis in patients with severe COVID-19. Cancers. 2020;12(7):1992.
Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65(8):1038–48.
Lee JK, An H, Koh Y, Lee CH. Clonal hematopoiesis of indeterminate potential is associated with current smoking status and history of exacerbation in patients with chronic obstructive pulmonary disease. Tuberc Respir Dis (Seoul). 2024;87(3):309–18.
Marchetti A, Pelusi S, Marella A, Malvestiti F, Ricchiuti A, Ronzoni L, et al. Impact of clonal hematopoiesis of indeterminate potential on hepatocellular carcinoma in individuals with steatotic liver disease. Hepatology. 2024;80(4):816–27.
Lareau SC, Fahy B, Meek P, Wang A. Chronic obstructive pulmonary disease (COPD). Am J Respir Crit Care Med. 2019;199(1):1–2.
Matatall KA, Wathan TK, Nguyen M, Chen H, McDonald A, Qi G, et al. TET2-mutant myeloid cells mitigate Alzheimer’s disease progression via CNS infiltration and enhanced phagocytosis in mice. Cell Stem Cell. 2025;32(8):1285-98.e8.
Yun J, Youn YC, Kim HR. Association between clonal hematopoiesis of indeterminate potential and brain β-amyloid deposition in Korean patients with cognitive impairment. Ann Lab Med. 2024;44(6):576–80.
Hajishengallis G. Aging and its impact on innate immunity and inflammation: implications for periodontitis. J Oral Biosci. 2014;56(1):30–7.
Ebersole JL, Graves CL, Gonzalez OA, Dawson D 3rd, Morford LA, Huja PE, et al. Aging, inflammation, immunity, and periodontal disease. Periodontol 2000. 2016;72(1):54–75.
Heyde A, Rohde D, McAlpine CS, Zhang S, Hoyer FF, Gerold JM, et al. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell. 2021;184(5):1348-61.e22.
Quiñones AR, Markwardt S, Botoseneanu A. Diabetes-multimorbidity combinations and disability among middle-aged and older adults. J Gen Intern Med. 2019;34(6):944–51.
Gajjala PR, Sanati M, Jankowski J. Cellular and molecular mechanisms of chronic kidney disease with diabetes mellitus and cardiovascular diseases as its comorbidities. Front Immunol. 2015;6:340.
Walsh K, Raghavachari N, Kerr C, Bick AG, Cummings SR, Druley T, et al. Clonal hematopoiesis analyses in clinical, epidemiologic, and genetic aging studies to unravel underlying mechanisms of age-related dysfunction in humans. Front Aging. 2022;3:841796.
Agathocleous M, Meacham CE, Burgess RJ, Piskounova E, Zhao Z, Crane GM, et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature. 2017;549(7673):476–81.
Gozdecka M, Dudek M, Wen S, Gu M, Stopforth RJ, Rak J, et al. Mitochondrial metabolism sustains DNMT3A-R882-mutant clonal haematopoiesis. Nature. 2025;642(8067):431–41.
Hosseini M, Voisin V, Chegini A, Varesi A, Cathelin S, Ayyathan DM, et al. Metformin reduces the competitive advantage of Dnmt3a(R878H) HSPCs. Nature. 2025;642(8067):421–30.