45
Views
0
CrossRef citations to date
0
Altmetric
Brief Report

Clonal hematopoiesis of indeterminate potential is rare in pediatric patients undergoing autologous stem cell transplantation

, , , , , , , & show all
Received 11 Mar 2024, Accepted 28 May 2024, Published online: 06 Jun 2024

References

  • Bick AG, Weinstock JS, Nandakumar SK, et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature. 2020;586(7831):763–768. doi:10.1038/s41586-020-2819-2.
  • Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–2498. doi:10.1056/NEJMoa1408617.
  • Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014; 371(26):2477–2487. doi:10.1056/NEJMoa1409405.
  • Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472–1478. doi:10.1038/nm.3733.
  • McKerrell T, Park N, Moreno T, et al. Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis. Cell Rep. 2015;10(8):1239–1245.
  • Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7(1):12484. doi:10.1038/ncomms12484.
  • Calvillo-Argüelles O, Jaiswal S, Shlush LI, et al. Connections between clonal hematopoiesis, cardiovascular disease, and cancer: a review. JAMA Cardiol. 2019;4(4):380–387. doi:10.1001/jamacardio.2019.0302.
  • Pardali E, Dimmeler S, Zeiher AM, et al. Clonal hematopoiesis, aging, and cardiovascular diseases. Exp Hematol. 2020;83:95–104.
  • Jaiswal S, Natarajan P, Silver AJ, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017; 377(2):111–121. doi:10.1056/NEJMoa1701719.
  • Wong TN, Miller CA, Jotte MRM, et al. Cellular stressors contribute to the expansion of hematopoietic clones of varying leukemic potential. Nat Commun. 2018;9(1):455. doi:10.1038/s41467-018-02858-0.
  • Coombs CC, Zehir A, Devlin SM, 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):374–382e4. doi:10.1016/j.stem.2017.07.010.
  • Pich O, Cortes-Bullich A, Muinos F, et al. The evolution of hematopoietic cells under cancer therapy. Nat Commun. 2021;12(1):4803. doi:10.1038/s41467-021-24858-3.
  • Bolton KL, Ptashkin RN, Gao T, et al. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat Genet. 2020;52(11):1219–1226. doi:10.1038/s41588-020-00710-0.
  • Swisher EM, Harrell MI, Norquist BM, et al. Somatic mosaic mutations in PPM1D and TP53 in the blood of women with ovarian carcinoma. JAMA Oncol. 2016;2(3):370–372. doi:10.1001/jamaoncol.2015.6053.
  • Gibson CJ, Lindsley RC, Tchekmedyian V, et al. Clonal hematopoiesis associated with adverse outcomes after autologous stem-cell transplantation for lymphoma. J Clin Oncol. 2017;35(14):1598–1605. doi:10.1200/JCO.2016.71.6712.
  • Mouhieddine TH, Sperling AS, Redd R, et al. Clonal hematopoiesis is associated with adverse outcomes in multiple myeloma patients undergoing transplant. Nat Commun. 2020;11(1):2996. doi:10.1038/s41467-020-16805-5.
  • Coorens THH, Collord G, Lu W, et al. Clonal hematopoiesis and therapy-related myeloid neoplasms following neuroblastoma treatment. Blood. 2021;137(21):2992–2997. doi:10.1182/blood.2020010150.
  • Acuna-Hidalgo R, Sengul H, Steehouwer M, et al. Ultra-sensitive sequencing identifies high prevalence of clonal hematopoiesis-associated mutations throughout adult life. Am J Hum Genet. 2017;101(1):50–64. doi:10.1016/j.ajhg.2017.05.013.
  • Collord G, Park N, Podestà M, et al. Clonal haematopoiesis is not prevalent in survivors of childhood cancer. Br J Haematol. 2018;181(4):537–539. doi:10.1111/bjh.14630.
  • Hagiwara K, Natarajan S, Wang Z, et al. Dynamics of age- versus therapy-related clonal hematopoiesis in long-term survivors of pediatric cancer. Cancer Discov. 2023;13(4):844–857. doi:10.1158/2159-8290.CD-22-0956.
  • Novetsky Friedman D, Chan ICC, Moskowitz CS, et al. Clonal hematopoiesis in survivors of childhood cancer. Blood Adv. 2023;7(15):4102–4106. doi:10.1182/bloodadvances.2023009817.
  • Takahashi K, Wang F, Kantarjian H, et al. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case–control study. Lancet Oncol. 2017;18(1):100–111.
  • Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–424. doi:10.1038/gim.2015.30.
  • Li MM, Datto M, Duncavage EJ, et al. Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer: A Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017;19(1):4–23.
  • Shlush LI. Age-related clonal hematopoiesis. Blood. 2018;131(5):496–504. doi:10.1182/blood-2017-07-746453.
  • Heini AD, Porret N, Zenhaeusern R, et al. Clonal hematopoiesis after autologous stem cell transplantation does not confer adverse prognosis in patients with AML. Cancers (Basel)). 2021;13(13):3190. doi:10.3390/cancers13133190.
  • Olivier M, Eeles R, Hollstein M, et al. The IARC TP53 database: new online mutation analysis and recommendations to users. Hum Mutat. 2002;19(6):607–614.
  • Petitjean A, Mathe E, Kato S, et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 2007;28(6):622–629.
  • Mitchell RL, Kosche C, Burgess K, et al. Misdiagnosis of Li–Fraumeni syndrome in a patient with clonal hematopoiesis and a somatic TP53 mutation. J Natl Compr Canc Netw. 2018;16(5):461–466. doi:10.6004/jnccn.2017.7058.
  • Batalini F, Peacock EG, Stobie L, et al. Li–Fraumeni syndrome: not a straightforward diagnosis anymore-the interpretation of pathogenic variants of low allele frequency and the differences between germline PVs, mosaicism, and clonal hematopoiesis. Breast Cancer Res. 2019;21(1):107. doi:10.1186/s13058-019-1193-1.
  • Weitzel JN, Chao EC, Nehoray B, et al. Somatic TP53 variants frequently confound germ-line testing results. Genet Med. 2018;20(8):809–816. doi:10.1038/gim.2017.196.
  • Vargas-Parra G, Del Valle J, Rofes P, et al. Comprehensive analysis and ACMG-based classification of CHEK2 variants in hereditary cancer patients. Hum Mutat. 2020;41(12):2128–2142.
  • Han FF, Guo CL, Liu LH. The effect of CHEK2 variant I157T on cancer susceptibility: evidence from a meta-analysis. DNA Cell Biol. 2013;32(6):329–335. doi:10.1089/dna.2013.1970.
  • Senol-Cosar O, Schmidt RJ, Qian E, et al. Considerations for clinical curation, classification, and reporting of low-penetrance and low effect size variants associated with disease risk. Genet Med. 2019;21(12):2765–2773. doi:10.1038/s41436-019-0560-8.
  • Meijers-Heijboer H, Wijnen J, Vasen H, et al. The CHEK2 1100delC mutation identifies families with a hereditary breast and colorectal cancer phenotype. Am J Hum Genet. 2003;72(5):1308–1314. doi:10.1086/375121.
  • Li J, Williams BL, Haire LF, et al. Structural and functional versatility of the FHA domain in DNA-damage signaling by the tumor suppressor kinase Chk2. Mol Cell. 2002;9(5):1045–1054. doi:10.1016/s1097-2765(02)00527-0.
  • Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555(7696):321–327. doi:10.1038/nature25480.
  • Ma X, Liu Y, Liu Y, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555(7696):371–376. doi:10.1038/nature25795.
  • Filbin M, Monje M. Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat Med. 2019;25(3):367–376. doi:10.1038/s41591-019-0383-9.
  • Young AL, Tong RS, Birmann BM, et al. Clonal hematopoiesis and risk of acute myeloid leukemia. Haematologica. 2019;104(12):2410–2417. doi:10.3324/haematol.2018.215269.
  • Abelson S, Collord G, Ng SWK, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559(7714):400–404. doi:10.1038/s41586-018-0317-6.
  • Desai P, Mencia-Trinchant N, Savenkov O, et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat Med. 2018;24(7):1015–1023. doi:10.1038/s41591-018-0081-z.
  • Wong TN, Ramsingh G, Young AL, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 2015;518(7540):552–555. doi:10.1038/nature13968.
  • Spitzer B, Rutherford KD, Gundem G, et al. Bone marrow surveillance of pediatric cancer survivors identifies clones that predict therapy-related leukemia. Clin Cancer Res. 2022;28(8):1614–1627. doi:10.1158/1078-0432.CCR-21-2451.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.