Clonal hematopoiesis in sickle cell disease

L Alexander Liggett, Liam D Cato, Joshua S Weinstock, Yingze Zhang, S Mehdi Nouraie, Mark T Gladwin, Melanie E Garrett, Allison Ashley-Koch, Marilyn J Telen, Brian Custer, Shannon Kelly, Carla L Dinardo, Ester C Sabino, Paula Loureiro, Anna B Carneiro-Proietti, Cláudia Maximo, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, Alexander P Reiner, Gonçalo R Abecasis, David A Williams, Pradeep Natarajan, Alexander G Bick, Vijay G Sankaran, L Alexander Liggett, Liam D Cato, Joshua S Weinstock, Yingze Zhang, S Mehdi Nouraie, Mark T Gladwin, Melanie E Garrett, Allison Ashley-Koch, Marilyn J Telen, Brian Custer, Shannon Kelly, Carla L Dinardo, Ester C Sabino, Paula Loureiro, Anna B Carneiro-Proietti, Cláudia Maximo, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, Alexander P Reiner, Gonçalo R Abecasis, David A Williams, Pradeep Natarajan, Alexander G Bick, Vijay G Sankaran

Abstract

BACKGROUNDCurative gene therapies for sickle cell disease (SCD) are currently undergoing clinical evaluation. The occurrence of myeloid malignancies in these trials has prompted safety concerns. Individuals with SCD are predisposed to myeloid malignancies, but the underlying causes remain undefined. Clonal hematopoiesis (CH) is a premalignant condition that also confers significant predisposition to myeloid cancers. While it has been speculated that CH may play a role in SCD-associated cancer predisposition, limited data addressing this issue have been reported.METHODSHere, we leveraged 74,190 whole-genome sequences to robustly study CH in SCD. Somatic mutation calling methods were used to assess CH in all samples and comparisons between individuals with and without SCD were performed.RESULTSWhile we had sufficient power to detect a greater than 2-fold increased rate of CH, we found no detectable variation in rate or clone properties between individuals affected by SCD and controls. The rate of CH in individuals with SCD was unaltered by hydroxyurea use.CONCLUSIONSWe did not observe an increased risk for acquiring detectable CH in SCD, at least as measured by whole-genome sequencing. These results should help guide ongoing efforts and further studies that seek to better define the risk factors underlying myeloid malignancy predisposition in SCD and help ensure that curative therapies can be more safely applied.FUNDINGNew York Stem Cell Foundation and the NIH.

Trial registration: ClinicalTrials.gov NCT00492531.

Keywords: Hematology; Leukemias.

Conflict of interest statement

Conflict of interest: MTG serves as a consultant for Actelion, Bayer Healthcare, Pfizer, Forma, and Fulcrum Therapeutics. GRA is an employee of Regeneron Pharmaceuticals and owns stock and stock options for Regeneron Pharmaceuticals. DAW serves on Novartis steering committee ETB115E2201, is a cofounder of Alerion Biosciences, chief scientific chair of Emerging Therapy Solutions, and serves as an advisor to Geneception, Insertion Site, Bluebird bio, and Beam Therapeutics. PN reports research grants from Amgen, Apple, and Boston Scientific, and is an advisor to Apple, AstraZeneca, Black Stone Life Sciences, Genexwell, Foresite Labs, Novartis, Roche/Genentech, TenSixteen Bio, and Blackstone Life Sciences. AGB serves as an advisor to and has equity in TenSixteen Bio. VGS serves as an advisor to and/or has equity in Novartis, Forma, Cellarity, Ensoma, and Branch Biosciences.

Figures

Figure 1. Prevalence of CH is similar…
Figure 1. Prevalence of CH is similar in unaffected and SCD populations.
(A) A generalized additive model was used here to fit rates of CH within WGS data from a total of 71,100 individuals unaffected by SCD and 3,090 individuals affected by SCD, which indicates no significantly increased prevalence of CH within individuals affected by SCD (OR = 1.30, P = 0.20). (B) Generalized additive model as in A was used to fit rates of CH using a genetically matched cohort without SCD. The matched cohort was created by selecting the 10 most similar individuals by the first 10 PCs for each individual in the SCD cohort. Resampling of individuals without SCD was permitted. SCD samples without sufficient matches were excluded. (C) Genes ranked by variant load across all individuals separated by SCD status into unaffected (blue) and affected (red). (D) Type of genetic change ranked by prevalence in all individuals separated by SCD status into unaffected (blue) and affected (red).
Figure 2. Mutation signatures do not vary…
Figure 2. Mutation signatures do not vary in SCD and hydroxyurea (HU) treatment does not impact the rate of CH development.
Percentages of total single-base substitutions made up by each possible 96 substitution and trinucleotide context pairs per individual (error is SD across individuals). Signatures are separated into (A) individuals without SCD and (B) individuals with SCD (data represent mean ± SD). (C) A generalized additive model is used here to fit rates of CH within WGS from individuals with SCD separated into never-HU-treated or HU-treated groups. There is no significant difference in the rate of CH in either the HU-treated or untreated groups adjusted for age, age2, sex, study, and the first 10 principal components (OR = 0.58, P = 0.23). (D) Genes ranked by variant load across all individuals with SCD separated by HU treatment status into untreated (green) and (E) treated (purple).

References

    1. DeBaun MR, et al. Decreased median survival of adults with sickle cell disease after adjusting for left truncation bias: a pooled analysis. Blood. 2019;133(6):615–617. doi: 10.1182/blood-2018-10-880575.
    1. Li Y, et al. Myeloid neoplasms in the setting of sickle cell disease: an intrinsic association with the underlying condition rather than a coincidence; report of 4 cases and review of the literature. Mod Pathol. 2019;32(12):1712–1726. doi: 10.1038/s41379-019-0325-6.
    1. Brunson A, et al. Increased risk of leukemia among sickle cell disease patients in California. Blood. 2017;130(13):1597–1599. doi: 10.1182/blood-2017-05-783233.
    1. Seminog OO, et al. Risk of individual malignant neoplasms in patients with sickle cell disease: English national record linkage study. J R Soc Med. 2016;109(8):303–309. doi: 10.1177/0141076816651037.
    1. Science. Gene Therapy Trials for Sickle Cell Disease Halted After Two Patients Develop Cancer. Updated February 16, 2021. Accessed May 7, 2021.
    1. Hsieh MM, et al. Myelodysplastic syndrome unrelated to lentiviral vector in a patient treated with gene therapy for sickle cell disease. Blood Adv. 2020;4(9):2058–2063. doi: 10.1182/bloodadvances.2019001330.
    1. Ghannam JY, et al. Baseline TP53 mutations in adults with SCD developing myeloid malignancy following hematopoietic cell transplantation. Blood. 2020;135(14):1185–1188.
    1. Bowman RL, et al. Clonal hematopoiesis and evolution to hematopoietic malignancies. Cell Stem Cell. 2018;22(2):157–170. doi: 10.1016/j.stem.2018.01.011.
    1. Jaiswal S, Ebert BL. Clonal hematopoiesis in human aging and disease. Science. 2019;366(6465):eaan4673. doi: 10.1126/science.aan4673.
    1. Jaiswal S, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–2498. doi: 10.1056/NEJMoa1408617.
    1. Abelson S, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559(7714):400–404. doi: 10.1038/s41586-018-0317-6.
    1. Desai P, 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.
    1. Taliun D, et al. Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature. 2021;590(7845):290–299. doi: 10.1038/s41586-021-03205-y.
    1. Bick AG 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.
    1. Jaiswal S, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017;377(2):111–121. doi: 10.1056/NEJMoa1701719.
    1. Laconi E, et al. Cancer as a disease of old age: changing mutational and microenvironmental landscapes. Br J Cancer. 2020;122(7):943–952. doi: 10.1038/s41416-019-0721-1.
    1. Liggett LA, Sankaran VG. Unraveling hematopoiesis through the lens of genomics. Cell. 2020;182(6):1384–1400. doi: 10.1016/j.cell.2020.08.030.
    1. Ware RE, et al. Sickle cell disease. Lancet. 2017;390(10091):311–323. doi: 10.1016/S0140-6736(17)30193-9.
    1. Ware RE, Dertinger SD. Absence of hydroxyurea-induced mutational effects supports higher utilisation for the treatment of sickle cell anaemia. Br J Haematol. 2021;194(2):252–266. doi: 10.1111/bjh.17323.
    1. Jones RJ, DeBaun MR. Leukemia after gene therapy for sickle cell disease: insertional mutagenesis, busulfan, both or neither. Blood. 2021;138(11):942–947. doi: 10.1182/blood.2021011488.
    1. Pincez T, et al. Clonal hematopoiesis in sickle cell disease. Blood. 2021;138(21):2148–2152. doi: 10.1182/blood.2021011121.
    1. Bergstrom EN, et al. SigProfilerMatrixGenerator: a tool for visualizing and exploring patterns of small mutational events. BMC Genomics. 2019;20(1):685. doi: 10.1186/s12864-019-6041-2.

Source: PubMed

3
Subscribe