Genomic profiling of a randomized trial of interferon-α vs hydroxyurea in MPN reveals mutation-specific responses

Trine Alma Knudsen, Vibe Skov, Kristen Stevenson, Lillian Werner, William Duke, Charles Laurore, Christopher J Gibson, Anwesha Nag, Aaron R Thorner, Bruce Wollison, Dennis Lund Hansen, Christina Ellervik, Daniel El Fassi, Karin de Stricker, Lukas Frans Ocias, Mette Brabrand, Ole Weis Bjerrum, Ulrik Malthe Overgaard, Mikael Frederiksen, Thomas Kielsgaard Kristensen, Torben A Kruse, Mads Thomassen, Torben Mourits-Andersen, Marianne Tang Severinsen, Jesper Stentoft, Joern Starklint, Donna S Neuberg, Lasse Kjaer, Thomas Stauffer Larsen, Hans Carl Hasselbalch, R Coleman Lindsley, Ann Mullally, Trine Alma Knudsen, Vibe Skov, Kristen Stevenson, Lillian Werner, William Duke, Charles Laurore, Christopher J Gibson, Anwesha Nag, Aaron R Thorner, Bruce Wollison, Dennis Lund Hansen, Christina Ellervik, Daniel El Fassi, Karin de Stricker, Lukas Frans Ocias, Mette Brabrand, Ole Weis Bjerrum, Ulrik Malthe Overgaard, Mikael Frederiksen, Thomas Kielsgaard Kristensen, Torben A Kruse, Mads Thomassen, Torben Mourits-Andersen, Marianne Tang Severinsen, Jesper Stentoft, Joern Starklint, Donna S Neuberg, Lasse Kjaer, Thomas Stauffer Larsen, Hans Carl Hasselbalch, R Coleman Lindsley, Ann Mullally

Abstract

Although somatic mutations influence the pathogenesis, phenotype, and outcome of myeloproliferative neoplasms (MPNs), little is known about their impact on molecular response to cytoreductive treatment. We performed targeted next-generation sequencing (NGS) on 202 pretreatment samples obtained from patients with MPN enrolled in the DALIAH trial (A Study of Low Dose Interferon Alpha Versus Hydroxyurea in Treatment of Chronic Myeloid Neoplasms; #NCT01387763), a randomized controlled phase 3 clinical trial, and 135 samples obtained after 24 months of therapy with recombinant interferon-alpha (IFNα) or hydroxyurea. The primary aim was to evaluate the association between complete clinicohematologic response (CHR) at 24 months and molecular response through sequential assessment of 120 genes using NGS. Among JAK2-mutated patients treated with IFNα, those with CHR had a greater reduction in the JAK2 variant allele frequency (median, 0.29 to 0.07; P < .0001) compared with those not achieving CHR (median, 0.27 to 0.14; P < .0001). In contrast, the CALR variant allele frequency did not significantly decline in those achieving CHR or in those not achieving CHR. Treatment-emergent mutations in DNMT3A were observed more commonly in patients treated with IFNα compared with hydroxyurea (P = .04). Furthermore, treatment-emergent DNMT3A mutations were significantly enriched in IFNα-treated patients not attaining CHR (P = .02). A mutation in TET2, DNMT3A, or ASXL1 was significantly associated with prior stroke (age-adjusted odds ratio, 5.29; 95% confidence interval, 1.59-17.54; P = .007), as was a mutation in TET2 alone (age-adjusted odds ratio, 3.03; 95% confidence interval, 1.03-9.01; P = .044). At 24 months, we found mutation-specific response patterns to IFNα: (1) JAK2- and CALR-mutated MPN exhibited distinct molecular responses; and (2) DNMT3A-mutated clones/subclones emerged on treatment.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Trial flowchart. NGS was performed on 202 primary MPN samples and 135 samples obtained 24 months after initiation of therapy with either HU or IFNα (IFNα-2a or IFNα-2b). One patient allocated to IFN-α died within 24 months.
Figure 2.
Figure 2.
Genomic profiling of somatic mutations in baseline samples by NGS (comutation plot). Each column represents 1 patient (n = 202), and the rows represent different somatic mutations. The VAF for each phenotypic driver mutation is color coded. The frequency of specific somatic mutations is listed on the right border of the figure. Somatic mutations in 34 different genes were detected in 191 (95%) patients, including 92% with MPN phenotypic driver mutations: JAK2, 74%; CALR, 14%; or MPL, 5%. JAK2-UPD was observed in 28% and was significantly associated with PV (Kruskal-Wallis test, P < .0001). The most frequent concomitant mutations affected 3 genes: TET2 (24%), DNMT3A (16%), and ASXL1 (10%). 9p-UPD, uniparental disomy of chromosome 9p.
Figure 3.
Figure 3.
Complete CHR response at 24 months. (A) Proportion of patients with complete CHR over time according to treatment group. Median time to CHR was 5.7 months (IQR, 1.8-10.5 months) for HU, and 4.9 months (IQR, 2.1-8.9 months) and 6.0 months (IQR, 1.8-10.1 months) for patients treated with IFNα-2a or IFNα-2b, respectively. The CHR rate reached a maximum after 12 months among patients treated with HU (47%), whereas the CHR rate increased almost gradually over time among patients treated with IFNα-2a or IFNα-2b. (B) Proportion of patients with CHR at 24 months according to treatment group. CHR was achieved in 8 of 38 (21%; 95% CI, 10-37) patients treated with HU, 25 of 82 (30%; 95% CI, 21-42) patients treated with IFNα-2a, and in 17 of 82 (17%; 95% CI, 13-31) patients treated with IFNα-2b. No significant difference in the CHR rate was detected between HU and the two IFNα groups combined. Patients considered nonevaluable (NE) at 24 months had all discontinued the study therapy to which they were allocated, except 3 patients in whom complete diagnostic workup was not available at 24 months (HU, n = 1; IFNα-2a, n = 2). Error bars are 95% CI upper limits.
Figure 4.
Figure 4.
Treatment-emergent mutations at 24 months. (A) Number of treatment-emergent mutations at 24 months. Thirty-eight treatment-emergent mutations were detected in 32 patients, 4 of whom had discontinued treatment. Mutations were defined as treatment-emergent if: (1) the VAF was <0.01 in the baseline sample and ≥0.01 in the 24-month sample (n = 36); or (2) if the VAF was ≥0.01 in the baseline sample and had a more than fourfold increase in the 24-month sample (n = 2). The most frequent treatment-emergent mutations were detected in DNMT3A (n = 15 [39%]), followed by TET2 (n = 4 [11%]), ASXL1 (n = 3 [8%]), PPM1D (n = 3 [8%]), and TP53 (n = 3 [8%]). (B) VAF of treatment-emergent mutations at baseline and posttreatment at 24 months. The median VAF of treatment-emergent mutations was low (median, 1.5%) and primarily occurred in JAK2-mutated patients (97%). Representative examples of treatment-emergent mutations detected in patients treated with HU (C) and IFNα (D). MPN phenotypic driver mutations are depicted with blue lines, treatment-emergent concomitant mutations with red lines, and other concomitant mutations with black lines. The upper rows of panels C and D also show uniparental disomy of chromosome 9p (9p-UPD) analysis. In all examples, 9p-UPD is no longer detectable posttreatment at 24 months, which is concordant with the decrease in mutant JAK2 VAF.
Figure 5.
Figure 5.
Association between somatic mutations and complete CHR on serial sampling. (A-B) Molecular response among patients allocated to IFNα at baseline (Pre) and at 24 months (Post) of treatment by complete CHR. (A) Among JAK2-mutated patients, those attaining CHR at 24 months had a greater reduction in the JAK2 VAF (median, 0.29 to 0.07; P < .0001) compared with JAK2-mutated patients who did not achieve CHR (median, 0.27 to 0.14; P < .0001). (B) The CALR VAF did not significantly decline among patients achieving CHR nor among those not achieving CHR at 24 months. The middle horizontal lines indicate the median value; box limits indicate the 5th and 95th percentiles, and whiskers indicate the range. All observations are represented by a dot. (C) Number of patients with no treatment-emergent mutations. Significantly more patients with no treatment-emergent mutations treated with IFNα achieved CHR (35 of 68 [51%]) compared with patients treated with HU (4 of 18 [22%]) (P = .03). (D) Number of patients with treatment-emergent mutations. No difference in the number of patients failing to achieve CHR was observed between patients treated with HU (10 of 13 [77%]) or IFNα (9 of 14 [64%]) (P = .68). (E) Number of patients with treatment-emergent DNMT3A mutations. (F) Number of patients with treatment-emergent non-DNMT3A mutations. Treatment-emergent DNMT3A mutations were significantly enriched in patients treated with IFNα failing to achieve CHR (8 of 9 [89%]) compared with treatment-emergent non-DNMT3A mutations (1 of 5 [20%]) (P = .02).

References

    1. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372-375.
    1. Arber DA, Orazi A, Hasserjian R, et al. . The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391-2405.
    1. Hultcrantz M, Kristinsson SY, Andersson TML, et al. . Patterns of survival among patients with myeloproliferative neoplasms diagnosed in Sweden from 1973 to 2008: a population-based study. J Clin Oncol. 2012;30(24):2995-3001.
    1. Kaifie A, Kirschner M, Wolf D, et al. ; Study Alliance Leukemia (SAL) . Bleeding, thrombosis, and anticoagulation in myeloproliferative neoplasms (MPN): analysis from the German SAL-MPN-registry. J Hematol Oncol. 2016;9(1):18.
    1. Hulegårdh E, Nilsson C, Lazarevic V, et al. . Characterization and prognostic features of secondary acute myeloid leukemia in a population-based setting: a report from the Swedish Acute Leukemia Registry. Am J Hematol. 2015;90(3):208-214.
    1. Mead AJ, Mullally A. Myeloproliferative neoplasm stem cells. Blood. 2017;129(12):1607-1616.
    1. Marneth AE, Mullally A. The molecular genetics of myeloproliferative neoplasms. Cold Spring Harb Perspect Med. 2020;10(2):a034876.
    1. Baxter EJ, Scott LM, Campbell PJ, et al. ; Cancer Genome Project . Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054-1061.
    1. James C, Ugo V, Le Couédic JP, et al. . A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144-1148.
    1. Kralovics R, Passamonti F, Buser AS, et al. . A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779-1790.
    1. Levine RL, Wadleigh M, Cools J, et al. . Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387-397.
    1. Scott LM, Tong W, Levine RL, et al. . JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459-468.
    1. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, et al. . Whole-exome sequencing identifies novel MPL and JAK2 mutations in triple-negative myeloproliferative neoplasms. Blood. 2016;127(3):325-332.
    1. Angona A, Fernández-Rodríguez C, Alvarez-Larrán A, et al. . Molecular characterisation of triple negative essential thrombocythaemia patients by platelet analysis and targeted sequencing. Blood Cancer J. 2016;6(8):e463.
    1. Skov V. Next generation sequencing in MPNs. Lessons from the past and prospects for use as predictors of prognosis and treatment responses. Cancers (Basel). 2020;12(8):1-38.
    1. Lundberg P, Karow A, Nienhold R, et al. . Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood. 2014; 123(14):2220-2228.
    1. Grinfeld J, Nangalia J, Baxter EJ, et al. . Classification and personalized prognosis in myeloproliferative neoplasms. N Engl J Med. 2018;379(15): 1416-1430.
    1. Vannucchi AM, Lasho TL, Guglielmelli P, et al. . Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27(9):1861-1869.
    1. Guglielmelli P, Lasho TL, Rotunno G, et al. . The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients. Leukemia. 2014;28(9):1804-1810.
    1. Tefferi A, Guglielmelli P, Lasho TL, et al. . Mutation-enhanced international prognostic systems for essential thrombocythaemia and polycythaemia vera. Br J Haematol. 2020;189(2):291-302.
    1. Ortmann CA, Kent DG, Nangalia J, et al. . Effect of mutation order on myeloproliferative neoplasms. N Engl J Med. 2015;372(7):601-612.
    1. Cortelazzo S, Finazzi G, Ruggeri M, et al. . Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med. 1995;332(17):1132-1136.
    1. Fruchtman SM, Mack K, Kaplan ME, Peterson P, Berk PD, Wasserman LR. From efficacy to safety: a Polycythemia Vera Study group report on hydroxyurea in patients with polycythemia vera. Semin Hematol. 1997;34(1):17-23.
    1. Harrison CN, Campbell PJ, Buck G, et al. ; United Kingdom Medical Research Council Primary Thrombocythemia 1 Study . Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med. 2005;353(1):33-45.
    1. Spanoudakis E, Bazdiara I, Kotsianidis I, et al. . Hydroxyurea (HU) is effective in reducing JAK2V617F mutated clone size in the peripheral blood of essential thrombocythemia (ET) and polycythemia vera (PV) patients. Ann Hematol. 2009;88(7):629-632.
    1. Larsen TS, Pallisgaard N, de Stricker K, Møller MB, Hasselbalch HC. Limited efficacy of hydroxyurea in lowering of the JAK2 V617F allele burden. Hematology. 2009;14(1):11-15.
    1. Antonioli E, Carobbio A, Pieri L, et al. . Hydroxyurea does not appreciably reduce JAK2 V617F allele burden in patients with polycythemia vera or essential thrombocythemia. Haematologica. 2010;95(8):1435-1438.
    1. Kiladjian J-JJ, Cassinat B, Turlure P, et al. . High molecular response rate of polycythemia vera patients treated with pegylated interferon α-2a. Blood. 2006;108(6):2037-2040.
    1. Quintás-Cardama A, Kantarjian H, Manshouri T, et al. . Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia vera. J Clin Oncol. 2009;27(32):5418-5424.
    1. Gisslinger H, Klade C, Georgiev P, et al. ; PROUD-PV Study Group . Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol. 2020;7(3):e196-e208.
    1. Kiladjian JJ, Cassinat B, Chevret S, et al. . Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008;112(8):3065-3072.
    1. Masarova L, Patel KP, Newberry KJ, et al. . Pegylated interferon alfa-2a in patients with essential thrombocythaemia or polycythaemia vera: a post-hoc, median 83 month follow-up of an open-label, phase 2 trial. Lancet Haematol. 2017;4(4):e165-e175.
    1. Quintás-Cardama A, Abdel-Wahab O, Manshouri T, et al. . Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon α-2a. Blood. 2013;122(6):893-901.
    1. Silver RT. Recombinant interferon-alpha for treatment of polycythaemia vera. Lancet. 1988;2(8607):403.
    1. Samuelsson J, Hasselbalch H, Bruserud O, et al. ; Nordic Study Group for Myeloproliferative Disorders . A phase II trial of pegylated interferon α-2b therapy for polycythemia vera and essential thrombocythemia: feasibility, clinical and biologic effects, and impact on quality of life. Cancer. 2006; 106(11):2397-2405.
    1. Stauffer Larsen T, Iversen KF, Hansen E, et al. . Long term molecular responses in a cohort of Danish patients with essential thrombocythemia, polycythemia vera and myelofibrosis treated with recombinant interferon alpha. Leuk Res. 2013;37(9):1041-1045.
    1. Utke Rank C, Weis Bjerrum O, Larsen TS, et al. . Minimal residual disease after long-term interferon-alpha2 treatment: a report on hematological, molecular and histomorphological response patterns in 10 patients with essential thrombocythemia and polycythemia vera. Leuk Lymphoma. 2016; 57(2):348-354.
    1. Larsen TS, Bjerrum OW, Pallisgaard N, Andersen MT, Møller MB, Hasselbalch HC. Sustained major molecular response on interferon alpha-2b in two patients with polycythemia vera. Ann Hematol. 2008;87(10):847-850.
    1. Larsen TS, Møller MB, De Stricker K, et al. . Minimal residual disease and normalization of the bone marrow after long-term treatment with alpha-interferon2b in polycythemia vera. A report on molecular response patterns in seven patients in sustained complete hematological remission. Hematology. 2009;14(6):331-334.
    1. Swerdlow S, Campo E, Harris N, et al. . World Classification of Tumors of Haematopoietic and Lymphoid Tissues. Lyon, France: IARC; 2008
    1. Barosi G, Mesa R, Finazzi G, et al. . Revised response criteria for polycythemia vera and essential thrombocythemia: an ELN and IWG-MRT consensus project. Blood. 2013;121(23):4778-4781.
    1. Barosi G, Bordessoule D, Briere J, et al. ; European Myelofibrosis Network . Response criteria for myelofibrosis with myeloid metaplasia: results of an initiative of the European Myelofibrosis Network (EUMNET). Blood. 2005;106(8):2849-2853.
    1. Segura-Díaz A, Stuckey R, Florido Y, et al. . Thrombotic risk detection in patients with polycythemia vera: the predictive role of DNMT3A/TET2/ASXL1 mutations. Cancers (Basel). 2020;12(4):1-9.
    1. Tefferi A, Lasho TL, Guglielmelli P, et al. . Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv. 2016;1(1):21-30.
    1. Cerquozzi S, Barraco D, Lasho T, et al. . Risk factors for arterial versus venous thrombosis in polycythemia vera: a single center experience in 587 patients. Blood Cancer J. 2017;7(12):662.
    1. Mosca M, Lamrani L, Marzac C, et al. . Differential impact of interferon alpha on JAK2V617F and CALR mutated hematopoietic stem and progenitor cells in classical MPN. Blood. 2018;132(suppl 1):4333.
    1. Mascarenhas J, Kosiorek HE, Prchal JT, et al. . Results of the Myeloproliferative Neoplasms–Research Consortium (MPN-RC) 112 randomized trial of pegylated interferon alfa-2a (PEG) versus hydroxyurea (HU) therapy for the treatment of high risk polycythemia vera (PV) and high risk essential thrombocythemia. Blood. 2018;132(suppl 1):577.
    1. Kjær L, Cordua S, Holmström MO, et al. . Differential dynamics of CALR mutant allele burden in myeloproliferative neoplasms during interferon alfa treatment. PLoS One. 2016;11(10):e0165336.
    1. Verger E, Cassinat B, Chauveau A, et al. . Clinical and molecular response to interferon-α therapy in essential thrombocythemia patients with CALR mutations. Blood. 2015;126(24):2585-2591.
    1. Czech J, Cordua S, Weinbergerova B, et al. . JAK2V617F but not CALR mutations confer increased molecular responses to interferon-α via JAK1/STAT1 activation. Leukemia. 2019;33(4):995-1010.
    1. Jia R, Kralovics R. Progress in elucidation of molecular pathophysiology of myeloproliferative neoplasms and its application to therapeutic decisions. Int J Hematol. 2020;111(2):182-191.
    1. Nangalia J, Nice FL, Wedge DC, et al. . DNMT3A mutations occur early or late in patients with myeloproliferative neoplasms and mutation order influences phenotype. Haematologica. 2015;100(11):e438-e442.
    1. Rao N, Butcher CM, Lewis ID, et al. . Clonal and lineage analysis of somatic DNMT3A and JAK2 mutations in a chronic phase polycythemia vera patient. Br J Haematol. 2012;156(2):268-270.
    1. Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7(1):12484.
    1. Stetka J, Hansen N, Kubovcakova L, Hao-shen H, Dirnhofer S, Skoda RC. Loss of Dnmt3a confers resistance to pegifn α in JAK2-V617F mouse model [abstract]. Blood. 2020;136(Supplement 1):8-9. Abstract 635.
    1. Jacquelin S, Straube J, Cooper L, et al. . Jak2V617F and Dnmt3a loss cooperate to induce myelofibrosis through activated enhancer-driven inflammation. Blood. 2018;132(26):2707-2721.
    1. Skoda RC. Accelerating myelofibrosis through loss of Dnmt3a. Blood. 2018;132(26):2703-2704.
    1. Lindsley RC, Saber W, Mar BG, et al. . Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N Engl J Med. 2017;376(6):536-547.
    1. 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.
    1. Kahn JD, Miller PG, Silver AJ, et al. . PPM1D-truncating mutations confer resistance to chemotherapy and sensitivity to PPM1D inhibition in hematopoietic cells. Blood. 2018;132(11):1095-1105.
    1. Hsu JI, Dayaram T, Tovy A, et al. . PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy. Cell Stem Cell. 2018;23(5):700-713.e6.
    1. Kubesova B, Pavlova S, Malcikova J, et al. . Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status. Leukemia. 2018;32(2):450-461.
    1. Gisslinger H, Klade C, Georgiev P, et al. . Long-term use of ropeginterferon alpha-2b in polycythemia vera: 5-year results from a randomized controlled study and its extension [abstract]. Blood. 2020;136(supplement 1):33. Abstract 481.
    1. Gisslinger H, Klade C, Georgiev P, et al. . Ropeginterferon alfa-2b: efficacy and safety in different age groups. HemaSphere. 2020;4(6):e485.
    1. Van Egeren D, Escabi J, Nguyen M, et al. . Reconstructing the lineage histories and differentiation trajectories of individual cancer cells in myeloproliferative neoplasms. Cell Stem Cell. 2021;28(3):514-523.e9.
    1. Williams N, Lee J, Moore L, et al. . Phylogenetic reconstruction of myeloproliferative neoplasm reveals very early origins and lifelong evolution. bioRxiv. Posted online November 9, 2020.
    1. 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.
    1. Cordua S, Kjaer L, Skov V, Pallisgaard N, Hasselbalch HC, Ellervik C. Prevalence and phenotypes of JAK2 V617F and calreticulin mutations in a Danish general population. Blood. 2019;134(5):469-479.
    1. Wolach O, Sellar RS, Martinod K, et al. . Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med. 2018;10(436):eaan8292.
    1. Pedersen RK, Andersen M, Knudsen TA, et al. . Data-driven analysis of JAK2 V617F kinetics during interferon- alpha2 treatment of patients with polycythemia vera and related neoplasms. Cancer Med. 2020;9(6):2039-2051.
    1. Steensma DP, Bolton KL. What to tell your patient with clonal hematopoiesis and why: insights from 2 specialized clinics. Blood. 2020;136(14): 1623-1631.

Source: PubMed

Подписаться