scholarly journals A Cure for Paroxysmal Nocturnal Hemoglobinuria Using Molecular Targeted Therapy Specific to a Driver Mutation

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1215-1215 ◽  
Author(s):  
Takamasa Katagiri ◽  
Ryo Tominaga ◽  
Keisuke Kataoka ◽  
Akio Maeda ◽  
Hiroshi Gomyo ◽  
...  

Abstract Background: The clonal expansion of PIGA mutant hematopoietic stem cells (HSCs) can be induced by secondary driver mutations in genes such as HMGA2 and JAK2 in some patients with paroxysmal nocturnal hemoglobinuria (PNH). Theoretically, this type of PNH may be cured by molecular targeted therapy if the therapy is specific for the driver mutations and can eliminate PIGA mutant HSCs that acquired a proliferative advantage. However, this theory has not been proven because of the lack of an appropriate targeted therapy for known driver mutations responsible for clonal expansion of PIGA mutant HSCs. We recently treated a case of PNH complicated by chronic myeloid leukemia (CML) with nilotinib and observed a complete molecular response of CML followed by a complete disappearance of glycosylphosphatidylinositol-anchored protein-deficient (GPI-AP-, PNH-type) cells after 19 months of treatment. Case report: The patient, a 27-year-old Japanese woman, developed severe anemia with leukocytosis and thrombocytosis in 2013. The laboratory findings on admission were as follows: white blood cell count of 18.7x109/L with 1.7% stab neutrophils, 5.4% segmented neutrophils, 9.3% basophils, 1.3% promyelocytes, 5.0% myelocytes, 3.0% metamyelocytes, 18.0% lymphocytes and 0.7% blasts; hemoglobin (Hb)=6.0 g/dL, platelets=1,000x109/L, 20.0% reticulocytes, total/direct bilirubin=1.9/0.3 mg/dL, LDH=1963 IU/L and haptoglobin <10 mg/dL. A high-sensitivity flow cytometry analysis of the patient's peripheral blood at diagnosis revealed that 99.2% of granulocytes, 75.7% of erythrocytes and 99.3% of monocytes were GPI-AP-, while no T cells, B cells or NK cells had the PNH-phenotype (Figure 1A). This GPI-AP- cell distribution pattern was in sharp contrast to that of a patient with typical PNH who showed various percentages of GPI-AP- cells in all lineages of leukocytes and erythrocytes (Figure 1B). A fluorescent in situ hybridization analysis showed that 98.0% of the patient's granulocytes were BCR-ABL gene-fusion positive. Deep sequencing of leukocytes obtained at diagnosis showed a G279T (Q93H) mutation in exon 4 of the PIGA gene. The patient was diagnosed with PNH complicated by CML in the chronic phase, and was treated with nilotinib at 400 mg/day. The percentage of GPI-AP- cells rapidly decreased in response to nilotinib, with 0.02% GPI-AP- granulocytes after six months of nilotinib therapy when the patient's BCR-ABL mRNA decreased to 0.007%. BCR-ABL mRNA decreased to less than 0.0035% 15 months after therapy; however, small populations of GPI-AP- granulocytes (0.01%) and erythrocytes (0.005%) were still detected at this time (Figure 1A). GPI-AP- cells became undetectable after 19 months of nilotinib therapy, suggesting that the BCR/ABL fusion occurred in a subclone of a PIGA mutant hematopoietic progenitor cell (HPC). The patient was continuing nilotinib as of August 2015 without any signs of an increase in the BCR-ABL mRNA copy number. Conclusions: This case indicates the BCR-ABL fusion can be a driver mutation capable of inducing the clonal expansion of PIGA mutant clones. More importantly, the origin of PNH in our case proved to be a minor HPC clone with a PIGA mutation, suggesting that PNH can be derived from an HPC with a limited life span only if a potent second hit occurs in the PIGA mutant HPC. The identification of driver mutations in patients with PNH may therefore lead to the development of targeted therapy capable of curing PNH. Disclosures No relevant conflicts of interest to declare.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 8528-8528
Author(s):  
Yufei Liu ◽  
Zhe Zhang ◽  
Waree Rinsurongkawong ◽  
Xiuning Le ◽  
Carl Michael Gay ◽  
...  

8528 Background: The use of durvalumab after chemoradiation in locally advanced non-small cell lung cancer (NSCLC) patients significantly improves overall survival. However, it is unclear whether this benefit applies to all genetic subtypes of lung cancer. We hypothesize that patients with driver mutation NSCLC may derive less benefit from consolidation durvalumab. Methods: Using the Genomic Marker-Guided Therapy Initiative (GEMINI) database at MD Anderson, we identified 134 patients who were treated with chemoradiation followed by durvalumab for NSCLC. We segregated patients with driver mutations to targetable (EGFR, ALK translocation, ROS1 fusion, MET exon 14 skipping, RET fusion, and/or BRAF) (N = 24) and those driven by canonical KRAS mutations (N = 26). The rest (N = 84) had none of these mutations. We gathered demographic, treatment, and outcome data and compared progression-free survival (PFS) and overall survival (OS) using the Kaplan-Meier method. We used multivariate regression analysis to account for demographic and treatment variables. Results: For our cohort, median age at diagnosis was 64.8, 52% were female (n = 70), and median follow up was 1.5 years. 86% of patients have a history of smoking (n = 115). 21% had squamous cell histology (n = 28). 2 patients had stage IIA disease, 6 had stage IIB, 48 had stage IIIA, 56 had stage IIIB, 13 had stage IIIC, and 9 had stage IV. 73 patients had progression after durvalumab and 37 patients died. Patients with driver mutations had significantly worse median PFS compared to those without driver mutations (8.9 mo vs 26.6 mo; HR 2.62 p < 0.001). Patients with KRAS mutations had particularly poor PFS (Median 7.9 mo, HR 3.34, p < 0.001), while patients with targetable driver mutations trended to worse PFS (Median 14.5 mo, HR 1.96, p = 0.056). The median OS for the cohort was 4.8 yrs with no significant differences based on driver mutation status. On multivariate analysis, only driver mutation status was associated with PFS, but not OS. For patients with first progression, we found the targetable driver group to have significantly improved time to second objective progression (PFS2) compared to the KRAS (HR 0.28, p = 0.011) or non-mutated group (HR 0.38, p = 0.025). All patients in the targetable driver group received targeted therapy after first progression. Conclusions: Our results suggest that patients with driver mutations have worse PFS compared to patients without these mutations after chemoradiation. However, patients with targetable oncogene driver mutations have significantly improved prognosis after initial progression compared to the other groups, likely due to targeted therapy, suggesting that these therapies, including novel approaches towards KRAS mutants, should be further explored in this setting.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5886-5886 ◽  
Author(s):  
Miroslaw Markiewicz ◽  
Malwina Rybicka-Ramos ◽  
Monika Dzierzak-Mietla ◽  
Anna Koclega ◽  
Krzysztof Bialas ◽  
...  

Abstract Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired clonal abnormality of hematopoietic stem cell leading to lack of phosphatidylinositol glycoproteins, sensitizing cells to complement-mediated lysis. Despite the efficient symptomatic treatment of hemolytic PNH with eculizumab, allo-HCT is the only curative treatment for the disease, although outcomes presented in the past were controversial. Material and methods: We report 41 allo-HCTs: 37 from MUD and 4 from MRD performed for PNH in 2004-2016. Median age of recipients was 29(20-62) years and donors 30(19-53), median time from diagnosis to allo-HCT was 16(2-307) months. Median size of PNH clone was 80% granulocytes (0.5%-100%). Indication for allo-HCT was PNH with aplastic/hypoplastic bone marrow (19 pts), MDS (2 pts), overlapping MDS/aplasia (3 pts), severe course of PNH with hemolytic crises and transfusion-dependency without access to eculizumab (17 pts). Additional risk factors were Budd-Chiari syndrome and hepatosplenomegaly (1 pt), history of renal insufficiency requiring hemodialyses (2 pts), chronic hepatitis B (1 pt) and C (1 pt). The preparative regimen consisted of treosulfan 3x14 g/m2 plus fludarabine 5x30 mg/m2 (31 pts) or treosulfan 2x10 g/m2 plus cyclophosphamide 4x40 mg/kg (10 pts). Standard GVHD prophylaxis consisted of cyclosporine-A, methotrexate and pre-transplant ATG in MUD-HCT. 2 pts instead of cyclosporine-A received mycophenolate mofetil and tacrolimus. Source of cells was bone marrow (13 pts) or peripheral blood (28 pts) with median 6.3x108NC/kg, 5.7x106CD34+cells/kg, 24.7x107CD3+cells/kg. Myeloablation was complete in all pts with median 9(1-20) days of absolute agranulocytosis <0.1 G/l. Median number of transfused RBC and platelets units was 9(0-16) and 8(2-18). Results: All pts engrafted, median counts of granulocytes 0.5 G/l, platelets 50 G/l and Hb 10 g/dl were achieved on days 17.5(10-33), 16(9-39) and 19.5(11-34). Acute GVHD grade I,II and III was present in 16, 7 and 3 pt, limited and extensive chronic GVHD respectively in 11 and 3 pts. LDH decreased by 73%(5%-91%) in first 30 days indicating disappearance of hemolysis. 100% donor chimerism was achieved in all pts. In 1 patient donor chimerism decreased to 81% what was treated with donor lymphocytes infusion (DLI). 3 patients died, 1 previously hemodialysed pt died on day +102 due to nephrotoxicity complicating adenoviral/CMV hemorrhagic cystitis, two other SAA patients with PNH clone<10% died on days +56 due to severe pulmonary infection and +114 due to aGvHD-III and multi organ failure. Complications in survivors were FUO (10 pts), CMV reactivation (13), VOD (1), neurotoxicity (1), venal thrombosis (1), hemorrhagic cystitis (4) and mucositis (8). 38 pts (92.7%) are alive 4.2 (0.4-12) years post-transplant and are doing well without treatment. Complete disappearance of PNH clone was confirmed by flow cytometry in all surviving pts. Conclusions: Allo-HCT with treosulfan-based conditioning is effective and well tolerated curative therapy for PNH. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 944-944
Author(s):  
Patricia Eiko Yamakawa ◽  
Ana Rita Da Fonseca ◽  
Caio Perez Gomes ◽  
Agatha Mendes ◽  
Fabiana Bettoni ◽  
...  

Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is a disorder due to an acquired loss-of-function mutation in the phosphatidylinositol glycan class A (PIG-A) gene. A large spectrum of acquired PIG-A mutations has been described, like insertions or deletions involving a single base or several bases, and single base substitution that are the most common. Usually there are more than one PIGA mutations and one clone is predominant. The clinical manifestations of PNH are intrinsically related to clonal expansion of hematopoietic stem cell deficient in GPI-anchored proteins. Some hypothesis failed to explain alone this clonal expansion. Here we try to identify and to correlate PIG-A gene mutations with clinical manifestations in a series of patients with PNH. Methods: We analyzed 31 patients with classical PNH (n=23) or aplastic anemia and PNH clone (n=8). The sequencing of the PIG-A gene was performed using the Sanger technique. The electropherograms were aligned against the reference sequence of the PIG-A gene deposited in GenBank (Accession number NG_009786), and analyzed using the Geneious R10 software (Biomatters). After analysis, a search was performed in the Clinvar, dbSNP and HGMD databases to verify the pathogenicity of the mutations. For variants without description in the literature, a pathogenicity prediction analysis was performed using Mutation Taster, Polyphen 2 and Human Splicing Finder software. Results: We found 29 different variants of the PIG-A gene in 27 patients: 23 were new mutations, with no previous description in the literature, 3 were previously described mutations, and 3 were single nucleotide polymorphism (SNP). There was great variation in the type and location of somatic mutations. Mutations were predominantly small deletions and simple base changes; 42% of the mutations were described as frameshift mutations and 31% missense mutations. We did not find any specific correlation between the clinical characteristics of hemolytic PNH patients and their mutations, due to the wide variety of mutations. According the pathogenicity prediction programs, the majority (22 of 29) of the variants found were classified as probably pathogenic. Among the 23 patients with hemolytic PNH, 19 patients had at least one mutation classified as pathogenic. In patients with subclinical PNH, only SNPs were found. Fifteen patients with hemolytic PNH had more than one concomitant mutation, most of which were probably pathogenic mutations associated with a polymorphism. Conclusion: We described PIG-A mutations in a series of PNH patients in Brazil and observed no correlation exists between mutation types and clinical features in hemolytic patients. Among subclinical PNH patients, only SNPs were observed, probably because of small clone sizes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-10
Author(s):  
Fangfei Chen ◽  
Bing Han ◽  
Jian Li

Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a disease presented with hemolysis, cytopenia and thrombosis. Apart from PIGA gene on hematopoietic stem cells which accounts for the glycosylphosphatidylinositol (GPI) anchor deficiency on the cell membrane, other mutations have also been detected in PNH through whole-exome sequencing (WES). However, the characteristics of mutations in patients with PNH and genes which may contribute to PNH clonal expansion have not been well defined. Methods: Peripheral blood samples were collected from 41 patients with PNH, among them samples from 6 patients were further separated into CD59- and CD59+ fractions by CD59 magnetic beads. Gene mutations were tested by whole-exome sequencing(WES). 178 genes commonly mutated in myeloid cancer were analyzed in the sequencing results, as well as their correlation with clinical indicators. Mutated genes correlated with cell proliferation were compared between sorted CD59+ and CD59- cells. Results: The most frequently mutated myeloid cancer-related genes were MAP3K4 and CSMD1 (12.2% respectively). Among them, RUNX1T1 mutation was found to be correlated with larger clone size, higher level of uncombined bilirubin, and lower level of hemoglobin (P&lt;0.05). No other correlation between clinical parameters and gene mutations were found. The proportion of mutations (DNMT3A、RUNX1、JAK2、JAK3、CSMD1) which have been shown to indicate poor outcome in patients with aplastic anemia decreased as PNH clone increased (p=0.026). Mutations related to cell proliferation tended to happen more frequently in CD59- fractions compared with CD59+ fractions of the same patient (P=0.062). Conclusions: Myeloid cancer-related mutations can be detected in patients with PNH with some correlation with clinical manifestations. Larger PNH clone may "save" patients from mutation indicating poor prognosis. CD59- fractions seemed to carry more proliferation related mutations, which may contribute to PNH clonal expansion. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3439-3439
Author(s):  
Yoshiko Murakami ◽  
Rieko Ohta ◽  
Norimitsu Inoue ◽  
Hideyoshi Noji ◽  
Tsutomu Shichishima ◽  
...  

Abstract Paroxysmal nocturnal hemoglobinuria (PNH) is caused by a somatic mutation of PIG-A gene in one or few hematopoietic stem cells and subsequent clonal expansion of mutant stem cells that leads to development of symptoms. It is known that PIG-A mutation is insufficient to account for the clonal expansion required for clinical manifestation of PNH. We are proposing a 3-step model of PNH pathogenesis. Step 1 involves the generation of a GPI-deficient hematopoietic stem cell by somatic mutation of the PIG-A gene. Step 2 involves the immunological selection of GPI-deficient hematopoietic stem cells. Based on the close association of PNH with aplastic anemia, it has been suggested that the selection pressure is immune mediated. However, in spite that over 60% of patients with aplastic anemia have subclinical population of GPI-deficient hematopoietic cells at diagnosis, only 10% develop clinical PNH, suggesting that step-1 and 2 are insufficient to cause PNH. Under immune mediated selection pressure, GPI-deficient cells not only survive, but also proliferate much more frequently than usual to compensate for anemia. This elevated proliferation rate increases the chance that additional genetic mutations are acquired, in turn leads to Step 3. Step 3 involves a second somatic mutation that bestows on PIG-A mutant stem cell a proliferative phenotype. According to this hypothesis, we searched for the candidate gene for Step 3. We reported 2 patients with PNH whose PIG-A mutant cells had an acquired rearrangement of chromosome12, making the break within the 3’ untranslated region in HMGA2. This gene encodes an architectural transcription factor which is deregulated in many benign mesenchymal tumors (Blood. 2006 vol.108 no.13, p4232). Recently, many reports show that truncation of 3’ untranslated region of HMGA2 disrupts binding of miRNA, let-7, which regulates both transcription and translation of HMGA2. In fact, these two PNH patients with chromosomal abnormalities had ectopic expression of HMGA2 in the bone marrow. Based on these, we consider HMGA2 as a candidate gene, ectopic expression of which causes proliferation. We have established the method for stable isolation of mRNA and miRNA from blood and bone marrow cells from PNH patients and analyzed the expression of HMGA2 and let-7 by quantitative RT-PCR. We have analyzed the peripheral blood from 8 healthy volunteers and 12 PNH patients. The samples from patients had significantly higher expression of HMGA2 than those from normal volunteers (relative mRNA expression, 4.8±2.4 vs 1.3±0.3, p<0.05). We analyzed the genomic sequence of three patients including one who has highest HMGA2 expression and found no mutation in 3’ untranslated region. We also analyzed the expression of miRNA and found significantly lower expression of let7b and c in patients. Surprisingly, truncated form without 3’ untranslated region is predominantly expressed in patients. There maybe deregulation of alternative splicing of HMGA2 gene in patients, which needs further investigations. We are now analyzing more PNH samples including bone marrow, where proliferation of stem cells takes place, to investigate whether high expression of HMGA2 contributes to the pathogenesis of PNH. In addition we are going to analyze whether high expression of HMGA2 causes the clonal expansion of PNH cells using PNH mouse model.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4786-4786
Author(s):  
Olivier Kosmider ◽  
Nicolas Chapuis ◽  
Sophie Kaltenbach ◽  
Romain Coriat ◽  
Pascaline boudou-Rouquette ◽  
...  

Abstract Abstract 4786 Background Chronic myelomonocytic leukemia (CMML) is a rare clonal hematopoietic stem cell disorder whom biology remains unclear.CMML is associated with many different somatic mutations in genes involved in key cellular processes including signaling (N/K-Ras, CBL, JAK2); differentiation (RUNX1, NPM1, CEBPa); epigenetic regulation (TET2, ASXL1, IDH1/2, EZH2, DNMT3A); and RNA splicing (SRSF2, U2AF1, SF3B1 and ZRSR2). FLT3 mutations are very rare but provide the rationale for FLT3 tyrosine kinase inhibitor use to treat this disease. We report the case of a 60-years-old patient diagnosed with a hepatocarcinoma metastasis which legitimized the introduction of anti-angiogenic therapy using the VEGF-R2 inhibitor, sorafenib. The patient was addressed to the hematology department with a myeloproliferative-like CMML in transformation. We show here that the molecular analyses of this hematological disorders allow us to use sorafenib as a targeted therapy to inhibit the consequences of a FLT3-ITD mutation. Methods Cytogenetic analysis and genome wide array-based comparative genomic hybridization (aCGH) were performed at diagnosis. The reference standard used for aCGH was matched genomic constitutional DNA (CD3+ T cells sorted from a blood sample). Serum samples collected from the patient before or under treatment with sorafenib were assessed for their plasma inhibitory activity by western blotting analyses of signaling molecules downstream the FLT3-ITD mutation. Genomic DNA samples extracted from BMMCs and peripheral blood (PB) cells at diagnosis were screened for mutations in 18 classical genes. To monitor the FLT3-ITD mutation, the exon 15 of FLT3 was amplified by a specific PCR using a 6FAM-labeled forward primer. Results The patient developed hyperleucocytosis (48.2 G/L) with neutrophilia (30.4 G/L), monocytosis (11.6 G/L) and basophilia (0.5 G/L) in January 2011. The BM was hypercellular with granulocytic and monocytic proliferation, dysgranulopoiesis and dysmegacaryopoiesis. Blast cells plus promonocytes accounted for 30% of the nucleated BM cells, leading to a diagnosis of AML secondary to CMML in the WHO classification BM karyotype identified no clonal abnormalities and aCGH analysis of BMMCs produced normal findings. BM and PB cells were screened for mutations in 18 CMML-associated genes. Only two abnormalities were identified: a 27 base pair (bp) insertion FLT3-ITD mutation (exon 15) detected in BM cells with near complete disappearance of the wild type (WT) FLT3 allele (FLT3-ITD/FLT3-WT ratio at 9.62) and a classical heterozygous mutation (dupG) was found in the exon 12 of ASXL1. ASXL1 and FLT3-ITD mutations were not detected in purified CD3+ T lymphocytes. Five months after sorafenib introduction, PB was strictly normal and BM examination demonstrated normal richness, blast cells and promonocytes accounting for 2% of the nucleated BM cells but persistent moderate dysgranulopoiesis and dysmegakaryopoiesis, indicative of complete remission. At this time, the FLT3-ITD/FLT3-WT ratio was 1.66 and 0.58 in the BM and PB, respectively. In January 2012, the WBC profile was still normal and a BM smear only showed moderate dysgranulopoiesis. On the molecular side, FLT3-ITD mutation was undetectable, indicative of complete molecular response. But ASXL1 mutation was evident at all time points. The serum of the patient, obtained before and under sorafenib was tested on cell line harboring FLT3-ITD mutation. Constitutive FLT3 Y591, Akt S473, STAT5 Y694 and ERK1/2 T202/Y204 phosphorylations were fully inhibited in the presence of the serum extracted under sorafenib treatment. Conclusion Our patient clearly had a myeloproliferative-CMML driven by an homozygous FLT3-ITD mutation. This is the first report of such a CMML patient achieving sustained CR and CMR after treatment with an FLT3-ITD tyrosine kinase inhibitor. In this case, the ASXL1 mutation remained detectable upon sorafenib treatment after the suppression of FLT3-ITD-driven malignant hematopoiesis, suggesting that it arose from a FLT3-WT subclone that contributed to the CMML phenotype with some dysplastic feature. In conclusion, we propose that mutations in the FLT3 gene should be examinated in all CMML cases, even their low frequency because FLT3 TKI may induce dramatic and sustained responses without significant toxicity and eventually allow for allogenic transplantation. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Jyoti Nangalia ◽  
Nicholas Williams ◽  
Joe Lee ◽  
Luiza Moore ◽  
E Baxter ◽  
...  

Abstract Mutations in cancer-associated genes drive tumour outgrowth. However, the timing of driver mutations and dynamics of clonal expansion that lead to human cancers are largely unknown. We used 448,553 somatic mutations from whole-genome sequencing of 843 clonal haematopoietic colonies to reconstruct the phylogeny of haematopoiesis, from embryogenesis to clinical disease, in 10 patients with myeloproliferative neoplasms which are blood cancers more common in older age. JAK2V617F, the pathognomonic mutation in these cancers, was acquired in utero or childhood, with upper estimates of age of acquisition ranging between 4.1 months and 11.4 years across 5 patients. DNMT3A mutations, which are associated with age-related clonal haematopoiesis, were also acquired in utero or childhood, by 7.9 weeks of gestation to 7.8 years across 4 patients. Subsequent driver mutation acquisition was separated by decades. The mean latency between JAK2V617F acquisition and clinical presentation was 34 years (range 20-54 years). Rates of clonal expansion varied substantially (<10% to >200% expansion/year), were affected by additional driver mutations, and predicted latency to clinical presentation. Driver mutations and rates of expansion would have been detectable in blood one to four decades before clinical presentation. This study reveals how driver mutation acquisition very early in life with life-long growth trajectories drive adult blood cancer, providing opportunities for early detection and intervention, and a new paradigm for cancer development.


Blood ◽  
2020 ◽  
Vol 136 (Supplement_2) ◽  
pp. LBA-1-LBA-1
Author(s):  
Nicholas Williams ◽  
Joe Lee ◽  
Luiza Moore ◽  
Joanna E Baxter ◽  
James Hewinson ◽  
...  

Background Recurrent mutations in cancer-associated genes drive tumour outgrowth, however, the timing of driver mutations and the dynamics of clonal expansion remain largely unknown. Philadelphia-negative myeloproliferative neoplasms (MPN) are unique cancers capturing the earliest stages of tumorigenesis through to disease evolution. Most patients harbor JAK2V617F, present as the only driver mutation or occurring in combination with driver mutations in genes such as DNMT3A or TET2. We aimed to identify the timing of driver mutations and clonal dynamics in adult MPN. Method We undertook whole-genome sequencing of individual single-cell derived hematopoietic colonies (n=952) together with targeted resequencing of longitudinal blood samples from 10 patients with MPN who presented with disease between ages 20 and 76 years. We identified 448,553 somatic mutations which were used to reconstruct phylogenetic trees of hematopoiesis, tracing blood cell lineages back to embryogenesis. We timed driver mutation acquisition, characterised the dynamics of tumour evolution and measured clonal expansion rates over the lifetime of patients. Resequencing of bulk blood samples corroborated clonal trajectories and provided population estimates. Results JAK2V617F was acquired in utero or childhood in all patients in whom JAK2V617F was the first or the only driver mutation. Earliest age estimates were within a few weeks post conception, and upper estimates of age of acquisition were between 4.1 months and 11.4 years, despite wide ranging ages of MPN presentation. The mean latency between JAK2V617F acquisition and clinical presentation was 34 years (range 20-54 years). Subsequent driver mutation acquisition, including for JAK2V617F, was separated by decades. Disease latency following acquisition of JAK2V617F as a second driver event was still 12-27 years. DNMT3A mutations, commonly associated with age-related clonal hematopoiesis (CH), occurred as the first driver event, subsequent to mutated-JAK2, and as independent clones representing CH in MPN patients. DNMT3A mutations were also first acquired in utero or childhood, at the earliest 1.2 weeks post conception, and the latest 7.9 weeks of gestation to 7.8 years across 4 patients. A recurrent feature of the clonal landscape in MPN was the observation of similar genetic changes repeatedly occurring in unrelated clones within the same patient. Such 'parallel evolution' was observed for chr9p loss-of-heterozygosity, chr1q+ and mutations in myeloid cancer genes, suggesting that patient-specific factors flavour selective landscapes in MPN. Normal hematopoietic stem cells accumulated ~18 somatic mutations/year, however, mutant clones, particularly those with mutant-JAK2, acquired 1.5-5.5 excess mutations/ year and had shorter telomeres, reflecting increased cell divisions during clonal expansion. We modelled the rates of clonal expansion and found that they varied substantially, both across patients and within individuals. In one patient, an in utero acquired DNMT3A-mutated clone expanded slowly at &lt;10%/year, taking 30 years to reach a clonal fraction of 1%, whilst a clone with mutated-JAK2, -DNMT3A and -TET2 expanded at &gt;200%/year, doubling in size every 7 months. JAK2V617F as a single driver mutation also expanded variably across patients, highlighting that other factors, which may include germline, cytokine or stem cell differences between individuals, also influence selection for driver mutations. JAK2V617F associated clonal expansion rates in MPN were greater than that reported for JAK2-CH. Furthermore, rates of expansion in the cohort predicted time to clinical presentation, more so than age of mutation acquisition or tumour burden at diagnosis. This suggests that JAK2-mutant clonal expansion rates determine both if and when clinical manifestations occur. Driver mutations and rates of clonal expansion would have been detectable in blood one to four decades before clinical presentation. Conclusions MPN originate from driver mutation acquisition very early in life, even before birth, with life-long clonal expansion and evolution, establishing a new paradigm for blood cancer development. Early detection of mutant-JAK2 together with determination of clonal expansion rates could provide opportunities for early interventions aimed at minimising thrombotic risk and targeting the mutant clone in at risk individuals. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document