Clonal evolution in patients developing therapy-related myeloid neoplasms following autologous stem cell transplantation

Author(s):  
Johannes Frasez Soerensen ◽  
Anni Aggerholm ◽  
Carina Agerbo Rosenberg ◽  
Marie Bill ◽  
Gitte Birk Kerndrup ◽  
...  
2021 ◽  
Vol 11 (3) ◽  
Author(s):  
Kalyan Nadiminti ◽  
M. Hasib Sidiqi ◽  
Kapil Meleveedu ◽  
Hassan B. Alkhateeb ◽  
William J. Hogan ◽  
...  

2020 ◽  
Vol 4 (5) ◽  
pp. 885-892 ◽  
Author(s):  
Johannes Frasez Soerensen ◽  
Anni Aggerholm ◽  
Gitte Birk Kerndrup ◽  
Marcus Celik Hansen ◽  
Ina Kathrine Lykke Ewald ◽  
...  

Abstract Therapy-related myeloid neoplasms (tMN) develop after exposure to cytotoxic and radiation therapy, and due to their adverse prognosis, it is of paramount interest to identify patients at high risk. The presence of clonal hematopoiesis has been shown to increase the risk of developing tMN. The value of analyzing hematopoietic stem cells harvested at leukapheresis before autologous stem cell transplantation (ASCT) with next-generation sequencing and immunophenotyping represents potentially informative parameters that have yet to be discovered. We performed a nested case-control study to elucidate the association between clonal hematopoiesis, mobilization potential, and aberrant immunophenotype in leukapheresis products with the development of tMN after ASCT. A total of 36 patients with nonmyeloid disease who were diagnosed with tMN after treatment with ASCT were included as case subjects. Case subjects were identified from a cohort of 1130 patients treated with ASCT and matched with 36 control subjects who did not develop tMN after ASCT. Case subjects were significantly poorer mobilizers of CD34+ cells at leukapheresis (P = .016), indicating that these patients possess inferior bone marrow function. Both clonal hematopoiesis (odds ratio, 5.9; 95% confidence interval, 1.8-19.1; P = .003) and aberrant expression of CD7 (odds ratio, 6.6; 95% confidence interval, 1.6-26.2; P = .004) at the time of ASCT were associated with an increased risk of developing tMN after ASCT. In conclusion, clonal hematopoiesis, present at low variant allele frequencies, and aberrant CD7 expression on stem cells in leukapheresis products from patients with nonmyeloid hematologic cancer hold potential for the early identification of patients at high risk of developing tMN after ASCT.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1529-1529
Author(s):  
Johannes Frasez Soerensen ◽  
Anni Aggerholm ◽  
Gitte Birk Kerndrup ◽  
Marcus C Hansen ◽  
Ina Kathrine Lykke Ewald ◽  
...  

Abstract Introduction: Therapy-related myeloid neoplasms (tMN) are high-risk conditions evolved after exposure to a number of agents, including cytotoxic therapy, and include myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPN). As such cytoreduction as part of autologous stem cell transplantation (ASCT) increases the risk of developing tMN. Importantly, both the use of ASCT and the incidence of tMN are rising. The recent characterization of clonal hematopoiesis of indeterminate potential (CHIP) has, in preliminary reports, been shown to increase the risk of developing de novo hematological disease as well as tMN. We hypothesized that patients with non-myeloid primary disease who develop tMN after ASCT, had detectable myeloid mutations at time of transplantation, and that these may represent a risk factor in the development of tMN. This study characterizes tMN patients previously subjected to ASCT and investigates whether CHIP mutations are present in hematopoietic stem cells at time of ASCT. Methods: The cohort of this observational study consists of patients treated with ASCT at the Department of Hematology, Aarhus University Hospital (Denmark) from 1989 to 2016. Cases were identified via the Danish Pathology Registry and all tMN diagnoses were verified by the same experienced hemopathologist (GBK). Only cases from patients with non-myeloid primary disease, who were diagnosed with tMN after being treated with ASCT (minimum latency being 90 days) were included. 36 cases with available leukapheresis products were identified out of a cohort of 1130 patients. Samples collected from leukapheresis prior to ASCT were subjected to targeted next-generation sequencing (NGS), using the commercially available panel "Myeloid Solution" (Sophia Genetics, Saint Sulpice, Switzerland), covering 30 genes relevant for myeloid neoplasms, joined with a bioinformatics pipeline from Sophia Genetics. Samples from 31 patients have been subjected to NGS. Variants residing in or within ± 25 nucleotides of coding exons and with coverage >5000 at the variant site were reported. Indels present in polynucleotide stretches were excluded. Data for continuous variables age, latency to tMN and survival were analyzed as one sample from a normal distribution based on the Students t-test. Normality was assessed via Q-Q plot. Estimates are reported with a 95% confidence intervals (Stata, version 15.1, StataCorp LLC, TX, USA). Results: The cohort consisted of 25 males (80.7%) and 6 females (19.3%), with an estimated median age at ASCT of 58 years (CI 95% 54;63). Estimated median time to tMN was 3.7 years (CI 95% 2.5;5.4) and estimated median survival after tMN diagnosis was 132 days (CI 95% 71;246). At time of tMN diagnosis, 14 patients had a poor risk karyotype and 9 patients had intermediate risk karyotype (hereof 4 normal karyotype). Karyotype was not evaluated in 8 patients. CHIP mutations were detected in stem cell enriched leukapheresis products from 21 patients (67.7%). Of these, we found multiple mutations in 14 patients (66%) and in one patient as many as 6 CHIP mutations. Point mutations were frequently found in the DNMT3A gene and was present in 16 out of 21 patients (76%). Six patients had more than one DNMT3A mutation, one of which had 5 separate DNMT3A mutations. Other mutations detected were TP53 (6/21), TET2 (5/21), ASXL1 (4/21), EZH2 (1/21), WT1 (1/21), JAK2 (1/21), NRAS (1/21), HRAS (1/21), BRAF (1/21), CSF3R (1/21), SF3B1 (1/21), ZRSR2 (1/21), CALR (1/21), SRSF2 (1/21). Conclusion: We found that CHIP mutations can be detected at time of ASCT in patients being treated for non-myeloid diseases. We hypothesize that presence of CHIP mutations at ASCT may predict the development of tMN and as such serve as a biomarker in this setting. We speculate that high-dose cytotoxic therapy may provide an evolutionary advantage for hematopoietic clones containing CHIP mutations. On the other hand, we cannot rule out that the cytoreduction administered prior at ASCT may be a main contributor to the tMN development. To address this as well as the development of tMN in the post-ASCT phase, a nested case-control study will be necessary. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 15-16
Author(s):  
Johannes Frasez Sørensen ◽  
Anni Aggerholm ◽  
Marcus H Hansen ◽  
Gitte Birk Kerndrup ◽  
Lene Hyldahl Ebbesen ◽  
...  

Introduction: Clonal hematopoiesis (CH) denotes somatic mutations in genes related to myeloid neoplasms present at any variant allele frequency (VAF). Clonal hematopoiesis increases the risk of cardiovascular disease, de novo myeloid neoplasms and therapy-related myeloid neoplasms (tMN). It is well established that CH can be detected years before disease onset. Furthermore, the impact of specific mutations with regards to progression from CH to tMN is currently being unraveled. When exposed to cytoreductive therapy, a proliferative advantage of stem cells with CH over normal hematopoietic stem cells (HSCs) has been demonstrated. However, it remains unclear whether CH is to be considered a mere tMN risk factor, or if the mutations directly impact or even drive the development of tMN. We hypothesized that CH contributes to the development of tMN, and pursued this by investigating the evolution of CH, present in patients with lymphoma and multiple myeloma, prior to autologous stem cell transplantation (ASCT) and at time of tMN diagnosis. Methods: Patients included were treated with ASCT at the Department of Hematology, Aarhus University Hospital, Denmark, between 1989 and 2016. Inclusion criteria were (i) treatment with ASCT on the indication of a non-myeloid primary disease; (ii) subsequent development of tMN, and (iii) available mononuclear cells (MNCs) at pre-ASCT and time of tMN. All tMN diagnoses were reviewed by an experienced pathologist. Data from time of ASCT of this cohort has previously been reported (Soerensen et al., 2020, PMID: 32150606). Twelve patients with available MNCs at both time points were identified out of 36 tMN patients. Samples (either leukapheresis products or bone marrow MNCs) were subjected to targeted next-generation sequencing, utilizing a 30-gene panel (Myeloid Tumor Solution, SOPHiA Genetics, Saint Sulpice, Switzerland). Variant exclusion criteria were (1) read depth < 3000; (2) VAF < 0.003; (3) variant location outside ±25 nucleotides of coding region; (4) indel present in homopolymeric stretch, and (5) potential germline variants at pre-ASCT with VAF > 0.95 or between 0.45 and 0.55, representing homo- and heterozygosity, and reported in the Exome Aggregation Consortium (ExAC) database. Results: The cohort included 12 patients with a median age at ASCT of 63 years (range 37-69) and male predominance (75%). Median time to tMN following ASCT was 3.9 years (range 0.7-15.3), with 7 patients developing therapy-related myelodysplastic syndrome and 5 therapy-related acute myeloid leukemia. A total of 36 and 38 mutations were detected at ASCT and tMN, respectively. Prior to ASCT, DNMT3A (39%) and TET2 (19%) were the most frequently mutated genes, whereas the mutational landscape at tMN proved to be more heterogenous, with TP53 (21%), DNMT3A (18%), RUNX1 (13%) and ASXL1 (13%) comprising the majority of mutated genes. Nine patients (75%) had one or more mutations that could be detected at pre-ASCT as well as at tMN. Seven patients (58%) had CH at pre-ASCT that were present at higher VAF (>0.15 VAF) in bone marrow samples at tMN. Of these, 6 patients had CH at VAF < 0.02 at baseline. We found a total of 14 mutations that were detected at both prior to ASCT and tMN diagnosis, distributed among TP53, SRSF2, DNMT3A, ASXL1, TET2, NRAS and EZH2. Importantly, all clones harboring mutations in non-DNMT3A genes expanded until diagnosis of tMN to VAF > 0.30, with the exception of TET2, which displayed only a modest increase in VAF from 0.01 to 0.15. Conclusion: In this cohort of patients treated with ASCT and who subsequently developed tMN, we found the majority of patients to harbor CH in HSCs pre-ASCT that, at time of tMN, completely dominated the malignant clone. Our data suggests both a persistency of CH identified in HSCs in peripheral blood prior to ASCT to the leukemic stem cells in bone marrow at tMN diagnosis, as well as an expansion of the clones over time. These findings provide evidence to support the emerging theories that tMNs are instigated by subsets of hematopoietic cells that gain malignant somatic mutations and drive the pathogenesis years before onset disease. Figure Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document