scholarly journals Clonal hematopoiesis predicts development of therapy-related myeloid neoplasms post–autologous stem cell transplantation

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 ◽  
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.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 491-491 ◽  
Author(s):  
Amrita Y. Krishnan ◽  
John A. Zaia ◽  
John J. Rossi ◽  
Arturo Molina ◽  
Mingjie Li ◽  
...  

Abstract Background: Optimal therapy of ARL relies on both effective anti-tumor chemotherapy and successful control of the underlying HIV infection. Management of HIV using HAART has been hampered by patient non-compliance with complex regimens, drug resistance and ongoing low level viral replication. Multiplexed RNA based anti-HIV gene transfer strategies to confer intrinsic cellular resistance may help circumvent these problems. Autologous stem cell transplantation for ARL is an ideal clinical platform for delivery of SiRNA transduced stem cells as combining gene transfer strategy with high dose antilymphoma therapy could provide control of both the HIV infection and the ARL. Methods: Gene transfer - anti-HIV RNA elements, including short hairpin RNA (shRNA) targeted to HIV tat/rev, a TAR-specific decoy sequence, and a ribozyme targeted to CCR5 were combined into a lentivirus vector (LV, rHIV7-shI-TAR-CCR5RZ). Using LV transduction methods, these anti-HIV RNAs were delivered into CD34+ hematopoietic progenitor cells (HPC). Results: Preclinical vector development - LV transduction allowed differentiation in liquid culture and in a SCID-hu model which produced macrophage and T cell progeny that were resistant to the HIV virus. Although HIV resistance can be induced in vitro with single anti-HIV shRNAs, no resistance was found in multiply passaged HIV in rHIV7-shI-TAR-CCR5RZ-transduced cells. In addition, cells were analyzed by microarray for mRNA and miRNA alterations and no significant changes were noted between CD34 transduced and untransduced cells. Copy number in transduced cells was 1–2/cell, and integration site analysis localized to transcriptionally active sites, usually away from terminal portions of gene sequences. This vector is proposed for use in ASCT for ARL. Update of ASCT in ARL: Between 1998 and 2006, 28 patients with high-risk ARL underwent ASCT at the City of Hope. Conditioning regimens included CBV (cyclophosphamide (Cy)100mg/kg, Carmustine150 mg/m2, VP16 60mg/kg) in 24 and FTBI/Cy/VP16 in 4. All patients engrafted, median time to ANC>500 was 11 days (range, 8–19). Regimen related toxicities included grade 3–4 hepatic toxicity n=3 and interstitial pneumonitis n=2. OI’s included PCP pneumonia n=2, CMV infection (2 viremias, 1 retinitis) and 2 cases of VZV. Therapy-related MDS was seen in one patient who ultimately died of MDS while in remission from his ARL. Median HIV viral load (VL) at ASCT was 6113 gc/ml with 22 having an undetectable VL. At two years only 9 pts had an undetectable VL. Median CD4 count at ASCT was 164 (range 25–1064), this rose to 263 (95–1164) at two years post ASCT. With a median f/u of 41 months, 2yr OS is 78% (95% CI 63–87) and PFS 78% (95% CI 63–87). Conclusions: ASCT can lead to durable remission for ARL. The fluctuation in VL seen post ASCT reflects the natural history of HIV infection and limitations of current antiviral therapy. Ultimately the system of gene transfer outlined above combined with ASCT as part of our next generation clinical trial could provide the optimal chance of cure for pts with high-risk ARL by offering both effective antilymphoma and anti-HIV therapy.


2021 ◽  
Vol 11 (3) ◽  
Author(s):  
Kalyan Nadiminti ◽  
M. Hasib Sidiqi ◽  
Kapil Meleveedu ◽  
Hassan B. Alkhateeb ◽  
William J. Hogan ◽  
...  

2005 ◽  
Vol 74 (5) ◽  
pp. 402-406 ◽  
Author(s):  
E. Jantunen ◽  
E. Koivunen ◽  
M. Putkonen ◽  
T. Siitonen ◽  
E. Juvonen ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document