scholarly journals Clonal Hematopoiesis Drives Therapy-Related Myeloid Neoplasms Following Autologous Stem Cell Transplantation and Propagates during Disease Evolution

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.

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.


2018 ◽  
Vol 8 (2) ◽  
pp. 177-180
Author(s):  
Mohammed Mosleh Uddin ◽  
Huque Mahfuz ◽  
Md Mostafil Karim

Haematopoietic stem cell transplantation (HSCT) involves the intravenous infusion of autologous or allogenic stem cells collected from bone marrow, peripheral blood or umbilical cord to re-establish haematopoietic function in patients whose bone marrow or immune system is damaged or defective. HSCT are mainly of two types –autologous stem cell transplantation (SCT) and allogenic SCT. Autologous SCT is mainly performed in multiple myeloma, Hodgkin lymphoma, non-Hodgkin lymphoma and less commonly in acute myeloid leukaemia. Haematopoietic stem cells are mobilized from bone marrow to the peripheral blood after the use of mobilizing agents, granulocyte colony stimulating factor (G-CSF) and plerixafor. Then the mobilized stem cells are collected from peripheral blood by apheresis and cryo-preserved. The patient is prepared by giving conditioning regimen (high dose melphelan). Stem cells, which are already collected, are re-infused into patient’s circulation by a blood transfusion set. Engraftment happens 7-14 days after auto SCT. Common side effects of this procedure include nausea, vomiting, diarrhoea, mucositis, infections etc. The first case of SCT performed in Combined Military Hospital, Dhaka, Bangladesh is presented here.Birdem Med J 2018; 8(2): 177-180


Cancers ◽  
2021 ◽  
Vol 13 (13) ◽  
pp. 3190
Author(s):  
Alexander D. Heini ◽  
Naomi Porret ◽  
Reinhard Zenhaeusern ◽  
Annette Winkler ◽  
Ulrike Bacher ◽  
...  

Introduction: Despite a 50% cure rate, relapse remains the main cause of death in patients with acute myeloid leukemia (AML) consolidated with autologous stem cell transplantation (ASCT) in first remission (CR1). Clonal hematopoiesis of indeterminate potential (CH) increases the risk for hematological and cardiovascular disorders and death. The impact of CH persisting after ASCT in AML patients is unclear. Materials and Methods: We retrospectively investigated the prognostic value of persisting DNMT3A, TET2, or ASXL1 (DTA) mutations after ASCT. Patients underwent stratification depending on the presence of DTA mutations. Results: We investigated 110 consecutive AML patients receiving ASCT in CR1 after two induction cycles at our center between 2007 and 2020. CH-related mutations were present in 31 patients (28.2%) after ASCT. The baseline characteristics were similar between patients with or without persisting DTA mutations after ASCT. The median progression free survival was 26.9 months in patients without DTA mutations and 16.7 months in patients with DTA mutations (HR 0.75 (0.42–1.33), p = 0.287), and the median overall survival was 80.9 and 54.4 months (HR 0.79 (0.41–1.51), p = 0.440), respectively. Conclusion: We suggest that DTA-CH after ASCT is not associated with an increased risk of relapse or death. The persistence of DTA mutations after induction should not prevent AML patients in CR1 from ASCT consolidation. Independent studies should confirm these data.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3888-3888
Author(s):  
Catharina Hazenberg ◽  
Fiona A.J. van den Heuvel ◽  
Edo Vellenga ◽  
Annet Z. Brouwers-Vos ◽  
Gerbrig Berger ◽  
...  

Abstract Autologous stem cell transplantation (ASCT) is frequently applied in patients with multiple myeloma and malignant lymphoma. Although adequate steady state hematopoiesis with normal peripheral blood counts is attained after ASCT, marked cytopenias may occur in times of stress such as sepsis or re-exposure to chemotherapy. Our group has previously shown impairment of the hematopoietic stem cell (HSC) compartment 1 year post ASCT (pASCT), reflected by reduced HSC frequency and quiescence, and increased ROS production (Haematologica 2013;98:1264). Considering the essential role for mesenchymal stem cells (MSCs) in supporting hematopoiesis, we studied the MSC compartment 1 year post ASCT. Bone marrow biopsies from pASCT patients (n=17) were studied and compared to normal bone marrow from healthy donors (NBM, n=20) by performing immunohistochemistry staining of endothelial cells by CD34 (indicating microvessel density, MVD) and MSCs by nestin, CD146 (Melanoma Cell Adhesion Molecule, MCAM) and CD271 (Nerve Growth Factor Receptor, NGFR). A significant increase in CD271+ MSCs was observed in pASCT bone marrow biopsies compared to NBM (p<0.0001), while the expression of additional markers did not differ between pASCT vs. NBM. MSCs were cultured from the CD34- fraction of bone marrow mononuclear cells, obtained from pASCT patients (n=17) and MSCs derived from NBM (n=20). MSCs were selected by their plastic-adherency and replated to generate MSCs. Although pASCT MSCs and NBM MSCs had similar population doubling times (1.92±0.22 and 3.52±1.02 in passage 4 (P4), pASCT MSCs cultured in vitro demonstrated a change in morphology from the onset of P4. We also observed premature exhaustion of growth in 45% of the studied patients at P5 (vs. 18% in NBM) and increased senescence shown by B-galactosidase staining in P5/P6 (p=0.04). Differentiation assays did not show impairment in differentiation towards osteoblasts or adipocytes of pASCT MSCs. Gene expression analysis on early passage MSCs showed upregulation of pro-inflammatory and cell cycle genes, such as IL6 and p21, in pASCT MSCs compared to NBM MSCs. Co-culture studies with cord blood-derived CD34+ cells on pASCT MSCs showed a significant reduction in output in CFC assays and significant reduction in number of cobblestone-area forming cells in pASCT co-cultures versus NBM (p < 0.05). Given the higher incidence of MDS and AML after ASCT, we questioned whether the observed phenotype of pASCT MSCs resembles MSCs from patients with MDS and AML. Therefore the endothelial and mesenchymal compartments of MDS (n=20) and AML (n=23) patients were studied. An increase in MVD was detected in MDS/AML bone marrow biopsies in contrast to NBM and pASCT (p < 0.05), while the expression of CD146, CD271 and nestin in MDS/AML patients was not significantly increased. 25% of AML MSC cultures showed no growth in the first passage. When MSC growth did occur, the remaining cultures did not show a difference in population doubling time or expansion. However, a change in morphology of MDS/AML MSCs similar to pASCT MSCs was observed. Studies of early passages of MDS/AML MSCs demonstrated a significantly increased gene expression of IL-6 and p21 comparable to pASCT MSCs. In addition PITX2 and Foxc1 expression was increased but no difference was observed in pASCT vs. MDS/AML MSCs. PITX2 has been linked to increased senescence of MDS MSCs while Foxc1 is linked to adipo-osteoprogenitor cell differentiation thereby affecting the HSC compartment. Since none of the pASCT patients did develop MDS, immunohistochemical stainings were also performed on bone marrow biopsies of patients that developed therapy related (t-)MDS/AML following ASCT for lymphoma and myeloma (n=7), after a mean of 117 (MDS) and 50 months (AML). An increase in MVD was observed shortly before or during MDS/AML development, which is probably related to the emergence of malignant cells. No major changes in the phenotype of the MSC compartment were observed before or during the emergence of t-MDS/AML, indicating that t-MDS/AML is preceded by an increase in MVD without distinct changes in the MSC compartment. In summary our results demonstrate that MSCs are affected after ASCT, as shown by expression pattern and functionality. These changes result in a pro-inflammatory phenotype with premature senescence and impaired support of hematopoietic cells, which may account for the reduced bone marrow reserve observed in pASCT patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1809-1809
Author(s):  
Parneet Cheema ◽  
Sunil Desai ◽  
Ronald Anderson ◽  
Max Coppes ◽  
Victor A. Lewis

Abstract INTRODUCTION: The use of peripheral blood as a source of stem cells for pediatric allogeneic stem cell transplantation remains controversial. We present a retrospective analysis evaluating the safety and efficacy of allogeneic peripheral blood stem cell transplantation (PBSCT) compared to allogeneic bone marrow transplantation (BMT) for the treatment of acute leukemia at our centre. The impact of ATG added to the treatment regimen was also evaluated in this analysis. METHODS: A chart review was conducted on all pediatric patients who underwent an allogeneic stem cell transplant for acute leukemia between April 1991 and August 2003 at the Alberta Children’s Hospital. All patients who received stem cells from HLA matched or one-antigen mismatched donors were included in this analysis. Data were analyzed using SPSS. RESULTS: 85 children (ages 9 months to 17 years) met study criteria. 24 patients underwent a PBSCT while 61 patients, a BMT for the treatment of either ALL (n=54) or AML/MDS (n=31). Median follow up time was 5.9 years. Six of 24 patients in the PBSCT group, and 30 of the 61 patients in the BMT group received ATG (p=.042). There was a trend towards increased overall survival (OS) at 5 years following PBSCT (p=.078). Disease free survival (DFS) at 5 years following PBSCT and BMT was similar (p=.297). Subgroup analysis found no difference in OS or DFS between PBSCT and BMT in patients receiving (p=.221, p=.820) or not receiving ATG as part of the conditioning regimen (p=.448, p=.820). Chronic GVHD was increased following PBSCT (45.8%) compared to BMT (19.7%) (p=.015). The rate of severe cGVHD, however, was not significantly different (25% vs. 11.5% for PBSCT and BMT, p=.119) between the two groups. No patients receiving ATG prior to PBSCT developed cGVHD (p=.009). Addition of ATG to the transplant regimen had no impact on the rate of cGVHD following BMT (p=.221). There was no increase in mortality due to cGVHD following PBSCT (p=.243) or BMT (p=.865). Although, there was a trend to improved DFS in patients who developed cGVHD following PBSCT, this was not statistically significant (p=.057). Despite a statistically significant increase in TRM found in all patients with cGVHD at one year post-transplantation (p=.048), there was no difference in TRM between the PBSCT (4.2%) and BMT (14.8%) groups (p=.173). CONCLUSION: Allogeneic PBSCT is a safe and effective alternative to allogeneic BMT for the treatment of acute leukemia in the pediatric population. Although there is an increased rate of cGVHD following PBSCT this does not appear to influence OS, DFS or TRM in our analysis. Addition of ATG into the transplant regimen decreases the occurrence of cGVHD after PBSCT. Studies to further assess the role of ATG in PBSCT are warranted.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 965-965 ◽  
Author(s):  
Christiane Pott ◽  
Eva Hoster ◽  
Kheira Beldjord ◽  
Elizabeth A. Macintyre ◽  
Sebastian Böttcher ◽  
...  

Abstract Abstract 965 Background and Methods: In the MCL younger trial of the European MCL Network, patients up to 65 years with stage II-IV mantle cell lymphoma were randomized to either a standard arm with 6 cycles of 3-weekly R-CHOP, stem cell mobilization with DexaBEAM and myeloablative treatment with 12 Gy TBI, 2×60mg/kg cyclophosphamide and autologous stem cell transplantation (ASCT) (Arm A), or an experimental treatment arm including 6 cycles of alternating R-CHOP/R-DHAP followed by a high dose Ara-C containing myeloablative therapy with 10 Gy TBI, 4×1.5 g/m2 Ara-C, 140mg/m2 melphalan and ASCT (Arm B). In addition to clinical response assessment, minimal residual disease (MRD) was prospectively monitored on the molecular level by real time quantitative (RQ-) PCR in both arms. MRD samples were collected at diagnosis, at midterm (after 4 induction cycles), after induction (6 cycles), and in 3-monthly intervals after ASCT until clinical progression. MRD results were evaluated according to ESG criteria (van der Velden, Leukemia 2007) and compared to clinical response and outcome. RQ-PCR was designed to reach a sensitivity of 10E-5, MRD negativity (MRD-) was defined as a negative RQ-PCR result with a technical assay sensitivity of at least 10E-04. Molecular response (MR) was defined as MRD- in peripheral blood (PB) and/or bone marrow (BM) at any sampling time point. Results: Until May 2010, 307/422 patients randomized in Germany or France had MRD samples available and a molecular marker for RQ-PCR (158 patients of the R-CHOP arm and 149 of the R-CHOP/R-DHAP arm). Overall 2374 samples, 1639 PB and 735 BM were evaluated for MRD. Clinical parameters as stage, LDH elevation, bone marrow infiltration and MIPI were distributed equally in both groups. Based on 173/307 (56%) patients with available midterm samples, MRD clearance was significantly higher in the experimental arm with 36/84 (43%) MRD- patients compared to only 12/89 (13%) in the standard arm; p<0.0001). This difference was confirmed in both, PB (50/80 (63%) vs. 18/84 (21%); p<0.0001) and BM (31/71 (44%) vs. 12/75 (16%); p=0.0003). After completion of induction 90% of R-CHOP and 94% of R-CHOP/R-DHAP patients achieved a clinical response with a CR rate of 26% and 39%, respectively. The rate patients achieving a MR was significantly higher in the R-CHOP/R-DHAP arm (60/82 (73%) vs. 29/92 (32%) after R-CHOP; p<0.0001). BM clearance from lymphoma cells was inferior after R-CHOP only (MRD- 17/71 (24%) compared to R-CHOP/R-DHAP (MRD- 39/57 (68%)). Accordingly, MRD- in the PB was achieved after R-CHOP in only 40/86 patients (47%) compared to 63/79 (80%) in the R-CHOP/R-DHAP arm. Achievement of BM MR after induction was associated with a significantly improved remission duration in the pooled treatment arms (89% vs. 74% at 24 months, p=0.0017). High-dose consolidation followed by ASCT demonstrated a high impact on tumor reduction in the pooled treatment arms and increased the BM MR rate from 50% to 75% (p = 0.0001, paired samples). This improvement was more prominent in the R-CHOP arm (29% to 65%; p = 0.0023) than in the experimental arm (76% to 88%; p=0.18). Remarkably, sustained PB MR during the first year after ASCT was predictive for outcome in both treatment arms (pooled cohort n = 135, 94% vs. 63% ongoing remissions at 2 years, p=0.0001) R-CHOP: 92% vs. 67%, p=0.0224 and R-CHOP/R-DHAP : 96% vs. 53%, p=0.0016). Conclusion: RQ-PCR is an excellent tool to quantitatively determine the impact of different treatment modalities on tumor clearance and revealed the superiority of the high dose Ara-C containing treatment arm early during treatment. Furthermore it underlines the importance of ASCT consolidation in MCL. Thus, MRD seems to be prognostically more relevant than clinical and morphological complete response assessment and was confirmed as a reliable early predictor of clinical outcome in MCL. Disclosures: Ribrag: LFB: Honoraria, Research Funding; servier: Research Funding; celgene: Research Funding; pfizer: Honoraria; novartis: Honoraria.


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

Sign in / Sign up

Export Citation Format

Share Document