bims-tremyl Biomed News
on Therapy resistance biology in myeloid leukemia
Issue of 2025–12–14
twenty-six papers selected by
Paolo Gallipoli, Barts Cancer Institute, Queen Mary University of London



  1. bioRxiv. 2025 Dec 01. pii: 2025.11.27.690982. [Epub ahead of print]
      Aging is the primary risk factor for clonal hematopoiesis and the development of hematologic malignancies ( 1-5 ), yet the selective pressures that shape stem cell behavior and clonal expansion during aging remain poorly defined. Here, we identify proteostasis stress as a central driver of hematopoietic stem cell (HSC) aging and clonal evolution. We show that Heat shock factor 1 (Hsf1) is activated in aging HSCs to preserve proteostasis and sustain self-renewal. However, this physiological, age-associated adaptive mechanism is co-opted by pre-leukemic Dnmt3a -mutant HSCs to resist proteostasis and inflammatory stress required to fuel clonal expansion during aging. In the context of co-occurring Dnmt3a and Nras mutations, which are frequently observed in human acute myeloid leukemia (AML) ( 6-13 ), mutant HSCs and progenitors exhibit heightened dependence on Hsf1 for expansion, malignant transformation and disease progression. Loss of Hsf1 , or disruption of proteostasis, impairs expansion of mutant progenitors, delays leukemia onset, and prolongs survival. Together, these findings reveal proteostasis as a key constraint in the aging hematopoietic system that imposes a selective bottleneck. Hsf1 activation enables both physiological adaptation in aging stem cells and pathological clonal outgrowth in pre-leukemic and leukemic states, establishing proteostasis control as a pivotal mechanism linking stem cell aging to clonal hematopoiesis and malignancy.
    DOI:  https://doi.org/10.1101/2025.11.27.690982
  2. Blood. 2025 Dec 08. pii: blood.2025030816. [Epub ahead of print]
      Measurable residual disease (MRD) can predict relapse in patients with advanced myelodysplastic neoplasms (MDS) or acute myeloid leukemia (AML). We report the long-term efficacy and safety of MRD-guided preemptive azacitidine treatment to prevent relapse in the phase 2 RELAZA2 trial. Patients with MDS or AML after either intensive chemotherapy only or consecutive allogeneic stem cell transplantation were prospectively screened for imminent relapse by molecular MRD assessment. Patients who became MRD positive (MRDpos) during screening received azacitidine for up to 2 years to prevent relapse. The primary endpoint was the proportion of patients alive and relapse-free six months after azacitidine start. Of 357 patients screened, 119 (33.3%) became MRDpos, of whom 95 (79.8%) were eligible for azacitidine treatment. The primary endpoint was met; 60 (63%) patients were relapse-free (95% confidence interval 54-71%, P<0.0001) six months after azacitidine initiation with no new safety signals. Of 60 patients achieving MRD response during the first six cycles of azacitidine, 31 (52%) maintained response without hematological relapse for ≥2 years following azacitidine initiation. The median treatment-free duration following azacitidine discontinuation was 20.8 months; the longest ongoing response was 104 months. After a median follow-up of 6.6 years, 15 initial responders (25%) remained alive and in remission. Among screened patients who remained continuously MRDneg, 60-month overall survival and relapse-free survival were 88% and 79%, respectively. Continuously MRDneg patients display a very favorable prognosis. A majority of MRDpos patients can be effectively treated with azacitidine with potential long-term remission even after termination of azacitidine. Clinicaltrials.gov: NCT01462578.
    DOI:  https://doi.org/10.1182/blood.2025030816
  3. Am J Hematol. 2025 Dec 06.
      Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative option for secondary acute myeloid leukemia (sAML). This study compared haploidentical donor (Haplo), matched sibling donor (MSD), and matched unrelated donor (MUD) HSCT in patients with sAML in first complete remission (CR1). Data from 3862 patients (Haplo = 643, MSD = 715, MUD = 2504) transplanted between 2010 and 2022 were analyzed, with a median follow-up of 3.3 years. Groups differed in patient and donor age, conditioning regimen, stem cell source, and graft-versus-host disease (GVHD) prophylaxis. Multivariate analysis showed that MSD-HSCT had a higher relapse risk than Haplo-HSCT (hazard ratio [HR]: 1.64) but lower non-relapse mortality (NRM, HR: 0.32) and acute GVHD risk (HR: 0.54 for grade II-IV), leading to an overall survival (OS) benefit (HR: 0.76). MUD-HSCT had lower NRM than Haplo-HSCT (HR: 0.63) but there were no significant differences in OS or GVHD risk. Donor type did not impact leukemia-free survival (LFS) or GVHD-free and relapse-free survival (GRFS). Adverse cytogenetics and reduced-intensity conditioning were associated with increased relapse risk, while lower Karnofsky scores, older age, and adverse cytogenetics independently predicted worse NRM, LFS, OS, and GRFS outcomes. Female-to-male donor-recipient pairs had an increased risk of chronic GVHD. In this registry-based analysis, MSD offered the best outcomes for sAML in CR1. Haplo-HSCT was comparable to MUD-HSCT, despite a higher NRM, and achieved long-term disease control in 60% of patients due to a low relapse incidence. In the absence of an MSD, both Haplo and MUD are viable alternatives.
    Keywords:  allogeneic hematopoietic cell transplantation; haploidentical donor; matched sibling donor; matched unrelated donor; secondary acute myeloid leukemia
    DOI:  https://doi.org/10.1002/ajh.70164
  4. Leuk Lymphoma. 2025 Dec 08. 1-23
      Myeloproliferative neoplasms (MPNs) are a spectrum of clonal hematologic malignancies, characterized by an acquired somatic mutation in hematopoietic stem cells (HSC). Consequent constitutive activation of the JAK/STAT signaling pathway ultimately leads to HSC clonal expansion, a heightened inflammatory state, and aberrant trafficking of the malignant stem cells to sites of extramedullary hematopoiesis. While polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF) are distinct disease entities, each with their own diagnostic criteria, risk stratification, and molecular profiles, they share a common pathogenesis and exist on a spectrum, with overlapping clinical features, propensity for thrombohemorrhagic events, and risk for transformation to acute leukemia. Interferon alpha (IFN-α) has both anti-proliferative and immunomodulatory effects on MPN HSCs, and therefore is an effective treatment modality for PV, ET, and MF. In this review, we discuss the rationale for IFN-α use in MPNs, examine the evidence supporting its use, and convey practical considerations.
    Keywords:  Interferon-alpha; essential thrombocythemia; myelofibrosis; myeloproliferative disorders; neoplasia; pegylated interferon alpha-2a; polycythemia vera
    DOI:  https://doi.org/10.1080/10428194.2025.2594038
  5. Haematologica. 2025 Dec 11.
      Acute myeloid leukemia (AML) remains difficult to cure, in part related to strong genetic and functional heterogeneity between and within individual patients. Metabolic reprogramming is emerging as an important feature of AML cells, allowing to explore alternative treatment strategies. Here, we describe a novel DHODH inhibitor, JNJ-74856665, that showed strong efficacy in a subset of AML samples. In a multi-omics approach, by combining label-free quantitative proteome data with drug sensitivity data in bone marrow stromal cocultures in a large cohort of primary AML patient samples we identified that sensitivity to DHODH inhibition (DHODHi) is linked to cholesterol and lipid metabolism. DHODHi resulted in an accumulation of cholesterol, mitochondrial ROS and lipid peroxidation. LC-MS/MS-based lipidomics studies revealed that DHODHi resulted in a strong increase in polyunsaturated fatty acids (PUFAs) and triglycerides (TGs), which are the primary lipid species stored in lipid droplets (LDs). We hypothesized that this might be the consequence of increased ROS and lipid peroxidation levels, prompting the cell to detoxify such toxic lipid species by storing them in LDs. Indeed, we could observed a marked increase in LD formation upon DHODHi. The transcriptional regulator SREBF2, known to control cholesterol and lipid metabolism, was upregulated in DHODHi sensitive AMLs, and a strong synergy was observed between combination of both DHODHi and the SREBP inhibitor dipyridamole. Our data indicate that combined DHODH and SREBP inhibition is of interest to explore further as a therapeutic target option in AML.
    DOI:  https://doi.org/10.3324/haematol.2025.287918
  6. Leukemia. 2025 Dec 12.
      Runt-related transcription factor 1 (RUNX1) is a key regulator of hematopoietic differentiation. RUNX1 mutations in acute myeloid leukemia (AML) are associated with poor prognosis. One-third are frameshift mutations encoding an oncogenic protein with an elongated C-terminus translated in an alternative reading frame. Here, we investigated whether the alternative reading frame of oncogenic RUNX1 can be targeted by immunotherapy. We introduced a construct with a RUNX1 frameshift mutation into B-cell lines with common HLA class I alleles and identified 13 neopeptides by immunopeptidomics. To investigate whether these peptides are neoantigens, peptide-MHC tetramers were used to screen healthy individuals for RUNX1 neoantigen-specific CD8 T cells. T-cell clones were isolated against 5 neoantigens in 4 HLA alleles. Two neoantigens were recognized on an HLA-B*07:02-positive AML cell line with an endogenous RUNX1 frameshift mutation. The T-cell receptors (TCRs) of these clones were sequenced, and analyzed after transfer into CD8 T cells. One TCR induced effective killing of RUNX1-mutated AML cells in vitro and in immunodeficient mice. TCR-engineered T cells also killed patient-derived AML cells, including leukemic stem cells. In conclusion, we showed that RUNX1 frameshift mutations can be effectively targeted, demonstrating the potential relevance of TCR-based immunotherapy to treat patients with RUNX1-mutated AML.
    DOI:  https://doi.org/10.1038/s41375-025-02817-x
  7. Exp Hematol. 2025 Dec 04. pii: S0301-472X(25)00621-6. [Epub ahead of print] 105332
      Acute myeloid leukemia (AML) is characterized by the proliferation of malignant myeloid progenitor cells and impairment of hematopoiesis. Although genetic abnormalities within leukemic cells have been investigated in detail, definitive explanations for the damage to the normal hematopoietic system are lacking. Here, we investigated the mechanisms underlying the impairment of the residual hematopoietic system in the bone marrow in AML. We evaluated the function of residual non-leukemic (nl)-hematopoietic stem / progenitor cell (HSPC) from the bone marrow of mice with MLL-AF9-induced AML. The nl-HSPC in the leukemic marrow showed a megakaryocyte (MgK) and myeloid-biased gene expression signature, with enrichment of TNF signaling and reduced repopulation ability. To investigate whether the upregulation of TNF signaling causes the MgK / myeloid lineage bias, we investigated the effects of TNF-α in normal hematopoietic stem cells (HSC) / HSPC under ex-vivo expansion condition. Single-cell transcriptome analysis of these cells revealed an increased frequency of cells expressing genes related to the MgK lineage and decreased repopulation capacity compared with those of ex vivo-expanded HSC / HSPC without TNF-α. Our data suggest that increased TNF-α in the leukemic bone marrow environment at least in part drives HSPC toward MgK / myeloid differentiation, resulting in the exhaustion of residual normal HSC / HSPC. These findings offer valuable insights into leukemic biology and normal hematopoiesis.
    Keywords:  Acute myeloid leukemia; bone marrow environment; hematopoietic stem progenitor cells; tumor necrosis factor
    DOI:  https://doi.org/10.1016/j.exphem.2025.105332
  8. Hemasphere. 2025 Dec;9(12): e70262
      Clonal hematopoiesis (CH), a prevalent and premalignant state in the elderly, has been detected in young individuals under selective pressures such as hematopoietic cell transplantation (HCT). However, the origin of CH and mutational processes underlying CH driver mutations in young blood systems remain unclear. Here, we used genome-wide somatic mutation profiles to retrospectively trace the origin of DNMT3A-mutant CH in three individuals, 14-41 years after childhood HCT. Both the rate and spectrum of somatic mutations in individuals with posttransplant CH were consistent with normal age-associated mutagenesis. Phylogenetic analysis revealed that DNMT3A-mutant HSPCs were present in the donor before 6.8 years of age, including during fetal development, despite being undetectable with a limit of detection of variant allele frequency of 0.001 at the time of transplantation. These findings were validated by comparing the observed mutations to expected age-dependent mutational signatures. Our results reveal that undetectable DNMT3A-mutant clones in young donors can expand into significant CH clones within decades upon transplantation. The rapid expansion of these clones in this context indicates that specific environmental pressures, rather than solely mutation acquisition, drive the development of CH.
    DOI:  https://doi.org/10.1002/hem3.70262
  9. Cancer Discov. 2025 Dec 06. OF1-OF30
    BDRL
      Accumulating evidence links pediatric cancers to prenatal transformation events, yet the influence of the developmental stage on oncogenesis remains elusive. We investigated how hematopoietic stem cell developmental stages affect leukemic transformation, disease progression, and therapy response using a novel, humanized model of NUP98::NSD1-driven pediatric acute myeloid leukemia that is particularly aggressive with WT1 comutations. Fetal-derived hematopoietic stem cells readily transform into leukemia, and WT1 mutations further enhance stemness and alter lineage hierarchy. In contrast, stem cells from later developmental stages become progressively resistant to transformation. Single-cell analyses revealed that fetal-origin leukemia stem cells exhibit greater quiescence and reliance on oxidative phosphorylation than their postnatal counterparts. These differences drive distinct therapeutic responses despite identical oncogenic mutations. In patients, onco-fetal transcriptional programs correlate with worse outcomes. By targeting key vulnerabilities of fetal-origin leukemia cells, we identified combination therapies that significantly reduce aggressiveness, highlighting the critical role of ontogeny in pediatric cancer treatment.
    SIGNIFICANCE: This study signifies the critical consequences of developmental timing in cancer initiation, revealing that identical driver mutations in fetal- versus postnatal-origin leukemias exhibit fundamentally distinct biology and treatment responses. Recognizing these developmental differences opens avenues for personalized therapeutic strategies, improving outcomes for pediatric patients with aggressive disease subtypes in leukemia.
    DOI:  https://doi.org/10.1158/2159-8290.CD-25-0556
  10. Hemasphere. 2025 Dec;9(12): e70270
      Atypical chronic myeloid leukemia (aCML) is a rare form of myelodysplastic (MDS)/myeloproliferative neoplasm (MPN) overlap disorder characterized by neutrophilic leukocytosis with circulating immature myeloid cells (IMC), frequent hepatosplenomegaly, and poor prognosis with high rates of leukemic transformation. In the 2022 World Health Organization (WHO) classification, aCML was therefore renamed to "MDS/MPN with neutrophilia." Diagnostic criteria for aCML include leukocytosis ≥ 13 × 109/L, circulating IMC ≥ 10%, dysgranulopoiesis, and at least one cytopenia for MDS-thresholds (per International Consensus Classification [ICC]). Bone marrow is hypercellular due to granulocytic proliferation, associated with dysplasia in granulocytes ± other cell lines. Mutations can be used to support the diagnosis in both classifications, that is, SETBP1 and ASXL1 or SETBP1 and/or ETNK1. Absence of monocytosis, basophilia, or eosinophilia, <20% blasts, and exclusion of other MPN, MDS/MPN, and tyrosine kinase fusions are mandatory. Cytogenetic abnormalities are identified in roughly 20%-40% of aCML patients, along with a complex genomic context with high rates of recurrent somatic mutations in ASXL1, SETBP1, SRSF2, TET2, EZH2, and, less frequently, in NRAS/KRAS, CBL, CSF3R, JAK2, and ETNK1. Unfortunately, effective risk stratification systems for identifying prognostic subgroups of aCML patients are lacking, resulting in the absence of a standard of care for its management. The most used agents include hydroxyurea, interferon, hypomethylating agents, and JAK inhibitors, although none of them are disease-modifying. Allogeneic hematopoietic stem cell transplant remains the only potentially curative approach and should be considered in all eligible patients. Actionable mutations (CSF3R, NRAS/KRAS, and KIT) have also been identified, supporting the development of new agents targeting the involved pathways.
    DOI:  https://doi.org/10.1002/hem3.70270
  11. Br J Haematol. 2025 Dec 12.
      Myelodysplastic syndromes (MDS) are heterogeneous stem cell disorders with a 30%-40% risk of transformation to acute myeloid leukaemia (AML). This study characterised the genetic and clinicopathological features of 437 newly diagnosed MDS patients. A predictive model for AML transformation was developed, which identified the bone marrow blast percentage, KRAS, STAG2 and TP53 mutations as significant predictors of disease progression. Novel prognostic models for overall survival (OS) and progression-free survival (PFS) were established and validated. The OS model incorporated age, sex and mutations in TP53, NF1, SRSF2, CSF3R, ETV6, CEBPA, BCOR, TET2, JAK2 and SF3B1. The PFS model incorporated age, sex, bone marrow blast percentage and mutations in STAT3, TP53, NF1, CEBPA, ETV6, CALR, DNMT3A, IDH2, GATA2, BCOR, JAK2, TET2 and SF3B1. These models stratify patients into favourable, intermediate-1, intermediate-2 and adverse risk groups, demonstrating superior risk stratification compared to the Revised/Molecular International Prognostic Scoring System (IPSS-R/M). This work provides the first genomic characterisation of a diagnosed MDS cohort in China and establishes the first risk prediction model for MDS-to-AML transformation, alongside novel prognostic models for OS and PFS. These tools offer improved prognostic prediction and potential guidance for therapeutic strategies in Chinese patients with MDS.
    Keywords:  acute myeloid leukaemia; gene mutation; myelodysplastic syndromes; next‐generation sequencing; prognostic score
    DOI:  https://doi.org/10.1111/bjh.70284
  12. JCO Precis Oncol. 2025 Dec;9 e2500494
       PURPOSE: Mixed phenotype acute leukemia (MPAL) is a rare clinical entity with historically poor outcomes.
    METHODS: We conducted a retrospective analysis of adults 18 years and older with newly diagnosed B-cell (B/M) or T-cell/myeloid (T/M) MPAL treated at our institution between 2017 and 2024.
    RESULTS: We identified 42 patients (median age 70 years); 20 (48%) had B/M MPAL, and 22 (52%) had T/M MPAL; 57% of patients had adverse risk cytogenetics, and 41% had a TP53 mutation. Sixty-two percent of patients were treated with a hybrid regimen, and 45% of patients received intensive therapy. A composite complete remission (CRc; CR + CRi) was achieved in 57% of patients (86% measurable residual disease [MRD]-negative). After a median follow-up of 27.9 months, the median relapse-free survival in patients achieving an overall response (CRc + morphological leukemia-free state) was 10.1 months, 17.8 months in those who achieved a CRc, and not reached (NR) in patients with MRD-negative CRc. The median overall survival (OS) for all patients was 9.5 months and NR for patients achieving a CRc. Although patients with T/M MPAL had a trend toward improved survival compared with those with B/M MPAL (median OS of 9.1 v 25 months P = .28), this difference abrogated when comparison was stratified by treatment intensity. Twelve patients (29%) underwent allogeneic hematopoietic stem-cell transplantation (HSCT); on landmark analysis, HSCT trended to improve OS (NR v 22.8, P = .12). Multivariate Cox analysis demonstrated that TP53 mutation was associated with increased hazards for death (hazard ratio [HR], 3.5, P = .01), whereas the use of intensive chemotherapy trended to be favorable (HR, 0.45, P = .11).
    CONCLUSION: Overall, these data demonstrate the need for treatment intensification in MPAL with HSCT in first remission for best outcomes.
    DOI:  https://doi.org/10.1200/PO-25-00494
  13. Blood Adv. 2025 Dec 09. pii: bloodadvances.2025016400. [Epub ahead of print]
      Interactions between acute myeloid leukemia (AML) and the bone marrow microenvironment (BMME) are critical to leukemia progression and chemoresistance. In the solid tumor microenvironment, altered metabolite levels contribute to cancer progression. We performed a metabolomic analysis of AML patient bone marrow serum, revealing increased metabolites compared to age- and sex-matched controls. The most highly elevated metabolite in the AML BMME was lactate. Lactate signaling in solid tumors induces immunosuppressive tumor-associated macrophages and correlates with poor prognosis. This has not yet been studied in the leukemic BMME. Herein, we describe the role of lactate in the polarization of leukemia-associated macrophages (LAMs). Using a murine AML model of blast crisis chronic myelogenous leukemia (bcCML), we characterize the suppressive phenotype of LAMs by surface markers, transcriptomics, and cytokine profiling. Then, mice genetically lacking GPR81, the extracellular lactate receptor, were used to demonstrate GPR81 signaling as a mechanism of both the polarization of LAMs and the direct support of leukemia cells. Furthermore, elevated lactate diminished the function of hematopoietic progenitors and reduced stromal support for normal hematopoiesis. We report microenvironmental lactate as a mechanism of AML-induced immunosuppression and leukemic progression, thus identifying GPR81 signaling as an exciting and novel therapeutic target for treating this devastating disease.
    DOI:  https://doi.org/10.1182/bloodadvances.2025016400
  14. Leuk Res. 2025 Dec 03. pii: S0145-2126(25)00635-6. [Epub ahead of print]160 108145
      We compared outcomes of patients ≥ 70 years old undergoing allogeneic stem cell transplantation (alloSCT) with graft-vs-host disease (GvHD) prophylaxis regimens either including post-transplant cyclophosphamide (PTCy) or without cyclophosphamide (non-Cy). The primary endpoint was GvHD-free, relapse-free survival (GRFS) at one and five years; secondary endpoints included clinically significant acute (grade III-IV) and chronic (extensive) GvHD, relapse, overall survival (OS), and non-relapse mortality (NRM). Among 61 patients, 41 received PTCy and 20 received non-Cy prophylaxis. Unrelated donors accounted for 80 % of allografts; all non-Cy patients had 10/10 HLA matches, while PTCy patients had 64 % matched, 29 % haploidentical, and 7 % mismatched unrelated donors. Acute GvHD occurred in 5 % of PTCy vs 15 % of non-Cy patients (p = ns). One-year chronic GvHD incidence was lower with PTCy (12 % vs 30 %, p = 0.03). One-year GRFS was similar (34 % PTCy, 35 % non-Cy; p = ns). At five years, OS was 20 % vs 30 % and GRFS 21 % vs 15 % for PTCy and non-Cy, respectively (p = ns). We observed similar outcomes among patients receiving GvHD prophylaxis with PTCy compared to non-Cy. Importantly, non-Cy patients had HLA-matched donors, whereas mismatched donors were possible for the PTCy group. In this way, PTCy seems to have equalized outcomes for fully matched and mismatched alloSCT by yielding similar one and five-year GRFS. We also found no significant difference in relapse rate, NRM, OS, and five-year GRFS between patients aged 70-74 and ages 75 + , showing that numerical age should not be a contraindication to alloSCT.
    Keywords:  Allogeneic; Cyclophosphamide; Elderly; GVHD; Geriatric; Leukemia; Myeloid; Transplantation
    DOI:  https://doi.org/10.1016/j.leukres.2025.108145
  15. Blood. 2025 Dec 12. pii: blood.2025031313. [Epub ahead of print]
      To prevent graft-versus-host disease (GVHD) in patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation (alloHSCT), a calcineurin inhibitor plus methotrexate is routinely used. Early phase studies suggested improved outcomes with Orca-T, an allogeneic T-cell immunotherapy that uses purified donor Treg cells to prevent GVHD with significantly less immunosuppression. This phase 3 trial randomized adult patients (N=187) with acute leukemias or myelodysplastic syndrome undergoing myeloablative conditioning to receive either Orca-T with tacrolimus or a conventional allograft with tacrolimus and methotrexate (Tac/MTX), using granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood from HLA-matched donors. The primary endpoint was survival free from moderate-to-severe chronic GVHD (cGFS). Using a stratified log-rank test, cGFS was significantly higher in the Orca-T arm compared to Tac/MTX (P<0.001; HR 0.26; 95% CI, 0.14 to 0.47). One-year estimates were as follows: cGFS was 78.0% with Orca-T versus 38.4% with Tac/MTX; cumulative incidence of moderate-to-severe chronic GVHD (cGVHD) was 12.6% with Orca-T and 44.0% with Tac/MTX (Gray's test P<0.001), while overall survival (OS) was 93.9% with Orca-T versus 83.1% with Tac/MTX (P=0.12); GVHD and relapse-free survival (GRFS) was 63.1% and 30.9% in the Orca-T and Tac/MTX arms (P<0.001), respectively; non-relapse mortality (NRM) was 3.4% with Orca-T versus 13.2% with Tac/MTX (P=0.03). Orca-T met the primary endpoint of improved survival free from cGVHD compared to Tac/MTX prophylaxis and should be considered a new therapeutic option with low toxicity for GVHD prophylaxis. Additionally, significantly less toxicity was observed with Orca-T patients including fewer serious infectious complications and less non-relapse mortality. (ClinicalTrials.gov number NCT05316701).
    DOI:  https://doi.org/10.1182/blood.2025031313
  16. Exp Hematol. 2025 Dec 07. pii: S0301-472X(25)00625-3. [Epub ahead of print] 105346
      Selenoproteins-a unique class of antioxidant enzymes characterized by the incorporation of selenocysteine at their catalytic core-function as pivotal regulators of redox homeostasis in hematopoiesis. This review elucidates how selenoprotein-mediated redox control orchestrates hematopoietic stem cell (HSC) fate decisions, maintaining the critical balance between self-renewal and differentiation. The glutathione peroxidase (GPX) family, particularly GPX1 and GPX4, plays indispensable roles in hydrogen peroxide detoxification and protection against lipid peroxidation-induced ferroptotic cell death, respectively. Similarly, thioredoxin reductases (TXNRDs) sustain critical redox equilibrium via thioredoxin regeneration. Recent studies demonstrate that these redox regulators facilitate the proliferation and differentiation of HSCs and mature lineages. Selenoprotein deficiency disrupts HSC fitness, impairs B- and erythroid-lineage maturation, and induces B-to-myeloid lineage switching-pathogenic features observed in aged hematopoiesis, highlighting the critical roles of selenoproteins in balanced, healthy hematopoiesis. Emerging evidence demonstrates that leukemic cells also exploit selenoprotein pathways to mitigate oxidative stress, suggesting that selective modulation of specific selenoproteins may constitute a promising therapeutic approach, provided we delineate their differential utilization between normal and malignant hematopoiesis. Selenoproteins function at the intersection of several transcriptional networks, including NRF2, whose orchestrated antioxidant responses may alter during aging and malignant transformation. Indeed, selenoproteins possess unique properties and function with tissue-specific expression patterns and non-redundant or redundant functions across different hematopoietic lineages. Understanding the contribution of selenoproteins to hematopoietic regulation offers promising avenues for developing targeted therapeutic strategies in hematological disorders and rejuvenating aged hematopoiesis, potentially through precision-guided modulation of selenoprotein-dependent pathways.
    DOI:  https://doi.org/10.1016/j.exphem.2025.105346
  17. Blood. 2025 Dec 08. pii: blood.2025030898. [Epub ahead of print]
      Recent studies have reached opposing conclusions about whether clonal hematopoiesis (CH) is increased or decreased in patients with sickle cell disease (SCD). Given that CH is typically age-related, its presence in children with SCD could offer unique insights into early-life mutagenesis and disease-related stressors. We tested the primary and secondary hypotheses, that children with SCD would have a higher prevalence of CH when compared to age, sex, and race matched children without SCD; and children with hydroxyurea would have a higher CH prevalence than children not treated with hydroxyurea. To address this, we conducted a cross-sectional study in two independent cohorts of children, ages 0-18 years, with SCD (n=1,025 and n=1,293, respectively) and a 2,957-person matched comparison group. Using a highly sensitive, error-corrected sequencing assay capable of detecting CH at a variant allele frequency ≥ 0.5%, we found that children with SCD have a significantly higher prevalence of CH in relation to the comparison group (odds ratio (OR)=4.2, p=7.4x10-13). Additionally, CH was not associated with exposure to hydroxyurea therapy (OR=0.76, p=0.44).
    DOI:  https://doi.org/10.1182/blood.2025030898
  18. Haematologica. 2025 Dec 11.
      The management of chronic myeloid leukemia in chronic phase (CML-CP) was transfigured with the introduction of imatinib in 2001. Since then, four other tyrosine kinase inhibitors (TKIs), dasatinib, nilotinib, bosutinib and most recently asciminib, have garnered approval for frontline management of CML-CP. The second generation TKIs (2G-TKIs) and asciminib have all been shown to be significantly superior to imatinib in attaining molecular responses, and asciminib possibly superior to 2G-TKIs. With limited prospective comparisons between the 2G-TKIs and similar survival outcomes with imatinib compared to 2G-TKIs, the selection of a TKI for patients with newly diagnosed CML-CP must be individualized to the needs of that specific patient. Important factors to consider when choosing a drug include patient related factors (age, comorbidities, lifestyle considerations, quality of life, patient preferences, shared-decision making and whether treatment free remission [TFR] is a goal), disease related factors (risk stratification, transcript type, presence of high risk gene mutations such as ASXL1) and drug related factors (major molecular response rates with each TKI, adverse events, rates of treatment discontinuation and TFR rates).
    DOI:  https://doi.org/10.3324/haematol.2025.287813
  19. Blood Adv. 2025 Dec 10. pii: bloodadvances.2025016338. [Epub ahead of print]
      TLR8 gain-of-function (GOF) somatic variants were recently identified as causing severe neutropenia, lymphoproliferation, and immune dysregulation. We report the expanded clinical and laboratory phenotype and management of 10 patients, including the original cohort of 6 male patients. We identify the first female patient with TLR8 GOF who presented during infancy with pure red cell aplasia due to a germline TLR8 variant, and two new disease-causing somatic variants in male patients. Eight patients had somatic mosaicism with peripheral blood variant allele fractions of 7-26% and age of disease onset of 9 months to 28 years. All patients had neutropenia, most with severe neutropenia refractory to medical therapy. Anemia and thrombocytopenia were common. Bone marrow characteristically demonstrated severe myeloid hypoplasia and activated T-cell infiltrates and/or aggregates. An increased number of large granular lymphocytes (LGLs) were identified in five patients. Seven patients underwent allogeneic hematopoietic cell transplantation (HCT). High rates of post-HCT cytopenia of unclear etiology and graft-versus-host disease were observed. Five are surviving at 1 to 3 years post-HCT with full donor myeloid and T-cell chimerism and resolution of disease phenotype. The two patients who presented during childhood and did not undergo HCT ultimately died from disease. In conclusion, TLR8 GOF is an X-linked dominant disorder that should be considered in male and female patients with cytopenia, particularly severe neutropenia, lymphoproliferation with immune dysregulation, increased LGLs, and new to this cohort, red cell aplasia. Disease is refractory to medical management, and curative, allogeneic HCT should be considered early after diagnosis. (NCT04339777).
    DOI:  https://doi.org/10.1182/bloodadvances.2025016338
  20. Commun Biol. 2025 Dec 10.
      Numerous strategies exist to isolate hematopoietic stem cells (HSCs) using complex combinations of markers and flow cytometry. However, robust identification of HSCs using imaging techniques is substantially more challenging which has prompted the recent development of HSC reporter mice. To date, very few molecules used in these reporters have been useful for human HSC identification. Here we report that PLXDC2 is a useful marker for both mouse and human cord blood HSCs. Using a green fluorescent protein (GFP) knock-in at the Plxdc2 locus in mice (hereafter denoted as Plxdc2-GFP), we showed that Plxdc2-GFP is highly expressed in HSCs with 1 in 2.8 Plxdc2-GFP+CD150+ cells giving long-term multi-lineage reconstitution in transplantation. Moreover, we developed a novel human PLXDC2 antibody and showed that human PLXDC2+ HSCs have higher long-term multilineage reconstitution ability compared with PLXDC2- HSCs in a xenograft model. This study identifies PLXDC2 as a highly relevant molecule in HSC identification.
    DOI:  https://doi.org/10.1038/s42003-025-09242-x
  21. J Exp Med. 2026 Mar 02. pii: e20250607. [Epub ahead of print]223(3):
      Mechanical force generated by blood flow stimulates emergence of the first hematopoietic stem cells (HSCs) that populate the blood system. Force drives the transition of HSC precursors from an endothelial to hematopoietic identity, yet the molecular regulation of this fate switch remains poorly understood. We report that shear stress triggers adaptation in mitochondrial composition, ultrastructure, and function, which are essential for hematopoietic fate and engraftment potential. Shear stress remodels mitochondria in hemogenic endothelium by promoting mitochondrial gene transcription and protein synthesis. Laminar flow selectively initiates translation of 5' terminal polypyrimidine (5'TOP) motif-containing transcripts, which commonly encode ribosome and translation machinery. Flow-responsive metabolic reprogramming depends upon mechanistic target of rapamycin (mTOR) activation and is stymied when ribosome activity or mTOR is blocked. Conversely, chemical induction of mTOR mimics the effects of force on mitochondria and blood reconstituting potential and also partially rescues hematopoiesis in heartbeat mutants in utero. These findings identify mechanometabolism as a determinant of hematopoietic fate that could inform engineering of HSCs for disease modeling and treatment.
    DOI:  https://doi.org/10.1084/jem.20250607
  22. Blood Adv. 2025 Dec 10. pii: bloodadvances.2025018011. [Epub ahead of print]
      Although phlebotomy and hydroxyurea (HU) are standard first-line therapies for polycythaemia vera (PV), they do not adequately address clonal expansion and chronic inflammation - key drivers of thrombosis, myelofibrosis and mortality. Biomarkers such as JAK2 V617F VAF and the neutrophil-to-lymphocyte ratio (NLR) are emerging as valuable tools for guiding therapy. Ropeginterferon alfa-2b has been shown to reduce both JAK2 VAF and NLR, improving event-free survival as demonstrated in the Low-PV and PROUD-PV/CONTINUATION-PV trials. In contrast, propensity score matching of the ECLAP trial showed that HU has limited effect on these biomarkers, suggesting weaker disease-modifying potential. Although no data on NLR dynamics with ruxolitinib have been published, the anti-inflammatory effects of ruxolitinib and its suppression of JAK2 VAF suggest it may exert similar biological activity. These findings support a shift towards biology-guided treatment in PV, recognising that inflammation and JAK2 VAF could serve as surrogate endpoints in future clinical trials.
    DOI:  https://doi.org/10.1182/bloodadvances.2025018011