bims-tremyl Biomed News
on Therapy resistance biology in myeloid leukemia
Issue of 2025–04–06
33 papers selected by
Paolo Gallipoli, Barts Cancer Institute, Queen Mary University of London



  1. Blood. 2025 Mar 31. pii: blood.2024027117. [Epub ahead of print]
      Acute myeloid leukemia (AML) that evolves from myeloproliferative neoplasm (MPN) is known as post-MPN AML. Current treatments don't significantly extend survival beyond 12 months. BCL-xL has been found to be overexpressed in leucocytes from MPN patients, making it a potential therapeutic target. We investigated the role of BCL-xL in post-MPN AML and tested the efficacy of DT2216, a platelet-sparing BCL-xL proteolysis-targeting chimera (PROTAC), in preclinical models of post-MPN AML. We found that BCL2L1, the gene encoding BCL-xL, is expressed at higher levels in post-MPN AML patients compared to those with de novo AML. Single-cell multi-omics analysis revealed that leukemia cells harboring both MPN-driver and TP53 mutations exhibited higher BCL2L1 expression, elevated scores for leukemia stem cell, megakaryocyte development, and erythroid progenitor than wild-type cells. BH3 profiling confirmed a strong dependence on BCL-xL in post-MPN AML cells. DT2216 alone, or in combination with standard AML/MPN therapies, effectively degraded BCL-xL, reduced the apoptotic threshold, and induced apoptosis in post-MPN AML cells. DT2216 effectively eliminated viable cells in JAK2-mutant AML cell lines, induced pluripotent stem cell-derived hematopoietic progenitor cells (iPSC-HPCs), primary samples, and reduced tumor burden in cell line-derived xenograft model in vivo by degrading BCL-xL. DT2216, either as a single agent or in combination with azacytidine, effectively inhibited the clonogenic potential of CD34+ leukemia cells from post-MPN AML patients. In summary, our data indicate that the survival of post-MPN AML is BCL-xL dependent, and DT2216 may offer therapeutic advantage in this high-risk leukemia subset with limited treatment options.
    DOI:  https://doi.org/10.1182/blood.2024027117
  2. Leuk Lymphoma. 2025 Mar 31. 1-13
      Monosomy 7 and 7q deletions (-7/del(7q)) are the most common adverse cytogenetic event in acute myeloid leukemia (AML), linked to high relapse rates. We analyzed 115 AML patients with -7/del(7q) who achieved remission after induction therapy to characterize the mutational landscape from diagnosis to relapse. Median overall survival (OS) was 10.4 months, with improved survival in patients without TP53 mutation (13.04 vs. 8.6 months) or complex karyotype (12.4 vs. 8.6 months). TP53 mutations were most frequent (67% of cases at diagnosis) and persisted in 97% of patients at relapse. At time of relapse, patients with TP53 mutations had fewer co-occurring mutations in ASXL1, RUNX1, NRAS, PTPN11 and SRSF2 compared to TP53 wild-type patients. Patients with mutated TP53 and co-mutation in NF1, BCORL1, GATA2, or RUNX1 had shorter relapse-free survival (2 vs. 5 months) and OS (7.2 vs. 10.4 months) than those with TP53 mutation alone. Allogeneic transplant improved OS significantly, regardless of TP53 status.
    Keywords:  Acute myeloid leukemia; chromosomal 7 deletions; mutational landscape
    DOI:  https://doi.org/10.1080/10428194.2025.2477723
  3. Blood. 2025 Mar 31. pii: blood.2024025670. [Epub ahead of print]
      Myelodysplastic syndromes (MDS) are myeloid malignancies often driven by mutations in genes encoding splicing factors (SFs). How these mutations drive the clonal expansion of MDS stem/progenitor cells to outcompete normal hematopoietic stem/progenitor cells (HSPCs) remains unexplained. Although a role for inflammatory processes in promoting clonal expansion of mutant HSPCs and MDS pathogenesis has been proposed, the specific mechanisms implicated remain incompletely understood. In this study, using human isogenic induced pluripotent stem cell (iPSC)-based models of SRSF2-mutant MDS and primary MDS patient cells, we show that the SRSF2 P95L mutation downregulates basal STAT1 expression. STAT1 downregulation dampens interferon (IFN) signaling in MDS stem/progenitor cells, which, unlike normal HSPCs, show resistance to suppression of clonogenic ability by IFNs. Treatment with the proteasome inhibitor bortezomib increased STAT1 protein levels and restored sensitivity of SRSF2-mutant cells to inflammatory stimuli. These results indicate that rewiring of STAT1 signaling by SRSF2 mutations blunts responsiveness to IFNs, conferring clonal fitness to SRSF2-mutant HSPCs against normal HSPCs in the presence of inflammatory stimuli. Our study provides a novel mechanistic link between SF mutations and inflammatory dysregulation and suggests proteasome inhibition as a potential strategy to treat MDS with SRSF2 mutations.
    DOI:  https://doi.org/10.1182/blood.2024025670
  4. Clin Cancer Res. 2025 Mar 31.
       PURPOSE: PPM1D, a central regulator of the DNA damage response, is commonly mutated in therapy-related clonal hematopoiesis (CH), acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). PPM1D mutations have been shown to expand under selective pressure of DNA-damaging chemotherapy. However, whether PPM1D mutations promote the development of hematologic malignancies remains unclear.
    EXPERIMENTAL DESIGN: We characterized the clinical and genomic profiles of 112 PPM1D-mutated patients across the spectrum of myeloid disorders using a combination of bulk and single-cell analyses on diagnostic and longitudinal samples.
    RESULTS: Among all patients, 78% had a history of primary cancer, with DNMT3A and TP53 being the most frequent co-mutated genes. In ten patients with high-grade serous ovarian cancer, longitudinal analysis showed variable dynamics of PPM1D-mutant clones, with 81% of clones expanding during exposure to alkylating agents. Clonal hierarchy estimation revealed that 44% of PPM1D-mutated acute myeloid leukemia (AML) patients had a PPM1D mutation in the founder clone, with rare TP53 co-mutations. Both TP53 wildtype and mutated AML patients had poor overall survival. Single cell DNA and surface protein analysis in seven patients confirmed that PPM1D mutations can arise in the founding clone, and were associated with expression of leukemic markers.
    CONCLUSIONS: PPM1D mutated CH clones can spontaneously regress after treatment discontinuation, however they can also be found in the dominant clone in AML/MDS.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-24-3683
  5. Leukemia. 2025 Mar 31.
      Myeloproliferative neoplasms (MPNs) are characterized by clonal proliferation of hematopoietic stem cells, which can lead to secondary myelofibrosis or acute myeloid leukemia. We explored the changes in genomic alterations during MPN transformation using whole-genome sequencing of samples from both the chronic and fibrotic or leukemic phases of 20 patients. We identified FOXP1 mutations in 3 of 14 (21.4%) patients with secondary myelofibrosis. This novel mutation was identified in another 5 of the 35 patients (14.3%) in an independent cohort. All these 8 patients with FOXP1 mutations did not experience leukemic transformation after a median follow-up of 5.1 years. The acquisition of non-canonical MPLY591 mutations was detected in the fibrotic or leukemic phase. Clonal expansion, involving both known and unknown driver genes (in 18 and 2 patients, respectively), was observed in all patients. We determined the patterns of clonal evolution based on myeloid driver mutations in 18 patients: linear clonal evolution in 11 patients and branched clonal evolution in 7 patients. Our results suggested that MPN patients carrying FOXP1 mutations are unlikely to have leukemia transformation and emphasized that the acquisition of specific genetic mutations and dynamic changes in clonal architecture underlie the pathogenesis in patients undergoing MPN transformation.
    DOI:  https://doi.org/10.1038/s41375-025-02576-9
  6. Blood. 2025 Apr 03. pii: blood.2024027244. [Epub ahead of print]
      Inflammation is increasingly recognized as a critical factor in acute myeloid leukemia (AML) pathogenesis. We performed blood-based proteomic profiling of 251 inflammatory proteins in 543 newly diagnosed AML patients. Using a machine learning model, we derived an eight-protein prognostic score termed Leukemia Inflammatory Risk Score (LIRS). Individual proteins were evaluated in multivariable cox models and model performance was assessed by cumulative concordance index. Findings were validated in internal and external cohorts across two institutions. Blood-based LIRS significantly outperformed the European LeukemiaNet (ELN) 2022 risk model and was independently prognostic of overall survival after accounting for known clinical and molecular prognostic factors. OSMR was uniquely identified as the strongest independent predictor of survival, early mortality, and induction chemotherapy response, and further validated in an independent assay. These blood-based biomarkers could have significant clinical implications for risk stratification and prognostication in patients with newly diagnosed AML.
    DOI:  https://doi.org/10.1182/blood.2024027244
  7. Leukemia. 2025 Apr 02.
      Internal tandem duplication mutations in the FMS-like tyrosine kinase 3 (FLT3-ITDs) occur in 25%-30% of acute myeloid leukemia (AML) cases and are associated with adverse prognosis. RNA-based therapeutics exhibit significant potential for treating diseases, prompting us to develop a novel circular RNA (circRNA)-based therapeutic strategy for FLT3-ITD AML. Here, we find circTADA2A is downregulated in FLT3-ITD AML patients. We further demonstrate that the downregulation of circTADA2A is critical for the proliferation of human FLT3-ITD AML cells, the sustenance of AML, and the self-renewal of leukemia stem/initiating cells (LSCs/LICs). Mechanistically, circTADA2A inhibits the TRIM28/MDM2 complexes formation by competitively binding to TRIM28, resulting in decreased levels of p53 ubiquitination and activating the p53 pathway. Importantly, in vitro transcription of circTADA2A and in vivo delivery via lipid nanoparticles (LNPs) significantly enhance the elimination of FLT3-ITD leukemia cells in combination with quizartinib treatment. In conclusion, our work uncovers the crucial functions of circTADA2A in the maintenance of FLT3-ITD AML and highlights a translationally important circTADA2A-based therapeutic approach for FLT3-ITD AML treatment.
    DOI:  https://doi.org/10.1038/s41375-025-02589-4
  8. Cell Rep. 2025 Apr 02. pii: S2211-1247(25)00265-7. [Epub ahead of print]44(4): 115494
      Blood cancers are generally more common in males, and the prevalence of most mutations that drive clonal hematopoiesis and myeloid malignancies is higher in males. In contrast, hematopoietic DNMT3A mutations are more common in females. Among ∼450,000 participants in the UK Biobank, the prevalence of DNMT3A mutations and copy-number abnormalities is higher in females than males. In a murine model, Dnmt3a-mutant hematopoietic stem cells (HSCs) from unperturbed female mice had increased stemness gene expression compared to male and wild-type (WT) mice. Estrogen regulates HSCs, and we found that Dnmt3a mutations maintain stemness in the setting of estrogen-induced proliferative stress. Dnmt3a-mutant myeloid cells outcompeted WT cells under chronic estrogen treatment, an effect that was dependent on cell-intrinsic estrogen receptor alpha activity. Our studies indicate that estrogen might contribute to the female predominance of DNMT3A-mutant clonal hematopoiesis.
    Keywords:  CHIP; CP: Stem cell research; DNMT3A; Esr1; HSC; clonal hematopoiesis; estrogen; female; hormones; sex; stem cell
    DOI:  https://doi.org/10.1016/j.celrep.2025.115494
  9. Blood Cancer Discov. 2025 Mar 31.
      Acute myeloid leukemia with complex karyotype (ckAML) is characterized by high genomic complexity, including frequent TP53 mutations and chromothripsis. Genomic rearrangements that reposition active enhancers near proto-oncogenes, leading to their aberrant expression, have not been systematically investigated in AML. To facilitate the discovery of such "enhancer hijacking" events, we developed pyjacker, a computational tool, and applied it to 39 ckAML samples. Pyjacker identified several enhancer hijacking events in AML patient samples, including aberrant expression of motor neuron and pancreas homeobox 1 (MNX1), which can result from del(7)(q22q36) and is associated with hijacking of a CDK6 enhancer. MNX1 activation occurs in 1.4% of AML patients and shows significant co-occurrence with BCOR mutations. Through a xenograft mouse model, we demonstrated that MNX1 is required for leukemia cell fitness. Pyjacker is an easy-to-use, accurate, and broadly applicable tool for identifying consequences of genomic events driving tumorigenesis, especially when germline genomic data is missing.
    DOI:  https://doi.org/10.1158/2643-3230.BCD-24-0278
  10. Nat Commun. 2025 Apr 03. 16(1): 3196
      Cancer stem cells are essential for initiation and therapy resistance of many cancers, including acute myeloid leukemias (AML). Here, we apply functional genomic profiling to diverse human leukemias, including high-risk MLL- and NUP98-rearranged specimens, using label tracing in vivo. Human leukemia propagation is mediated by a rare quiescent label-retaining cell (LRC) population undetectable by current immunophenotypic markers. AML quiescence is reversible, preserving genetic clonal competition and epigenetic inheritance. LRC quiescence is defined by distinct promoter-centered chromatin and gene expression dynamics controlled by an AP-1/ETS transcription factor network, where JUN is necessary and sufficient for LRC quiescence and associated with persistence and chemotherapy resistance in diverse patients. This enables prospective isolation and manipulation of immunophenotypically-varied leukemia stem cells, establishing the functions of epigenetic plasticity in leukemia development and therapy resistance. These findings offer insights into leukemia stem cell quiescence and the design of therapeutic strategies for their clinical identification and control.
    DOI:  https://doi.org/10.1038/s41467-025-58370-9
  11. Blood Cancer J. 2025 Mar 31. 15(1): 50
      Hypomethylating agents (HMA) are indicated in the treatment of higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). The combination of hypomethylating agents with venetoclax (Ven) has demonstrated promising results in these diseases, although randomized clinical trials are needed for validation. In this retrospective study, we compared two matched cohorts of patients with MDS or CMML: one receiving oral decitabine-cedazuridine (DEC-C, n = 73) and one receiving DEC-C and Ven (DEC-C-Ven, n = 51), in three contemporary clinical trials. The aim is to determine the impact of the addition of Ven to HMA in MDS and CMML. Individuals were matched using a propensity score approach that was based on the IPSS-M score and age. All patients had excess blasts; 84% were diagnosed with MDS and 16% with CMML. Most patients had high- or very high-risk disease, according to the revised IPSS-R. The overall response rate was superior in the DEC-C-Ven cohort (90% vs 64%, P = 0.002). The median times to best response were 1.1 and 2.7 months for the DEC-C-Ven and DEC-C cohorts, respectively (P < 0.001). More patients underwent hematopoietic stem cell transplantation in the DEC-C-Ven cohort (47%) than in the DEC-C cohort (16%, P < 0.001). The 4- and 8-week mortality did not significantly differ between the DEC-C and DEC-C-Ven cohorts. Patients in the DEC-C-Ven cohort had a more profound neutropenia at days 15 and 21 of the first cycle. The median overall survival was 24 and 19 months for the DEC-C-Ven and DEC-C cohorts, respectively (P = 0.89), and the median event-free survival durations were 18 and 10 months (P = 0.026). In conclusion, the addition of Ven resulted in improved response rates and outcomes in specific subgroups; prospective clinical trials are needed to confirm these findings.
    DOI:  https://doi.org/10.1038/s41408-025-01245-5
  12. Exp Hematol. 2025 Mar 29. pii: S0301-472X(25)00059-1. [Epub ahead of print] 104768
      Hematopoietic stem cells (HSCs) are rare cells residing at the top of the haematopoietic hierarchy capable of reconstituting all blood cell populations through their ability of self-renewal and differentiation. Their ability to maintain haematopoiesis can be majorly depleted by chemotherapeutic agents, leading to a long-term bone marrow injury. However, pre-clinical studies have focused on the acute effects of chemotherapy, leaving the lasting impact on healthy cells poorly understood. To study this, we combined rapid ex vivo models to study the long-term/late-stage effects of a cyclin-dependent kinase subunit 1 (CKS1) inhibitor. Inhibition of CKS1 has been shown to protect healthy HSCs from chemotherapy during acute myeloid leukaemia, and here we show a dose-dependent role of CKS1 inhibition on haematopoiesis, either boosting B lymphopoiesis or ablating HSC proliferation capacity, dependent on the context. Mechanistically, low doses of the CKS1 inhibitor (CKS1i) activates Foxo1 signalling potentiating B-cell differentiation, but impairing HSC proliferation. These results reveal a novel role for the SCF-CKS1 complex in boosting haematopoiesis and propose the use of rapid ex vivo models to investigate the long-term effects of chemotherapeutic treatments targeting HSCs with the potential of reducing late adverse effects.
    DOI:  https://doi.org/10.1016/j.exphem.2025.104768
  13. bioRxiv. 2025 Mar 20. pii: 2025.03.19.643917. [Epub ahead of print]
    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 co-mutations. 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.
    DOI:  https://doi.org/10.1101/2025.03.19.643917
  14. bioRxiv. 2025 Mar 14. pii: 2025.03.11.642680. [Epub ahead of print]
      MLL rearrangements ( MLL r) are the most common cause of congenital and infant leukemias. MLL r arise prior to birth and require few cooperating mutations for transformation, yet congenital leukemias are 10-fold less common than infant leukemias and >100-fold less common than childhood leukemias overall. This raises the question of whether mechanisms exist to suppress leukemic transformation during fetal life, thereby protecting the developing fetus from malignancy during a period of rapid hematopoietic progenitor expansion. Here, we use mouse models to show that fetal MLL::ENL exposure creates a heritable, leukemia-resistant state. MLL::ENL imposes a negative selective pressure on fetal hematopoietic progenitors. It leads to postnatal loss of self-renewal gene expression and enhanced myeloid differentiation that precludes transformation. These changes do not occur when MLL::ENL is induced shortly after birth, and transformation proceeds efficiently in this context. The fetal barrier to transformation is enforced by the histone methyltransferase MLL3. It can be overcome by cooperating mutations, such as Nras G12D , or through somatic or germline inactivation of MLL3. Heritable fetal protection against leukemic transformation may explain the low incidence of congenital leukemias in humans despite prenatal MLL rearrangement.
    DOI:  https://doi.org/10.1101/2025.03.11.642680
  15. J Clin Invest. 2025 Apr 03. pii: e181659. [Epub ahead of print]
      Telomere biology disorders (TBD) are genetic diseases caused by defective telomere maintenance. TBD patients often develop bone marrow failure and have an increased risk of myeloid neoplasms. To better understand the factors underlying hematopoietic outcomes in TBD, we comprehensively evaluated acquired genetic alterations in hematopoietic cells from 166 pediatric and adult TBD patients. 47.6% of patients (28.8% of children, 56.1% of adults) had clonal hematopoiesis. Recurrent somatic alterations involved telomere maintenance genes (7.6%), spliceosome genes (10.4%, mainly U2AF1 p.S34), and chromosomal alterations (20.2%), including 1q gain (5.9%). Somatic variants affecting the DNA damage response (DDR) were identified in 21.5% of patients, including 20 presumed loss-of-function variants in ATM. Using multimodal approaches, including single-cell sequencing, assays of ATM activation, telomere dysfunction-induced foci analysis, and cell growth assays, we demonstrate telomere dysfunction-induced activation of ATM-dependent DDR pathway with increased senescence and apoptosis in TBD patient cells. Pharmacologic ATM inhibition, modeling the effects of somatic ATM variants, selectively improved TBD cell fitness by allowing cells to bypass DDR-mediated senescence without detectably inducing chromosomal instability. Our results indicate that ATM-dependent DDR induced by telomere dysfunction is a key contributor to TBD pathogenesis and suggest dampening hyperactive ATM-dependent DDR as a potential therapeutic intervention.
    Keywords:  Clonal selection; Hematology; Hematopoietic stem cells; Oncology; Telomeres
    DOI:  https://doi.org/10.1172/JCI181659
  16. bioRxiv. 2025 Mar 21. pii: 2025.03.20.644389. [Epub ahead of print]
      Normal and oncogenic Ras proteins are functionally dependent on one or more lipid modifications 1,2 . Whereas K-Ras4b farnesylation is sufficient for stable association with the plasma membrane, farnesylated H-Ras, K-Ras4a, and N-Ras traffic to the Golgi where they must undergo palmitoylation before regulated translocation to cell membranes. N-Ras palmitoylation by the DHHC family of palmitoyl acyl transferases (PATs) and depalmitoylation by ABHD17 serine hydrolases is a dynamic process that is essential for the growth of acute myeloid leukemias (AMLs) harboring oncogenic NRAS mutations 3-6 . Here, we have tested whether co-targeting ABHD17 enzymes and Ras signal output would cooperatively inhibit the proliferation and survival of NRAS -mutant AMLs while sparing normal tissues that retain K-Ras4b function. We show that ABD778, a potent and selective ABHD17 inhibitor with in vivo activity, selectively reduces the growth of NRAS -mutant AML cells in vitro and is synergistic with the allosteric MEK inhibitor PD0325901 (PD901) 7,8 . Similarly, ABD778 and PD901 significantly extended the survival of recipient mice transplanted with three independent primary mouse AMLs harboring an oncogenic Nras G12D driver mutation. Resistant leukemias that emerged during continuous drug treatment acquired by-pass mutations that confer adaptive drug resistance and increase mitogen activated protein kinase (MAPK) signal output. ABD778 augmented the anti-leukemia activity of the pan-PI3 kinase inhibitor pictilisib 9 , the K/N-Ras G12C inhibitor sotorasib 10 , and the FLT3 inhibitor gilteritinib 11 . Co-treatment with ABD778 and gilteritinib restored drug sensitivity in a patient-derived xenograft model of adaptive resistance to FLT3 inhibition. These data validate the palmitoylation cycle as a promising therapeutic target in AML and support exploring it in other NRAS -mutant cancers.
    DOI:  https://doi.org/10.1101/2025.03.20.644389
  17. Cancer Cell. 2025 Mar 20. pii: S1535-6108(25)00108-4. [Epub ahead of print]
      Myeloid cancers such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) remain resistant to standard of care (SOC) and targeted therapies. In this study, we demonstrate that responsiveness to therapy is associated with activation of β-catenin-JAG1 in osteoblastic cells of patients treated with all-trans-retinoic acid (ATRA). ATRA suppresses β-catenin activity in patients and leukemic mice. Consequently, it inhibits the growth and survival of MDS/AML cells from patients with active β-catenin-JAG1 signaling and promotes their differentiation. This occurs independently of cytogenetics and mutational profile. ATRA also improves disease outcome in mice with no evidence of relapse and a superior safety profile to SOC. A human anti-JAG1 antibody improves efficacy in leukemic mice and patient-derived MDS/AML cells. β-catenin activation provides an explanation for the differential response to ATRA and a mechanistic biomarker for ATRA repurposing in myeloid malignancies, potentially evading relapse and extending across a broad range of cancers.
    Keywords:  AML; ATRA; MDS; anti-JAGGED1; b-catenin; bone marrow microenvironment; humanized antibody; myeloid malignancies; therapy
    DOI:  https://doi.org/10.1016/j.ccell.2025.03.007
  18. Blood Cancer J. 2025 Apr 03. 15(1): 56
      Venotoclax schedule in acute myeloid leukemia.
    DOI:  https://doi.org/10.1038/s41408-025-01270-4
  19. Science. 2025 Apr 04. 388(6742): 52-59
      Most phenotype-associated genetic variants map to noncoding regulatory regions of the human genome, but their mechanisms remain elusive in most cases. We developed a highly efficient strategy, Perturb-multiome, to simultaneously profile chromatin accessibility and gene expression in single cells with CRISPR-mediated perturbation of master transcription factors (TFs). We examined the connection between TFs, accessible regions, and gene expression across the genome throughout hematopoietic differentiation. We discovered that variants within TF-sensitive accessible chromatin regions in erythroid differentiation, although representing <0.3% of the genome, show a ~100-fold enrichment for blood cell phenotype heritability, which is substantially higher than that for other accessible chromatin regions. Our approach facilitates large-scale mechanistic understanding of phenotype-associated genetic variants by connecting key cis-regulatory elements and their target genes within gene regulatory networks.
    DOI:  https://doi.org/10.1126/science.ads7951
  20. Nat Cell Biol. 2025 Apr 02.
      Myocardial infarction (MI) is a major global health concern. Although myeloid cells are crucial for tissue repair in emergency haematopoiesis after MI, excessive myelopoiesis can exacerbate scarring and impair cardiac function. Bone marrow (BM) haematopoietic stem cells (HSCs) have the unique capability to replenish the haematopoietic system, but their role in emergency haematopoiesis after MI has not yet been established. Here we collected human sternal BM samples from over 150 cardiac surgery patients, selecting 49 with preserved cardiac function. We show that MI causes detrimental transcriptional and functional changes in human BM HSCs. Lineage tracing experiments suggest that HSCs are contributors of pro-inflammatory myeloid cells infiltrating cardiac tissue after MI. Therapeutically, enforcing HSC quiescence with the vitamin A metabolite 4-oxo-retinoic acid dampens inflammatory myelopoiesis, thereby modulating tissue remodelling and preserving long-term cardiac function after MI.
    DOI:  https://doi.org/10.1038/s41556-025-01639-4
  21. Expert Rev Hematol. 2025 Apr 02. 1-9
       INTRODUCTION: Older patients with acute myeloid leukemia (AML) are often unsuitable for standard treatments and traditionally have a dismal prognosis. For 20 years, hypomethylating agents (HMAs), as single agents and recently as a backbone with venetoclax, have been used in this setting. The oral combination of decitabine and cedazuridine (C-DEC), which is therapeutically and pharmacologically equivalent to the intravenous (IV) formulation (IV-DEC), has expanded the therapeutic arsenal for AML, allowing for better convenience of administration. This review provides an overview of C-DEC, current clinical applications, and ongoing clinical studies, highlighting its potential role in managing AML in older patients.
    AREAS COVERED: For the aim of this review, the authors searched the current English literature on C-DEC in AML patients through PubMed, using several pertinent keywords. To summarize the most recent developments as up-to-date as possible, we considered, with a few exceptions, only papers published over the last three years until March 2025.
    EXPERT OPINION: Compared to IV-DEC, C-DEC provides similar therapeutic exposure and efficacy with an optional safety profile, enhanced treatment adherence, reduced treatment burden, and required healthcare facilities compared to those associated with IV-DEC, providing a precious tool to manage older and unfit AML patients.
    Keywords:  AML; Acute myeloid leukemia; QOL; clinical treatment; managed care; novel agents; older patients; oral decitabine; quality of life
    DOI:  https://doi.org/10.1080/17474086.2025.2487605
  22. Ann Hematol. 2025 Apr 04.
      Venetoclax (VEN)--based induction therapy has demonstrated considerable promise in treating acute myeloid leukemia (AML); however, the optimal VEN-based combination therapy remains to be established. This study evaluated the efficacy and safety of the venetoclax-homoharringtonine-cytarabine (VHA) regimen in patients with newly diagnosed (ND) AML. A retrospective analysis was conducted on 55 patients treated with the VHA regimen. The overall response rate (ORR) was 92.7% (51/55, 95% CI 82%-98%), and the composite complete remission (CRc) rate was 87.3% (48/55, 95% CI 76%-95%). Among the 48 patients who achieved CRc, 91.7% (44/48, 95% CI 67%-90%) achieved complete remission (CR), and 85.4% (41/48, 95% CI 72%-94%) reached measurable residual disease (MRD)-negative CR. In the adverse-risk group, ORR and CRc were 95% (19/20, 95% CI 75%-100%) and 75% (15/20, 95% CI 51%-91%), respectively. The most common grade 3-4 adverse events were febrile neutropenia (32.7%), pneumonia (16.3%), and sepsis (9.1%). Median overall survival (OS) was 26 months, while event-free survival (EFS) was not reached. One-year OS was 83%, and one-year EFS was 82%. These preliminary data suggest that the VHA regimen achieves a very high rate of CR and low toxicity, particularly for adverse-risk AML patients.
    Keywords:  Acute myeloid leukemia; Cytarabine; Homoharringtonine; Venetoclax
    DOI:  https://doi.org/10.1007/s00277-025-06250-z
  23. Leukemia. 2025 Apr 02.
      Acute myeloid leukemia stem cells (LSCs) express major histocompatibility complex (MHC) class I and II and many different immune checkpoint ligands and receptors, in which respect they resemble professional antigen-presenting cells. In addition, LSCs reside in the bone marrow (BM), a primary and secondary lymphoid organ, surrounded by immune cells. The function of these immune checkpoints (ICs) in the regulation of an anti-tumor immune response is well studied and IC inhibitors (ICIs) became a standard of care in many solid tumors. However, ICIs have very limited efficacy in AML. Nevertheless, the expression especially of immune activating ligands and receptors on LSCs is somewhat unexpected, since these cells have to evade protective immunity. Many ICs have been shown to mediate direct signaling in AML blasts and LSCs and thereby regulate self-renewal, differentiation and expansion of leukemic cells. Thus, the expression of ICs on the cell surface or their soluble forms often correlate with worse survival. In this review we summarize recent data on selected ICs of the immunoglobulin superfamily (IgSF) and the tumor necrosis factor receptor superfamily (TNFRSF) that have a documented role in the regulation of LSCs, independent of their immune regulatory role, and might become novel therapeutic targets.
    DOI:  https://doi.org/10.1038/s41375-025-02566-x
  24. STAR Protoc. 2025 Mar 31. pii: S2666-1667(25)00128-5. [Epub ahead of print]6(2): 103722
      Long-range correction strategies require ex vivo activation of hematopoietic stem and progenitor cells (HSPCs) to engage the homology-directed repair (HDR) mechanism, but prolonged culture causes harmful cellular responses, reducing the long-term functionality of gene-edited (GE) HSPCs. Here, we present a protocol for optimizing culture conditions for ex vivo activation during CRISPR-Cas9 gene editing in human HSPCs. We describe steps for HSPC thawing, ex vivo treatments, gene editing, and downstream in vitro and in vivo analyses to assess the functionality of GE-HSPCs. For complete details on the use and execution of this protocol, please refer to della Volpe et al.1.
    Keywords:  CRISPR; Cell Biology; Cell culture; Stem Cells
    DOI:  https://doi.org/10.1016/j.xpro.2025.103722
  25. Cell Commun Signal. 2025 Apr 02. 23(1): 162
      Mutation in the internal tandem duplication sequence of the FLT3 gene (FLT3-ITD) is linked to a poor clinical prognosis in acute myeloid leukemia (AML) patients. FLT3 inhibitors have demonstrated efficacy in improving the prognosis of AML patients with FLT3-ITD. However, the efficacy of FLT3 inhibitors is short-lived, and is often limited by secondary drug resistance when used alone. Recent investigations have provided an innovative approach for treating FLT3-ITD AML by targeting FLT3 protein degradation. Our study revealed that decursin selectively impaired the viability of FLT3-ITD-positive AML cells. Subsequent analysis revealed that decursin preferentially induced cell cycle arrest and apoptosis in FLT3-ITD-positive AML cells through proteasome-mediated FLT3-ITD degradation. Further research revealed that decursin significantly increased the expression of UBE2L6, an e2-conjugating enzyme that degrades FLT3-ITD. Downregulation of UBE2L6 by small hairpin RNA (shRNA) reduced decursin-induced FLT3-ITD-linked apoptosis and degradation. The anti-FLT3-ITD AML effect of decursin was also validated in cell lines and patient-derived mouse models. Moreover, decursin synergistically enhanced venetoclax-induced apoptosis.
    Keywords:  AML; Decursin; Degradation; FLT3-ITD; UBE2L6
    DOI:  https://doi.org/10.1186/s12964-025-02157-4
  26. medRxiv. 2025 Mar 23. pii: 2025.03.21.25324408. [Epub ahead of print]
      Somatic mosaicism (SM), the presence of somatic mutations, is classified as clonal hematopoiesis (CH) when it occurs in hematopoietic cells at an age-related rate. CH is associated with risk for hematologic malignancies and cardiovascular disease, but most studies are predominately based on individuals of European ancestry. Using peripheral blood whole exome sequencing data from 125,748 individuals of diverse genetic ancestries, we cataloged 503,703 SM mutations based on low variant allele frequency distributions and 89,361 CH variants based on age-skewing. We examined CH prevalence across ancestry groups, including commonly recognized pathogenic variants in myeloid (M-CHIP) and lymphoid (L-CHIP) malignancies. CH and M-CHIP variants had the highest prevalence in the European non-Finnish ancestry group, and males trended toward more M-CHIP variants. Ancestry differences in CH included more mutations in NF1 in African/African American, TP53 in European, and CUX1 in Asian and Latino/Admixed American ancestry groups. Linking the identified CH variants to a cancer database, CH was detected in 14% (55,190/391,102) of patient tumors. Prevalence of CH variants in some solid tumors ranged from 25% - 40%. M-CHIP variants in solid tumors were associated with younger age (61 vs 63, p <0.001), while M-CHIP in hematologic malignancies were linked to older age (60 vs 50, p <0.001), suggesting differences in disease biology. This study provides a catalog of SM, CH, and CHIP variants across diverse ancestry groups, highlighting differences that are important to inform clinical care, drug discovery, and study design to maximize generalizability across individuals.
    KEY POINTS: Establish a large-scale catalog of somatic mosaicism and clonal hematopoiesis mutations across genetic ancestry groups.Mutation occurrences within clonal hematopoiesis genes vary across ancestry groups, with age, and across tumor types.
    DOI:  https://doi.org/10.1101/2025.03.21.25324408
  27. Haematologica. 2025 Apr 03.
      Not available.
    DOI:  https://doi.org/10.3324/haematol.2024.287135