bims-tremyl Biomed News
on Therapy resistance biology in myeloid leukemia
Issue of 2025–02–02
thirty-six papers selected by
Paolo Gallipoli, Barts Cancer Institute, Queen Mary University of London



  1. Am J Hematol. 2025 Jan 28.
      The clinical relevance of TP53 mutations (TP53MUT) in myeloproliferative neoplasms (MPN) and their prognostic interaction with MPN subtype designation has not been systematically studied. In the current study, 114 patients with MPN harboring TP53MUT (VAF ≥ 2%) were evaluated for overall survival (OS), calculated from the time of TP53MUT detection: chronic phase myelofibrosis (MF-CP; N = 61); blast-phase (MPN-BP; N = 31) or accelerated-phase (MPN-AP; N = 16) MPN, and polycythemia vera/essential thrombocythemia (PV/ET; N = 6). Sixty-five (57%) patients harbored International Consensus Classification (ICC)-defined multihit TP53MUT and 56 (49%) monosomal/complex karyotype (MK/CK). Majority of MPN-BP (90%) and MPN-AP (81%) while 39% of MF-CP and none of PV/ET patients harbored multihit TP53MUT. OS in MPN-BP and MPN-AP was equally dismal (median 6 vs. 4.5 months, respectively; p = 1.0), regardless of multihit configuration (p = 0.44), while OS in TP53MUT MPN-BP/AP (N = 47; median 4 months) was inferior to that of a separate cohort (N = 49) with TP53 wild-type MPN-BP/AP (median 11 months; p < 0.01). OS in MF-CP was significantly shorter with multihit versus non-multihit TP53MUT (median 10 vs. 35 months; HR 2.9; p < 0.01), independent of other MF-relevant genetic risk factors, including ASXL1/SRSF2/U2AF1 mutations. Multihit TP53MUT was also associated with inferior survival following allogeneic stem cell transplant (ASCT): median 9 months versus "not reached" in patients with (N = 9) versus without (N = 8) multihit TP53MUT (p < 0.01). The presence of multihit or non-multihit TP53MUT in MPN-BP/AP or multihit TP53MUT in MF-CP is associated with exceptionally poor prognosis and justifies inclusion into the ICC category of "myeloid neoplasms with mutated TP53." By contrast, detection of non-multihit TP53MUT, by itself, might not endanger short-term survival in MF-CP, PV, or ET.
    Keywords:  ICC; WHO; karyotype; prognosis; survival
    DOI:  https://doi.org/10.1002/ajh.27609
  2. Exp Hematol. 2025 Jan 22. pii: S0301-472X(25)00018-9. [Epub ahead of print] 104727
      Signaling responses to cytokines are disrupted in clonal hematopoiesis and myeloid malignancies. To better identify specific signaling response alterations in the presence or absence of TET2, we developed a 36-parameter CyTOF panel of both surface marker and phosphoprotein antigens in murine BM. We show diverse, cell-type specific inflammatory cytokine responses in healthy hematopoietic cells. We next investigated changes associated with bone marrow cells from Tet2KO mice. High dimensional surface marker phenotyping revealed expansion of HSPCs, committed cKIT+Ly6C+ myeloid progenitors, and monocytes. Loss of TET2 function increased the magnitude of response to extracellular perturbations, including IFNγ and H2O2. Response time courses revealed that IFNγ-mediated pSTAT1 remains elevated over time in Tet2KO. Further, IFNγ resulted in a more significant increase in major histocompatibility complex class II (MHCII) expression in Tet2KO immortalized progenitor cells than in Tet2WT. Inhibition of Janus kinase 1 and 2 (JAK1/2) with ruxolitinib significantly reduced STAT1 phosphorylation and MHCII expression in Tet2KO cells. Our results identify targetable disrupted signaling responses in Tet2KO cells.
    Keywords:  TET2; hematopoiesis; inflammation; interferon signaling; mass cytometry; myeloid differentiation; single-cell signaling responses
    DOI:  https://doi.org/10.1016/j.exphem.2025.104727
  3. Am J Hematol. 2025 Jan 27.
      Chromoanagenesis (CAG) encompasses a spectrum of catastrophic genomic events, including chromothripsis, chromoanasynthesis, and chromoplexy. We studied CAG in 410 patients with a diagnosis of acute myeloid leukemia (AML), 292 newly diagnosed (ND), and 118 refractory/relapsed, using optical genome mapping. CAG was identified by the presence of clusters (with 10 or more breakpoints) of structural abnormalities and/or segmental copy number alterations within one or more chromosomal regions. CAG was detected in 65 (16%) patients. Compared with patients without CAG, those with CAG showed significantly (p < 0.0001) higher frequencies of highly complex karyotype (92% vs. 11%), monosomal karyotype (88% vs. 12%), extensive clonal heterogeneity (75% vs. 7%), gene amplification (49% vs. 1%), and TP53 deletion/mutation (92% vs. 9%). Overall, CAG was detected in about two-thirds of AML patients who exhibited the abovementioned high-risk cytogenetic abnormalities/karyotype. Among the 42 patients with ND AML and CAG, 36 received treatments and follow-ups, and 28 (78%) had no or only partial response to therapy. Among patients with ND AML, those with CAG had a shorter overall survival than those without CAG (median survival: 5 vs. 14 months, p < 0.0001). However, in multivariate analysis, CAG did not appear to be an independent risk factor for survival. These results indicate that CAG is frequently associated with high-risk chromosomal alterations and genomic instability in AML and may contribute to treatment refractoriness and inferior survival in this subset of AML patients.
    Keywords:  AML; chromoanagenesis; clonal heterogeneity; complex karyotype; treatment refractory
    DOI:  https://doi.org/10.1002/ajh.27575
  4. Exp Hematol Oncol. 2025 Jan 30. 14(1): 10
      Clonal hematopoiesis of indeterminate potential (CHIP) is a condition where blood or bone marrow cells carry mutations associated with hematological malignancies. Individuals with CHIP have an increased risk of developing hematological malignancies, atherosclerotic cardiovascular disease, and all-cause mortality. Bone marrow transplantation (BMT) of cells carrying CHIP mutations into irradiated mice are useful procedures to investigate the dynamics of clonal expansion and potential therapeutic strategies, but myeloablative conditioning can induce confounding effects. We established a non-conditioned BMT model using C57BL/6J-KitW-41J/J (W41) recipient mice to overcome the unwanted effects of irradiation. Conditional Tet2 deletion using tamoxifen was used to obtain Tet2-/- cells from donor mice. Total BM Tet2-/- cells were transplanted into W41 recipients, and longitudinal and terminal analyses at 10 months post-BMT were performed. We showed that W41 mice can be used for BMT procedures without myeloablative pre-conditioning. The transplantation of Tet2-/- BM cells led to a progressive expansion of the donor cells in W41 recipients. By modulating the numbers of Tet2-/- cells transplanted, recipient mice developed features of clonal hematopoiesis or myeloid malignancies. In conclusion, our model is an alternative to conventional irradiation-based transplantation models to study mechanisms underlying malignant hematopoiesis without confounding effects derived from pre-conditioning regimen.
    DOI:  https://doi.org/10.1186/s40164-025-00598-8
  5. Biomedicines. 2025 Jan 15. pii: 202. [Epub ahead of print]13(1):
      Acute myeloid leukemias (AMLs) comprise a group of genetically heterogeneous hematological malignancies that result in the abnormal growth of leukemic cells and halt the maturation process of normal hematopoietic stem cells. Despite using molecular and cytogenetic risk classification to guide treatment decisions, most AML patients survive for less than five years. A deeper comprehension of the disease's biology and the use of new, targeted therapy approaches could potentially increase cure rates. RAS oncogene mutations are common in AML patients, being observed in about 15-20% of AML cases. Despite extensive efforts to find targeted therapy for RAS-mutated AMLs, no effective and tolerable RAS inhibitor has received approval for use against AMLs. The frequency of RAS mutations increases in the context of AMLs' chemoresistance; thus, novel anti-RAS strategies to overcome drug resistance and improve patients' therapy responses and overall survival are the need of the hour. In this article, we aim to update the current knowledge on the role of RAS mutations and anti-RAS strategies in AML treatments.
    Keywords:  AML; RAF-MEK-ERK1/2; RAS; RAS-like proteins; chemoresistance; oncogene
    DOI:  https://doi.org/10.3390/biomedicines13010202
  6. J Biol Chem. 2025 Jan 27. pii: S0021-9258(25)00087-0. [Epub ahead of print] 108240
      Forkhead box M1 (FOXM1), a Forkhead family transcription factor, is often overexpressed in a variety of human cancers, including AML and strongly associated with therapy resistance and unfavourable outcomes. In AML with NPM1 mutations NPM1/FOXM1 complex sequesters FOXM1 in the cytoplasm and confers favourable treatment outcomes for AML patients, because of FOXM1 inactivation. Inhibition of FOXM1 in AML cell lines and animal models of AML sensitizes AML cells to the Bcl2-inhibitor, venetoclax. In a recent study the upregulation of the BCL2-family protein, BCL2A1 conferred resistance to venetoclax and multiple venetoclax combinations.In this study, we investigated FOXM1/BCL2A1 axis and determined that FOXM1 specifically inhibits venetoclax-induced apoptosis in AML via upregulation of BCL2A1.The knockdown of BCL2A1 in AML in the presence of high levels of FOXM1 led to sensitization of AML cells to venetoclax, suggesting that BCL2A1 is a major target of FOXM1 responsible for resistance to venetoclax. Venetoclax in combination with FOXM1 inhibitor STL001 inhibited BCL2A1 and circumvented venetoclax resistance. Pharmacological inhibition of FOXM1/BCL2A1 axis represents a therapeutic strategy to sensitize AML cells to venetoclax-induced apoptosis.
    DOI:  https://doi.org/10.1016/j.jbc.2025.108240
  7. J Clin Invest. 2025 Jan 30. pii: e184743. [Epub ahead of print]
      Although nucleoporin 98 (NUP98) fusion oncogenes often drive aggressive pediatric leukemia by altering chromatin structure and expression of HOX genes, underlying mechanisms remain elusive. Here, we report that a Hoxb-associated lncRNA HoxBlinc was aberrantly activated in NUP98-PHF23 fusion-driven leukemias. HoxBlinc chromatin occupancies led to elevated MLL1 recruitment and aberrant homeotic topologically associated domains (TADs) that enhanced chromatin accessibilities and activated homeotic/hematopoietic oncogenes. HoxBlinc-depletion in NUP98 fusion-driven leukemia impaired HoxBlinc binding, TAD integrity, MLL1 recruitment, and MLL1-driven chromatin signature within HoxBlinc-defined TADs in a CTCF-independent manner, leading to inhibited homeotic/leukemic oncogenes that mitigated NUP98 fusion-driven leukemogenesis in xenografted mouse models. Mechanistically, HoxBlinc overexpression in mouse hematopoietic compartment induced leukemias resembling those in NUP98-PHF23 knock-in mice via enhancing HoxBlinc chromatin binding, TAD formation, and Hox gene aberration leading to expansion of hematopoietic stem and progenitor cell (HSPC) and myeloid/lymphoid subpopulations. Thus, our studies reveal a CTCF-independent role of HoxBlinc in leukemic TAD organization and oncogene regulatory networks.
    Keywords:  Epigenetics; Genetics; Hematology; Hematopoietic stem cells; Oncogenes
    DOI:  https://doi.org/10.1172/JCI184743
  8. Leukemia. 2025 Jan 27.
      Refractory disease and relapse are major challenges in acute myeloid leukemia (AML) therapy attributed to survival of leukemic stem cells (LSC). To target LSCs, antibody-drug conjugates (ADCs) provide an elegant solution, combining the specificity of antibodies with highly potent payloads. We aimed to investigate if FLT3-20D9h3-ADCs delivering either the DNA-alkylator duocarmycin (DUBA) or the microtubule-toxin monomethyl auristatin F (MMAF) can eradicate quiescent LSCs. We show here that DUBA more potently kills cell-cycle arrested AML cells compared to microtubule-targeting auristatins. Due to limited stability of 20D9h3-DUBA ADC in vivo, we analyzed both ADCs in advanced in vitro stem cell assays. 20D9h3-DUBA successfully eliminated leukemic progenitors in vitro in colony-forming unit and long-term culture initiating cell assays, both in patient cells and in patient-derived xenograft (PDX) cells. Further, it completely prevented engraftment of AML PDX leukemia-initiating cells in NSG mice. 20D9h3-MMAF had a similar effect in engraftment assays, but a less prominent effect in colony assays. Both ADCs did not affect healthy stem and progenitor cells at comparable doses providing the rationale for FLT3 as therapeutic LSC target. Collectively, we show that FLT3-directed ADCs with DUBA or MMAF have potent activity against AML LSCs and represent promising candidates for further clinical development.
    DOI:  https://doi.org/10.1038/s41375-024-02510-5
  9. medRxiv. 2025 Jan 13. pii: 2025.01.12.25320422. [Epub ahead of print]
      Clonal hematopoiesis of indeterminate potential (CHIP) is associated with increased mortality and malignancy risk, yet the determinants of clonal expansion remain poorly understood. We performed sequencing at >4,000x depth of coverage for CHIP mutations in 6,986 postmenopausal women from the Women's Health Initiative at two timepoints approximately 15 years apart. Among 3,685 mutations detected at baseline (VAF ≥ 0.5%), 50% progressed to CHIP (VAF ≥ 2%) at follow-up. We confirmed that clonal expansion is highly dependent on initial clone size and CHIP driver gene, with SF3B1 and JAK2 mutations exhibiting the fastest growth rate. We identified germline variants in TERT , IL6R , TCL1A , and MSI2 that modulate clonal expansion rate. Measured baseline leukocyte telomere length showed differential effects on incident CHIP risk, with shorter baseline leukocyte telomere length predisposing to incident PPM1D mutations and longer baseline leukocyte telomere length favoring incident DNMT3A mutations. We discovered that the IL6R missense variant p.Asp358Ala specifically impairs TET2 clonal expansion, supported by direct measurements of soluble interleukin-6 receptor and interleukin-6. Faster clonal growth rate was associated with increased risk of cytopenia, leukemia, and all-cause mortality. Notably, CHIP clonal expansion rate mediated 34.4% and 43.7% of the Clonal Hematopoiesis Risk Score's predictive value for leukemia and all-cause mortality, respectively. These findings reveal key biological determinants of CHIP progression and suggest that incorporating growth rate measurements could enhance risk stratification.
    DOI:  https://doi.org/10.1101/2025.01.12.25320422
  10. Nat Commun. 2025 Jan 13. 16(1): 617
      Acute myeloid leukemia (AML) featuring retinoic acid receptor-gamma (RARG) rearrangements exhibits morphological features resembling those of acute promyelocytic leukemia but is associated with drug resistance and poor clinical outcomes. However, the mechanisms underlying the role of RARG fusions in leukemogenesis remain elusive. Here, we show that RARG fusions disrupt myeloid differentiation and promote proliferation and self-renewal of hematopoietic stem and progenitor cells (HSPCs) by upregulating BCL2 and ATF3. RARG fusions overexpression leads to preleukemic phenotypes but fails to induce oncogenic transformation. However, the co-occurrence of RARG fusions and heterozygous Wt1 loss induce fully penetrant AML by activating MYC and HOXA9/MEIS1 targets. Leveraging Connectivity Map resources and high-throughput screening, we identify venetoclax, homoharringtonine, and daunorubicin as potential therapeutic options for RARG-AML. Overall, our findings provide pivotal insights into the molecular mechanisms governed by RARG fusions and enhanced by WT1 loss in AML development and propose a rational therapeutic strategy for RARG-AML.
    DOI:  https://doi.org/10.1038/s41467-024-55047-7
  11. JAMA. 2025 Jan 27.
       Importance: Essential thrombocythemia, a clonal myeloproliferative neoplasm with excessive platelet production, is associated with an increased risk of thrombosis and bleeding. The annual incidence rate of essential thrombocythemia in the US is 1.5/100 000 persons.
    Observations: Patients with essential thrombocythemia have a persistent platelet count of 450 × 109/L or greater. The differential diagnosis includes myeloproliferative neoplasms (polycythemia vera, primary myelofibrosis, chronic myeloid leukemia); inflammatory conditions such as rheumatoid arthritis and systemic lupus erythematosus; infections; splenectomy; iron deficiency anemia; and solid tumors such as lung cancer. Approximately 90% of individuals with essential thrombocythemia have genetic variants that upregulate the JAK-STAT (signal transducer and activator of transcription 5) signaling pathway, including Janus kinase 2 (JAK2, 64%), calreticulin (CALR, 23%), and myeloproliferative leukemia virus oncogene (MPL, 4%). The median age at diagnosis of essential thrombocythemia is 59 years. The median overall survival exceeds 35 years in those diagnosed at 40 years or younger. Patients with essential thrombocythemia are at increased risk of arterial thrombosis (11%), venous thrombosis (7%), and hemorrhagic complications (8%). Thrombosis risk is increased among those with a history of thrombosis, age older than 60 years, a JAK2 gene variant, and cardiovascular risk factors (eg, hypertension, diabetes mellitus, hyperlipidemias, tobacco use). Use of aspirin (81-100 mg/d) is suggested for most patients with essential thrombocythemia to lower thrombosis risk. In a retrospective study of 300 affected patients with a low thrombosis risk (younger than 60 years with no prior thrombosis), those not taking aspirin (100 mg/d) had a risk of arterial thrombosis of 9.4/1000 patient-years and a venous thrombosis risk of 8.2/1000 patient years; cardiovascular risk factors were associated with a higher risk of arterial thrombi (incidence rate ratio, 2.5 [95% CI, 1.02-6.1]), and a JAK2 gene variant was associated with increased risk of venous thrombosis (incidence rate ratio, 4.0 [95% CI, 1.2-12.9]). In a randomized trial of 114 patients at higher risk for thrombosis (age older than 60 years or a prior thrombotic event), cytoreduction with hydroxyurea significantly lowered the risk of arterial or venous thrombotic events compared with no cytoreductive therapy (3.6% vs 24%; P < .01). At a median of 8.5 years from diagnosis, approximately 10% of patients with essential thrombocythemia develop myelofibrosis and about 3% develop acute myeloid leukemia.
    Conclusions: Essential thrombocythemia is a rare clonal myeloproliferative neoplasm associated with an increased risk of venous and arterial thrombosis, hemorrhage, myelofibrosis, and acute myeloid leukemia. Based on individual risk factors for thrombosis, persons with essential thrombocythemia may be treated with low-dose aspirin, either alone or in combination with a cytoreductive drug such as hydroxyurea.
    DOI:  https://doi.org/10.1001/jama.2024.25349
  12. Trends Cancer. 2025 Jan 24. pii: S2405-8033(25)00007-X. [Epub ahead of print]
      The AUGMENT-101 clinical trial reported that the use of revumenib led to improved overall survival (OS) and complete remission (CR)/CR with partial hematologic recovery (CRh) in patients with acute leukemias and has released updated data with longer follow-up of Phase 2 results. Additionally, revumenib has received FDA approval for its use in relapsed or refractory (r/r) lysine methyltransferase 2A rearranged (KMT2A-r) acute leukemias.
    Keywords:  AUGMENT-101 trial; acute leukemias; menin inhibitors; revumenib
    DOI:  https://doi.org/10.1016/j.trecan.2025.01.006
  13. Nat Aging. 2025 Jan 29.
      Somatic stem cell pools comprise diverse, highly specialized subsets whose individual contribution is critical for the overall regenerative function. In the bone marrow, myeloid-biased hematopoietic stem cells (myHSCs) are indispensable for replenishment of myeloid cells and platelets during inflammatory response but, at the same time, become irreversibly damaged during inflammation and aging. Here we identify an extrinsic factor, semaphorin 4A (Sema4A), which non-cell-autonomously confers myHSC resilience to inflammatory stress. We show that, in the absence of Sema4A, myHSC inflammatory hyper-responsiveness in young mice drives excessive myHSC expansion, myeloid bias and profound loss of regenerative function with age. Mechanistically, Sema4A is mainly produced by neutrophils, signals via a cell surface receptor, plexin D1, and safeguards the myHSC epigenetic state. Our study shows that, by selectively protecting a distinct stem cell subset, an extrinsic factor preserves functional diversity of somatic stem cell pool throughout organismal lifespan.
    DOI:  https://doi.org/10.1038/s43587-024-00798-7
  14. Haematologica. 2025 01 30.
    MINHEMON, FRENVEX
      Not available.
    DOI:  https://doi.org/10.3324/haematol.2024.286641
  15. Cell Commun Signal. 2025 Jan 25. 23(1): 47
      One hallmark of cancer is the upregulation and dependency on glucose metabolism to fuel macromolecule biosynthesis and rapid proliferation. Despite significant pre-clinical effort to exploit this pathway, additional mechanistic insights are necessary to prioritize the diversity of metabolic adaptations upon acute loss of glucose metabolism. Here, we investigated a potent small molecule inhibitor to Class I glucose transporters, KL-11743, using glycolytic leukemia cell lines and patient-based model systems. Our results reveal that while several metabolic adaptations occur in response to acute glucose uptake inhibition, the most critical is increased mitochondrial oxidative phosphorylation. KL-11743 treatment efficiently blocks the majority of glucose uptake and glycolysis, yet markedly increases mitochondrial respiration via enhanced Complex I function. Compared to partial glucose uptake inhibition, dependency on mitochondrial respiration is less apparent suggesting robust blockage of glucose uptake is essential to create a metabolic vulnerability. When wild-type and oncogenic RAS patient-derived induced pluripotent stem cell acute myeloid leukemia (AML) models were examined, KL-11743 mediated induction of mitochondrial respiration and dependency for survival associated with oncogenic RAS. Furthermore, we examined the therapeutic potential of these observations by treating a cohort of primary AML patient samples with KL-11743 and witnessed similar dependency on mitochondrial respiration for sustained cellular survival. Together, these data highlight conserved adaptations to acute glucose uptake inhibition in diverse leukemic models and AML patient samples, and position mitochondrial respiration as a key determinant of treatment success.
    Keywords:  Adaptations; Bioenergetics; Cancer; Chemotherapy; Glucose; Leukemia; Metabolism; Mitochondria; Oncogenes; Stem cells
    DOI:  https://doi.org/10.1186/s12964-025-02044-y
  16. Int J Cancer. 2025 Jan 30.
      Chromosomal rearrangements involving the Mixed Lineage Leukemia gene (MLL1, KMT2A) are defining a genetically distinct subset in about 10% of human acute leukemias. Translocations involving the KMT2A-locus at chromosome 11q23 are resulting in the formation of a chimeric oncogene, where the N-terminal part of KMT2A is fused to a variety of translocation partners. The most frequently found fusion partners of KMT2A in acute leukemia are the C-terminal parts of AFF1, MLLT3, MLLT1 and MLLT10. Unfortunately, the presence of an KMT2A-rearrangements is associated with adverse outcomes in leukemia patients. Moreover, non-rearranged KMT2A-complexes have been demonstrated to be crucial for disease development and maintenance in NPM1-mutated and NUP98-rearranged leukemia, expanding the spectrum of genetic disease subtypes that are dependent on KMT2A. Recent advances in the development of targeted therapy strategies to disrupt the function of KMT2A-complexes in leukemia have led to the establishment of Menin-KMT2A interaction inhibitors that effectively eradicate leukemia in preclinical model systems and show favorable tolerability and significant efficacy in early-phase clinical trials. Indeed, one Menin inhibitor, Revumenib, was recently approved for the treatment of patients with relapsed or refractory KMT2A-rearranged acute leukemia. However, single agent therapy can lead to resistance. In this Review article we summarize our current understanding about the biology of pathogenic KMT2A-complex function in cancer, specifically leukemia, and give a systematic overview of lessons learned from recent clinical and preclinical studies using Menin inhibitors.
    Keywords:  ALL; AML; Bleximenib; KMT2A; MLL1; Menin; NPM1; NUP98; Revumenib; Ziftomenib; leukemia; resistance
    DOI:  https://doi.org/10.1002/ijc.35332
  17. Eur J Haematol. 2025 Jan 28.
       INTRODUCTION: Iron overload (IOL) accumulates in myelodysplastic syndromes (MDS) from expanded erythropoiesis and transfusions. Somatic mutations (SM) are frequent in MDS and stratify patient risk. MDS treatments reversing or limiting transfusion dependence are limited.
    METHODS: The literature was reviewed on how IOL and oxidative stress interact with specific SM in MDS to influence cellular physiology. PubMed searches included keywords of each specific mutation combined with iron, oxidative stress, and reactive oxygens species (ROS). Papers relevant to hematopoietic stem/progenitor cells, the bone marrow microenvironment, MDS, AML or other myeloid disorders were preferred. Included were the most frequent SM in MDS, SM of the International Prognostic Scoring System-Molecular (IPSS-M), of familial predisposing conditions and the CMML PSS-molecular.
    RESULTS: About 31 SM plus four familial conditions were searched. Discussed are the frequency of each SM, whether function is gained or lost, early or late SM status, a function of the unmutated gene, and function considering iron and oxidative stress.
    DISCUSSION: Given limited effective MDS therapies, considering how IOL and ROS interact with SM to influence cellular physiology in the hematopoietic system, increasing bone marrow failure progression or malignant transformation may be of benefit and support optimization of measures to reduce IOL or neutralize ROS.
    Keywords:  IOL; MDS; ROS; iron overload; oxidative stress; reactive oxygen species; somatic mutations
    DOI:  https://doi.org/10.1111/ejh.14385
  18. Blood Cancer J. 2025 Jan 30. 15(1): 7
      GATA2 germline mutations lead to a syndrome characterized by immunodeficiency, vascular disorders and myeloid malignancies. To elucidate how these mutations affect hematopoietic homeostasis, we created a knock-in mouse model expressing the recurrent Gata2 R396Q missense mutation. Employing molecular and functional approaches, we investigated the mutation's impact on hematopoiesis, revealing significant alterations in the hematopoietic stem and progenitor (HSPC) compartment in young age. These include increased LT-HSC numbers, reduced self-renewal potential, and impaired response to acute inflammatory stimuli. The mature HSPC compartment was primarily affected at the CMP sub-population level. In the mutant LT-HSC population, we identified an aberrant subpopulation strongly expressing CD150, resembling aging, but occurring prematurely. This population showed hyporesponsiveness, accumulated over time, and exhibited allele-specific expression (ASE) favoring the mutated Gata2 allele, also observed in GATA2 mutated patients. Our findings reveal the detrimental impact of a Gata2 recurrent missense mutation on the HSC compartment contributing to its functional decline. Defects in the CMP mature compartment, along with the inflammatory molecular signature, explain the loss of heterogeneity in HPC compartment observed in patients. Finally, our study provides a valuable model that recapitulates the ASE-related pathology observed in GATA2 deficiency, shedding light on the mechanisms contributing to the disease's natural progression.
    DOI:  https://doi.org/10.1038/s41408-025-01213-z
  19. Bone Marrow Transplant. 2025 Jan 29.
      Methodological advancements now allow older adults with AML to receive allografts although conflicting data exist regarding relative outcomes across age groups and benefits of different conditioning intensities. We retrospectively analyzed 495 adults aged 60-64 (n = 184), 65-69 (n = 189), or ≥70 (n = 122) allografted for AML in remission at our institution from 2006 to 2023. There were no significant differences in relapse or relapse-free survival (RFS) among the 3 age cohorts after multivariable adjustment. Patients aged ≥70 years had higher non-relapse mortality (NRM) than those aged ≥60-64 (P = 0.022) but their overall survival (OS) was only statistically non-significantly shorter (P = 0.11). There was an important interplay between age, conditioning intensity, and outcomes. Relative to age 60-64, age ≥70 years was associated with a higher risk of relapse (hazard ratio [HR] = 3.47; P = 0.012) and NRM (HR = 3.88; P = 0.001) with reduced intensity conditioning (RIC), leading to shorter RFS (HR = 3.79; P < 0.001) and OS (HR = 3.46; P < 0.001), while no such associations were found with nonmyeloablative (NMA) conditioning. Underlying, patients aged 60-64 and 65-69, but not those aged ≥70, had a significantly lower relapse risk with RIC relative to NMA conditioning, whereas NRM risks increased across all age cohorts. Our findings support allografting for adults ≥70 with AML in remission, especially with NMA conditioning.
    DOI:  https://doi.org/10.1038/s41409-025-02516-2
  20. medRxiv. 2025 Jan 15. pii: 2025.01.14.25320566. [Epub ahead of print]
       Background: Clonal hematopoiesis of indeterminate potential (CHIP) is the age-related presence of expanded somatic clones secondary to leukemogenic driver mutations and is associated with cardiovascular (CV) disease and mortality. We sought to evaluate relationships between CHIP with cardiometabolic diseases and incident outcomes in high-risk individuals.
    Methods: CHIP genotyping was performed in 8469 individuals referred for cardiac catheterization at Duke University (CATHGEN study) to identify variants present at a variant allele fraction (VAF) ≥2%. Associations were tested among any CHIP variant, large CHIP clones (VAF ≥10%) and individual CHIP genes with prevalent cardiometabolic traits. Cox proportional hazard models tested CHIP associations with time-to-overall mortality and Fine-Gray analyses tested CHIP associations with incident cardiovascular outcomes.
    Results: We identified 463 CHIP variants in 427 individuals (5.0%) of which 268 (3.2%) harbored large CHIP clones. CHIP and large CHIP were associated with lower odds of obesity (OR 0.79 [95% CI 0.65-0.98], p=0.03; OR 0.76 [95% CI 0.57-0.99], p=0.04, respectively). CHIP was associated with prevalent HF (OR 1.25 [95% CI 1.01 - 1.55], p=0.04; especially for non- DNMT3A CHIP (OR 1.38 [95% CI 1.04-1.82], p=0.02). CHIP was also associated with incident events: Non- DNMT3A CHIP was associated with increased risk of time-to-HF hospitalization (HR 1.29 [95% CI 1.02-1.63], p=0.03).
    Conclusions: In high-risk individuals referred for cardiac catheterization, large CHIP and non- DNTM3A CHIP were associated with obesity, prevalent HF, incident CV events. These findings strengthen the importance of CHIP as a biomarker for CV disease and highlight the contributing risk of large CHIP clones and non- DNMT3A CHIP variants.
    Condensed Abstract: CHIP, the presence of somatic expanded mutations in myeloid driver genes in hematopoietic cells, is an emerging CVD biomarker. Using whole exome sequencing of peripheral blood derived DNA from participants in the CATHGEN cohort, we identified significant associations with obesity, prevalent HF, incident mortality, HF hospitalization and AF after adjusting for established clinical risk factors. These findings add strength to the growing literature of CHIP as a CVD biomarker, emphasizing large CHIP and non- DNMT3A CHIP variants for driving risk. Future studies should aim to further elucidate gene-specific risk and the inflammatory and metabolic mechanisms possibly mediating these relationships.
    Clinical Perspective: What Is New?: In a cohort with high prevalence of CAD, CHIP is inversely associated with obesity and associated with higher odds of prevalent HF and subsequent mortality, even after adjustment for relevant clinical comorbidities. Risk of incident events of mortality, HF hospitalization and AF were driven by large CHIP variants (VAF≥10%) and CHIP variants in genes other than DNMT3A . What are the Clinical Implications?: Though more research is needed, as the evidence around the risk associated with specific CHIP variants continues to grow, clinicians should be prepared to provide gene- specific counseling for cardiometabolic disease risk.
    DOI:  https://doi.org/10.1101/2025.01.14.25320566
  21. Am J Hematol. 2025 Jan 29.
      We retrospectively analyzed a large international cohort of 1113 patients with aplastic anemia to evaluate treatment choice and outcome in elderly patients as compared with a younger population. Overall, 319 (29%) patients were > 60 years old at diagnosis (60-64 years (n = 85), 106 65-69 years (n = 106), and 128 > 70 years (n = 128)). Elderly patients showed a more severe thrombocytopenia at onset and a significantly lower overall response (complete plus partial) to first-line therapy at 6 months as compared to younger patients (47% vs. 65%, p < 0.0001), irrespective of treatment modality (ATG or CyA combinations, eltrombopag, or androgens); 27 (8%) received transplant as second line therapy and 11 (41%) died, mainly due to transplant complications. The rate of evolution to MDS was greater in elderly patients (12% vs. 7% in younger AA, p = 0.002), whilst the rate of evolution to AML was similar (1.8 vs. 1.3%). By multivariable analysis, older age remained the main factor associated with mortality [HR 1.64 (95% CI 1.5-1.7), p < 0.001], followed by disease severity by Camitta classification [HR 2.24 (95% CI 1.6-3.1) for severe AA; HR 3.8 (95% CI 2.4-6) for very severe AA], and male gender [1.45 (95% CI 1.1-1.8), p < 0.001]. In this large study, elderly AA was associated with inferior outcome even in the TPO-RA era, highlighting the need for further optimization of clinical management.
    Keywords:  anti‐thymocyte globulin; aplastic anemia; cyclosporine; elderly; eltrombopag
    DOI:  https://doi.org/10.1002/ajh.27611
  22. Cell Commun Signal. 2025 Jan 28. 23(1): 53
      FLT3 mutations occur in approximately 25% of all acute myeloid leukemia (AML) patients. While several FLT3 inhibitors have received FDA approval, their use is currently limited to combination therapies with chemotherapy, as resistance occurs, and efficacy decreases when the inhibitors are used alone. Given the highly heterogeneous nature of AML, there is an urgent need for novel targeted therapies that address the disease from multiple angles. Recent research has identified the NLRP3 inflammasome as a potential new driver in AML. Here, we investigated the efficacy of different NLRP3 inhibitors in targeting AML cells in vitro. Our findings reveal that NLRP3 inhibition induces cell cycle arrest as well as signs of senescence in multiple AML cell lines. In contrast, NLRP3 inhibition selectively induced apoptosis in FLT3 mutant AML cell lines, but not in FLT3 wild-type AML cells. Moreover, we show that NLRP3 inhibition impairs FLT3 signaling by reducing both FLT3 expression as well as downstream signaling in FLT3 mutant cells. A database analysis revealed a strong positive correlation between FLT3 and NLRP3 in cancer, which was particularly evident in AML patients. Strikingly, the simultaneous inhibition of NLRP3 and FLT3 markedly enhanced apoptosis in FLT3-ITD mutant AML cells, but not in FLT3 wild-type cells. In summary, this study reveals a promising combined therapeutic strategy specifically targeting NLRP3/FLT3-ITD positive AML blasts in vitro, highlighting a potential new avenue for AML treatment.
    DOI:  https://doi.org/10.1186/s12964-025-02046-w
  23. J Exp Med. 2025 Mar 03. pii: e20241248. [Epub ahead of print]222(3):
      Hematopoietic stem cells (HSCs) are susceptible to replication stress, which is a major contributor to HSC defects in Fanconi anemia (FA). Here, we report that HSCs relax the global chromatin by downregulating the expression of a chromatin architectural protein, DEK, in response to replication stress. DEK is abnormally accumulated in bone marrow (BM) CD34+ cells from patients with FA and in Fancd2-deficient HSCs. DEK haploinsufficiency promotes chromatin relaxation, replication stress relief, and function recovery of Fancd2-deficient HSCs. Furthermore, inhibition of DEK restores the proliferation of FA CD34+ cells in vitro and enhances their engraftment in vivo. Mechanistically, the activating transcription factor 2 (ATF2), specifically phosphorylated ATF2 at Thr69/71, was identified as a promoter of DEK transcription. Fancd2 deficiency results in p38 hyperphosphorylation, which in turn phosphorylates ATF2 at Thr69/71, leading to DEK accumulation in HSCs. In conclusion, our findings establish a functional link between chromatin relaxation and replication stress tolerance in HSCs and highlight DEK as a target for FA.
    DOI:  https://doi.org/10.1084/jem.20241248
  24. Am J Hematol. 2025 Jan 28.
      The influence of t(v;22) sole, major route ACAs all (+8, n = 14; +Ph, n = 10; +19, n = 1), and -Y sole on progression-free survival. Survival curves are compared with those of patients with the standard t(9;22) translocation. Other ACAs or complex karyotypes did not influence survival.
    Keywords:  additional chromosomal abnormalities; chronic myeloid leukemia; cytogenetics; prognosis
    DOI:  https://doi.org/10.1002/ajh.27608
  25. Ann Hematol. 2025 Jan 31.
      The prefibrotic phase of primary myelofibrosis (pre-PMF) represents a distinct subentity within the spectrum of myeloproliferative neoplasms (MPNs), recognized by the World Health Organization (WHO) and the International Consensus Classification (ICC). Pre-PMF is characterized by unique morphological, clinical, and molecular features, distinguishing it from essential thrombocythemia (ET) and overt myelofibrosis (overt-PMF). The diagnostic process for pre-PMF relies on bone marrow histology, identification of molecular mutations and exclusion of other myeloid neoplasms. Misclassification remains a significant challenge due to overlapping phenotypes and the heterogeneity of clinical presentations, which range from asymptomatic cases to severe cytopenias and a high thrombotic risk. Management strategies for pre-PMF focus on mitigating symptom burden, reducing thromboembolic events, and preventing disease progression. Low-risk patients often benefit from observational approaches or low-dose aspirin, while cytoreductive therapies, such as hydroxyurea or interferon-alpha, are utilized in symptomatic or high-risk cases. JAK inhibitors like ruxolitinib have shown promise in addressing splenomegaly and systemic symptoms, although their role in pre-PMF requires further investigation. Advances in artificial intelligence are enhancing diagnostic precision by refining bone marrow histopathological analysis, paving the way for more accurate disease classification and tailored therapeutic strategies. This position paper integrates insights from a German expert panel discussion, underscoring the need for interdisciplinary collaboration, adherence to updated WHO/ICC diagnostic criteria, and personalized treatment approaches. By addressing diagnostic challenges and therapeutic nuances, it seeks to improve outcomes and quality of life for patients navigating the complexities of pre-PMF.
    Keywords:  MPN; Myeloproliferative neoplasm; Pre-PMF; Prefibrotic myelofibrosis; Targeted therapy
    DOI:  https://doi.org/10.1007/s00277-025-06191-7
  26. Blood Adv. 2025 Jan 28. pii: bloodadvances.2024012600. [Epub ahead of print]
      Interferon alpha (IFNa) is approved for the therapy of patients (pts) with polycythemia vera (PV), a subtype of myeloproliferative neoplasms (MPN). Some pts achieve molecular responses (MR), but clonal factors sensitizing for MR remain elusive. We integrated colony formation and differentiation assays with single-cell RNA seq and genotyping in PV-derived cells vs. healthy controls (HC) to dissect how IFNa targets diseased clones during erythroid differentiation. IFNa significantly decreased colony growth in MPN and HC, with variable transcriptional responses observed in individual colonies. scRNAseq of colonies demonstrated more mature erythroid PV-derived colonies compared with HC. JAK2V617F-mutant cells exhibited upregulated STAT5A, heme, and G2M checkpoint pathways compared to JAK2WT cells from the same pts. Subgroup analysis revealed that IFNa significantly decreased immature erythrocytic cells in PV (basophilic erythroblasts p<0.05; polychromatic erythroblasts p<0.05) but not in HC. CD71-/CD235a+ cells from HC (p<0.05) but not from PV pts were inhibited by IFNa, and number of reticulocytes was less affected in PV. Robust IFNa responses persisted throughout differentiation, leading to significant apoptosis in PV. Apoptotic cells displayed downregulation of ribosomal genes. This link between apoptosis and ribosomal genes was corroborated through analysis of mitochondrial variants demonstrating IFNa-induced eradication of specific clones, characterized by elevated expression of ribosomal genes. Our findings indicate that PV-derived clones either undergo apoptosis or pass through differentiation, overall reducing cycling mutant cells over long-term treatment. Further, the significance of ribosomal genes and clonal prerequisites in IFNa's therapeutic mechanism is underscored, shedding light on the intricate dynamics of IFNa treatment in PV.
    DOI:  https://doi.org/10.1182/bloodadvances.2024012600
  27. Blood Adv. 2025 Feb 11. 9(3): 533-544
       ABSTRACT: Little is known about the impact of recent advances in acute myeloid leukemia (AML) treatment on racial/ethnic disparities in survival outcomes. We performed a retrospective cohort study of patients with newly diagnosed AML using data from a nationwide electronic health record-derived deidentified database. Patients were categorized based on their diagnosis date relative to venetoclax approval, as pre-novel therapy era (Pre era; 2014-2018; n = 2998) or post-novel therapy era (Post era; 2019-2022; n = 2098). Patients in the Post era were older and had more comorbidities than Pre era. Non-Hispanic Black (NHB) and Hispanic patients were younger and more likely to have lower socioeconomic status than non-Hispanic White (NHW) patients, with no differences in the distributions of key disease features. After accounting for age and comorbidity, overall survival (OS) was higher in patients in Post era than Pre era (adjusted hazard ratio [aHR], 0.90; 95% confidence interval [CI], 0.83-0.96). In Pre era, NHB had a 22% higher hazard of death than NHW (aHR, 1.22; 95% CI, 1.04-1.43), whereas worse OS was not observed for NHB in Post era (aHR, 0.86; 95% CI, 0.69-1.08; predicted 2-year survival, 45.3% vs 39.9%). Utilization of novel therapeutics in frontline therapy did not differ by race/ethnicity. Among patients receiving venetoclax-based induction, particularly those without TP53, RAS, or FLT3-ITD mutations, results suggested higher OS for NHB than NHW patients (aHR, 0.67; 95% CI, 0.45-1.01). Additional studies are needed to elucidate factors contributing to these observed survival differences and to inform strategies to optimize outcomes for all patients with AML.
    DOI:  https://doi.org/10.1182/bloodadvances.2024014127
  28. EJHaem. 2025 Feb;6(1): e1052
       Background: Acute myeloid leukemia (AML) with RAM immunophenotype is a newly recognized high-risk AML immunophenotypic subcategory characterized by blasts with bright expression of CD56 and weak to absent expression of CD45, HLA-DR, and CD38, as first described by the Children's Oncology Group (COG). The relationship between AML-RAM and other CD56-positive acute leukemias is unclear. The goal of this study is to characterize the clinicopathological characteristics of AML with RAM phenotype and compare them with other CD56 co-expressing acute leukemias.
    Methods: From a multi-institutional search, we identified a total of 160 CD56+ acute leukemia cases, including AML-RAM (n = 28), CD56+ acute undifferentiated leukemia (AUL) (n = 11), CD56+ T-lymphoblastic leukemia (n = 39), and CD56+ AML (n = 81). We compared the clinical and pathologic findings of these groups.
    Results: AML-RAM patients were significantly younger and presented with significantly higher platelet and white blood cell counts and bone marrow (BM) blast percentages when compared to AUL (p > 0.05) and had higher median BM blast percentages than T-ALL and CD56+ AML groups (both p < 0.05). Flow cytometry showed significantly brighter expression of CD56 on blasts as compared to other CD56+ AML cases, partial CD34 expression compared to AUL, and AML, weak-to-absent CD38 expression compared to all groups, and absent HLA-DR and terminal deoxynucleotidyl transferase as compared to AUL and T-ALL (all p < 0.05). The frequency of abnormal karyotypes was significantly higher among RAM when compared to all groups (p < 0.05). Next-generation sequencing profiles differed among the leukemia groups, with significant enrichment of CBFA2T3::GLIS2 fusions (p < 0.05) and TP53 mutations (p < 0.05) in RAM cases compared to other AML control groups, and U2AF1 (p < 0.05), serine and arginine-rich splicing factor 2 (p < 0.05), and BCL6 co-repressor (p < 0.05) mutations compared to AUL. Clinical outcome analysis demonstrated significantly lower 3-year overall survival of the RAM subgroup (36 months) compared to control groups (p = 0.002).
    Conclusion: We find that AML with RAM phenotype occurs primarily in younger ages, with distinct clinicopathological, immunophenotypic, and mutational presentations, and worse prognosis. This diagnosis should be considered in the clinical differential diagnosis of CD56-positive acute leukemias.
    Keywords:  AML; flow cytometry; genomic
    DOI:  https://doi.org/10.1002/jha2.1052
  29. Cell Mol Biol Lett. 2025 Jan 29. 30(1): 14
       BACKGROUND: The protein cereblon (CRBN) mediates the antileukemia effect of lenalidomide (Len). Len binds to CRBN, recruits IKZF1/IKZF3, and promotes their ubiquitination and degradation, through which Len exhibits its antileukemia and antimyeloma activity. Therefore, the protein level of CRBN might affect the antiproliferative effect of Len. In this study, we explored the interactome for CRBN using proximity labeling technique TurboID and quantitative proteomics, and then investigated the antileukemia effect of Len.
    METHODS: The primary acute myeloid leukemia (AML) cells and AML cell lines were used to explore the functions of histone demethylase KDM5C on the antileukemia effect of Len. The cell viability and CRBN protein levels were evaluated in these cell lines. In addition, the KDM5C inhibitors were used to determine the effects of KDM5C enzymatic activity on the viability of AML cell lines.
    RESULTS: We identified that histone demethylase KDM5C was a CRBN-interacting protein. Biochemical experiments found that the CRBN-interacting protein KDM5C could stabilize CRBN and enhance the antileukemia effect of Len in an enzyme activity-independent manner. Furthermore, our studies revealed that the small-molecule compound MLN4924 could increase CRBN by elevating KDM5C.The combination of MLN4924 and Len can further increase the sensitivity of primary AML cells and AML cell lines to Len.
    CONCLUSIONS: This study provides a possible strategy for a combination treatment with MLN4924 and Len for leukemia.
    Keywords:  Cell viability; Cereblon (CRBN); KDM5C; Lenalidomide; Leukemia
    DOI:  https://doi.org/10.1186/s11658-025-00697-8
  30. Nutr Cancer. 2025 Jan 31. 1-7
      Hematopoietic stem cells (HSCs) maintain production of all functional blood cells and are located within the bone marrow. In pathological conditions, such as obesity or leukemia, changes in these cells contribute to disease pathophysiology. In this study, we examined the impact of metabolic modulation of stem and progenitor cells within the bone marrow during diet-induced obesity (DIO) and leukemia relapse. Avocatin B (Avo), an inhibitor of fatty acid oxidation (FAO), was provided in the diet and its impact on stem cells using two disease models was tested. In DIO, high fat diet(HFD)-induced alterations in HSC number and function were attenuated with Avo (HFD: 46.9% decrease compared to control; p < 0.001; whereas DIO + Avo: 58.8% recovery; p < 0.05). In leukemia relapse, dietary Avo delayed disease reoccurrence. Taken together, addition of Avo into the diet imparts protection in the bone marrow.
    DOI:  https://doi.org/10.1080/01635581.2025.2459445
  31. Nature. 2025 Jan 29.
      During normal cellular homeostasis, unfolded and mislocalized proteins are recognized and removed, preventing the build-up of toxic byproducts1. When protein homeostasis is perturbed during ageing, neurodegeneration or cellular stress, proteins can accumulate several forms of chemical damage through reactive metabolites2,3. Such modifications have been proposed to trigger the selective removal of chemically marked proteins3-6; however, identifying modifications that are sufficient to induce protein degradation has remained challenging. Here, using a semi-synthetic chemical biology approach coupled to cellular assays, we found that C-terminal amide-bearing proteins (CTAPs) are rapidly cleared from human cells. A CRISPR screen identified FBXO31 as a reader of C-terminal amides. FBXO31 is a substrate receptor for the SKP1-CUL1-F-box protein (SCF) ubiquitin ligase SCF-FBXO31, which ubiquitylates CTAPs for subsequent proteasomal degradation. A conserved binding pocket enables FBXO31 to bind to almost any C-terminal peptide bearing an amide while retaining exquisite selectivity over non-modified clients. This mechanism facilitates binding and turnover of endogenous CTAPs that are formed after oxidative stress. A dominant human mutation found in neurodevelopmental disorders reverses CTAP recognition, such that non-amidated neosubstrates are now degraded and FBXO31 becomes markedly toxic. We propose that CTAPs may represent the vanguard of a largely unexplored class of modified amino acid degrons that could provide a general strategy for selective yet broad surveillance of chemically damaged proteins.
    DOI:  https://doi.org/10.1038/s41586-024-08475-w