bims-tremyl Biomed News
on Therapy resistance biology in myeloid leukemia
Issue of 2025–02–16
thirty papers selected by
Paolo Gallipoli, Barts Cancer Institute, Queen Mary University of London



  1. Am J Hematol. 2025 Feb 08.
      Ontogeny of acute myeloid leukemia (AML) provides prognostic information, however closer interrogation with respect to AML characteristics, genomics, and various treatments are warranted. We defined untreated clinical secondary (CS) AML as AML with a diagnosis of antecedent myelodysplastic syndrome (MDS) or MDS/myeloproliferative neoplasm (MDS-MPN) without exposure to hypomethylating agents or chemotherapy; genomic secondary (GS) AML included patients with myelodysplasia related cytogenetics (MRC) or myelodysplasia related mutations (MRM) without a known antecedent myeloid neoplasm or prior chemo-radiotherapy for non-myeloid neoplasms. Among newly diagnosed AML patients classified as untreated CS-AML (n = 133) or GS-AML (n = 389), median relapse-free survival (RFS) (11.9 vs. 12.4 months, p = 0.36) and overall survival (OS) (11.6 vs. 14.4 months, p = 0.75) were similar. No difference in RFS and OS between these groups treated with low-intensity therapy (LIT) and venetoclax regimens was seen, but both were inferior to de novo (DN) AML without secondary-type genomics (pure DN-AML). GS-AML defined by the presence of only MRM had superior OS compared with MRM ± MRC with LIT+ venetoclax therapy (RFS 19.5 vs. 6.8 months [p < 0.01] and OS 29.6 vs. 8.4 [p < 0.01]) and had similar RFS (29.8 months, p = 0.48) and OS (32.0 months, p = 0.48) to pure DN-AML treated with LIT+ venetoclax. On multivariate analysis in patients treated with LIT+ venetoclax, untreated CS-AML (vs. GS-AML), adverse cytogenetics and ELN 2024 adverse-risk disease (mutated TP53) were associated with higher hazard of death. Adverse cytogenetics was the strongest prognostic variable predicting survival. Mutation-driven genomic ontogeny of newly diagnosed AML with MRM appears less prognostic than cytogenetic-driven ontogeny with venetoclax-based therapy.
    Keywords:  Venetoclax; cytogenetics; myelodysplasia related genomics; secondary AML; stem cell transplantation
    DOI:  https://doi.org/10.1002/ajh.27628
  2. Bone Marrow Transplant. 2025 Feb 12.
      We retrospectively analyzed the impact of conditioning intensity on transplant outcomes according to their cytogenetic/molecular risk in a cohort of 1823 patients with acute myeloid leukemia (AML) and intermediate- or adverse-risk cytogenetics in first complete remission (CR1). These patients received their first hematopoietic stem cell transplantation (HSCT) using post-transplant cyclophosphamide (PTCy). The intermediate-risk cytogenetic group included 1386 (76%) patients, and 608 (34%) had mutated FLT3-ITD. Myeloablative conditioning was used in 930 patients (51%), while 1130 (62%) received an intensified conditioning (score ≥2.5) based on the transplant conditioning intensity (TCI) score. Conditioning intensity using the myeloablative/reduced intensity stratification did not impact transplant outcomes across the entire cohort. However, a higher TCI score was associated with a lower risk of relapse, with no effect on survival. In specific cytogenetic risk groups, a higher TCI score did not influence outcomes in the adverse-risk group. In the intermediate-risk group, the impact varied with FLT3-ITD status. Patients with FLT3-ITD mutation who received a higher TCI showed a beneficial effect on relapse, leukemia-free survival (LFS), and overall survival. Conversely, in FLT3-ITD wild-type patients, more intense conditioning had a detrimental effect on graft-versus-host disease-free, and relapse-free survival with no effect on other outcomes. In conclusion, for AML patients in CR1 undergoing HSCT with PTCy, it is crucial to consider cytogenetic risk and molecular status when selecting the conditioning regimen. Intensive conditioning should be considered for patients with intermediate-risk cytogenetics and mutated FLT3-ITD but should probably be avoided for those with wild-type FLT3-ITD.
    DOI:  https://doi.org/10.1038/s41409-025-02527-z
  3. Blood Adv. 2025 Feb 12. pii: bloodadvances.2024015149. [Epub ahead of print]
      Mutations found in AML such as DNMT3A, TET2 and ASXL1 can be found in the peripheral blood of healthy adults - a phenomenon termed clonal hematopoiesis (CH). These mutations are thought to represent the earliest genetic events in the evolution of AML. Genomic studies on samples acquired at diagnosis, remission, and at relapse have demonstrated significant stability of CH mutations following induction chemotherapy. Meanwhile, later mutations in genes such as NPM1 and FLT3, have been shown to contract at remission and in the case of FLT3 often are absent at relapse. We sought to understand how early CH mutations influence subsequent evolutionary trajectories throughout remission and relapse in response to induction chemotherapy. We assembled a retrospective cohort of patients diagnosed with de novo AML at our institution that underwent genomic sequencing at diagnosis, remission and/or relapse (total n=182 patients). FLT3 and NPM1 mutations were generally eliminated at complete remission but subsequently reemerged upon relapse, whereas DNMT3A, TET2 and ASXL1 mutations often persisted through remission. CH-related mutations exhibited distinct constellations of co-occurring genetic alterations, with NPM1 and FLT3 mutations enriched in DNMT3Amut AML, while CBL and SRSF2 mutations were enriched in TET2mut and ASXL1mut AML, respectively. In the case of NPM1 and FLT3 mutations, these differences vanished at the time of complete remission yet readily reemerged upon relapse, indicating the reproducible nature of these genetic interactions. Thus, CH-associated mutations that likely precede malignant transformation subsequently shape the evolutionary trajectories of AML through diagnosis, therapy, and relapse.
    DOI:  https://doi.org/10.1182/bloodadvances.2024015149
  4. Blood Cancer Discov. 2025 Feb 10.
      Despite the curative potential of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia (AML), its efficacy is limited by intrinsic resistance of cancer cells to donor-derived T-cell cytotoxicity. Using a genome-wide CRISPR screen, we identified the SOCS1-JAK1-STAT1 pathway as a mediator of AML susceptibility to T cells. SOCS1 knockdown in AML cells sensitized them to killing by allogeneic T cells, whereas SOCS1 overexpression in AML cells induced resistance to T-cell anti-leukemic activity. Mechanistically, SOCS1 protected AML cells from T-cell killing by antagonizing IFNγ-JAK1-induced ICAM-1 expression. Furthermore, primary AML cells with lower SOCS1 expression correlated with better overall survival in patients, especially those with a lower exhausted CD8+ T-cell score. Thus, this study reveals SOCS1 and its downstream mediators as a potential targetable pathway to enhance T cell-based immunotherapy for AML.
    DOI:  https://doi.org/10.1158/2643-3230.BCD-24-0140
  5. Leuk Res. 2025 Feb 06. pii: S0145-2126(25)00021-9. [Epub ahead of print]150 107661
      Myelodysplastic syndrome (MDS) with isolated deletion 11q is a rare favorable cytogenetic abnormality with a low risk of progression to acute myeloid leukemia (AML). The aim of this study is to describe the clinical characteristics and long-term outcomes of patients with isolated del(11q) MDS. Between August 1997 and January 2024, 52 patients with MDS and isolated del(11q) were diagnosed, representing 0.4 % of the cohort. The median age was 69 years, with a mild male predominance (62 %). By the World Health Organization (WHO) 2022, 42 % of patients had MDS with low blasts. With a median follow-up of 96 months, the median survival was 71 months with a 5-year survival rate of 53 %. The 5-year survival rates were 45 % and 68 % in the hypomethylating agents and best supportive care group, respectively (P = 0.63). Multivariate Cox regression analyses identified age, absolute neutrophil count, hemoglobin, and blast percentages as significant prognostic factors. Despite isolated del(11q) MDS being classified as a very-good-risk cytogenetic abnormality, long-term survival is poor with the risk of progression to AML and complications from cytopenias. The poor long-term survival indicates the need for the investigation of effective supportive care and early intervention to benefit patients with lower-risk MDS and high-risk features.
    Keywords:  Del(11q); HMA; MDS; Overall survival; Progression-free survival
    DOI:  https://doi.org/10.1016/j.leukres.2025.107661
  6. Blood Adv. 2025 Feb 10. pii: bloodadvances.2024015176. [Epub ahead of print]
      We conducted a multi-center, open-label, randomized phase II study to assess the efficacy of Nivolumab as maintenance therapy for patients with AML in first complete remission (CR) or CR with incomplete hematologic recovery (CRi) who were not candidates for SCT. Patients were stratified and randomized to Observation (Obs) or Nivolumab (Nivo, 3mg/kg IV every 2 weeks for 46 doses). The primary endpoint was progression-free survival (PFS) defined as time to disease relapse or death due to any reason. Secondary endpoints included overall survival (OS), and evaluation of adverse events following Nivolumab administration. Eighty patients were enrolled with median duration of follow-up of 24 months (33 months among survivors). PFS was 13.2 months in the Nivolumab arm (95% CI: 8.5-21.8) and 10.9 months in the Observation arm (5.4-14.9 months). Overall PFS curves were not statistically significantly different ((Nivo/Obs)= 0.92; 95% CI: 0.54, 1.56; one-sided p = 0.38). The median OS was 53.9 months in the Nivolumab arm and 30.9 months in the Observation arm. Cox regression model HR (Nivo/Obs)= 0.78; 95% CI: 0.40, 1.51; p=0.23 (one-sided). There were more adverse events (AEs) of any type (regardless of attribution) on the Nivolumab arm; 27 (71%) patients on the Nivolumab arm had a grade 3 or higher AE compared to 5 patients (12%) on the Observation arm (p<0.001). Nivolumab maintenance after AML chemotherapy failed to improve the PFS and OS in this randomized Phase II study. There were increased AEs and SAEs with nivolumab, but these AEs and SAEs were expected and manageable. ClinicalTrials.gov ID NCT02275533.
    DOI:  https://doi.org/10.1182/bloodadvances.2024015176
  7. Cell Rep. 2025 Jan 28. pii: S2211-1247(24)01502-X. [Epub ahead of print]44(1): 115151
      In acute myeloid leukemia (AML), malignant cells surviving chemotherapy rely on high mRNA translation and their microenvironmental metabolic support to drive relapse. However, the role of translational reprogramming in the niche is unclear. Here, we found that relapsing AML cells increase translation in their bone marrow (BM) niches, where BM mesenchymal stromal cells (BMSCs) become a source of eIF4A-cap-dependent translation machinery that is transferred to AML cells via extracellular vesicles (EVs) to meet their translational demands. In two independent models of highly chemo-resistant AML driven by MLL-AF9 or FLT3-ITD (internal tandem duplication) and nucleophosmin (NPMc) mutations, protein synthesis levels increase in refractory AML dependent on nestin+ BMSCs. Inhibiting cap-dependent translation in BMSCs abolishes their chemoprotective ability, while EVs from BMSCs carrying eIF4A boost AML cell translation and survival. Consequently, eIF4A inhibition synergizes with conventional chemotherapy. Together, these results suggest that AML cells rely on BMSCs to maintain an oncogenic translational program required for relapse.
    Keywords:  CP: Cancer; acute myeloid leukemia; bone marrow mesenchymal stromal cells; chemotherapy; extracellular vesicles; microenvironment; niche; protein synthesis; refractory; relapse; translation
    DOI:  https://doi.org/10.1016/j.celrep.2024.115151
  8. Am J Hematol. 2025 Feb 11.
      
    Keywords:  BMP; SMAD; TGF‐beta; myelodysplastic; myelofibrosis
    DOI:  https://doi.org/10.1002/ajh.27638
  9. Signal Transduct Target Ther. 2025 Feb 10. 10(1): 50
      The importance of MCL-1 in leukemogenesis has prompted development of MCL-1 antagonists e.g., S63845, MIK665. However, their effectiveness in acute myeloid leukemia (AML) is limited by compensatory MCL-1 accumulation via the ubiquitin proteasome system. Here, we investigated mechanisms by which kinase inhibitors with Src inhibitory activity e.g., bosutinib (SKI-606) might circumvent this phenomenon. MCL-1 antagonist/SKI-606 co-administration synergistically induced apoptosis in diverse AML cell lines. Consistently, Src or MCL-1 knockdown with shRNA markedly sensitized cells to MCL-1 inhibitors or SKI-606 respectively, while ectopic MCL-1 expression significantly diminished apoptosis. Mechanistically, MCL-1 antagonist exposure induced MCL-1 up-regulation, an event blocked by Src inhibitors or Src shRNA knock-down. MCL-1 down-regulation was associated with diminished transcription and increased K48-linked degradative ubiquitination. Enhanced cell death depended functionally upon down-regulation of phosphorylated STAT3 (Tyr705/Ser727) and cytoprotective downstream targets c-Myc and BCL-xL, as well as BAX/BAK activation, and NOXA induction. Importantly, the Src/MCL-1 inhibitor regimen robustly killed primary AML cells, including primitive progenitors, but spared normal hematopoietic CD34+ cells and human cardiomyocytes. Notably, the regimen significantly improved survival in an MV4-11 cell xenograft model, while reducing tumor burden in two patient-derived xenograft (PDX) AML models and increased survival in a third. These findings argue that Src inhibitors such as SKI-606 potentiate MCL-1 antagonist anti-leukemic activity in vitro and in vivo by blocking MCL-1 antagonist-mediated cytoprotective MCL-1 accumulation by promoting degradative ubiquitination, disrupting STAT-3-mediated transcription, and inducing NOXA-mediated MCL-1 degradation. They also suggest that this strategy may improve MCL-1 antagonist efficacy in AML and potentially other malignancies.
    DOI:  https://doi.org/10.1038/s41392-025-02125-x
  10. Blood Adv. 2025 Feb 10. pii: bloodadvances.2024015173. [Epub ahead of print]
      Post-transplant high-dose cyclophosphamide (PTCy) is effective in overcoming the negative impact of HLA disparity in the haploidentical setting. In the light of these results, we investigated the efficacy and safety of PTCy, with a calcineurine inhibitor and mycophenolate mofetil, in improving clinical outcomes of haematopoietic cell transplantation (HCT) from mismatched unrelated donor (MMUD) in patients with acute myeloid malignancies by reducing aGvHD incidence and severity. A prospective single arm, phase II study (PHYLOS - NCT03270748) was conducted by the Italian GITMO. The primary objective was the cumulative incidence (CI) of grade II-IV aGvHD. Conditioning regimen for all patients was busulfan (total dose 12.8mg/kg) and fludarabine (total dose 160mg/m2). The ethical committees of the participating centers approved the study (EURODRACT 2017-003530-85). Seventy-seven consecutive patients (AML: 64; MDS: 13) were enrolled at 26 Italian transplant centers (January 2020-November 2022). Median age was 53 years (range 19-65). The 100-day cumulative incidence (CI) of grade II-IV aGvHD was 18.2% (95%CI: 10.6-27.6) and 6.5% (95%CI: 3.1-15.1) for grade III-IV. Seventy-one patients (92%) had full-donor chimerism with complete neutrophil engraftment by day +30.One-year CI of chronic GvHD (cGvHD) was 13.4% (95%CI: 6.9-22.1). One-year CI of non-relapse mortality was 9.1% (95%CI: 4.0-16.9), and the relapse rate was 23.8% (95%CI: 14.9-33.9). One-year overall survival and graft-relapse-free survival were 78.6.% (95%CI: 67.4-86.3) and 55.3% (95%CI: 43.4-65.7), respectively. Our study in a homogeneous patient cohort suggests that PTCy leads to a low rate of aGvHD and improves clinical outcomes of MMUD transplantation.
    DOI:  https://doi.org/10.1182/bloodadvances.2024015173
  11. Genomics Proteomics Bioinformatics. 2025 Feb 13. pii: qzaf005. [Epub ahead of print]
      Precise mapping of leukemic cells onto the known hematopoietic hierarchy is important for understanding the cell-of-origin and mechanisms underlying disease initiation and development. However, this task remains challenging because of the high interpatient and intrapatient heterogeneity of leukemia cell clones as well as the differences existed between leukemic and normal hematopoietic cells. Using single-cell RNA sequencing (scRNA-seq) data with a curated clustering approach, we constructed a comprehensive reference hierarchy of normal hematopoiesis. This reference hierarchy was accomplished through multistep clustering and annotating over 100,000 bone marrow mononuclear cells derived from 25 healthy donors. We further employed the cosine distance algorithm to develop a likelihood score, determining the similarities of leukemic cells to their putative normal counterparts. Using our scoring strategies, we mapped the cells of acute myeloid leukemia (AML) and B cell precursor acute lymphoblastic leukemia (BCP-ALL) samples to their corresponding counterparts. The reference hierarchy also facilitated bulk RNA sequencing (RNA-seq) analysis, enabling the development of a least absolute shrinkage and selection operator (LASSO) score model to reveal subtle differences in lineage aberrancy within AML or BCP-ALL patients. To facilitate interpretation and application, we have established an R-based package (HematoMap) that offers a fast, convenient, and user-friendly tool for identifying and visualizing lineage aberrations in leukemia from scRNA-seq and bulk RNA-seq data. Our tool provides curated resources and data analytics for understanding leukemogenesis, with the potential to enhance leukemia risk stratification and personalized treatments. The HematoMap is available at https://github.com/NRCTM-bioinfo/HematoMap.
    Keywords:  Acute leukemia; Bioinformatics; Hematopoietic hierarchy; Lineage aberration; Single-cell RNA sequencing
    DOI:  https://doi.org/10.1093/gpbjnl/qzaf005
  12. Blood Adv. 2025 Feb 14. pii: bloodadvances.2024015365. [Epub ahead of print]
      Dysregulated JAK/STAT signaling underlies the pathogenesis of myelofibrosis, a myeloproliferative neoplasm characterized by cytopenias, splenomegaly and constitutional symptoms. JAK inhibitors, such as fedratinib, are the primary therapeutic option for patients with high-risk or symptomatic myelofibrosis. Fedratinib has characteristics that distinguish it from the other commercially available JAK inhibitors, such as its preferential inhibition of JAK2 and its inhibitory effects on kinases such as FLT3 and BRD4. Fedratinib is most often used in the second-line setting after intolerance or resistance to other JAK inhibitors, but there is substantial evidence that it is an effective first-line option in the appropriate patient population. Prevention and early treatment of fedratinib-related gastrointestinal toxicity is key to maintaining adequate drug exposure, and clinicians must remain vigilant for Wernicke encephalopathy during treatment. Fedratinib's JAK2 selectivity and kinome profile make it an appealing agent for alternative indications, such as myelodysplastic/myeloproliferative neoplasms and maintenance after bone marrow transplantation, which are under active investigation.
    DOI:  https://doi.org/10.1182/bloodadvances.2024015365
  13. Br J Haematol. 2025 Feb 10.
      Relapse after allogeneic stem cell transplantation (alloSCT) represents a frequent complication in patients with myelofibrosis (MF). This retrospective study analysed 38 patients with JAK2-mutated MF who underwent alloSCT. Serial sensitive molecular assessments at regular intervals of JAK2 V617F revealed that a ≥2-fold increase in JAK2 variant allele frequency (VAF) at days +100 and +180 post-alloSCT was highly predictive of eventual morphological relapse. The integration of JAK2 VAF monitoring with chimerism analysis into post-alloSCT care represents a novel strategy for early detection of MF relapse, which may enable timely interventions and improve outcomes in such high-risk patients.
    Keywords:  molecular pathology; myelofibrosis; relapse; stem cell transplantation
    DOI:  https://doi.org/10.1111/bjh.20003
  14. Haematologica. 2025 Feb 13.
      Germline loss of function (LoF) DDX41 variants predispose to late-onset hematopoietic malignancies (HMs), predominantly of myeloid lineage. Among 43 families with germline DDX41 LoF variants, bone marrow (BM) biopsies in those without (n=8) or with malignancies (n=21) revealed mild dysplasia in peripheral blood (57%) and BM (88%), long before the average age of DDX41-related HM onset. Therefore, we recommend baseline bone marrow biopsies in people with germline DDX41LoF alleles to avoid over-diagnosis of myelodysplastic syndromes. A variety of solid tumors were also observed in our cohort, with 24% penetrance by age 75. Although acquired DDX41 mutations are common in HMs, we failed to identify such alleles in solid tumors arising in those with germline DDX41LoF variants (n=15), suggesting an alternative mechanism driving solid tumor development. Furthermore, 33% of pedigrees in which >15% of first-degree relatives including the proband were diagnosed with a solid tumor had second germline deleterious variants in other cancerpredisposition genes, likely serving as primary cancer drivers. Finally, both lymphoblastoid cell lines and primary peripheral blood from individuals with germline DDX41LoF variants exhibited differential levels of inflammation-associated proteins. These data provide evidence of inflammatory dysfunction mediated by germline DDX41LoF alleles that may contribute to solid tumor growth in the context of additional germline cancer-associated variants. For those with HMs and personal/family histories of solid tumors, we recommend broad germline testing. DDX41 may be an indirect modifier of solid tumor pathogenesis compared to its tumor suppressor function within hematopoietic tissues, a hypothesis that can be addressed in future work.
    DOI:  https://doi.org/10.3324/haematol.2024.286887
  15. Hemasphere. 2025 Feb;9(2): e70086
      We explored the impact of luspatercept therapy on overall survival (OS) and possible predictors of response in low-risk (LR) myelodysplastic syndrome (MDS) patients. We evaluated 331 anemic patients treated with luspatercept. Hematological response (HI) was defined as (i) hemoglobin (Hb) increase of ≥1.5 g/dL in nontransfusion-dependent (NTD) patients, and (ii) red blood cell (RBC) transfusion independence (TI) with a concomitant Hb increase of ≥1.5 g/dL, or RBC-TI without an Hb increase of 1.5 g/dL, or >50% reduction in RBC transfusion burden (TB) for TD patients. Response was observed in 166 patients (50.2%), with significantly higher response in NTD and low TB versus high TB patients (p < 0.001). A significant correlation between lower Molecular International Prognostic Scoring System (IPSS-M) risk scores and response was observed. No statistically significant difference in HI was found in SF3B1-mutated versus wild-type MDS patients (53.8% vs. 40.1%, respectively). SF3B1mut hotspots (K700E vs. others) and variant allele frequencies (VAFs; <38% VAF vs. ≥38% VAF) did not impact on HI. SF3B1-mutated MDS with del5q showed inferior HI compared to other LR-MDS (p = 0.046). The median treatment duration overall was 35 weeks (20.86-90.29), the median time to response was 11 weeks (8.71-21.86), and the median duration of response was 65 weeks (26.5-114). After a median follow-up of 13 months, median OS was not reached (NR) for responders and 24 months for nonresponders (hazard ratio [HR] 0.25, 95% confidence interval 0.14-0.44, p < 0.001). This analysis of 331 luspatercept real-life-treated LR-MDS patients demonstrated a significant OS benefit upon luspatercept response. Low baseline RBC-TB and lower risk IPSS-M scores correlated with higher HI and could constitute predictive markers of response.
    DOI:  https://doi.org/10.1002/hem3.70086
  16. Nat Genet. 2025 Feb 13.
      The impact of genetic ancestry on the development of clonal hematopoiesis (CH) remains largely unexplored. Here, we compared CH in 136,401 participants from the Mexico City Prospective Study (MCPS) to 416,118 individuals from the UK Biobank (UKB) and observed CH to be significantly less common in MCPS compared to UKB (adjusted odds ratio = 0.59, 95% confidence interval (CI) = [0.57, 0.61], P = 7.31 × 10-185). Among MCPS participants, CH frequency was positively correlated with the percentage of European ancestry (adjusted beta = 0.84, 95% CI = [0.66, 1.03], P = 7.35 × 10-19). Genome-wide and exome-wide association analyses in MCPS identified ancestry-specific variants in the TCL1B locus with opposing effects on DNMT3A-CH versus non-DNMT3A-CH. Meta-analysis of MCPS and UKB identified five novel loci associated with CH, including polymorphisms at PARP11/CCND2, MEIS1 and MYCN. Our CH study, the largest in a non-European population to date, demonstrates the power of cross-ancestry comparisons to derive novel insights into CH pathogenesis.
    DOI:  https://doi.org/10.1038/s41588-025-02085-6
  17. Lancet Haematol. 2025 Feb 07. pii: S2352-3026(24)00354-5. [Epub ahead of print]
       BACKGROUND: Resistance or intolerance to the available tyrosine kinase inhibitors (TKIs) remains a treatment challenge for patients with chronic myeloid leukaemia. We aimed to report the safety, antileukaemic activity, and pharmacokinetics of oral vodobatinib, a novel selective BCR::ABL1 TKI, in patients with Philadelphia chromosome-positive (Ph-positive) chronic myeloid leukaemia who previously received at least three TKIs, including ponatinib and asciminib.
    METHODS: This open-label, multicentre, phase 1/2 trial was conducted at 28 clinical sites across ten countries (Belgium, France, Hungary, India, Italy, Romania, South Korea, Spain, UK, and the USA). Patients aged 18 years or older with Ph-positive chronic myeloid leukaemia or acute lymphoblastic leukaemia (eligible only for the phase 1 study), and an Eastern Cooperative Oncology Group performance status of 2 or lower were eligible. Phase 1 included patients who previously received at least three TKIs or had no other available treatment options. Phase 2 required patients to have treatment resistance or intolerance (or both) with loss of response to at least three TKIs and previous ponatinib use. A key exclusion criterion for both phases was presence of the Thr315Ile mutation. Patients self-administered oral vodobatinib (12-240 mg) once per day for each 28-day treatment cycle and for up to 60 months (ie, 65 cycles) unless patient discontinuation due to adverse events, progressive disease, lost to follow-up, or death. The primary endpoints were to determine the maximum tolerated dose (based on dose-limiting toxicities in phase 1) and antileukaemic activity of vodobatinib (ie, major cytogenetic response for chronic-phase and major haematological response for accelerated-phase or blast-phase in phase 2). Assessment of vodobatinib safety, activity, and pharmacokinetics were determined based on the pooled analysis of data from the phase 1 and 2 studies. This trial is registered with ClinicalTrials.gov, NCT02629692 (active). At data cutoff (July 15, 2023), phase 2 enrolment was closed early on June 22, 2023, due to recruitment-related challenges.
    FINDINGS: 78 patients were enrolled and received at least one vodobatinib dose (safety and efficacy analysis set). Between April 6, 2017, and June 20, 2023, phase 1 enrolled 58 patients and phase 2 enrolled 20 patients between March 3, 2020, and March 29, 2023. We included 66 (85%) patients with chronic-phase, eight (10%) with accelerated-phase, and four (5%) with blast-phase chronic myeloid leukaemia. 43 (55%) of 78 patients were male and 35 (45%) were female. The median age was 59·0 years (IQR 47·0-66·0). The median follow-up was 22·3 months (IQR 11·1-43·9). Two patients receiving vodobatinib 240 mg had dose-limiting toxicities (one had grade 3 dyspnoea and the other had grade 2 fluid overload), thus the 204 mg dose was considered to be the maximum tolerated dose. 73 (94%) patients had one or more treatment-emergent adverse events, with most events being haematological or gastrointestinal that were grade 2 or lower in severity. Grade 3 or higher treatment-emergent adverse events occurred in 47 (60%) patients and included thrombocytopenia (14 [18%]), neutropenia (10 [13%]), anaemia (nine [12%]), and increased lipase (eight [10%]). Seven (9%) patients died during the study; one death was considered related to treatment by the clinical investigator. At data cutoff, major cytogenetic response was observed in 44 (70%) of 63 patients with chronic-phase chronic myeloid leukaemia, of which 12 (75%) of 16 patients in the phase 2 study had major cytogenetic response. For patients with accelerated-phase chronic myeloid leukaemia, six (86%) of seven patients had a major haematological response (median duration 17·8 [IQR 10·2-24·3]) at data cutoff; major haematological response was observed in three (100%) evaluable patients in the phase 2 study. Major haematological response was reached by two (50%) of four patients with blast-phase chronic myeloid leukaemia and the median duration of response was 6·2 months (IQR 3·2-9·3); no blast-phase patients were enrolled in the phase 2 study.
    INTERPRETATION: Pooled analysis of the phase 1 and 2 studies showed clinically meaningful antileukaemic activity of vodobatinib and a tolerable safety profile in patients with advanced chronic myeloid leukaemia who previously received multiple TKIs, including ponatinib and asciminib, addressing an otherwise unmet clinical need. The phase 2 study was statistically underpowered and warrants further investigation in a phase 3, randomised controlled trial and in an earlier treatment setting of the disease.
    FUNDING: Sun Pharma Advanced Research Company.
    DOI:  https://doi.org/10.1016/S2352-3026(24)00354-5
  18. Mol Oncol. 2025 Feb 10.
      Acute myeloid leukemia (AML) is a heterogeneous cancer, making outcomes prediction challenging. Several predictive and prognostic models are used but have considerable inaccuracy at individual level. We tried to increase prediction accuracy using a multi-omics strategy. We interrogated data from 1391 consecutive, newly diagnosed subjects with AML, integrating information on mutation topography, DNA methylation, and transcriptomics. We developed an unsupervised multi-omics classification system (UAMOCS) with these data. UAMOCS provides a multidimensional understanding of AML heterogeneity and stratifies subjects into three cohorts: (a) UAMOCS1 [high lymphocyte activating 3 (LAG3) expression, chromosome instability, myelodysplasia-related mutations]; (b) UAMOCS2 (monocytic-like profile, immune suppression and activated angiogenesis and hypoxia pathways); and (c) UAMOCS3 [CCAAT enhancer binding protein alpha (CEBPA) mutations and MYC pathway activation]. UAMOCS distinguishes overall survival rates across the cohorts (TCGA P = 0.042; GSE71014 P = 0.043; ihCAMs-AML, GSE102691 and GSE37642 all P < 0.001). The model's C-statistic is comparable to the 2022 ELN risk classification (0.87 vs 0.82; P = 0.162), but offers a more nuanced distinction between intermediate- and high-risk groups. When combined with high-throughput drug sensitivity testing, UAMOCS can accurately predict sensitivity to azacitidine (AZA) and venetoclax. The UAMOCS system is available as an R package. The UAMOCS system has the potential to redefine AML subtypes, enhance prognostic predictions, and guide treatment strategies based on patients' immune status and expected responses to therapies.
    Keywords:  acute myeloid leukemia; classification; multi‐omics; prognosis
    DOI:  https://doi.org/10.1002/1878-0261.70000
  19. Blood Adv. 2025 Feb 10. pii: bloodadvances.2024015016. [Epub ahead of print]
      The treatment of monogenetic disorders, such as hemoglobinopathies and lysosomal storage diseases, has markedly improved with the advent of cell and gene therapies, particularly allogeneic or gene-modified autologous stem cell transplantations. However, therapeutic efficacy is reliant on maintaining engraftment above a critical threshold. To maintain such engraftment levels, we and others have pursued approaches to shield edited cells from antibody or CAR T-cell mediated selection. Here we focused on CD33, which is expressed early on hematopoietic stem and progenitor cells (HSPC) as well as on myeloid progenitors. Rhesus macaques were engrafted with HSPCs edited to ablate CD33 utilizing either CRISPR/Cas9 or adenine base editor. Both editing strategies showed similar post-transplant recovery kinetics and yielded equivalent levels of engraftment. We then created a V-set domain specific chimeric antigen receptor construct (CAR33), validated its functionality in vitro, and treated both animals with autologous CAR33 T cells. CAR33 T cells expanded after infusion and caused specific depletion of CD33WT but not CD33null progeny - leading to a transient enrichment for gene-edited cells in the blood. No depletion was seen in the bone marrow stem cell compartment with CD34+CD90+ HSCs expressing lower levels of CD33 in comparison to monocytes. Thus, we show proof of concept and safety of an epitope editing based enrichment/protection strategy in macaques.
    DOI:  https://doi.org/10.1182/bloodadvances.2024015016
  20. Cancer Cell. 2025 Feb 10. pii: S1535-6108(25)00022-4. [Epub ahead of print]43(2): 269-291.e19
      Immune-related adverse events (irAEs) in cancer patients receiving immune checkpoint inhibitors (ICIs) cause morbidity and necessitate cessation of treatment. Comparing irAE treatments, we find that anti-tumor immunity is preserved in mice after extracorporeal photopheresis (ECP) but reduced with glucocorticosteroids, TNFα blockade, and α4β7-integrin inhibition. Local adiponectin production elicits a tissue-specific effect by reducing pro-inflammatory T cell frequencies in the colon while sparing tumor-specific T cell development. A prospective phase-1b/2 trial (EudraCT-No.2021-002073-26) with 14 patients reveals low ECP-related toxicity. Overall response rate for all irAEs is 92% (95% confidence interval [CI]: 63.97%-99.81%); colitis-specific complete remission rate is 100% (95% CI: 63.06%-100%). Glucocorticosteroid dosages could be reduced for all patients after ECP therapy. The ECP-adiponectin axis reduces intestinal tissue-resident memory T cell activation and CD4+IFN-γ+ T cells in patients with ICI-induced colitis without evidence of loss of anti-tumor immunity. In conclusion, we identify adiponectin as an immunomodulatory molecule that controls ICI-induced irAEs without blocking anti-tumor immunity.
    Keywords:  adiponectin; anti-tumor immunity; arginase-1; cancer immunotherapy; colitis; extracorporeal photopheresis; immune checkpoint inhibition; immune-related adverse events; immunomodulation; immunosuppression
    DOI:  https://doi.org/10.1016/j.ccell.2025.01.004
  21. Oncogene. 2025 Feb 10.
      AML is a complex disease caused by multiple molecular mechanisms. As an important regulatory molecule, the role of circRNA in AML is not fully understood. By performing high-throughput sequencing on clinical samples, we systematically identified the differences in circRNA expression and distribution between AML and healthy donor samples. One circular RNA, circAFF2, was found to be significantly upregulated in AML patients. Functional studies showed that knockdown of circAFF2 could significantly inhibit the proliferation of AML cells and promote their apoptosis. Overexpression of circAFF2 can have opposite effects. In vivo experiments showed that transplantation of AML cells with circAFF2 knockdown slowed the proliferation and infiltration and prolonged the survival time of mice compared to controls. Further studies showed that circAFF2 can promote the degradation of PML mRNA by binding to the 3'UTR of PML mRNA, thereby affecting the proliferation and apoptosis of AML cells. In conclusion, our work demonstrates that circAFF2 can bind to PML mRNA to regulate AML cell function, providing new insights into the mechanism of AML development and potential targets for clinical diagnosis and treatment.
    DOI:  https://doi.org/10.1038/s41388-025-03299-y
  22. Leuk Res. 2025 Feb 04. pii: S0145-2126(25)00022-0. [Epub ahead of print]150 107662
      CMML is a heterogenous myelodysplastic/myeloproliferative neoplasm (MDS/MPN) sharing both diseases' molecular and clinical phenotypes. Several models are used to risk-stratify patients diagnosed with CMML. Inflammation plays a pivotal role in developing the disease or its progression and has been linked to worse outcomes. Serum albumin (SA) is an inflammatory marker and/or surrogate for co-morbidities. While the role of SA has been investigated in myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), multiple myeloma, and other cancers, its prognostic value in CMML remains unclear. We identified 919 patients diagnosed with CMML with known SA levels at the time of diagnosis or prior to any therapy. We divided patients into three groups based on SA levels: < 3.5 g/dL, 3.5-4.0 g/dL and > 4.0 g/dL. We then compared the baseline characteristics and outcomes of these three groups. Patients with SA < 3.5 g/dL had higher risk disease according to the CPSS-Molecular model, WHO 2022 classification, and FAB classification. Additionally, patients with SA < 3.5 g/dL had a higher median blast percentage, ferritin levels, WBC, and monocyte count (P < 0.001). These patients were also more likely to be cytopenic and RBC transfusion-dependent (RBC-TD) (P < 0.001). In multivariable Cox regression analysis, SA was independently significant for predicting overall survival (OS) after adjusting for CPSS-Molecular risk, WHO 2022 subtype, proliferative CMML (FAB classification), RBC-TD, and bi/pancytopenia. Therefore, SA is an independent prognostic factor for OS among patients with CMML. Low SA may reflect inflammatory disease status or a surrogate for co-morbidities. Risk stratification models should incorporate serum albumin levels to refine their prognostic value.
    Keywords:  [CMML]; [clinical]; [myeloid malignancy]
    DOI:  https://doi.org/10.1016/j.leukres.2025.107662
  23. Blood Adv. 2025 Feb 14. pii: bloodadvances.2024014905. [Epub ahead of print]
      The pursuit of ex vivo erythrocyte generation has led to the development of various culture systems that simulate the bone marrow microenvironment. However, these models often fail to fully replicate the hematopoietic niche's complex dynamics. In our research, we employ a comprehensive strategy that emphasizes physiological red blood cell (RBC) differentiation using a minimal cytokine regimen. A key innovation in our approach is the integration of a three-dimensional (3D) silk-based scaffold engineered to mimic both the physical and chemical properties of human bone marrow. This scaffold facilitates critical macrophage-RBC interactions and incorporates fibronectin functionalization to support the formation of erythroblastic island (EBI)-like niches. We observed diverse stages of erythroblast maturation within these niches, driven by the activation of autophagy, which promotes organelle clearance and membrane remodeling. This process leads to reduced surface integrin expression and significantly enhances RBC enucleation. Using a specialized bioreactor chamber, millions of RBCs can be detached from the EBIs and collected in transfusion bags via dynamic perfusion. Inhibition of autophagy through pharmacological agents or alpha4 integrin blockade, disrupted EBI formation, preventing cells from completing their final morphological transformations and remaining trapped in the erythroblast stage. Our findings underscore the importance of the bone marrow niche in maintaining the structural integrity of EBIs and highlight the critical role of autophagy in facilitating organelle clearance during RBC maturation. RNA sequencing analysis further confirmed that these processes are uniquely supported by the 3D silk scaffold, which is essential for enhancing RBC production ex vivo.
    DOI:  https://doi.org/10.1182/bloodadvances.2024014905
  24. Mol Cell. 2025 Feb 04. pii: S1097-2765(24)01067-0. [Epub ahead of print]
      Recurrent cancer-causing fusions of NUP98 produce higher-order assemblies known as condensates. How NUP98 oncofusion-driven condensates activate oncogenes remains poorly understood. Here, we investigate NUP98-PHF23, a leukemogenic chimera of the disordered phenylalanine-and-glycine (FG)-repeat-rich region of NUP98 and the H3K4me3/2-binding plant homeodomain (PHD) finger domain of PHF23. Our integrated analyses using mutagenesis, proteomics, genomics, and condensate reconstitution demonstrate that the PHD domain targets condensate to the H3K4me3/2-demarcated developmental genes, while FG repeats determine the condensate composition and gene activation. FG repeats are necessary to form condensates that partition a specific set of transcriptional regulators, notably the KMT2/MLL H3K4 methyltransferases, histone acetyltransferases, and BRD4. FG repeats are sufficient to partition transcriptional regulators and activate a reporter when tethered to a genomic locus. NUP98-PHF23 assembles the chromatin-bound condensates that partition multiple positive regulators, initiating a feedforward loop of reading-and-writing the active histone modifications. This network of interactions enforces an open chromatin landscape at proto-oncogenes, thereby driving cancerous transcriptional programs.
    Keywords:  MLL; NUP98; WDR5; biomolecular condensate; coactivator; fusion protein; leukemia; phase separation; selective partitioning; transcriptional regulation
    DOI:  https://doi.org/10.1016/j.molcel.2024.12.026
  25. Blood Adv. 2025 Feb 12. pii: bloodadvances.2024014632. [Epub ahead of print]
      Pathogenic germline variants affecting proper telomere maintenance result in premature telomere shortening and cause telomere biology disorders (TBDs). While classical dyskeratosis congenita in children is rather well defined, late-onset ("cryptic") TBDs remain underrecognized, resulting in underdiagnosis and inadequate treatment in affected adults. Here, we present a series of adult TBD cases collected through the German TBD reference center between 2014 and 2024. Patients ≥18 years with an age-matched telomere length (TL) < 10th percentile in lymphocytes and detection of either a variant of uncertain significance, a pathogenic or a likely pathogenic variant in TBD-associated genes, and available clinical data were included in this analysis. On this basis, a novel point-based algorithm for categorization into proven, probable and suspected-only TBD cases, respectively, was developed. Out of a total of 1,537 TL analyses, 42 patients with proven (n=29) or probable (n=13) TBD were identified. Median age at first clinical manifestation and at diagnosis was 20.0 years and 34.1 years, respectively. Bone marrow failure (BMF) was the most frequent manifestation observed in our cohort (73.8%), followed by liver or interstitial lung diseases (50.0% and 41.5%, respectively). Immunosuppressive therapy was carried out in six patients with BMF, none of them responded. In comparison, eight of eight evaluable patients treated with androgen derivatives showed hematologic response. Our data provide novel real-world insight into the clinical manifestation spectrum, diagnosis as well as clinical course and treatment of TBD in adult, late-onset cases of this hereditary disease.
    DOI:  https://doi.org/10.1182/bloodadvances.2024014632