Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,984)

Search Parameters:
Keywords = acute myeloid leukemia

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2441 KB  
Article
Aberrant CD25 and Increased CD123 Expression Are Common in Acute Myeloid Leukemia with KMT2A Partial Tandem Duplication and Are Associated with FLT3 Internal Tandem Duplication
by Qing Wei, Guilin Tang, Shaoying Li, Sa A. Wang, Pei Lin, Wei Wang, Sanam Loghavi, Wei J. Wang, L. Jeffrey Medeiros and Jie Xu
Cancers 2026, 18(2), 282; https://doi.org/10.3390/cancers18020282 - 16 Jan 2026
Abstract
Background: KMT2A partial tandem duplication (PTD) occurs in approximately 5–10% of acute myeloid leukemia (AML) cases and is associated with poor prognosis. While its cytogenetic and molecular features are well described, the immunophenotypic characteristics of AML with KMT2A-PTD remain incompletely defined. Methods: [...] Read more.
Background: KMT2A partial tandem duplication (PTD) occurs in approximately 5–10% of acute myeloid leukemia (AML) cases and is associated with poor prognosis. While its cytogenetic and molecular features are well described, the immunophenotypic characteristics of AML with KMT2A-PTD remain incompletely defined. Methods: We identified 47 cases of AML with KMT2A-PTD by optical genome mapping. All cases underwent flow cytometric immunophenotypic analysis and next-generation sequencing using an 81-gene panel. Results: The cohort included 32 men and 15 women with a median age of 67 years (range, 19–87). Thirty-eight cases were de novo AML, and nine were secondary to myelodysplastic syndrome and/or myeloproliferative neoplasm. Most cases (93%) demonstrated a normal or non-complex karyotype. The most frequent mutations involved FLT3-ITD (47%), DNMT3A (43%), and RUNX1 (23%). Thirty-one cases (66%) were granulocytic, while 16 (34%) showed granulocytic and/or monocytic differentiation. Blasts uniformly expressed HLA-DR and frequently expressed CD117 (91%) and CD34 (79%). Increased expression of CD123 (74%) and CD117 (43%) and decreased expression of HLA-DR (74%) and CD38 (69%) were common. Aberrant CD25 expression was observed in 51% of cases. Increased CD123 and aberrant CD25 expression were significantly associated with FLT3-ITD mutations (both p < 0.0001) but not with other recurrent mutations. There was no correlation between FLT3-ITD mutation and expression levels of CD117, CD38 or HLA-DR (all p > 0.05). Conclusions: AML with KMT2A-PTD shows distinctive immunophenotypic features with increased CD123 and aberrant CD25 expression, both associated with FLT3-ITD. These markers may have diagnostic and therapeutic relevance in this AML subtype. Full article
(This article belongs to the Special Issue Advances in Pathology of Lymphoma and Leukemia)
Show Figures

Figure 1

19 pages, 1529 KB  
Review
Marrow Microenvironmental Pathobiology and Therapeutic Opportunities for TP53-Mutated Myelodysplastic Syndrome/Acute Myeloid Leukemia
by Cameron J. Hunter, Annie P. Im and Rory M. Shallis
Cancers 2026, 18(2), 275; https://doi.org/10.3390/cancers18020275 - 16 Jan 2026
Abstract
Mutations in TP53 inhibit p53 protective behaviors including cell cycle arrest, DNA damage repair protein recruitment, and apoptosis. The ubiquity of p53 in genome-stabilizing functions leads to an aberrant tumor microenvironment in TP53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Profound [...] Read more.
Mutations in TP53 inhibit p53 protective behaviors including cell cycle arrest, DNA damage repair protein recruitment, and apoptosis. The ubiquity of p53 in genome-stabilizing functions leads to an aberrant tumor microenvironment in TP53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Profound immunosuppression mediated by myeloid-derived suppressor cells, the upregulation of cytokines and cell-surface receptors on leukemic cells, the suppression of native immune regulator cells, and metabolic aberrations in the bone marrow are features of the TP53-mutated AML/MDS marrow microenvironment. These localized changes in the bone marrow microenvironment (BMME) explain why traditional therapies for MDS/AML, including chemotherapeutics and hypomethylating agents, are not as effective in TP53-mutated myeloid neoplasms and demonstrate the dire need for new treatments in this patient population. The unique pathophysiology of TP53-mutated disease also provides new therapeutic approaches which are being studied, including intracellular targets (MDM2, p53), cell-surface protein biologics (immune checkpoint inhibitors, BiTE therapy, and antibody–drug conjugates), cell therapies (CAR-T, NK-cell), signal transduction pathways (Hedgehog, Wnt, NF-κB, CCRL2, and HIF-1α), and co-opted biologic pathways (cholesterol synthesis and glycolysis). In this review, we will discuss the pathophysiologic anomalies of the tumor microenvironment in TP53-mutant MDS/AML, the hypothesized mechanisms of chemoresistance it imparts, and how novel therapies are leveraging diverse therapeutic targets to address this critical area of need. Full article
Show Figures

Figure 1

15 pages, 631 KB  
Review
Cell Lines in Myelodysplastic Syndromes/Neoplasms (MDS) Research: A Review of Existing Models and Their Applications
by Karolina Maślińska-Gromadka, Małgorzata Palusińska, Julia Weronika Łuczak, Rafał Skopek, Leszek Kraj, Tino Schenk, Artur Zelent and Łukasz Szymański
Int. J. Mol. Sci. 2026, 27(2), 898; https://doi.org/10.3390/ijms27020898 - 16 Jan 2026
Abstract
Myelodysplastic syndromes/neoplasms (MDS) are clonal hematopoietic disorders characterized by ineffective hematopoiesis, cytopenias, and a variable risk of progression to secondary acute myeloid leukemia (sAML). Despite major advances in the molecular and clinical characterization of MDS, mechanistic and translational research remains constrained by the [...] Read more.
Myelodysplastic syndromes/neoplasms (MDS) are clonal hematopoietic disorders characterized by ineffective hematopoiesis, cytopenias, and a variable risk of progression to secondary acute myeloid leukemia (sAML). Despite major advances in the molecular and clinical characterization of MDS, mechanistic and translational research remains constrained by the limited availability of well-validated in vitro models. Many historically used cell lines are difficult to maintain, exhibit restricted proliferative capacity, or represent advanced disease stages rather than bona fide MDS, while others have been affected by misidentification or cross-contamination. This review provides a comprehensive and critical overview of currently available MDS and MDS-related cell lines, including MDS92, MDS-L and its sublines, M-TAT, TER-3, SKK-1, SKM-1, and MOLM-17/18. We summarize their clinical origin, cytogenetic and molecular features, growth factor dependence, differentiation capacity, and experimental applications, with particular emphasis on their relevance to disease stage, clonal evolution, and leukemic transformation. In addition, we discuss the controversy surrounding misidentified models such as PC-MDS and highlight the importance of rigorous cell line authentication. Full article
Show Figures

Figure 1

13 pages, 2358 KB  
Article
Low-VAF TP53-Mutated AML Displays Distinct Biological Features in a Single-Center Cohort
by Xiaoxuan Lu, Xiaohang Ma, Kainan Zhang, Shun Zhang, Fangfang Wei, Hao Jiang, Qian Jiang, Yingjun Chang, Xiaojun Huang and Xiaosu Zhao
Biomedicines 2026, 14(1), 180; https://doi.org/10.3390/biomedicines14010180 - 14 Jan 2026
Viewed by 83
Abstract
Background: The International Consensus Classification (ICC) currently proposes an empirical variant allele frequency (VAF) threshold of 10% to define TP53-mutated acute myeloid leukemia (AML) and to distinguish oncogenic driver from concomitant mutations. However, the optimal cutoff remains uncertain, and the biological and [...] Read more.
Background: The International Consensus Classification (ICC) currently proposes an empirical variant allele frequency (VAF) threshold of 10% to define TP53-mutated acute myeloid leukemia (AML) and to distinguish oncogenic driver from concomitant mutations. However, the optimal cutoff remains uncertain, and the biological and clinical features of low-VAF cases have not been systematically characterized. Methods: In this single-center retrospective cohort study, we stratified TP53-mutated AML by a 10% VAF cutoff and compared clinical characteristics, cytogenetic and molecular profiles, and survival outcomes between groups. Results: The VAF < 10% group exhibited a distinctive profile: fewer adverse cytogenetic abnormalities [complex karyotype, −7, −5/del(5q)], a more adverse molecular profile (EVI1 overexpression, greater co-mutation burden, higher frequencies of ASXL1 and SRSF2 mutations), and a higher proportion of CD34+CD38 blast immunophenotype. TP53 hotspot mutations were also more common. Survival analyses showed poor prognosis in both groups, and the VAF < 10% group showed numerically longer survival without statistical significance, indicating no clear survival advantage for low VAF. Conclusions: These data support the clinical relevance of the ICC 10% threshold. TP53-mutated AML with VAF < 10% may represent a biologically distinct subgroup. Further multicenter studies with larger cohorts are needed to validate and refine the VAF threshold for prognostic evaluation and individualized management. Full article
Show Figures

Figure 1

16 pages, 366 KB  
Review
Emerging Applications of Triazole Antifungal Drugs
by Luiz Ricardo Soldi, Ana Paula de Lima Oliveira and Marcelo José Barbosa Silva
Int. J. Mol. Sci. 2026, 27(2), 817; https://doi.org/10.3390/ijms27020817 - 14 Jan 2026
Viewed by 74
Abstract
Patients with leukemia are at heightened risk for invasive fungal infections (IFIs) due to profound immunosuppression caused by both the malignancy and its treatment. Chemotherapy-induced neutropenia, mucosal barrier disruption, and impaired innate and adaptive immune responses create a highly permissive environment for opportunistic [...] Read more.
Patients with leukemia are at heightened risk for invasive fungal infections (IFIs) due to profound immunosuppression caused by both the malignancy and its treatment. Chemotherapy-induced neutropenia, mucosal barrier disruption, and impaired innate and adaptive immune responses create a highly permissive environment for opportunistic fungal pathogens. Antifungal prophylaxis, particularly in acute myeloid leukemia (AML), has become a cornerstone in reducing IFI-related morbidity and mortality. This review outlines the immunopathogenic mechanisms underlying susceptibility to IFI and discusses current evidence on the optimal timing and therapeutic strategies for antifungal intervention. The clinical utility of key antifungal agents, namely, posaconazole, isavuconazole, and voriconazole, is critically evaluated. We also examine the potential role of emerging agents such as opelconazole, which enables targeted pulmonary delivery and prolonged epithelial retention, representing a promising approach to IFI prevention. Drug-specific considerations, including pharmacokinetics, drug–drug interactions, toxicity profiles, and cost-effectiveness, are analyzed in the context of clinical decision-making. Finally, we emphasize the importance of tailoring antifungal strategies based on leukemia subtype, immunosuppressive status, and individual patient factors to optimize outcomes and support antifungal stewardship in hematologic malignancies. Full article
(This article belongs to the Collection Feature Papers in Molecular Pharmacology)
Show Figures

Graphical abstract

25 pages, 2645 KB  
Review
Moving Beyond Somatic Alterations: Uncovering the Germline Basis of Myeloid Malignancies
by Ismail Elbaz Younes, Lynh Nguyen and Ling Zhang
Cancers 2026, 18(2), 240; https://doi.org/10.3390/cancers18020240 - 13 Jan 2026
Viewed by 124
Abstract
Myeloid neoplasms (MNs) with germline predisposition represent a distinct, increasingly recognized category in the WHO classification, encompassing myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) arising in the context of an inherited genetic alteration or mutation. While often presenting at a younger age [...] Read more.
Myeloid neoplasms (MNs) with germline predisposition represent a distinct, increasingly recognized category in the WHO classification, encompassing myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) arising in the context of an inherited genetic alteration or mutation. While often presenting at a younger age or with characteristic cytopenias with or without organ dysfunction, some can manifest in adulthood, highlighting the need for vigilance regardless of age or family history. Key predisposing genes include transcription factors (e.g., RUNX1, CEBPA) and genes involved in RNA splicing and telomere biology disorders. Identification of these germline mutations is critical as MNs with germline predisposition dictate specific therapeutic strategies—particularly for hematopoietic stem cell transplantation (HSCT)—and require genetic counseling and surveillance for at-risk relatives. Accurate diagnosis often requires non-hematopoietic germline DNA testing, which provides important biological insights into the development of different myeloid neoplasms and directs personalized patient care. Full article
Show Figures

Figure 1

15 pages, 593 KB  
Review
Beyond XX and XY, Understanding Sex Differences in Leukemia
by Mai Mostafa, Alaa Elhaddad, Mohamed Z. Gad, Rasha Hanafi, Hanaa Rashad and Sami El Deeb
Med. Sci. 2026, 14(1), 38; https://doi.org/10.3390/medsci14010038 - 11 Jan 2026
Viewed by 2298
Abstract
The major subtypes of leukemia show sex differences. This review summarizes current knowledge and identifies gaps regarding sex differences across acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, and chronic lymphoblastic leukemia in epidemiology, mortality and survival rates, risk factors, and epigenetic, [...] Read more.
The major subtypes of leukemia show sex differences. This review summarizes current knowledge and identifies gaps regarding sex differences across acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, and chronic lymphoblastic leukemia in epidemiology, mortality and survival rates, risk factors, and epigenetic, metabolomic, and sex-specific patterns. Males have higher incidence and mortality rates of leukemia compared to females, emphasizing the importance of biological sex. Underreporting of sex differences in leukemia is highlighted, suggesting that sex is often overlooked as a research variable. A significant clinical observation is that women demonstrate higher overall survival rates but experience more severe treatment-related toxicity. Clinically, women tend to survive longer but experience more severe side effects. In contrast, a significant clinical observation in pediatric leukemia contradicts this enigma, suggesting that sex differences may be less pronounced during childhood. These differences play a significant role in how the disease develops. This review presents a sex-based perspective for hematological and biochemical patterns, genetic risk factors, environmental, lifestyle, and parental risk factors, epigenetics and metabolites. Furthermore, males and females might have different responses to the same toxic, environmental, and hormonal exposures. Trying to understand these disparities better based on molecular mechanisms is considered an approach for precision medicine. Full article
Show Figures

Graphical abstract

33 pages, 24811 KB  
Article
Demystifying Deep Learning Decisions in Leukemia Diagnostics Using Explainable AI
by Shahd H. Altalhi and Salha M. Alzahrani
Diagnostics 2026, 16(2), 212; https://doi.org/10.3390/diagnostics16020212 - 9 Jan 2026
Viewed by 261
Abstract
Background/Objectives: Conventional workflows, peripheral blood smears, and bone marrow assessment supplemented by LDI-PCR, molecular cytogenetics, and array-CGH, are expert-driven in the face of biological and imaging variability. Methods: We propose an AI pipeline that integrates convolutional neural networks (CNNs) and transfer [...] Read more.
Background/Objectives: Conventional workflows, peripheral blood smears, and bone marrow assessment supplemented by LDI-PCR, molecular cytogenetics, and array-CGH, are expert-driven in the face of biological and imaging variability. Methods: We propose an AI pipeline that integrates convolutional neural networks (CNNs) and transfer learning-based models with two explainable AI (XAI) approaches, LIME and Grad-Cam, to deliver both high diagnostic accuracy and transparent rationale. Seven public sources were curated into a unified benchmark (66,550 images) covering ALL, AML, CLL, CML, and healthy controls; images were standardized, ROI-cropped, and split with stratification (80/10/10). We fine-tuned multiple backbones (DenseNet-121, MobileNetV2, VGG16, InceptionV3, ResNet50, Xception, and a custom CNN) and evaluated the accuracy and F1-score, benchmarking against the recent literature. Results: On the five-class task (ALL/AML/CLL/CML/Healthy), MobileNetV2 achieved 97.9% accuracy/F1, with DenseNet-121 reaching 97.66% F1. On ALL subtypes (Benign, Early, Pre, Pro) and across tasks, DenseNet121 and MobileNetV2 were the most reliable, achieving state-of-the-art accuracy with the strongest, nucleus-centric explanations. Conclusions: XAI analyses (LIME, Grad-CAM) consistently localized leukemic nuclei and other cell-intrinsic morphology, aligning saliency with clinical cues and model performance. Compared with baselines, our approach matched or exceeded accuracy while providing stronger, corroborated interpretability on a substantially larger and more diverse dataset. Full article
Show Figures

Figure 1

12 pages, 247 KB  
Article
Incidence and Characteristics of Perianal Infections in CPX-351-Treated AML Patients
by Elisa Buzzatti, Cristina Mauro, Cristiano Tesei, Giovangiacinto Paterno, Raffaele Palmieri, Fabiana Esposito, Elisa Meddi, Federico Moretti, Marco Zomparelli, Lucia Cardillo, Carmelo Gurnari, Luca Maurillo, Francesco Buccisano, Adriano Venditti and Maria Ilaria Del Principe
Cancers 2026, 18(2), 208; https://doi.org/10.3390/cancers18020208 - 9 Jan 2026
Viewed by 182
Abstract
Background: Perianal infections (PIs) are a serious threat in patients with acute myeloid leukemia (AML). While CPX-351 is designed to reduce gastrointestinal toxicity, its impact on the incidence of PIs is unknown. This study aims to evaluate the incidence and characteristics of PIs [...] Read more.
Background: Perianal infections (PIs) are a serious threat in patients with acute myeloid leukemia (AML). While CPX-351 is designed to reduce gastrointestinal toxicity, its impact on the incidence of PIs is unknown. This study aims to evaluate the incidence and characteristics of PIs in a cohort of CPX-351-treated AML patients. Methods: We enrolled 22 adult patients diagnosed with secondary AML receiving CPX-351 between May 2020 and July 2025 at Policlinico Tor Vergata Hospital. Statistical analysis used descriptive statistics and multivariate analysis. Results: The incidence of PIs in the cohort was 31.8%. Microbiological cultures from the lesions commonly yielded Klebsiella pneumoniae and Enterococcus species. The development of a PI was associated with a significantly longer hospital stay (mean, 49.6 vs. 37.7 days; p = 0.034). An increased odds ratio of having PIs was noted for mucositis and positive rectal swabs (17.961, p = 0.062; 5.554, p = 0.391, respectively), with two patients (28.5%) having a positive pre-infection swab for Klebsiella pneumoniae. Surgical intervention was guided by patient pain levels and hematological criteria. Surgical patients had significantly higher pain levels (p = 0.001) and a platelet count greater than 20 × 109/L (p = 0.028). All patients were alive at 30 days, with low rates of septic shock (14.2%, n = 1) and no infection-related mortality or recurrence. Conclusions: Despite CPX-351’s known reduced gastrointestinal toxicity, our study showed a significantly higher incidence of PIs compared to literature data. While the outcomes were favorable, PIs led to prolonged hospitalization. Routine rectal swab surveillance could be a valuable tool for risk stratification and preemptive strategies. Full article
(This article belongs to the Special Issue The Unseen Burden: Incidence and Outcomes of Infections in Leukemia)
54 pages, 3566 KB  
Review
Implementation of Natural Products and Derivatives in Acute Myeloid Leukemia Management: Current Treatments, Clinical Trials and Future Directions
by Faten Merhi, Daniel Dauzonne and Brigitte Bauvois
Cancers 2026, 18(2), 185; https://doi.org/10.3390/cancers18020185 - 6 Jan 2026
Viewed by 531
Abstract
Bioactive natural products (NPs) may play a critical role in cancer progression by targeting nucleic acids and a wide array of proteins, including enzymes. Furthermore, a large number of derivatives (NPDs), including semi-synthetic products and pharmacophores from NPs, have been developed to enhance [...] Read more.
Bioactive natural products (NPs) may play a critical role in cancer progression by targeting nucleic acids and a wide array of proteins, including enzymes. Furthermore, a large number of derivatives (NPDs), including semi-synthetic products and pharmacophores from NPs, have been developed to enhance the solubility and stability of NPs. Acute myeloid leukemia (AML) is a poor-prognosis hematologic malignancy characterized by the clonal accumulation in the blood and bone marrow of myeloid progenitors with high proliferative capacity, survival and propagation abilities. A number of potential pathways and targets have been identified for development in AML, and include, but are not limited to, Fms-like tyrosine kinase 3 (FLT3) and isocitrate dehydrogenases resulting from genetic mutations, BCL2 family members, various signaling kinases and histone deacetylases, as well as tumor-associated antigens (such as CD13, CD33, P-gp). By targeting nucleic acids, FLT3 or CD33, several FDA-approved NPs and NPDs (i.e., cytarabine, anthracyclines, midostaurin, melphalan and calicheamicin linked to anti-CD33) are the major agents of upfront treatment of AML. However, the effective treatment of the disease remains challenging, in part due to the heterogeneity of the disease but also to the involvement of the bone marrow microenvironment and the immune system in favoring leukemic stem cell persistence. This review summarizes the current state of the art, and provides a summary of selected NPs/NPDs which are either entering or have been investigated in preclinical and clinical trials, alone or in combination with current chemotherapy. With multifaceted actions, these biomolecules may target all hallmarks of AML, including multidrug resistance and deregulated metabolism. Full article
(This article belongs to the Special Issue Study on Acute Myeloid Leukemia)
Show Figures

Figure 1

16 pages, 707 KB  
Review
AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations
by Sarvath Aafreen Sanaullah, Pierre-Alexandre Vidi and Timothy S. Pardee
Int. J. Mol. Sci. 2026, 27(1), 510; https://doi.org/10.3390/ijms27010510 - 3 Jan 2026
Viewed by 292
Abstract
Racial and ethnic disparities in acute myeloid leukemia (AML) survival persist despite advances in treatment, with non-Hispanic black (NHB) patients and Hispanic patients often experiencing worse outcomes than Non-Hispanic White (NHW) patients due to a combination of clinical, socioeconomic, and biological factors. This [...] Read more.
Racial and ethnic disparities in acute myeloid leukemia (AML) survival persist despite advances in treatment, with non-Hispanic black (NHB) patients and Hispanic patients often experiencing worse outcomes than Non-Hispanic White (NHW) patients due to a combination of clinical, socioeconomic, and biological factors. This review focuses on these disparities and emphasizes potential contributions of biology, as illustrated by the effects of the nucleophosmin 1 (NPM1) mutation. Mutation landscapes and chromosomal abnormalities strongly influence AML patient outcomes. While AML cases with NPM1 mutations are associated with favorable prognoses for NHW patients, NHB patients with NPM1-mutated AML have adverse outcomes. Thus, treatment algorithms and prognostic systems based on outcomes from a single racial ancestry group are inadequate. Beyond the more traditional socioeconomic determinants of health, addressing disparities in AML to achieve equity in care requires exploring biological factors linked to ancestry that shape treatment response. Full article
(This article belongs to the Special Issue Molecular Research in Hematologic Malignancies)
Show Figures

Figure 1

39 pages, 825 KB  
Review
Reawakening Differentiation Therapy in Acute Myeloid Leukemia: A Comprehensive Review of ATRA-Based Combination Strategies
by Shinichiro Takahashi
Curr. Oncol. 2026, 33(1), 25; https://doi.org/10.3390/curroncol33010025 - 2 Jan 2026
Viewed by 351
Abstract
(1) Background: All-trans retinoic acid (ATRA) has transformed the treatment of acute promyelocytic leukemia (APL) by inducing terminal myeloid differentiation. However, its efficacy in non-APL acute myeloid leukemia (AML) is limited. Exploring combination strategies that enhance ATRA-induced differentiation may broaden its therapeutic potential. [...] Read more.
(1) Background: All-trans retinoic acid (ATRA) has transformed the treatment of acute promyelocytic leukemia (APL) by inducing terminal myeloid differentiation. However, its efficacy in non-APL acute myeloid leukemia (AML) is limited. Exploring combination strategies that enhance ATRA-induced differentiation may broaden its therapeutic potential. (2) Methods: A literature search of PubMed using the keywords “ATRA,” “myeloid,” and “differentiation inducer or enhancer” identified more than 500 published papers as of November 2025. Pre-clinical and clinical studies were reviewed, with a focus on mechanisms, combination partners, and translational relevance. (3) Results: Clinical evidence confirms that ATRA combined with arsenic trioxide or epigenetic modulators achieves high remission rates in APL and selected AML subtypes. Pre-clinical studies show synergistic differentiation effects when ATRA is combined with CDK and kinase inhibitors, nucleotide synthesis inhibitors, DNA-damaging agents, Bcl-2/MDM2 inhibitors, proteasome inhibitors, cytokines, glycosylation modifiers, natural products, and antibiotic-derived compounds. Mechanistically, these combinations modulate key signaling pathways (MAPK, Akt, JAK/STAT), stabilize RARα, remodel chromatin, and perturb nucleotide metabolism. Although translation to non-APL AML remains limited, these findings provide a rational basis for future clinical trials. (4) ATRA-based combination therapies represent a promising strategy to extend differentiation therapy beyond APL. This review, authored solely by the investigator, highlights molecular targets and potential enhancers warranting further clinical evaluation in AML. Full article
(This article belongs to the Section Hematology)
Show Figures

Figure 1

12 pages, 715 KB  
Article
Characterizing KMT2A Rearrangement in Acute Myeloid Leukemia: A Comprehensive Genomic Study
by Osama Batayneh, Mahmoudreza Moein, Nour Sabiha Naji, Ansy Patel, Anupa R. Mandava, Alexandra Goodman, Jeffrey S. Ross, Caleb Ho, Chelsea Marcus, Zheng Zhou, Gillian Kupakuwana-Suk, Teresa Gentile and Krishna B. Ghimire
Cancers 2026, 18(1), 161; https://doi.org/10.3390/cancers18010161 - 2 Jan 2026
Viewed by 363
Abstract
Background: The KMT2A (MLL1) gene is altered in a variety of hematological malignancies and solid tumors. KMT2A-rearranged (KMT2Ar) AML represents a distinct subtype associated with poor outcomes and high relapse rate despite initial responsiveness to chemotherapy. Methods [...] Read more.
Background: The KMT2A (MLL1) gene is altered in a variety of hematological malignancies and solid tumors. KMT2A-rearranged (KMT2Ar) AML represents a distinct subtype associated with poor outcomes and high relapse rate despite initial responsiveness to chemotherapy. Methods: A total of 3863 cases of AML peripheral blood samples were analyzed using the FoundationOne Heme combined comprehensive hybrid capture-based DNA and RNA sequencing assay. Results: Of the 3863 AML cases, 521 (13.4%) featured genomic alterations (GAs) in the KMT2A gene, 99.1% of which were large rearrangements (KMT2Ar). A total of 56.9% were males with a median age of 62 years. Of the KMT2Ar cases, there were 43.1% KMT2A duplications, 52.7% fusions, and 4.2% not otherwise specified rearrangements. A total of 0.9% of the KMT2A-altered AML cases were short variant mutations. There were no KMT2A (0%) amplifications or deletions. KMT2Ar cases were associated with increased GA frequencies in FLT3 (27.3% vs. 19.8%; p = 0.0002), KRAS (17.2% vs. 7.8%; p < 0.0001) (overall; 1.1% KRAS G12C), and IDH2 (16.0% vs. 10.4%; p < 0.0001), while KMT2A wild-type AML (KMT2Awt) had significantly increased GA frequencies in RUNX1 (20.7% vs. 15.8%; p = 0.0081), ASXL1 (16.6% vs. 10.5%; p = 0.0003), and TET2 (16.4% vs. 10.1%; p = 0.0002), NPM1 (17.5% vs. 0.2%; p < 0.0001), and TP53 (17.8% vs. 7.9%; p < 0.0001). Conclusions: KMT2A rearrangements are common in AML (13.4% of cases featured KMT2Ar). A total of 99.1% of alterations in KMT2A are large rearrangements, with fusions being the most commonly observed alteration (52.7% of total rearrangements). No amplifications or deletions were seen. This genomic landscape study highlights significant genomic differences between KMT2Ar and KMT2Awt AML patients, which may enrich our understanding of the molecular profile and clusters of mutations in AML. Full article
Show Figures

Figure 1

14 pages, 467 KB  
Article
15-Day Duration of Venetoclax Combined with Azacitidine in Treatment-Naive Higher-Risk Myelodysplastic Syndromes: A Prospective Multicenter Study
by Binbin Lai, Chen Mei, Xiao Yan, Lieguang Chen, Yi Wang, Lixia Sheng, Shanhao Tang, Liping Mao, Ping Zhang, Yongcheng Sun, Wanzhuo Xie, De Zhou, Wenyuan Mai, Huafeng Wang, Liya Ma, Yinjun Lou, Wenjun Wu, Huifang Jiang, Jin Zhang, Baodong Ye, Hongyan Tong and Guifang Ouyangadd Show full author list remove Hide full author list
Cancers 2026, 18(1), 159; https://doi.org/10.3390/cancers18010159 - 2 Jan 2026
Viewed by 341
Abstract
Background: Higher-risk myelodysplastic syndromes (HR-MDS) carry a high risk of progression to acute myeloid leukemia and poor overall survival. Hypomethylating agents (HMAs), such as azacitidine, remain the standard of care but have limited efficacy. A 15-day venetoclax-azacitidine regimen has shown promising objective response [...] Read more.
Background: Higher-risk myelodysplastic syndromes (HR-MDS) carry a high risk of progression to acute myeloid leukemia and poor overall survival. Hypomethylating agents (HMAs), such as azacitidine, remain the standard of care but have limited efficacy. A 15-day venetoclax-azacitidine regimen has shown promising objective response rates (ORR) and potential as a bridge to allogeneic hematopoietic stem cell transplantation (HSCT) in relapsed/refractory HR-MDS. We conducted a prospective multicenter trial to evaluate its efficacy and safety in previously untreated patients. Methods: This multicenter prospective study enrolled treatment-naïve HR-MDS patients (IPSS-R > 3.5). Venetoclax was administered on days 1–15 (escalated from 100 to 400 mg), combined with azacitidine (75 mg/m2) on days 1–7 of each 28-day cycle. The primary endpoint was ORR (2006 IWG criteria); secondary endpoints included complete remission (CR), overall survival (OS), and AML progression. Results: Twenty-eight patients (median age: 63 years) were enrolled, with a median follow-up of 8.5 months. ORR was 85.7% per 2006 IWG (CR: 35.7%, marrow CR: 50.0%), and 78.6% per 2023 IWG (CR: 35.7%). Responses were consistent across molecular and IPSS-R subgroups. Median OS was not reached. High neutrophil count and high cytogenetic risk were favorable factors; TP53 mutation/deletion was an adverse prognostic marker. Grade 3–4 hematologic toxicities included neutropenia (96.4%), anemia (71.4%), and thrombocytopenia (64.3%). Serious adverse events (35.7%) were mainly infections. No dose-limiting or unexpected toxicities were observed. Conclusions: The 15-day venetoclax plus azacitidine regimen demonstrated high efficacy and manageable toxicity in treatment-naïve HR-MDS. It may be particularly beneficial for patients with high neutrophil counts, adverse cytogenetics, or those eligible for HSCT, supporting further investigation in larger trials. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

11 pages, 704 KB  
Article
cellMCD Effectively Discovers Drug Resistance and Sensitivity Genes for Acute Myeloid Leukemia
by Dora Obodo, Nam H. K. Nguyen, Xueyuan Cao, Phani Krishna Parcha, Christopher D. Vulpe, Jatinder K. Lamba and Stanley B. Pounds
Genes 2026, 17(1), 49; https://doi.org/10.3390/genes17010049 - 1 Jan 2026
Viewed by 218
Abstract
Background: Rapid advances in biotechnology provide researchers with the opportunity to integrate omics profiles (genomics, epigenomics, transcriptomics, proteomics, etc.) with multiple phenotypes or experimental conditions. In cancers such as acute myeloid leukemia (AML), where combination therapies are standard of care, identifying genetic drivers [...] Read more.
Background: Rapid advances in biotechnology provide researchers with the opportunity to integrate omics profiles (genomics, epigenomics, transcriptomics, proteomics, etc.) with multiple phenotypes or experimental conditions. In cancers such as acute myeloid leukemia (AML), where combination therapies are standard of care, identifying genetic drivers of drug resistance requires evaluating how genes are associated with multiple drug response phenotypes. Statistical analyses associating omics profiles with multiple phenotypes yield multiple significance values and rankings for each of many genes. There is a great need to consolidate these multiple rankings into a consensus ranking to prioritize specific genes for detailed follow-up wet-lab or clinical studies. Methods/Results: Here, we evaluate the well-known Fisher’s method, the sum of squared z-statistics (SSz), and the recently published cellMCD method as tools for gene prioritization. In simulation studies, cellMCD showed very similar or highly superior performance to the widely used Fisher’s and SSz methods. These advantages were also observed in an example application involving a CRISPR drug screen of an acute myeloid leukemia cell line. Conclusions: In summary, our results indicate that cellMCD should be more widely used for prioritizing discoveries from multiple omic association studies. These methods are available as an R package on github. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Back to TopTop