Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (136)

Search Parameters:
Keywords = myelodysplastic neoplasms

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 1324 KiB  
Review
Advances and Challenges in the Management of Myelodysplastic Syndromes
by Jessica M. Stempel, Tariq Kewan and Amer M. Zeidan
Cancers 2025, 17(15), 2469; https://doi.org/10.3390/cancers17152469 - 25 Jul 2025
Viewed by 986
Abstract
Myelodysplastic syndromes/neoplasms (MDS) represent a biologically and clinically diverse group of myeloid malignancies marked by cytopenias, morphological dysplasia, and an inherent risk of progression to acute myeloid leukemia. Over the past two decades, the field has made significant advances in characterizing the molecular [...] Read more.
Myelodysplastic syndromes/neoplasms (MDS) represent a biologically and clinically diverse group of myeloid malignancies marked by cytopenias, morphological dysplasia, and an inherent risk of progression to acute myeloid leukemia. Over the past two decades, the field has made significant advances in characterizing the molecular landscape of MDS, leading to refined classification systems to reflect the underlying genetic and biological diversity. In 2025, the treatment of MDS is increasingly individualized, guided by integrated clinical, cytogenetic, and molecular risk stratification tools. For lower-risk MDS, the treatment paradigm has evolved beyond erythropoiesis-stimulating agents (ESAs) with the introduction of novel effective agents such as luspatercept and imetelstat, as well as shortened schedules of hypomethylating agents (HMAs). For higher-risk disease, monotherapy with HMAs continue to be the standard of care as combination therapies of HMAs with novel agents have, to date, failed to redefine treatment paradigms. The recognition of precursor states like clonal hematopoiesis of indeterminate potential (CHIP) and the increasing use of molecular monitoring will hopefully enable earlier intervention/prevention strategies. This review provides a comprehensive overview of the current treatment approach for MDS, highlighting new classifications, prognostic tools, evolving therapeutic options, and ongoing challenges. We discuss evidence-based recommendations, treatment sequencing, and emerging clinical trials, with a focus on translating biological insights into improved outcomes for patients with MDS. Full article
(This article belongs to the Special Issue New Insights of Hematology in Cancer)
Show Figures

Figure 1

14 pages, 3307 KiB  
Article
Expanding the Spectrum of CSF3R-Mutated Myeloid Neoplasm Beyond Chronic Neutrophilic Leukemia and Atypical Chronic Myeloid Leukemia: A Comprehensive Analysis of 13 Cases
by Neha Seth, Judith Brody, Peihong Hsu, Jonathan Kolitz, Pratik Q. Deb and Xinmin Zhang
J. Clin. Med. 2025, 14(15), 5174; https://doi.org/10.3390/jcm14155174 - 22 Jul 2025
Viewed by 309
Abstract
Background: Genetic alterations in CSF3R, typically associated with chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML), rarely occur in other myeloid neoplasms. Methods: This study characterized the clinical, morphologic, cytogenetic, and molecular features of 13 patients with non-CNL non-aCML myeloid [...] Read more.
Background: Genetic alterations in CSF3R, typically associated with chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML), rarely occur in other myeloid neoplasms. Methods: This study characterized the clinical, morphologic, cytogenetic, and molecular features of 13 patients with non-CNL non-aCML myeloid neoplasms with CSF3R alterations. Patients (median age, 77 years) were categorized into groups with a myelodysplastic/myeloproliferative neoplasm (MDS/MPN) (n = 5), acute leukemia (n = 4), and other myeloid neoplasms (n = 4) based on the WHO 2022 and ICC criteria. Results: The CSF3R p.Thr618Ile mutation was most frequent (11/13), with additional pathogenic variants including p.Gln743Ter and frameshift mutations affecting the cytoplasmic tail. Variant allele frequencies (VAFs) ranged from 2% to 49%, with the highest median VAF in the MDS/MPN group. Co-mutations varied by subtype; MDS/MPN, NOS, and CMML cases frequently harbored mutations in epigenetic regulators (ASXL1, TET2) and splicing factors (SF3B1, SRSF2, ZRSR2), while acute leukemia cases showed alterations in JAK3, STAT3, and NRAS. Survival analysis revealed distinct patterns across the three diagnostic groups, with MDS/MPN having the poorest prognosis. Conclusion: This study expands the recognized spectrum of CSF3R-related myeloid neoplasms and highlights the clinical and molecular heterogeneity associated with these mutations, emphasizing the need for comprehensive molecular profiling and the potential for targeted therapies. Full article
(This article belongs to the Special Issue Novel Therapeutic Strategies for Acute Myeloid Leukemia)
Show Figures

Figure 1

18 pages, 1674 KiB  
Article
CD34+ Cell Dose, Measurable Residual Disease, and Outcome After Myeloablative HLA-Matched Peripheral Blood Hematopoietic Cell Transplantation for Adults with Acute Myeloid Leukemia
by Margery Gang, Megan Othus, Anne-Chloe Olix, Kate A. Markey, Derek L. Stirewalt, Laura S. Connelly-Smith, Stephanie J. Lee, Filippo Milano and Roland B. Walter
Cancers 2025, 17(14), 2323; https://doi.org/10.3390/cancers17142323 - 12 Jul 2025
Viewed by 396
Abstract
Background: The impact of donor graft cell composition on post-HCT outcomes in AML remains controversial. Furthermore, it is unknown whether this interacts with pre-HCT MRD status. We evaluated the impact of CD34+ and CD3+ cell doses on outcomes of myeloablative conditioning (MAC) [...] Read more.
Background: The impact of donor graft cell composition on post-HCT outcomes in AML remains controversial. Furthermore, it is unknown whether this interacts with pre-HCT MRD status. We evaluated the impact of CD34+ and CD3+ cell doses on outcomes of myeloablative conditioning (MAC) HCT in patients with myelodysplastic neoplasm (MDS)/AML or AML with and without detectable MRD in pre-HCT bone marrow specimens. Methods: We utilized an electronic database to identify all adults ≥18 years with MDS/AML or AML who underwent MAC and received 10/10 HLA-matched sibling or unrelated donor mobilized PBSC allografts in first morphologic remission between 2006 and 2023 at the University of Washington/Fred Hutchinson Cancer Center. Results: Among 385 adults, we found a progressive decrease in relapse incidence and improved survival with increasing CD34+ doses up to a threshold of 5.61 × 106/kg, above which the relapse risk no longer decreased. After multivariable adjustment, a low CD34+ dose was associated with increased risk of relapse as well as lower overall and relapse-free survival. Similar results were obtained for patients with and without pre-HCT MRD. Higher CD3+ doses were linearly associated with an increased incidence of moderate–severe chronic GVHD. Conclusions: Our data identify a non-linear relationship between CD34+ cell dose and relapse risk in AML patients undergoing myeloablative allogeneic HCT, with no apparent added benefit beyond a CD34+ dose threshold. Our findings suggest that donor graft composition impacts outcomes in adults with AML undergoing allogeneic HCT after MAC, independent of pre-HCT MRD status; however, additional studies are needed for other donor cell scenarios. Full article
Show Figures

Figure 1

7 pages, 4563 KiB  
Case Report
Diagnosis and Treatment of Langerhans Cell Sarcoma: A Case Report and Review of the Literature
by Giulia Pileggi, Sabrina Mariani, Valentina De Santis, Gianluca Maiorana, Federica Lubrano Lobianco, Chiara Togni, Monica Piedimonte, Caterina Tatarelli, Esmeralda Conte, Arianna di Napoli, Emanuela Pilozzi, Evelina Rogges, Agostino Tafuri and Giovanna Palumbo
Hemato 2025, 6(3), 18; https://doi.org/10.3390/hemato6030018 - 26 Jun 2025
Viewed by 360
Abstract
Langerhans cell sarcoma (LCS) is a rare and aggressive neoplasm characterized by a clonal proliferation of Langerhans cells (LCs), with multi-organ involvement and poor prognosis. Diagnostic challenges arise from its rarity and overlapping features with Langerhans cell histiocytosis (LCH), requiring immunophenotypic and histological [...] Read more.
Langerhans cell sarcoma (LCS) is a rare and aggressive neoplasm characterized by a clonal proliferation of Langerhans cells (LCs), with multi-organ involvement and poor prognosis. Diagnostic challenges arise from its rarity and overlapping features with Langerhans cell histiocytosis (LCH), requiring immunophenotypic and histological analysis for differentiation. This case report discusses a 67-year-old male with multi-organ LCS involvement. Diagnosis was confirmed via liver biopsy and genetic analysis, revealing a MAP2K1 mutation. Treatment with subcutaneous cladribine and dexamethasone resulted in significant clinical and radiological improvement, despite hematological toxicity due to an underlying myelodysplastic neoplasm (MDS). This case proves the potential efficacy of cladribine for disseminated LCS and highlights the necessity for further research into optimal therapeutic approaches for this rare malignancy. Full article
(This article belongs to the Special Issue Hematopathology: Rare Hematological Diseases)
Show Figures

Figure 1

19 pages, 768 KiB  
Article
From Sanger to Oxford Nanopore MinION Technology: The Impact of Third-Generation Sequencing on Genetic Hematological Diagnosis
by María José Larráyoz, Pablo Luri-Martin, Amagoia Mañu, Oihane Churruca, Natalia Gordillo, Irache Erdozain, Ada Esteban-Figuerola, Carlos de Miguel, Diego Robles, María García-Fortes, José Rifón Roca, Ana Alfonso-Pierola, Felipe Prósper, Beñat Ariceta and María José Calasanz
Cancers 2025, 17(11), 1811; https://doi.org/10.3390/cancers17111811 - 29 May 2025
Viewed by 791
Abstract
Background: Sanger sequencing remains the gold standard for characterizing genetic variants in short DNA fragments (<700 bp). However, the increasing demand for short TATs and high sensitivities in variant detection, particularly in oncohematology, is driving the need for more efficient methods. Next-generation sequencing [...] Read more.
Background: Sanger sequencing remains the gold standard for characterizing genetic variants in short DNA fragments (<700 bp). However, the increasing demand for short TATs and high sensitivities in variant detection, particularly in oncohematology, is driving the need for more efficient methods. Next-generation sequencing (NGS) has improved sensitivity and allows for the simultaneous analysis of multiple genes, but it is still costly and time-consuming. Consequently, Sanger sequencing continues to be widely used. In this study, we have compared Sanger sequencing with Oxford Nanopore technology (ONT), which offers enhanced sensitivity and faster sequencing, delivering diagnostic results within 24 h. Methods: This study involves 164 samples (for a total of 174 analyzed regions of interest) previously characterized using either Sanger sequencing or a next-generation sequencing (NGS) panel, categorized by their genetic alterations. Validation was conducted on 15 genes crucial for the diagnosis, prognosis, or identification of drug resistance in myeloproliferative neoplasms (MPN), myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), and chronic myeloid leukemia (CML). The primary objective was to assess whether MinION could identify the same variants previously detected in these patients. Results and Conclusions: With a 99.43% concordance observed in our comparison, our results support the implementation of MinION technology in routine variant detection in MPN, MDS, AML, and CML cases due to its significant advantages over Sanger sequencing. Full article
(This article belongs to the Special Issue Long-Read Sequencing in Cancer)
Show Figures

Figure 1

20 pages, 2793 KiB  
Article
High Burden of Non-Clonal Chromosome Aberrations Before Onset of Detectable Neoplasia in Fanconi Anemia Bone Marrow
by Silvia Sánchez, Benilde García-de-Teresa, Marco A. Mejía-Barrera, Pedro V. Reyes-Jiménez, Antonio Paz-Martínez, Miguel A. Martínez, Moisés Ó. Fiesco-Roa, Angélica Monsiváis-Orozco, Bertha Molina, Leda Torres, Alfredo Rodríguez and Sara Frias
Cancers 2025, 17(11), 1805; https://doi.org/10.3390/cancers17111805 - 28 May 2025
Viewed by 973
Abstract
Background/objectives: Fanconi anemia (FA) is an inherited bone marrow failure syndrome characterized by chromosome instability and predisposition to develop myelodysplastic neoplasm (MDS) and acute myeloid leukemia (AML). Clonal chromosome aberrations (CCAs) in chromosomes 1, 3, and 7 frequently appear in the bone marrow [...] Read more.
Background/objectives: Fanconi anemia (FA) is an inherited bone marrow failure syndrome characterized by chromosome instability and predisposition to develop myelodysplastic neoplasm (MDS) and acute myeloid leukemia (AML). Clonal chromosome aberrations (CCAs) in chromosomes 1, 3, and 7 frequently appear in the bone marrow (BM) of patients with FA and are associated with MDS/AML progression. Given the underlying DNA repair defect that characterizes FA, non-clonal chromosomal abnormalities (NCCAs) are expected to be common events in the FA BM; in this study, we investigated the presence and significance of NCCA and CCA in the bone marrow (BM) of patients with FA. Methods: Here, we transversally examined the BM karyotypes of 43 non-transplanted patients with FA, 41 with non-clinically detectable hematologic neoplasia and two with diagnosed MDS. We searched for the presence of NCCAs, complex karyotypes (CKs), and CCAs as well as their association with the natural history of the disease, including age, degree of BM failure, and neoplastic transformation. Results: NCCAs were observed in the metaphase spreads of 41/43 FA patients; CKs were observed in 25/43 patients; CCAs were found in 15/43 patients; CCAs involving chromosomes 1, 3 and/or 7 were found in four patients; and other autosomes were found in the remaining 11 patients. Overall, we observed a baseline large karyotypic heterogeneity in the BM of FA patients, demonstrated by the ubiquitous presence of NCCA; such karyotypic heterogeneity precedes the eventual emergence of CKs and selection of cells carrying fitness-improving CCAs. Finally, CCAs involving chromosomes 1, 3 and 7, well-known drivers of hematological malignancy in FA, become established. Overall, we observed that the frequency of NCCAs and CCAs increased with age, even though a significant correlation was not found. Conclusions: These observations fit the model of evolution towards cancer that comprises a first phase of macroevolution represented by NCCAs and karyotypic heterogeneity, followed by the establishment of clones with CCAs, leading to microevolution and cancer. NCCAs are the most frequent chromosomal alterations in the bone marrow of patients with AF and constitute a genome with extensive karyotypic heterogeneity that evolves into clones with more complex genomes and can eventually progress to cancer. Full article
(This article belongs to the Special Issue The Role of Chromosomal Instability in Cancer: 2nd Edition)
Show Figures

Figure 1

30 pages, 1369 KiB  
Review
Myelodysplastic Neoplasms (MDS): Pathogenesis and Therapeutic Prospects
by Xuefeng Li, Chaoyu Zou, Xinrong Xiang, Lei Zhao, Mengran Chen, Chenlu Yang and Yu Wu
Biomolecules 2025, 15(6), 761; https://doi.org/10.3390/biom15060761 - 25 May 2025
Cited by 1 | Viewed by 1169
Abstract
Myelodysplastic neoplasms (MDS) are a group of hematological malignancies originating from hematopoietic stem cells (HSCs), characterized by distinct clinical and/or molecular heterogeneity across different MDS subtypes. This review elucidates the pathogenesis of MDS from two main perspectives: the bone marrow microenvironment and recurrent [...] Read more.
Myelodysplastic neoplasms (MDS) are a group of hematological malignancies originating from hematopoietic stem cells (HSCs), characterized by distinct clinical and/or molecular heterogeneity across different MDS subtypes. This review elucidates the pathogenesis of MDS from two main perspectives: the bone marrow microenvironment and recurrent genetic abnormalities. Abnormal bone marrow microenvironment initiates aberrant innate immune response in HSCs, with quantitative and/or functional alterations of immune cells that collectively establish an immunosuppressive microenvironment, and abnormal bone marrow mesenchymal stromal cells that support and promote the progression of MDS. In addition, this review synthesizes current evidence on the biological functions and pathogenic mechanisms of frequently mutated genes in MDS. Furthermore, emerging therapies based on the pathogenesis of MDS are evaluated and summarized. In summary, aberrant innate immune responses promote pyroptosis of HSCs and acquisition of recurrent genetic abnormalities, resulting in the transformation of HSCs into MDS blasts; the immunosuppressive milieu (especially in higher-risk MDS) facilitates immune evasion of MDS blasts, ultimately leading to disease progression. Future research should focus on the interplay between different genetic abnormalities and immune dysregulation, coupled with the development of novel therapies targeting multiple nodes of the pathogenic network, to overcome current challenges in the treatment of MDS. Full article
Show Figures

Figure 1

11 pages, 1418 KiB  
Article
The Role of BCL-2 Expression in Patients with Myelodysplastic Neoplasms
by Bartłomiej Kuszczak, Krzysztof Zduniak, Angela Jendzierowska, Tomasz Wróbel, Piotr Ziółkowski and Justyna Rybka
Curr. Issues Mol. Biol. 2025, 47(5), 346; https://doi.org/10.3390/cimb47050346 - 10 May 2025
Viewed by 494
Abstract
Myelodysplastic neoplasms (MDS) represent a heterogeneous group of neoplastic bone marrow disorders. A crucial component in regulating bone marrow cell apoptosis is the B-cell CLL/lymphoma 2 (BCL-2) protein. This retrospective study aimed to assess BCL-2 expression by immunohistochemistry in trephine biopsy specimens from [...] Read more.
Myelodysplastic neoplasms (MDS) represent a heterogeneous group of neoplastic bone marrow disorders. A crucial component in regulating bone marrow cell apoptosis is the B-cell CLL/lymphoma 2 (BCL-2) protein. This retrospective study aimed to assess BCL-2 expression by immunohistochemistry in trephine biopsy specimens from 76 patients diagnosed with MDS. The obtained retrospective results were correlated with clinical parameters, including age, sex, MDS subtype, IPSS, IPSS-R, bone marrow blast percentage, Ogata score, response to treatment, blood morphology parameters, and overall survival (OS). The median follow-up duration was 16 months. During the observation period, 58 patients died (median OS of this group: 14.6 months), and 25 patients experienced progression to acute myeloid leukemia. The median BCL-2 expression assessed using the Histoscore (H-score) was 10. Patients with BCL-2 expression below 10 had better survival outcomes than those with expression ≥ 10. Furthermore, patients without detectable BCL-2 expression had significantly better survival compared to those with detectable BCL-2 expression (p = 0.0084). Higher BCL-2 expression was significantly associated with high and very high cytogenetic risk, as defined by IPSS-R. BCL-2 immunohistochemistry should be viewed as a complementary biomarker that, when integrated with IPSS-R and mutational data, could refine therapeutic algorithms. Full article
(This article belongs to the Special Issue Targeting Tumor Microenvironment for Cancer Therapy, 3rd Edition)
Show Figures

Figure 1

16 pages, 794 KiB  
Article
Unmet Needs and Their Impact on Quality of Life and Symptoms in Myelodysplastic Neoplasm Patients and Caregivers
by Elena Crisà, Daniela Cilloni, Marta Riva, Enrico Balleari, Daniela Barraco, Beatrice Manghisi, Lorenza Borin, Michela Calmasini, Anna Calvisi, Isabella Capodanno, Matteo Giovanni Della Porta, Elisa Diral, Bruno Fattizzo, Susanna Fenu, Stefania Paolini, Carlo Finelli, Claudio Fozza, Chiara Frairia, Valentina Giai, Mauro Turrini, Maria Antonia Isoni, Federico Itri, Luca Maurillo, Alfredo Molteni, Giuseppe Alberto Palumbo, Anna Maria Pelizzari, Federica Pilo, Antonella Poloni, Costanza Bosi, Grazia Sanpaolo, Rosaria Sancetta, Cristina Amato, Valeria Santini, Maria Teresa Voso, Sam Salek, Tatyana Ionova, Annamaria Nosari and Esther Natalie Olivaadd Show full author list remove Hide full author list
Cancers 2025, 17(9), 1587; https://doi.org/10.3390/cancers17091587 - 7 May 2025
Viewed by 686
Abstract
Background/Objectives: The aim of this study was to assess the unmet needs of myelodysplastic neoplasm (MDS) patients and their caregivers, focusing on how these needs impact quality of life (QoL) and daily functioning. MDS predominantly affects older adults. It is often complicated by [...] Read more.
Background/Objectives: The aim of this study was to assess the unmet needs of myelodysplastic neoplasm (MDS) patients and their caregivers, focusing on how these needs impact quality of life (QoL) and daily functioning. MDS predominantly affects older adults. It is often complicated by severe red blood cell transfusion-dependent anemia and may require frequent hospital visits, conferring a substantial burden on patients and caregivers. Methods: A national survey was conducted between June 2022 and May 2023 in 46 hematology centers across Italy, involving 259 patients and 105 caregivers. The survey included validated QoL tools (QOL-E and HM-PRO) to measure the impact of disease and treatments on health-related QoL and symptoms. Results: Of the 259 patients surveyed, 42% were transfusion-dependent, with 45% reporting distress related to hospital travel, which was significantly associated with lower QoL scores (QOL-E physical score 50.0 vs. 62.5, p < 0.001). Transfusion dependency led to worse outcomes across physical, emotional, and social domains (HM-PRO Part A score 59.8 vs. 23.7, p < 0.001). Anxiety affected 66% of patients, while 56% reported feeling emotionally distressed. Forty-eight percent of patients required a caregiver, and among caregivers, 29% reported significant disruption to their work, including changing their job or reduced hours. Patients requiring frequent hospital visits showed notably worse QoL scores (HM-PRO emotional score 56.8 vs. 31.8, p < 0.001). Conclusions: This study identified substantial unmet needs for MDS patients, particularly in addressing the heavy burden of transfusions and hospital visits. Both patients and caregivers experienced significant impact on daily life and on QoL, highlighting the urgent need for treatments that reduce hospital dependency, improve patient outcomes, and alleviate the caregiver burden. Full article
(This article belongs to the Special Issue Symptom Burden in Cancer: Assessment and Management)
Show Figures

Graphical abstract

15 pages, 1577 KiB  
Article
Clinical Utility of Optical Genome Mapping as an Additional Tool in a Standard Cytogenetic Workup in Hematological Malignancies
by Gokce A. Toruner, Shimin Hu, Sanam Loghavi, Chi Young OK, Zhenya Tang, Qing Wei, Rashmi Kanagal-Shamanna, L. Jeffrey Medeiros and Guilin Tang
Cancers 2025, 17(9), 1436; https://doi.org/10.3390/cancers17091436 - 25 Apr 2025
Cited by 1 | Viewed by 952
Abstract
Background and Objective: The primary objective of this study is to evaluate the added value of optical genome mapping (OGM) when integrated into the standard cytogenetic workup (SCGW) for hematological malignancies. Methods: The study cohort comprised 519 cases with different types of hematological [...] Read more.
Background and Objective: The primary objective of this study is to evaluate the added value of optical genome mapping (OGM) when integrated into the standard cytogenetic workup (SCGW) for hematological malignancies. Methods: The study cohort comprised 519 cases with different types of hematological malignancies. OGM and SCGW (including G-banded karyotyping and fluorescence in situ hybridization) were performed on blood and/or bone marrow. The analytical sensitivity of OGM, defined as the detection of all additional cytogenomic aberrations, and its clinical utility, referring to aberrations with diagnostic, prognostic, or therapeutic significance, were assessed. Results: OGM led to increased analytical sensitivity and clinical utility in 58% and 15% of the cases, respectively. The clinical utility varied across different malignancies, with the highest utility in T-lymphoblast leukemia (52%), followed by mixed phenotype acute leukemia (43%), B-lymphoblastic leukemia (37%), other B-cell lymphomas (22%), mature T-cell leukemia/lymphoma (20%), chronic lymphocytic leukemia (14%), acute myeloid leukemia (13%), multiple myeloma (13%), mantle cell lymphoma (8%), myelodysplastic/myeloproliferative neoplasms (6%), myelodysplastic syndrome (5%), and myeloproliferative neoplasms (0%). Conclusion: Compared to SCGW, OGM detects additional cytogenomic aberrations in approximately 58% of cases. OGM provides clinical utility at varying rates across different types of hematological malignancies. Given these differences, strategic triaging can help maximize the clinical value of OGM by focusing on diseases where it offers the most significant benefit. Full article
(This article belongs to the Special Issue Diagnostic Biomarkers in Cancers Study)
Show Figures

Figure 1

19 pages, 8160 KiB  
Article
High-Efficiency Enrichment of Megakaryocytes and Identification of Micromegakaryocytes from Human Bone Marrow by Imaging Flow Cytometry
by Maya Nautrup Pedersen, Trine Engelbrecht Hybel, Jens Haugbølle Bjerre, Anne Sofie Borg Hammer, Anja Bille Bohn, Marie Bill, Carina Agerbo Rosenberg and Maja Ludvigsen
Cells 2025, 14(8), 588; https://doi.org/10.3390/cells14080588 - 12 Apr 2025
Viewed by 967
Abstract
Megakaryocytes (MKs) are rare, large, polyploid bone marrow (BM) cells responsible for the production of platelets. The identification and characterization of MKs is widely recognized as challenging. Manual microscopy is especially difficult due to the rarity and complex morphology of MKs, while flow [...] Read more.
Megakaryocytes (MKs) are rare, large, polyploid bone marrow (BM) cells responsible for the production of platelets. The identification and characterization of MKs is widely recognized as challenging. Manual microscopy is especially difficult due to the rarity and complex morphology of MKs, while flow cytometry faces additional challenges from MKs’ large size, fragility, and platelet adhesion, causing false positives. We present a novel approach to accurately enrich MKs from human BM aspirates with a specific focus on the detection and quantification of microMKs. By integrating CD41+ cell enrichment, immunophenotyping, and morphometric analysis, we identified cells of the megakaryocytic lineage. To increase accuracy, a convolutional neural network was trained to identify CD41 cells falsely displaying an MK-like immunophenotype due to adhesive CD41+ platelets. This allowed for exclusion of 94.9% of false positive events, considerably enhancing specificity. CD41 positive enrichment prior to imaging flow cytometry acquisition increased the MK frequency nearly 200-fold, yielding a population of both mature and immature MKs, thus supporting analysis of MK progenitors. Overall, this advanced approach enables enrichment of MKs from human BM, considerably increasing the accuracy and statistical power of the MK analysis. This may provide an important addition in the context of MK-related diagnostics and research. Full article
(This article belongs to the Special Issue Insight into Developments and Applications of Flow Cytometry)
Show Figures

Figure 1

26 pages, 971 KiB  
Review
The Emerging Role of CD8+ T Cells in Shaping Treatment Outcomes of Patients with MDS and AML
by Athanasios Tasis, Theodoros Spyropoulos and Ioannis Mitroulis
Cancers 2025, 17(5), 749; https://doi.org/10.3390/cancers17050749 - 22 Feb 2025
Cited by 2 | Viewed by 1321
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) [...] Read more.
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

14 pages, 2915 KiB  
Article
Effects of Daratumumab (Anti-CD38) Monoclonal Antibody Therapy on Flow Cytometry Analysis in Multiple Myeloma
by Sharon Koorse Germans, Christine Wamsley Kahlow, Weina Chen and Franklin Fuda
Therapeutics 2025, 2(1), 2; https://doi.org/10.3390/therapeutics2010002 - 18 Feb 2025
Viewed by 1686
Abstract
Background: Plasma cell myeloma is an incurable malignancy of clonal plasma cells. Recent success in immunotherapeutic strategies has altered the landscape of myeloma treatment. Daratumumab is an anti-CD38 IgG kappa monoclonal antibody that has shown great efficacy in the treatment of myeloma. However, [...] Read more.
Background: Plasma cell myeloma is an incurable malignancy of clonal plasma cells. Recent success in immunotherapeutic strategies has altered the landscape of myeloma treatment. Daratumumab is an anti-CD38 IgG kappa monoclonal antibody that has shown great efficacy in the treatment of myeloma. However, Daratumumab brought with it new challenges in post-therapeutic laboratory assessment, including therapeutic antibody interference with serum protein electrophoresis and serum immunofixation electrophoresis assays. In this study, we highlight the interference identified in post-therapeutic flow cytometry analysis related to bound Daratumumab on normal hematopoietic cells. We also highlight the methods of detection of residual plasma cell neoplasm, post-Daratumumab therapy.: A total of 28 patients with refractory plasma cell myeloma who received Daratumumab (2016–2018) were included in this study. Flow cytometry was performed using 4- or 10-color antibody panels (BD FASC Canto) and analyzed by cluster analysis (Cytopaint Classic software) using four tube panels including VS38c for measurable residual disease (MRD) testing. Pretreatment and post-Daratumumab follow-up bone marrow flow cytometry samples were analyzed. In addition, 10 multiple myeloma patient samples were reflexed to multi-epitope CD38 analysis by flow cytometric analysis of post-Daratumumab residual disease. When discussing CD38 expression, we will refer to CD38 as being detected by conventional reagents. Results: All post-Daratumumab-treated cases (100%) showed negative staining for CD38 using conventional reagents on all plasma cells in the specimens. MRD testing successfully identified small clonal plasma cell populations using VS38C and multi-epitope CD38 (meCD38) antibodies, despite the absence of demonstrable CD38 expression. Additionally, all cases exhibited weak kappa light chain staining on hematogones, attributed to the binding of Daratumumab kappa monoclonal antibody. This interaction can create the appearance of a CD10+ monotypic B-cell population. We also noted diminished CD38 staining on myeloblasts, resulting in an atypical CD34/CD38 staining pattern. This alteration could potentially be misinterpreted as indicative of a myelodysplastic neoplasm (MDS). Furthermore, decreased staining of CD38 was noted on T cells, natural killer (NK) cells, basophils, monocytes, and plasmacytoid dendritic cells. Conclusions: With the emergence of successfully targeted immunotherapies, such as anti-CD38 antibodies, it is important to understand and correctly interpret variations in flow cytometry that may arise from the therapy. Hematogones exhibit high-intensity levels of CD38 expression; thus, Daratumumab binds to them, creating the appearance of kappa expression on all hematogones. Stage I/early stage II hematogones normally lack surface immunoglobulin light chain expression, but in the presence of Daratumumab, they appear to be a CD10(+) monotypic population of B cells. The misinterpretation of these normal cells as a CD10(+) B-cell clone can lead to inaccurate assessment, unnecessary bone marrow immunohistochemical evaluation, and unwarranted anxiety. Additionally, artefacts on various other hematopoietic cells can result in inaccurate assessments of immunophenotypic aberrancy due to binding of the drug. This may lead to the false interpretation of a secondary/therapy-related myeloid neoplasm. This study highlights in detail the interferences that must be considered when assessing residual disease in the era of targeted drug therapies. Full article
Show Figures

Figure 1

21 pages, 2013 KiB  
Review
Diagnostic Approaches in Myeloid Sarcoma
by Elzbieta Patkowska, Agnieszka Krzywdzinska, Iwona Solarska, Magdalena Wojtas and Monika Prochorec-Sobieszek
Curr. Issues Mol. Biol. 2025, 47(2), 111; https://doi.org/10.3390/cimb47020111 - 10 Feb 2025
Viewed by 1869
Abstract
Myeloid sarcoma (MS), or extramedullary acute myeloid leukaemia tumour (eAML), is a rare hematopoietic neoplasm. Recognised as a distinct entity within acute myeloid leukaemia (AML), MS presents significant diagnostic challenges due to its rarity, clinical heterogeneity, and variable immunophenotypic and genetic characteristics. The [...] Read more.
Myeloid sarcoma (MS), or extramedullary acute myeloid leukaemia tumour (eAML), is a rare hematopoietic neoplasm. Recognised as a distinct entity within acute myeloid leukaemia (AML), MS presents significant diagnostic challenges due to its rarity, clinical heterogeneity, and variable immunophenotypic and genetic characteristics. The mechanisms by which leukaemic stem cells (LSCs) migrate to form solid tumours in extramedullary (EM) sites remain unclear. MS can occur de novo, precede AML, and manifest alongside AML relapse. It can also develop with myelodysplastic syndromes (MDSs) or myeloproliferative neoplasms (MPNs). MS frequently presents in organs such as the skin, lymph nodes, gastrointestinal (GI) tract, and central nervous system (CNS), often resulting in diverse clinical manifestations. Diagnosis relies on a comprehensive approach, including tissue biopsy, bone marrow (BM) evaluation, and advanced imaging modalities. Accurate diagnosis is crucial for risk stratification and treatment selection. Prognosis is influenced by several factors: MS’s anatomical location, timing of MS diagnosis, genetic profile, and possible treatment. This review emphasises the need for comprehensive diagnostic methods to better define individual MS characteristics and prognosis. It explores the role of novel targeted therapies in improving patient outcomes and further highlights the critical need for future multicentre data collection to optimise diagnostic and therapeutic approaches. Full article
Show Figures

Figure 1

20 pages, 954 KiB  
Review
Unveiling Myelodysplastic Syndromes: Exploring Pathogenic Mechanisms and Therapeutic Advances
by Nishanth Thalambedu, Bhavesh Mohan Lal, Brent Harbaugh, Daisy V. Alapat, Mamatha Gaddam, Cesar Giancarlo Gentille Sanchez, Muthu Kumaran and Ankur Varma
Cancers 2025, 17(3), 508; https://doi.org/10.3390/cancers17030508 - 3 Feb 2025
Viewed by 2040
Abstract
Myelodysplastic syndromes (MDSs), either primary or secondary, are a heterogeneous group of clonal hematological neoplasms characterized by bone marrow dyshematopoiesis, peripheral blood cytopenia, and the potential risk of acute myeloid leukemia (AML) transformation. The clinical heterogeneity in MDS is a reflection of the [...] Read more.
Myelodysplastic syndromes (MDSs), either primary or secondary, are a heterogeneous group of clonal hematological neoplasms characterized by bone marrow dyshematopoiesis, peripheral blood cytopenia, and the potential risk of acute myeloid leukemia (AML) transformation. The clinical heterogeneity in MDS is a reflection of the underlying multitude of genetic defects playing a role in the pathogenesis. Recent advances in the clinicopathological, immunophenotypic, and molecular landscape in understanding the pathophysiology of MDS lead to evolving and refined classification systems with newer entities. Evolving MDS therapies will target the disease’s core mechanisms, allowing for personalized treatment based on individual patient’s genes and leading to better outcomes. This review provides an overview of MDS pathogenesis to enhance comprehension of its various subgroups. Additionally, we examine the updated classification systems of the World Health Organization (WHO) and the International Consensus Classification (ICC) pertaining to MDS, along with relevant therapeutic approaches. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Back to TopTop