Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (20)

Search Parameters:
Keywords = clonal leukemogenesis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 3454 KiB  
Article
RNA Sequencing Identifies WT1 Overexpression as a Predictor of Poor Outcomes in Acute Myeloid Leukemia
by Harsh Goel, Avanish Kumar Pandey, Rahul Kumar, Rakesh Kumar, Somorjit Singh Ningombam, Farhat Naz, Harshita Makkar, Jay Singh, Shadab Ali, Anita Chopra, Amar Ranjan, Aditya Kumar Gupta, Jagdish Prasad Meena, Ganesh Kumar Viswanathan, Sameer Bakhshi, Ranjit Kumar Sahoo, Atul Batra, Goura Kishor Rath, Showket Hussain, Abhimanyu Kumar Jha and Pranay Tanwaradd Show full author list remove Hide full author list
Cancers 2025, 17(11), 1818; https://doi.org/10.3390/cancers17111818 - 29 May 2025
Viewed by 1731
Abstract
Background/Objectives: AML is a heterogeneous hematological malignancy distinguished by the clonal expansion of immature myeloid progenitor cells. Despite advances in therapy, relapse rates remain high, and outcomes are poor. The WT1 gene has emerged as a potential contributor to leukemogenesis, but its clinical [...] Read more.
Background/Objectives: AML is a heterogeneous hematological malignancy distinguished by the clonal expansion of immature myeloid progenitor cells. Despite advances in therapy, relapse rates remain high, and outcomes are poor. The WT1 gene has emerged as a potential contributor to leukemogenesis, but its clinical relevance at the transcriptional level is not fully understood. This study employed RNA sequencing as a discovery tool to identify WT1 gene expression in AML and further investigated its role in diagnosis, prognosis, and treatment response. Methods: Between 2020 and 2024, 345 diagnostic, 259 post-induction, and 70 relapse-stage BM or PB samples were prospectively collected from de novo AML patients at AIIMS, New Delhi. RNA sequencing was initially performed on five paired diagnosis-relapse samples to profile transcriptomic changes and assess WT1 expression dynamics. WT1 expression was further validated by qPCR. The relationship between WT1 expression and various clinical parameters was evaluated using Cox regression analysis to determine its impact on prognosis. Results: RNA sequencing and qPCR confirmed WT1 overexpression at diagnosis, which significantly declined following induction therapy. High WT1 expression at diagnosis was linked with adverse clinical characteristics, including elevated WBC counts and higher blast percentages and predicted poor survival outcomes. WT1 expression was identified as a significant prognostic marker, correlating with OS and EFS. Conclusions: By integrating RNA sequencing with targeted validation, this study highlights WT1 expression as a critical biomarker for AML diagnosis, prognosis, and treatment response. The findings suggest that WT1 expression may serve as a valuable tool for monitoring disease status, risk stratification, and guiding treatment decisions in AML, with potential applications for WT1-targeted precision therapies. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia in Adults)
Show Figures

Figure 1

27 pages, 1035 KiB  
Review
The Role of the Sirtuin Family Histone Deacetylases in Acute Myeloid Leukemia—A Promising Road Ahead
by Piotr Strzałka, Kinga Krawiec, Aneta Wiśnik, Dariusz Jarych, Magdalena Czemerska, Izabela Zawlik, Agnieszka Pluta and Agnieszka Wierzbowska
Cancers 2025, 17(6), 1009; https://doi.org/10.3390/cancers17061009 - 17 Mar 2025
Viewed by 1041
Abstract
Acute myeloid leukemia (AML) corresponds to a heterogeneous group of clonal hematopoietic diseases, which are characterized by uncontrolled proliferation of malignant transformed myeloid precursors and their inability to differentiate into mature blood cells. The prognosis of AML depends on many variables, including the [...] Read more.
Acute myeloid leukemia (AML) corresponds to a heterogeneous group of clonal hematopoietic diseases, which are characterized by uncontrolled proliferation of malignant transformed myeloid precursors and their inability to differentiate into mature blood cells. The prognosis of AML depends on many variables, including the genetic features of the disease. Treatment outcomes, despite the introduction of new targeted therapies, are still unsatisfactory. Recently, there have been an increasing number of reports on enzymatic proteins of the sirtuin family and their potential importance in cancer in general. Sirtuins are a group of 7 (SIRT1-7) NAD+-dependent histone deacetylases with pleiotropic effects on metabolism, aging processes, and cell survival. They are not only responsible for post-translational modification of histones but also play various biochemical functions and interact with other proteins regulating cell survival, such as p53. Thus, their role in key mechanisms of tumorigenesis makes them a worthwhile topic in AML. Different sirtuins have been shown to act oppositely depending on the biological context, the mechanism of which requires further exploration. This review provides a comprehensive description of the significance and role of sirtuins in AML in light of the current state of knowledge. It focuses in particular on molecular mechanisms regulated by sirtuins and signaling pathways involved in leukemogenesis, as well as clinical aspects and potential therapeutic targets in AML. Full article
(This article belongs to the Special Issue Exploring the Genetic and Epigenetic Factors in Leukemia and Lymphoma)
Show Figures

Figure 1

28 pages, 1972 KiB  
Review
Unraveling the Genetic Heterogeneity of Acute Lymphoblastic Leukemia Based on NGS Applications
by Valentina Ramírez Maldonado, Josgrey Navas Acosta, Iván Maldonado Marcos, Ángela Villaverde Ramiro, Alberto Hernández-Sánchez, Jesús M. Hernández Rivas and Rocío Benito Sánchez
Cancers 2024, 16(23), 3965; https://doi.org/10.3390/cancers16233965 - 26 Nov 2024
Viewed by 1965
Abstract
Acute lymphoblastic leukemia (ALL) is a hematological neoplasm characterized by the clonal expansion of abnormal lymphoid precursors in bone marrow, which leads to alterations in the processes of cell differentiation and maturation as a consequence of genetic alterations. The integration of conventional methods, [...] Read more.
Acute lymphoblastic leukemia (ALL) is a hematological neoplasm characterized by the clonal expansion of abnormal lymphoid precursors in bone marrow, which leads to alterations in the processes of cell differentiation and maturation as a consequence of genetic alterations. The integration of conventional methods, such as cytogenetics and immunophenotyping, and next-generation sequencing (NGS) has led to significant improvements at diagnosis and patient stratification; this has also allowed the discovery of several novel molecular entities with specific genetic variants that may drive the processes of leukemogenesis. Nevertheless, the understanding of the process of leukemogenesis remains a challenge since this disease persists as the most frequent cancer in children; it accounts for approximately one-quarter of adult acute leukemias, and the patient management may take into consideration the high intra- and inter-tumor heterogeneity and the relapse risk due to the various molecular events that can occur during clonal evolution. Some germline variants have been identified as risk factors or have been found to be related to the response to treatment. Therefore, better knowledge of the genetic alterations in B-ALL will have a prognostic impact from the perspective of personalized medicine. This review aims to compare, synthesize, and highlight recent findings concerning ALL obtained through NGS that have led to a better understanding of new molecular subtypes based on immunophenotypic characteristics, mutational profiles, and expression profiles. Full article
(This article belongs to the Special Issue Algorithms and Data Analysis of High Throughput Sequencing in Cancers)
Show Figures

Figure 1

19 pages, 2010 KiB  
Review
Immunotherapeutic Potential of Mutated NPM1 for the Treatment of Acute Myeloid Leukemia
by Jochen Greiner, Eithar Mohamed, Daniel M. Fletcher, Patrick J. Schuler, Hubert Schrezenmeier, Marlies Götz and Barbara-ann Guinn
Cancers 2024, 16(20), 3443; https://doi.org/10.3390/cancers16203443 - 10 Oct 2024
Cited by 2 | Viewed by 3100
Abstract
Acute myeloid leukemia (AML) is a malignant disease of the blood and bone marrow that is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Nucleophosmin 1 (NPM1) gene mutations are the most common genetic abnormality in AML, detectable in blast cells [...] Read more.
Acute myeloid leukemia (AML) is a malignant disease of the blood and bone marrow that is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Nucleophosmin 1 (NPM1) gene mutations are the most common genetic abnormality in AML, detectable in blast cells from about one-third of adults with AML. AML NPM1mut is recognized as a separate entity in the World Health Organization classification of AML. Clinical and survival data suggest that patients with this form of AML often have a more favorable prognosis, which may be due to the immunogenicity created by the mutations in the NPM1 protein. Consequently, AML with NPM1mut can be considered an immunogenic subtype of AML. However, the underlying mechanisms of this immunogenicity and associated favorable survival outcomes need to be further investigated. Immune checkpoint molecules, such as the programmed cell death-1 (PD-1) protein and its ligand, PD-L1, play important roles in leukemogenesis through their maintenance of an immunosuppressive tumor microenvironment. Preclinical trials have shown that the use of PD-1/PD-L1 checkpoint inhibitors in solid tumors and lymphoma work best in novel therapy combinations. Patients with AML NPM1mut may be better suited to immunogenic strategies that are based on the inhibition of the PD-1 immune checkpoint pathway than patients without this mutation, suggesting the genetic landscape of patients may also inform best practice for the use of PD-1 inhibitors. Full article
(This article belongs to the Special Issue The Role of NPM1 Mutation in Acute Myeloid Leukemia)
Show Figures

Graphical abstract

11 pages, 1462 KiB  
Article
MicroRNA: A Signature for the Clinical Progression of Chronic Lymphocytic Leukemia
by Yuliya A. Veryaskina, Sergei E. Titov, Igor B. Kovynev, Tatiana I. Pospelova, Sofya S. Fyodorova, Yana Yu. Shebunyaeva, Sergei A. Demakov, Pavel S. Demenkov and Igor F. Zhimulev
Lymphatics 2024, 2(3), 157-167; https://doi.org/10.3390/lymphatics2030013 - 13 Aug 2024
Viewed by 1123
Abstract
Chronic lymphocytic leukemia (CLL) is the most common human leukemia. The disease is caused by abnormal proliferation and development of lymphocytes and their precursors in the blood and bone marrow (BM). Recent studies have shown that the CLL’s clinical course and outcome depend [...] Read more.
Chronic lymphocytic leukemia (CLL) is the most common human leukemia. The disease is caused by abnormal proliferation and development of lymphocytes and their precursors in the blood and bone marrow (BM). Recent studies have shown that the CLL’s clinical course and outcome depend not only on genetic but also epigenetic factors. MicroRNAs (miRNAs) are involved in the development of hematological tumors, including CLL. The aim of this study is to identify the miRNA expression profile in CLL and determine the role of miRNAs in biological pathways associated with leukemogenesis in CLL. The following samples were used in this study: (1) samples obtained by sternal puncture and aspiration biopsy of BM (n = 115). They included samples from 21 CLL patients with anemia and indications for therapy and 45 CLL patients without anemia and with indications for therapy. The control group for the CLL BM samples consisted of patients with non-cancerous blood diseases (n = 35). (2) Lymph node (LN) samples (n = 20) were collected from CLL patients. The control group for the CLL LN samples consisted of patients with lymphadenopathy (n = 37). All cases were patients before treatment. We demonstrated a significant upregulation of miRNA-34a and miRNA-150 in CLL BM samples (p < 0.05) and downregulation of miRNA-451a in CLL LN samples (p < 0.05). We noted a dynamic increase in the levels of miRNA-150 and miRNA-34a in BM at various stages of tumor progression of CLL. We concluded that a dynamic picture of clinical manifestations of CLL closely correlates with changes in epigenetic characteristics of the tumor. Progression of the lymphoproliferative process and indications for cytoreductive therapy are associated with changes in the miRNA profile generated by cancer cells in different sites of clonal expansion. Full article
Show Figures

Figure 1

12 pages, 1753 KiB  
Article
The Leukemic Isocitrate Dehydrogenase (IDH) 1/2 Mutations Impair Myeloid and Erythroid Cell Differentiation of Primary Human Hematopoietic Stem and Progenitor Cells (HSPCs)
by Sara Pierangeli, Serena Donnini, Valerio Ciaurro, Francesca Milano, Valeria Cardinali, Sofia Sciabolacci, Gaetano Cimino, Ilaria Gionfriddo, Roberta Ranieri, Sabrina Cipriani, Eleonora Padiglioni, Roberta Iacucci Ostini, Tiziana Zei, Antonio Pierini and Maria Paola Martelli
Cancers 2024, 16(15), 2675; https://doi.org/10.3390/cancers16152675 - 27 Jul 2024
Cited by 1 | Viewed by 1808
Abstract
How hematopoietic stem and progenitor cell (HSPC) fate decisions are affected by genetic alterations acquired during AML leukemogenesis is poorly understood and mainly explored in animal models. Here, we study isocitrate dehydrogenase (IDH) gene mutations in the human model of HSPC [...] Read more.
How hematopoietic stem and progenitor cell (HSPC) fate decisions are affected by genetic alterations acquired during AML leukemogenesis is poorly understood and mainly explored in animal models. Here, we study isocitrate dehydrogenase (IDH) gene mutations in the human model of HSPC and discuss the available literature on this topic. IDH1/2 mutations occur in ~20% of AML cases, are recognized among the mutations earliest acquired during leukemogenesis, and are targets of specific inhibitors (ivosidenib and enasidenib, respectively). In order to investigate the direct effects of these mutations on HSPCs, we expressed IDH1-R132H or IDH2-R140Q mutants into human CD34+ healthy donor cells via lentiviral transduction and analyzed the colony-forming unit (CFU) ability. CFU ability was dramatically compromised with a complete trilineage block of differentiation. Strikingly, the block was reversed by specific inhibitors, confirming that it was a specific effect induced by the mutants. In line with this observation, the CD34+ leukemic precursors isolated from a patient with IDH2-mutated AML at baseline and during enasidenib treatment showed progressive and marked improvements in their fitness over time, in terms of CFU ability and propensity to differentiate. They attained clonal trilinear reconstitution of hematopoiesis and complete hematological remission. Full article
(This article belongs to the Special Issue Blood Stem Cell and Hematological Malignancies)
Show Figures

Figure 1

14 pages, 770 KiB  
Review
Homing and Engraftment of Hematopoietic Stem Cells Following Transplantation: A Pre-Clinical Perspective
by Tanvir Hasan, Ajay Ratan Pasala, Dhuha Hassan, Justine Hanotaux, David S. Allan and Harinad B. Maganti
Curr. Oncol. 2024, 31(2), 603-616; https://doi.org/10.3390/curroncol31020044 - 23 Jan 2024
Cited by 2 | Viewed by 5508
Abstract
Hematopoietic stem-cell (HSC) transplantation (HSCT) is used to treat various hematologic disorders. Use of genetically modified mouse models of hematopoietic cell transplantation has been critical in our fundamental understanding of HSC biology and in developing approaches for human patients. Pre-clinical studies in animal [...] Read more.
Hematopoietic stem-cell (HSC) transplantation (HSCT) is used to treat various hematologic disorders. Use of genetically modified mouse models of hematopoietic cell transplantation has been critical in our fundamental understanding of HSC biology and in developing approaches for human patients. Pre-clinical studies in animal models provide insight into the journey of transplanted HSCs from infusion to engraftment in bone-marrow (BM) niches. Various signaling molecules and growth factors secreted by HSCs and the niche microenvironment play critical roles in homing and engraftment of the transplanted cells. The sustained equilibrium of these chemical and biologic factors ensures that engrafted HSCs generate healthy and durable hematopoiesis. Transplanted healthy HSCs compete with residual host cells to repopulate stem-cell niches in the marrow. Stem-cell niches, in particular, can be altered by the effects of previous treatments, aging, and the paracrine effects of leukemic cells, which create inhospitable bone-marrow niches that are unfavorable for healthy hematopoiesis. More work to understand how stem-cell niches can be restored to favor normal hematopoiesis may be key to reducing leukemic relapses following transplant. Full article
(This article belongs to the Section Cell Therapy)
Show Figures

Graphical abstract

18 pages, 2207 KiB  
Article
A Comprehensive Metabolism-Related Gene Signature Predicts the Survival of Patients with Acute Myeloid Leukemia
by Yujia Zhai, Heng Shen and Hui Wei
Genes 2024, 15(1), 63; https://doi.org/10.3390/genes15010063 - 31 Dec 2023
Cited by 2 | Viewed by 2805
Abstract
(1) Background: Acute myeloid leukemia (AML) is a clonal malignancy with heterogeneity in genomics and clinical outcome. Metabolism reprogramming has been increasingly recognized to play an important role in the leukemogenesis and prognosis in AML. A comprehensive prognostic model based on metabolism signatures [...] Read more.
(1) Background: Acute myeloid leukemia (AML) is a clonal malignancy with heterogeneity in genomics and clinical outcome. Metabolism reprogramming has been increasingly recognized to play an important role in the leukemogenesis and prognosis in AML. A comprehensive prognostic model based on metabolism signatures has not yet been developed. (2) Methods: We applied Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) normalization to establish a metabolism-related prognostic gene signature based on glycolysis, fatty acid metabolism, and the tricarboxylic acid cycle gene signatures. The Cancer Genome Atlas-Acute Myeloid Leukemia-like (TCGA-LAML) cohort was set as the training dataset for model construction. Three independent AML cohorts (GSE37642, GSE10358, and GSE12417) combined from Gene Expression Omnibus (GEO) datasets and the Beat-AML dataset were retrieved as two validation sets to test the robustness of the model. The transcriptome data and clinic information of the cohorts were enrolled for the analysis. (3) Results: Divided by the median value of the metabolism risk score, the five-year overall survival (OS) of the high-risk and low-risk groups in the training set were 8.2% and 41.3% (p < 0.001), respectively. The five-year OS of the high-risk and low-risk groups in the combined GEO cohort were 25.5% and 37.3% (p = 0.002), respectively. In the Beat-AML cohort, the three-year OS of the high-risk and low-risk groups were 16.2% and 40.2% (p = 0.0035), respectively. The metabolism risk score showed a significantly negative association with the long-term survival of AML. Furthermore, this metabolism risk score was an independent unfavorable factor for OS by univariate analysis and multivariate analysis. (4) Conclusions: Our study constructed a comprehensive metabolism-related signature with twelve metabolism-related genes for the risk stratification and outcome prediction of AML. This novel signature might contribute to a better use of metabolism reprogramming factors as prognostic markers and provide novel insights into potential metabolism targets for AML treatment. Full article
Show Figures

Figure 1

14 pages, 972 KiB  
Review
NPM 1 Mutations in AML—The Landscape in 2023
by Naman Sharma and Jane L. Liesveld
Cancers 2023, 15(4), 1177; https://doi.org/10.3390/cancers15041177 - 12 Feb 2023
Cited by 26 | Viewed by 9086
Abstract
Acute myeloid leukemia (AML) represents 80% of acute leukemia in adults and is characterized by clonal expansion of hematopoietic stem cells secondary to genomic mutations, rendering a selective growth advantage to the mutant clones. NPM1mut is found in around 30% of AML [...] Read more.
Acute myeloid leukemia (AML) represents 80% of acute leukemia in adults and is characterized by clonal expansion of hematopoietic stem cells secondary to genomic mutations, rendering a selective growth advantage to the mutant clones. NPM1mut is found in around 30% of AML and clinically presents with leukocytosis, high blast percentage and extramedullary involvement. Considered as a “gate-keeper” mutation, NPM1mut appears to be a “first hit” in the process of leukemogenesis and development of overt leukemia. Commonly associated with other mutations (e.g., FLT 3, DNMT3A, TET2, SF3B1), NPM1 mutation in AML has an important role in diagnosis, prognosis, treatment and post-treatment monitoring. Several novel therapies targeting NPM1 are being developed in various clinical phases with demonstration of efficacy. In this review, we summarize the pathophysiology of the NPM1 gene mutation in AML, clinical implications and the novel targeted therapies to date. Full article
(This article belongs to the Collection Acute Myeloid Leukemia (AML))
Show Figures

Graphical abstract

36 pages, 2669 KiB  
Review
Old and New Facts and Speculations on the Role of the B Cell Receptor in the Origin of Chronic Lymphocytic Leukemia
by Davide Bagnara, Andrea Nicola Mazzarello, Fabio Ghiotto, Monica Colombo, Giovanna Cutrona, Franco Fais and Manlio Ferrarini
Int. J. Mol. Sci. 2022, 23(22), 14249; https://doi.org/10.3390/ijms232214249 - 17 Nov 2022
Cited by 10 | Viewed by 3661
Abstract
The engagement of the B cell receptor (BcR) on the surface of leukemic cells represents a key event in chronic lymphocytic leukemia (CLL) since it can lead to the maintenance and expansion of the neoplastic clone. This notion was initially suggested by observations [...] Read more.
The engagement of the B cell receptor (BcR) on the surface of leukemic cells represents a key event in chronic lymphocytic leukemia (CLL) since it can lead to the maintenance and expansion of the neoplastic clone. This notion was initially suggested by observations of the CLL BcR repertoire and of correlations existing between certain BcR features and the clinical outcomes of single patients. Based on these observations, tyrosine kinase inhibitors (TKIs), which block BcR signaling, have been introduced in therapy with the aim of inhibiting CLL cell clonal expansion and of controlling the disease. Indeed, the impressive results obtained with these compounds provided further proof of the role of BcR in CLL. In this article, the key steps that led to the determination of the role of BcR are reviewed, including the features of the CLL cell repertoire and the fine mechanisms causing BcR engagement and cell signaling. Furthermore, we discuss the biological effects of the engagement, which can lead to cell survival/proliferation or apoptosis depending on certain intrinsic cell characteristics and on signals that the micro-environment can deliver to the leukemic cells. In addition, consideration is given to alternative mechanisms promoting cell proliferation in the absence of BcR signaling, which can explain in part the incomplete effectiveness of TKI therapies. The role of the BcR in determining clonal evolution and disease progression is also described. Finally, we discuss possible models to explain the selection of a special BcR set during leukemogenesis. The BcR may deliver activation signals to the cells, which lead to their uncontrolled growth, with the possible collaboration of other still-undefined events which are capable of deregulating the normal physiological response of B cells to BcR-delivered stimuli. Full article
(This article belongs to the Collection Feature Papers in Molecular Immunology)
Show Figures

Figure 1

18 pages, 2009 KiB  
Review
Beyond Pathogenic RUNX1 Germline Variants: The Spectrum of Somatic Alterations in RUNX1-Familial Platelet Disorder with Predisposition to Hematologic Malignancies
by Alisa Förster, Melanie Decker, Brigitte Schlegelberger and Tim Ripperger
Cancers 2022, 14(14), 3431; https://doi.org/10.3390/cancers14143431 - 14 Jul 2022
Cited by 10 | Viewed by 3548
Abstract
Pathogenic loss-of-function RUNX1 germline variants cause autosomal dominantly-inherited familial platelet disorder with predisposition to hematologic malignancies (RUNX1-FPD). RUNX1-FPD is characterized by incomplete penetrance and a broad spectrum of clinical phenotypes, even within affected families. Heterozygous RUNX1 germline variants set the basis for leukemogenesis, [...] Read more.
Pathogenic loss-of-function RUNX1 germline variants cause autosomal dominantly-inherited familial platelet disorder with predisposition to hematologic malignancies (RUNX1-FPD). RUNX1-FPD is characterized by incomplete penetrance and a broad spectrum of clinical phenotypes, even within affected families. Heterozygous RUNX1 germline variants set the basis for leukemogenesis, but, on their own, they are not transformation-sufficient. Somatically acquired secondary events targeting RUNX1 and/or other hematologic malignancy-associated genes finally lead to MDS, AML, and rarely other hematologic malignancies including lymphoid diseases. The acquisition of different somatic variants is a possible explanation for the variable penetrance and clinical heterogeneity seen in RUNX1-FPD. However, individual effects of secondary variants are not yet fully understood. Here, we review 91 cases of RUNX1-FPD patients who predominantly harbor somatic variants in genes such as RUNX1, TET2, ASXL1, BCOR, PHF6, SRSF2, NRAS, and DNMT3A. These cases illustrate the importance of secondary events in the development and progression of RUNX1-FPD-associated hematologic malignancies. The leukemia-driving interplay of predisposing germline variants and acquired variants remain to be elucidated to better understand clonal evolution and malignant transformation and finally allow risk-adapted surveillance and targeted therapeutic measures to prevent leukemia. Full article
(This article belongs to the Special Issue Genomics of Rare Hematologic Cancers)
Show Figures

Figure 1

16 pages, 798 KiB  
Article
The Cytogenetic Profile of Primary and Secondary Plasma Cell Leukemia: Etiopathogenetic Perspectives, Prognostic Impact and Clinical Relevance to Newly Diagnosed Multiple Myeloma with Differential Circulating Clonal Plasma Cells
by Stefanos I. Papadhimitriou, Evangelos Terpos, Konstantinos Liapis, Dimitrios Pavlidis, Theodoros Marinakis, Efstathios Kastritis, Meletios-Athanasios Dimopoulos, Ourania E. Tsitsilonis and Ioannis V. Kostopoulos
Biomedicines 2022, 10(2), 209; https://doi.org/10.3390/biomedicines10020209 - 19 Jan 2022
Cited by 18 | Viewed by 3152
Abstract
Plasma cell leukemia (PCL) is a rare and aggressive plasma cell dyscrasia that may appear as de-novo leukemia (pPCL) or on the basis of a pre-existing multiple myeloma (MM), called secondary plasma cell leukemia (sPCL). In this prospective study, we have applied a [...] Read more.
Plasma cell leukemia (PCL) is a rare and aggressive plasma cell dyscrasia that may appear as de-novo leukemia (pPCL) or on the basis of a pre-existing multiple myeloma (MM), called secondary plasma cell leukemia (sPCL). In this prospective study, we have applied a broad panel of FISH probes in 965 newly diagnosed MM (NDMM) and 44 PCL cases of both types to reveal the particular cytogenetic differences among the three plasma cell dyscrasias. In order to evaluate the frequency and patterns of clonal evolution, the same FISH panel was applied both at diagnosis and at the time of first relapse for 81 relapsed MM patients and both at MM diagnosis and during sPCL transformation for the 19 sPCL cases described here. pPCL was characterized by frequent MYC translocations and t(11;14) with a 11q13 breakpoint centered on the MYEOV gene, not commonly seen in MM. sPCL had a higher number of FISH abnormalities and was strongly associated with the presence of del(17p13), either acquired at the initial MM stage or as a newly acquired lesion upon leukemogenesis in the context of the apparent clonal evolution observed in sPCL. In clinical terms, sPCL showed a shorter overall survival than pPCL with either standard or high-risk (t(4;14) and/or t(14;16) and/or del(17p13) and/or ≥3 concomitant aberrations) abnormalities (median 5 months vs. 21 and 11 months respectively, p < 0.001), suggesting a prognostic stratification based on cytogenetic background. These observations proved relevant in the NDMM setting, where higher levels of circulating plasma cells (CPCs) were strongly associated with high-risk cytogenetics (median frequency of CPCs: 0.11% of peripheral blood nucleated cells for high-risk vs. 0.007% for standard-risk NDMM, p < 0.0001). Most importantly, the combined evaluation of CPCs (higher or lower than a cut-off of 0.03%), together with patients’ cytogenetic status, could be used for an improved prognostic stratification of NDMM patients. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

14 pages, 1889 KiB  
Article
Telomerase (hTERT) Overexpression Reveals a Promising Prognostic Biomarker and Therapeutical Target in Different Clinical Subtypes of Pediatric Acute Lymphoblastic Leukaemia
by Beatriz Maria Dias Nogueira, Laudreísa da Costa Pantoja, Emerson Lucena da Silva, Fernando Augusto Rodrigues Mello Júnior, Eliel Barbosa Teixeira, Alayde Vieira Wanderley, Jersey Heitor da Silva Maués, Manoel Odorico de Moraes Filho, Maria Elisabete Amaral de Moraes, Raquel Carvalho Montenegro, André Salim Khayat and Caroline Aquino Moreira-Nunes
Genes 2021, 12(10), 1632; https://doi.org/10.3390/genes12101632 - 17 Oct 2021
Cited by 6 | Viewed by 2647
Abstract
Acute Lymphoblastic Leukemia (ALL) is a neoplasm of the hematopoietic system defined as a clonal expansion of an abnormal lymphoid precursor cell. It mostly affects children under five years of age and is the most common tumor to afflict pediatric patients. The expression [...] Read more.
Acute Lymphoblastic Leukemia (ALL) is a neoplasm of the hematopoietic system defined as a clonal expansion of an abnormal lymphoid precursor cell. It mostly affects children under five years of age and is the most common tumor to afflict pediatric patients. The expression of the human telomerase gene (hTERT) in patients with ALL has been studied as a biomarker and could become a new therapeutic target. We evaluate the role of hTERT gene expression in ALL pediatric patients, through quantitative real-time PCR technique, and the possible correlation between hTERT expression and clinical variables: gender, age, white blood cells (WBC), gene fusions, and immunophenotyping. The analysis between healthy controls and ALL patients (N = 244) was statistically significant (p < 0.001), demonstrating hTERT overexpression in these patients. In comparison with the usual set of clinical variables, the data were not statistically significant (p > 0.05), indicating that hTERT is equally overexpressed among patients regardless of gender, age, gene fusions, and immunophenotyping. Moreover, patients who presented a higher hTERT expression level had a significant (p < 0.0001) lower overall survival rate. In summary, hTERT expression emerges as an important molecular pathway in leukemogenesis regardless patient’s clinical variables, thus, the data here presented pointed it as a valuable biomarker in pediatric acute lymphoblastic leukemia and a promising target for new therapeutic and prognostic measures. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

41 pages, 3681 KiB  
Review
Transcription Factors, R-Loops and Deubiquitinating Enzymes: Emerging Targets in Myelodysplastic Syndromes and Acute Myeloid Leukemia
by Silvia M. L. Barabino, Elisabetta Citterio and Antonella Ellena Ronchi
Cancers 2021, 13(15), 3753; https://doi.org/10.3390/cancers13153753 - 26 Jul 2021
Cited by 3 | Viewed by 6541
Abstract
Myeloid neoplasms encompass a very heterogeneous family of diseases characterized by the failure of the molecular mechanisms that ensure a balanced equilibrium between hematopoietic stem cells (HSCs) self-renewal and the proper production of differentiated cells. The origin of the driver mutations leading to [...] Read more.
Myeloid neoplasms encompass a very heterogeneous family of diseases characterized by the failure of the molecular mechanisms that ensure a balanced equilibrium between hematopoietic stem cells (HSCs) self-renewal and the proper production of differentiated cells. The origin of the driver mutations leading to preleukemia can be traced back to HSC/progenitor cells. Many properties typical to normal HSCs are exploited by leukemic stem cells (LSCs) to their advantage, leading to the emergence of a clonal population that can eventually progress to leukemia with variable latency and evolution. In fact, different subclones might in turn develop from the original malignant clone through accumulation of additional mutations, increasing their competitive fitness. This process ultimately leads to a complex cancer architecture where a mosaic of cellular clones—each carrying a unique set of mutations—coexists. The repertoire of genes whose mutations contribute to the progression toward leukemogenesis is broad. It encompasses genes involved in different cellular processes, including transcriptional regulation, epigenetics (DNA and histones modifications), DNA damage signaling and repair, chromosome segregation and replication (cohesin complex), RNA splicing, and signal transduction. Among these many players, transcription factors, RNA splicing proteins, and deubiquitinating enzymes are emerging as potential targets for therapeutic intervention. Full article
(This article belongs to the Special Issue Blood Stem Cells in Pre-leukaemia)
Show Figures

Figure 1

19 pages, 29101 KiB  
Article
Development of BCR-ABL1 Transgenic Zebrafish Model Reproducing Chronic Myeloid Leukemia (CML) Like-Disease and Providing a New Insight into CML Mechanisms
by Daniela Zizioli, Simona Bernardi, Marco Varinelli, Mirko Farina, Luca Mignani, Katia Bosio, Dario Finazzi, Eugenio Monti, Nicola Polverelli, Michele Malagola, Elisa Borsani, Giuseppe Borsani and Domenico Russo
Cells 2021, 10(2), 445; https://doi.org/10.3390/cells10020445 - 19 Feb 2021
Cited by 8 | Viewed by 4348
Abstract
Zebrafish has proven to be a versatile and reliable experimental in vivo tool to study human hematopoiesis and model hematological malignancies. Transgenic technologies enable the generation of specific leukemia types by the expression of human oncogenes under specific promoters. Using this technology, a [...] Read more.
Zebrafish has proven to be a versatile and reliable experimental in vivo tool to study human hematopoiesis and model hematological malignancies. Transgenic technologies enable the generation of specific leukemia types by the expression of human oncogenes under specific promoters. Using this technology, a variety of myeloid and lymphoid malignancies zebrafish models have been described. Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia characterized by the BCR-ABL1 fusion gene, derived from the t (9;22) translocation causing the Philadelphia Chromosome (Ph). The BCR-ABL1 protein is a constitutively activated tyrosine kinas inducing the leukemogenesis and resulting in an accumulation of immature leukemic cells into bone marrow and peripheral blood. To model Ph+ CML, a transgenic zebrafish line expressing the human BCR-ABL1 was generated by the Gal4/UAS system, and then crossed with the hsp70-Gal4 transgenic line. The new line named (BCR-ABL1pUAS:CFP/hsp70-Gal4), presented altered expression of hematopoietic markers during embryonic development compared to controls and transgenic larvae showed proliferating hematopoietic cells in the caudal hematopoietic tissue (CHT). The present transgenic zebrafish would be a robust CML model and a high-throughput drug screening tool. Full article
(This article belongs to the Special Issue Signaling Pathway Analysis and Disease Modeling in Zebrafish)
Show Figures

Figure 1

Back to TopTop