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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (468)

Search Parameters:
Keywords = IDH mutation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
10 pages, 3826 KiB  
Communication
Circulating Tumour DNA Is a Biomarker of Response in Angioimmunoblastic T-Cell Lymphoma
by Costas Kleanthes Yannakou, Simon Wu, Karthik Rajah, Chathuri Abeyakoon, Caitlyn Nguyen-Ngo, Yan Zhuang Yap, James Sheldon, Piers Blombery and Henry Miles Prince
Int. J. Mol. Sci. 2025, 26(14), 6719; https://doi.org/10.3390/ijms26146719 - 13 Jul 2025
Viewed by 179
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a rare and aggressive subtype of non-Hodgkin lymphoma, the monitoring of which is largely restricted to radiological methods. Diagnosis relies on identifying characteristic clinicopathological features, supported by the detection of recurrent somatic mutations in RHOA, TET2, [...] Read more.
Angioimmunoblastic T-cell lymphoma (AITL) is a rare and aggressive subtype of non-Hodgkin lymphoma, the monitoring of which is largely restricted to radiological methods. Diagnosis relies on identifying characteristic clinicopathological features, supported by the detection of recurrent somatic mutations in RHOA, TET2, IDH2 and DNMT3A. The characteristic molecular profile of AITL and the high levels of circulating tumour DNA (ctDNA) measurable in AITL before treatment makes this an attractive lymphoma subtype in which to further investigate the role of ctDNA monitoring. The detection of somatic mutations in pre-treatment AITL-containing tissue samples was compared to those detected in pre-treatment ctDNA samples in a cohort of 12 patients. Changes in ctDNA somatic mutation burden over time were then correlated with radiological response. All six paired pre-treatment ctDNA and tissue samples had variants in common. All (8/8) previously ctDNA-detectable IDH2 and RHOA variants were undetectable in ctDNA samples at the time of end-of-treatment complete metabolic response (CMR). In comparison, the majority of both previously ctDNA-detectable DNMT3A variants (3/4) and TET2 variants (6/11) were detectable in ctDNA samples at the time of end-of-treatment CMR. These observations suggest that IDH2/RHOA variants may be more reliable markers of measurable residual disease in AITL than DNMT3A/TET2 variants. Full article
(This article belongs to the Special Issue Leukemia and Lymphoma: A Focus on Molecular Genetics Research)
Show Figures

Figure 1

19 pages, 1510 KiB  
Review
Updated Insights into the Molecular Pathophysiology of Olfactory Neuroblastoma Using Multi-Omics Analysis
by Enes Demir, Deondra Montgomery, Varun Naravetla and Michael Karsy
J. Pers. Med. 2025, 15(7), 309; https://doi.org/10.3390/jpm15070309 - 13 Jul 2025
Viewed by 189
Abstract
Background/Objectives: Olfactory neuroblastoma (ONB), also known as esthesioneuroblastoma, is a rare neuroectodermal malignancy of the nasal cavity characterized by aggressive local invasion and variable metastatic potential, with diverse clinical behavior, often presenting at advanced stages. ONB poses challenges for targeted therapeutic strategies, [...] Read more.
Background/Objectives: Olfactory neuroblastoma (ONB), also known as esthesioneuroblastoma, is a rare neuroectodermal malignancy of the nasal cavity characterized by aggressive local invasion and variable metastatic potential, with diverse clinical behavior, often presenting at advanced stages. ONB poses challenges for targeted therapeutic strategies, despite advances in surgical and multimodal treatment strategies, because of the rarity of this disease and the limited understanding of its molecular pathophysiology. Methods: A comprehensive review of genomic, multi-omic, and molecular studies was performed to integrate known targeted sites in ONB with the current understanding of its pathophysiology. Results: Recent genetic and molecular studies have identified significant epigenetic and signaling pathway alterations that are critical in pathogenesis and treatment resistance and may serve as potential therapeutic targets. Additionally, novel discovered immunohistochemical and transcriptomic markers, such as IDH2, NEUROD1, and OTX2, offer improved diagnostic specificity and prognostication. Multi-genomic platforms (i.e., multi-omics), involving the combined integration of transcriptomics, epigenetics, and proteomics findings, have led to several recent insights, including the subclassification of neural and basal genomic subtypes, the identification of key driver mutations, and new insights into disease development. This review synthesizes current knowledge on the molecular landscape of ONB, including its tumor origin, immune microenvironment, genetic alterations, and key molecular pathways involved in its pathogenesis. Conclusions: Future research may benefit from integrating these findings into precision medicine approaches, enabling earlier diagnosis and more accurate prognosis. Full article
(This article belongs to the Section Mechanisms of Diseases)
Show Figures

Figure 1

21 pages, 453 KiB  
Review
Precision Medicine in Hematologic Malignancies: Evolving Concepts and Clinical Applications
by Rita Khoury, Chris Raffoul, Christina Khater and Colette Hanna
Biomedicines 2025, 13(7), 1654; https://doi.org/10.3390/biomedicines13071654 - 7 Jul 2025
Viewed by 451
Abstract
Precision medicine is transforming hematologic cancer care by tailoring treatments to individual patient profiles and moving beyond the traditional “one-size-fits-all” model. This review outlines foundational technologies, disease-specific advances, and emerging directions in precision hematology. The field is enabled by molecular profiling techniques, including [...] Read more.
Precision medicine is transforming hematologic cancer care by tailoring treatments to individual patient profiles and moving beyond the traditional “one-size-fits-all” model. This review outlines foundational technologies, disease-specific advances, and emerging directions in precision hematology. The field is enabled by molecular profiling techniques, including next-generation sequencing (NGS), whole-exome sequencing (WES), and RNA sequencing (RNA-seq), as well as epigenomic and proteomic analyses. Complementary tools such as liquid biopsy and minimal residual disease (MRD) monitoring have improved diagnosis, risk stratification, and therapeutic decision making. We discuss major molecular targets and personalized strategies across hematologic malignancies: FLT3 and IDH1/2 in acute myeloid leukemia (AML); Philadelphia chromosome–positive and Ph-like subtypes in acute lymphoblastic leukemia (ALL); BCR-ABL1 in chronic myeloid leukemia (CML); TP53 and IGHV mutations in chronic lymphocytic leukemia (CLL); molecular subtypes and immune targets in diffuse large B-cell lymphoma (DLBCL) and other lymphomas; and B-cell maturation antigen (BCMA) in multiple myeloma. Despite significant progress, challenges remain, including high costs, disparities in access, a lack of standardization, and integration barriers in clinical practice. However, advances in single-cell sequencing, spatial transcriptomics, drug repurposing, immunotherapies, pan-cancer trials, precision prevention, and AI-guided algorithms offer promising avenues to refine treatment and improve outcomes. Overcoming these barriers will be critical for ensuring the equitable and widespread implementation of precision medicine in routine hematologic oncology care. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnosis and Treatment of Hematologic Malignancies)
Show Figures

Figure 1

26 pages, 2124 KiB  
Article
Integrating Boruta, LASSO, and SHAP for Clinically Interpretable Glioma Classification Using Machine Learning
by Mohammad Najeh Samara and Kimberly D. Harry
BioMedInformatics 2025, 5(3), 34; https://doi.org/10.3390/biomedinformatics5030034 - 30 Jun 2025
Viewed by 547
Abstract
Background: Gliomas represent the most prevalent and aggressive primary brain tumors, requiring precise classification to guide treatment strategies and improve patient outcomes. Purpose: This study aimed to develop and evaluate a machine learning-driven approach for glioma classification by identifying the most relevant genetic [...] Read more.
Background: Gliomas represent the most prevalent and aggressive primary brain tumors, requiring precise classification to guide treatment strategies and improve patient outcomes. Purpose: This study aimed to develop and evaluate a machine learning-driven approach for glioma classification by identifying the most relevant genetic and clinical biomarkers while demonstrating clinical utility. Methods: A dataset from The Cancer Genome Atlas (TCGA) containing 23 features was analyzed using an integrative approach combining Boruta, Least Absolute Shrinkage and Selection Operator (LASSO), and SHapley Additive exPlanations (SHAP) for feature selection. The refined feature set was used to train four machine learning models: Random Forest, Support Vector Machine, XGBoost, and Logistic Regression. Comprehensive evaluation included class distribution analysis, calibration assessment, and decision curve analysis. Results: The feature selection approach identified 13 key predictors, including IDH1, TP53, ATRX, PTEN, NF1, EGFR, NOTCH1, PIK3R1, MUC16, CIC mutations, along with Age at Diagnosis and race. XGBoost achieved the highest AUC (0.93), while Logistic Regression recorded the highest testing accuracy (88.09%). Class distribution analysis revealed excellent GBM detection (Average Precision 0.840–0.880) with minimal false negatives (5–7 cases). Calibration analysis demonstrated reliable probability estimates (Brier scores 0.103–0.124), and decision curve analysis confirmed substantial clinical utility with net benefit values of 0.36–0.39 across clinically relevant thresholds. Conclusions: The integration of feature selection techniques with machine learning models enhances diagnostic precision, interpretability, and clinical utility in glioma classification, providing a clinically ready framework that bridges computational predictions with evidence-based medical decision-making. Full article
Show Figures

Figure 1

20 pages, 3490 KiB  
Article
Isocitrate Dehydrogenase-Wildtype Glioma Adapts Toward Mutant Phenotypes and Enhanced Therapy Sensitivity Under D-2-Hydroxyglutarate Exposure
by Geraldine Rocha, Clara Francés-Gómez, Javier Megías, Lisandra Muñoz-Hidalgo, Pilar Casanova, Jose F. Haro-Estevez, Vicent Teruel-Martí, Daniel Monleón and Teresa San-Miguel
Biomedicines 2025, 13(7), 1584; https://doi.org/10.3390/biomedicines13071584 - 28 Jun 2025
Viewed by 432
Abstract
Background/Objectives: Isocitrate dehydrogenase (IDH) mutations are hallmark features in subsets of gliomas, producing the oncometabolite D-2-hydroxyglutarate (2HG). Although IDH mutations are associated with better clinical outcomes, their relationship with tumor progression is complex. This study aimed to investigate, in vitro [...] Read more.
Background/Objectives: Isocitrate dehydrogenase (IDH) mutations are hallmark features in subsets of gliomas, producing the oncometabolite D-2-hydroxyglutarate (2HG). Although IDH mutations are associated with better clinical outcomes, their relationship with tumor progression is complex. This study aimed to investigate, in vitro and in vivo, the phenotypic consequences of IDH mutation and 2HG exposure in glioblastoma (GBM) under normoxic and hypoxic conditions and under temozolomide (TMZ) and radiation exposure. Methods: Experiments were conducted using IDH-wildtype (IDH-wt) and IDH-mutant (IDH-mut) glioma cell lines under controlled oxygen conditions. Functional assays included cell viability, cell cycle analysis, apoptosis profiling, migration, and surface marker expression via flow cytometry. Orthotopic xenografts were established in immunocompromised mice to assess in vivo tumor growth and morphology, followed by MRI and histological analysis. Treatments included TMZ, radiation, and 2HG at varying concentrations. Statistical analyses were performed using SPSS and RStudio. Results:IDH-wt cells exhibited faster proliferation and greater adaptability under hypoxia, while IDH-mut cells showed cell cycle arrest and limited growth. 2HG recapitulated IDH-mut features in IDH-wt cells, including increased apoptosis under TMZ, reduced proliferation, and altered CD24/CD44 expression. In vivo, IDH-wt tumors were larger and more infiltrative, while 2HG administration reduced tumor volume and promoted compact morphology. Notably, migration was initially similar across genotypes but increased in IDH-mut and 2HG-treated IDH-wt cells over time, though suppressed under therapeutic stress. Conclusions: IDH mutation and 2HG modulate glioma cell biology, including cell cycle dynamics, proliferation rates, migration, and apoptosis. While the IDH mutation and its metabolic product confer initial growth advantages, they enhance treatment sensitivity and reduce invasiveness, highlighting potential vulnerabilities for targeted therapy. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapy of Gliomas)
Show Figures

Figure 1

25 pages, 1441 KiB  
Review
From Tumor to Network: Functional Connectome Heterogeneity and Alterations in Brain Tumors—A Multimodal Neuroimaging Narrative Review
by Pablo S. Martínez Lozada, Johanna Pozo Neira and Jose E. Leon-Rojas
Cancers 2025, 17(13), 2174; https://doi.org/10.3390/cancers17132174 - 27 Jun 2025
Viewed by 289
Abstract
Intracranial tumors such as gliomas, meningiomas, and brain metastases induce complex alterations in brain function beyond their focal presence. Modern connectomic and neuroimaging approaches, including resting-state functional MRI (rs-fMRI) and diffusion MRI, have revealed that these tumors disrupt and reorganize large-scale brain networks [...] Read more.
Intracranial tumors such as gliomas, meningiomas, and brain metastases induce complex alterations in brain function beyond their focal presence. Modern connectomic and neuroimaging approaches, including resting-state functional MRI (rs-fMRI) and diffusion MRI, have revealed that these tumors disrupt and reorganize large-scale brain networks in heterogeneous ways. In adult patients, diffuse gliomas infiltrate neural circuits, causing both local disconnections and widespread functional changes that often extend into structurally intact regions. Meningiomas and metastases, though typically well-circumscribed, can perturb networks via mass effect, edema, and diaschisis, sometimes provoking global “dysconnectivity” related to cognitive deficits. Therefore, this review synthesizes interdisciplinary evidence from neuroscience, oncology, and neuroimaging on how intracranial tumors disrupt functional brain connectivity pre- and post-surgery. We discuss how functional heterogeneity (i.e., differences in network involvement due to tumor type, location, and histo-molecular profile) manifests in connectomic analyses, from altered default mode and salience network activity to changes in structural–functional coupling. The clinical relevance of these network effects is examined, highlighting implications for pre-surgical planning, prognostication of neurocognitive outcomes, and post-operative recovery. Gliomas demonstrate remarkable functional plasticity, with network remodeling that may correlate with tumor genotype (e.g., IDH mutation), while meningioma-related edema and metastasis location modulate the extent of network disturbance. Finally, we explore future directions, including imaging-guided therapies and “network-aware” neurosurgical strategies that aim to preserve and restore brain connectivity. Understanding functional heterogeneity in brain tumors through a connectomic lens not only provides insights into the neuroscience of cancer but also informs more effective, personalized approaches to neuro-oncologic care. Full article
Show Figures

Figure 1

14 pages, 1586 KiB  
Article
Genomic Profile and Clinical Outcomes in Acute Myeloid Leukemia with Monosomal Karyotype
by Collins Wangulu, Ehsan Bahrami Hezaveh, Mojgan Zarif, Qianghua Zhou, Winnie Lo, Cuihong Wei, Hassan Sibai and Hong Chang
Int. J. Mol. Sci. 2025, 26(12), 5845; https://doi.org/10.3390/ijms26125845 - 18 Jun 2025
Viewed by 460
Abstract
The biology of Monosomal Karyotype Acute Myeloid Leukemia (MK AML) remains unclear, and its mutational profile has not been exclusively assessed. We sought to determine the genomic profile of MK AML patients and its correlation with overall survival (OS). We conducted a retrospective [...] Read more.
The biology of Monosomal Karyotype Acute Myeloid Leukemia (MK AML) remains unclear, and its mutational profile has not been exclusively assessed. We sought to determine the genomic profile of MK AML patients and its correlation with overall survival (OS). We conducted a retrospective study involving 664 AML patients, identifying 156 (23.5%) with MK AML. The most common monosomies were -17 (41%) and -7 (37%), with 149 (95%) and 138 (88%) having myelodysplasia-related (MR) cytogenetics and complex karyotype (CK), respectively. Frequent mutations included TP53 (69%), DNMT3A (19%), TET2 (13%), and IDH1 (7%). Patients with MK AML with TP53 mutation (TP53 Mut) had shorter OS compared to those with TP53 wild-type (WT) (median OS, 3.9 versus 9.2 months, p = 0.002). Our validation study further supports this finding. There was no significant difference in OS related to the presence or absence of CK (p = 0.252), MR mutations (p = 0.252), DNMT3A (p = 0.264), TET2 (p = 0.264), and IDH1 (p = 0.183) alterations. Co-mutation with novel EPI6 and TAZI signature alterations did not significantly impact OS among MK AML TP53 Mut patients, suggesting that TP53 Mut remains the dominant driver of outcome in this subgroup. In conclusion, MK AML is a genotypically diverse and high-risk group, with MK AML TP53 Mut indicating worse prognosis. Full article
(This article belongs to the Special Issue Leukemia and Lymphoma: A Focus on Molecular Genetics Research)
Show Figures

Figure 1

12 pages, 1031 KiB  
Article
IDH1 Mutation Impacts DNA Repair Through ALKBH2 Rendering Glioblastoma Cells Sensitive to Artesunate
by Olivier Switzeny, Stefan Pusch, Markus Christmann and Bernd Kaina
Biomedicines 2025, 13(6), 1479; https://doi.org/10.3390/biomedicines13061479 - 16 Jun 2025
Viewed by 623
Abstract
Background: Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are enzymes that catalyze the oxidative decarboxylation of isocitrate to alpha-ketoglutarate (α-KG), which is essential for many metabolic processes, including some steps in DNA repair. In tumors, notably in gliomas, IDH1 and IDH2 [...] Read more.
Background: Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are enzymes that catalyze the oxidative decarboxylation of isocitrate to alpha-ketoglutarate (α-KG), which is essential for many metabolic processes, including some steps in DNA repair. In tumors, notably in gliomas, IDH1 and IDH2 are frequently mutated. The mutation found in different cancers is functionally active, causing, instead of α-KG, the formation of 2-hydroxyglutarate (2-HG), which inhibits α-KG-dependent enzymes. Gliomas harboring mutated IDH1/2 show a better prognosis than IDH1 wild-type (wt) tumors of the same grade, which might result from the inhibition of DNA repair functions. A DNA repair enzyme dependent on α-KG is alkB homolog 2 (ALKBH2), which removes several lesions from DNA. These findings prompted us to investigate the response of glioma cells to artesunate (ART), a plant ingredient with genotoxic and anticancer activity currently used in several trials. Materials and Methods: We used isogenic glioblastoma cell lines that express IDH1 wild-type or, based on a TET-inducible system, the IDH1 mutant (mt) protein, and treated them with increasing doses of artesunate. We also treated glioblastoma cells with 2-HG, generated ALKBH2 knockout cells, and checked their sensitivity to the cytotoxic effects of artesunate. Results: We show that the cell-killing effect of ART is enhanced if the IDH1 mutant (R132H) is expressed in glioblastoma cells. Further, we show that 2-HG imitates the effect of IDH1mt as 2-HG ameliorates the cytotoxicity of ART. Finally, we demonstrate that the knockout of ALKBH2 causes the sensitization of glioblastoma cells to ART. Conclusions: The data indicate that ALKBH2 protects against the anticancer effect of ART, and the mutation of IDH1/2 commonly occurring in low-grade gliomas sensitizes to ART via an ALKBH2-dependent mechanism. The data support the use of ART in the therapy of IDH1/2-mutated cancers both in combination with chemotherapy and adjuvant treatment. Full article
(This article belongs to the Special Issue Glioma Therapy: Current Status and Future Prospects)
Show Figures

Figure 1

12 pages, 1910 KiB  
Article
Diagnostic Utility of Intratumoral Susceptibility Signals in Adult Diffuse Gliomas: Tumor Grade Prediction and Correlation with Molecular Markers Within the WHO CNS5 (2021) Classification
by José Ignacio Tudela Martínez, Victoria Vázquez Sáez, Guillermo Carbonell, Héctor Rodrigo Lara, Florentina Guzmán-Aroca and Juan de Dios Berna Mestre
J. Clin. Med. 2025, 14(11), 4004; https://doi.org/10.3390/jcm14114004 - 5 Jun 2025
Viewed by 592
Abstract
Background/Objectives: This study evaluates intratumoral susceptibility signals (ITSS) as imaging markers for glioma grade prediction and their association with molecular and histopathologic features, in the context of the fifth edition of the World Health Organization Classification of Tumors of the Central Nervous [...] Read more.
Background/Objectives: This study evaluates intratumoral susceptibility signals (ITSS) as imaging markers for glioma grade prediction and their association with molecular and histopathologic features, in the context of the fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System (WHO CNS5). Methods: We retrospectively analyzed patients with adult diffuse gliomas who underwent pretreatment magnetic resonance imaging. ITSS were semiquantitatively graded by two radiologists: grade 0 (no signal), grade 1 (1–5), grade 2 (6–10), and grade 3 (≥11). Relative cerebral blood volume (rCBV) and tumor volume were also obtained. Histopathologic features included tumor grade, Ki-67, mitotic count, necrosis, microvascular proliferation, and molecular alterations (isocitrate dehydrogenase [IDH], 1p/19q, cyclin-dependent kinase inhibitors 2A and 2B [CDKN2A/B], and p53). Regression models predicted tumor grade (low: 1–2, high: 3–4) using ITSS, tumor volume, and rCBV. ROC curves and diagnostic performance metrics were analyzed. Results: 99 patients were included. ITSS grading correlated with rCBV, tumor volume, mitotic count, Ki-67, and tumor grade (p < 0.001). ITSS grades 0–1 were associated with oligodendrogliomas and astrocytomas (p < 0.001), IDH mutations (p < 0.001), and 1p/19q co-deletions (p = 0.01). ITSS grades 2–3 were linked to glioblastomas (p < 0.001), necrosis (p < 0.001), microvascular proliferation (p < 0.001), and CDKN2A/B homozygous deletions (p = 0.02). Models combining ITSS with rCBV and volume showed AUC of 0.94 and 0.96 (p < 0.001), outperforming univariate models. Conclusions: Semiquantitative ITSS grading correlates with key histopathologic and molecular glioma features. Combined with perfusion and volumetric parameters, ITSS enhance non-invasive glioma grading, in alignment with WHO CNS5. Full article
(This article belongs to the Section Nuclear Medicine & Radiology)
Show Figures

Graphical abstract

19 pages, 2933 KiB  
Article
Role of Amide Proton Transfer Weighted MRI in Predicting MGMTp Methylation Status, p53-Status, Ki-67 Index, IDH-Status, and ATRX Expression in WHO Grade 4 High Grade Glioma
by Faris Durmo, Jimmy Lätt, Anna Rydelius, Elisabet Englund, Tim Salomonsson, Patrick Liebig, Johan Bengzon, Peter C. M. van Zijl, Linda Knutsson and Pia C. Sundgren
Tomography 2025, 11(6), 64; https://doi.org/10.3390/tomography11060064 - 31 May 2025
Viewed by 502
Abstract
Objectives: To assess amide proton transfer weighted (APTw) MR imaging capabilities in differentiating high-grade glial tumors across alpha-thalassemia/mental retardation X-linked (ATRX) expression, tumor-suppressor protein p53 expression (p53), O6-methylguanine-DNA methyltransferase promoter (MGMTp) methylation, isocitrate dehydrogenase (IDH) status, and proliferation marker Ki-67 (Ki-67 index) as [...] Read more.
Objectives: To assess amide proton transfer weighted (APTw) MR imaging capabilities in differentiating high-grade glial tumors across alpha-thalassemia/mental retardation X-linked (ATRX) expression, tumor-suppressor protein p53 expression (p53), O6-methylguanine-DNA methyltransferase promoter (MGMTp) methylation, isocitrate dehydrogenase (IDH) status, and proliferation marker Ki-67 (Ki-67 index) as a preoperative diagnostic aid. Material & Methods: A total of 42 high-grade glioma WHO grade 4 (HGG) patients were evaluated prospectively (30 males and 12 females). All patients were examined using conventional MRI, including the following: T1w-MPRAGE pre- and post-contrast administration, conventional T2w and 3D FLAIR, and APTw imaging with a 3T MR scanner. Receiver operating characteristic (ROC) curves were calculated for the APTw% mean, median, and max signal for the different molecular biomarkers. A logistic regression model was constructed for combined mean and median APTw% signals for p53 expression. Results: The whole-tumor max APTw% signal could significantly differentiate MGMTp from non-MGMTp HGG, p = 0.035. A cutoff of 4.28% max APTw% signal yielded AUC (area under the curve) = 0.702, with 70.6% sensitivity and 66.7% specificity. The mean/median APTw% signals differed significantly in p53 normal versus p53-overexpressed HGG s: 1.81%/1.83% vs. 1.15%/1.18%, p = 0.002/0.006, respectively. Cutoffs of 1.25%/1.33% for the mean/median APTw% signals yielded AUCs of 0.786/0.757, sensitivities of 76.9%/76.9%, and specificities of 50%/66.2%, p = 0.002/0.006, respectively. A logistic regression model with a combined mean and median APTw% signal for p53 status yielded an AUC = 0.788 and 76.9% sensitivity and 66.2% specificity. ATRX-, IDH- wild type (wt) vs. mutation (mut), and the level of Ki-67 did not differ significantly, but trends were found: IDH-wt and low Ki-67 showed higher mean/median/max APTw% signals vs. IDH-mut and high Ki-67, respectively. ATRX-wt vs. mutation showed higher mean and median APTw% signals but lower max APTw% signal. Conclusions: APTw imaging can potentially be a useful marker for the stratification of p53 expression and MGMT status in high-grade glioma in the preoperative setting and potentially aid surgical decision-making. Full article
Show Figures

Figure 1

25 pages, 6990 KiB  
Review
Non-Coding RNAs in Diagnostic Pathology of High-Grade Central Osteosarcoma
by Albert Roessner, Sabine Franke, Julian Schreier, Sarah R. Ullmann and Franziska S. Karras
Diagnostics 2025, 15(11), 1355; https://doi.org/10.3390/diagnostics15111355 - 28 May 2025
Viewed by 381
Abstract
A histological evaluation remains the cornerstone of diagnosing highly malignant osteosarcoma, having demonstrated its efficacy and reliability over several decades. However, despite these advancements, misdiagnoses with severe consequences, including inadequate surgical procedures, continue to occur. Consequently, there is a pressing need to further [...] Read more.
A histological evaluation remains the cornerstone of diagnosing highly malignant osteosarcoma, having demonstrated its efficacy and reliability over several decades. However, despite these advancements, misdiagnoses with severe consequences, including inadequate surgical procedures, continue to occur. Consequently, there is a pressing need to further enhance diagnostic security. Adjunct immunohistochemical approaches have demonstrated significant effectiveness in regard to cancer diagnostics, generally. However, their utility for identifying highly malignant osteosarcoma is limited. Molecular genetic findings have significantly improved the diagnosis of Ewing’s sarcoma by identifying specific translocations and have been used to detect specific IDH gene mutations in chondrosarcoma. Nevertheless, molecular genetic alterations in highly malignant osteosarcoma exhibit a high degree of complexity, thereby limiting their diagnostic utility. Given that only 1–2% of the human genome comprises protein-coding sequences, the growing number of non-coding regulatory RNAs, which are increasingly being elucidated, has garnered substantial attention in the field of clinical cancer diagnostics. Over the past several years, patterns of altered non-coding RNA expression have been identified that facilitate the distinction between benign and malignant tumors in various organs. In the field of bone tumors, the experience of this approach has been limited thus far. The divergent expression of microRNAs has demonstrated utility for differentiating osteosarcoma from osteoblastoma and discriminating between osteosarcoma and giant-cell tumors of bone and fibrous dysplasia. However, the application of non-coding RNA expression patterns for the differential diagnosis of osteosarcoma is still in its preliminary stages. This review provides an overview of the current status of non-coding RNAs in osteosarcoma diagnostics, in conjunction with a histological evaluation. The potential of this approach is discussed comprehensively. Full article
(This article belongs to the Special Issue Bone Tumours: From Molecular Pathology to Clinical Practice)
Show Figures

Figure 1

11 pages, 1542 KiB  
Article
Feasible and Rapid Screening of IDH1/2 and FLT3-TKD2 Mutations by High-Resolution Melting for Patients with Acute Myeloid Leukemia
by José Vicente Gil, Sandra de las Heras, Alberto Miralles, Claudia Sargas, Marta Llop, Rebeca Rodríguez-Veiga, Laura Torres-Miñana, Blanca Boluda, Isabel Cano-Ferri, Evelyn Acuña-Cruz, Irene Navarro, Pilar Lloret-Madrid, Pau Montesinos and Eva Barragán
Diagnostics 2025, 15(10), 1230; https://doi.org/10.3390/diagnostics15101230 - 14 May 2025
Viewed by 484
Abstract
Background: In recent years, numerous recurrently mutated genes have been identified in acute myeloid leukemia (AML), some of which, such as FLT3 and IDH1/2, serve as therapeutic targets, offering new treatment options. Rapid mutational analysis is crucial for timely and optimal [...] Read more.
Background: In recent years, numerous recurrently mutated genes have been identified in acute myeloid leukemia (AML), some of which, such as FLT3 and IDH1/2, serve as therapeutic targets, offering new treatment options. Rapid mutational analysis is crucial for timely and optimal therapy selection. This study aims to develop and validate a rapid, cost-effective, and sensitive screening method for detecting IDH1, IDH2, and FLT3-TKD2 mutations using polymerase chain reaction (PCR) and high-resolution melting curve analysis (HRM). Methods: A PCR-HRM assay was developed to simultaneously detect mutations in IDH1, IDH2, and FLT3-TKD2. The method was applied to a cohort of 1363 AML patients, and its performance, including turnaround time, was evaluated through comparison with next-generation sequencing (NGS) results. Results: The PCR-HRM method demonstrated a positive percent agreement of 98%, 98%, and 92% for IDH1, IDH2, and FLT3-TKD2, respectively, and a negative percent agreement of 100% for all three genes compared to NGS. No false positives were observed, and false negatives were detected in less than 1% of cases, mostly in FLT3-TKD2, all occurring below the established limit of detection. The turnaround time and cost of PCR-HRM were significantly lower than those of NGS. Conclusions: This method offers a highly sensitive, specific, and time-efficient approach for the simultaneous detection of IDH1, IDH2, and FLT3-TKD2 mutations in AML patients. Its rapid turnaround time and cost-effectiveness make it a valuable tool for routine clinical screening, facilitating timely and targeted treatment decisions. Full article
(This article belongs to the Special Issue Diagnosis, Prognosis and Management of Hematologic Malignancies)
Show Figures

Figure 1

20 pages, 1818 KiB  
Review
Oligodendroglioma: Advances in Molecular Mechanisms and Immunotherapeutic Strategies
by Yongxin Zhao, Yan Yu, Weizhi Chen, Xiaojun Zhang, Jing Lv and Heping Zhao
Biomedicines 2025, 13(5), 1133; https://doi.org/10.3390/biomedicines13051133 - 7 May 2025
Viewed by 1027
Abstract
Oligodendroglioma is a central nervous system tumor defined by IDH1/2 mutations and 1p/19q co-deletion. Current management involves maximal resection followed by radiotherapy/chemotherapy, yielding a 20-year survival rate of 37% for grade 3 tumors according to the WHO 2021 classification. As these tumors primarily [...] Read more.
Oligodendroglioma is a central nervous system tumor defined by IDH1/2 mutations and 1p/19q co-deletion. Current management involves maximal resection followed by radiotherapy/chemotherapy, yielding a 20-year survival rate of 37% for grade 3 tumors according to the WHO 2021 classification. As these tumors primarily affect young to middle-aged patients, novel therapies are urgently needed to improve outcomes. Immunotherapy has revolutionized tumor treatment by modulating immune responses. However, its application in oligodendrogliomas faces two major hurdles, including the immunosuppressive tumor microenvironment (TME) and the blood–brain barrier’s restrictive properties. This review first examines oligodendroglioma’s molecular alterations to refine diagnosis and guide targeted therapies. Next, we focus on the oligodendroglioma TME to evaluate emerging immunotherapies, including oncolytic viruses, immune checkpoint blockade, chimeric antigen receptor (CAR) T-cell therapy, and cancer vaccines. Finally, we discuss current challenges and future directions to overcome therapeutic limitations and advance treatment strategies. Full article
(This article belongs to the Special Issue Feature Reviews in Tumor Immunology)
Show Figures

Figure 1

24 pages, 1770 KiB  
Review
Unraveling Venetoclax Resistance: Navigating the Future of HMA/Venetoclax-Refractory AML in the Molecular Era
by Theodora Chatzilygeroudi, Theodoros Karantanos and Vasiliki Pappa
Cancers 2025, 17(9), 1586; https://doi.org/10.3390/cancers17091586 - 7 May 2025
Cited by 1 | Viewed by 1908
Abstract
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with [...] Read more.
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with the hypomethylating agent azacitidine (AZA/VEN) has become a standard treatment for this group of AML patients, demonstrating a 65% overall response rate and a median overall survival of 14.7 months, compared to 22% and 8 months with azacitidine monotherapy, respectively. However, resistance and relapses remain common, representing a significant clinical challenge. Recent studies have identified molecular alterations, such as mutations in FLT3-ITD, NRAS/KRAS, TP53, and BAX, as major drivers of resistance. Additionally, other factors, including metabolic changes, anti-apoptotic protein expression, and monocytic or erythroid/megakaryocytic differentiation status, contribute to treatment failure. Clinical trials are exploring strategies to overcome venetoclax resistance, including doublet or triplet therapies targeting IDH and FLT3 mutations; novel epigenetic approaches; menin, XPO1, and MDM2 inhibitors; along with immunotherapies like monoclonal antibodies and antibody–drug conjugates. A deeper understanding of the molecular mechanisms of resistance through single-cell analysis will be crucial for developing future therapeutic strategies. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia in Adults)
Show Figures

Figure 1

45 pages, 15819 KiB  
Review
The Molecular Basis of Pediatric Brain Tumors: A Review with Clinical Implications
by Elias Antoniades, Nikolaos Keffes, Stamatia Vorri, Vassilios Tsitouras, Nikolaos Gkantsinikoudis, Parmenion Tsitsopoulos and John Magras
Cancers 2025, 17(9), 1566; https://doi.org/10.3390/cancers17091566 - 4 May 2025
Viewed by 1671
Abstract
Central nervous system (CNS) tumors are the most common solid malignancy in the pediatric population. These lesions are the result of the aberrant cell signaling step proteins, which normally regulate cell proliferation. Mitogen-activated protein kinase (MAPK) pathways and tyrosine kinase receptors are involved [...] Read more.
Central nervous system (CNS) tumors are the most common solid malignancy in the pediatric population. These lesions are the result of the aberrant cell signaling step proteins, which normally regulate cell proliferation. Mitogen-activated protein kinase (MAPK) pathways and tyrosine kinase receptors are involved in tumorigenesis of low-grade gliomas. High-grade gliomas may carry similar mutations, but loss of epigenetic control is the dominant molecular event; it can occur either due to histone mutations or inappropriate binding or unbinding of DNA on histones. Therefore, despite the absence of genetic alteration in the classic oncogenes or tumor suppressor genes, uncontrolled transcription results in tumorigenesis. Isocitric dehydrogenase (IDH) mutations do not predominate compared to their adult counterpart. Embryonic tumors include medulloblastomas, which bear mutations of transcription-regulating pathways, such as wingless-related integration sites or sonic hedgehog pathways. They may also relate to high expression of Myc family genes. Atypical teratoid rhabdoid tumors harbor alterations of molecules that contribute to ATP hydrolysis of chromatin. Embryonic tumors with multilayered rosettes are associated with microRNA mutations and impaired translation. Ependymomas exhibit great variability. As far as supratentorial lesions are concerned, the major events are mutations either of NFkB or Hippo pathways. Posterior fossa tumors are further divided into two types with different prognoses. Type A group is associated with mutations of DNA damage repair molecules. Lastly, germ cell tumors are a heterogeneous group. Among them, germinomas manifest KIT receptor mutations, a subgroup of the tyrosine kinase receptor family. Full article
(This article belongs to the Special Issue New Advances in the Treatment of Pediatric Solid Tumors)
Show Figures

Figure 1

Back to TopTop