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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (372)

Search Parameters:
Keywords = cancer progenitor cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 2489 KiB  
Article
Cabozantinib, an Anti-Aging Agent, Prevents Bone Loss in Estrogen-Deficient Mice by Suppressing Senescence-Associated Secretory Phenotype Factors
by Jueun Lee, Jiin Oh, Jae-Ryong Kim, Hyunil Ha, Taesoo Kim and Daewon Jeong
Int. J. Mol. Sci. 2025, 26(15), 7123; https://doi.org/10.3390/ijms26157123 - 24 Jul 2025
Viewed by 311
Abstract
Senescent cells secrete pro-inflammatory cytokines, collectively referred to as the senescence-associated secretory phenotype (SASP). Certain pro-inflammatory SASP factors are known to inhibit the differentiation of bone-forming osteoblast while promoting the differentiation of bone-resorbing osteoclasts, thereby causing osteoporosis. In this study, we screened cabozantinib, [...] Read more.
Senescent cells secrete pro-inflammatory cytokines, collectively referred to as the senescence-associated secretory phenotype (SASP). Certain pro-inflammatory SASP factors are known to inhibit the differentiation of bone-forming osteoblast while promoting the differentiation of bone-resorbing osteoclasts, thereby causing osteoporosis. In this study, we screened cabozantinib, a tyrosine kinase inhibitor used to treat medullary thyroid cancer, for its ability to reduce doxorubicin-induced cellular senescence in both osteoblast and osteoclast progenitors. This non-cytotoxic agent suppressed the secretion of SASP factors (e.g., TNFα, IL1α, IL1β, IL6, and CCL2) from senescent osteoblast and osteoclast progenitors, resulting in enhanced osteoblast differentiation and reduced osteoclast differentiation. Furthermore, intraperitoneal administration of cabozantinib to age-related estrogen-deficient mice subjected to ovariectomy prevented bone loss without apparent side effects, increasing osteoblast numbers and reducing osteoclast numbers along the surface of the trabecular bone. In summary, our findings suggest that anti-aging cabozantinib has potential as a preventive anti-osteoporotic agent by promoting osteogenesis and inhibiting osteoclastogenesis through the repression of SASP. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

18 pages, 2145 KiB  
Review
Expression of Aldehyde Dehydrogenase 1A1 in Relapse-Associated Cells in Acute Myeloid Leukemia
by Régis Costello, Garrett M. Dancik, Anaïs Dubiau, Lamia Madaci and Spiros Vlahopoulos
Cells 2025, 14(13), 1038; https://doi.org/10.3390/cells14131038 - 7 Jul 2025
Viewed by 752
Abstract
In acute myeloid leukemia (AML) it is important to elucidate the biological events that lead from remission to relapse, which have a high probability of leading to an adverse disease outcome. The cancer stem cell marker aldehyde dehydrogenase 1 (ALDH1A1) is underexpressed in [...] Read more.
In acute myeloid leukemia (AML) it is important to elucidate the biological events that lead from remission to relapse, which have a high probability of leading to an adverse disease outcome. The cancer stem cell marker aldehyde dehydrogenase 1 (ALDH1A1) is underexpressed in AML cells when compared to healthy cells, both at the RNA level and at the protein level, and at least in the former, both in the bone marrow and in peripheral blood. Nonetheless, ALDH1A1/ALDH1A2 activity increases in AML cells during disease relapse and is higher in adverse prognosis AML in comparison with favorable prognosis AML. Furthermore, especially in relapsed AML and in unfavorable AML, AML cells rich in ALDH1A1 can contain high levels of reactive oxygen species (ROS), in parallel with high ALDH1A1/2 activity. This metabolic feature is clearly incompatible with normal stem cells. The term “stem-like” therefore is useful to coin malignant cells with a variety of genetic makeups, metabolic programming and biomarkers that converge in the function of survival of clones sufficient to sustain, spread and re-establish neoplastic disease. Therefore, AML “stem-like” cells survive cancer treatment that eradicates other malignant cell clones. This fact differentiates AML “stem-like” cells from normal stem and progenitor cells that function in tissue regeneration as part of a distinct hierarchical order of cell phenotypes. The ODYSSEY clinical trial is a Phase I/II study designed to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of ABD-3001, a novel therapeutic agent, in patients with AML who have relapsed or are refractory to standard treatments. In this context, ABD-3001 is used as an inhibitor of cytosolic ALDH1 enzymes, such as ALDH1A1 and ALDH1A2. Full article
(This article belongs to the Section Tissues and Organs)
Show Figures

Figure 1

20 pages, 5423 KiB  
Article
Integrative Transcriptomic Meta-Analysis Reveals Risk Signatures and Immune Infiltration Patterns in High-Grade Serous Ovarian Cancer
by Paula D. Morales-Suárez, Yina T. Zambrano-O, Alejandro Mejía-Garcia, Hsuan Megan Tsao, Liliana Lopez-Kleine, Diego A. Bonilla, Alba L. Combita, Rafel Parra-Medina, Patricia Lopez-Correa, Silvia J. Serrano-G, Juliana L. Rodriguez and Carlos A. Orozco
Immuno 2025, 5(3), 23; https://doi.org/10.3390/immuno5030023 - 25 Jun 2025
Viewed by 621
Abstract
Background: High-grade serous ovarian cancer (HGSOC) is a highly aggressive malignancy with poor prognosis due to late-stage diagnosis and limited treatments. Identifying differentially expressed genes (DEGs), and immune cell infiltration patterns may improve prognostic assessment and therapeutic strategies. Methods: We conducted a meta-analysis [...] Read more.
Background: High-grade serous ovarian cancer (HGSOC) is a highly aggressive malignancy with poor prognosis due to late-stage diagnosis and limited treatments. Identifying differentially expressed genes (DEGs), and immune cell infiltration patterns may improve prognostic assessment and therapeutic strategies. Methods: We conducted a meta-analysis of gene expression data from the GEO (Gene Expression Omnibus, NCBI). DEGs were identified, functionally enriched, and analyzed for protein-protein interactions. Overlaps with oncogenes and tumor suppressor genes were examined. Cox survival analysis and a gene expression-based risk stratification model were developed. Immune infiltration differences were assessed using deconvolution methods. Results: A total of 11 studies (291 HGSOC, 96 controls) identified 892 DEGs, mainly involved in mitochondrial function, vesicle trafficking, and immune regulation. Key oncogenes (EZH2, PDK1, ERBB2) and tumor suppressor genes (BRCA1, DUSP22) were identified. Survival analysis associated the expression of SEC24B, TGOLN2, TRAK1, and CAST with poor prognosis. Low-risk patients had higher activated dendritic cells and CD4+ memory T cells while high-risk patients were enriched in common lymphoid progenitors and megakaryocyte-erythroid progenitors. Conclusions: This study identifies key DEGs in HGSOC progression and presents a risk stratification model predicting patient outcomes. Full article
Show Figures

Figure 1

19 pages, 1216 KiB  
Review
Dysregulation of the Bone Marrow Microenvironment in Pediatric Tumors: The Role of Extracellular Vesicles in Acute Leukemias and Neuroblastoma
by Giovanna D’Amico, Rita Starace, Martina Della Lastra, Danilo Marimpietri, Erica Dander, Fabio Morandi and Irma Airoldi
Int. J. Mol. Sci. 2025, 26(11), 5380; https://doi.org/10.3390/ijms26115380 - 4 Jun 2025
Cited by 1 | Viewed by 832
Abstract
The role of extracellular vesicles has been extensively studied in physiological and pathological conditions, and growing evidence has pinpointed them as key players in tumor progression, regulation of the metastatic niche, and modulation of anti-tumor immune responses. Indeed, a dynamic transfer of extracellular [...] Read more.
The role of extracellular vesicles has been extensively studied in physiological and pathological conditions, and growing evidence has pinpointed them as key players in tumor progression, regulation of the metastatic niche, and modulation of anti-tumor immune responses. Indeed, a dynamic transfer of extracellular vesicles between cancer cells and immunological or non-immunological cells homing in the tumor microenvironment exists, and the balance between their release by cancer cells and by normal cells determines cancer progression. Here, we focused on the role of extracellular vesicles in the dysregulation of the bone marrow environment in pediatric tumors such as acute leukemias and neuroblastomata, whose poor prognosis is strictly related to the involvement of such anatomical site. Acute leukemias arise from bone marrow progenitors, whereas approximately 50% of neuroblastoma patients have bone marrow metastases at diagnosis. Thus, here, we discuss the mechanisms underlying the bone marrow dysregulation in pediatric acute leukemias and neuroblastomata with particular emphasis on the involvement of extracellular vesicles. Full article
Show Figures

Figure 1

29 pages, 3898 KiB  
Article
Bone Marrow Myeloid–Lymphatic Progenitors Expand Tumor Lymphatic Vasculature Through Cell Fusion
by Shaswati Athaiya, Lisa Volk-Draper, Emma Cox, Kathy Robinson, Natalya Zinkevich and Sophia Ran
Cancers 2025, 17(11), 1804; https://doi.org/10.3390/cancers17111804 - 28 May 2025
Viewed by 683
Abstract
Background: Bone marrow (BM)-derived myeloid–lymphatic endothelial cell progenitors (M-LECPs) promote formation of tumor lymphatics that are responsible for metastasis to lymph nodes. The regenerative capacity of BM progenitors to other lineages is mediated through cell fusion, a process that delivers a pro-mitotic message [...] Read more.
Background: Bone marrow (BM)-derived myeloid–lymphatic endothelial cell progenitors (M-LECPs) promote formation of tumor lymphatics that are responsible for metastasis to lymph nodes. The regenerative capacity of BM progenitors to other lineages is mediated through cell fusion, a process that delivers a pro-mitotic message directly to division-restricted cells. This suggested that M-LECPs might use a similar mechanism to induce division of lymphatic endothelial cells (LECs). Methods: To test this hypothesis, we determined expression of fusogenic markers in M-LECP produced in vitro and recruited to human or mouse tumors in vivo as well as quantified their fusion with LECs in both settings. Fusion in vivo was determined in female chimera mice grafted with male BM that have been implanted with MDA-MB-231 or EMT6 breast tumors. Co-staining for Y-chromosome and LEC-specific markers allowed us to quantify tumor lymphatic vessels fused with BM progenitors. Results: We found that both tumor-recruited and in-vitro-produced M-LECPs expressed multiple fusogenic regulators and possessed a significant fusogenic activity towards cultured and vessel-lining LECs. Y-chromosomes, a marker of fusion, were detected in nearly half of tumor lymphatics and were associated with mitotic division, vessel formation, and node metastasis. Both in vitro and in vivo assays showed dependency of fusion on Th2 and Toll-like receptor-4 (TLR4) pathways. Conclusions: This novel mechanism of tumor lymphatic formation triggered by fusion with BM myeloid–lymphatic progenitors suggests a variety of new targets for inhibition of metastatic spread. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

18 pages, 3181 KiB  
Article
Transcriptome-Wide Analysis of Brain Cancer Initiated by Polarity Disruption in Drosophila Type II Neuroblasts
by Simona Paglia, Patrizia Morciano, Dario de Biase, Federico Manuel Giorgi, Annalisa Pession and Daniela Grifoni
Int. J. Mol. Sci. 2025, 26(11), 5115; https://doi.org/10.3390/ijms26115115 - 26 May 2025
Viewed by 617
Abstract
Brain tumors, in particular gliomas and glioblastoma multiforme (GBM), are thought to originate from different cells facing specific founding insults, a feature that partly justifies the complexity and heterogeneity of these severe forms of cancer. However, gliomas and GBM are usually reproduced in [...] Read more.
Brain tumors, in particular gliomas and glioblastoma multiforme (GBM), are thought to originate from different cells facing specific founding insults, a feature that partly justifies the complexity and heterogeneity of these severe forms of cancer. However, gliomas and GBM are usually reproduced in animal models by inducing molecular alterations in mature glial cells, which, though being part of the puzzle, do not represent the whole picture. To fill this conceptual gap, we previously developed a neurogenic model of brain cancer in Drosophila, demonstrating that the loss of cell polarity in neural stem cells (called neuroblasts in the fruit fly) is sufficient to promote the formation of malignant masses that continue to grow in the adult, displaying several phenotypic traits typical of human GBM. Here, we expand on previous work by restricting polarity disruption to Drosophila type II neuroblasts, whose self-renewal is comparable to that of mammalian neural progenitors, with the aim to capture the molecular signature of the resulting cancers in a specific and reproducible context. A comparison of the most deregulated transcripts with those found in human primary GBMs confirmed that our model can be proficiently used to delve into the roots of human brain tumorigenesis. Full article
(This article belongs to the Special Issue Drosophila: A Model System for Human Disease Research)
Show Figures

Figure 1

48 pages, 2181 KiB  
Review
Tumor-Associated Macrophages: Polarization, Immunoregulation, and Immunotherapy
by Abdullah Farhan Saeed
Cells 2025, 14(10), 741; https://doi.org/10.3390/cells14100741 - 19 May 2025
Cited by 1 | Viewed by 3304
Abstract
Tumor-associated macrophages’ (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based [...] Read more.
Tumor-associated macrophages’ (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based on cytokines and metabolic signals. Recent advances in TAM heterogeneity, polarization, characterization, immunological responses, and therapy are described here. The manuscript details TAM functions and their role in resistance to PD-1/PD-L1 blockade. Similarly, TAM-targeted approaches, such as CSF-1R inhibition or PI3Kγ-driven reprogramming, are discussed to address anti-tumor immunity suppression. Furthermore, innovative biomarkers and combination therapy may enhance TAM-centric cancer therapies. It also stresses the relevance of this distinct immune cell in human health and disease, which could impact future research and therapies. Full article
Show Figures

Graphical abstract

28 pages, 8689 KiB  
Article
Extracellular-Vesicle-Associated UBE2NL and HIST2H3PS2 Promote Tumor Aggressiveness and Metastasis in Gynecologic Cancer
by Chih-Ming Ho, Ting-Lin Yen, Tzu-Hao Chang and Shih-Hung Huang
Int. J. Mol. Sci. 2025, 26(10), 4833; https://doi.org/10.3390/ijms26104833 - 18 May 2025
Viewed by 549
Abstract
Extracellular vesicles (EVs) play pivotal roles in tumor progression and metastasis by mediating intercellular communication within the tumor microenvironment. In this study, we identified two novel EX cargo proteins—UBE2NL and HIST2H3PS2—derived from highly aggressive epithelial ovarian cancer (EOC) cells and mesenchymal-type ovarian stromal [...] Read more.
Extracellular vesicles (EVs) play pivotal roles in tumor progression and metastasis by mediating intercellular communication within the tumor microenvironment. In this study, we identified two novel EX cargo proteins—UBE2NL and HIST2H3PS2—derived from highly aggressive epithelial ovarian cancer (EOC) cells and mesenchymal-type ovarian stromal progenitor cells (MSC-OCSPCs) but absent in less aggressive SKOV3 cells. Quantitative proteomic profiling via LC-MS/MS and TCGA-integrated analysis revealed that high expression of these genes correlated with advanced tumor stages and poor overall survival in EOC, and high HIST2H3PS2 expression predicted poor survival in endometrial cancer (EC). Functionally, UBE2NL and HIST2H3PS2 overexpression promoted EOC cell invasiveness, which was further enhanced by EX-mediated autocrine and paracrine effects. In contrast, the knockdown of UBE2NL reduced cell invasiveness and prolonged mouse survival in vivo. Moreover, HIST2H3PS2-enriched EXs significantly increased peritoneal dissemination and ascites in murine models. These findings suggest that EX-packaged UBE2NL and HIST2H3PS2 drive tumor aggressiveness and metastasis in gynecologic cancers, highlighting their potential as prognostic biomarkers and therapeutic targets. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

30 pages, 2663 KiB  
Review
IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis
by Abigail J. Harland and Claire M. Perks
Int. J. Mol. Sci. 2025, 26(10), 4749; https://doi.org/10.3390/ijms26104749 - 15 May 2025
Viewed by 1121
Abstract
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation—most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent [...] Read more.
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation—most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent of a neurodevelopmental hierarchy, in which neural stem and progenitor markers are widely expressed by tumour stem-like cells. However, NSC fates and the cues that drive them are poorly understood. Studying the crosstalk within NSC niches may better inform our understanding of glioblastoma initiation and development. Insulin-like growth factor binding protein 2 (IGFBP-2) has a well-established prognostic role in glioblastoma, and cell-based mechanistic studies show the independent activation of downstream oncogenic pathways. However, IGFBP-2 is more commonly recognised as a modulator of insulin-like growth factors (IGFs) for receptor tyrosine kinase signal propagation or attenuation. In the adult human brain, both IGFBP-2 and IGF-II expression are retained in the choroid plexus (ChP) and secreted into the cerebral spinal fluid (CSF). Moreover, secretion by closely associated cells and NSCs themselves position IGFBP-2 and IGF-II as interesting factors within the NSC niche. In this review, we will highlight the experimental findings that show IGFBP-2 and IGF-II influence NSC behaviour. Moreover, we will link this to glioblastoma biology and demonstrate the requirement for further analysis of these factors in glioma stem cells (GSCs). Full article
(This article belongs to the Special Issue The Role of the IGF Axis in Disease, 4th Edition)
Show Figures

Figure 1

19 pages, 6195 KiB  
Article
Identification of Novel HPK1 Hit Inhibitors: From In Silico Design to In Vitro Validation
by Israa H. Isawi, Rayan M. Obeidat, Soraya Alnabulsi and Rufaida Al Zoubi
Int. J. Mol. Sci. 2025, 26(9), 4366; https://doi.org/10.3390/ijms26094366 - 4 May 2025
Viewed by 849
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a negative regulator of T-cells, B-cells, and dendritic cells, has gained attention in antitumor immunotherapy research over the past decade. No HPK1 inhibitor has yet reached clinical approval, largely due to selectivity and drug-like limitations. Leveraging the available [...] Read more.
Hematopoietic progenitor kinase 1 (HPK1), a negative regulator of T-cells, B-cells, and dendritic cells, has gained attention in antitumor immunotherapy research over the past decade. No HPK1 inhibitor has yet reached clinical approval, largely due to selectivity and drug-like limitations. Leveraging the available structural insights into HPK1, we conducted a rational hit identification using a structure-based virtual screening of over 600,000 drug-like molecules from ASINEX and OTAVA databases. A series of molecular docking studies, in vitro kinase assays, and molecular dynamics simulations were conducted to identify viable HPK1 inhibitor hits. This approach resulted in two promising novel hit scaffolds, 4H-Pyrido[1,2-a] thieno[2,3-d] pyrimidin-4-one (ISR-05) and quinolin-2(1H)-one (ISR-03), neither of which has previously been reported as an HPK1 inhibitor. ISR-05 and ISR-03 exhibited IC50 values of 24.2 ± 5.07 and 43.9 ± 0.134 µM, respectively, in kinase inhibition assays. These hits constitute tractable starting points for future hit-to-lead optimization aimed at developing more effective HPK1 inhibitors for cancer therapy. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Graphical abstract

22 pages, 2431 KiB  
Perspective
Safely Targeting Cancer, the Wound That Never Heals, Utilizing CBP/Beta-Catenin Antagonists
by Yusuke Higuchi, Jia-Ling Teo, Daniel Yi and Michael Kahn
Cancers 2025, 17(9), 1503; https://doi.org/10.3390/cancers17091503 - 29 Apr 2025
Viewed by 977
Abstract
Stem cells, both normal somatic (SSC) and cancer stem cells (CSC) exist in minimally two states, i.e., quiescent and activated. Regulation of these two states, including their reliance on different metabolic processes, i.e., FAO and glycolysis in quiescent versus activated stem cells respectively, [...] Read more.
Stem cells, both normal somatic (SSC) and cancer stem cells (CSC) exist in minimally two states, i.e., quiescent and activated. Regulation of these two states, including their reliance on different metabolic processes, i.e., FAO and glycolysis in quiescent versus activated stem cells respectively, involves the analysis of a complex array of factors (nutrient and oxygen levels, adhesion molecules, cytokines, etc.) to initiate the epigenetic changes to either depart or enter quiescence. Quiescence is a critical feature of SSC that is required to maintain the genomic integrity of the stem cell pool, particularly in long lived complex organisms. Quiescence in CSC, whether they are derived from mutations arising in SSC, aberrant microenvironmental regulation, or via dedifferentiation of more committed progenitors, is a critical component of therapy resistance and disease latency and relapse. At the beginning of vertebrate evolution, approximately 450 million years ago, a gene duplication generated the two members of the Kat3 family, CREBBP (CBP) and EP300 (p300). Despite their very high degree of homology, these two Kat3 coactivators play critical and non-redundant roles at enhancers and super-enhancers via acetylation of H3K27, thereby controlling stem cell quiescence versus activation and the cells metabolic requirements. In this review/perspective, we discuss the unique regulatory roles of CBP and p300 and how specifically targeting the CBP/β-catenin interaction utilizing small molecule antagonists, can correct lineage infidelity and safely eliminate quiescent CSC. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

16 pages, 12668 KiB  
Article
Molecular Iodine Exhibited Differential Antiproliferative Actions in Progenitor and Stem Populations from Chemoresistant Cancer Cells
by Irasema Mendieta, Jazmin Leon-Pichardo, Gustavo Orizaga-Osti, Edgar R. Juvera-Avalos, Uriel Rangel-Chavez, Evangelina Delgado-Gonzalez, Brenda Anguiano and Carmen Aceves
Int. J. Mol. Sci. 2025, 26(9), 4020; https://doi.org/10.3390/ijms26094020 - 24 Apr 2025
Viewed by 764
Abstract
Cancer stem cells (CSCs) are described as a subpopulation of cells with capabilities of self-renewal, chemoresistance, and invasiveness. CSCs reside in tumor niches and can be studied in vitro through their enrichment in spheroids (Stem). Molecular iodine (I2) induces apoptosis and [...] Read more.
Cancer stem cells (CSCs) are described as a subpopulation of cells with capabilities of self-renewal, chemoresistance, and invasiveness. CSCs reside in tumor niches and can be studied in vitro through their enrichment in spheroids (Stem). Molecular iodine (I2) induces apoptosis and differentiation in various cancer cells. I2 can activate peroxisome proliferator-activated receptors type gamma (PPARγ), and its pathways are associated with its oxidant/antioxidant capacity. This work aimed to compare the effect of I2 supplementation in progenitor and CSC populations with low (MCF-7 and S-K-NAS) and high invasiveness (MDA-MB231 and SK-N-BE2) in mammary and neuroblastoma (NB) cell lines. Results showed that the CSC population enriched by the spheroid culture overexpressed stem messengers CD44, SOX2, and NMYC and exhibited the highest mitochondrial metabolism (membrane mitochondrial potential and O2). The presence of I2 increases PPARγ expression and induces apoptosis through the Bax/Bcl2 index in all populations but silences NMYC expression and reduces mitochondrial metabolism in Stem NB. I2 also enhances the expression of nuclear erythroid factor 2 (Nrf2) in all populations, but the target antioxidant superoxide dismutase 2 (SOD2) is only elevated in progenitor cells. In contrast, the mitophagy inductors PTEN-induced putative kinase 1 (Pink1) and microtubule-associated protein1 light chain3 alpha (LC3) were overexpressed in Stem populations. I2-preselected SK-N-BE2 populations exhibited minor implantation and invasion capacities in the in vivo zebrafish model. These data indicate that I2 interferes with viability, implantation, and invasion capacity in all cell lines, but the molecular mechanisms vary depending on the progenitor or Stem condition. Full article
(This article belongs to the Special Issue The Role of Iodinated Compounds and Molecular Iodine in Human Disease)
Show Figures

Figure 1

26 pages, 4125 KiB  
Article
Adult B-Cell Acute Lymphoblastic Leukaemia Antigens and Enriched Pathways Identify New Targets for Therapy
by Eithar Mohamed, Sara Goodman, Leah Cooksey, Daniel M. Fletcher, Olivia Dean, Viktoriya B. Boncheva, Ken I. Mills, Kim H. Orchard and Barbara-ann Guinn
Onco 2025, 5(2), 19; https://doi.org/10.3390/onco5020019 - 22 Apr 2025
Viewed by 1569
Abstract
Background: Adult B-cell acute lymphoblastic leukaemia (aB-ALL) is characterised by abnormal differentiation and proliferation of lymphoid progenitors. Despite a significant improvement in relapse-free and overall survival for children with B-ALL, aB-ALL has a particularly poor prognosis with a 5-year survival rate of 20%. [...] Read more.
Background: Adult B-cell acute lymphoblastic leukaemia (aB-ALL) is characterised by abnormal differentiation and proliferation of lymphoid progenitors. Despite a significant improvement in relapse-free and overall survival for children with B-ALL, aB-ALL has a particularly poor prognosis with a 5-year survival rate of 20%. First remission is achieved for most patients, but relapse is common with a high associated mortality. New treatments such as immunotherapy offer an opportunity to extend remission and prevent relapse. Methods: aB-ALL antigens were identified using different sources—immunoscreening, protoarrays, two microarrays and one cancer-testis antigen database, and a review of the genomic analyses of aB-ALL. A total of 385 aB-ALL-associated gene products were examined for their association with patient survival. Results: We identified 87 transcripts with differential expression between aB-ALL and healthy volunteers (peripheral blood, bone marrow and purified CD19+ cells), and 42 that were associated with survival. Enrichr analysis showed that the Transforming Growth Factor-β (TGFβ), Wnt and Hippo pathways were highly represented (p < 0.02). We found that SOX4 and ROCK1 were upregulated in all types of B-ALL (ROCK1 having a p < 0.001 except in t(8;14) patients), as well as SMAD3 and TEAD4 upregulation being associated with survival (p = 0.0008, 0.05 and 0.001, respectively). Expression of each aB-ALL antigen was verified by qPCR, but only TEAD4 showed significant transcript upregulation in aB-ALL compared to healthy volunteer CD19+ cells (p = 0.01). Conclusions: We have identified a number of antigens and their pathways that play key roles in aB-ALL and may act as useful targets for future immunotherapy strategies. Full article
Show Figures

Figure 1

27 pages, 1100 KiB  
Review
CD8+ T Cell Subsets as Biomarkers for Predicting Checkpoint Therapy Outcomes in Cancer Immunotherapy
by Rosaely Casalegno Garduño, Alf Spitschak, Tim Pannek and Brigitte M. Pützer
Biomedicines 2025, 13(4), 930; https://doi.org/10.3390/biomedicines13040930 - 9 Apr 2025
Cited by 2 | Viewed by 1556
Abstract
The advent of immune checkpoint blockade (ICB) has transformed cancer immunotherapy, enabling remarkable long-term outcomes and improved survival, particularly with ICB combination treatments. However, clinical benefits remain confined to a subset of patients, and life-threatening immune-related adverse effects pose a significant challenge. This [...] Read more.
The advent of immune checkpoint blockade (ICB) has transformed cancer immunotherapy, enabling remarkable long-term outcomes and improved survival, particularly with ICB combination treatments. However, clinical benefits remain confined to a subset of patients, and life-threatening immune-related adverse effects pose a significant challenge. This limited efficacy is attributed to cancer heterogeneity, which is mediated by ligand–receptor interactions, exosomes, secreted factors, and key transcription factors. Oncogenic regulators like E2F1 and MYC drive metastatic tumor environments and intertwine with immunoregulatory pathways, impairing T cell function and reducing immunotherapy effectiveness. To address these challenges, FDA-approved biomarkers, such as tumor mutational burden (TMB) and programmed cell death-ligand 1 (PD-L1) expression, help to identify patients most likely to benefit from ICB. Yet, current biomarkers have limitations, making treatment decisions difficult. Recently, T cells—the primary target of ICB—have emerged as promising biomarkers. This review explores the relationship between cancer drivers and immune response, and emphasizes the role of CD8+ T cells in predicting and monitoring ICB efficacy. Tumor-infiltrating CD8+ T cells correlate with positive clinical outcomes in many cancers, yet obtaining tumor tissue remains complex, limiting its practical use. Conversely, circulating T cell subsets are more accessible and have shown promise as predictive biomarkers. Specifically, memory and progenitor exhausted T cells are associated with favorable immunotherapy responses, while terminally exhausted T cells negatively correlate with ICB efficacy. Ultimately, combining biomarkers enhances predictive accuracy, as demonstrated by integrating TMB/PD-L1 expression with CD8+ T cell frequency. Computational models incorporating cancer and immune signatures could further refine patient stratification, advancing personalized immunotherapy. Full article
(This article belongs to the Special Issue Roles of T Cells in Immunotherapy, 2nd Edition)
Show Figures

Figure 1

18 pages, 2345 KiB  
Article
An Analysis of the mRNA Expression of Peripheral-Blood Stem and Progenitor Cell Markers in Pancreatic Neoplastic Disorders
by Krzysztof Dąbkowski, Maciej Tarnowski, Krzysztof Safranow, Maria Dąbkowska, Alicja Kosiorowska, Kamila Pukacka and Teresa Starzyńska
Curr. Issues Mol. Biol. 2025, 47(4), 236; https://doi.org/10.3390/cimb47040236 - 28 Mar 2025
Viewed by 586
Abstract
Background: Our aim was to assess the expression profiles of the messenger RNA (mRNA) expression profiles of stem-cell genes (POU5F1, NANOG) and pancreatic progenitor genes (CK19, HES1, INS, PDX1) in peripheral-blood mononuclear cells (PBMCs) in [...] Read more.
Background: Our aim was to assess the expression profiles of the messenger RNA (mRNA) expression profiles of stem-cell genes (POU5F1, NANOG) and pancreatic progenitor genes (CK19, HES1, INS, PDX1) in peripheral-blood mononuclear cells (PBMCs) in selected neoplastic pancreatic diseases, such as cancer and neuroendocrine tumors, to identify neoplastic disease markers in the pancreas. Methods: In this study, 49 patients diagnosed with pancreatic neoplastic diseases (37 with cancer and 12 with neuroendocrine tumors) and 34 control patients, all of whom were hospitalized at a tertiary center, were enrolled. Venous blood samples were collected from the participants, and RNA was extracted from PBMCs. The mRNA expression levels of six stem-cell and pancreatic progenitor markers— POU5F1 (POU class 5 homeobox 1), NANOG, CK19 (keratin 19), HES1 (HES family bHLH transcription factor 1), INS (insulin), and PDX1 (pancreatic and duodenal homeobox 1)—were quantified via real-time quantitative PCR. The data were statistically analyzed to explore associations between gene-expression levels and various clinical, biochemical, and morphological parameters (including full blood count, Ca 19-9, weight, height, and BMI) via the Kruskal–Wallis test, Mann–Whitney U test, and Spearman rank correlation coefficient. Results: The results revealed that the expression of the gene associated with early stem cells, NANOG (median= 0.002, p = 0.03), as well as the genes encoding insulin INS (median = 0.004, p = 0.02) and CK19 (median 0.0003, p = 0.005), was significantly elevated in patients with pancreatic cancer. However, the gene-expression levels in patients with neuroendocrine tumors did not exhibit statistically significant differences compared to those observed in the control group. Additionally, no significant differences in gene expression were observed among patients at different stages of pancreatic cancer. Furthermore, CK19 overexpression was found to be positively correlated with inflammatory markers, specifically C-reactive protein (CRP) and WBC, in patients with pancreatic cancer. Conclusions: An elevated mRNA expression of specific stem and pancreatic progenitor genes (NANOG, INS, CK19) in PBMCs may serve as a potential markers for pancreatic cancer, reflecting the disease’s interplay with systemic inflammation. Full article
Show Figures

Figure 1

Back to TopTop