Sign in to use this feature.

Years

Between: -

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,878)

Search Parameters:
Journal = Cancers
Section = Cancer Biomarkers

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1060 KB  
Article
Expression Analysis of JCAD and IL-33 in Gingival Cancer Tumor Angiogenesis
by Tatsuya Shirai, Yasumasa Kakei, Yumi Muraki, Tatsuya Nagano, Ratoe Suraya, Kaito Uryu, Daisuke Takeda, Manabu Shigeoka, Akira Kimoto, Takumi Hasegawa, Tetsuya Hara, Noriaki Emoto and Masaya Akashi
Cancers 2025, 17(23), 3732; https://doi.org/10.3390/cancers17233732 - 21 Nov 2025
Abstract
Background: Tumor angiogenesis is a key step in oral squamous cell carcinoma (OSCC) development. Here, we evaluated the expression patterns of junctional cadherin 5-associated (JCAD), a pathological angiogenesis protein, and interleukin-33 (IL-33) in OSCC to investigate their roles in pathological angiogenesis. Methods [...] Read more.
Background: Tumor angiogenesis is a key step in oral squamous cell carcinoma (OSCC) development. Here, we evaluated the expression patterns of junctional cadherin 5-associated (JCAD), a pathological angiogenesis protein, and interleukin-33 (IL-33) in OSCC to investigate their roles in pathological angiogenesis. Methods: Wound healing assays were performed to evaluate pathological angiogenesis in JCAD knockout (JCAD-KO) mice. In human mandibular gingival SCC and lymph nodes specimens, the numbers of blood vessels positive for CD34 (a vascular endothelial cell marker), CD105 (a well-established tumor angiogenesis marker), JCAD, and IL-33 were counted. We also evaluated the effects of tumor necrosis factor-α (TNF-α) stimulation as a pro-angiogenic factor on human umbilical vein endothelial cells (HUVECs) with JCAD knockdown. Results: In JCAD-KO mouse skin, wound healing and angiogenesis were significantly disturbed. In the clinical samples, the number of microvessels in which CD105 and JCAD were expressed but intranuclear IL-33 expression was lost significantly increased in the intratumoral area compared with the normal area. JCAD knockdown restored the TNF-α-induced loss of intranuclear IL-33 expression in HUVECs. Conclusions: Our combined assessment of JCAD and IL-33 supports the evaluation of tumor angiogenesis in OSCC. JCAD is a potential target for controlling tumor angiogenesis mediated by TNF-α. Full article
(This article belongs to the Special Issue The Biomarkers of Oral Cancer)
17 pages, 1996 KB  
Article
AdhesionScore: A Prognostic Predictor of Breast Cancer Patients Based on a Cell Adhesion-Associated Gene Signature
by Catarina Esquível, Rogério Ribeiro, Ana Sofia Ribeiro, Pedro G. Ferreira and Joana Paredes
Cancers 2025, 17(23), 3731; https://doi.org/10.3390/cancers17233731 - 21 Nov 2025
Abstract
Background: Aberrant or loss of cell adhesion drives invasion and metastasis, key hallmarks of cancer progression. In this work, we hypothesized that a gene signature related to cell adhesion could predict breast cancer prognosis. Methods: Highly variant genes were tested for association with [...] Read more.
Background: Aberrant or loss of cell adhesion drives invasion and metastasis, key hallmarks of cancer progression. In this work, we hypothesized that a gene signature related to cell adhesion could predict breast cancer prognosis. Methods: Highly variant genes were tested for association with overall survival using Cox regression. Adhesion-related genes were identified through gene ontology analysis and multivariate Cox regression, with AIC selection, defined the prognostic signature. The AdhesionScore was then calculated as a weighted sum of gene expression, with risk stratification assessed by Kaplan–Meier and log-rank tests. Results: We found that the AdhesionScore was a significant independent predictor of poor survival in three large independent datasets, as it provided a robust stratification of patient prognosis in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) (HR: 2.65; 95% CI: 2.33–3.0, p = 2.34 × 10−51), The Cancer Genome Atlas (TCGA) (HR: 3.46; 95% CI: 2.35–5.09, p = 3.50 × 10−10), and the GSE96058 (HR: 2.83; 95% CI: 2.20–3.65, p = 6.29 × 10−16) datasets. The 5-year risk of death in the high-risk group was 32.41% for METABRIC, 27.8% for TCGA, and 17.54% for GSE96058 datasets. Consistently, HER2-enriched and triple-negative breast carcinomas (TNBC) cases showed higher AdhesionScores than luminal subtypes, indicating an association with aggressive tumor biology. Conclusions: We have developed, for the first time, a molecular signature based on cell adhesion, as well as an associated AdhesionScore that can predict patient prognosis in invasive breast cancer, with potential clinical application. We developed a novel adhesion-based molecular signature, the AdhesionScore, that robustly predicts prognosis in breast cancer across independent cohorts, highlighting its potential clinical utility for patient risk stratification. Full article
Show Figures

Figure 1

21 pages, 2674 KB  
Article
Exosomal Thomsen–Friedenreich Glycoantigen as a Sensitive and Specific Biomarker for Colon, Ovarian and Prostate Cancer Diagnosis
by Yafei Su, Man Qi, Shoaib Vasini, Mary E. Reid, Kate Rittenhouse-Olson, Grace K. Dy and Yun Wu
Cancers 2025, 17(23), 3729; https://doi.org/10.3390/cancers17233729 - 21 Nov 2025
Abstract
Background/Objectives: The screening and diagnosis of colon, ovarian, and prostate cancers are challenged by invasive procedures and lack of effective screening tests. To address these challenges, we developed an exosome-based liquid biopsy assay that utilized a new exosomal biomarker, α-linked Thomsen–Friedenreich glycoantigen (TF-Ag-α, [...] Read more.
Background/Objectives: The screening and diagnosis of colon, ovarian, and prostate cancers are challenged by invasive procedures and lack of effective screening tests. To address these challenges, we developed an exosome-based liquid biopsy assay that utilized a new exosomal biomarker, α-linked Thomsen–Friedenreich glycoantigen (TF-Ag-α, Galβ1-3GalNAc-α). This assay provides a minimally invasive method with high sensitivity and specificity for the screening and diagnosis of colon, ovarian and prostate cancers. Methods: Exosomal TF-Ag-α levels in 10 μL serum samples were measured using a compact, surface plasmon resonance-based biosensor developed to facilitate clinical translation. The diagnostic performance of exosomal TF-Ag-α was initially evaluated in a training set consisting of normal controls (n = 80) and cancer patients (n = 120, 40 for prostate, 40 for colon, and 40 for ovarian cancer), and then validated in an independent test set including normal controls (n = 29) and cancer patients (n = 60, 20 for each cancer type). Receiver operating characteristic (ROC) analysis was performed to assess diagnostic sensitivity, specificity and area under the curve (AUC) of exosomal TF-Ag-α. Results: Serum samples from cancer patients showed significantly elevated levels of exosomal TF-Ag-α compared with normal controls. Exosomal TF-Ag-α distinguished all cancer patients from normal controls with a sensitivity of 99.2%, specificity of 100% and an overall accuracy of 99.5% at a cutoff value of 0.406. Conclusions: These findings demonstrated the clinical translational promise of exosomal TF-Ag-α as a liquid biopsy biomarker for the detection of colon, ovarian and prostate cancers. Full article
(This article belongs to the Special Issue Advances in Exosomes and Cancer Biomarkers)
Show Figures

Figure 1

24 pages, 1325 KB  
Review
Bridging Discovery and Treatment: Cancer Biomarker
by Hengrui Liu, Ilayda Karsidag, Rebecca Golin and Guangzhen Wu
Cancers 2025, 17(22), 3720; https://doi.org/10.3390/cancers17223720 - 20 Nov 2025
Abstract
Biomarkers have transformed cancer care by linking molecular insights to personalized treatment strategies. This review first surveys the spectrum of detection modalities, including tissue genomics and histopathology; liquid biopsies such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and extracellular vesicles; imaging [...] Read more.
Biomarkers have transformed cancer care by linking molecular insights to personalized treatment strategies. This review first surveys the spectrum of detection modalities, including tissue genomics and histopathology; liquid biopsies such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and extracellular vesicles; imaging and radiomics; and multi-omic approaches spanning epigenomics, transcriptomics, proteomics, metabolomics, and microbiomics. We then critically appraise the translational challenges that hinder clinical implementation, assay standardization, data integration, analytical and clinical validation, regulatory pathways, and cost-effectiveness. Finally, we explore emerging solutions, including artificial intelligence (AI)-driven multi-modal phenotyping, adaptive trial designs, and point-of-care assays, and outline future directions for expanding access and equitable adoption. By emphasizing the central role of biomarkers in guiding targeted and immune-based therapies, we underscore their potential to overcome tumor heterogeneity, preempt resistance, and ultimately improve patient outcomes. Full article
(This article belongs to the Special Issue Diagnostic Biomarkers in Cancers Study)
Show Figures

Figure 1

15 pages, 6659 KB  
Article
Results of the First Folate Receptor Alpha Testing Trial by the German Quality Assurance Initiative in Pathology (QuIP®)
by Alexander Scheiter, Sven Mattern, Verena Gassenmaier, Hans-Ulrich Schildhaus, Matthias Christgen, Hans Kreipe, Hermann Herbst, Bettina Lambert, Guido Sauter, Maximilian Lennartz, Korinna Jöhrens, Florian Sperling, Afschin Soleiman, Ramona Erber, Stephan Singer, Annette Staebler and Kirsten Utpatel
Cancers 2025, 17(22), 3703; https://doi.org/10.3390/cancers17223703 - 19 Nov 2025
Viewed by 137
Abstract
Background: Folate receptor alpha (FRα) is a glycosylphosphatidylinositol-anchored membrane protein encoded by the FOLR1 gene. Its overexpression in various cancers, including ovarian carcinoma, makes it a promising target for antibody-drug conjugates (ADC). Mirvetuximab soravtansine-gynx, an FRα-targeting ADC, has been approved by the FDA [...] Read more.
Background: Folate receptor alpha (FRα) is a glycosylphosphatidylinositol-anchored membrane protein encoded by the FOLR1 gene. Its overexpression in various cancers, including ovarian carcinoma, makes it a promising target for antibody-drug conjugates (ADC). Mirvetuximab soravtansine-gynx, an FRα-targeting ADC, has been approved by the FDA and EMA for the treatment of FRα-positive, platinum-resistant ovarian cancer. In the United States, patient selection is tied to the VENTANA FOLR1 (FOLR1-2.1) RxDx Assay, an immunohistochemical (IHC) test that identifies tumors with ≥75% moderate-to-strong membrane staining. However, in the European Union, no specific IHC test is mandated, and alternative antibodies are frequently used in routine pathology, necessitating validation of their diagnostic performance. Methods and Results: We report the results of the first interlaboratory proficiency trial on FRα testing conducted by the German Quality Assurance Initiative in Pathology (QuIP®). Sixty-eight pathology institutes participated across internal and open trials using a variety of antibodies and staining platforms. The VENTANA FOLR1 RxDx Assay demonstrated the highest reliability, with 83% of participating laboratories achieving a successful result. In contrast, alternative clones such as BN3.2 (Leica/Novocastra) and EPR20277 (Abcam) showed substantially weaker staining intensity, lower concordance with reference values, and success rates of only 22–25%, while other antibodies failed entirely. Problem analysis revealed that failures with the VENTANA FOLR1 (FOLR1-2.1) RxDx Assay were mainly due to interpretative challenges, whereas weak staining was the predominant issue with alternative clones. Participation in a preparatory online seminar improved pass rates, underscoring the importance of training. Conclusions: These findings highlight the critical importance of standardized, validated assays for FRα detection to ensure accurate patient selection for targeted therapies. The study emphasizes the need for further optimization of alternative antibodies before clinical implementation. Full article
Show Figures

Figure 1

19 pages, 1377 KB  
Review
Circulating Tumor DNA (ctDNA) in Gastroesophageal Adenocarcinoma (GEA): Evidence and Emerging Applications
by Oudai Sahwan, Lin Batha, Fares Jamal and Mohamad Bassam Sonbol
Cancers 2025, 17(22), 3692; https://doi.org/10.3390/cancers17223692 - 18 Nov 2025
Viewed by 281
Abstract
The role of circulating tumor DNA (ctDNA) in gastroesophageal adenocarcinoma (GEA) has expanded in recent years. In resectable disease, postoperative ctDNA is able to detect patients at highest risk of recurrence months before scans. Tumor-informed assays provide the best sensitivity and emerging methylation [...] Read more.
The role of circulating tumor DNA (ctDNA) in gastroesophageal adenocarcinoma (GEA) has expanded in recent years. In resectable disease, postoperative ctDNA is able to detect patients at highest risk of recurrence months before scans. Tumor-informed assays provide the best sensitivity and emerging methylation assays are useful when tissue is scarce. In metastatic GEA, baseline ctDNA burden correlates with prognosis, and a decrease in ctDNA level after treatment initiation reflects therapeutic response. It can also uncover actionable targets, including ERBB2, FGFR2, and MSI-H, and detect resistance that can arise after starting treatment. Limitations include variable assay performance, low shedding in some tumors, clonal hematopoiesis confounding, and a lack of randomized data showing that ctDNA-guided changes improve outcomes. Ongoing trials are testing MRD-guided escalation/de-escalation and ctDNA-directed biomarker therapy. In this review, we evaluate the role of ctDNA in GEA cancers over recent years. Full article
Show Figures

Figure 1

19 pages, 7499 KB  
Article
Caught in the Act: Tumor-Immune Interactions in Circulation of Patients with Immune Marker Positive Circulating Tumor Cells
by Amin Naghdloo, Mohamed Kamal, Dean Tessone, Valerie Hennes, James Hicks and Peter Kuhn
Cancers 2025, 17(22), 3667; https://doi.org/10.3390/cancers17223667 - 15 Nov 2025
Viewed by 252
Abstract
Background/Objectives: Circulating tumor cells (CTCs) and large extracellular vesicles (LEVs) are key components of the liquid biopsy that provide minimally invasive access to tumor biology. A clinically relevant subset of CTCs coexpressing epithelial and immune markers (im.CTCs) has been described, yet the origin [...] Read more.
Background/Objectives: Circulating tumor cells (CTCs) and large extracellular vesicles (LEVs) are key components of the liquid biopsy that provide minimally invasive access to tumor biology. A clinically relevant subset of CTCs coexpressing epithelial and immune markers (im.CTCs) has been described, yet the origin of this phenotype remains unclear. In this study, we investigated the cellular and molecular context underlying the emergence of immune marker expression on CTCs and LEVs. Methods: Using high-resolution immunofluorescence microscopy of patient-derived blood samples, we identified direct physical interactions between white blood cells (WBCs) and both im.CTCs and im.LEVs, exclusively in patients harboring im.CTCs. Results: In several cases, WBCs partially encapsulated CTCs and LEVs, and quantitative analysis revealed localized enrichment of immune membrane markers at the contact interface, distinguishing these events from random proximity. Proteomic profiling further identified CD4+ T cells as the predominant interacting immune cell type and confirmed the presence of CD45, CD3, and CD4 on the interacting CTCs and LEVs, matching their WBC counterparts. Conclusion: These findings support membrane transfer as a potential mechanism for the acquisition of immune markers by CTCs and LEVs and provide in vivo evidence of contact-dependent tumor-immune interactions in circulation with implications for immune modulation and clinical interpretation of the im.CTC phenotype. Full article
(This article belongs to the Special Issue Recent Advances in Liquid Biopsy Biomarkers of Cancer)
Show Figures

Figure 1

12 pages, 865 KB  
Article
Loss of Argininosuccinate Synthetase-1 (ASS1) Occurs in Esophageal Adenocarcinoma and Represents a Promising Biomarker for Therapy with Pegargiminase
by Karl Knipper, Su Ir Lyu, Eleni Tzitzili, Sarah-Michele Spielmann, Christiane J. Bruns, Thomas Schmidt, Felix C. Popp and Alexander Quaas
Cancers 2025, 17(22), 3624; https://doi.org/10.3390/cancers17223624 - 11 Nov 2025
Viewed by 320
Abstract
Background/Objectives: Despite the introduction of targeted therapies such as Nivolumab, survival outcomes for patients with esophageal adenocarcinoma remain poor. During tumorigenesis, some tumors develop auxotrophy by downregulation of Argininosuccinate Synthetase-1 (ASS1), making them reliant on external arginine supply and thus potentially susceptible [...] Read more.
Background/Objectives: Despite the introduction of targeted therapies such as Nivolumab, survival outcomes for patients with esophageal adenocarcinoma remain poor. During tumorigenesis, some tumors develop auxotrophy by downregulation of Argininosuccinate Synthetase-1 (ASS1), making them reliant on external arginine supply and thus potentially susceptible to arginine deprivation therapy. Arginine deprivation therapy with agents such as pegargiminase has shown improved survival in patients with pleural mesothelioma exhibiting ASS1 loss in tumor cells. Therefore, we investigated the prevalence of ASS1 loss in esophageal adenocarcinoma. Methods: First, we compared the staining patterns of three antibodies for ASS1 with RNA in situ Scope analysis results to identify the most reliable antibody for ASS1 immunohistochemical staining in esophageal adenocarcinoma. Subsequently, we performed ASS1 immunohistochemical staining on samples from 97 patients who underwent curative resection. The staining results were classified into three categories based on expression levels: negative, low-positive, and positive. Results: Among all included patients, 6.2% exhibited an ASS1 loss, and 6.2% showed low ASS1 expression. Notably, patients with an ASS1 loss did not demonstrate a response to neoadjuvant therapy. Patients with ASS1 loss or low expression were significantly younger. Conclusions: Our findings indicate that approximately 12.4% of patients with esophageal adenocarcinoma may be eligible and could potentially benefit from arginine deprivation therapy. This underscores the urgent need for clinical trials evaluating the efficacy of pegargiminase in this patient population. Additionally, incorporating ASS1 immunohistochemical staining into pre-neoadjuvant biopsy assessments should be considered to optimize neoadjuvant treatment strategies and advance the implementation of personalized cancer therapy. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

13 pages, 962 KB  
Article
SARIFA Is Associated with Lymph Node Metastases in PT3 and PT4 Gastric Cancers
by Krešimir Mustapić, Petar Đolonga, Tomislav Ivanović, Ana Paparella Karaman, Luka Minarik, Katarina Vukojević and Merica Glavina Durdov
Cancers 2025, 17(21), 3593; https://doi.org/10.3390/cancers17213593 - 6 Nov 2025
Viewed by 308
Abstract
Background/Objectives: Advanced gastric cancer usually has an unfavorable prognosis. Stroma AReactive Invasion Front Area (SARIFA) is a newly recognized biomarker of aggressiveness, easily recognized as five tumor cells in direct contact with adipocytes in perigastric, submucosal, and perivascular adipose tissue. We investigated [...] Read more.
Background/Objectives: Advanced gastric cancer usually has an unfavorable prognosis. Stroma AReactive Invasion Front Area (SARIFA) is a newly recognized biomarker of aggressiveness, easily recognized as five tumor cells in direct contact with adipocytes in perigastric, submucosal, and perivascular adipose tissue. We investigated this phenomenon and correlated it with other pathohistological variables. Material and Methods: The sample includes 102 Croatian patients with locally advanced gastric cancer, who underwent total gastrectomy/lymphadenectomy between 2012–2018 and in 2023 at University Hospital Split, Croatia, and had pathological stage pT3 or pT4. Representative histological specimens were analyzed for SARIFA, and results were compared with other variables and overall survival. External validation and gene expression analysis of CD36 and FABP4 were performed using the TCGA-STAD cohort. Results: SARIFA was significantly associated with positive pN status (p = 0.009) and perineural invasion (p = 0.043). Patients with SARIFA had a more than fivefold increased risk of nodal involvement (OR = 6.35; 95% CI: 1.35–29.84; p = 0.019). Lymphovascular invasion (LVI) was associated with nodal disease (OR = 4.39; 95% CI: 1.194–16.143; p = 0.026), and SARIFA was marginally associated (OR = 4.886; 95% CI: 0.985–24.241; p = 0.052). Patients who had both LVI and SARIFA had a higher proportion of affected lymph nodes (p = 0.009). SARIFA status did not significantly affect overall survival. Gene expression analysis showed a significant increase in CD36 expression, while FABP4 expression was elevated but not statistically significant, in SARIFA-positive cases. Conclusions: SARIFA could be used as a marker for invasiveness and further investigated due to its predictive potential. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

13 pages, 1991 KB  
Article
Revisiting p53 Immunohistochemical Staining and Its Prognostic Implications in Advanced EGFR-Mutated Lung Adenocarcinoma
by Feng-Che Kuan, Shun-Fu Chang, Yao-Ren Yang, Yu-Ying Wu, Fen-Fen Chen, Kam-Fai Lee, Chen-Lin Chi, Meng-Hung Lin and Chung-Sheng Shi
Cancers 2025, 17(21), 3577; https://doi.org/10.3390/cancers17213577 - 5 Nov 2025
Viewed by 375
Abstract
Background/Objectives: TP53 mutations in advanced epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer could worsen prognosis. Therefore, we aimed to investigate the clinical significance of TP53 mutations and p53 expression in these patients. Methods: Patients with advanced/metastatic EGFR-mutated lung adenocarcinoma treated with [...] Read more.
Background/Objectives: TP53 mutations in advanced epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer could worsen prognosis. Therefore, we aimed to investigate the clinical significance of TP53 mutations and p53 expression in these patients. Methods: Patients with advanced/metastatic EGFR-mutated lung adenocarcinoma treated with first-line tyrosine kinase inhibitors were retrospectively enrolled. Sanger sequencing was performed to detect TP53 mutations and immunohistochemical staining was used to verify p53 protein expression levels. Kaplan-Meier and Cox proportional hazards analyses were used to estimate survival and hazard ratio (HR) with 95% confidence interval (CI). Results: The study involved 83 patients with adequate tumor samples for TP53/p53 analysis. Patients with tumor p53 immunostaining ≥50% showed significantly better overall survival (OS) (HR: 0.49 [95% CI: 0.30–0.81], p < 0.001), but TP53 mutations were not associated with inferior progression-free survival (PFS) or OS (missense vs. wild-type [PFS, HR: 0.68 (95% CI: 0.40–1.15), p = 0.151; OS, HR: 0.88 (95% CI: 0.56–1.42), p = 0.599]). Areas under the receiver operating characteristic curves of TP53 mutations with different cut-off values for p53 positivity were 0.51–0.56. The Kaplan-Meier survival analysis revealed significant survival benefits in patients with EGFR L858R substitution and tumor p53 immunostaining ≥50% (median PFS: 8.0 vs. 5.3; median OS: 20.4 vs. 15.3 months; log-rank p = 0.025 and 0.049, respectively). Conclusions: Tumor p53 immunostaining (≥50%) was associated with better OS, especially in patients with TP53 mutations or L858R. Prospective clinical trials are required to explore the prognostic significance of p53 expression in the genomic era of TP53 mutations. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

24 pages, 1042 KB  
Review
Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers
by Neda Dadgar, Muhammad Anees, Christopher Sherry, Hyun Young Park, Erin E. Grayhack, Arul Goel, Alisha F. Khan, Ashten Omstead, David L. Bartlett, Patrick L. Wagner and Ali H. Zaidi
Cancers 2025, 17(21), 3552; https://doi.org/10.3390/cancers17213552 - 2 Nov 2025
Viewed by 627
Abstract
Gastroesophageal cancers including esophageal and gastric cancer remain major causes of global cancer mortality, primarily due to late diagnosis and high recurrence rates after curative treatment. Current surveillance methods, such as endoscopy and imaging, are invasive, costly, and often inadequate for detection. Blood-based [...] Read more.
Gastroesophageal cancers including esophageal and gastric cancer remain major causes of global cancer mortality, primarily due to late diagnosis and high recurrence rates after curative treatment. Current surveillance methods, such as endoscopy and imaging, are invasive, costly, and often inadequate for detection. Blood-based biomarkers (“liquid biopsies”) offer a minimally invasive alternative capable of real-time tumor monitoring. In this review, we summarize recent advances across all major classes of blood-derived biomarkers: circulating tumor DNA (ctDNA), methylated DNA, cell-free RNAs (microRNAs, lncRNAs, circRNAs), circulating proteins, autoantibodies, circulating tumor cells, extracellular vesicles, and metabolites. Reviewing the existing literature on gastroesophageal cancers, we highlight current evidence, validation phases, performance metrics, and limitations. Special attention is given to clinical trial evidence, including ctDNA monitoring studies, that demonstrated earlier recurrence detection compared to imaging. While blood-based biomarker analysis has not yet supplanted endoscopy as standard of care in gastroesophageal cancer surveillance, the convergence of multi-analyte assays, AI, and clinical validation trials positions liquid biopsy as a transformative tool in the surveillance of gastroesophageal cancers. Full article
Show Figures

Figure 1

20 pages, 32904 KB  
Article
Identification of Phenotypically Distinct Cancer Stem Cell Subpopulations in Oral Squamous Cell Carcinoma
by Tarig Al-Hadi Osman, Oddveig Rikardsen, Muy-Teck Teh, Dipak Sapkota, Kristina Xiao Liang, Evelyn Neppelberg, Adrian Biddle, Ian Mackenzie, Lars Uhlin-Hansen, Anne Christine Johannessen and Daniela Elena Costea
Cancers 2025, 17(21), 3547; https://doi.org/10.3390/cancers17213547 - 1 Nov 2025
Viewed by 492
Abstract
Background: Several markers have been shown to define subpopulations enriched for cancer stem cells (CSCs) and correlate with poor clinical outcomes in oral squamous cell carcinoma (OSCC). Objective: To investigate the pattern of expression and correlation with clinical parameters of two CSC markers, [...] Read more.
Background: Several markers have been shown to define subpopulations enriched for cancer stem cells (CSCs) and correlate with poor clinical outcomes in oral squamous cell carcinoma (OSCC). Objective: To investigate the pattern of expression and correlation with clinical parameters of two CSC markers, namely p75NTR and ALDH1A1, in both patient samples and cell lines. Methods: Archival formalin-fixed paraffin-embedded samples from normal human oral mucosa (NHOM, n = 31), oral dysplasia (OD, n = 10) and OSCC (n = 177) were subjected to multiple immunohistochemistry and some to qRT-PCR for expression of CSC and proliferation-related markers, BMI1 and Ki67. Correlations between CSC marker expression and clinical parameters were investigated. Primary cells and cell lines derived from NHOM, OD or OSCC were FACS-analyzed for the same markers. Results: A higher frequency of cells positive for CSC markers was detected in OD and OSCC compared to NHOM. Co-localization of the two markers was a rare finding in OSCC as compared to NHOM or OD and was more heterogeneous in OSCC cell lines than in OD and NHOM cells. Cells positive for p75NTR exhibited higher expression of proliferative and self-renewal markers in comparison to ALDH1A1+ or double ALDH1A1+/p75NTR+ cells. Cells positive for p75NTR were more frequent in small-size tumors and poorly to moderately differentiated tumors and correlated with poor survival of patients otherwise (clinically) deemed as of better prognosis. Higher frequency of ALDH1A1+ cells was found to be associated with lymph node metastasis. Both p75NTR+ cells and ALDH1A1+ cells could emerge de novo from the respective negative subpopulation after FACS and in vitro growth, but with different kinetics. Conclusions: Here, we show that several stem cell subpopulations with distinct phenotypes co-exist in a tumor, each having impact on different clinical parameters. The cell subpopulations identified by the use of different CSC markers were found to be dynamic populations, able to switch between phenotypes. In addition, our data suggest that the stem cell heterogeneity is acquired and evolves parallel with carcinoma progression. Full article
(This article belongs to the Special Issue Molecular and Genetic Biomarkers in Oral Squamous Cell Carcinoma)
Show Figures

Figure 1

13 pages, 2607 KB  
Article
SMARCD3 Promotes Epithelial–Mesenchymal Transition in Gastric Cancer by Integrating PI3K-AKT and WNT/β-Catenin Pathways
by Ji-Ho Park, Sun Yi Park, Eun-Jung Jung, Young-Tae Ju, Chi-Young Jeong, Ju-Yeon Kim, Taejin Park, Miyeong Park, Young-Joon Lee and Sang-Ho Jeong
Cancers 2025, 17(21), 3526; https://doi.org/10.3390/cancers17213526 - 31 Oct 2025
Viewed by 314
Abstract
Background: Epithelial–mesenchymal transition (EMT) is a fundamental process that drives invasion and metastasis in patients with diffuse-type gastric cancer (DGC). The role of SMARCD3, a subunit of the SWI/SNF chromatin remodeling complex, in this process is largely unknown. The aim of this [...] Read more.
Background: Epithelial–mesenchymal transition (EMT) is a fundamental process that drives invasion and metastasis in patients with diffuse-type gastric cancer (DGC). The role of SMARCD3, a subunit of the SWI/SNF chromatin remodeling complex, in this process is largely unknown. The aim of this study is to elucidate the molecular mechanism through which SMARCD3 integrates with the PI3K-AKT and WNT/β-catenin signaling pathways to promote EMT and gastric cancer progression. Methods: Stable SMARCD3-overexpressing MKN45 and MKN74 cell lines were established. RNA sequencing (RNA-seq) was performed to investigate signaling alterations. Western blot analysis confirmed the expression of EMT markers (Snail and Slug) and the phosphorylation of AKT (Ser473) and GSK3β (Ser9). PI3K dependency was tested using the inhibitor LY294002. Cooperative effects were examined by activating the WNT pathway with WNT3A. Results: SMARCD3 overexpression upregulated PI3K-AKT and WNT signaling, which correlated with increased Snail/Slug expression and increased AKT/GSK3β phosphorylation. GSK3β inactivation (pSer9) stabilizes Snail, driving EMT. LY294002 treatment suppressed Snail/Slug expression, attenuated AKT activation, and reversed the mesenchymal phenotype. Furthermore, WNT3A treatment synergistically increased nuclear Snail accumulation. Conclusions: SMARCD3 acts as a critical epigenetic regulator that promotes EMT in patients with gastric cancer through the integration of the PI3K-AKT and WNT/β-catenin pathways. Targeting this SMARCD3-mediated mechanism offers a promising therapeutic strategy to inhibit metastasis and improve outcomes for patients with gastric cancer. Full article
(This article belongs to the Special Issue Advancements in “Cancer Biomarkers” for 2025–2026)
Show Figures

Figure 1

46 pages, 10549 KB  
Review
Detection of Protein and Metabolites in Cancer Analyses by MALDI 2000–2025
by Dorota Bartusik-Aebisher, Daniel Roshan Justin Raj and David Aebisher
Cancers 2025, 17(21), 3524; https://doi.org/10.3390/cancers17213524 - 31 Oct 2025
Viewed by 655
Abstract
Cancer metabolomics has become a powerful way of understanding tumor biology, identifying biomarkers and metabolites, and helping precision oncology. Matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), among many other analytical platforms, has gained popularity over the past two and a half decades due to [...] Read more.
Cancer metabolomics has become a powerful way of understanding tumor biology, identifying biomarkers and metabolites, and helping precision oncology. Matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), among many other analytical platforms, has gained popularity over the past two and a half decades due to its unique ability of directly analyzing metabolites in tissue with spatial resolution. This review will study 2000–2025 MALDI-based strategies for cancer metabolite detection, spanning from early proof-of-concept protein profiling to the development of high-resolution MALDI-MS imaging (MALDI-MSI), which is capable of mapping thousands of metabolites at near single-cell resolution. Its applications include the differentiation of tumor versus normal tissue, discovery of stage and subtype specific biomarkers, mapping of metabolic heterogeneity, and the visualization of drug metabolism in situ. Breakthrough technological milestones, such as the advanced matrices, on-tissue derivatization, MALDI-2 post-ionization, and the integration with Orbitrap or Fourier-transform ion cyclotron resonance (FT-ICR) platforms, have significantly improved the overall sensitivity, metabolite coverage, and spatial fidelity. Clinically, MALDI-MS has shown its purpose in breast, prostate, colorectal, lung, and liver cancers by providing metabolic fingerprints that are linked to tumor microenvironments, hypoxia, and therapeutic response. However, challenges such as the inclusion of matrix interface with low-mass metabolites, limited quantitation, ion suppression, and the lack of standardized procedures do not yet allow for the transition from translation to routine diagnostics. Even with these hurdles, the future of MALDI-MS in oncology remains in a good position with major advancements in multimodal imaging, machine learning-based data integration, portable sampling devices, and clinical validation studies that are pushing the field towards precision treatment. Full article
(This article belongs to the Special Issue New Biomarkers in Cancers 2nd Edition)
Show Figures

Figure 1

28 pages, 1759 KB  
Review
Liquid Biopsy in Lung Cancer: Tracking Resistance to Targeted Therapies
by Scarlet B. Urtecho, Beatriz Jimenez Munarriz, Mary R. Rabey and Natasha B. Leighl
Cancers 2025, 17(21), 3474; https://doi.org/10.3390/cancers17213474 - 29 Oct 2025
Viewed by 1062
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related mortality worldwide. While target therapies have changed the outcomes of patients harboring actionable mutations, resistance inevitably emerges. Circulating tumor DNA (ctDNA) offers a minimally invasive tool for capturing tumor evolution in real [...] Read more.
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related mortality worldwide. While target therapies have changed the outcomes of patients harboring actionable mutations, resistance inevitably emerges. Circulating tumor DNA (ctDNA) offers a minimally invasive tool for capturing tumor evolution in real time. This approach enables the rapid detection of resistance mechanisms, complements or substitutes for tissue re-biopsy, and reduces the burden of invasive procedures for patients. In this review, we summarize the current evidence on the use of liquid biopsy to uncover resistance mechanisms in patients progressing on targeted therapies, with a focus on its role in dynamic tumor profiling and longitudinal disease monitoring. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

Back to TopTop