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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (990)

Search Parameters:
Keywords = tumour immunity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 1537 KB  
Review
Comparative Insights into Cutaneous, Mucosal, and Vulvovaginal Melanomas: Biology, Targeted Therapies, and Survival with a Focus on Immune Checkpoint Inhibitors
by Danielle Christmas, Christina Pappa, Catherine Howell, Mohammad Daas, Keith Howell, Sunanda Dhar, Binamra Sigdel, Sanjiv Manek and Moiad Alazzam
J. Pers. Med. 2025, 15(11), 551; https://doi.org/10.3390/jpm15110551 - 12 Nov 2025
Abstract
Background/Objectives: Melanoma is a malignant tumour of melanocytes. Cutaneous melanoma accounts for the vast majority of cases and has benefitted from advances in targeted and immune checkpoint inhibitor therapies, leading to substantial improvements in survival. In contrast, mucosal and vulvovaginal melanomas are [...] Read more.
Background/Objectives: Melanoma is a malignant tumour of melanocytes. Cutaneous melanoma accounts for the vast majority of cases and has benefitted from advances in targeted and immune checkpoint inhibitor therapies, leading to substantial improvements in survival. In contrast, mucosal and vulvovaginal melanomas are rare, aggressive subtypes with distinct molecular and immune profiles and poor prognoses. This review synthesises evidence comparing cutaneous, mucosal, and vulvovaginal melanoma, with emphasis on biology, treatment, and outcomes Methods: A narrative comparative review was undertaken, examining the published literature on the epidemiology, molecular and immune characteristics, and treatment outcomes of cutaneous, mucosal, and vulvovaginal melanoma, including systemic therapies and surgical approaches. Results: Cutaneous melanoma demonstrates high tumour mutational burden and frequent BRAF and NRAS mutations, underpinning the success of targeted therapy and immunotherapy. Mucosal and vulvovaginal melanomas exhibit lower mutational burden, distinct mutation patterns, and reduced immunogenicity, correlating with poorer treatment responses. Surgery remains the mainstay of management, though optimal margins in vulvovaginal melanoma are unclear. Recurrence rates are high, and five-year survival remains poor. Evidence for systemic therapy is limited to small retrospective cohorts and subgroup analyses, showing lower response and survival rates compared with cutaneous melanoma. Chemotherapy has minimal benefit. Conclusions: Mucosal and vulvovaginal melanomas are biologically and clinically distinct from cutaneous melanoma and continue to have poor survival outcomes. Their rarity restricts high-quality evidence, highlighting the need for collaborative, innovative research to inform effective treatment strategies. Full article
(This article belongs to the Special Issue Clinical Updates and Challenges of Gynecological Cancers)
Show Figures

Figure 1

48 pages, 3688 KB  
Review
The Role of Porphyromonas gingivalis in Oral Carcinogenesis and Progression by Remodelling the Tumour Microenvironment: A Narrative Review
by Katarzyna Starska-Kowarska
Cancers 2025, 17(21), 3478; https://doi.org/10.3390/cancers17213478 - 29 Oct 2025
Viewed by 356
Abstract
(1) Background: Oral squamous cell carcinoma (OSCC) is the most common type of head and neck malignancy worldwide. Despite the prevalence of modern diagnostic and prognostic techniques, late diagnosis and resistance to treatment still result in a low 5-year survival rate, high recurrence [...] Read more.
(1) Background: Oral squamous cell carcinoma (OSCC) is the most common type of head and neck malignancy worldwide. Despite the prevalence of modern diagnostic and prognostic techniques, late diagnosis and resistance to treatment still result in a low 5-year survival rate, high recurrence rate, and frequent malignant metastases. Increasing evidence indicates that bacteria of the oral microbiome, such as the Gram-negative anaerobic Porphyromonas gingivalis, may play a crucial role in the initiation and development of OSCC by inducing periodontitis. Indeed, epithelial-to-mesenchymal transition (EMT) and dysregulated immune response have been attributed to the activity of a dysbiotic microbiota. This comprehensive review examines the influence of P. gingivalis on oral carcinogenesis and progression, which has been associated with tumour microenvironment remodelling and the dysregulation of key signalling pathways related to epithelial-to-mesenchymal transition (EMT), cell-cycling, autophagy, and apoptosis. (2) Methods: The article reviews current literature on the possible role of P. gingivalis and induced dysbiosis, periodontitis and a pro-inflammatory environment as key mechanisms driving neoplastic epithelial changes and chemoresistance to anticancer agents in patients with OSCC; the research corpus was acquired from the Pub-Med/Medline/EMBASE/Cochrane Library databases. (3) Results: The identification of virulence factors and key mechanisms used by P. gingivalis to promote the development and progression of OSCC may support traditional diagnostic methods and factors related to treatment response and prevention of OSCC. (4) Conclusions: Emerging evidence suggests a possible association between periodontal bacteria and oral carcinogenesis. P. gingivalis may be an important potential target for future strategies aimed at treating oral cancer. Full article
Show Figures

Figure 1

21 pages, 879 KB  
Review
New Horizons with Growth Differentiation Factor 15 in Oncology: From Cancer Cachexia and Tumour Immunity to Novel Therapeutic Strategies
by Keiji Sugiyama, Naureen Starling and Ian Chau
Curr. Oncol. 2025, 32(11), 604; https://doi.org/10.3390/curroncol32110604 - 29 Oct 2025
Viewed by 572
Abstract
Growth differentiation factor 15 (GDF-15) is a stress-induced cytokine produced by tumour cells and peripheral cells. It is implicated in the development of cancer cachexia, a debilitating condition for which no effective pharmacological therapy currently exists. GDF-15 regulates appetite and metabolic processes through [...] Read more.
Growth differentiation factor 15 (GDF-15) is a stress-induced cytokine produced by tumour cells and peripheral cells. It is implicated in the development of cancer cachexia, a debilitating condition for which no effective pharmacological therapy currently exists. GDF-15 regulates appetite and metabolic processes through complex neural and hormonal networks. Furthermore, it has been implicated in chemotherapy-induced nausea and vomiting, representing a potential therapeutic target. GDF-15 negatively affects tumour immunity, suggesting that anti-GDF-15 therapy could potentially enhance immune responses and help overcome resistance to immunotherapy. Recently, early clinical trials have reported preliminary results of GDF-15-targeted therapies in alleviating cancer cachexia and potentially enhancing the efficacy of immunotherapy. This review aims to provide an overview of the role of GDF-15 in cancer cachexia, including the underlying neural mechanisms and their involvement in tumour immunity. This review also summarises recent clinical trial findings and discusses future perspectives on GDF-15-targeted therapy in oncology, offering important insights for future research. Full article
Show Figures

Figure 1

22 pages, 8382 KB  
Article
A Prognostic Neuromodulation-Related Gene Signature Identifies Immunomodulation and Tumour-Associated Hallmarks in Glioblastoma
by Min Yee Chow, Sylvia Sue Xian Liew, Mastura Monif, Muhamad Noor Alfarizal Kamarudin and Brandon Wee Siang Phon
Biomedicines 2025, 13(11), 2640; https://doi.org/10.3390/biomedicines13112640 - 28 Oct 2025
Viewed by 407
Abstract
Background and Objective: Neuromodulators such as neuropeptide, neurotrophic factors and neurotransmitters are increasingly reported to be involved in glioblastoma (GBM) progression. Nonetheless, the association between neuromodulation-related genes (NMRGs) and GBM prognosis remains elusive. Hence, this study aims to identify clinically significant NMRGs [...] Read more.
Background and Objective: Neuromodulators such as neuropeptide, neurotrophic factors and neurotransmitters are increasingly reported to be involved in glioblastoma (GBM) progression. Nonetheless, the association between neuromodulation-related genes (NMRGs) and GBM prognosis remains elusive. Hence, this study aims to identify clinically significant NMRGs that can form a prognostic gene signature for GBM patients. Methods and Results: Differential expression analysis of transcriptomic profiles extracted from GSE147352, GSE165595, TCGA and CGGA determined 272 differentially expressed NMRGs (deNMRGs) in GBM compared to normal brain tissue. The subsequent Kaplan–Meier survival analysis and Cox proportional hazard model further identified ten common deNMRGs (IGF2, RETN, EDNRB, C3AR1, CLCF1, NTRK1, OSMR, KCNN4, SLC18A3 and HTR7), forming a 10-NMRG signature. This signature stratifies GBM patients and consistently predicts poorer survival outcomes for the high-risk score group compared to the low-risk score group in the TCGA and CGGA cohorts. The gene set enrichment analysis and active-subnetwork-oriented enrichment analysis identified a connection between immunomodulation and tumour-associated hallmarks with the high-risk GBM patient group. Next, the correlation proportionality analysis identified a positive association between the signature genes with immune activators, immune suppressors and pro-motility genes. Additionally, high expressions of the 10-NMRGs were noted in the mesenchymal GBM subtype. Conclusions: Collectively, our analysis highlights the potential use of the 10-NMRG signature to stratify the high-risk GBM group with a strong association of immunomodulation and tumour-associated hallmarks that can contribute to the poor survival outcomes. Full article
(This article belongs to the Special Issue Epigenetic Regulation in Cancer Progression)
Show Figures

Graphical abstract

18 pages, 4151 KB  
Article
The Effects of PAK-Regulated Tumour Vasculature on Gemcitabine Response of Pancreatic Cancer
by Arian Ansardamavandi, Chelsea Dumesny, Yi Ma, Li Dong, Sarah Ellis, Ching-Seng Ang, Mehrdad Nikfarjam and Hong He
Cancers 2025, 17(21), 3434; https://doi.org/10.3390/cancers17213434 - 26 Oct 2025
Viewed by 289
Abstract
Background/Objectives: The tumour microenvironment in pancreatic ductal adenocarcinoma (PDA) is highly complex, influencing both vascular function and therapy response. P21-activated kinases (PAKs) are key regulators of the cellular and immune system, but the specific roles of PAK1 and PAK4 in pancreatic tumour [...] Read more.
Background/Objectives: The tumour microenvironment in pancreatic ductal adenocarcinoma (PDA) is highly complex, influencing both vascular function and therapy response. P21-activated kinases (PAKs) are key regulators of the cellular and immune system, but the specific roles of PAK1 and PAK4 in pancreatic tumour vasculature and chemotherapy sensitivity are unclear. This study investigated the effects of PAK1 and PAK4 on tumour vasculature and therapeutic response in an immunocompromised mouse model. Methods: KPC-derived wild type (WT), PAK1 knockout (KO), PAK4KO, or PAK1&4KO pancreatic cancer cells were injected subcutaneously into SCID mice, followed by gemcitabine treatment. Tumour growth, vessel density, pericyte coverage, and endothelial adhesion molecule expression were analysed by histology and immunostaining. A proteomic study was used to identify protein changes. Results: PAK1KO significantly reduced tumour growth, enhanced vascular normalisation, upregulated stromal ICAM-1 and VCAM-1, but reduced gemcitabine efficacy. PAK4KO did not inhibit tumour growth but increased vessel diameter and enhanced gemcitabine efficacy. Proteomics study indicated that PAK1KO downregulated proteins involved in the VEGF pathway, while PAK4KO upregulated most proteins involved in the VEGF pathway and downregulated DNA repair proteins, contributing to improved chemotherapy sensitivity. The double knockout of PAK1 and PAK4 did not inhibit tumour growth, although it stimulated vascular normalisation, indicating an outcome balanced between PAK1 and PAK4. Conclusions: PAK1 and PAK4 differentially regulated pancreatic tumour vasculature and chemotherapy response. PAK1KO suppressed tumour growth by reducing angiogenesis and enhancing vascular normalisation, whereas PAK4KO enhanced gemcitabine efficacy through vessel dilation. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

18 pages, 4090 KB  
Article
Expression of TIM-3 and Gal-9 Immune Checkpoints in Chronic Lymphocytic Leukemia: The Potential Role of Interleukin-27
by Ewelina Wędrowska, Tomasz Wandtke, Bartosz Ulaszewski, Edyta Cichocka, Robert Dębski, Piotr Kopiński, Jan Styczyński and Grzegorz Przybylski
Curr. Issues Mol. Biol. 2025, 47(11), 881; https://doi.org/10.3390/cimb47110881 - 23 Oct 2025
Viewed by 325
Abstract
Background: Chronic lymphocytic leukemia (CLL) is characterized by malignant B lymphocyte accumulation and progressive immune dysfunction. The immune checkpoint molecule TIM-3 and its ligand galectin-9 (Gal-9) contribute to T cell exhaustion, impairing anti-tumour immunity. Interleukin-27 (IL-27) has pleiotropic immunomodulatory properties, but its impact [...] Read more.
Background: Chronic lymphocytic leukemia (CLL) is characterized by malignant B lymphocyte accumulation and progressive immune dysfunction. The immune checkpoint molecule TIM-3 and its ligand galectin-9 (Gal-9) contribute to T cell exhaustion, impairing anti-tumour immunity. Interleukin-27 (IL-27) has pleiotropic immunomodulatory properties, but its impact on TIM-3 and Gal-9 expression in CLL remains unclear. Methods: Peripheral blood mononuclear cells (PBMCs) from 20 treatment-naive CLL patients were cultured with or without IL-27 (100 ng/mL) for 72 h. Flow cytometry assessed TIM-3 and Gal-9 expression on CD4+, CD8+, and CD19+ cells. Results: IL-27 stimulation significantly increased TIM-3 expression on CD8+ T cells (2.18 ± 0.32% vs. 3.09 ± 0.49%, p = 0.009), a hallmark of T cell exhaustion. IL-27 also modestly increased intracellular Gal-9 levels in total lymphocytes (93.91 ± 1.17% vs. 96.55 ± 0.67%, p = 0.005). Additionally, IL-27 reduced CD4+ T cell proportions (26.71 ± 4.19% vs. 22.01 ± 3.23%, p = 0.010). Although numerically modest, these changes may be biologically pertinent in the context of checkpoint-mediated CD8+ T-cell exhaustion. Conclusions: IL-27 may enhance immunosuppressive mechanisms in CLL by modulating immune checkpoint expression, potentially contributing to disease progression. These ex vivo findings in PBMCs from CLL patients indicate the IL-27-associated modulation of checkpoint expression under the conditions tested. In the absence of parallel healthy-donor controls, CLL specificity cannot be established in this study. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

21 pages, 584 KB  
Review
Beyond Imaging: Integrating Radiomics, Genomics, and Multi-Omics for Precision Breast Cancer Management
by Xiaorong Wu and Wei Dai
Cancers 2025, 17(21), 3408; https://doi.org/10.3390/cancers17213408 - 23 Oct 2025
Viewed by 819
Abstract
Radiomics has emerged as a promising tool for non-invasive tumour phenotyping in breast cancer, providing valuable insights into tumour heterogeneity, response prediction, and risk stratification. However, traditional radiomic approaches often rely on correlative patterns of image analysis to clinical data and lack direct [...] Read more.
Radiomics has emerged as a promising tool for non-invasive tumour phenotyping in breast cancer, providing valuable insights into tumour heterogeneity, response prediction, and risk stratification. However, traditional radiomic approaches often rely on correlative patterns of image analysis to clinical data and lack direct biological interpretability. Combining information provided by radiomics with genomics or other multi-omics data can be important to personalise diagnostic and therapeutic work up in breast cancer management. This review aims to explore the current progress in integrating radiomics with multi-omics data—genomics and transcriptomics—to establish biologically grounded, multidimensional models for precision management of breast cancer. We will review recent advances in integrative radiomics and radiogenomics, highlight the synergy between imaging and molecular profiling, and discuss emerging machine learning methodologies that facilitate the integration of high-dimensional data. Applications of radiogenomics, including breast cancer subtype and molecular mutation prediction, radiogenomic mapping of the tumour immune microenvironment, and response forecasting to immunotherapy and targeted therapies, as well as lymph nodes involvement, will be evaluated. Challenges in technical limitations including imaging modalities harmonization, interpretability, and advancing machine learning methodologies will be addressed. This review positions integrative radiogenomics as a driving force for next-generation breast cancer care. Full article
(This article belongs to the Special Issue Radiomics in Cancer)
Show Figures

Figure 1

20 pages, 3046 KB  
Article
ASMase Activation in Ultrasound-Stimulated Radiation Enhancement Using MRI-Guided Focused Ultrasound
by Tera N. Petchiny, Deepa Sharma, Anoja Giles, Kai Xuan Leong, Wenyi Yang, Lakshmanan Sannachi, David Alberico and Gregory J. Czarnota
Cells 2025, 14(20), 1618; https://doi.org/10.3390/cells14201618 - 17 Oct 2025
Viewed by 425
Abstract
Focused ultrasound-stimulated microbubble (MB + FUS) therapy is a promising radiation enhancement strategy, utilizing vascular disruption to enhance radiation efficacy. However, its mechanistic effects in large tumour volumes and clinical translatability remain insufficiently characterized. This study evaluates the synergistic impact of MB + [...] Read more.
Focused ultrasound-stimulated microbubble (MB + FUS) therapy is a promising radiation enhancement strategy, utilizing vascular disruption to enhance radiation efficacy. However, its mechanistic effects in large tumour volumes and clinical translatability remain insufficiently characterized. This study evaluates the synergistic impact of MB + FUS combined with radiation therapy (XRT) in a large-scale, immunosuppressed rabbit xenograft model using a clinically adaptable, MRI-guided 6144-element focused ultrasound (MRgFUS) system. Tumours were treated with MB + FUS, XRT, or both, with real-time image-guided MB activation and evaluation of treatment effects on vascular integrity, proliferation, and cellular stress responses. Assessments included Power Doppler ultrasound, histology, and immunohistochemistry targeting TUNEL, ASMase, Ki-67, Factor VIII, HIF-1α, and VEGF. Combination therapy induced significant vascular collapse, reduced perfusion, and decreased Factor VIII expression, alongside increased nuclear condensation, TUNEL positivity, and ASMase expression, consistent with ASMase-mediated endothelial apoptosis and vascular disruption. Upregulation of HIF-1α and VEGF indicated hypoxia-driven angiogenic signalling, while Ki-67 suppression reflected sustained tumour growth inhibition. Although immune responses were limited by host immunosuppression, the larger tumour burden provided clinically relevant constraints. The MRgFUS platform enabled precise and reproducible MB activation, reinforcing MB + FUS as a potent radio-enhancement modality. These findings support the continued development of MB + FUS toward clinical translation and highlight its potential as a complementary strategy to radiation therapy. Full article
Show Figures

Figure 1

22 pages, 3624 KB  
Article
The Immunomodulatory Role of Gemcitabine in Triple Negative Breast Cancer
by Cory Fines, Syed Umbreen, Elaine Gilmore, Helen McCarthy and Niamh Buckley
Cells 2025, 14(20), 1604; https://doi.org/10.3390/cells14201604 - 16 Oct 2025
Viewed by 480
Abstract
Triple negative breast cancer (TNBC), defined for its lack of expression/amplification of three major receptors, makes up ~15% of all BC cases but a majority of all BC deaths. TNBC has been found to be the most immune-rich among BC subtypes, and progress [...] Read more.
Triple negative breast cancer (TNBC), defined for its lack of expression/amplification of three major receptors, makes up ~15% of all BC cases but a majority of all BC deaths. TNBC has been found to be the most immune-rich among BC subtypes, and progress has been made in the development of immunotherapies; however, not all patients are eligible, and response can be limited. Therefore, there is a significant clinical need to enhance the response to these treatments. Given chemotherapy is the core component of TNBC treatment, and is given in combination with immunotherapy, its potential immunomodulatory impact warrants exploration. Gemcitabine, currently used for the treatment of metastatic TNBC, has been reported to have potential immunomodulatory properties that create a more immune-favourable TME for combination with immunotherapies and/or improved outcome. We therefore investigated the use of gemcitabine as an immunomodulator in a primary 4T1 TNBC mouse model. Gemcitabine was able to reduce pro-tumour immune cells including macrophages and MDSCs while increasing T-cell abundance, therefore resulting in a less immunosuppressive TME. We demonstrated that this immune response was both temporal and dose-dependent, which has impact for planning and scheduling combination treatments. In conclusion, we have demonstrated that gemcitabine modulates the TME in ways that could not only enhance the direct anti-tumour effects of gemcitabine itself but also potentially enhance responsiveness to immunotherapy. This work has laid the foundation for further studies investigating combination therapy for the treatment of TNBC. Full article
Show Figures

Figure 1

20 pages, 5082 KB  
Article
Oncolytic Maraba Virus MG1 Mediates Direct and Natural Killer Cell-Dependent Lysis of Ewing Sarcoma
by Tyler Barr, Victoria A. Jennings, Elizabeth A. Roundhill, Richard T. Baugh, Maisa Yamrali, Heather E. Owston, Dennis McGonagle, Peter V. Giannoudis, Natasha J. Caplen, Javed Khan, John C. Bell, Susan A. Burchill, Fiona Errington-Mais and Graham P. Cook
Cancers 2025, 17(20), 3319; https://doi.org/10.3390/cancers17203319 - 14 Oct 2025
Viewed by 526
Abstract
Background: Ewing sarcoma (EWS) is a rare cancer of the bone and soft tissue, most prevalent in children and young adults. The treatment of EWS has progressed relatively little in over 30 years. Survival rates for patients, particularly those with metastatic and/or relapsed [...] Read more.
Background: Ewing sarcoma (EWS) is a rare cancer of the bone and soft tissue, most prevalent in children and young adults. The treatment of EWS has progressed relatively little in over 30 years. Survival rates for patients, particularly those with metastatic and/or relapsed disease remain poor, highlighting the urgent need for innovative treatment options. Methods: Here, we have explored the therapeutic potential of the oncolytic Maraba virus strain MG1 using various in vitro models of EWS, including established cell lines, doxorubicin-resistant derivatives, spheroid cultures and primary patient-derived Ewing sarcoma cell cultures. We examined the direct oncolytic activity of MG1 and its ability to stimulate the immune-mediated killing of EWS by human healthy donor peripheral blood mononuclear cells. Results: We show that MG1 undergoes productive replication and exerts direct oncolysis of established EWS cell lines, doxorubicin-resistant EWS cell lines and patient-derived Ewing sarcoma cell cultures more recently established from tumours. In contrast, primary mesenchymal stem cells (the likely cell of origin of EWS) were resistant to MG1, with IFN-I being a major determinant of tumour cell selectivity. MG1-treated PBMC produced IFN-I and killed EWS cells in vitro, in a natural killer (NK) cell-dependent manner. Conclusions: The ability of MG1 to kill EWS cells directly and stimulate NK cell cytotoxicity against this tumour suggests that MG1 may provide therapeutic benefit for EWS patients where the efficacy of conventional treatments is currently limited. Full article
(This article belongs to the Special Issue Advances in Soft Tissue and Bone Sarcoma (2nd Edition))
Show Figures

Graphical abstract

31 pages, 25829 KB  
Article
The Hepatoprotective Properties of the Revised Formulation of Dahuang Xiaoshi Tang, an Ancient Chinese Herbal Decoction, Are Probed by Integrated Metabolomics and Network Pharmacology
by Xiangpeng Kong, Xiaoyang Wang, Haiqin Ren, Yajun Yao, Hui Zhang, Huifeng Li, Huifang Li, Yangang Cheng, Zhuqing Song, Miaorong Pei and Karl Wah Keung Tsim
Pharmaceuticals 2025, 18(10), 1534; https://doi.org/10.3390/ph18101534 - 13 Oct 2025
Viewed by 799
Abstract
Background: Dahuang Xiaoshi Tang (DXT), an ancient Chinese herbal remedy dating back to 220 AD, as documented initially in “Treatise on Febrile and Miscellaneous Diseases,” is used to treat damp-heat jaundice with interior sthenia syndrome. In DXT, anthraquinones and alkaloids form insoluble [...] Read more.
Background: Dahuang Xiaoshi Tang (DXT), an ancient Chinese herbal remedy dating back to 220 AD, as documented initially in “Treatise on Febrile and Miscellaneous Diseases,” is used to treat damp-heat jaundice with interior sthenia syndrome. In DXT, anthraquinones and alkaloids form insoluble complexes, reducing its effectiveness. A revised herbal extract, DXT-M, was developed, and its hepatoprotective properties were demonstrated in animal models using pharmacodynamic, metabolomic, network pharmacological, and toxicological approaches. Methods: The α-naphthalene isothiocyanate was utilised to establish the acute liver injury rat model. The assays of glutamate pyruvate transaminase, glutamic oxalacetic transaminase, alkaline phosphatase, bilirubin, total bile acid, complement 3 (C3) and C4, interleukin-2 (IL-2) and IL-6, tumour necrosis factor α (TNF-α), and pathological morphology were used to evaluate the hepatoprotection of DXT in comparison to DXT-M. The 1H-NMR-based serum and urine metabolomics were performed to identify potential biomarkers and metabolic pathways of DXT-M in treating hepatitis. The intrinsic regulatory mechanisms of DXT in liver protection, as well as the combination of network toxicology, were elucidated. Statistical analyses included RM two-way ANOVA with Geisser–Greenhouse correction and Dunnett’s post hoc test for longitudinal data, and one-way ANOVA with Dunnett’s post hoc test for group comparisons. Data were shown as mean ± SD. Results: Liver-injured animals exhibited weight loss, ruffled fur, and liver damage, accompanied by elevated liver function indicators. DXT-M effectively improved these symptoms, repaired liver damage, restored liver function, and regulated immune status by modulating complement 3. Metabonomics and other analyses indicated the CYP/GST-ROS axis is key to its hepatoprotective effects. DXT-M outperformed DXT in efficacy. Conclusions: DXT-M demonstrated significant effectiveness in restoring liver pathological damage, correcting abnormal biochemical indicators of liver function, and regulating complement factors. The pathway of CYP/GST-ROS served as the shared regulatory axis and transformation site for DXT-M’s liver protective effects. These findings suggest that DXT-M has potential as a treatment for acute liver injury, highlighting the need for further research into its underlying molecular mechanisms as well as its complete material basis. This study’s main limitation is its focus on acute models; future research should include other liver diseases and clinical observation to evaluate its full potential. Full article
(This article belongs to the Special Issue Network Pharmacology of Natural Products, 2nd Edition)
Show Figures

Graphical abstract

21 pages, 2804 KB  
Article
Clinicopathological Significance of Transcription Factor p73 in Breast Cancers: Protein Expression and Transcriptomic Study
by Ahmed Shoqafi, Asmaa Ibrahim, Ayat Lashen, Michael S. Toss, Shatha Alqahtani, Islam Miligy, Mashael Algethami, Amera Sheha, Jennie N. Jeyapalan, Nigel P. Mongan, Andrew R. Green, Emad A. Rakha and Srinivasan Madhusudan
Biomedicines 2025, 13(10), 2484; https://doi.org/10.3390/biomedicines13102484 - 12 Oct 2025
Viewed by 376
Abstract
Background: p73, a member of the p53 family of transcription factors, plays important roles in DNA repair, cell proliferation, angiogenesis, invasion, metastasis, immune evasion, and cytotoxic therapy response. The clinicopathological significance of p73 in breast cancer, particularly in the context of TP53 [...] Read more.
Background: p73, a member of the p53 family of transcription factors, plays important roles in DNA repair, cell proliferation, angiogenesis, invasion, metastasis, immune evasion, and cytotoxic therapy response. The clinicopathological significance of p73 in breast cancer, particularly in the context of TP53 mutation, remains largely unknown. Methods: Clinicopathological significance of p73 and p53 protein expression was evaluated in 1369 invasive BC and 317 ductal carcinomas in situ (DCIS), including in p53 wild-type or p53 mutant tumours. p73 transcripts and splice variants were investigated in breast cancer genomes (TCGA). Results: High cytoplasmic p73 was significantly associated with high tumour grades, high pleomorphism scores, high mitotic scores, high risk Nottingham prognostic index, negative expression of oestrogen receptors (ERs), triple negative phenotypes (all p values ≤ 0.01), and poor breast cancer specific survival (BCSS) (p = 0.013). In TP53 mutant breast cancers, high p73 was significantly associated with aggressive histopathological features (all p ≤ 0.001) and poor BCSS (p = 0.001) but not in p53 wild-type tumours. Conclusions: Cytoplasmic p73 may be a marker of aggressive phenotype and worse prognosis, particularly in p53 mutant breast cancer. p73, in conjunction with altered p53 expression, may be involved in breast cancer pathogenesis. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

16 pages, 255 KB  
Article
Tumour SNPs Associated with Immune-Related Hepatitis in Patients with Melanoma Receiving Immune Checkpoint Inhibitors
by Jose María Rodríguez-Piñas, Alicia Romero-Lorca, Maria Gaibar, Apolonia Novillo, Margarita Rubio, Diego Malón, Beatriz Antón-Pascual, Francisca Inmaculada Camacho, Mercedes Cavanagh, David Ricardo Luján, Ana Manuela Martín, Radia Khedaoui, Diana Moreno, Fernando Pinedo, Macarena Boiza, Silvia García-Adrián, Patricia Gómez, David Marrupe and Ana Fernández-Santander
Biomedicines 2025, 13(10), 2351; https://doi.org/10.3390/biomedicines13102351 - 25 Sep 2025
Viewed by 376
Abstract
Background and Aims: Immune checkpoint inhibitors (ICIs) have significantly improved survival rates for patients with metastatic melanoma. However, these treatments can lead to immune-related adverse events (irAEs), including hepatitis. This exploratory study sought to identify tumour single-nucleotide polymorphisms (SNPs) associated with the risk [...] Read more.
Background and Aims: Immune checkpoint inhibitors (ICIs) have significantly improved survival rates for patients with metastatic melanoma. However, these treatments can lead to immune-related adverse events (irAEs), including hepatitis. This exploratory study sought to identify tumour single-nucleotide polymorphisms (SNPs) associated with the risk of ICI-induced hepatitis in melanoma patients. Methods: The study cohort comprised 69 patients with malignant melanoma treated with ICIs at several hospitals in Madrid, Spain. DNA was extracted from formalin-fixed paraffin-embedded tumour biopsies and SNP genotyping was conducted using a MassARRAY platform. Results: Significant associations were found between hepatitis risk and 4 of the 20 SNPs examined. A possible risk effect was shown for the variant GABRP SNP alleles rs11743438 and rs11743735. Among patients homozygous for these variant alleles (v/v), significantly higher proportions developed hepatitis, 75% and 71.4%, respectively, compared to 32.8% and 21.4, respectively, not developing hepatitis (p = 0.046; p = 0.013). However, in the same genotype group comparisons, the RGMA SNP rs4778080 seemed to have a protective effect, as 100% of patients who developed hepatitis were not in the v/v group for this allele (p = 0.043). Additionally, in genotype group comparisons wt/wt versus wt/v + v/v, the PACRG SNP rs55733913 was also associated with a higher risk of ICI-induced hepatitis: 66.7% of patients with hepatitis versus 22.8% without hepatitis in genotype group wt/v + v/v (p = 0.041). Conclusions: This exploratory study identifies candidate tumour SNPs as possible biomarkers to predict the risk of ICI-induced hepatitis, warranting their validation in larger patient cohorts. Full article
(This article belongs to the Special Issue Genetic Insights and Pharmacogenomic Approaches in Cancer Treatment)
27 pages, 2788 KB  
Review
The Role of Intracellular Lipid-Binding Proteins in Digestive System Neoplasms
by Christos Kakouratos, Adriana Fernandez Garcia, Pramod Darvin and Hemant M. Kocher
Curr. Oncol. 2025, 32(10), 531; https://doi.org/10.3390/curroncol32100531 - 24 Sep 2025
Viewed by 856
Abstract
Intracellular lipid-binding proteins (iLBPs) are key mediators of intracellular transport for fatty acids and retinoids, functioning as lipid chaperones. Beyond lipid transport, iLBPs regulate signalling pathways, gene expression, oxidative balance, and inflammation. Furthermore, they are increasingly recognised for their involvement in gastrointestinal (GI) [...] Read more.
Intracellular lipid-binding proteins (iLBPs) are key mediators of intracellular transport for fatty acids and retinoids, functioning as lipid chaperones. Beyond lipid transport, iLBPs regulate signalling pathways, gene expression, oxidative balance, and inflammation. Furthermore, they are increasingly recognised for their involvement in gastrointestinal (GI) diseases, especially in cancer. iLBPs are classified into four different subfamilies, each displaying distinct tissue distributions and ligand preferences. Functional roles are context-dependent, for instance, CRABP2 may act as either tumour suppressor or promoter, and FABP4 exhibits metabolic state dependent effects. These proteins also influence drug resistance, immune evasion, and lipid-mediated signalling. Overall, iLBPs extend beyond lipid trafficking to intersect with oncogenic pathways, influence cell fate, and affect treatment response, highlighting their potential as biomarkers and therapeutic targets in GI oncology. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Graphical abstract

27 pages, 2143 KB  
Review
Targeting the CXCR4/CXCL12 Axis to Overcome Drug Resistance in Triple-Negative Breast Cancer
by Desh Deepak Singh, Dharmendra Kumar Yadav and Dongyun Shin
Cells 2025, 14(18), 1482; https://doi.org/10.3390/cells14181482 - 22 Sep 2025
Viewed by 1400
Abstract
Triple-negative breast cancer (TNBC) remains one of the most aggressive and treatment-resistant forms. TNBC is an aggressive and therapeutically resistant subtype of breast cancer, marked by the absence of estrogen, progesterone, and HER2 receptors. The lack of defined molecular targets significantly limits treatment [...] Read more.
Triple-negative breast cancer (TNBC) remains one of the most aggressive and treatment-resistant forms. TNBC is an aggressive and therapeutically resistant subtype of breast cancer, marked by the absence of estrogen, progesterone, and HER2 receptors. The lack of defined molecular targets significantly limits treatment options and contributes to high recurrence rates. Among the key pathways involved in TNBC progression and resistance, the CXCR4/CXCL12 chemokine axis has emerged as a critical player. CXCR4, a G-protein-coupled receptor, binds specifically to its ligand CXCL12, promoting tumour cell proliferation, metastasis, immune evasion, and stromal remodelling. Its overexpression is frequently associated with poor prognosis, disease progression, and resistance to conventional therapies in TNBC. This review explores how the chemokine receptor type 4 (CXCR4/CXCL12) axis facilitates drug resistance through mechanisms such as epithelial–mesenchymal transition (EMT), cancer stemness, and microenvironmental interactions. Notably, CXCR4 antagonists like plerixafor, balixafortide, and POL5551 have shown encouraging preclinical and clinical results, particularly when combined with chemotherapy or immunotherapy. Additionally, innovative strategies, including radiopharmaceuticals, peptide inhibitors, and nanotechnology-based delivery platforms, offer expanded therapeutic avenues. Despite persistent challenges such as tumour heterogeneity and potential toxicity, growing clinical evidence supports the translational relevance of this axis. This manuscript provides an in-depth analysis of CXCR4/CXCL12-mediated drug resistance in TNBC and evaluates current and emerging therapeutic interventions. Full article
(This article belongs to the Special Issue Unlocking the Secrets Behind Drug Resistance at the Cellular Level)
Show Figures

Figure 1

Back to TopTop