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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (782)

Search Parameters:
Keywords = human pancreatic cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 1726 KB  
Review
CILP2: From ECM Component to a Pleiotropic Modulator in Metabolic Dysfunction, Cancer, and Beyond
by Zheqiong Tan, Suotian Liu and Zhongxin Lu
Biomolecules 2026, 16(1), 167; https://doi.org/10.3390/biom16010167 - 19 Jan 2026
Viewed by 32
Abstract
Initially characterized as a component of the extracellular matrix (ECM) in cartilage, cartilage intermediate layer protein 2 (CILP2) is now recognized as a pleiotropic secretory protein with far-reaching roles in physiology and disease. This review synthesizes evidence establishing CILP2 as a key modulator [...] Read more.
Initially characterized as a component of the extracellular matrix (ECM) in cartilage, cartilage intermediate layer protein 2 (CILP2) is now recognized as a pleiotropic secretory protein with far-reaching roles in physiology and disease. This review synthesizes evidence establishing CILP2 as a key modulator at the nexus of metabolic dysfunction, cancer, and other pathologies. Genomic studies have firmly established the NCAN-CILP2 locus as a hotspot for genetic variants influencing dyslipidemia and cardiovascular risk. Functionally, CILP2 is upregulated by metabolic stress, including high glucose and oxidatively modified LDL (oxLDL), and actively contributes to pathologies such as dyslipidemia, diabetes, and sarcopenia by impairing glucose metabolism and mitochondrial function. Its role extends to fibrosis and neurodevelopment, promoting hypertrophic scar formation and neurogenesis through interactions with ATP citrate lyase (ACLY) and Wnt3a, respectively. More recently, CILP2 has emerged as an oncoprotein, overexpressed in multiple cancers, including pancreatic ductal adenocarcinoma and colorectal cancer. It drives tumor proliferation and metastasis and correlates with tumor microenvironment remodeling through mechanisms involving Akt/EMT signaling and immune infiltration. The dysregulation of CILP2 in patient serum and its correlation with disease severity and poor prognosis highlight it as a promising biomarker and a compelling therapeutic target across a spectrum of human diseases. Full article
(This article belongs to the Section Biomacromolecules: Proteins, Nucleic Acids and Carbohydrates)
Show Figures

Figure 1

21 pages, 5725 KB  
Article
The Synergistic Effects of rhArg with Bcl-2 Inhibitors or Metformin Co-Treatment in Multiple Cancer Cell Models
by Lai-Pan Sze, Vicky Mei-Ki Ho, Wing-Ki Fung, Kin-Ho Law, Yifan Tu, Yik-Hing So, Sai-Fung Chung, Wing-Leung Wong, Zhen Liu, Alisa Sau-Wun Shum, Leo Man-Yuen Lee and Yun-Chung Leung
Cells 2026, 15(2), 164; https://doi.org/10.3390/cells15020164 - 16 Jan 2026
Viewed by 267
Abstract
Background: Recombinant human arginase (rhArg) has been proven to exhibit an anticancer effect via arginine starvation. To further improve the efficacy of rhArg, we examined the feasibility of a combination strategy with Bcl-2 inhibitors (ABT263 and ABT199) or an antidiabetic drug (metformin) and [...] Read more.
Background: Recombinant human arginase (rhArg) has been proven to exhibit an anticancer effect via arginine starvation. To further improve the efficacy of rhArg, we examined the feasibility of a combination strategy with Bcl-2 inhibitors (ABT263 and ABT199) or an antidiabetic drug (metformin) and investigated the mechanistic basis for these strategies. Methods: The combination effects were evaluated in a panel of human cancer cell lines modeling pancreatic ductal carcinoma (PDAC), triple-negative breast cancer (TNBC), colorectal cancer (CRC) and glioblastoma (GBM). Western blot analysis was used to evaluate the expression of apoptotic and cell cycle markers. MTT assay was used to evaluate the combination efficacy. Flow cytometric assays were used to investigate the apoptotic and cell cycle effects. Results: The combination of rhArg with sublethal doses of ABT263 significantly induced dose-dependent apoptosis, with elevated expression of apoptotic markers and a CI of 0.47 in U251. The combination inhibited CDK2 and cyclin A expression, indicating that the observed synergy also resulted from cell cycle arrest. We also found that rhArg + metformin was synergistic in a time-dependent manner. Compared to other amino acid depletion agents, rhArg + ABT263 was the most favorable combination pair. Conclusions: The combination of rhArg and ABT263 enhanced apoptosis and cell cycle arrest, demonstrating a potential broad-spectrum antitumor strategy. Full article
Show Figures

Figure 1

21 pages, 13737 KB  
Article
Adipose Stromal Cell-Derived Cancer-Associated Fibroblasts Promote Pancreatic Adenocarcinoma Progression Through SFRP4 Signaling
by Joseph Rupert, Lingyi Cai, Alexes C. Daquinag, Dimitris Anastassiou and Mikhail G. Kolonin
Cancers 2026, 18(2), 233; https://doi.org/10.3390/cancers18020233 - 12 Jan 2026
Viewed by 236
Abstract
Background/objectives: Progression of pancreatic ductal adenocarcinoma (PDAC) and other carcinomas relies on cancer-associated fibroblasts (CAFs). A subset of CAFs is derived from adipose stromal cells (ASCs) recruited by tumors and the ASC-CAF conversion has been associated with invasiveness and poor prognosis. Methods: To [...] Read more.
Background/objectives: Progression of pancreatic ductal adenocarcinoma (PDAC) and other carcinomas relies on cancer-associated fibroblasts (CAFs). A subset of CAFs is derived from adipose stromal cells (ASCs) recruited by tumors and the ASC-CAF conversion has been associated with invasiveness and poor prognosis. Methods: To explore the underlying molecular mechanisms, we used a model based on primary ASCs derived from human visceral adipose tissue co-cultured with human PDAC cell line Capan-1. To investigate cancer progression in vivo, we also used mice orthotopically grafted with mouse KPC cells. Results: Genomic analysis revealed that Capan-1 co-culture induces Wnt and TGFβ signaling and extracellular matrix (ECM) gene expression in ASC. We investigated the function of two markers of the fibroblastic transition highly induced by cancer cells: a long non-coding RNA LINC01614 and a Wnt signaling modulator SFRP4. By using ASCs with either SFRP4 or LINC01614 knocked out (ko), we showed that both genes are required for Wnt/TGFβ signaling and ECM induction in ASCs by Capan-1. Analysis of changes in Capan-1 genes that rely on LINC01614 and SFRP4 expression in ASCs also identified the Wnt and TGF pathways. SFRP4 ko in ASCs suppressed both migration and invasion of Capan-1 cells. We show that tumors in SFRP4 ko mice have less desmoplasia, less epithelial dedifferentiation, reduced growth rate, and reduced progression to metastasis. Conclusions: We conclude that SFRP4 promotes cancer progression in pancreatic cancer and is a promising therapeutic target. Full article
Show Figures

Figure 1

23 pages, 575 KB  
Review
A Bird’s Eye View: A Close Look into Avian CAM Models for Translational Blood Cancer Research
by Izabela M. Cymer, Niamh McAuley, Cathy E. Richards, Hanne Jahns, Siobhan V. Glavey and Ann M. Hopkins
Cancers 2026, 18(2), 209; https://doi.org/10.3390/cancers18020209 - 9 Jan 2026
Viewed by 339
Abstract
The chorioallantoic membrane (CAM) is a well-vascularised extra-embryonic membrane that supports avian embryonic development and can be used as an implantation site for xenograft models of various cancers. CAM tumour research models are powerful and versatile, offering a rapid, cost-effective and ethical complement [...] Read more.
The chorioallantoic membrane (CAM) is a well-vascularised extra-embryonic membrane that supports avian embryonic development and can be used as an implantation site for xenograft models of various cancers. CAM tumour research models are powerful and versatile, offering a rapid, cost-effective and ethical complement to mouse xenograft studies. Their capacity for real-time observation of tumour growth, angiogenesis and metastasis within an immunocompetent living organism is particularly compelling. While CAM models have been extensively utilised for investigating solid cancers, such as breast, lung and pancreatic, their potential for haematological malignancy research remains comparatively underexplored. This review examines the relevance, advantages and translational potential of avian CAM models in studying blood cancers. Their applications across three primary categories are discussed—leukaemias, lymphomas and myelomas—highlighting experimental approaches that replicate aspects of human disease progression and therapeutic responsiveness. Moreover, the review evaluates species-specific considerations relevant to model fidelity, including evolutionary distance and functional parallels between avian and human haematopoiesis. These comparisons underscore both the opportunities and limitations for utilising CAM models in haematologic malignancy research. For their potential to investigate mechanisms of cancer development and treatment in simple but immunocompetent in vivo settings, we propose that CAM tumour models offer high value as a bridge between in vitro and mammalian in vivo studies for haematology translational research. Full article
Show Figures

Figure 1

23 pages, 2214 KB  
Review
Role of Gut Microbiome in Oncogenesis and Oncotherapies
by Renuka Sri Sai Peddireddi, Sai Kiran Kuchana, Rohith Kode, Saketh Khammammettu, Aishwarya Koppanatham, Supriya Mattigiri, Harshavardhan Gobburi and Suresh K. Alahari
Cancers 2026, 18(1), 99; https://doi.org/10.3390/cancers18010099 - 29 Dec 2025
Viewed by 607
Abstract
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering [...] Read more.
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering bile acid metabolism, and disrupting epithelial barrier integrity. At the same time, the gut microbiome significantly modulates the host response to oncotherapies including chemotherapy, radiotherapy, and especially immunotherapy, where microbial diversity and specific taxa determine treatment efficacy and toxicity. This review synthesizes current evidence on the role of the gut microbiome in both oncogenesis and oncotherapies, focusing on thirteen cancers with the strongest and most clinically relevant microbiome associations, colorectal cancer, gastric cancer, hepatocellular carcinoma, gallbladder cancer, esophageal cancer, pancreatic cancer, oral squamous cell carcinoma, cervical cancer, prostate cancer, breast cancer, lung cancer, brain cancer, and melanoma. These cancers were selected based on robust mechanistic data linking microbial alterations to tumor initiation, progression, and therapy modulation, as well as their global health burden and translational potential. In addition, we have provided mechanistic insights or clinical correlations between the microbiome and cancer outcomes. Across cancers, common microbial mechanisms included pro-inflammatory signaling (e.g., NF-κB and STAT3 pathways), DNA damage from bacterial toxins (e.g., colibactin, nitrosating species), and metabolite-driven tumor promotion (e.g., secondary bile acids, trimethylamine N-oxide). Conversely, beneficial commensals such as Faecalibacterium prausnitzii and Akkermansia muciniphila supported antitumor immunity and improved responses to immune checkpoint inhibitors. In conclusion, the gut microbiome functions as both a driver of malignancy and a modifiable determinant of therapeutic success. Integrating microbiome profiling and modulation strategies such as dietary interventions, probiotics, and fecal microbiota transplantation into oncology practice may pave the way for personalized and more effective cancer care. Full article
Show Figures

Figure 1

21 pages, 3493 KB  
Article
Downregulation of the CCK-B Receptor in Pancreatic Stellate Cells Blocks Molecular Proliferative Pathways and Increases Apoptosis to Decrease Pancreatic Cancer Growth In Vitro
by Miranda Ortega, Eri Agena, Wenqiang Chen, Hong Cao, Sona Vasudevan, Narayan Shivapurkar, Mariaelena Pierobon and Jill P. Smith
Int. J. Mol. Sci. 2025, 26(23), 11699; https://doi.org/10.3390/ijms262311699 - 3 Dec 2025
Viewed by 597
Abstract
Pancreatic cancer is characterized by an extensive fibrotic stroma largely driven by activated pancreatic stellate cells (PSCs)/fibroblasts, which also function to support tumor growth and metastasis. Cholecystokinin-B receptors (CCK-BRs) are expressed on pancreatic stellate cells (PSCs) and have emerged as a key regulator [...] Read more.
Pancreatic cancer is characterized by an extensive fibrotic stroma largely driven by activated pancreatic stellate cells (PSCs)/fibroblasts, which also function to support tumor growth and metastasis. Cholecystokinin-B receptors (CCK-BRs) are expressed on pancreatic stellate cells (PSCs) and have emerged as a key regulator of PSC activation and tumor-stromal interactions. We hypothesized that disrupting CCK-BR function shifts PSCs to a more quiescent phenotype and reduces their pro-fibrotic and tumor-supportive activity to decrease growth of pancreatic cancer. Murine PSCs were genetically engineered with CRISPR-Cas9 to knockout the CCK-BR. In a series of experiments, the role of the CCK-BR expression was evaluated on cell migration, proliferation, differentially expressed genes, molecular signaling pathways, and in co-culture with murine pancreatic cancer epithelial cells. Next, primary human pancreatic stellate cells were treated with proglumide, a CCK-BR antagonist, to study the effects of pharmacologic blockade of the CCK-BR on cellular signaling and proliferative pathways by RNA sequencing. Knockout of the CCK-BR led to significant decreases in PSC activation and the ability to stimulate growth of pancreatic cancer cells in co-culture. Both genetic knockdown and pharmacologic blockade of the CCK-BR downregulated genes implicated in fibrosis, proliferation, fibroblast activation, and tumorigenesis, while genes implicated in apoptosis and tumor suppression were upregulated. Flow cytometry showed increased apoptosis markers in CCK-BR-knockout cells compared to controls. These experiments combine transcriptomic profiling with functional validation to provide a comprehensive analysis of how targeting CCK-BR interrupts the cross-communication between cancer cells and fibroblasts. Blockade or downregulation of the CCK-BR on pancreatic fibroblasts may provide a strategy to disrupt oncogenic signaling pathways and reprogram the tumor microenvironment. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Pancreatic Cancer: 2nd Edition)
Show Figures

Figure 1

19 pages, 2720 KB  
Article
Evaluation of Nanoparticle-Based Plasma Enrichment on Individuals with Primary and Metastatic Pancreatic Cancer
by Ching-Seng Ang, Nicholas A. Williamson, Chelsea Dumesny, Michael G. Leeming, Keshava Datta, Swati Varshney, Mehrdad Nikfarjam and Hong He
Cancers 2025, 17(23), 3765; https://doi.org/10.3390/cancers17233765 - 25 Nov 2025
Viewed by 802
Abstract
Background and Methods: Using a nanoparticle-based enrichment (Proteonano) methodology on human plasma samples, we achieved a substantial increase in identified proteins from ~700 to >5000 proteins compared to neat plasma digest. In a small-scale pilot test, we applied this methodology to a [...] Read more.
Background and Methods: Using a nanoparticle-based enrichment (Proteonano) methodology on human plasma samples, we achieved a substantial increase in identified proteins from ~700 to >5000 proteins compared to neat plasma digest. In a small-scale pilot test, we applied this methodology to a small cohort of plasma samples from pancreatic cancer (PC) patients with different disease stages: (I) primary tumor and (II) metastases, and compared them with healthy controls. Most identified proteins are within the Human Plasma Proteome Project (HPPP) database, and more than 300 proteins are on the list of FDA-approved drug targets. Results: We observed a large and significant increase in ribosomal proteins in the plasma of patients with metastatic PC. ADH1C and ADH1B, both members of the alcohol dehydrogenase family, were particularly upregulated in patients with liver metastasis. Fifteen other predicted secreted and/or cell surface–associated proteins with known cancer associations are also significantly altered and would otherwise go undetected in neat, digested plasma. Conclusions: The significant increase in proteome depth allows a strong foundation for future large-scale experimental and comparative analysis. Lastly, similar conclusions could be reached from comparing different mass spectrometers (Orbitrap Astral and Orbitrap Ascend) and columns (depth and throughput) setups on the same dataset, although the depth approach on the newer Orbitrap Astral instrumentations can reveal additional insights in the plasma proteome. Full article
(This article belongs to the Special Issue The Progress of Pancreatectomy for Pancreatic Cancer Treatment)
Show Figures

Figure 1

23 pages, 1019 KB  
Review
Role of Probiotics in Enhancing Immune Function and Improving the Effectiveness of Treatments for Pancreatic Cancer
by Kawaljit Kaur
Microorganisms 2025, 13(12), 2687; https://doi.org/10.3390/microorganisms13122687 - 25 Nov 2025
Viewed by 1701
Abstract
Pancreatic cancer often goes unnoticed in its early stages because it causes few or no symptoms, leading to late diagnoses and limited treatment options. The main challenges are delayed detection, drug resistance, and the tumor’s complexity, though progress is being made in targeted [...] Read more.
Pancreatic cancer often goes unnoticed in its early stages because it causes few or no symptoms, leading to late diagnoses and limited treatment options. The main challenges are delayed detection, drug resistance, and the tumor’s complexity, though progress is being made in targeted therapies, immunotherapy, metabolism-based strategies, and early detection methods. Current treatments aim to boost immune responses, extend survival, and improve quality of life. In pancreatic cancer patients, peripheral blood-derived natural killer (NK) cells show reduced numbers, decreased cytotoxic activity, and lower cytokine secretion, which may contribute to tumor growth and spread. NK cell-based immunotherapies have gained attention, with in vitro and mouse studies showing that NK cells can slow the growth of pancreatic tumor stem-like cells and encourage tumor differentiation through cytokines. Preclinical research in humanized mice suggests that NK cell therapies could reduce tumor load and restore immune function. Probiotics are also being studied in preclinical models as a potential adjuvant in therapy to restore immunity, slow tumor growth, and improve outcomes. This review compiles preclinical evidence on the benefits of combining probiotics with NK cell-based treatments for pancreatic cancer. In vitro studies indicate that probiotics can activate immune cells like peripheral blood mononuclear cells (PBMCs), NK cells, T cells, and antigen-presenting cells to help fight tumors. In humanized mouse models, combining probiotics with NK cell therapy has shown promise in reducing tumor burden, restoring immune function, and even reversing tumor-induced bone damage. The exact probiotic formulations and mechanisms are still under study, and clinical trials are in early stages without conclusive results yet. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

38 pages, 3171 KB  
Review
Emerging Role of Calycosin in Inflammatory Diseases: Molecular Mechanisms and Potential Therapeutic Applications
by Tongzhan Liu, Yifei Ye, Yu Hu and Meixiu Jiang
Biomolecules 2025, 15(12), 1643; https://doi.org/10.3390/biom15121643 - 22 Nov 2025
Viewed by 1289
Abstract
Inflammatory diseases are a type of disease caused by multiple factors, which are characterized by local or systemic tissue inflammatory reactions, commonly including atherosclerosis, osteoarthritis, non-alcoholic fatty liver, chronic kidney diseases, acute pancreatitis, and tumors. The prevalence of the above diseases is globally [...] Read more.
Inflammatory diseases are a type of disease caused by multiple factors, which are characterized by local or systemic tissue inflammatory reactions, commonly including atherosclerosis, osteoarthritis, non-alcoholic fatty liver, chronic kidney diseases, acute pancreatitis, and tumors. The prevalence of the above diseases is globally high and a growing threat to human health, as well as a huge healthcare burden. In recent years, Chinese herbal medicines have become an important reservoir for the discovery of new drugs and applications due to their unique molecular structures and potential biotherapeutic effects. Numerous studies have confirmed the beneficial effects of natural products in the prevention and treatment of different diseases. Scientific studies on the therapeutic potential of natural products have become a hot topic nowadays, especially regarding the active ingredients of herbs. Calycosin is a kind of isoflavonoid extracted from the root of Radix astragali, exhibiting anti-inflammatory, antioxidant, anti-cancer, cardioprotective, hepatoprotective, and neuroprotective activities. Therefore, this review aims to discuss the emerging roles, molecular mechanisms and therapeutic potential of calycosin in resolving inflammatory diseases. Full article
(This article belongs to the Special Issue Inflammation—The Surprising Bridge between Diseases)
Show Figures

Figure 1

22 pages, 10255 KB  
Article
Targeting PAK1 or PAK4 Uncovers Different Mechanisms of Vascular Reprogramming in Pancreatic Cancer
by Arian Ansardamavandi, Chelsea Dumesny, Sarah Ellis, Ching-Seng Ang, Mehrdad Nikfarjam and Hong He
Cells 2025, 14(22), 1806; https://doi.org/10.3390/cells14221806 - 17 Nov 2025
Viewed by 806
Abstract
The tumour microenvironment in pancreatic ductal adenocarcinoma (PDA) regulates vascular function and therapeutic response. P21-activated kinases (PAKs) regulate cytoskeletal dynamics and angiogenesis; however, their roles in vascular reprogramming and chemotherapy responses remain unclear. This study examined the effects of a PAK1 knockdown (PAK1KD) [...] Read more.
The tumour microenvironment in pancreatic ductal adenocarcinoma (PDA) regulates vascular function and therapeutic response. P21-activated kinases (PAKs) regulate cytoskeletal dynamics and angiogenesis; however, their roles in vascular reprogramming and chemotherapy responses remain unclear. This study examined the effects of a PAK1 knockdown (PAK1KD) and a PAK4 knockout (PAK4KO) on vascular remodelling in PDA. Human PANC-1 wild-type (WT), PAK1KD, and PAK4KO cells were injected subcutaneously into the flanks of SCID mice followed gemcitabine treatment. The tumour growth, vascular density, pericyte coverage, adhesion molecules, and hypoxia were determined. A proteomics study was used to identify the molecular changes involved in the vascular pathways. PAK1KD suppressed tumour growth and angiogenesis, promoted vascular normalisation, reduced hypoxia, and increased stromal ICAM-1. PAK4KO inhibited tumour growth, enlarged vessels, enhanced angiogenesis, and reduced hypoxia. PAK4KO did not affect adhesion molecules in the absence of gemcitabine, but markedly upregulated ICAM-1 and VCAM-1 with gemcitabine. Additionally, PAK4KO promoted vascular mimicry (VM) with a compromised integrity in tumour-derived vessels, but enhanced the integrity in endothelial-derived vessels. The proteomics study confirmed the enrichment of molecules in fibronectin and the VEGF pathway in PAK4KO cancer cells, along with the upregulation of EphA2, RhoA, ROCK1, ROCK2, and components of the EPH-ephrin signalling pathway, linking to enhanced VM. Neither PAK1KD nor PAK4KO increased the gemcitabine efficacy. In conclusion, PAK1KD and PAK4KO suppressed tumour growth with distinct vascular effects, but failed to enhance the gemcitabine responses, suggesting that PAK targeting reprograms the PDA vasculature, but offers limited benefit in chemotherapy-resistant models. Full article
(This article belongs to the Special Issue Molecular, Cellular and Biochemical Approaches of Anti-Cancer Drugs)
Show Figures

Graphical abstract

12 pages, 2184 KB  
Review
Structural and Functional Perspectives of Optineurin in Autophagy, Immune Signaling, and Cancer
by Gianluca Medigovic, Hari Krishnareddy Rachamala, Shamit Kumar Dutta and Krishnendu Pal
Cells 2025, 14(22), 1746; https://doi.org/10.3390/cells14221746 - 7 Nov 2025
Viewed by 1223
Abstract
Optineurin (OPTN) is a multifunctional adaptor protein that regulates diverse cellular processes, including inflammatory signaling, autophagy, vesicular trafficking, and immune responses. This multifaceted role of OPTN is made possible by the presence of a complex structure comprising multiple domains that interact with different [...] Read more.
Optineurin (OPTN) is a multifunctional adaptor protein that regulates diverse cellular processes, including inflammatory signaling, autophagy, vesicular trafficking, and immune responses. This multifaceted role of OPTN is made possible by the presence of a complex structure comprising multiple domains that interact with different proteins to exert various functions important for modulating key signaling processes. Mutations in OPTN are linked with several human pathologies including glaucoma, Paget’s disease of bone, Crohn’s disease, and neurodegenerative diseases such as amyotrophic lateral sclerosis, and dementia. Emerging evidence suggests that OPTN has a complex and context-dependent role in cancer biology as well. It is upregulated in pancreatic ductal adenocarcinoma and hepatocellular carcinoma but downregulated in lung and colorectal cancers, indicating its dual role as a potential oncogene or tumor suppressor depending on the cellular environment. Additionally, OPTN plays a critical role in preventing immune evasion in colorectal cancer by maintaining interferon-gamma receptor 1 (IFNGR1) expression and supporting dendritic cell-mediated T-cell priming, thereby enhancing antitumor immune responses. Despite its significance in oncogenic pathways and immune regulation, the therapeutic potential of targeting OPTN in cancer remains largely unexplored. This review aims to provide a comprehensive understanding of OPTN’s pleiotropic functions, highlighting its role in autophagy, inflammation, immune surveillance, and cancer progression. By elucidating its diverse regulatory mechanisms, we seek to encourage further research into the therapeutic implications of OPTN in cancer treatment and immunotherapy. Full article
(This article belongs to the Section Autophagy)
Show Figures

Graphical abstract

31 pages, 11464 KB  
Article
Mechanochemical Solvent-Free Synthesis and Biological Profiling of Novel 2-Hydrazone-Bridged Benzothiazoles as Potent Anticancer Agents
by Ivana Sokol, Hanja Mlinar, Dajana Kučić Grgić, Leentje Persoons, Dirk Daelemans, Moris Mihovilović and Tatjana Gazivoda Kraljević
Sustain. Chem. 2025, 6(4), 41; https://doi.org/10.3390/suschem6040041 - 3 Nov 2025
Viewed by 909
Abstract
This study reports the solvent-free mechanochemical synthesis of a novel series of 2-hydrazone-bridged benzothiazole derivatives 1952 via the reaction of 2-hydrazinylbenzothiazole derivatives 46 with O-alkylated benzaldehydes 718. The stereostructure of the E-isomers was confirmed [...] Read more.
This study reports the solvent-free mechanochemical synthesis of a novel series of 2-hydrazone-bridged benzothiazole derivatives 1952 via the reaction of 2-hydrazinylbenzothiazole derivatives 46 with O-alkylated benzaldehydes 718. The stereostructure of the E-isomers was confirmed by 2D NOESY spectroscopy. The antiproliferative potential of these newly prepared 2-hydrazone derivatives of benzothiazole 1952 was evaluated in vitro against eight human cancer cell lines. Several compounds demonstrated low micromolar IC50 values, with some outperforming the reference drug etoposide. Among the most potent compounds, the 6-chloro-2-hydrazone(3-fluorophenyl)benzothiazole derivative 38 exhibited remarkable activity against pancreatic adenocarcinoma (Capan-1, IC50 = 0.6 µM) and non-small cell lung cancer (NCI-H460, IC50 = 0.9 µM). Structure–activity relationship analysis revealed that derivatives 4552, featuring a methoxy group at position 6 of the benzothiazole ring and either a methoxy or fluorine substituent at position 3 of the phenyl ring, showed consistently strong antiproliferative effects across all tested cell lines (IC50 = 1.3–12.8 µM). Furthermore, compounds bearing N,N-diethylamino or N,N-dimethylamino groups at position 4 of the phenyl ring generally exhibited superior activity compared to those with morpholine or piperidine moieties. However, as this study represents an initial screening, further mechanistic investigations are required to confirm specific anticancer pathways and therapeutic relevance. In addition to their in vitro anticancer properties, the antibacterial activity of the compounds was assessed against both Gram-positive and Gram-negative bacteria. Notably, compound 37 demonstrated selective antibacterial activity against Pseudomonas aeruginosa (MIC = 4 µg/mL). Overall, this work highlights the efficiency of a green, mechanochemical approach for synthesizing E-isomer hydrazone-bridged benzothiazoles and underscores their potential as promising scaffolds for the development of potent antiproliferative agents. Full article
Show Figures

Graphical abstract

20 pages, 1135 KB  
Review
Proteomic Perspectives on KRAS-Driven Cancers and Emerging Therapeutic Approaches
by Ramesh Karki, Ru Chen and Sheng Pan
Curr. Oncol. 2025, 32(11), 614; https://doi.org/10.3390/curroncol32110614 - 2 Nov 2025
Viewed by 1579
Abstract
KRAS mutations are implicated in approximately 23% of all human malignancies, with particularly high prevalence in pancreatic ductal adenocarcinoma (PDAC) (~92%), colorectal cancer (CRC) (~49%), and non-small cell lung cancer (NSCLC) (~35%). The recent approval of the KRASG12C-specific inhibitors for NSCLC [...] Read more.
KRAS mutations are implicated in approximately 23% of all human malignancies, with particularly high prevalence in pancreatic ductal adenocarcinoma (PDAC) (~92%), colorectal cancer (CRC) (~49%), and non-small cell lung cancer (NSCLC) (~35%). The recent approval of the KRASG12C-specific inhibitors for NSCLC represents a pivotal advancement in KRAS-targeted therapy. Nevertheless, the emergence of intrinsic and acquired resistance to KRAS-targeted therapies poses a significant clinical obstacle to targeting KRAS, which necessitates a deeper understanding of the resistance mechanisms. Recent progress in proteomic studies has enabled comprehensive profiling of the proteomic alterations driven by KRAS mutations, offering valuable insights into the disrupted KRAS interactome, aberrant signaling pathways and dysregulated cellular processes contributing to tumorigenesis. This review discusses current knowledge on proteomic alterations associated with oncogenic KRAS mutations, with particular focus on allele-specific proteome signatures and the roles of post-translational modifications (PTMs) of KRAS in modulating the functional networks. Furthermore, we highlight recent therapeutic advances targeting KRAS variants and discuss emerging resistance mechanisms from a proteomics-informed perspective. Full article
Show Figures

Figure 1

20 pages, 4468 KB  
Brief Report
Modified Hematopoietic Stem Cell-Derived Dendritic Cell Therapy Retained Tumor-Inhibitory Function and Led to Regression of Primary and Metastatic Pancreatic Tumors in Humanized Mouse Models
by Jose D. Gonzalez, Saleemulla Mahammad, Senay Beraki, Ariel Rodriguez-Frandsen, Neha Sheik, Elango Kathirvel, Francois Binette, David Weinstein, Anahid Jewett and Lu Chen
Vaccines 2025, 13(11), 1131; https://doi.org/10.3390/vaccines13111131 - 2 Nov 2025
Viewed by 1583
Abstract
Background/Objectives: Dendritic cell (DC)-based immunotherapies offer a promising strategy for cancer treatment but are limited by inefficient activation of cytotoxic T cells and, in turn, the host immune system. This report demonstrated that CD34+ hematopoietic stem cell (HSC)-derived allogeneic DCs engineered [...] Read more.
Background/Objectives: Dendritic cell (DC)-based immunotherapies offer a promising strategy for cancer treatment but are limited by inefficient activation of cytotoxic T cells and, in turn, the host immune system. This report demonstrated that CD34+ hematopoietic stem cell (HSC)-derived allogeneic DCs engineered by an optimized lentiviral vector (LVV) expressing CD93, CD40-ligand (CD40L), and Chemokine (C-X-C motif) ligand-13 (CXCL13) significantly enhanced the host immune system, activated tumor-specific cytotoxic T cells, and led to complete regression of both primary and metastatic pancreatic tumors in humanized mouse models. This LVV shows comparable pre-clinical efficacy compared to the first-generation vector, in addition to being compliant for clinical use, which allows further pre-clinical development towards the human trials. Methods: This 2nd generation (Gen) LVV incorporates codon-optimized transgenes (CD40L, CD93, and CXCL13) with rearranged sequence to enhance expression, driven by a strong EF1α promoter. CD34+ HSCs were transduced with this modified 2nd Gen LVV and differentiated to Engineered DCs. Therapeutic efficacy of the DC therapy with the modified vector was tested on humanized mouse models of pancreatic tumors. This was accomplished by establishing an early-stage disease model (using MIA PaCa-2 (MP2)-tumors) and late-stage metastatic disease model of the pancreatic tumors to mimic the clinical setting using luciferase-expressing MP2-(Luc)-pancreatic tumor-bearing humanized mice. Results: The modified lentiviral construct had 6-fold greater expression of CD40L, 2% less toxicity, 4.5-fold greater CD40L, and 2.2-fold greater CXCL13 secretion than its predecessor. In vitro, Engineered DCs induced robust T cell proliferation in up to 20% of T cells, up to 4-fold greater interferon-gamma (IFN-γ) secretion than controls, and showcased antigen-specific cytotoxicity by CD8+ T cells. In vivo, two intradermal doses of the 2nd Gen DCs led to complete regression of primary pancreatic tumors and metastases. Treated mice exhibited prolonged survival, indicating the induction of durable anti-tumor immunity. Conclusions: Vector optimization retained the efficacy of DC-based therapy, achieving curative responses in pancreatic tumor models. These findings support the clinical development of this 2nd Gen DC immunotherapy for pancreatic and potentially other tumors. Full article
Show Figures

Figure 1

19 pages, 7649 KB  
Article
Comparative Oncology: Cross-Sectional Single-Cell Transcriptomic Profiling of the Tumor Microenvironment Across Seven Human Cancers
by Riku Okamoto, Kota Okuno, Akiko Watanabe, Kanako Naito, Hiroyuki Minoura, Shumpei Shibaki, Kyonosuke Ikemura, Keiko Oki, Yu Kuroda, Shiori Fujino, Yusuke Nie, Nobuyuki Nishizawa, Eiichiro Watanabe, Mariko Kikuchi, Koshi Kumagai, Takahiro Yamanashi, Hiroshi Katoh, Hajime Takayasu, Takeo Sato, Takafumi Sangai, Yusuke Kumamoto, Takeshi Naitoh, Naoki Hiki and Keishi Yamashitaadd Show full author list remove Hide full author list
Cancers 2025, 17(21), 3527; https://doi.org/10.3390/cancers17213527 - 31 Oct 2025
Viewed by 1188
Abstract
Background/Objectives: To elucidate the differential transcriptional and intercellular signaling features of tumor components across various cancers, we conducted a comparative analysis using single-cell RNA sequencing (scRNA-seq). This technology enables detailed characterization of tumor ecosystems and may explain variations in tumor behavior among [...] Read more.
Background/Objectives: To elucidate the differential transcriptional and intercellular signaling features of tumor components across various cancers, we conducted a comparative analysis using single-cell RNA sequencing (scRNA-seq). This technology enables detailed characterization of tumor ecosystems and may explain variations in tumor behavior among distinct cancer types. Methods: We analyzed publicly available scRNA-seq datasets (GEO) from seven cancer types—pancreatic ductal adenocarcinoma (PDAC), hepatocellular carcinoma (HCC), esophageal squamous cell carcinoma (ESCC), breast cancer (BC), thyroid cancer (TC), gastric cancer (GC), and colorectal cancer (CRC)—to define their unique molecular characteristics and intercellular interactions. Results: PDAC displayed a distinct tumor microenvironment (TME) dominated by myeloid cells (~42%), including abundant CXCR1/CXCR2-expressing tumor-associated neutrophils (TANs) that preferentially interacted with immune rather than cancer cells. The competitive receptor ACKR1 was minimally expressed on endothelial cells, consistent with PDAC hypo-vascularity. In HCC, tumor cells lacked EPCAM and expressed complement and stem cell markers; cancer-associated fibroblasts (CAFs) were scarce, and stellate cells expressed the pericyte marker RGS5. CAFs were abundant in ESCC and BC, with IGF1/2 expression, while in GC, these markers were uniquely found in plasma cells. Since BC and GC subtypes exhibit distinct TME patterns, it is necessary to perform subtype-specific analyses for these cancers. TC showed high expression of tumor-suppressor genes, including HOPX, in tumor cells. Differential interactions and the presence of “dominant signaling cell populations “ with dominant outgoing signals may underlie the heterogeneity in tumor aggressiveness across these cancers. Conclusions: Comparative scRNA-seq analysis of multiple cancers reveals distinct tumor phenotypes and cell–cell communication patterns, offering insights into the molecular architecture of human solid tumors. Full article
Show Figures

Figure 1

Back to TopTop