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
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 (1,270)

Search Parameters:
Keywords = H460 lung cancer cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 3474 KB  
Article
A Marine Anticancer Cinnamyloxyl Derivative with Unique Binding Sites at Carbonic Anhydrase IX (CAIX) Inhibits Adenocarcinomic A549 Cells
by Shailaja Vommi Lakshmipathy, Christina Vijayaraghavan Sathyanathan, Mohanapriya Dandapani Chinambedu, Mohanraj Gopikrishnan, Abhinand Ponneri Adithavarman, Sadras Panchatcharam Thyagarajan and Mary Elizabeth Gnanambal Krishnan
Pharmaceuticals 2026, 19(1), 132; https://doi.org/10.3390/ph19010132 - 12 Jan 2026
Viewed by 182
Abstract
Background: Many inhibitors have been discovered to target hypoxia-induced carbonic anhydrase IX (CAIX) due to its critical role in lung cancers. This study discovers a novel compound, 3-(E-3,4-dihydroxycinnamaoyloxyl)-2-hydroxypropyl-9Z,12Z-octadeca-9,12-dienoate, which is produced by the seagrass Cymodocea serrulata and has binding sites at CAIX that [...] Read more.
Background: Many inhibitors have been discovered to target hypoxia-induced carbonic anhydrase IX (CAIX) due to its critical role in lung cancers. This study discovers a novel compound, 3-(E-3,4-dihydroxycinnamaoyloxyl)-2-hydroxypropyl-9Z,12Z-octadeca-9,12-dienoate, which is produced by the seagrass Cymodocea serrulata and has binding sites at CAIX that are distinct from those of current inhibitors. Methods: Compound and reference drug treatment for cell lines; Cell viability: MTT; Staining: Ao/PI/DAPI; MMP shifts and cell cycle: FACS; Gene and protein expression of CAIX, BAX, BAD: qPCR and Western blotting. Results: The compound binds to the CAIX protein, raises extracellular pH, and kills A549 cells [IC50: 11.61 µM], producing results that are lower than those of the reference drug doxorubicin [13.7 µM]. The substance depolarised the electrical potential of the mitochondrial membrane, caused S-phase arrest, and fragmented DNA. Additionally, it downregulated CAIX by 0.9 times while increasing apoptotic mRNA, BAX and BAD by 5.2 and 3.08 times, respectively, as demonstrated by qPCR. Between 0 and 24 h, the untreated hypoxic cells had a ΔpHe of 0.15, but the compound-treated cells had a ΔpHe of 0.6 indicative of intracellular acidosis. MD simulations verify the stability of the CAIX–C1 complex for more than 100 ns, and in silico studies show a strong binding affinity of the molecule to CAIX [−7.55 kcal/mol]. Conclusions: This implies that the amount of extracellular alkalosis was increased by the combination of treatment and hypoxia induction. As a result, when the cells were deprived of O2, the compound provided less defense against ROS. The compound binds to the glutamine and alanine amino acids at positions 242 and 392, respectively, at the central Zn atom of CAIX, which sets it apart from conventional sulphonamide CAIX inhibitors. This naturally occurring compound may be a potent CAIX inhibitor with newer binding sites, which could help treat hypoxic lung cancers. Full article
(This article belongs to the Special Issue Identification and Extraction of Bioactive Compounds from Marine Life)
Show Figures

Graphical abstract

30 pages, 5568 KB  
Article
Anticancer Activity of 2,3′-Dihydroxy-5′-Methoxystilbene Against NSCLC Cell Lines Through AKT-Dependent Mechanisms: A Comprehensive In Vitro and Computational Analysis
by Phisit Pouyfung, Nonthalert Lertnitikul, Noriyoshi Ogino, Achitphol Chookaew, Varisa Pongrakhananon, Piriya Chonsut, Natthaporn Sueangoen and Suwichak Chaisit
Int. J. Mol. Sci. 2026, 27(2), 719; https://doi.org/10.3390/ijms27020719 - 10 Jan 2026
Viewed by 153
Abstract
Lung cancer remains a major clinical challenge, with therapy resistance in non-small-cell lung cancer (NSCLC) driving the search for novel selective agents. This study demonstrates that 2,3′-dihydroxy-5′-methoxystilbene exhibits significant anticancer activity in NSCLC cell lines (A549, H23, and H460) while displaying substantially lower [...] Read more.
Lung cancer remains a major clinical challenge, with therapy resistance in non-small-cell lung cancer (NSCLC) driving the search for novel selective agents. This study demonstrates that 2,3′-dihydroxy-5′-methoxystilbene exhibits significant anticancer activity in NSCLC cell lines (A549, H23, and H460) while displaying substantially lower toxicity toward normal NIH/3T3 fibroblasts. The compound reduced the viability of H23 and H460 cells after 48 h. (IC50: 23.39 ± 3.27 μM and 24.20 ± 2.61 μM, respectively), with NIH/3T3 cells remaining comparatively resistant (IC50 > 100 μM). At 25 μM, it suppressed proliferation by approximately 40% in H23, 30% in H460, and 20% in A549 cells, and dose-dependently impaired colony formation and migration, leading to near-complete migration arrest in H460 cells. Apoptosis induction peaked at 19% in H23, 17% in H460, and 8% in A549 cells at 25 μM. Mechanistic studies and molecular modeling revealed AKT-dependent activity, with decreased p-AKT and p-GSK3β levels (0.70 and 0.75 in H23; 0.65 and 0.70 in H460 at 25 μM), without changes in total protein expression. Combination treatment with cisplatin yielded synergistic effects in A549 (CI = 0.83) and H460 (CI = 0.94) cells, but antagonistic effects in H23 cells (CI = 1.32). These findings identify 2,3′-dihydroxy-5′-methoxystilbene as a selective AKT-targeting stilbene with promising anticancer potential and context-dependent chemosensitizing activity in NSCLC cells. Full article
Show Figures

Graphical abstract

17 pages, 1405 KB  
Article
Heat-Assisted Extraction and Bioactivity Evaluation of a Dinactin-Associated Compound from Streptomyces UP Strains
by Grissana Pook-In, Somsak Tammawong, Chorpaka Phuangsri, Khwanla Seansupa, Sontaya Sookying, Tomoko Takahashi and Anchalee Rawangkan
Microbiol. Res. 2026, 17(1), 16; https://doi.org/10.3390/microbiolres17010016 - 9 Jan 2026
Viewed by 147
Abstract
Streptomyces is a versatile genus widely used in drug production and biotechnological applications. This study aimed to identify and characterize bioactive compounds produced by Streptomyces UP-AC4 and UP-3.2 strains and evaluate their antibacterial and anticancer activities. The strains were identified as Streptomyces californicus [...] Read more.
Streptomyces is a versatile genus widely used in drug production and biotechnological applications. This study aimed to identify and characterize bioactive compounds produced by Streptomyces UP-AC4 and UP-3.2 strains and evaluate their antibacterial and anticancer activities. The strains were identified as Streptomyces californicus and Streptomyces purpurascens via chemotaxonomy, 16S rRNA sequencing, amplified ribosomal DNA restriction analysis, and phylogenetic analysis. Bioactive compounds were extracted using heat treatments at 63 °C for 30 min or 73–110 °C for 10 min. Antibacterial activity against Staphylococcus aureus, Bacillus cereus, and Escherichia coli was assessed by agar disc assay, with MICs of 0.024–0.195 mg/mL and MBCs of 0.098–0.391 mg/mL for the most effective extracts. Anticancer activity against A549, H1299, and Lu99 lung cancer cells was evaluated using the MTT assay, showing IC50 values of 0.23 ± 0.06 to 4.85 ± 0.64 mg/mL, while exhibiting no toxicity to normal fibroblast cells. HPLC analysis indicated that heat-assisted extraction of UP-AC4 at 73 °C for 10 min enriched a dinactin-associated compound as a predominant metabolite with antibiotic and anticancer activities. In conclusion, Streptomyces UP-AC4 and UP-3.2 produce promising low-cost bioactive compounds with strong potential for pharmaceutical and healthcare applications. Full article
Show Figures

Figure 1

22 pages, 5268 KB  
Article
Herba Patriniae Component Linarin Induces Cell Cycle Arrest and Senescence in Non-Small-Cell Lung Cancer Associated with Cyclin A2 Downregulation
by Wen Xie, Xia Li, Dongmei Huang, Jiana Xu, Minghan Yu, Yanping Li and Qing K. Wang
Pharmaceuticals 2026, 19(1), 111; https://doi.org/10.3390/ph19010111 - 8 Jan 2026
Viewed by 177
Abstract
Background: Non-small-cell lung cancer (NSCLC) remains a major therapeutic challenge due to its high incidence and mortality. Herba Patriniae (HP), a traditional Chinese medicine, has long been used for respiratory disorders and exhibits anti-cancer potential. However, the therapeutic effects of HP on [...] Read more.
Background: Non-small-cell lung cancer (NSCLC) remains a major therapeutic challenge due to its high incidence and mortality. Herba Patriniae (HP), a traditional Chinese medicine, has long been used for respiratory disorders and exhibits anti-cancer potential. However, the therapeutic effects of HP on NSCLC and the underlying mechanisms have not been fully elucidated. Methods: Network pharmacology was applied to identify the core active components of HP and their potential targets in NSCLC. The anti-cancer effects of the core HP component Linarin on the malignant phenotypes of NSCLC cells were characterized using Tumor Protein P53 (p53) wild-type A549 and p53-null H1299 cell lines with Cell Counting Kit-8 (CCK-8), EdU fluorescence staining, colony formation, apoptosis analysis, cell cycle analysis, and senescence-associated β-galactosidase (SA-β-gal) staining, together with molecular docking and Western blotting analyses. Results: Network pharmacology analysis identified Linarin as the core active component of HP and screened out six hub targets, including Cyclin Dependent Kinase 1/4 (CDK1/4), Cyclin A2/B1 (CCNA2/B1), and Checkpoint Kinase 1/2 (CHEK1/2), which were found to be mainly enriched in cell cycle and senescence pathways. In vitro assays showed that Linarin dose-dependently (0–200 μM) inhibited NSCLC cell proliferation, induced G0/G1 phase arrest, and promoted cellular senescence and apoptosis in both cell lines, irrespective of p53 status. Molecular docking confirmed strong binding affinities between Linarin and the hub targets, and Western blotting confirmed that Linarin downregulated CCNA2/B1 and CHEK1. Conclusions: This study demonstrates that Linarin, the core active component of HP, exerts potent anti-NSCLC effects by inducing G0/G1 arrest, senescence, and apoptosis. These effects are associated with the downregulation of key cell cycle regulators, including CCNA2/B1 and CHEK1. Together, these findings highlight the potential of Linarin as a promising therapeutic option for NSCLC. Full article
(This article belongs to the Special Issue Adjuvant Therapies for Cancer Treatment: 2nd Edition)
Show Figures

Graphical abstract

17 pages, 783 KB  
Review
Updates on Antibody Drug Conjugates and Bispecific T-Cell Engagers in SCLC
by Kinsley Wang, Kyle Taing and Robert Hsu
Antibodies 2026, 15(1), 4; https://doi.org/10.3390/antib15010004 - 4 Jan 2026
Viewed by 486
Abstract
Background/Objectives: Small-cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy characterized by rapid proliferation, early metastasis, and near-universal relapse after initial therapy. While chemo-immunotherapy modestly improves first-line outcomes, survival after progression remains poor and highlights the urgent need for biomarker-directed strategies. Methods [...] Read more.
Background/Objectives: Small-cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy characterized by rapid proliferation, early metastasis, and near-universal relapse after initial therapy. While chemo-immunotherapy modestly improves first-line outcomes, survival after progression remains poor and highlights the urgent need for biomarker-directed strategies. Methods: A comprehensive literature search was conducted using major medical databases looking at key relevant studies on SCLC antibody studies. All authors reviewed the literature, assessed study quality, and interpreted the results from each study. Results: Recent advances in antibody–drug conjugates (ADCs) and T-cell engagers (TCEs) have transformed therapeutic development by targeting antigens selectively expressed on SCLC cells, enabling more precise and potentially durable tumor control. DLL3 has emerged as the most clinically relevant target to date, with the bispecific TCE tarlatamab demonstrating meaningful and durable response, manageable cytokine-release toxicity, and ultimately achieving accelerated FDA approval for previously treated extensive-stage SCLC. Concurrently, DLL3-directed ADCs have shown variable efficacy, underscoring the importance of payload selection, linker chemistry, and antigen density. Beyond DLL3, next-generation ADCs targeting TROP2, B7-H3, and SEZ6 have reported encouraging early-phase activity, including response rates exceeding those of existing second-line cytotoxic options, though myelosuppression, interstitial lung disease, and hepatic toxicity remain key considerations. Conclusions: Collectively, these emerging immunotherapies illustrate a shift toward antigen-specific targeting in a disease historically defined by limited therapeutic innovation. Continued optimization of antigen selection, payload and linker engineering, and biomarker-driven trial design will be critical for translating early promise into durable clinical benefit and reshaping the treatment landscape for SCLC. Full article
(This article belongs to the Section Antibody-Based Therapeutics)
Show Figures

Figure 1

21 pages, 1595 KB  
Article
Evaluation of the Cellsway Microfluidic CTC Enrichment and Identification Platform for CTC Detection in Metastatic NSCLC
by Ebru Özgür, Ayça Çırçır, Begüm Şen Doğan, Şebnem Şahin, Gizem Karayalçın, Mehmet Alper Demir, Başak Erkek, Enes Demirtaş, Özge Zorlu, Furkan Ceylan, Haluk Külah, Nuri Karadurmuş, Mehmet Ali Nahit Şendur and Saadettin Kılıçkap
Biosensors 2026, 16(1), 34; https://doi.org/10.3390/bios16010034 - 2 Jan 2026
Viewed by 424
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide, with non-small-cell lung cancer (NSCLC) accounting for the majority of cases. Standard tissue biopsies are invasive and unsuitable for repeated monitoring. Liquid biopsy technologies, particularly circulating tumor cell (CTC) analysis, offer a minimally [...] Read more.
Lung cancer is the leading cause of cancer-related mortality worldwide, with non-small-cell lung cancer (NSCLC) accounting for the majority of cases. Standard tissue biopsies are invasive and unsuitable for repeated monitoring. Liquid biopsy technologies, particularly circulating tumor cell (CTC) analysis, offer a minimally invasive alternative for real-time disease tracking. To address the need for efficient and reproducible CTC isolation, we developed the Cellsway microfluidic CTC enrichment and identification platform, which employs inertial hydrodynamics in a spiral-shaped microfluidic channel comprising hydrofoil-shaped pillars to enable high-throughput, label-free enrichment of CTCs while preserving cell integrity, followed by an optimized CTC identification assay. Analytical performance assessed through spiking experiments using NSCLC cell lines demonstrated recovery rates of 91.9% for H1975 cells and 78.3% for A549 cells. Clinical validation was performed on blood samples from 51 stage IV NSCLC patients. A 7.5 mL volume of peripheral blood was processed with the SwayBox platform, and enriched CTCs were identified through an optimized multiplex immunofluorescence protocol. CTCs were detected in 47% of NSCLC patients, with counts ranging from 0 to 72 cells per 7.5 mL of blood. At a cutoff of 1 CTC per 7.5 mL, the assay achieved a specificity of 95%. Patient-derived CTCs exhibited smaller mean diameters compared to cultured NSCLC cell lines, yet were effectively enriched through hydro-dynamic tuning. These findings demonstrate that the Cellsway platform enables efficient and re-producible CTC isolation with high specificity, supporting its potential utility for clinical monitoring and precision oncology in NSCLC. Full article
(This article belongs to the Section Nano- and Micro-Technologies in Biosensors)
Show Figures

Graphical abstract

15 pages, 1638 KB  
Article
Screening of Bioactive Microalgae from Freshwaters, Collected in Hue, Vietnam: Cytotoxic Constituents from Dolichospermum smithii HU04
by Nguyen Thi Minh Hang, Nguyen Thi Thu Ha, Hoang Duc Manh, Duong Thi Thuy, Hoang Thi Quynh, Nguyen Thi Thu Lien, Nguyen Thi Tu Oanh, Tran Huu Giap, Buu Huu Tai, Doan Thi Mai Huong, Ngo Quoc Anh and Nguyen Xuan Nhiem
Molecules 2026, 31(1), 165; https://doi.org/10.3390/molecules31010165 - 1 Jan 2026
Viewed by 265
Abstract
Background/Objectives: Microalgae are recognized as prolific producers of bioactive metabolites with pharmaceutical potential. This study aimed to isolate and characterize cytotoxic constituents from selected cytotoxic microalgae, collected in Hue city, Vietnam. Methods: Microalgal samples were collected from freshwater bodies, morphologically identified, and maintained [...] Read more.
Background/Objectives: Microalgae are recognized as prolific producers of bioactive metabolites with pharmaceutical potential. This study aimed to isolate and characterize cytotoxic constituents from selected cytotoxic microalgae, collected in Hue city, Vietnam. Methods: Microalgal samples were collected from freshwater bodies, morphologically identified, and maintained in laboratory culture. Thirteen strains were successfully isolated and cultivated in BG11, Z8, and BBM media to determine optimal growth conditions. Cytotoxic effects of extracts/compounds were determined using the sulforhodamine B assay on human lung cancer (SK-LU-1) and human liver cancer (HepG2) cell lines. The methanol extract was partitioned with n-hexane and CH2Cl2, followed by extensive chromatographic separation and HPLC purification to afford twelve compounds, including two new and ten known compounds. The structures were elucidated by HR-ESI-MS and NMR spectra, chemical methods, and comparing compounds in the literature. Results: From the phytoplankton samples collected across six freshwater bodies in Hue city, Vietnam, thirteen microalgal strains were successfully isolated and purified under laboratory conditions. These strains were morphologically and taxonomically identified to be Microcystis aeruginosa HU05, Microcystis viridis HU13, Anabaena circinalis HU08, Aphanizomenon flos-aquae HU02, Dolichospermum smithii HU04, Calothrix braunii HU14, Nostoc muscorum HU12, Nostoc punctiforme HU11, Raphidiopsis raciborskii HU03, Lyngbya spiralis HU15, Planktothrix stagnina HU16, Phormidium subtilis HU06, and Scenedesmus quadricauda HU07. All methanol extracts of those microalgae were evaluated for cytotoxic activity. The MeOH extracts of M. viridis (HU13) and D. smithii (HU04) exhibited significant cytotoxic effects, with IC50 values of 6.19 ± 0.80 and 4.89 ± 0.76 µg/mL for M. viridis, and 9.51 ± 0.84 and 8.32 ± 0.94 µg/mL for D. smithii against SK-LU-1 and HepG2 cell lines, respectively. Furthermore, chemical studies of D. smithii HU04 led to the isolation of two new compounds, smithioside A (1) and smithioside B (2) and ten known ones, 3,4,5-trimethoxyphenyl-1-O-β-D-glucopyranoside (3), 4′-hydroxy-3′-methoxyphenol-β-D-[6-O-(4″-hydroxy-3″,5″-dimethoxylbenzoate)]-glucopyranoside (4), 4′-hydroxy-2′,6′-dimethoxyphenol 1-O-β-D-(6-O-syringoyl)glucopyranoside (5), mallophenol B (6), pisoninol II (7), guaiacylglycerol (8), (E)-asarone (9), deacetylsarmentamide B (10), (E)-2-hexenyl-β-D-glucopyranoside (11), and 5,6-dihydropyridin-2(1H)-one (12). The cytotoxic activity of all isolated compounds was also evaluated against SK-LU-1 and HepG2 cancer cell lines. Compound 12 showed the strongest activity, with IC50 values of 9.13 ± 0.89 µM (SK-LU-1) and 7.64 ± 0.46 µM (HepG2). Compounds 5 and 6 exhibited moderate cytotoxic activity on both human cancer cell lines with IC50 values ranging from 25.99 to 51.47 µM. Conclusions: These results highlight the potential of Dolichospermum smithii HU04 as a source of bioactive compounds, particularly in anticancer applications. These findings suggest that D. smithii HU04 extracts could be developed for therapeutic purposes targeting cancer. Full article
Show Figures

Figure 1

23 pages, 1747 KB  
Article
The Use of Benzoin as a Privileged Structure: Synthesis, Characterization, Crystalline Form and an In Vitro Biological Evaluation of 1,2-Diphenyl-2-[1,2,3]triazol-1-yl-ethanol Derivatives
by Noé Martínez-Romero, Mario Valle-Sánchez, Marco A. García-Eleno, Carlos A. González-González, David Corona-Becerril, Lizbeth Triana-Cruz, Diego Martínez-Otero, María Teresa Ramírez-Apan, David Morales-Morales, Jorge Andrés Ornelas-Guillén and Erick Cuevas-Yañez
Molecules 2026, 31(1), 170; https://doi.org/10.3390/molecules31010170 - 1 Jan 2026
Viewed by 392
Abstract
A collection of 40 derivatives of 1,2-diphenyl-2-[1,2,3]triazol-1-yl-ethanol was obtained through a sequence of reactions, starting with benzoin as the initial raw material and using the CuAAC reaction as the key step in this process. The structure of a pair of these compounds was [...] Read more.
A collection of 40 derivatives of 1,2-diphenyl-2-[1,2,3]triazol-1-yl-ethanol was obtained through a sequence of reactions, starting with benzoin as the initial raw material and using the CuAAC reaction as the key step in this process. The structure of a pair of these compounds was ultimately corroborated by single-crystal X-ray diffraction studies, which also reveals important O-H···N interactions. The antimicrobial activity of synthesized 1,2,3-triazoles was assessed against strains that include Candida albicans and Staphylococcus aureus. The antiproliferative properties of some of these novel compounds were also tested using a variety of tumor cell lines, including U251 (human glioblastoma), PC-3 (human prostate cancer cell line), K562 (human leukemia), HCT-15 (human colorectal adenocarcinoma), MCF-7 (human breast adenocarcinoma), and SKLU (human lung adenocarcinoma). Full article
Show Figures

Figure 1

26 pages, 3200 KB  
Article
A Novel Quinolone JH62 (E-2-(Tridec-4-en-1-yl)-quinolin-4(1H)-one) from Pseudomonas aeruginosa Exhibits Potent Anticancer Activity
by Qunyi Chen, Jianhe Wang, Xiaoyan Wu, Lantu Xiong, Lianhui Zhang and Zining Cui
Microorganisms 2026, 14(1), 78; https://doi.org/10.3390/microorganisms14010078 - 30 Dec 2025
Viewed by 357
Abstract
Cancer remains a leading cause of mortality worldwide, and new chemical leads are essential for developing potent anticancer therapies. Evidence suggests that Pseudomonas aeruginosa (Pa) may suppress tumorigenesis, although the underlying mechanisms remain largely unclear. This study characterized a novel small [...] Read more.
Cancer remains a leading cause of mortality worldwide, and new chemical leads are essential for developing potent anticancer therapies. Evidence suggests that Pseudomonas aeruginosa (Pa) may suppress tumorigenesis, although the underlying mechanisms remain largely unclear. This study characterized a novel small molecule quinolone, JH62 (E-2-(tridec-4-en-1-yl)-quinolin-4(1H)-one, C22H31NO), from Pa. JH62 exhibited broad-spectrum anticancer activity, inhibiting the proliferation of A549 lung cancer cells in a time- and dose-dependent manner with an IC50 of 15 μM, while showed low cytotoxicity toward normal cells. In xenograft mice model, treatment with JH62 (10 mg/kg) reduced tumor weight and volume by 73% and 79%, respectively. Mechanistically, treatment with JH62 induced structural and functional disruption of mitochondria in cancer cells, triggered autophagic cell death, and did not cause DNA damage. Genetic analysis confirmed that JH62 biosynthesis depends on the pqsABCDE gene cluster and that JH62 positively regulates its own production. ADMET profiling further indicated promising drug-like properties for future development. These findings establish JH62 as a promising anticancer lead compound derived from microbial metabolism. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

24 pages, 5708 KB  
Article
Hypoxia-Inducible Factor-1α, a Novel Molecular Target for a 2-Aminopyrrole Derivative: Biological and Molecular Modeling Study
by Svetlana S. Zykova, Tatyana Gessel, Aigul Galembikova, Evgenii S. Mozhaitsev, Sophia S. Borisevich, Nazim Igidov, Emiliya S. Egorova, Ekaterina Mikheeva, Natalia Khromova, Pavel Kopnin, Alina Galyautdinova, Vladimir Luzhanin, Maxim Shustov and Sergei Boichuk
Cancers 2026, 18(1), 115; https://doi.org/10.3390/cancers18010115 - 30 Dec 2025
Viewed by 335
Abstract
Background: Hypoxia-inducible factor-1α (HIF-1α) is a well-known transcriptional regulator that mediates a broad spectrum of cellular responses to hypoxia, including angiogenesis, extracellular matrix remodeling, and metabolic reprogramming. These activities can be achieved by upregulation of numerous genes, such as vascular endothelial growth factors, [...] Read more.
Background: Hypoxia-inducible factor-1α (HIF-1α) is a well-known transcriptional regulator that mediates a broad spectrum of cellular responses to hypoxia, including angiogenesis, extracellular matrix remodeling, and metabolic reprogramming. These activities can be achieved by upregulation of numerous genes, such as vascular endothelial growth factors, fibroblast growth factors, and platelet-derived growth factors, which are involved in the growth regulation of normal tissues and solid tumors. Notably, HIF-1α-mediated regulation of the solid tumor’s microenvironment effectively modulates tumor sensitivity to anticancer therapies and thereby can contribute to disease progression. Methods: The study was performed on breast, lung and prostate cancer cell lines. Protein expression was examined by western blotting. Antitumor activity of 2-ANPC was measured by syngeneic 4T1 breast cancer mouse model. Results: We show here that a 2-aminopyrrole derivative (2-amino-1-benzamido-5-(2-(naphthalene-2-yl)-2-oxoethylidene)-4-oxo-4,5-dihydro-1-H-pyrrole-3-carboxamide—2-ANPC), previously shown as a potent microtubule-targeting agent, effectively downregulates HIF-1α expression in a broad spectrum of cancer cell lines, including breast, lung, and prostate cancer. The downregulation of HIF-1α expression in 2-ANPC-treated cancer cells was due to enhanced proteasome-mediated degradation, whereas the proteasome inhibitor MG-132 effectively reversed this downregulation. 2-ANPC’s potency in downregulating HIF-1α was also shown in vivo by using the 4T1 breast cancer syngraft model. Importantly, this 2-aminopyrrole derivative also downregulated the expression of vascular endothelial growth factor receptors 1 and 3 (VEGFR1 and 3) in 4T1 tumors, which correlated with decreased tumor weight and size. As expected, an increase in apoptotic (i.e., cleaved caspase-3-positive) cells was detected in 4T1 tumors treated with 2-aminopyrrole derivative. Lastly, using various computational tools, we identified four potential binding sites for 2-ANPC to interact with HIF-1α, HIF-1β, and the p300 complex. Conclusions: Collectively, we show here, for the first time, that HIF-1α is a novel molecular target for the 2-aminopyrrole derivative (2-ANPC), thereby illustrating it as a potential scaffold for the development of potent chemotherapeutic agents with anti-angiogenic activity. Full article
(This article belongs to the Special Issue Hypoxia-Targeting Strategies to Improve Cancer Therapy Outcomes)
Show Figures

Figure 1

17 pages, 2193 KB  
Article
Adamantyl-Substituted Chalcone CA13 Induces Cytoprotective Autophagy and JNK-Dependent Apoptosis in Lung Cancer Cells
by Yuting Chen, Yaxin Liu, Jing Zhou, Tingting Bao, Jing Wang and Mingtao Ao
Biomolecules 2026, 16(1), 54; https://doi.org/10.3390/biom16010054 - 30 Dec 2025
Viewed by 260
Abstract
Lung cancer remains a leading cause of cancer mortality worldwide, highlighting the need for novel therapeutics. Here, we designed and synthesized a series of adamantyl-substituted chalcones and identified CA13 as a lead compound with potent and selective antiproliferative activity against non–small cell lung [...] Read more.
Lung cancer remains a leading cause of cancer mortality worldwide, highlighting the need for novel therapeutics. Here, we designed and synthesized a series of adamantyl-substituted chalcones and identified CA13 as a lead compound with potent and selective antiproliferative activity against non–small cell lung cancer (NSCLC) cells. CA13 triggered both apoptosis and autophagy in H292 cells. Western blotting and confocal imaging confirmed the activation of complete autophagic flux, while inhibition of autophagy markedly enhanced CA13-induced apoptosis, suggesting a cytoprotective role of autophagy. Mechanistically, CA13 activated JNK phosphorylation in a dose- and time-dependent manner, and pharmacological blockade of JNK significantly attenuated apoptotic signaling. In vivo, CA13 effectively suppressed H292 xenograft tumor growth without apparent systemic toxicity. Collectively, these results demonstrate that CA13 exerts its antitumor effects through JNK-dependent apoptosis accompanied by cytoprotective autophagy, providing a promising structural framework for the development of chalcone-based anticancer agents targeting programmed cell death pathways. Full article
Show Figures

Figure 1

19 pages, 5171 KB  
Article
Interferon-Type-I Response and Autophagy Independently Regulate Radiation-Induced HLA-Class-I Molecule Expression in Lung Cancer
by Erasmia T. Xanthopoulou, Ioannis Lamprou, Ioannis M. Koukourakis, Achilleas G. Mitrakas, Georgios D. Michos, Anastasia Polyzoidou, Filippos G. Antoniadis, Alexandra Giatromanolaki and Michael I. Koukourakis
Curr. Issues Mol. Biol. 2026, 48(1), 28; https://doi.org/10.3390/cimb48010028 - 25 Dec 2025
Viewed by 245
Abstract
Background/Objectives: The enhancement of antitumor immune responses by radiotherapy (RT) is partially attributed to the activation of the IFN-type-I pathway. However, the loss of HLA-class-I molecules, which occurs in a large percentage of non-small-cell lung cancers (NSCLCs), may block the cytotoxic effect [...] Read more.
Background/Objectives: The enhancement of antitumor immune responses by radiotherapy (RT) is partially attributed to the activation of the IFN-type-I pathway. However, the loss of HLA-class-I molecules, which occurs in a large percentage of non-small-cell lung cancers (NSCLCs), may block the cytotoxic effect of T-cells and immunotherapy (IO). Moreover, autophagy is also involved in HLA downregulation. We investigated the complex interactions between RT, HLA molecules, autophagy, and IFN-type-I responses. Methods: The A549, H1299, and ATG7-deficient NSCLC cell lines, along with the modified shLC3A H1299 cell line, were used for in vitro experiments. The effect of RT (8 and 3 × 8 Gy) on Interferon beta (IFNβ), IFN-stimulated genes (ISGs), and HLA-class-I expression in combination with IFN-type-I-response inhibitors (Ruxolitinib, Tofacitinib, Amlexanox) targeting the JAK and TBK1 was studied with Flow cytometry and RT-PCR. Results: RT significantly induced HLA-class-I expression. A parallel upregulation of IFNβ and ISGs mRNA levels was also documented. Although the IFN-type-I-response inhibitors suppressed the RT-induced IFNβ and ISGs expression, their effect on HLA-class-I expression was minimal. Blockage of LC3A autophagy (shLC3A cell line) significantly upregulated HLA-class-I basal levels, and RT further enhanced HLA expression. IFN-type-I-response inhibitors blocked the RT-inductive effect in the shLC3A H1299, but had no effect in the ATG7-deficient H1650 cell line. Conclusions: The current study supports the theory that baseline autophagy, RT-induced autophagy blockage, and IFN-type-I response enhancement define the HLA-class-I levels in NSCLC cells. This complex interplay emerges as a promising target for the development of radio-vaccination strategies to enhance the efficacy of radio-immunotherapy. Full article
(This article belongs to the Special Issue Molecular Insights into Radiation Oncology)
Show Figures

Figure 1

22 pages, 12677 KB  
Article
Biomass-Haze PM2.5 from Northern Thailand Drives Genotype-Specific Oxidative Stress and Transcriptomic Remodeling in Non-Small-Cell Lung Cancer Cells
by Sakawwarin Prommana, Sitthisak Intarasit, Saruda Thongyim, Nuttipon Yabueng, Somporn Chantara, Pachara Sattayawat, Aussara Panya and Sahutchai Inwongwan
Toxics 2026, 14(1), 21; https://doi.org/10.3390/toxics14010021 - 25 Dec 2025
Viewed by 500
Abstract
Fine particulate matter (PM2.5) is a major air pollutant linked to lung cancer progression. In Southeast Asia, seasonal smoke-haze produces biomass-derived PM2.5, yet its acute effects on genetically diverse lung tumours remain unclear. We investigate how Chiang Mai haze-derived PM2.5 impacts oxidative stress [...] Read more.
Fine particulate matter (PM2.5) is a major air pollutant linked to lung cancer progression. In Southeast Asia, seasonal smoke-haze produces biomass-derived PM2.5, yet its acute effects on genetically diverse lung tumours remain unclear. We investigate how Chiang Mai haze-derived PM2.5 impacts oxidative stress and gene expression in three non-small-cell lung cancer (NSCLC) cell lines: A549 (KRAS-mutant), NCI-H1975 (EGFR-mutant), and NCI-H460 (KRAS/PIK3CA-mutant). Cells were exposed to PM2.5 (0–200 µg/mL) and assessed for viability (MTT), reactive oxygen species (ROS; H2O2, •OH) and malondialdehyde (MDA) levels, mitochondrial-associated fluorescence, and whole-transcriptome responses. Acute exposure caused dose- and time-dependent viability loss, with A549 and NCI-H1975 more sensitive than NCI-H460. ROS profiling normalized to viable cells revealed genotype-specific oxidative patterns: cumulative increases in A549, sharp reversible spikes in NCI-H1975, and modest changes in NCI-H460. MitoTracker intensity trended downward without significance, with subtle fluorescence changes and particulate uptake. RNA-seq identified robust induction of xenobiotic metabolism (CYP1A1, CYP1B1), oxidative/metabolic stress mediators (GDF15, TIPARP), and tumour-associated genes (FOSB, VGF), alongside repression of tumour suppressors (FAT1, LINC00472). Pathway enrichment analyses highlighted oxidative stress, IL-17, NF-κB, and immune checkpoint signaling. Together, biomass haze-derived PM2.5 from Northern Thailand drives genotype-dependent oxidative stress and transcriptional remodeling in NSCLC cells. Full article
Show Figures

Figure 1

35 pages, 5474 KB  
Article
BRG1 (SMARCA4) Status Dictates the Response to EGFR Inhibitors in Wild-Type EGFR Non-Small Cell Lung Cancer
by Rebaz Ahmed, Ranganayaki Muralidharan, Narsireddy Amreddy, Akhil Srivastava, Meghna Mehta, Janani Panneerselvam, Rodrigo Orlandini de Castro, William L. Berry, Susmita Ghosh, Murali Ragothaman, Pawan Acharya, Yan D. Zhao, Roberto Jose Pezza, Anupama Munshi and Rajagopal Ramesh
Cancers 2026, 18(1), 62; https://doi.org/10.3390/cancers18010062 - 24 Dec 2025
Viewed by 374
Abstract
Background: Epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors (TKIs) have exhibited efficacy in EGFR-mutant non-small cell lung cancer (NSCLC) patients. However, the response is modest in patients with wild-type (wt)-EGFR, and approximately 30–40% of patients develop TKI resistance. Recently, a role [...] Read more.
Background: Epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors (TKIs) have exhibited efficacy in EGFR-mutant non-small cell lung cancer (NSCLC) patients. However, the response is modest in patients with wild-type (wt)-EGFR, and approximately 30–40% of patients develop TKI resistance. Recently, a role for BRG1 (SMARCA4) in regulating gene expression and its frequent alteration in various cancers, including NSCLC, has been reported. Yet, its specific function in response to EGFR-TKI therapy remains elusive. Herein, we investigated the role of BRG1 in EGFR-TKI response in vitro and in vivo using lung cancer models. Methods: In vitro, A549, H358, and HCC827 cell lines that varied in their EGFR and BRG1 status were assessed for response to EGFR-TKI upon overexpression or gene silencing of BRG1 through cell viability, cell migration, and Western blotting assays. In vivo, A549 and H358 tumor xenografts that overexpressed BRG1 or had BRG1 silenced were investigated for tumor growth response to EGFR-TKI. Results: EGFRwt/BRG1mt (A549) cells were resistant to TKI, and restoration of wt-BRG1 expression reverted them to TKI sensitivity both in vitro and in vivo. In contrast, silencing of BRG1wt in H358 cells showed a tendency toward TKI resistance. Additionally, wt-EGFR and pAKTSer473 protein complex formation in A549 cells was disrupted with an AKT inhibitor (MK2206), resulting in enhanced cytotoxicity in vitro. Conclusions: Our study demonstrates that EGFR-TKI response in wt-EGFR cells is dictated by BRG1 status. These findings propose screening of wt-EGFR NSCLC patients for BRG1 status for identifying individuals likely to benefit from EGFR-TKI therapy versus patients who will benefit from AKT inhibitor treatment. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

17 pages, 9440 KB  
Article
Gedunin Impacts Pancreatic Cancer Stem Cells Through the Sonic Hedgehog Signaling Pathway
by Karla Perez, Sheryl Rodriguez, Jose Barragan, Poornimadevi Narayanan, Alberto Ruiseco, Preetha Rajkumar, Nallely Ramirez, Victor Vasquez, Rajkumar Lakshmanaswamy and Ramadevi Subramani
Pharmaceuticals 2026, 19(1), 19; https://doi.org/10.3390/ph19010019 - 22 Dec 2025
Viewed by 300
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with a high rate of recurrence and a dismal prognosis. Studies have shown that pancreatic cancer stem cells (PCSCs) are a subpopulation that contributes to tumor progression, resistance to therapeutics, and metastasis, making [...] Read more.
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with a high rate of recurrence and a dismal prognosis. Studies have shown that pancreatic cancer stem cells (PCSCs) are a subpopulation that contributes to tumor progression, resistance to therapeutics, and metastasis, making them a key subpopulation to target for treatment. Gedunin (GD), a natural compound derived from Azadirachta indica (neem), has shown anticancer properties in pancreatic cancer cells, but its effects on PCSCs remains unclear. This study evaluated the effects of GD in pancreatic cancer stem cells, highlighting its impacts on tumor growth and progression and focusing on its impact on the sonic hedgehog (Shh) signaling pathway. Methods: Functional assays were performed to assess the effect of GD on the sphere-forming ability, colony formation, and self-renewal of PCSCs. Athymic mice xenograft models were utilized to evaluate the tumor suppression effect of GD in vivo. Furthermore, the anticancer effect of GD on PCSCs was assessed using both in vitro and in vivo limiting dilution assay. GD-induced changes in Shh signaling and key stem cell marker expressions in PCSCs were evaluated. Results: GD effectively inhibited tumor growth in xenograft models and reduced the percentage of PCSCs. GD was effective in decreasing PCSCs’ proliferative, self-renewal, and colony-forming capacity. GD decreased the protein expression levels of key Shh signaling markers Gli1 and Shh, stem cell markers SOX2, Nanog, and Oct4, metastasis-related proteins MMP-2, MMP-3, and MMP-9, and EMT markers Tgf1, Slug, Snail, and Twist in both PDAC cells and PCSCs. We demonstrated a significant decrease in the spheroid formation and self-renewal capacity of the (ALDH+) PCSC population following GD treatment in HPAC cells, indicating its potential antagonistic effects on PCSCs. GD was highly effective in reducing tumor volume, stemness, and metastasis in both early and late chemotherapy. In vivo limiting dilution assay using CD133+/LGR5+ PCSC xenografts demonstrated that GD reduces tumor growth, metastasis, and stemness associated with PCSCs by downregulating the expression of Shh and Gli1. GD treatment also reduced micrometastatic lesions in the lung, liver, and brain, as identified using H&E staining. Conclusions: The findings highlight GD’s potential as a promising therapeutic candidate for PDAC, with the ability to target both bulk tumor cells and PCSCs. By simultaneously suppressing tumor growth, stemness, and metastatic spread, GD may contribute to more effective treatment strategies and improved patient outcomes. Full article
(This article belongs to the Special Issue Anticancer Compounds in Medicinal Plants—4th Edition)
Show Figures

Graphical abstract

Back to TopTop