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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (249)

Search Parameters:
Keywords = on-target

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 2209 KB  
Article
Association Between Lipid-Lowering Therapy and Differences in the Distribution of LDL-C, apoB and non-HDL-C
by Marcin Ziółkowski, Karolina Obońska, Jakub Ratajczak, Piotr Adamski, Maciej Banach, Krzysztof Chlebus, Klaudyna Grzelakowska, Piotr Jankowski, Magdalena Krintus, Jacek Kryś, Ewa Laskowska, Natalia Mrzywka, Piotr Niezgoda, Małgorzata Ostrowska, Przemysław Podhajski, Grzegorz Skonieczny, Bożena Sosnowska, Łukasz Szarpak, Małgorzata Topolska, Julia Umińska, Alicja Rzepka-Cholasińska, Eliano Pio Navarese and Jacek Kubicaadd Show full author list remove Hide full author list
J. Clin. Med. 2026, 15(1), 26; https://doi.org/10.3390/jcm15010026 - 20 Dec 2025
Viewed by 208
Abstract
Background: The diagnosis of hypercholesterolemia relies on the laboratory assessment of lipid parameters. This study aimed to evaluate differences in the distribution of low-density lipoprotein cholesterol (LDL-C), non-high-density lipoprotein cholesterol (non-HDL-C), and apolipoprotein B (apoB) concentrations according to the presence and type of [...] Read more.
Background: The diagnosis of hypercholesterolemia relies on the laboratory assessment of lipid parameters. This study aimed to evaluate differences in the distribution of low-density lipoprotein cholesterol (LDL-C), non-high-density lipoprotein cholesterol (non-HDL-C), and apolipoprotein B (apoB) concentrations according to the presence and type of lipid-lowering therapy (LLT). Methods: This retrospective analysis included consecutive patients who had at least one measurement of LDL-C, apoB, and non-HDL-C between March and November 2024 in a high-volume tertiary hospital. All lipid fractions were expressed as the percentages of measurements above or below cut-off values established by the recent ESC guidelines. Subgroup analysis based on LLT type was performed, with patients categorized as receiving either single or combined LLT. Results: A total of 5048 patients were included in the analysis. Among patients receiving LLT, most were on statin monotherapy (77.3%), predominantly atorvastatin. Combined therapy, primarily statin plus ezetimibe, was used in 22.7% of treated patients. Discordance between on-target apoB levels and elevated LDL-C concentrations occurred in 26.6% of untreated and 13.6% of all treated patients, and in 15.1% and 8.6% of single and combined-LLT patients, respectively. Similarly, discordance between on-target non-HDL-C and elevated LDL-C levels was observed in 13.5% of untreated and 7.5% of all treated patients, and in 8.4% and 4.8% of single and combined-LLT patients, respectively. Conclusions: Classification of hyperlipidemia based on LDL-C, non-HDL-C, and apoB concentrations reveals significant discrepancies between these markers, especially between LDL-C and apoB. LLT reduces these discrepancies with combined LLT being particularly effective. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

18 pages, 841 KB  
Review
Cutaneous Adverse Events of Tyrosine Kinase Inhibitors in Endocrine Tumors: Clinical Features, Mechanisms, and Management Strategies
by Marta Marino, Francois Rosset, Alice Nervo, Alessandro Piovesan, Valentina Pala, Elisa Vaccaro, Luca Mastorino, Aldo E. Calogero and Emanuela Arvat
Biomedicines 2025, 13(12), 3044; https://doi.org/10.3390/biomedicines13123044 - 11 Dec 2025
Viewed by 386
Abstract
Background: Tyrosine kinase inhibitors (TKIs) are crucial to treating endocrine-related malignancies, including advanced thyroid cancers and neuroendocrine tumors, but their benefit is tempered by cutaneous adverse events (CAEs) that impair adherence and quality of life. Objective: To summarize the dermatologic toxicities of TKIs [...] Read more.
Background: Tyrosine kinase inhibitors (TKIs) are crucial to treating endocrine-related malignancies, including advanced thyroid cancers and neuroendocrine tumors, but their benefit is tempered by cutaneous adverse events (CAEs) that impair adherence and quality of life. Objective: To summarize the dermatologic toxicities of TKIs used in endocrine oncology and provide practical, multidisciplinary guidance for prevention and management. Methods: Narrative synthesis of clinical trial reports, post-marketing studies, and specialty guidelines pertinent to lenvatinib, vandetanib, cabozantinib, and other commonly used TKIs, integrating dermatologic and endocrine perspectives on mechanisms and care pathways. Results: VEGFR-targeted TKIs frequently cause hand–foot skin reaction, xerosis, fissuring, paronychia, and impaired wound healing; multikinase inhibition also produces alopecia, pigmentary changes, and mucositis. Epidermal growth factor receptor (EGFR) and rearranged during transfection (RET) inhibition with vandetanib is associated with acneiform eruption, photosensitivity, and nail fragility. Pathogenesis reflects on-target inhibition of VEGF/EGFR signaling leading to keratinocyte dysfunction, vascular fragility, and altered eccrine mechanics. Early risk stratification, patient education, and bundle-based prophylaxis (emollients, keratolytics, urea-based creams, sun protection) reduce incidence and severity. Grade-based algorithms combining topical corticosteroids/antibiotics, dose interruptions or reductions, and short systemic courses (e.g., doxycycline, antihistamines) enable symptom control while maintaining anticancer intensity. Close coordination around procedures minimizes wound-healing complications. Conclusions: Dermatologic toxicities are predictable, mechanism-linked, and manageable with proactive, multidisciplinary care. Standardized prevention and treatment pathways tailored to specific TKIs—particularly lenvatinib, vandetanib, and cabozantinib—can preserve dose intensity, optimize quality of life, and sustain antineoplastic efficacy. Full article
Show Figures

Figure 1

15 pages, 622 KB  
Review
Target Discovery in Head-and-Neck Squamous Cell Carcinoma: Genome-Wide CRISPR Screens Illuminate Therapeutic Resistance and Actionable Dependencies
by Vui King Vincent-Chong
Biomedicines 2025, 13(12), 3012; https://doi.org/10.3390/biomedicines13123012 - 8 Dec 2025
Viewed by 416
Abstract
Head-and-neck squamous cell carcinoma (HNSCC) remains a lethal malignancy with stagnant survival despite advances in surgery, radiotherapy, and systemic therapy. Beyond cetuximab and PD-1 inhibitors, there are only a few targeted options, which benefit only a minority of patients, underscoring the need for [...] Read more.
Head-and-neck squamous cell carcinoma (HNSCC) remains a lethal malignancy with stagnant survival despite advances in surgery, radiotherapy, and systemic therapy. Beyond cetuximab and PD-1 inhibitors, there are only a few targeted options, which benefit only a minority of patients, underscoring the need for new biomarkers and druggable dependencies. Genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) Cas9 screening now enables systematic, high-specificity investigation of gene function to reveal determinants of tumor proliferation, survival, and therapy response. Compared with RNA interference, CRISPR provides cleaner on-target knockout and more interpretable phenotypes, allowing efficient discovery of essential genes and synthetic-lethal interactions. Although the Cancer Dependency Map profiled 89 OSCC/HNSCC lines to nominate baseline dependencies, drug-perturbed states critical for understanding platinum resistance remain underexplored. Only a handful of HNSCC studies have applied genome-wide CRISPR cas9 screening: two mapped core essential genes; two mapped cisplatin resistance and radiation resistance; and others uncovered synthetic-lethal targets, including vulnerabilities to mTOR inhibition, EGFR inhibition, glutamine metabolism inhibition, and host determinants of oncolytic HSV-1 efficacy. This review synthesizes these findings, highlights methodological considerations (library design, coverage, and treatment duration), and integrates complementary functional data to prioritize targets for rational combinations. This review also provides information on the TCGA database and in vivo CRISPR screening that can accelerate precision therapeutics for patients with HNSCC. Full article
Show Figures

Figure 1

12 pages, 1798 KB  
Article
Mitochondrial Base Editing of the m.8993T>G Mutation Restores Bioenergetics and Neural Differentiation in Patient iPSCs
by Luke Yin, Angel Yin and Marjorie Jones
Genes 2025, 16(11), 1298; https://doi.org/10.3390/genes16111298 - 1 Nov 2025
Viewed by 576
Abstract
Background: Point mutations in mitochondrial DNA (mtDNA) cause a range of neurometabolic disorders that currently have no curative treatments. The m.8993T>G mutation in the Homo sapiens MT-ATP6 gene leads to neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP) when heteroplasmy exceeds approximately [...] Read more.
Background: Point mutations in mitochondrial DNA (mtDNA) cause a range of neurometabolic disorders that currently have no curative treatments. The m.8993T>G mutation in the Homo sapiens MT-ATP6 gene leads to neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP) when heteroplasmy exceeds approximately 70%. Methods: We engineered a split DddA-derived cytosine base editor (DdCBE), each half fused to programmable TALE DNA-binding domains and a mitochondrial targeting sequence, to correct the m.8993T>G mutation in patient-derived induced pluripotent stem cells (iPSCs). Seven days after plasmid delivery, deep amplicon sequencing showed 35 ± 3% on-target C•G→T•A conversion at position 8993, reducing mutant heteroplasmy from 80 ± 2% to 45 ± 3% with less than 0.5% editing at ten predicted off-target loci. Results: Edited cells exhibited a 25% increase in basal oxygen consumption rate, a 50% improvement in ATP-linked respiration, and a 2.3-fold restoration of ATP synthase activity. Directed neural differentiation yielded 85 ± 2% Nestin-positive progenitors compared to 60 ± 2% in unedited controls. Conclusions: Edits remained stable over 30 days in culture. These results establish mitochondrial base editing as a precise and durable strategy to ameliorate biochemical and cellular defects in NARP patient cells. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

39 pages, 4245 KB  
Review
Coumarin Derivatives as Anticancer Agents: Mechanistic Landscape with an Emphasis on Breast Cancer
by Veda B. Hacholli, Shubha M. R., Prabhanajan B. H., Lavanya M., Pramod S., Abhishek Kumar, Łukasz Szeleszczuk and Marcin Gackowski
Molecules 2025, 30(21), 4167; https://doi.org/10.3390/molecules30214167 - 23 Oct 2025
Cited by 1 | Viewed by 1574
Abstract
Coumarin derivatives constitute a versatile small-molecule chemotype with broad anticancer potential. This narrative review synthesizes recent in vitro and in vivo evidence on coumarin-based scaffolds, emphasizing breast cancer and covering lung, prostate, and colorectal models. We summarize major mechanisms of action—including induction of [...] Read more.
Coumarin derivatives constitute a versatile small-molecule chemotype with broad anticancer potential. This narrative review synthesizes recent in vitro and in vivo evidence on coumarin-based scaffolds, emphasizing breast cancer and covering lung, prostate, and colorectal models. We summarize major mechanisms of action—including induction of apoptosis (caspase activation and BAX/BCL-2 balance), modulation of PI3K/Akt/mTOR signaling, inhibition of angiogenesis (VEGFR-2), interference with estrogen biosynthesis (aromatase/ER axis), chaperone targeting (Hsp90), and attenuation of multidrug resistance (efflux pumps/autophagy)—and highlight representative chemotypes (e.g., benzimidazole, triazole, furocoumarins, topoisomerase- and CDK-oriented hybrids). Where available, we contrast potency and selectivity across models (e.g., MCF-7 vs. MDA-MB-231; A549; PC-3; colon lines) and discuss structure–activity trends linking substituent patterns (heteroaryl linkers, judicious halogenation, polar handles) to pathway engagement. We also delineate translational gaps limiting clinical progress—selectivity versus non-malignant cells, incomplete pharmacokinetic and safety characterization, and limited validation beyond xenografts. Finally, we outline priorities for preclinical optimization: biology-aligned target selection with biomarkers, resistance-aware combinations (e.g., PI3K/mTOR ± autophagy modulation; MDR mitigation), and early integration of ADME/tox and PK/PD to confirm on-target exposure. Collectively, the evidence supports coumarins as adaptable, multi-target anticancer leads, particularly promising in hormone-dependent breast cancer while remaining relevant to other tumor types. Full article
Show Figures

Figure 1

15 pages, 2416 KB  
Article
Engineering a High-Fidelity MAD7 Variant with Enhanced Specificity for Precision Genome Editing via CcdB-Based Bacterial Screening
by Haonan Zhang, Ying Yang, Tianxiang Yang, Peiyao Cao, Cheng Yu, Liya Liang, Rongming Liu and Zhiying Chen
Biomolecules 2025, 15(10), 1413; https://doi.org/10.3390/biom15101413 - 4 Oct 2025
Cited by 1 | Viewed by 972
Abstract
CRISPR (clustered regularly interspaced short palindromic repeats)-Cas (CRISPR-associated protein) nucleases enable precise genome editing, but off-target cleavage remains a critical challenge. Here, we report the development of MAD7_HF, a high-fidelity variant of the MAD7 nuclease engineered through a bacterial screening system leveraging the [...] Read more.
CRISPR (clustered regularly interspaced short palindromic repeats)-Cas (CRISPR-associated protein) nucleases enable precise genome editing, but off-target cleavage remains a critical challenge. Here, we report the development of MAD7_HF, a high-fidelity variant of the MAD7 nuclease engineered through a bacterial screening system leveraging the DNA gyrase-targeting toxic gene ccdB. This system couples survival to efficient on-target cleavage and minimal off-target activity, mimicking the transient action required for high-precision editing. Through iterative selection and sequencing validation, we identified MAD7_HF, harboring three substitutions (R187C, S350T, K1019N) that enhanced discrimination between on- and off-target sites. In Escherichia coli assays, MAD7_HF exhibited a >20-fold reduction in off-target cleavage across multiple mismatch contexts while maintaining on-target efficiency comparable to wild-type MAD7. Structural modeling revealed that these mutations stabilize the guide RNA-DNA hybrid at on-target sites and weaken interactions with mismatched sequences. This work establishes a high-throughput bacterial screening strategy that allows the identification of Cas12a variants with improved specificity at a given target site, providing a useful framework for future efforts to develop precision genome-editing tools. Full article
(This article belongs to the Special Issue Advances in Microbial CRISPR Editing)
Show Figures

Figure 1

16 pages, 2388 KB  
Article
Generation Using Phage-Display of pH-Dependent Antibodies Against the Tumor-Associated Antigen AXL
by Tristan Mangeat, Célestine Mairaville, Myriam Chentouf, Madeline Neiveyans, Martine Pugnière, Giang Ngo, Vincent Denis, Corentin Catherine, Alexandre Pichard, Emmanuel Deshayes, Margaux Maurel, Matthieu Gracia, Anne Bigot, Vincent Mouly, Sébastien Estaran, Alain Chavanieu, Pierre Martineau and Bruno Robert
Antibodies 2025, 14(4), 83; https://doi.org/10.3390/antib14040083 - 30 Sep 2025
Viewed by 1248
Abstract
Background/Objectives: Tumor-associated antigens are not tumor-specific antigens but proteins that are overexpressed by tumor cells and also weakly expressed at the surface of healthy tissues. Therefore, some side effects are observed when targeted by therapeutic antibodies, a phenomenon named “on-target, off-tumor toxicity”. As [...] Read more.
Background/Objectives: Tumor-associated antigens are not tumor-specific antigens but proteins that are overexpressed by tumor cells and also weakly expressed at the surface of healthy tissues. Therefore, some side effects are observed when targeted by therapeutic antibodies, a phenomenon named “on-target, off-tumor toxicity”. As tumors generate an acidic microenvironment, we investigated whether we could generate pH-dependent antibodies to increase their tumor specificity. For this proof-of-concept study, we selected the tyrosine kinase receptor AXL because we already developed several antibodies against this target. Methods: To generate a pH-dependent anti-AXL antibody, we performed classical panning of a single-chain variable fragment (scFv) library using phage display at an acidic pH throughout the process. Results: After the third round of panning, 9 scFvs, among the 96 picked clones, bound to AXL at acidic pH and showed very low binding at a neutral pH. After reformatting them into IgG, two clones were selected for further study due to their strong pH-sensitive binding. Using molecular docking and alanine scanning, we found that their binding strongly depended on two histidine residues present on AXL at positions 61 and 116. Conclusions: To conclude, we set-up an easy process to generate pH-dependent antibodies that may increase their tumor-binding specificity and potentially decrease toxicity towards healthy tissues. Full article
(This article belongs to the Section Antibody Discovery and Engineering)
Show Figures

Figure 1

24 pages, 884 KB  
Review
Nanopesticides in Brazilian Crops: Classes, Mechanisms, Efficacy, Risks, and Photocatalytic Remediation
by Tatiana Cardoso e Bufalo, Victor Hugo Buttrós, Aline Bastos de Paiva, Deyne Dehon de Oliveira, Caio Silas Ferreira Ribeiro and Joyce Dória
Plants 2025, 14(18), 2880; https://doi.org/10.3390/plants14182880 - 16 Sep 2025
Viewed by 928
Abstract
Brazil leads tropical agriculture, yet annual yield losses from insect pests and concerns over water contamination, non-target impacts, and resistance sustain demand for safer, more efficient control tools. This review synthesizes advances in nanopesticides for Brazil’s major crops (soybean, sugarcane, coffee, and citrus) [...] Read more.
Brazil leads tropical agriculture, yet annual yield losses from insect pests and concerns over water contamination, non-target impacts, and resistance sustain demand for safer, more efficient control tools. This review synthesizes advances in nanopesticides for Brazil’s major crops (soybean, sugarcane, coffee, and citrus) and is organized into five parts, comprising concepts and definitions; formulation families; modes of action; efficacy evidence from laboratory, greenhouse, and field studies; and environmental and human health risk considerations. Evidence indicates that nano-enabled delivery can increase on-target deposition, prolong residual activity, and match or exceed control at reduced active ingredient loads by improving foliar adhesion, transcuticular transport, plant uptake, and spatiotemporal targeting with pheromone-releasing nanofibers and other dispensers. Because nanoformulations can alter exposure pathways and environmental fate, this review emphasizes nano-specific physicochemical characterization under use conditions, fate and transport in Oxisols and Ultisols, and tropical waters, ecotoxicity panels that include pollinators, aquatic invertebrates, soil biota, and vertebrate surrogates, and scenario-based exposure assessment for applicators, residents, and consumers. The review closes with practical guidance for Brazil: head-to-head efficacy benchmarks against commercial standards, the standardized reporting of release and characterization data, and a nano-specific environmental risk assessment checklist to help realize efficacy gains while protecting environmental and human health. Full article
Show Figures

Figure 1

19 pages, 5487 KB  
Review
Recent Advances in the Development of Pro-PROTAC for Selective Protein Degradation
by Fady Hakem, Ahmad Abdelwaly, Reem Alshaman, Abdullah Alattar, Fawaz E. Alanazi, Sawsan A. Zaitone and Mohamed A. Helal
Pharmaceutics 2025, 17(9), 1160; https://doi.org/10.3390/pharmaceutics17091160 - 4 Sep 2025
Viewed by 2887
Abstract
PROTACs are trimeric small molecules consisting of a specific modulator of the target protein connected to a ligase-recruiting ligand via a suitably flexible linker. Ligase-recruiting ligands deliver ubiquitin ligases like E3 ligase to the Protein of Interest (POI). The vicinity of the POI-PROTAC-E3 [...] Read more.
PROTACs are trimeric small molecules consisting of a specific modulator of the target protein connected to a ligase-recruiting ligand via a suitably flexible linker. Ligase-recruiting ligands deliver ubiquitin ligases like E3 ligase to the Protein of Interest (POI). The vicinity of the POI-PROTAC-E3 ternary complex enables the E3 ligase to ubiquitinate the surface lysine residues of the POI. The Ubiquitin–Proteasome System (UPS) then degrades the POI. However, despite the considerable advances in the design of PROTACs targeting several types of enzymes and receptors, this strategy is still facing the challenges of precision target delivery and duration of action. In this review, we highlight the recent approaches for the development of PROTAC prodrugs or pro-PROTAC to control the delivery of PROTACs and achieve the required on-target exposure. This strategy may facilitate the application of the PROTAC technology and expand its clinical benefits. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

52 pages, 1118 KB  
Review
Advancing CAR T-Cell Therapy in Solid Tumors: Current Landscape and Future Directions
by Saeed Rafii, Deborah Mukherji, Ashok Sebastian Komaranchath, Charbel Khalil, Faryal Iqbal, Siddig Ibrahim Abdelwahab, Amin Abyad, Ahmad Y. Abuhelwa, Lakshmikanth Gandikota and Humaid O. Al-Shamsi
Cancers 2025, 17(17), 2898; https://doi.org/10.3390/cancers17172898 - 3 Sep 2025
Cited by 2 | Viewed by 11045
Abstract
Background: Chimeric Antigen Receptor (CAR) T-cell therapy has transformed the treatment of hematological malignancies, yet its application in solid tumors remains constrained by unique biological and logistical barriers. Objective: This review critically examines the evolving landscape of CAR T-cell therapy in solid malignancies, [...] Read more.
Background: Chimeric Antigen Receptor (CAR) T-cell therapy has transformed the treatment of hematological malignancies, yet its application in solid tumors remains constrained by unique biological and logistical barriers. Objective: This review critically examines the evolving landscape of CAR T-cell therapy in solid malignancies, with a focus on antigen heterogeneity, the immunosuppressive tumor microenvironment, and risks of on-target, off-tumor toxicity. Methods: We outline recent advances in CAR engineering, including co-stimulatory optimization, dual- and multi-antigen targeting, armored CARs, and gene-edited constructs designed to enhance persistence and anti-tumor activity. Clinical progress is highlighted by recent FDA approvals of genetically modified T-cell therapies in synovial sarcoma and melanoma, underscoring the potential for broader solid tumor application. Additionally, we synthesize early-phase clinical trial findings across multiple solid tumor types (e.g., lung, colorectal, ovarian, glioblastoma), and discuss innovative approaches such as regional delivery, checkpoint blockade combinations, and incorporation of chemokine receptors for improved tumor infiltration. The review also considers future strategies, including artificial intelligence-guided target discovery and rational trial design to overcome translational bottlenecks. Conclusions: With expanding clinical experience and continued technological innovation, CAR T-cell therapy is steadily transitioning from an experimental strategy to a therapeutic reality in solid tumors, poised to reshape the future of cancer immunotherapy. Full article
(This article belongs to the Special Issue CAR T Cells in Lymphoma and Multiple Myeloma)
Show Figures

Figure 1

21 pages, 2295 KB  
Article
Discovery of a Promising Hydroxyamino-Piperidine HDAC6 Inhibitor via Integrated Virtual Screening and Experimental Validation in Multiple Myeloma
by Federica Chiera, Antonio Curcio, Roberta Rocca, Ilenia Valentino, Massimo Gentile, Stefano Alcaro, Nicola Amodio and Anna Artese
Pharmaceuticals 2025, 18(9), 1303; https://doi.org/10.3390/ph18091303 - 29 Aug 2025
Cited by 1 | Viewed by 1400
Abstract
Background: Histone deacetylase 6 (HDAC6) is a unique class IIb HDAC isozyme characterized by two catalytic domains and a zinc finger ubiquitin-binding domain. It plays critical roles in various cellular processes, including protein degradation, autophagy, immune regulation, and cytoskeletal dynamics. Due to its [...] Read more.
Background: Histone deacetylase 6 (HDAC6) is a unique class IIb HDAC isozyme characterized by two catalytic domains and a zinc finger ubiquitin-binding domain. It plays critical roles in various cellular processes, including protein degradation, autophagy, immune regulation, and cytoskeletal dynamics. Due to its multifunctional nature and overexpression in several cancer types, HDAC6 has emerged as a promising therapeutic target. Methods: In this study, we employed a ligand-based pharmacophore modeling approach using a structurally diverse set of known HDAC6 inhibitors. This was followed by the virtual screening of over 140,000 commercially available compounds from both the MolPort and Asinex databases. The screening workflow incorporated pharmacophore filtering, molecular docking, and molecular dynamic (MD) simulations. Binding free energies were estimated using Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analysis to prioritize top candidates. A fluorometric enzymatic assay was used to measure HDAC6 activity, while cell viability assay by Cell Titer Glo was used to assess the anti-tumor activity against drug-sensitive and -resistant multiple myeloma (MM) cells. Western blotting was used to evaluate the acetylation of tubulin or histone H4 after treatment with selected compounds. Results: Three promising compounds were identified based on stable binding conformations and favorable interactions within the HDAC6 catalytic pocket. Among them, Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analysis identified Compound 10 (AKOS030273637) as the top theoretical binder, with a ΔGbind value of −45.41 kcal/mol. In vitro enzymatic assays confirmed its binding to the HDAC6 catalytic domain and inhibitory activity. Functional studies on MM cell lines, including drug-resistant variants, showed that Compound 10 reduced cell viability. Increased acetylation of α-tubulin, a substrate of HDAC6, likely suggested on-target mechanism of action. Conclusions: Compound 10, featuring a benzyl 4-[4-(hydroxyamino)-4-oxobutylidene] piperidine-1-carboxylate scaffold, demonstrates potential drug-like properties and a predicted bidentate zinc ion coordination, supporting its potential as an HDAC6 inhibitor for further development in hematologic malignancies. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

13 pages, 2203 KB  
Article
A Cancer-Specific Anti-Podocalyxin Monoclonal Antibody (humPcMab-60) Demonstrated Antitumor Efficacy in Pancreatic and Colorectal Cancer Xenograft Models
by Hiroyuki Suzuki, Tomokazu Ohishi, Takuro Nakamura, Miyuki Yanaka, Saori Handa, Tomohiro Tanaka, Mika K. Kaneko and Yukinari Kato
Antibodies 2025, 14(3), 67; https://doi.org/10.3390/antib14030067 - 11 Aug 2025
Viewed by 1207
Abstract
Background: Podocalyxin (PODXL) has been identified as a promising therapeutic target and a potential diagnostic biomarker in various tumors. Despite the therapeutic potential of anti-PODXL monoclonal antibodies (mAbs), their further development has been limited by concerns regarding potential on-target off-tumor toxicities. To [...] Read more.
Background: Podocalyxin (PODXL) has been identified as a promising therapeutic target and a potential diagnostic biomarker in various tumors. Despite the therapeutic potential of anti-PODXL monoclonal antibodies (mAbs), their further development has been limited by concerns regarding potential on-target off-tumor toxicities. To minimize adverse effects on normal tissues, developing a cancer-specific mAb (CasMab) against PODXL is essential. Methods: Our group established a cancer-specific anti-PODXL mAb, PcMab-60 (IgM, κ), through the screening of over one hundred hybridoma clones. In this study, PcMab-60 was engineered into a humanized IgG1-type mAb (humPcMab-60), and its antitumor activity was examined using mouse xenograft models of pancreatic ductal adenocarcinoma (PDAC) and colorectal cancer. Results: HumPcMab-60 retains cancer-specific reactivity; humPcMab-60 reacted to PDAC cell lines (PK-45H and MIA PaCa-2) and the colorectal cancer cell line (Caco-2), but not to a normal lymphatic endothelial cell line in flow cytometry. Furthermore, humPcMab-60 exerted antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity against PODXL-expressing cell lines and showed antitumor effects against the tumor xenografts. Conclusions: A humanized anti-PODXL CasMab, humPcMab-60, could be a promising mAb-based tumor therapy. Full article
Show Figures

Figure 1

14 pages, 546 KB  
Review
Belzutifan-Associated Hypoxia: A Review of the Novel Therapeutic, Proposed Mechanisms of Hypoxia, and Management Recommendations
by John Kucharczyk, Anshini Bhatt, Laura Bauer and Minas Economides
Int. J. Mol. Sci. 2025, 26(15), 7094; https://doi.org/10.3390/ijms26157094 - 23 Jul 2025
Viewed by 5827
Abstract
Belzutifan is a hypoxia-inducible factor-2α (HIF-2α) inhibitor that received FDA approval in 2021 for treating cancers resulting from von Hippel-Lindau (VHL) disease, including clear cell renal cell carcinoma (ccRCC), followed by approval in 2023 for sporadic ccRCC that has progressed through multiple lines [...] Read more.
Belzutifan is a hypoxia-inducible factor-2α (HIF-2α) inhibitor that received FDA approval in 2021 for treating cancers resulting from von Hippel-Lindau (VHL) disease, including clear cell renal cell carcinoma (ccRCC), followed by approval in 2023 for sporadic ccRCC that has progressed through multiple lines of therapy. HIF-2α is a promising drug target, as VHL is commonly inactivated in ccRCC, which results in HIF-2α-mediated signaling that is considered central to tumorigenesis. Belzutifan has demonstrated efficacy in clinical trials in the first-line and subsequent line settings, and in combination with tyrosine kinase inhibitors. Despite being overall well tolerated, belzutifan has a distinct safety profile because of its unique mechanism of action. Anemia was the most common adverse event observed in clinical trials and is considered an on-target effect. Hypoxia is also frequently observed and commonly results in dose reductions, treatment discontinuation, and supplemental oxygen use. This review summarizes the rates of hypoxia seen in clinical trials of belzutifan in ccRCC. As the cause of hypoxia is not well understood, this review also discusses possible mechanisms of hypoxia based on preclinical studies of the HIF pathway and HIF-2α inhibitors. Finally, this review proposes monitoring and management recommendations for clinicians prescribing belzutifan to ccRCC patients. Full article
(This article belongs to the Special Issue Recent Advances in Urological Cancer)
Show Figures

Figure 1

29 pages, 2844 KB  
Review
Hsp90 pan and Isoform-Selective Inhibitors as Sensitizers for Cancer Immunotherapy
by Shiying Jia, Neeraj Maurya, Brian S. J. Blagg and Xin Lu
Pharmaceuticals 2025, 18(7), 1025; https://doi.org/10.3390/ph18071025 - 10 Jul 2025
Cited by 2 | Viewed by 3499
Abstract
The 90 kDa heat shock proteins (Hsp90) are molecular chaperones that regulate the stability and maturation of numerous client proteins implicated in the regulation of cancer hallmarks. Despite the potential of pan-Hsp90 inhibitors as anticancer therapeutics, their clinical development has been hindered [...] Read more.
The 90 kDa heat shock proteins (Hsp90) are molecular chaperones that regulate the stability and maturation of numerous client proteins implicated in the regulation of cancer hallmarks. Despite the potential of pan-Hsp90 inhibitors as anticancer therapeutics, their clinical development has been hindered by on-target toxicities, particularly ocular and cardiotoxic effects, as well as the induction of pro-survival, compensatory heat shock responses. Together, these and other complications have prompted the development of isoform-selective Hsp90 inhibitors. In this review, we discuss the molecular bases for Hsp90 function and inhibition and emphasize recent advances in isoform-selective targeting. Importantly, we highlight how Hsp90 inhibition can sensitize tumors to cancer immunotherapy by enhancing antigen presentation, reducing immune checkpoint expression, remodeling the tumor microenvironment, and promoting innate immune activation. Special focus is given to Hsp90β-selective inhibitors, which modulate immunoregulatory pathways without eliciting the deleterious effects observed with pan-inhibition. Preclinical and early clinical data support the integration of Hsp90 inhibitors with immune checkpoint blockade and other immunotherapeutic modalities to overcome resistance mechanisms in immunologically cold tumors. Therefore, the continued development of isoform-selective Hsp90 inhibitors offers a promising avenue to potentiate cancer immunotherapy with improved efficacy. Full article
Show Figures

Graphical abstract

11 pages, 2422 KB  
Article
Cross-Activity Analysis of CRISPR/Cas9 Editing in Gene Families of Solanum lycopersicum Detected by Long-Read Sequencing
by Ofri Kutchinsky, Dongqi Li, Guy Assa, Asaph Aharoni and Zohar Yakhini
Curr. Issues Mol. Biol. 2025, 47(7), 507; https://doi.org/10.3390/cimb47070507 - 2 Jul 2025
Viewed by 816
Abstract
CRISPR/Cas9 genome editing holds promise for precise genetic modifications, yet off-target effects remain a concern—particularly in gene families with high sequence similarity. In this study, we present a computational framework for analyzing editing specificity and cross-reactivity in gene families using long-read sequencing data. [...] Read more.
CRISPR/Cas9 genome editing holds promise for precise genetic modifications, yet off-target effects remain a concern—particularly in gene families with high sequence similarity. In this study, we present a computational framework for analyzing editing specificity and cross-reactivity in gene families using long-read sequencing data. The pipeline integrates multiplex PCR, NGS, and CRISPECTOR-based analysis to detect and quantify on- and off-target events with high sensitivity. As a use case, we applied this framework to Solanum lycopersicum, evaluating on-target editing in thirteen gene families and analyzing off-target cross-reactivity in five representative families. While the biological results are illustrative, the primary contribution lies in the generalizable analysis approach, which can support genome editing studies in complex plant genomes and beyond. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

Back to TopTop