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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (873)

Search Parameters:
Keywords = tumor penetration

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 10639 KB  
Article
Taming the Tumor Stroma: A Two-Stage Targeted Nanocapsule for Potent Deep Chemo-Immunotherapy in Triple-Negative Breast Cancer
by Bin Xing, Xinru Shen, Xintao Jia, Ying Zhang, Zhongyan Liu, Xueli Guo, Xin Li and Zhidong Liu
Pharmaceutics 2026, 18(2), 184; https://doi.org/10.3390/pharmaceutics18020184 - 30 Jan 2026
Abstract
Background: The tumor microenvironment (TME) poses significant challenges to effective therapy, with cancer-associated fibroblasts (CAFs) playing a key role in tumor progression and drug resistance in triple-negative breast cancer (TNBC). Herein, a TME responsive nanocapsule, NPC-ABS/FDS, was developed utilizing baicalein, a CAFs [...] Read more.
Background: The tumor microenvironment (TME) poses significant challenges to effective therapy, with cancer-associated fibroblasts (CAFs) playing a key role in tumor progression and drug resistance in triple-negative breast cancer (TNBC). Herein, a TME responsive nanocapsule, NPC-ABS/FDS, was developed utilizing baicalein, a CAFs modulator, and the cytotoxic drug doxorubicin to selectively target CAFs and tumor cells, respectively, in a stepwise manner. Methods: NPC-ABS/FDS was designed with CD13-mediated primary targeting for tumor accumulation and secondary targeting via σ-receptor binding (ABS nanoparticles) for CAFs and folate modification (FDS nanoparticles) for cancer cells. Physicochemical properties were assessed using TEM, particle size, and ζ-potential analyses. Fluorescence imaging evaluated tumor retention, while cellular uptake and TME modulation were analyzed in vitro and in vivo. Results: The successful preparation of NPC-ABS/FDS was demonstrated by its uniform morphology, stable characteristics, charge reversal, and increased particle size. Fluorescence imaging confirmed prolonged peritumoral retention. Cellular uptake increased 2.5-fold for baicalein in CAFs and 4.3-fold for doxorubicin in cancer cells. NPC-ABS/FDS downregulated α-SMA and FAP, reducing CAFs activation, improving intratumoral drug penetration, and enhancing CD8+ and CD4+ T cell infiltration while decreasing regulatory T cells. Conclusions: NPC-ABS/FDS effectively modulates multiple TME components, including CAFs and immune cells, and improves drug delivery in TNBC. These findings may support the development of improved therapeutic approaches for TNBC. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

29 pages, 890 KB  
Review
Nanotechnology-Enabled Precision Therapy for Lung Cancer in Never-Smokers
by Cristian Cojocaru, Adina Magdalena Țurcanu, Ruxandra Cojocaru and Elena Cojocaru
Pharmaceutics 2026, 18(2), 161; https://doi.org/10.3390/pharmaceutics18020161 - 26 Jan 2026
Viewed by 104
Abstract
Lung cancer in never-smokers (LCINS) represents a distinct clinical entity driven by dominant oncogenic alterations and characterized by a low tumor mutational burden. Although tyrosine kinase inhibitors (TKIs) achieve high initial response rates, their long-term efficacy is limited by suboptimal pharmacokinetics, restricted central [...] Read more.
Lung cancer in never-smokers (LCINS) represents a distinct clinical entity driven by dominant oncogenic alterations and characterized by a low tumor mutational burden. Although tyrosine kinase inhibitors (TKIs) achieve high initial response rates, their long-term efficacy is limited by suboptimal pharmacokinetics, restricted central nervous system (CNS) penetration, tumor microenvironment barriers, and acquired resistance. In this review, we critically assess the current state of nanotechnology-assisted drug delivery systems for LCINS, with a primary focus on how rationally designed nanocarriers can overcome biological barriers, enable molecular subtype-specific therapeutic strategies, and address mechanisms that limit clinical efficacy and durability of response. We conducted a structured literature search using PubMed and Web of Science (January 2022 to November 2025), focusing on primary studies reporting the preparation, physicochemical properties, and therapeutic performance of nanocarriers in in vitro and in vivo models, as well as available pharmacokinetic and clinical data. LCINS is characterized by inefficient vasculature, high extracellular matrix density, active efflux transporters, and immunosuppressive niches, and is frequently complicated by brain metastases. Nanocarrier-based platforms can enhance aqueous solubility, prolong systemic circulation, and improve tumor or CNS targeting. Co-delivery systems combining TKIs with nucleic acid-based therapeutics, together with stimuli-responsive platforms, offer the potential for simultaneous modulation of multiple oncogenic pathways and partial mitigation of resistance mechanisms. In summary, nanotechnology provides a promising strategy to improve both the efficacy and specificity of targeted therapies in LCINS. Successful clinical translation will depend on biologically aligned carrier–payload combinations, scalable and reproducible manufacturing processes, and biomarker-guided patient selection. Full article
Show Figures

Graphical abstract

15 pages, 3536 KB  
Review
Extracellular Matrix in Human Disease and Therapy: From Pathogenic Remodeling to Biomaterial Platforms and Precision Diagnostics
by Jun-Hyeog Jang
Biomedicines 2026, 14(1), 247; https://doi.org/10.3390/biomedicines14010247 - 21 Jan 2026
Viewed by 177
Abstract
The extracellular matrix (ECM) is a dynamic, tissue-specific network that integrates biochemical and mechanical cues to regulate cell behavior and organ homeostasis. Increasing evidence indicates that dysregulated ECM remodeling is an upstream driver of chronic human diseases rather than a passive consequence of [...] Read more.
The extracellular matrix (ECM) is a dynamic, tissue-specific network that integrates biochemical and mechanical cues to regulate cell behavior and organ homeostasis. Increasing evidence indicates that dysregulated ECM remodeling is an upstream driver of chronic human diseases rather than a passive consequence of injury. This review summarizes principles of ECM organization, mechanotransduction, and pathological remodeling and highlights translational opportunities for ECM-targeted therapies, biomaterial platforms, and precision diagnostics. We conducted a narrative synthesis of foundational and recent literature covering ECM composition and turnover, stiffness-dependent signaling, and disease-associated remodeling across fibrosis/cardiovascular disease, cancer, and metabolic disorders, together with advances in ECM-based biomaterials, drug delivery, and ECMderived biomarkers and imaging. Across organs, a self-reinforcing cycle of altered matrix composition, excessive crosslinking, and stiffness-dependent mechanotransduction (including integrin–FAK and YAP/TAZ pathways) sustains fibroinflammation, myofibroblast persistence, and progressive tissue dysfunction. In tumors, aligned and crosslinked ECM promotes invasion, immune evasion, and therapy resistance while also shaping perfusion and drug penetration. Translational strategies increasingly focus on modulating ECM synthesis and crosslinking, normalizing rather than ablating matrix architecture, and targeting ECM–cell signaling axes in combination with anti-fibrotic, cytotoxic, or immunotherapeutic regimens. ECM biology provides a unifying framework linking pathogenesis, therapy, and precision diagnostics across chronic diseases. Clinical translation will benefit from standardized quantitative measures of matrix remodeling, mechanism-based biomarkers of ECM turnover, and integrative imaging–omics approaches for patient stratification and treatment monitoring. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

18 pages, 1428 KB  
Review
The Glymphatic–Immune Axis in Glioblastoma: Mechanistic Insights and Translational Opportunities
by Joaquin Fiallo Arroyo and Jose E. Leon-Rojas
Int. J. Mol. Sci. 2026, 27(2), 928; https://doi.org/10.3390/ijms27020928 - 16 Jan 2026
Viewed by 344
Abstract
Glioblastoma (GBM) remains one of the most treatment-resistant human malignancies, largely due to the interplay between disrupted fluid dynamics, immune evasion, and the structural complexity of the tumor microenvironment; in addition to these, treatment resistance is also driven by intratumoral heterogeneity, glioma stem [...] Read more.
Glioblastoma (GBM) remains one of the most treatment-resistant human malignancies, largely due to the interplay between disrupted fluid dynamics, immune evasion, and the structural complexity of the tumor microenvironment; in addition to these, treatment resistance is also driven by intratumoral heterogeneity, glioma stem cell persistence, hypoxia-induced metabolic and epigenetic plasticity, adaptive oncogenic signaling, and profound immunosuppression within the tumor microenvironment. Emerging evidence shows that dysfunction of the glymphatic system, mislocalization of aquaporin-4, and increased intracranial pressure compromise cerebrospinal fluid–interstitial fluid exchange and impair antigen drainage to meningeal lymphatics, thereby weakening immunosurveillance. GBM simultaneously remodels the blood–brain barrier into a heterogeneous and permeable blood–tumor barrier that restricts uniform drug penetration yet enables tumor progression. These alterations intersect with profound immunosuppression mediated by pericytes, tumor-associated macrophages, and hypoxic niches. Advances in imaging, including DCE-MRI, DTI-ALPS, CSF-tracing PET, and elastography, now allow in vivo characterization of glymphatic function and interstitial flow. Therapeutic strategies targeting the fluid-immune interface are rapidly expanding, including convection-enhanced delivery, intrathecal and intranasal approaches, focused ultrasound, nanoparticle systems, and lymphatic-modulating immunotherapies such as VEGF-C and STING agonists. Integrating barrier modulation with immunotherapy and nanomedicine holds promise for overcoming treatment resistance. Our review synthesizes the mechanistic, microenvironmental, and translational advances that position the glymphatic–immune axis as a new frontier in glioblastoma research. Full article
Show Figures

Figure 1

26 pages, 2484 KB  
Review
Effective Non-Invasive Delivery of Epigenetic Drugs Using Functionalized Accessory Unit Conjugates
by Toshihiko Tashima
Pharmaceutics 2026, 18(1), 115; https://doi.org/10.3390/pharmaceutics18010115 - 15 Jan 2026
Viewed by 344
Abstract
Epigenetics involves heritable changes in gene expression—such as DNA methylation (5-methylcytosine; 5mC), histone modifications, and regulation by non-coding RNAs at the mRNA translation level—without altering the underlying DNA sequence. As targeting these mechanisms enables intervention at the root cause of disease rather than [...] Read more.
Epigenetics involves heritable changes in gene expression—such as DNA methylation (5-methylcytosine; 5mC), histone modifications, and regulation by non-coding RNAs at the mRNA translation level—without altering the underlying DNA sequence. As targeting these mechanisms enables intervention at the root cause of disease rather than the symptoms alone, epigenetics has become a rapidly advancing field in pharmaceutical sciences. Various epigenetic modulators, including histone deacetylase (HDAC) inhibitors, histone acetyltransferase (HAT) inhibitors, DNA methyltransferase (DNMT) inhibitors, and microRNAs (miRNAs), have been developed, and some have already been approved for cancer therapy. However, these agents often face significant challenges such as poor membrane permeability, enzymatic instability, and suboptimal biodistribution. Incorporating functionalized accessory units—serving as vectors (e.g., transporter recognition units, cell-penetrating peptides, tumor-homing peptides, monoclonal antibodies) or as carriers (e.g., monoclonal antibodies, nanoparticles)—into epigenetic modulators may help overcome these delivery barriers. In this narrative review, I discuss the potential and advantages of effective non-invasive delivery of epigenetic drugs using such functionalized accessory unit conjugates. Full article
(This article belongs to the Special Issue Development and Drug Delivery in Epigenetic Therapy)
Show Figures

Figure 1

37 pages, 2140 KB  
Review
Functional Peptide-Based Biomaterials for Pharmaceutical Application: Sequences, Mechanisms, and Optimization Strategies
by Dedong Yu, Nari Han, Hyejeong Son, Sun Jo Kim and Seho Kweon
J. Funct. Biomater. 2026, 17(1), 37; https://doi.org/10.3390/jfb17010037 - 13 Jan 2026
Viewed by 619
Abstract
Peptide-based biomaterials have emerged as versatile tools for pharmaceutical drug delivery due to their biocompatibility and tunable sequences, yet a comprehensive overview of their categories, mechanisms, and optimization strategies remains lacking to guide clinical translation. This review systematically collates advances in peptide-based biomaterials, [...] Read more.
Peptide-based biomaterials have emerged as versatile tools for pharmaceutical drug delivery due to their biocompatibility and tunable sequences, yet a comprehensive overview of their categories, mechanisms, and optimization strategies remains lacking to guide clinical translation. This review systematically collates advances in peptide-based biomaterials, covering peptide excipients (cell penetrating peptides, tight junction modulating peptides, and peptide surfactants/stabilizers), self-assembling peptides (peptide-based nanospheres, cyclic peptide nanotubes, nanovesicles and micelles, peptide-based hydrogels and depots), and peptide linkers (for antibody drug-conjugates, peptide drug-conjugates, and prodrugs). We also dissect sequence-based optimization strategies, including rational design and biophysical optimization (cyclization, stapling, D-amino acid incorporation), functional motif integration, and combinatorial discovery with AI assistance, with examples spanning marketed drugs and research-stage candidates. The review reveals that cell-penetrating peptides enable efficient intracellular payload delivery via direct penetration or endocytosis; self-assembling peptides form diverse nanostructures for controlled release; and peptide linkers achieve site-specific drug release by responding to tumor-associated enzymes or pH cues, while sequence optimization enhances stability and targeting. Peptide-based biomaterials offer precise, biocompatible and tunable solutions for drug delivery, future advancements relying on AI-driven design and multi-functional modification will accelerate their transition from basic research to clinical application. Full article
Show Figures

Figure 1

16 pages, 1343 KB  
Article
GPA33-Targeted Trimeric Immunotoxin Exhibits Enhanced Antitumor Activity in Human Colorectal Cancer Xenografts
by Javier Ruiz-de-la-Herrán, Javier Narbona, Rubén G. Gordo, Laura Sanz and Javier Lacadena
Int. J. Mol. Sci. 2026, 27(2), 764; https://doi.org/10.3390/ijms27020764 - 12 Jan 2026
Viewed by 315
Abstract
Immunotoxins are chimeric molecules with high potential as therapeutic candidates that combine antibody specificity to recognize and bind tumor-associated antigens and the cytotoxic potency of the enzymatic activity of a toxin, leading to the selective death of target cells. The use of immunotoxins [...] Read more.
Immunotoxins are chimeric molecules with high potential as therapeutic candidates that combine antibody specificity to recognize and bind tumor-associated antigens and the cytotoxic potency of the enzymatic activity of a toxin, leading to the selective death of target cells. The use of immunotoxins as therapeutic tools remains limited by various issues, such as selecting the appropriate tumor-associated antigen (TAA), penetration difficulties in solid tumors, low renal clearance, and low toxic payload. For this purpose, in this work we have designed a novel trimeric immunotoxin (IMTXTriA33αS) against colorectal cancer, combining the scFv against GPA33 as a targeting domain and the fungal ribotoxin α-sarcin (αS) as the toxic fragment, linked by a trimerization domain (TIEXVIII). Our results demonstrate that IMTXTriA33αS has greater avidity and toxic load, showing a very significant increase in its in vitro and in vivo antitumor efficacy, due to its trimeric structure. Full article
(This article belongs to the Special Issue Molecular Diagnosis and Treatment of Colorectal Cancer)
Show Figures

Graphical abstract

27 pages, 1311 KB  
Review
Peptide-Functionalized Iron Oxide Nanoparticles for Cancer Therapy: Targeting Strategies, Mechanisms, and Translational Opportunities
by Andrey N. Kuskov, Lydia-Nefeli Thrapsanioti, Ekaterina Kukovyakina, Anne Yagolovich, Elizaveta Vlaskina, Petros Tzanakakis, Aikaterini Berdiaki and Dragana Nikitovic
Molecules 2026, 31(2), 236; https://doi.org/10.3390/molecules31020236 - 10 Jan 2026
Viewed by 453
Abstract
Therapeutic peptides have emerged as promising tools in oncology due to their high specificity, favorable safety profile, and capacity to target molecular hallmarks of cancer. Their clinical translation, however, remains limited by poor stability, rapid proteolytic degradation, and inefficient biodistribution. Iron oxide nanoparticles [...] Read more.
Therapeutic peptides have emerged as promising tools in oncology due to their high specificity, favorable safety profile, and capacity to target molecular hallmarks of cancer. Their clinical translation, however, remains limited by poor stability, rapid proteolytic degradation, and inefficient biodistribution. Iron oxide nanoparticles (IONPs) offer a compelling solution to these challenges. Owing to their biocompatibility, magnetic properties, and ability to serve as both drug carriers and imaging agents, IONPs have become a versatile platform for precision nanomedicine. The integration of peptides with IONPs has generated a new class of hybrid systems that combine the biological accuracy of peptide ligands with the multifunctionality of magnetic nanomaterials. Peptide functionalization enables selective tumor targeting and deeper tissue penetration, while the IONP core supports controlled delivery, MRI-based tracking, and activation of therapeutic mechanisms such as magnetic hyperthermia. These hybrids also influence the tumor microenvironment (TME), facilitating stromal remodeling and improved drug accessibility. Importantly, the iron-driven redox chemistry inherent to IONPs can trigger regulated cell death pathways, including ferroptosis and autophagy, inhibiting opportunities to overcome resistance in aggressive or refractory tumors. As advances in peptide engineering, nanotechnology, and artificial intelligence accelerate design and optimization, peptide–IONP conjugates are poised for translational progress. Their combined targeting precision, imaging capability, and therapeutic versatility position them as promising candidates for next-generation cancer theranostics. Full article
Show Figures

Graphical abstract

19 pages, 6074 KB  
Article
Albumin Nanoparticles Harness Activated Neutrophils to Cross Vascular Barriers for Targeted Subcutaneous and Orthotopic Colon Cancer Therapy
by Zhifan Luo, Liuqing Dong, Yujie Zhang and Mingzhen Zhang
J. Funct. Biomater. 2026, 17(1), 36; https://doi.org/10.3390/jfb17010036 - 10 Jan 2026
Viewed by 436
Abstract
Colorectal cancer (CRC) therapy faces challenges due to limited drug penetration across the blood–tumor barrier. Neutrophils, with their natural ability to migrate to inflamed and tumor sites, offer a promising cell-mediated delivery strategy. This study developed albumin nanoparticles loaded with 6-shogaol (NPs/6-shogaol) and [...] Read more.
Colorectal cancer (CRC) therapy faces challenges due to limited drug penetration across the blood–tumor barrier. Neutrophils, with their natural ability to migrate to inflamed and tumor sites, offer a promising cell-mediated delivery strategy. This study developed albumin nanoparticles loaded with 6-shogaol (NPs/6-shogaol) and utilized activated neutrophils as carriers to transport the nanoparticles across vascular barriers for colon cancer therapy. The physicochemical properties, biocompatibility, and targeting efficiency of the NPs were evaluated in vitro and in vivo. The formulated NPs/6-shogaol exhibited favorable physicochemical properties, including a uniform nano-scale size (~150 nm), negative zeta potential, and high drug loading efficiency. In both subcutaneous and orthotopic colon cancer models, neutrophil-mediated delivery significantly enhanced tumor accumulation of 6-shogaol, inhibited tumor growth, and induced apoptosis by suppressing neutrophil elastase (NE) expression. Notably, no significant systemic toxicity was observed. This neutrophil-hitchhiking albumin nanoplatform provides a targeted and biocompatible strategy for effective colon cancer therapy. Full article
(This article belongs to the Special Issue Nanomaterials for Drug Targeting and Drug Delivery (2nd Edition))
Show Figures

Figure 1

39 pages, 1558 KB  
Review
Rewriting Tumor Entry Rules: Microfluidic Polyplexes and Tumor-Penetrating Strategies—A Literature Review
by Simona Ruxandra Volovat, Iolanda Georgiana Augustin, Constantin Volovat, Ingrid Vasilache, Madalina Ostafe, Diana Ioana Panaite, Alin Burlacu and Cristian Constantin Volovat
Pharmaceutics 2026, 18(1), 84; https://doi.org/10.3390/pharmaceutics18010084 - 9 Jan 2026
Viewed by 423
Abstract
Cancer immunotherapy increasingly relies on nucleic acid-based vaccines, yet achieving efficient and safe delivery remains a critical limitation. Polyplexes—electrostatic complexes of cationic polymers and nucleic acids—have emerged as versatile carriers offering greater chemical tunability and multivalent targeting capacity compared to lipid nanoparticles, with [...] Read more.
Cancer immunotherapy increasingly relies on nucleic acid-based vaccines, yet achieving efficient and safe delivery remains a critical limitation. Polyplexes—electrostatic complexes of cationic polymers and nucleic acids—have emerged as versatile carriers offering greater chemical tunability and multivalent targeting capacity compared to lipid nanoparticles, with lower immunogenicity than viral vectors. This review summarizes key design principles governing polyplex performance, including polymer chemistry, architecture, and assembly route—emphasizing microfluidic fabrication for improved size control and reproducibility. Mechanistically, effective systems support stepwise delivery: tumor targeting, cellular uptake, endosomal escape (via proton-sponge, membrane fusion, or photochemical disruption), and compartment-specific cargo release. We discuss therapeutic applications spanning plasmid DNA, siRNA, miRNA, mRNA, and CRISPR-based editing, highlighting preclinical data across multiple tumor types and early clinical evidence of on-target knockdown in human cancers. Particular attention is given to physiological barriers and engineering strategies—including size-switching systems, charge-reversal polymers, and tumor-penetrating peptides—that improve intratumoral distribution. However, significant challenges persist, including cationic toxicity, protein corona formation, manufacturing variability, and limited clinical responses to date. Current evidence supports polyplexes as a modular platform complementary to lipid nanoparticles in selected oncology indications, though realizing this potential requires continued optimization alongside rigorous translational development. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

20 pages, 4835 KB  
Article
Cell-Penetrating Peptide-Mediated siRNA Targeting of LDHC Suppresses Tumor Growth in a Triple-Negative Breast Cancer Zebrafish Xenograft Model
by Hanan Qasem, Adviti Naik, Tricia Gomez, Janarthanan Ponraj, Umar Jafar, Martin Sikhondze, Remy Thomas, Khaled A. Mahmoud and Julie Decock
Pharmaceutics 2026, 18(1), 78; https://doi.org/10.3390/pharmaceutics18010078 - 7 Jan 2026
Viewed by 368
Abstract
Background: Lactate Dehydrogenase C (LDHC) is a promising therapeutic target due to its highly tumor-specific expression, immunogenicity, and oncogenic functions. We previously showed that LDHC silencing in triple-negative breast cancer (TNBC) cells enhances treatment response to DNA-damage response-related drugs, supporting its therapeutic [...] Read more.
Background: Lactate Dehydrogenase C (LDHC) is a promising therapeutic target due to its highly tumor-specific expression, immunogenicity, and oncogenic functions. We previously showed that LDHC silencing in triple-negative breast cancer (TNBC) cells enhances treatment response to DNA-damage response-related drugs, supporting its therapeutic potential. However, no selective LDHC inhibitors exist, highlighting the need for innovative targeting strategies. Methods: We assessed the physicochemical properties and evaluated the delivery efficiency, anti-tumor activity, and safety of four cell-penetrating peptides (CPPs)—R10, 10R-RGD, cRGD-10R, and iRGD-10R—for siRNA-mediated LDHC silencing in TNBC. Clonogenic assays were used to evaluate effects on olaparib sensitivity, and TNBC zebrafish xenografts were utilized to study in vivo anti-tumor activity. Results: All CPP:siRNA complexes formed uniform nanocomplexes (129–168 nm) with low polydispersity indices (<0.25) and positive zeta potentials (+6.47 to +29.6 mV). Complexes remained stable in human serum for 24 h and showed no significant cytotoxicity in TNBC and non-cancerous cell lines. The 10R-RGD and cRGD-10R:siLDHC complexes achieved 40% LDHC protein knockdown, reduced TNBC clonogenicity by 30–36%, and enhanced olaparib sensitivity. Treatment of TNBC zebrafish xenografts with 10R-RGD or cRGD-10R:siLDHC complexes significantly reduced tumor growth by approximately 50% without major toxicity. Conclusions: These results demonstrate that CPP-mediated siRNA delivery enables selective LDHC silencing with tumor growth inhibition in triple-negative breast cancer models. This approach represents a novel, effective, and safe proof-of-concept therapeutic strategy to target LDHC, with potential translational relevance as a standalone therapy or in combination with common anti-cancer drugs. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

29 pages, 1038 KB  
Review
Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities
by Dimitrios Vrachas, Elisavet Kosma, Angeliki-Ioanna Giannopoulou, Angeliki Margoni, Antonios N. Gargalionis, Elias A. El-Habr, Christina Piperi and Christos Adamopoulos
Cancers 2026, 18(1), 156; https://doi.org/10.3390/cancers18010156 - 2 Jan 2026
Viewed by 593
Abstract
Central nervous system (CNS) tumors consist of a diverse set of malignancies that remain clinically challenging due to their biological complexity, high morbidity, and limited responsiveness to current therapies. A growing body of genomic evidence has revealed that dysregulation of the mitogen-activated protein [...] Read more.
Central nervous system (CNS) tumors consist of a diverse set of malignancies that remain clinically challenging due to their biological complexity, high morbidity, and limited responsiveness to current therapies. A growing body of genomic evidence has revealed that dysregulation of the mitogen-activated protein kinase (MAPK) signaling pathway is a recurrent and unifying characteristic across many pediatric and adult CNS tumor entities. Alterations affecting upstream receptor tyrosine kinases (RTKs), RAS GTPases, RAF kinases, and other associated regulators contribute to MAPK signaling pathway hyperactivation, shaping tumor behavior, therapy response and resistance. These aberrations ranging from hotspot mutations such as BRAF V600E and oncogenic fusions like BRAF–KIAA1549 are particularly enriched in gliomas and glioneuronal tumors, highlighting MAPK signaling as a key oncogenic driver. The expanding availability of molecularly targeted compounds, including selective inhibitors of RAF, MEK and ERK, has begun to transform treatment approaches for specific molecular subtypes. However, the clinical benefit of MAPK-directed therapies is frequently limited by restricted blood–brain barrier (BBB) penetration, intratumoral heterogeneity, parallel pathway reactivation, and an immunosuppressive tumor microenvironment (TME). In this review, we synthesize current knowledge on MAPK pathway alterations in CNS tumors and evaluate the therapeutic landscape of MAPK inhibition, with emphasis on approved agents, emerging compounds, combination strategies, and novel drug-delivery technologies. We also discuss mechanisms that undermine treatment efficacy and highlight future directions aimed at integrating MAPK-targeted therapy into precision-based management of brain tumors. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

14 pages, 2313 KB  
Article
Ultrasound Imaging Properties of Heterologously Synthesized Gas Vesicles from Halophilic Archaeon
by Wenze Ou, Chenxing Liu, Yuanyuan Wang, Qiuxia Fu, Wei Liu, Huan Long and Fei Yan
Nanomaterials 2026, 16(1), 62; https://doi.org/10.3390/nano16010062 - 31 Dec 2025
Viewed by 368
Abstract
Biosynthetic gas vesicles (GVs), as novel nanoscale ultrasound contrast agents, exhibit unique potential in biomedical ultrasound imaging. For example, they are expected to have better tissue penetration through the tumor vasculature for detecting tumor cells by the design of GV-based acoustic probes. Of [...] Read more.
Biosynthetic gas vesicles (GVs), as novel nanoscale ultrasound contrast agents, exhibit unique potential in biomedical ultrasound imaging. For example, they are expected to have better tissue penetration through the tumor vasculature for detecting tumor cells by the design of GV-based acoustic probes. Of all these GVs, GVs from Halobacterium sp. NRC-1 possess the largest size (over 200 nm) and are nearly spherical in shape, endowing them with stronger acoustic signals and better tumor penetration. However, their genetic manipulation is relatively difficult due to the requirement of a high-salt cytoplasmic environment for their expression and assembly, limiting the application of biosynthetic technology for modulating their structural features in heterologous host cells. In this study, we cloned the gene cluster encoding GVs from Halobacterium sp. NRC-1 and transformed it into Haloferax volcanii, an archaeal species naturally incapable of producing GVs. The genetically engineered Haloferax volcanii successfully synthesized functional GVs (GVvol) with a similar size and shape to naturally synthesized GVs from Halobacterium sp. NRC-1 (GVhalo). The ultrasound imaging properties of GVvol heterologously synthesized in Haloferax volcanii were compared with naturally synthesized GVhalo in vitro and in vivo, showing that GVvol could achieve a mean signal intensity of 113.6 ± 0.9 a.u. in vitro and a peak intensity of 121.5 ± 0.8 a.u. in vivo in the kidney, compared with 115.7 ± 0.5 a.u. and 119.0 ± 0.5 a.u. for GVhalo, respectively. These findings confirm the functional integrity of heterologously synthesized GVvol and its potential for biomedical applications. Our study provides a solid experimental foundation for genetically tailoring Halobacterium GV properties to optimize biomedical imaging performance. Full article
(This article belongs to the Special Issue Advanced Nanomaterials for Bioimaging: 2nd Edition)
Show Figures

Figure 1

37 pages, 2896 KB  
Review
Targeting Cancer-Associated Fibroblasts in Prostate Cancer: Recent Advances and Therapeutic Opportunities
by Peng Chen, Junhao Chen, Peiqin Zhan, Xinni Ye, Li Zhao, Zhongsong Zhang, Jieming Zuo, Hongjin Shi, Xiangyun Li, Songhong Wu, Yuanzhi Fu, Haifeng Wang and Shi Fu
Cancers 2026, 18(1), 151; https://doi.org/10.3390/cancers18010151 - 31 Dec 2025
Viewed by 504
Abstract
Advanced prostate cancer, particularly castration-resistant disease, remains challenging to treat due to intratumoral heterogeneity, immune exclusion, and a suppressive tumor microenvironment. Within this ecosystem, cancer-associated fibroblasts shape tumor–stroma communication, but their marked heterogeneity and plasticity complicate classification and make indiscriminate fibroblast depletion potentially [...] Read more.
Advanced prostate cancer, particularly castration-resistant disease, remains challenging to treat due to intratumoral heterogeneity, immune exclusion, and a suppressive tumor microenvironment. Within this ecosystem, cancer-associated fibroblasts shape tumor–stroma communication, but their marked heterogeneity and plasticity complicate classification and make indiscriminate fibroblast depletion potentially ineffective or even harmful. This review summarizes recent progress in fibroblast origins, functional subtypes, and fibroblast-driven mechanisms that promote tumor progression and therapy resistance, as well as emerging therapeutic opportunities in prostate cancer. We conducted a structured literature search of PubMed, ScienceDirect, and major publisher platforms (including Nature and SpringerLink) from database inception to 15 February 2025, supplemented by targeted manual screening of reference lists. Evidence from single-cell/spatial-omics and mechanistic studies indicates that prostate tumors contain multiple fibroblast programs that occupy distinct niches yet can interconvert. Across these studies, it was found that these fibroblasts contribute to immune suppression, extracellular matrix remodeling and stromal barrier formation, angiogenesis, and metabolic support, collectively limiting drug penetration and reinforcing immune evasion; therapeutic pressure can further rewire fibroblast states and resistance-associated signaling. Overall, the literature supports a shift toward function- and subtype-directed intervention rather than “one-size-fits-all” targeting, with promising directions including precision targeting and reversible reprogramming, rational combination strategies, and localized delivery approaches that reduce stromal barriers while preserving tissue homeostasis in high-risk and treatment-refractory prostate cancer. Full article
Show Figures

Figure 1

27 pages, 4988 KB  
Review
Recent Advances in Functionalized Gold Nanoprobes for Photoacoustic Imaging Analysis of Diseases
by Zhiwan Huang, Hanying Ye, Haiting Cao, Yao Ma, Kecheng Lou, Yao He and Binbin Chu
Sensors 2026, 26(1), 203; https://doi.org/10.3390/s26010203 - 28 Dec 2025
Viewed by 610
Abstract
Photoacoustic imaging (PAI) integrates the high-contrast merits of optical imaging with the high-spatial-resolution advantages of acoustic imaging, enabling the acquisition of three-dimensional images with deep tissue penetration (up to several centimeters) for in vivo disease detection and diagnosis. Among various photoacoustic nanoagents, gold [...] Read more.
Photoacoustic imaging (PAI) integrates the high-contrast merits of optical imaging with the high-spatial-resolution advantages of acoustic imaging, enabling the acquisition of three-dimensional images with deep tissue penetration (up to several centimeters) for in vivo disease detection and diagnosis. Among various photoacoustic nanoagents, gold nanomaterials (GNMs) have been widely explored for the PAI-based imaging analysis and photothermal therapy of diseases, owing to their strong near-infrared (NIR) absorption, which can generate distinct photoacoustic signals in deep tissues. This review focuses on recent advances and achievements in the development of functionalized gold nanoprobes, including Janus gold nanoprobes, gold nanocomposite probes (such as functionally coated GNMs and GNMs-loaded nanocarriers), and gold nanoaggregate probes (e.g., pre-assembly of GNMs and in situ aggregation of GNMs). The multifunctionalization of GNMs can enhance their PAI performance by shifting absorption to the NIR-I and NIR-II regions, while simultaneously imparting additional functionalities such as targeted delivery to disease sites and specific responsiveness to disease biomarkers. These features can render functionalized GNMs-based nanoprobes highly suitable for PAI-based analysis and the precise detection of various pathological conditions, including bacterial infections, tumors, kidney injury, and disorders affecting the ocular, gastrointestinal, cardiovascular, visceral, and lymphatic systems. Finally, this review provides a concise summary of biosafety evaluation and outlines the current challenges and future perspectives in optimizing the GNMs-based PAI methods, highlighting their potential to enhance the rapid and precise diagnosis of diseases in the future. Full article
(This article belongs to the Special Issue Photoacoustic and Photothermal Sensing and Imaging)
Show Figures

Figure 1

Back to TopTop