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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,563)

Search Parameters:
Keywords = cytotoxic immune response

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 985 KiB  
Review
From Molecular Precision to Clinical Practice: A Comprehensive Review of Bispecific and Trispecific Antibodies in Hematologic Malignancies
by Behzad Amoozgar, Ayrton Bangolo, Maryam Habibi, Christina Cho and Andre Goy
Int. J. Mol. Sci. 2025, 26(11), 5319; https://doi.org/10.3390/ijms26115319 - 1 Jun 2025
Abstract
Multispecific antibodies have redefined the immunotherapeutic landscape in hematologic malignancies. Bispecific antibodies (BsAbs), which redirect cytotoxic T cells toward malignant targets via dual antigen engagement, are now established components of treatment for diseases such as acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma [...] Read more.
Multispecific antibodies have redefined the immunotherapeutic landscape in hematologic malignancies. Bispecific antibodies (BsAbs), which redirect cytotoxic T cells toward malignant targets via dual antigen engagement, are now established components of treatment for diseases such as acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and multiple myeloma (MM). Clinical trials of agents like blinatumomab, glofitamab, mosunetuzumab, and teclistamab have demonstrated deep and durable responses in heavily pretreated populations. Trispecific antibodies (TsAbs), although still investigational, represent the next generation of immune redirection therapies, incorporating additional tumor antigens or co-stimulatory domains (e.g., CD28, 4-1BB) to mitigate antigen escape and enhance T-cell persistence. This review provides a comprehensive evaluation of BsAbs and TsAbs across hematologic malignancies, detailing molecular designs, mechanisms of action, therapeutic indications, resistance pathways, and toxicity profiles including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), cytopenias, and infections. We further discuss strategies to mitigate adverse effects and resistance, such as antigen switching, checkpoint blockade combinations, CELMoDs, and construct optimization. Notably, emerging platforms such as tetrafunctional constructs, checkpoint-integrated multispecifics, and protease-cleavable masking designs are expanding the therapeutic index of these agents. Early clinical evidence also supports the feasibility of applying multispecific antibodies to solid tumors. Finally, we highlight the transformative role of artificial intelligence (AI) and machine learning (ML) in multispecific antibody development, including antigen discovery, biomarker-driven treatment selection, toxicity prediction, and therapeutic optimization. Together, BsAbs and TsAbs illustrate the convergence of molecular precision, clinical innovation, and AI-driven personalization, establishing a new paradigm for immune-based therapy across hematologic and potentially solid tumor malignancies. Full article
(This article belongs to the Special Issue Antibody Therapy for Hematologic Malignancies)
Show Figures

Figure 1

28 pages, 1534 KiB  
Review
T-Cell Engager Therapy in Prostate Cancer: Molecular Insights into a New Frontier in Immunotherapy
by Whi-An Kwon and Jae Young Joung
Cancers 2025, 17(11), 1820; https://doi.org/10.3390/cancers17111820 - 29 May 2025
Viewed by 201
Abstract
Advanced prostate cancer (PCa) remains lethal despite standard therapies, and immune checkpoint inhibitors offer limited benefit in its “immune-cold” microenvironment. T-cell engagers (TCEs)—bispecific antibodies linking CD3 on T-cells to tumor-associated antigens (TAAs)—provide potent, MHC-independent cytotoxicity, overcoming a key resistance mechanism. While early PSMA-targeted [...] Read more.
Advanced prostate cancer (PCa) remains lethal despite standard therapies, and immune checkpoint inhibitors offer limited benefit in its “immune-cold” microenvironment. T-cell engagers (TCEs)—bispecific antibodies linking CD3 on T-cells to tumor-associated antigens (TAAs)—provide potent, MHC-independent cytotoxicity, overcoming a key resistance mechanism. While early PSMA-targeted TCEs established proof-of-concept, recent data, notably for six transmembrane epithelial antigen of the prostate 1 (STEAP1)-targeting agents like Xaluritamig, demonstrate more substantial objective responses, highlighting progress through improved target selection and molecular design. This review synthesizes the evolving landscape of TCEs targeting PSMA, STEAP1, and DLL3 in PCa. We critically evaluate emerging clinical evidence, arguing that realizing the significant therapeutic potential of TCEs requires overcoming key challenges, including cytokine release syndrome (CRS), limited response durability, and antigen escape. We contend that future success hinges on sophisticated engineering strategies (e.g., affinity tuning, masking, multispecific constructs) and rationally designed combination therapies tailored to disease-specific hurdles. Strategies for toxicity mitigation, the crucial role of biomarker-driven patient selection, and potential integration with existing treatments are also discussed. Accumulating evidence supports TCEs becoming a new therapeutic pillar for advanced PCa, but achieving this demands sustained innovation focused on optimizing efficacy and safety. This review critically connects molecular engineering advancements with clinical realities and future imperatives. Full article
Show Figures

Figure 1

17 pages, 3651 KiB  
Article
Polarization of THP-1-Derived Human M0 to M1 Macrophages Exposed to Flavored E-Liquids
by Raivat Shah, Emily D. Luo, Carly A. Shaffer, Maya Tabakha, Sophie Tomov, Siara H. Minton, Mikaela K. Brown, Dominic L. Palazzolo and Giancarlo A. Cuadra
Toxics 2025, 13(6), 451; https://doi.org/10.3390/toxics13060451 - 29 May 2025
Viewed by 415
Abstract
Electronic cigarettes (ECIGs) are widely used but their effects on the immune system need to be further investigated. Macrophages are white blood cells central to the immune response. Using THP-1-derived M0 macrophages, this study aims to determine the effects of ECIG liquids (E-liquids) [...] Read more.
Electronic cigarettes (ECIGs) are widely used but their effects on the immune system need to be further investigated. Macrophages are white blood cells central to the immune response. Using THP-1-derived M0 macrophages, this study aims to determine the effects of ECIG liquids (E-liquids) on the polarization of M0 to the pro-inflammatory M1 macrophage subtype. THP-1 cells were cultured and differentiated to M0 macrophages using RPMI media. E-liquids ± cinnamon, menthol, strawberry and tobacco flavors were added to cell cultures at 1% (v/v) during polarization with lipopolysaccharides and interferon γ for 24 to 72 h. Morphology, viability, gene expression and cytokine production were measured using light microscopy, the LDH cytotoxicity assay, qPCR and ELISA, respectively. The results show that cells present little to no LDH activity under any treatments. In addition, cinnamon-flavored E-liquid severely affects morphology (i.e., abolishing pseudopodia formation), gene expression of all genes tested, and cytokine production. Other E-liquid flavors also affect some of these parameters, but to a lesser extent. Our data suggest that E-liquids can affect the polarization from M0 to M1, thus affecting the immune response in ECIG-exposed tissues such as the mucosa in the oral cavity and airways, ultimately mitigating the health status. Full article
(This article belongs to the Special Issue Health Risk Assessment of Exposure to Emerging Contaminants)
Show Figures

Graphical abstract

13 pages, 2024 KiB  
Article
Changes in NK Cells and Exhausted Th Cell Phenotype in RA Patients Treated with Janus Kinase Inhibitors: Implications for Adverse Effects
by Juan José Fernández-Cabero, Carmen Lasa-Teja, David San Segundo, Alejandra Comins-Boo, Juan Irure-Ventura, David Walias Rivera, Jose Luis Martín-Varillas, Cristina Mata, Montserrat Santos, Elena Aurrecoechea, Ricardo Blanco and Marcos López-Hoyos
Int. J. Mol. Sci. 2025, 26(11), 5160; https://doi.org/10.3390/ijms26115160 - 28 May 2025
Viewed by 48
Abstract
Recent concerns regarding the safety of Janus kinase inhibitors (JAKis) have prompted investigation into their impact on immune cell subsets in rheumatoid arthritis (RA) patients. This study aims to analyse alterations in immune cell populations induced by JAKis that may contribute to adverse [...] Read more.
Recent concerns regarding the safety of Janus kinase inhibitors (JAKis) have prompted investigation into their impact on immune cell subsets in rheumatoid arthritis (RA) patients. This study aims to analyse alterations in immune cell populations induced by JAKis that may contribute to adverse events, such as infections or malignancies. This study included 78 RA patients meeting ACR/EULAR criteria with an established treatment with JAKis (tofacitinib, baricitinib, upadacitinib, or filgotinib), 20 healthy donors, and 20 RA patients treated with biological disease-modifying antirheumatic drugs (bDMARDs). Peripheral blood mononuclear cells (PBMCs) were immunophenotyped directly after isolation using multiparametric flow cytometry to characterise innate and adaptive immune-cell subsets. JAKi-treated patients showed a significant reduction in cytotoxic NK Dim (CD3−CD56+CD16+) cells and in the percentage of NK Dim cells expressing the activation marker Nkp30. In CD4+ T cells, the percentage of Th17 (CD3+CD4+CD45RA+CCR6+CXCR3−), Th1-17 (CD3+CD4+CD45RA+CCR6+CXCR3+), and central memory (CM, CD3+CD4+CD45RA+CD62L+) cells was lower in the JAKi group, while effector memory (EM, CD3+CD4+CD45RA−CD62L−) and terminally differentiated CD45RA (TEMRA, CD3+CD4+CD45RA+CD62L−) T helper cells were increased compared to healthy and bDMARD-treated controls. The reduction in NK Dim and Th1-17 cells and the increase in exhausted Th subsets suggest a potential compromise in antiviral immunity and balanced immune responses in JAKi-treated RA patients. These alterations may contribute to an increased risk of infections or malignancies. Full article
(This article belongs to the Special Issue Mechanisms of Immune Tolerance and Autoimmune Diseases)
Show Figures

Figure 1

18 pages, 8713 KiB  
Article
Protective Potential and Functional Role of Antibodies Against SARS-CoV-2 Nucleocapsid Protein
by Alexandra Rak, Ekaterina Bazhenova, Polina Prokopenko, Victoria Matyushenko, Yana Orshanskaya, Konstantin V. Sivak, Arina Kostromitina, Larisa Rudenko and Irina Isakova-Sivak
Antibodies 2025, 14(2), 45; https://doi.org/10.3390/antib14020045 - 28 May 2025
Viewed by 38
Abstract
Cases of new COVID-19 infection, which manifested in 2019 and caused a global socioeconomic crisis, still continue to be registered worldwide. The high mutational activity of SARS-CoV-2 leads to the emergence of new antigenic variants of the virus, which significantly reduces the effectiveness [...] Read more.
Cases of new COVID-19 infection, which manifested in 2019 and caused a global socioeconomic crisis, still continue to be registered worldwide. The high mutational activity of SARS-CoV-2 leads to the emergence of new antigenic variants of the virus, which significantly reduces the effectiveness of COVID-19 vaccines, as well as the sensitivity of diagnostic test systems based on variable viral antigens. These problems may be solved by focusing on highly conserved coronavirus antigens, for example nucleocapsid (N) protein, which is actively expressed by coronavirus-infected cells and serves as a target for the production of virus-specific antibodies and T cell responses. It is known that anti-N antibodies are non-neutralizing, but their protective potential and functional activity are not sufficiently studied. Here, the protective effect of anti-N antibodies was studied in Syrian hamsters passively immunized with polyclonal sera raised to N(B.1) recombinant protein. The animals were infected with 105 or 104 TCID50 of SARS-CoV-2 (B.1, Wuhan or BA.2.86.1.1.18, Omicron) 6 h after serum passive transfer, and protection was assessed by weight loss, clinical manifestation of disease, viral titers in the respiratory tract, as well as by the histopathological evaluation of lung tissues. The functional activity of anti-N(B.1) antibodies was evaluated by complement-dependent cytotoxicity (CDC) and antibody-dependent cytotoxicity (ADCC) assays. The protection of anti-N antibodies was evident only against a lower dose of SARS-CoV-2 (B.1) challenge, whereas almost no protection was revealed against BA.2.86.1.1.18 variant. Anti-N(B.1) monoclonal antibodies were able to stimulate both CDC and ADCC. Thus, anti-N(B.1) antibodies possess protective activity against homologous challenge infection, which is possibly mediated by innate Fc-mediated immune reactions. These data may be informative for the development of N-based broadly protective COVID-19 vaccines. Full article
(This article belongs to the Section Humoral Immunity)
Show Figures

Figure 1

21 pages, 1612 KiB  
Review
CD300a: An Innate Immune Checkpoint Shaping Tumor Immunity and Therapeutic Opportunity
by Jei-Ming Peng and Hui-Ying Liu
Cancers 2025, 17(11), 1786; https://doi.org/10.3390/cancers17111786 - 27 May 2025
Viewed by 202
Abstract
CD300 family members are immunoglobulin superfamily receptors that regulate immune cell function through either activating or inhibitory signals. Among them, CD300a is a prototypical inhibitory receptor, highly expressed in both myeloid and lymphoid lineages, and plays a pivotal role in the pathogenesis of [...] Read more.
CD300 family members are immunoglobulin superfamily receptors that regulate immune cell function through either activating or inhibitory signals. Among them, CD300a is a prototypical inhibitory receptor, highly expressed in both myeloid and lymphoid lineages, and plays a pivotal role in the pathogenesis of inflammation and tumor immunity. CD300a transduces inhibitory signals in several immune cells—including mast cells, eosinophils, monocytes, dendritic cells (DCs), neutrophils, and natural killer (NK) cells—by recruiting SHP-1 phosphatase to immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and suppressing activation pathways such as Toll-like receptor (TLR)-MyD88 and FcεRI signaling. Recent studies suggest that tumor cells may hijack CD300a-associated pathways to establish an immunosuppressive microenvironment that facilitates immune evasion, tumor survival, and potentially metastatic spread. Proposed mechanisms include reduced DC-mediated type I interferon (IFN) production, diminished NK cell cytotoxicity, and negative regulation of mast cell– and eosinophil-dependent anti-tumor responses. Although some of these findings are derived from in vivo models, the cumulative evidence positions CD300a as a critical immune checkpoint in tumor-associated immune regulation. In addition to its established roles in hematologic malignancies—including chronic lymphocytic leukemia, acute lymphoblastic leukemia, and acute myeloid leukemia—CD300a has also been implicated in modulating tumor-associated immune responses in other pathological contexts. While most studies emphasize its immune cell–mediated effects, emerging evidence suggests that CD300a may directly influence tumor progression by regulating immune homeostasis, intracellular signaling, and tumor microenvironment interactions. Collectively, these findings establish CD300a as a pleiotropic immunoregulatory molecule in both hematologic and non-hematologic malignancies, underscoring the need to further explore its broader relevance and therapeutic potential in cancer immunology. Full article
Show Figures

Figure 1

17 pages, 2221 KiB  
Article
Nanoparticle-Based mRNA Vaccine Induces Protective Neutralizing Antibodies Against Infectious Bronchitis Virus in In-Vivo Infection
by Aseno Sakhrie, Ankarao Kalluri, Zeinab H. Helal, Challa V. Kumar and Mazhar I. Khan
Vaccines 2025, 13(6), 568; https://doi.org/10.3390/vaccines13060568 - 26 May 2025
Viewed by 283
Abstract
Background: Live attenuated and inactivated virus vaccines are commonly used against infectious bronchitis virus (IBV) in chickens, but they have limitations such as mutation risks and short efficacy. This study explores cationic bovine serum albumin (BSA) polyamine nanoparticles (NPs) for delivering IBV spike [...] Read more.
Background: Live attenuated and inactivated virus vaccines are commonly used against infectious bronchitis virus (IBV) in chickens, but they have limitations such as mutation risks and short efficacy. This study explores cationic bovine serum albumin (BSA) polyamine nanoparticles (NPs) for delivering IBV spike protein mRNA, aiming to develop a safer and more effective vaccine. Methods: A BSA-based nanoparticle system was designed with positive surface charges and characterized using dynamic light scattering (DLS), Zetasizer, and transmission electron microscopy (TEM). Its cytotoxicity, cellular uptake, and ability to deliver IBV spike protein mRNA were evaluated in macrophage-like chicken cell lines (HD11), followed by immunogenicity studies in SPF chickens to assess immune responses. Results: The study demonstrated successful binding and transfection efficiency of the in vitro transcription (IVT)-mRNA complexed with the NPs, which was enhanced with chloroquine. Immunogenicity studies in SPF chickens showed a significant increase in antibody titers in chickens vaccinated with the mRNA vaccine compared to the PBS control, indicating an effective immune response against the IBV S protein. Furthermore, the neutralization index doubled after a higher-dose mRNA booster with chloroquine, and PBMCs from immunized chickens exhibited a threefold higher stimulation index than the PBS control. Conclusions: BSA-based NPs effectively deliver IBV spike protein mRNA, enhancing immune responses and offering a promising strategy for a safer, more effective IBV vaccine. Full article
Show Figures

Graphical abstract

24 pages, 9856 KiB  
Article
Roseburia intestinalis Modulates Immune Responses by Inducing M1 Macrophage Polarization
by Anna Bircher, Egle Katkeviciute, Yasser Morsy, Silvia Lang, Ana Montalban-Arques and Michael Scharl
Int. J. Mol. Sci. 2025, 26(11), 5049; https://doi.org/10.3390/ijms26115049 - 23 May 2025
Viewed by 255
Abstract
In recent years, the gut microbiome has been recognized as one influential factor in cancer development. Particularly in colorectal cancer (CRC), several studies observed a major imbalance of the intestinal microbiota, marked by a reduction in beneficial bacterial species, such as Roseburia intestinalis [...] Read more.
In recent years, the gut microbiome has been recognized as one influential factor in cancer development. Particularly in colorectal cancer (CRC), several studies observed a major imbalance of the intestinal microbiota, marked by a reduction in beneficial bacterial species, such as Roseburia intestinalis, and an increase in opportunistic pathobionts, like Peptostreptococcus stomatis. We previously observed that specific Eubacteriales, including R. intestinalis, were significantly reduced in CRC patients and have a potent anti-tumor immune effect when applied as oral monotherapy in mice. Here, we investigate the molecular mechanism of R. intestinalis on various cell types in vitro, highlighting its potential therapeutic value in CRC. Co-culture experiments with macrophages demonstrated that R. intestinalis exposure induced an increase in the M1 phenotype and decreased the M2 phenotype, suggesting macrophage-polarizing properties of these bacteria. R. intestinalis also triggered a gene expression profile resembling M1 macrophages and led to distinct chemokine and cytokine secretion in cancer cells, suggesting an immune-activating environment. However, we did not observe direct cytotoxic effects in cancer cells. Our research provides insights into the potential of R. intestinalis to activate immune responses, supporting further investigation into its therapeutic role in CRC. These findings underscore the need for deeper studies on the bacterium’s impact on CRC pathogenesis and treatment. Full article
(This article belongs to the Special Issue Molecular Advances in Gut Microbiota and Intestinal Diseases)
Show Figures

Figure 1

25 pages, 2873 KiB  
Article
Ergolide Regulates Microglial Activation and Inflammatory-Mediated Dysfunction: A Role for the Cysteinyl Leukotriene Pathway
by Danielle M. Galvin, Sara Fernandez-Garcia, Emma Dawson, Ciara Pryce, Billy P. Egan, Niamh C. Clarke, Alison L. Reynolds and Derek A. Costello
Int. J. Mol. Sci. 2025, 26(11), 5050; https://doi.org/10.3390/ijms26115050 - 23 May 2025
Viewed by 303
Abstract
Neurodegenerative diseases are characterised by the progressive loss of neurons, leading to a decline in specific brain functions. Alzheimer’s disease (AD) and Parkinson’s disease (PD) are the most prevalent, affecting approximately 60 million people worldwide. The pathogenesis of these diseases is complex, combining [...] Read more.
Neurodegenerative diseases are characterised by the progressive loss of neurons, leading to a decline in specific brain functions. Alzheimer’s disease (AD) and Parkinson’s disease (PD) are the most prevalent, affecting approximately 60 million people worldwide. The pathogenesis of these diseases is complex, combining inflammatory, oxidative, and excitotoxic processes that result in neuronal dysfunction and death. Despite recent advances, there is currently no cure for AD and PD. Available therapies demonstrate limited efficacy, highlighting the continuing need for novel therapeutic approaches. Ergolide, a naturally occurring sesquiterpene lactone from the Inula brittanica plant, has shown immunoregulatory properties in systemic immune cells and potential applications in certain cancers. This study examines whether the therapeutic effects of ergolide extend to the brain. We explored its mechanisms of action in vitro, and its capacity to restore behavioural integrity in zebrafish models of inflammation and neurotoxicity in vivo. We report the ability of ergolide to attenuate inflammatory cytokine and nitric oxide (NO) production from microglia in response to toll-like receptor activation. We further propose a role for the NFκB and cysteinyl leukotriene (CysLT) pathways in ergolide-mediated regulation of microglial activation. Ergolide did not protect against oxidative-induced neuronal death in vitro or mitigate seizure activity in zebrafish. Instead, we revealed a pro-oxidant and cytotoxic effect on neuroblastoma cells. Importantly, ergolide improved survival and alleviated the dysfunction in sensorimotor behaviour in a zebrafish model of inflammation. Our findings reveal a neuroprotective effect of ergolide, likely stemming from its immunoregulatory capacity. We also support further investigation of the CysLT pathway as a therapeutic target for neuroinflammatory-related disease. Full article
(This article belongs to the Collection Feature Papers in Molecular Neurobiology)
Show Figures

Figure 1

35 pages, 5451 KiB  
Review
Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer
by Juliane Blümke, Moritz Schameitat, Atul Verma, Celina Limbecker, Elise Arlt, Sonja M. Kessler, Heike Kielstein, Sebastian Krug, Ivonne Bazwinsky-Wutschke and Monika Haemmerle
Cancers 2025, 17(10), 1689; https://doi.org/10.3390/cancers17101689 - 17 May 2025
Viewed by 480
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar–ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes. Full article
(This article belongs to the Special Issue Management of Pancreatic Cancer)
Show Figures

Figure 1

23 pages, 3484 KiB  
Article
In Silico Design, Optimization, and Evaluation of a Multi-Epitope Vaccine Targeting the Clostridium perfringens Collagen Adhesin Protein
by Dhiraj Chundru, Shailes Bhattrai, Madhusudan Timilsina, Hyun Lillehoj, Zhifeng Sun, Mostafa Ghanem and Charles Li
Microorganisms 2025, 13(5), 1147; https://doi.org/10.3390/microorganisms13051147 - 16 May 2025
Viewed by 257
Abstract
Necrotic enteritis (NE), caused by pathogenic Clostridium perfringens, poses a significant threat to global poultry health, with estimated annual losses exceeding USD 6 billion. The rising incidence of NE has been associated with the reduced use of antibiotic growth promoters, underscoring the [...] Read more.
Necrotic enteritis (NE), caused by pathogenic Clostridium perfringens, poses a significant threat to global poultry health, with estimated annual losses exceeding USD 6 billion. The rising incidence of NE has been associated with the reduced use of antibiotic growth promoters, underscoring the urgent need for alternative control measures such as vaccination. Collagen adhesin protein (CNA), a key virulence factor in NE pathogenesis, represents a promising vaccine target. The US Food and Drug Administration has begun phasing out animal testing requirements for biologics and monoclonal antibody drugs. In this study, a computational multi-epitope vaccine (MEV) targeting CNA was designed by integrating predicted Cluster of Differentiation (CD)4+ helper T lymphocyte (Th), CD8+ cytotoxic T lymphocyte (CTL), and B-cell epitopes. Bioinformatics tools were used to identify immunogenic, antigenic, and non-allergenic epitopes assembled into a 115-amino-acid peptide vaccine construct. The candidate demonstrated strong stability and solubility. In silico immune simulation predicted robust immune responses, including elevated IgG and IgM antibody levels, plasma cell proliferation, Th memory formation, and CTL activation, comparable to responses elicited by a full-length CNA. These findings support the potential of the designed peptide as one of the multiple effective NE vaccine components, offering a promising alternative to antibiotic-based approaches in poultry disease management. Full article
(This article belongs to the Special Issue Poultry Pathogens and Poultry Diseases, 2nd Edition)
Show Figures

Figure 1

20 pages, 2617 KiB  
Article
Evaluation of the PP6D5 Polymer as a Novel Non-Viral Vector in the Development of a CRISPR/nCas9-Based Gene Therapy for Tay–Sachs Disease
by Jacky M. Guerrero-Vargas, Diego A. Suarez-Garcia, Andrés F. Leal, Ivonne L. Diaz-Ariza, León D. Pérez-Pérez, Angela J. Espejo-Mojica and Carlos J. Alméciga-Díaz
Pharmaceutics 2025, 17(5), 628; https://doi.org/10.3390/pharmaceutics17050628 - 9 May 2025
Viewed by 439
Abstract
Background/Objectives: Tay–Sachs disease (TSD) is a neurodegenerative disorder caused by a deficiency in β-hexosaminidase A (HexA), which accumulates GM2 gangliosides, primarily in neurons. Currently, therapeutic options are limited, highlighting the need for new strategies such as gene therapy. Despite their effectiveness, viral vectors [...] Read more.
Background/Objectives: Tay–Sachs disease (TSD) is a neurodegenerative disorder caused by a deficiency in β-hexosaminidase A (HexA), which accumulates GM2 gangliosides, primarily in neurons. Currently, therapeutic options are limited, highlighting the need for new strategies such as gene therapy. Despite their effectiveness, viral vectors can elicit adverse immune responses; consequently, non-viral vectors are being explored as an alternative. We have previously investigated the use of CRISPR/Cas9 nickase (nCas9) as a potential tool for treating TSD. Here, we expanded our study by evaluating the PP6D5 polymer as a novel non-viral vector for delivering the CRISPR/nCas9 system to restore HexA activity. Methods: First, we evaluated the PP6D5-mediated CRISPR/nCas9 system’s transfection efficiency in NIH-3T3 fibroblasts, U87MG astrocytoma, SHSY5Y neuroblastoma, and TSD fibroblasts. We then evaluated the potential of PP6D5 to correct the gene defect in TSD fibroblasts. Results: The results showed that PP6D5 exhibited significantly higher transfection efficiency compared to lipofectamine 3000 in all tested cell models. In TSD fibroblasts, transfection with both HEXA and HEXB cDNAs increased the HexA activity levels by up to 7.4-fold, compared to a 3.2-fold increase in cells transfected only with HEXA cDNA after 15 days post-transfection. These levels were up to 4.5-fold higher than those observed in lipofectamine-mediated transfection. Additionally, PP6D5-mediated CRISPR/nCas9-based genome editing led to a significant reduction in the lysosomal mass of TSD fibroblasts. Conclusions: This study provides promising evidence for the use of the PP6D5 polymer as a non-viral vector for delivering CRISPR/nCas9-based gene therapy in TSD. The use of the PP6D5 polymer may offer some advantages that viral vectors cannot, such as a reduction in cytotoxicity and higher TE in difficult-to-transfect cell lines. Furthermore, this type of polymeric vector has not been extensively explored for gene therapy, making this study an important contribution to the development of non-viral delivery systems for the treatment of neurodegenerative diseases. Full article
Show Figures

Graphical abstract

16 pages, 4550 KiB  
Article
PI3K/mTOR Signaling Pathway Dual Inhibition for the Management of Neuroinflammation: Novel Insights from In Vitro Models
by Alessio Ardizzone, Sarah Adriana Scuderi, Giovanna Casili, Rossella Basilotta, Emanuela Esposito and Marika Lanza
Biomolecules 2025, 15(5), 677; https://doi.org/10.3390/biom15050677 - 7 May 2025
Viewed by 239
Abstract
Neuroinflammatory responses are central to the pathogenesis of neurodegenerative diseases, affecting cells of both neuronal and glial origin that respond to immune-driven inflammatory stimuli. The PI3K/mTOR signaling pathway is essential for the regulation of these neuroinflammatory processes and is therefore a promising target [...] Read more.
Neuroinflammatory responses are central to the pathogenesis of neurodegenerative diseases, affecting cells of both neuronal and glial origin that respond to immune-driven inflammatory stimuli. The PI3K/mTOR signaling pathway is essential for the regulation of these neuroinflammatory processes and is therefore a promising target for therapeutic intervention. Here, we investigated the consequences of PI3K/mTOR pathway inhibition on neuroinflammation employing PF-04691502, an agent with combined PI3K and mTOR inhibitory activity. We treated SH-SY5Y, C6, BV-2, and Mo3.13 cell lines with PF-04691502 at concentrations of 0.1, 0.5, and 1 µM to assess the modulation of neuroinflammatory responses. To induce inflammation, cells were stimulated with lipopolysaccharide (LPS, 1 μg/mL) and interferon-gamma (IFN-γ, 100 U/mL). The results from the MTT assays demonstrated that PI3K/mTOR inhibition preserved cell viability at 0.5 and 1 µM across all of the cell lines, indicating its potential to mitigate inflammation-driven cytotoxicity. Subsequent ELISA assays revealed a marked decrease in the NF-κB and pro-inflammatory cytokine levels, confirming the effective suppression of inflammation through PI3K/mTOR inhibition. In addition, the SH-SY5Y cell line was exposed to MPP+ to simulate Parkinson’s disease (PD)-like toxicity; then, cell viability, PD-associated markers, and apoptotic indicators were assessed. Our results indicate that inhibition of the PI3K/mTOR signaling axis may alleviate neurodegenerative processes by modulating both neuroinflammatory responses and apoptotic pathways. These findings highlight the therapeutic promise of targeting PI3K/mTOR in the context of neurodegenerative disorders and support the need for further validation through in vivo and clinical investigations. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

18 pages, 2822 KiB  
Article
Iron Oxide Magnetic Nanoparticles Synthesized by Laser Target Evaporation Method for the Needs of Cancer Immunotherapy
by Felix Blyakhman, Fedor Fadeyev, Alexander Safronov, Tatiana Terziyan, Ekaterina Burban, Tatyana Shklyar and Galina Kurlyandskaya
Materials 2025, 18(9), 2142; https://doi.org/10.3390/ma18092142 - 6 May 2025
Viewed by 346
Abstract
Administration of monocyte-derived dendritic cells (moDCs) sensitized by cancer-associated antigens to the patient is applied to boost the T-cell mediated anti-tumor immune response. Loading moDCs with magnetic nanoparticles (MNPs) and controlling their migration to lymph nodes by an external magnetic field is a [...] Read more.
Administration of monocyte-derived dendritic cells (moDCs) sensitized by cancer-associated antigens to the patient is applied to boost the T-cell mediated anti-tumor immune response. Loading moDCs with magnetic nanoparticles (MNPs) and controlling their migration to lymph nodes by an external magnetic field is a way to improve the effectiveness of immunotherapy. In this study, spherical MNPs of maghemite iron oxide with a diameter of about 14 nm were synthesized by laser target evaporation method (LTE) and examined in the context of their prospective use for the needs of moDCs immunotherapy. Characterization of the physicochemical properties of MNPs and their stabilization in physiological media, as well as the magnetic properties of MNPs in the suspensions were considered in detail. The cytotoxic effect of MNPs in growth medium on the human moDCs and MNPs uptake by the cells were also estimated. We show that up-taken MNPs and MNPs in growth medium demonstrated cytotoxic effect only at high concentrations. At the same time, at low concentrations MNPs up-taken by moDCs increased their viability causing the stimulation effect. The evaluation of the quantity of MNPs, up-taken by cells, is possible by magnetometry even for the smallest γ-Fe2O3 concentrations. Full article
(This article belongs to the Special Issue Magnetic Nanomaterials: Synthesis, Characterization and Applications)
Show Figures

Figure 1

24 pages, 818 KiB  
Review
Deficient Mismatch Repair and Microsatellite Instability in Solid Tumors
by Joy A. Awosika, James L. Gulley and Danielle M. Pastor
Int. J. Mol. Sci. 2025, 26(9), 4394; https://doi.org/10.3390/ijms26094394 - 6 May 2025
Viewed by 327
Abstract
The integrity of the genome is maintained by mismatch repair (MMR) proteins that recognize and repair base mismatches and insertion/deletion errors generated during DNA replication and recombination. A defective MMR system results in genome-wide instability and the progressive accumulation of mutations. Tumors exhibiting [...] Read more.
The integrity of the genome is maintained by mismatch repair (MMR) proteins that recognize and repair base mismatches and insertion/deletion errors generated during DNA replication and recombination. A defective MMR system results in genome-wide instability and the progressive accumulation of mutations. Tumors exhibiting deficient MMR (dMMR) and/or high levels of microsatellite instability (termed “microsatellite instability high”, or MSI-H) have been shown to possess fundamental differences in clinical, pathological, and molecular characteristics, distinguishing them from their “microsatellite stable” (MSS) counterparts. Molecularly, they are defined by a high mutational burden, genetic instability, and a distinctive immune profile. Their distinct genetic and immunological profiles have made dMMR/MSI-H tumors particularly amenable to treatment with immune checkpoint inhibitors (ICIs). The ongoing development of biomarker-driven therapies and the evaluation of novel combinations of immune-based therapies, with or without the use of conventional cytotoxic treatment regimens, continue to refine treatment strategies with the goals of maximizing therapeutic efficacy and survival outcomes in this distinct patient population. Moreover, the resultant knowledge of the mechanisms by which these features are suspected to render these tumors more responsive, overall, to immunotherapy may provide information regarding the potential optimization of this therapeutic approach in tumors with proficient MMR (pMMR)/MSS tumors. Full article
Show Figures

Figure 1

Back to TopTop