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
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
remove_circle_outline

Search Results (2,902)

Search Parameters:
Keywords = antigen-presenting cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 2459 KB  
Review
The Immunoregulatory Mechanisms of Human Cytomegalovirus from Primary Infection to Reactivation
by Xiaodan Liu, Chang Liu and Ting Zhang
Pathogens 2025, 14(10), 998; https://doi.org/10.3390/pathogens14100998 (registering DOI) - 2 Oct 2025
Abstract
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses [...] Read more.
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses major histocompatibility complex (MHC)-dependent antigen presentation to evade T cell recognition. As the downregulation of MHC molecules may trigger NK cell activation, the virus compensates for this by expressing proteins such as UL40 and IL-10, which engage inhibitory NK cell receptors and block activating signals, thereby suppressing NK cell immune surveillance. Viral proteins like UL36 and UL37 block host cell apoptosis and necroptosis, allowing HCMV to persist undetected and avoid clearance. In settings of profound immunosuppression, such as after allogeneic hematopoietic stem cell transplantation (allo-HSCT) or solid organ transplantation, slow immune reconstitution creates a window for viral reactivation. Likewise, immunosenescence and chronic low-grade inflammation during aging increases the risk of reactivation. Once reactivated, HCMV triggers programmed cell death, releasing viral PAMPs (pathogen-associated molecular patterns) and host-derived DAMPs (damage-associated molecular patterns). This release fuels a potent inflammatory response, promoting further viral reactivation and exacerbating tissue damage, creating a vicious cycle. This cycle of inflammation and reactivation contributes to both transplant-related complications and the decline of antiviral immunity in the elderly. Therefore, understanding the immune regulatory mechanisms that govern the switch from latency to reactivation is critical, especially within the unique immune landscapes of transplantation and aging. Elucidating these pathways is essential for developing strategies to prevent and treat HCMV-related disease in these high-risk populations. Full article
(This article belongs to the Special Issue Pathogen–Host Interactions: Death, Defense, and Disease)
Show Figures

Figure 1

25 pages, 3573 KB  
Article
Chronic Folliculitis Associated with Ovine gammaherpesvirus 2-Induced Infections in Dairy Cows from Southern Brazil
by Selwyn Arlington Headley, Flávia Helena Pereira Silva, Mariana da Silva Marques, Juliana Torres Tomazi Fritzen, Fernanda Pinto Ferreira, Geovana Depieri Yoshitani, Ana Aparecida Correa Xavier, Pedro Paulo Benyunes Vieira and Amauri Alcindo Alfieri
Animals 2025, 15(19), 2883; https://doi.org/10.3390/ani15192883 - 1 Oct 2025
Abstract
Ovine gammaherpesvirus 2 (OvGHV2) is a Macavirus and the cause of sheep-associated malignant catarrhal fever (SA-MCF) in susceptible mammalian hosts worldwide. OvGHV2 may produce typical clinical manifestations of SA-MCF or subclinical infections. Additionally, OvGHV2 is associated with cutaneous lesions in ruminants, with few [...] Read more.
Ovine gammaherpesvirus 2 (OvGHV2) is a Macavirus and the cause of sheep-associated malignant catarrhal fever (SA-MCF) in susceptible mammalian hosts worldwide. OvGHV2 may produce typical clinical manifestations of SA-MCF or subclinical infections. Additionally, OvGHV2 is associated with cutaneous lesions in ruminants, with few documented reports of this unusual manifestation worldwide. This paper presents the pathological, immunohistochemical (IHC), and molecular findings observed in outbreaks of OvGHV2-related skin infections in dairy cattle from Southern Brazil. Cutaneous scrapings (n = 35) and biopsies (n = 6) were obtained from dairy cows derived from three farms. All cows (n = 35) developed widespread, ulcerative to scaly and erythematous skin lesions, and had no contact with sheep or goats. The biopsies were evaluated for histopathological diagnosis and then used in IHC analyses designed to detect malignant catarrhal fever virus (MCFV) antigens and to evaluate the inflammatory response. All scrapings and biopsies were used in PCR assays to amplify OvGHV2. Additionally, all biopsies were used in PCR assays to detect bovine gammaherpesvirus 6 (BoGHV6), bovine alphaherpesvirus 1 (BoAHV1), and poxvirus. Histopathology revealed chronic folliculitis in all biopsies. IHC detected intralesional, intracytoplasmic MCFV antigens in most (83.3%; 5/6) of the cutaneous lesions with folliculitis. These skin lesions showed a strong T-cell response, macrophage clusters, and caspase-positive follicular keratinocytes. OvGHV2 DNA was detected in 66.7% (4/6) of the cutaneous biopsies that contained MCFV antigens and in 8.6% (3/35) of the cutaneous scrapings. The DNA of BoGHV6, BoAHV1, and Poxvirus was not amplified from any of the cutaneous biopsies. These findings demonstrated that OvGHV2 was associated with the cutaneous lesions in dairy cows at these farms and represent the first description of OvGHV2-related skin disease in ruminants from Brazil and the entire Latin America. A review of previous cases of skin lesions associated with infections by OvGHV2 revealed that most cases had a histological diagnosis of folliculitis, suggesting that folliculitis may be associated with OvGHV2-related skin infections. Additionally, this investigation contrasts all previous reports of OvGHV2-related skin disease in ruminants, since the infected cows herein identified were not reared concomitantly or within proximity of the asymptomatic reservoir host. Furthermore, the possible form of OvGHV2 dissemination to the susceptible cows during this study is discussed. Full article
(This article belongs to the Section Cattle)
24 pages, 19724 KB  
Article
Endothelial Cell Transition: Preliminary Data on Cross-Organ Shift from Brain to Liver
by Alexey Larionov, Luis Filgueira and Christian M. Hammer
Cells 2025, 14(19), 1538; https://doi.org/10.3390/cells14191538 - 1 Oct 2025
Abstract
Background: Endothelial cells (EC), crucial components of the vascular system, are adaptable cells that maintain homeostasis and respond to pathological events through structural and functional plasticity. Hepatocyte growth factor (HGF) is a multifunctional cytokine that has been demonstrated to have protective and [...] Read more.
Background: Endothelial cells (EC), crucial components of the vascular system, are adaptable cells that maintain homeostasis and respond to pathological events through structural and functional plasticity. Hepatocyte growth factor (HGF) is a multifunctional cytokine that has been demonstrated to have protective and disruptive influence on the blood barrier function. In endothelial biology, its role is also poorly characterized. The present study explores the impact of supraphysiological concentrations of HGF on mouse brain endothelial cells (MBECs), scrutinizing how it alters their integrity and morphology. Methods: Two groups of MBECs—control (CTR) and experimental (EXP)—were analyzed at two time points: early passage (p5) and late passage (p41). The EXP-groups (p5 and p41) were treated with HGF at a concentration of 4 µL/mL. Cellular morphology was assessed with brightfield microscopy; protein expression and localization of the tight junction marker (ZO-1) and the endothelial marker (Factor VII related antigen/von Willebrand factor, vWf) were analyzed using Western blotting, immunocytochemistry, and confocal microscopy. Intercellular barrier function was estimated via Transendothelial Electric Resistance (TEER) and Transendothelial Dextran Permeability (TEDP) assays. Results: Microscopical analysis demonstrated a change in the morphology of the MBECs from a longitudinal, spindle-like shape to a rounded, more spheroid, cobblestone-like morphology under high-dose HGF treatment. Western blotting revealed a progressive decrease of ZO-1 expression in the EXP-groups. The expression of vWf did not show significant differences. Qualitative immunocytochemical staining: vWf showed consistent expression across all groups. ZO-1 displayed a punctate, well-defined membrane and cytoplasmic localization pattern in the CTR-groups at p5 and p41. In contrast, the p5 EXP-group demonstrated a shift to a more diffuse cytoplasmic pattern. At p41, the EXP-group displayed a markedly reduced ZO-1 signal with no clear-cut membrane localization. Confocal analysis: ZO-1: punctate membrane-associated localization in CTR-groups at p5 and 41. The EXP-groups at p5 and p41 confirmed the diffuse cytoplasmic ZO-1 distribution. Phalloidin: well-organized actin cytoskeleton in CTR-groups, but rearrangement and stress fiber disorganization in the EXP-groups, especially at p41. The merged images confirmed reduced co-localization of ZO-1 with actin structures. Barrier function: TEER values dropped significantly in HGF-treated cells. TEDP to small and medium molecular weight dextran increased markedly under HGF treatment. Conclusions: Our data demonstrate that supraphysiological doses of HGF in an in vitro MBEC-barrier-like model disrupt TJ organization, leading to morphological changes and functional weakening of the MBEC-barrier-like structure, as shown by uncoupling between ZO-1/F-actin cytoskeleton, reduced TEER, and increased size-selective paracellular permeability (TEDP). Full article
Show Figures

Figure 1

15 pages, 434 KB  
Review
Dendritic Cell Immunotherapy for Solid Tumors: Advances in Translational Research and Clinical Application
by Mi Eun Kim and Jun Sik Lee
Curr. Issues Mol. Biol. 2025, 47(10), 806; https://doi.org/10.3390/cimb47100806 - 1 Oct 2025
Abstract
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based [...] Read more.
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based vaccines for solid tumors. Clinical studies across melanoma, glioblastoma, prostate cancer, and non-small cell lung cancer have demonstrated safety and immunogenicity, with encouraging signals of clinical efficacy, particularly when DC vaccination is combined with immune checkpoint blockade or personalized neoantigen approaches. However, translational barriers remain, including the immunosuppressive tumor microenvironment, inefficient DC migration, and variability in manufacturing protocols. Developing solutions such as in vivo DC targeting, biomaterials-based delivery systems, high-resolution single-cell analyses, and artificial intelligence-driven epitope prediction are controlled to overcome these challenges. Together, these innovations highlight the evolving role of DC immunotherapy as a foundation of precision oncology, offering the potential to integrate personalized vaccination strategies into standard treatment paradigms for solid tumors. Therefore, in this review, we specifically focus on these advances in dendritic cell immunotherapy for solid tumors and their translational implications. Full article
(This article belongs to the Special Issue Future Challenges of Targeted Therapy of Cancers: 2nd Edition)
Show Figures

Figure 1

23 pages, 2559 KB  
Review
Mechanisms of Interleukin-10-Mediated Immunosuppression in Viral Infections
by Zijing Guo, Qifu He, Yan Zhang, Yuling Li and Zhidong Zhang
Pathogens 2025, 14(10), 989; https://doi.org/10.3390/pathogens14100989 - 1 Oct 2025
Abstract
Interleukin-10 (IL-10), a potent anti-inflammatory cytokine, plays a vital role in regulating immune responses across various infectious and inflammatory conditions. While IL-10 is essential for preventing excessive tissue damage and maintaining immune homeostasis (e.g., respiratory syncytial virus), its elevated levels could result in [...] Read more.
Interleukin-10 (IL-10), a potent anti-inflammatory cytokine, plays a vital role in regulating immune responses across various infectious and inflammatory conditions. While IL-10 is essential for preventing excessive tissue damage and maintaining immune homeostasis (e.g., respiratory syncytial virus), its elevated levels could result in immunosuppression during viral infections, enabling viruses to evade host defenses (e.g., foot-and-mouth disease virus). This review aims to elucidate the mechanisms through which IL-10 mediates immunosuppression in viral infections and to explore the implications of these mechanisms for therapeutic intervention. The key scientific concepts outlined in this review include the mechanisms of IL-10 production and its varied impacts on the immune response during viral infections. Specifically, we discuss the multifaceted inhibitory effects of IL-10 on innate and adaptive immunity, including its implications for antigen presentation, T cells activation, pro-inflammatory cytokine production, immune cell differentiation, trafficking, apoptosis, and co-inhibitory expression related to T cells exhaustion. Finally, we discuss the therapeutic potential of targeting IL-10, such as monoclonal antibodies and small molecule inhibitors, and their potential to restore effective immune responses. By summarizing current knowledge on IL-10’s role in viral infections, this review offers a thorough insight into its immunosuppressive mechanisms and their therapeutic potential, paving the way for innovative treatment strategies in viral diseases. Full article
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
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

26 pages, 6802 KB  
Article
Multifunctional Polymer-Modified P-CaO2@Au@OVA@Cu@DHPs Nanoparticles Enhance SARS-CoV-2 mRNA Vaccine-Induced Immunity via the cGAS–STING Signaling Pathway
by Yanle Zhi, Shengchao Wang, Haibo Zhang, Guimin Xue and Zhiqiang Zhang
Polymers 2025, 17(19), 2636; https://doi.org/10.3390/polym17192636 - 30 Sep 2025
Abstract
The success of mRNA-based SARS-CoV-2 vaccines has been confirmed in both preclinical and clinical settings. However, the development of safe and efficient mRNA vaccine delivery platforms remains challenging. In this report, PBAE-G-B-SS-modified CaO2 nanofibers and Au@OVA@Cu@Dendrobium huoshanense polysaccharides were employed to establish [...] Read more.
The success of mRNA-based SARS-CoV-2 vaccines has been confirmed in both preclinical and clinical settings. However, the development of safe and efficient mRNA vaccine delivery platforms remains challenging. In this report, PBAE-G-B-SS-modified CaO2 nanofibers and Au@OVA@Cu@Dendrobium huoshanense polysaccharides were employed to establish novel self-assembling polymeric micelles (CaO2@Au@OVA@Cu@DHPs) capable of serving as both an adjuvant and a delivery system for mRNA vaccines. In vitro, CaO2@Au@OVA@Cu@DHPs nanoparticles (NPs) were conducive to effective macrophage antigen uptake and efficient antigen processing. In vivo, P-CaO2@Au@OVA@Cu@DHPs NP administration was associated with a reduction in the ovalbumin (OVA) release rate that was conducive to the sustained induction of long-term immunity and to the production of higher levels of different IgG subtypes, suggesting that these effects were attributable to enhanced antigen uptake by antigen-presenting cells. Overall, these present data highlight the promise of these P-CaO2@Au@OVA@Cu@DHPs NPs as an effective and safe platform amenable to vaccine delivery through their ability to provide robust adjuvant activity and sustained antigen release capable of eliciting long-term immunological memory while potentiating humoral and cellular immune responses. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Figure 1

31 pages, 1560 KB  
Review
Overcoming Immune Therapy Resistance in Cancer Through Innate Immune Reprogramming
by Giada Mandracci, Nardine Soliman and Nadia El Khawanky
Int. J. Mol. Sci. 2025, 26(19), 9554; https://doi.org/10.3390/ijms26199554 - 30 Sep 2025
Abstract
Overcoming immune resistance remains the critical barrier to durable immunotherapy responses. Tumors with non-inflamed, “cold” microenvironments exclude cytotoxic lymphocytes and evade checkpoint blockade. Innate nucleic acid-sensing pathways—including TLRs, RIG-I-like RNA sensors, and the cGAS–STING DNA-sensing axis—can recondition this hostile landscape by licensing dendritic [...] Read more.
Overcoming immune resistance remains the critical barrier to durable immunotherapy responses. Tumors with non-inflamed, “cold” microenvironments exclude cytotoxic lymphocytes and evade checkpoint blockade. Innate nucleic acid-sensing pathways—including TLRs, RIG-I-like RNA sensors, and the cGAS–STING DNA-sensing axis—can recondition this hostile landscape by licensing dendritic cells, restoring antigen presentation, and recruiting effector T and NK cells. In this review, we synthesize mechanistic insights into how these receptors function across tumor and immune compartments and evaluate recent translational advances spanning small-molecule and nucleic acid agonists, engineered delivery systems, and clinical trials. We highlight challenges that have limited clinical impact, including pathway silencing, systemic toxicity, and lack of predictive biomarkers, while emphasizing emerging solutions such as tumor-intrinsic targeting, CAR-T/NK engineering, and biomarker-guided patient selection. By integrating innate activation into rational combination regimens, innate immune reprogramming offers a blueprint to convert resistant disease into one susceptible to durable immune control. Full article
Show Figures

Figure 1

13 pages, 1961 KB  
Article
A CpG 1018S/QS-21-Adjuvanted HBsAg Therapeutic Vaccine as a Novel Strategy Against HBV
by Zixuan Wang, Jing Wu, Xiaohan Meng, He Weng, Qiang Li, Lin Li, Zhenhao Ma, Sirong Bi, Qiuju Han, Huajun Zhao, Cunbao Liu and Deping Meng
Vaccines 2025, 13(10), 1014; https://doi.org/10.3390/vaccines13101014 - 29 Sep 2025
Abstract
Chronic hepatitis B virus (HBV) infection remains a major global health challenge, substantially contributing to liver-related morbidity and mortality. Background/Objectives: Developing therapeutic strategies that overcome immune tolerance and achieve functional cures is an urgent priority. Methods: In this study, we report [...] Read more.
Chronic hepatitis B virus (HBV) infection remains a major global health challenge, substantially contributing to liver-related morbidity and mortality. Background/Objectives: Developing therapeutic strategies that overcome immune tolerance and achieve functional cures is an urgent priority. Methods: In this study, we report a therapeutic vaccine comprising hepatitis B surface antigen (HBsAg) formulated with the dual adjuvant system CpG 1018S and QS-21. The immunogenicity and therapeutic efficacy of this vaccine were systematically evaluated in an rAAV8-HBV1.3-established chronic HBV mouse model. Results: The vaccine elicited a robust Th1-skewed immune response, characterized by elevated anti-HBs IgG2b titers and an increased IgG2b/IgG1 ratio. Notably, immunized mice showed markedly reduced circulating HBsAg levels. Mechanistically, the CpG 1018S and QS-21 adjuvant system enhanced dendritic cell activation, maturation, and antigen presentation, expanded HBV-specific CD4+ and CD8+ T cell populations, and attenuated the expression of the exhaustion markers TIM-3 and TIGIT. Additionally, immunized mice exhibited restored T cell polyfunctionality, with an increased secretion of effector cytokines, including TNF-α and IL-21. These responses collectively contributed to the reversal of T cell exhaustion and breakdown of immune tolerance, facilitating sustained viral suppression. Conclusions: Our findings demonstrate that the CpG 1018S/QS-21-adjuvanted vaccine induces potent humoral and cellular immunity against chronic HBV infection and represents a promising candidate for clinical chronic HBV (CHB) treatment. Full article
(This article belongs to the Section Hepatitis Virus Vaccines)
Show Figures

Figure 1

11 pages, 1425 KB  
Review
Challenges in the Treatment of HIV-Related Lymphomas Complicated by COVID-19
by Kinga Siewiorek, Marcin Jasiński, Błażej Izdebski, Maciej Przybylski, Małgorzata Kobylecka, Joanna Mączewska, Krzysztof Jamroziak and Joanna Drozd-Sokołowska
Pharmaceuticals 2025, 18(10), 1461; https://doi.org/10.3390/ph18101461 - 28 Sep 2025
Abstract
Lymphomas remain a significant cause of morbidity and mortality among patients living with HIV. Although the introduction of antiretroviral therapy has led to a reduction in the incidence of AIDS-related lymphomas (ARL) and an overall improvement in prognosis, these malignancies continue to pose [...] Read more.
Lymphomas remain a significant cause of morbidity and mortality among patients living with HIV. Although the introduction of antiretroviral therapy has led to a reduction in the incidence of AIDS-related lymphomas (ARL) and an overall improvement in prognosis, these malignancies continue to pose a considerable clinical challenge. Beyond the inherent complexity of lymphoma treatment itself, the management of comorbidities, particularly infections, represents a therapeutic obstacle. Here, we review the published evidence on ARL complicated by COVID-19. Despite the fact that nearly 800 million confirmed cases of SARS-CoV-2 infection have been reported so far, only five cases of ARL and COVID-19 have been published, among whom most patients experienced a mild course of SARS-CoV-2 infection, with only one case progressing to severe COVID-19 that required oxygen therapy and prolonged hospitalization. Additionally, we present another case of a 49-year-old male patient with newly diagnosed ARL, Epstein–Barr virus (EBV)-positive, diffuse large B-cell lymphoma, not otherwise specified, complicated by prolonged SARS-CoV-2 infection. Although initially asymptomatic, the patient subsequently experienced transient respiratory failure. Despite administration of molnupiravir, both SARS-CoV-2 antigen and RT-qPCR tests remained positive for a minimum of 113 days. The prolonged SARS-CoV-2 infection, in conjunction with other opportunistic infections, impeded the delivery of adequate chemotherapy dose intensity and contributed to disease progression and ultimately the patient’s death. This case and review of the literature underscores the diversity of the clinical course of SARS-CoV-2 infection in patients with ARL and highlights the associated challenges in delivering optimal anti-lymphoma therapy in those patients. Full article
Show Figures

Figure 1

16 pages, 512 KB  
Review
CAR-T in the Treatment of Solid Tumors—A Review of Current Research and Future Perspectives
by Natalia Picheta, Julia Piekarz, Karolina Daniłowska, Katarzyna Szklener and Sławomir Mańdziuk
Int. J. Mol. Sci. 2025, 26(19), 9486; https://doi.org/10.3390/ijms26199486 - 28 Sep 2025
Abstract
The aim of this narrative review is to present the current state of knowledge regarding the use of chimeric antigen receptor T-cell (CAR-T) therapy in solid tumors. Phase I clinical trials and side effects are discussed. The review is based on an analysis [...] Read more.
The aim of this narrative review is to present the current state of knowledge regarding the use of chimeric antigen receptor T-cell (CAR-T) therapy in solid tumors. Phase I clinical trials and side effects are discussed. The review is based on an analysis of available scientific publications, primarily phase I trials, Food and Drug Administration (FDA) reports, and PubMed, Scopus, and Google Scholar sources. It includes clinical trials and review articles from 2016 to 2025. Accumulated data indicate promising efficacy of CAR-T therapy in the treatment of certain solid tumors, particularly those of the gastrointestinal tract, although clinical responses were often limited to disease stabilization. The therapy was generally well tolerated, with a low incidence of serious adverse events. Efficacy was found to depend on factors such as the type of target antigen, the presence of conditioning therapy, and the ability to overcome the immunosuppressive tumor microenvironment. CAR-T therapy remains experimental outside of hematological malignancies, but further development, refinement of receptor design, and the search for better molecular targets may make it an effective treatment option for solid tumors as well. Current studies are in early phase and require confirmation in larger-scale randomized trials. Full article
(This article belongs to the Special Issue 25th Anniversary of IJMS: Updates and Advances in Molecular Oncology)
Show Figures

Graphical abstract

35 pages, 2417 KB  
Review
Insights into Persistent SARS-CoV-2 Reservoirs in Chronic Long COVID
by Swayam Prakash, Sweta Karan, Yassir Lekbach, Delia F. Tifrea, Cesar J. Figueroa, Jeffrey B. Ulmer, James F. Young, Greg Glenn, Daniel Gil, Trevor M. Jones, Robert R. Redfield and Lbachir BenMohamed
Viruses 2025, 17(10), 1310; https://doi.org/10.3390/v17101310 - 27 Sep 2025
Abstract
Long COVID (LC), also known as post-acute sequelae of COVID-19 infection (PASC), is a heterogeneous and debilitating chronic disease that currently affects 10 to 20 million people in the U.S. and over 420 million people globally. With no approved treatments, the long-term global [...] Read more.
Long COVID (LC), also known as post-acute sequelae of COVID-19 infection (PASC), is a heterogeneous and debilitating chronic disease that currently affects 10 to 20 million people in the U.S. and over 420 million people globally. With no approved treatments, the long-term global health and economic impact of chronic LC remains high and growing. LC affects children, adolescents, and healthy adults and is characterized by over 200 diverse symptoms that persist for months to years after the acute COVID-19 infection is resolved. These symptoms target twelve major organ systems, causing dyspnea, vascular damage, cognitive impairments (“brain fog”), physical and mental fatigue, anxiety, and depression. This heterogeneity of LC symptoms, along with the lack of specific biomarkers and diagnostic tests, presents a significant challenge to the development of LC treatments. While several biological abnormalities have emerged as potential drivers of LC, a causative factor in a large subset of patients with LC, involves reservoirs of virus and/or viral RNA (vRNA) that persist months to years in multiple organs driving chronic inflammation, respiratory, muscular, cognitive, and cardiovascular damages, and provide continuous viral antigenic stimuli that overstimulate and exhaust CD4+ and CD8+ T cells. In this review, we (i) shed light on persisting virus and vRNA reservoirs detected, either directly (from biopsy, blood, stool, and autopsy samples) or indirectly through virus-specific B and T cell responses, in patients with LC and their association with the chronic symptomatology of LC; (ii) explore potential mechanisms of inflammation, immune evasion, and immune overstimulation in LC; (iii) review animal models of virus reservoirs in LC; (iv) discuss potential T cell immunotherapeutic strategies to reduce or eliminate persistent virus reservoirs, which would mitigate chronic inflammation and alleviate symptom severity in patients with LC. Full article
(This article belongs to the Special Issue SARS-CoV-2, COVID-19 Pathologies, Long COVID, and Anti-COVID Vaccines)
Show Figures

Figure 1

43 pages, 6032 KB  
Article
Modulation of mTOR Within Retinal Pigment Epithelium Affects Cell Viability and Mitochondrial Pathology
by Gloria Lazzeri, Michela Ferrucci, Paola Lenzi, Maria Anita Giambelluca, Francesca Biagioni, Carla Letizia Busceti, Alessandro Frati and Francesco Fornai
Int. J. Mol. Sci. 2025, 26(19), 9442; https://doi.org/10.3390/ijms26199442 - 26 Sep 2025
Abstract
The relevance of well-structured mitochondria in sustaining the integrity of the retinal pigment epithelium (RPE) is increasingly evident. Conversely, altered mitochondria are a culprit of age-related macular degeneration (AMD), which is influenced by the activity of mechanistic target of rapamycin (mTOR). In the [...] Read more.
The relevance of well-structured mitochondria in sustaining the integrity of the retinal pigment epithelium (RPE) is increasingly evident. Conversely, altered mitochondria are a culprit of age-related macular degeneration (AMD), which is influenced by the activity of mechanistic target of rapamycin (mTOR). In the present manuscript, the mitochondrial status of RPE cells was investigated by light and electron microscopy following the administration of various doses of compounds, which modulate mTOR. The study combines MitoTracker dyes and mitochondrial immunohistochemistry with in situ mitochondrial morphometry. Various doses of 3-methyladenine (3-MA), curcumin, and rapamycin were administered alone or in combination. The activity of autophagy and mTOR was quantified following each treatment. Administration of 3-MA led to activation of mTOR, which was associated with severe cell death, altered membrane permeability, and altered ZO-1 expression. In this condition, mitochondrial mass was reduced, despite a dramatic increase in damaged mitochondria being reported. The decrease in healthy mitochondria was concomitant with alterations in key mitochondria-related antigens such as Tomm20, Pink1, and Parkin. Specific mitochondrial alterations were quantified through in situ ultrastructural morphometry. Both curcumin and rapamycin counteract mTOR activation and rescue mitochondrial status, while preventing RPE cell loss and misplacement of decreased ZO-1 expression. Mitigation of mTOR may protect mitochondria in retinal degeneration. Full article
(This article belongs to the Special Issue Molecular Pathways of Proteostasis in Aging and Diseases)
Show Figures

Figure 1

17 pages, 7798 KB  
Article
Dendritic Cell-Cytokine-Induced Killer Cells Co-Loaded with WT1/MUC1/Poly(I:C) Enhance Antitumor Immune Responses In Vitro and In Vivo
by Huimin Liu, Chenlong Wang, Hongtao Chang, Liangliang Dong, Guoqing Yang, Cailing Tong and Lin Mao
Biomolecules 2025, 15(10), 1356; https://doi.org/10.3390/biom15101356 - 24 Sep 2025
Viewed by 72
Abstract
Dendritic cell-cytokine-induced killer (DC-CIK) therapy faces limitations due to antigenic heterogeneity and suboptimal immune activation. In this study, we developed a multi-antigen-loaded DC-CIK (Ag-DC-CIK) system that co-targets Wilms’ tumor 1 (WT1), mucin-1 (MUC1), and the TLR3 agonist poly(I:C) to improve therapeutic outcomes. Utilizing [...] Read more.
Dendritic cell-cytokine-induced killer (DC-CIK) therapy faces limitations due to antigenic heterogeneity and suboptimal immune activation. In this study, we developed a multi-antigen-loaded DC-CIK (Ag-DC-CIK) system that co-targets Wilms’ tumor 1 (WT1), mucin-1 (MUC1), and the TLR3 agonist poly(I:C) to improve therapeutic outcomes. Utilizing umbilical cord blood-derived DC and CIK cells, we demonstrated that Ag-DC-CIK significantly enhanced cytotoxicity, as evidenced by the lactate dehydrogenase (LDH) assay, and increased apoptosis induction, indicated by elevated Bax and reduced Bcl-2 expression, in various tumor cell lines (HeLa, HCT116, MKN45) and organoids generated from a gastric cancer patient. Furthermore, Ag-DC-CIK effectively suppressed tumor cell migration and reduced the viability of the organoid. In MKN45 xenograft models, Ag-DC-CIK treatment inhibited tumor growth without inducing systemic toxicity, as shown by decreased Ki67 cell proliferation. This tripartite strategy synergistically enhances DC-CIK therapy by expanding antigen recognition and augmenting immune responses, presenting a promising translational approach for the treatment of gastric cancer. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

13 pages, 1630 KB  
Article
Nodal Spread Prediction in Human Oral Tongue Squamous Cell Carcinoma Using a Cancer-Testis Antigen Genes Signature
by Yoav Smith, Amit Cohen, Tzahi Neuman, Yoram Fleissig and Nir Hirshoren
Int. J. Mol. Sci. 2025, 26(18), 9258; https://doi.org/10.3390/ijms26189258 - 22 Sep 2025
Viewed by 164
Abstract
Cervical lymph node metastasis is the strongest prognostic factor in oral tongue carcinoma, yet current clinical guidelines rely primarily on depth of invasion to guide elective neck dissection. This approach results in unnecessary surgery in up to 70% of patients. Cancer-testis antigens (CTAs) [...] Read more.
Cervical lymph node metastasis is the strongest prognostic factor in oral tongue carcinoma, yet current clinical guidelines rely primarily on depth of invasion to guide elective neck dissection. This approach results in unnecessary surgery in up to 70% of patients. Cancer-testis antigens (CTAs) are a family of genes associated with tumor aggressiveness and may serve as predictive biomarkers for nodal spread. A multi-step analysis integrating large-scale public datasets, including microarray (GSE78060), bulk RNA-seq emerging from the cancer genome atlas (TCGA), and single-cell RNA-seq (GSE103322), was employed to identify CTA genes active in oral tongue cancer. Selected genes were validated using NanoString nCounter RNA profiling of 16 patients undergoing curative glossectomy with elective neck dissection. Machine learning algorithms, including decision trees, t-distributed stochastic neighbor embedding (t-SNE), and convolutional neural networks (CNN), were applied to assess predictive power for nodal metastasis. Computational analysis initially identified 40 cancer-active CTA genes, of which four genes (LY6K, MAGEA3, CEP55, and ATAD2) were most indicative of nodal spread. In our patient cohort, NanoString nCounter profiling combined with machine learning confirmed these four genes as highly predictive. We present a proof-of-concept CTA-based genetic diagnostic tool capable of discriminating nodal involvement in oral tongue cancer. This approach may reduce unnecessary neck dissections, minimizing surgical morbidity. Full article
(This article belongs to the Special Issue The Role of Genome in Cancer Therapy)
Show Figures

Figure 1

Back to TopTop