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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (518)

Search Parameters:
Keywords = immunosuppressive molecules

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
42 pages, 939 KiB  
Review
B7-H3 in Cancer Immunotherapy—Prospects and Challenges: A Review of the Literature
by Sylwia Mielcarska, Anna Kot, Miriam Dawidowicz, Agnieszka Kula, Piotr Sobków, Daria Kłaczka, Dariusz Waniczek and Elżbieta Świętochowska
Cells 2025, 14(15), 1209; https://doi.org/10.3390/cells14151209 - 6 Aug 2025
Abstract
In today’s oncology, immunotherapy arises as a potent complement for conventional cancer treatment, allowing for obtaining better patient outcomes. B7-H3 (CD276) is a member of the B7 protein family, which emerged as an attractive target for the treatment of various tumors. The molecule [...] Read more.
In today’s oncology, immunotherapy arises as a potent complement for conventional cancer treatment, allowing for obtaining better patient outcomes. B7-H3 (CD276) is a member of the B7 protein family, which emerged as an attractive target for the treatment of various tumors. The molecule modulates anti-cancer immune responses, acting through diverse signaling pathways and cell populations. It has been implicated in the pathogenesis of numerous malignancies, including melanoma, gliomas, lung cancer, gynecological cancers, renal cancer, gastrointestinal tumors, and others, fostering the immunosuppressive environment and marking worse prognosis for the patients. B7-H3 targeting therapies, such as monoclonal antibodies, antibody–drug conjugates, and CAR T-cells, present promising results in preclinical studies and are the subject of ongoing clinical trials. CAR-T therapies against B7-H3 have demonstrated utility in malignancies such as melanoma, glioblastoma, prostate cancer, and RCC. Moreover, ADCs targeting B7-H3 exerted cytotoxic effects on glioblastoma, neuroblastoma cells, prostate cancer, and craniopharyngioma models. B7-H3-targeting also delivers promising results in combined therapies, enhancing the response to other immune checkpoint inhibitors and giving hope for the development of approaches with minimized adverse effects. However, the strategies of B7-H3 blocking deliver substantial challenges, such as poorly understood molecular mechanisms behind B7-H3 protumor properties or therapy toxicity. In this review, we discuss B7-H3’s role in modulating immune responses, its significance for various malignancies, and clinical trials evaluating anti-B7-H3 immunotherapeutic strategies, focusing on the clinical potential of the molecule. Full article
Show Figures

Figure 1

12 pages, 814 KiB  
Review
Cancer Resistance to Immunotherapy
by Rita Khoury, Annoir Shayya, Cendrella Bou Orm, Osama Zein Deen and Hady Ghanem
Immuno 2025, 5(3), 32; https://doi.org/10.3390/immuno5030032 - 5 Aug 2025
Abstract
Immunotherapy has revolutionized cancer treatment. Despite its success across various malignancies, a significant proportion of patients either fail to respond (primary resistance) or relapse after an initial response (acquired resistance). This review explores the different mechanisms underlying resistance to immunotherapy, including tumor-intrinsic factors [...] Read more.
Immunotherapy has revolutionized cancer treatment. Despite its success across various malignancies, a significant proportion of patients either fail to respond (primary resistance) or relapse after an initial response (acquired resistance). This review explores the different mechanisms underlying resistance to immunotherapy, including tumor-intrinsic factors such as loss of antigen presentation, genetic, and epigenetic mutations. It also examines tumor-extrinsic contributors, such as immunosuppressive cells in the tumor microenvironment, checkpoint molecule upregulation, and microbiome influences. A comprehensive understanding of resistance mechanisms is essential for improving patient selection, developing combination therapies, and ultimately enhancing the efficacy and durability of immunotherapeutic interventions. Full article
Show Figures

Figure 1

15 pages, 1064 KiB  
Article
Targeting RARγ Decreases Immunosuppressive Macrophage Polarization and Reduces Tumor Growth
by Jihyeon Park, Jisun Oh, Sang-Hyun Min, Ji Hoon Yu, Jong-Sup Bae and Hui-Jeon Jeon
Molecules 2025, 30(15), 3099; https://doi.org/10.3390/molecules30153099 - 24 Jul 2025
Viewed by 273
Abstract
Tumor-associated macrophages (TAMs) play a critical role in the tumor microenvironment (TME), interacting with cancer cells and other components to promote tumor growth. Given the influence of TAMs on tumor progression and resistance to therapy, regulating the activity of these macrophages is crucial [...] Read more.
Tumor-associated macrophages (TAMs) play a critical role in the tumor microenvironment (TME), interacting with cancer cells and other components to promote tumor growth. Given the influence of TAMs on tumor progression and resistance to therapy, regulating the activity of these macrophages is crucial for improving cancer treatment outcomes. TAMs often exhibit immunosuppressive phenotypes (commonly referred to as M2-like macrophages), which suppress immune responses and contribute to drug resistance. Therefore, inhibiting immunosuppressive polarization offers a promising strategy to impede tumor growth. This study revealed retinoic acid receptor gamma (RARγ), a nuclear receptor, as a key regulator of immunosuppressive polarization in THP-1 macrophages. Indeed, the inhibition of RARγ, either by a small molecule or gene silencing, significantly reduced the expression of immunosuppressive macrophage markers. In a three-dimensional tumor spheroid model, immunosuppressive macrophages enhanced the proliferation of HCT116 colorectal cancer cells, which was significantly hindered by RARγ inhibition. These findings suggest that targeting RARγ reprograms immunosuppressive macrophages and mitigates the tumor-promoting effects of TAMs, highlighting RARγ as a promising therapeutic target for developing novel anti-cancer strategies. Full article
Show Figures

Figure 1

17 pages, 277 KiB  
Review
Harnessing miRNA-Containing Extracellular Vesicles from Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Regeneration of Bone Defects: A Narrative Review of Mechanisms, Biomaterials, and Clinical Translation
by Kashia Goto, Daisuke Watanabe, Kazuki Yanagida, Tatsuya Takagi and Akio Mizushima
Cancers 2025, 17(15), 2438; https://doi.org/10.3390/cancers17152438 - 23 Jul 2025
Viewed by 277
Abstract
We present a narrative review focusing on the therapeutic potential of mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) in regenerating bone defects, particularly those resulting from surgical treatment of malignant bone and soft tissue tumors. These large bone defects pose significant challenges for reconstruction [...] Read more.
We present a narrative review focusing on the therapeutic potential of mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) in regenerating bone defects, particularly those resulting from surgical treatment of malignant bone and soft tissue tumors. These large bone defects pose significant challenges for reconstruction and functional recovery, highlighting the need for innovative regenerative strategies. Background: MSCs, which can differentiate into various cell types, are known for their immunosuppressive properties and ability to promote tissue repair. MSC-EVs, rich in bioactive molecules like microRNAs and proteins, play a crucial role in bone regeneration by mediating intercellular communication and modulating inflammation. Methods: This narrative review compiles data from various studies, including systematic reviews and individual research, focusing on the application of MSC-EVs in bone defect treatment. It examines the characteristics, mechanisms of action, and therapeutic effects of MSC-EVs, as well as the microRNAs involved in bone regeneration. Results: The findings indicate that MSC-EVs can enhance both osteogenesis and angiogenesis, highlighting their potential as promising candidates for clinical applications in bone defects. However, many mechanisms remain unclear; therefore, further investigation is needed. Conclusions: The review emphasizes the potential of MSC-EVs in improving patient outcomes for severe bone defects. It also highlights future challenges, including formulation, standardization, safety, and delivery methods, particularly in conjunction with biomaterials. Overall, MSC-EVs represent a significant advancement in regenerative medicine for bone defects. Full article
(This article belongs to the Special Issue Advances in Soft Tissue and Bone Sarcoma (2nd Edition))
20 pages, 1400 KiB  
Review
Novel Therapeutics and the Path Toward Effective Immunotherapy in Malignant Peripheral Nerve Sheath Tumors
by Joshua J. Lingo, Elizabeth C. Elias and Dawn E. Quelle
Cancers 2025, 17(14), 2410; https://doi.org/10.3390/cancers17142410 - 21 Jul 2025
Viewed by 485
Abstract
Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are a deadly subtype of soft tissue sarcoma for which effective therapeutic options are lacking. Currently, the best treatment for MPNSTs is complete surgical resection with wide negative margins, but this is often complicated by the tumor [...] Read more.
Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are a deadly subtype of soft tissue sarcoma for which effective therapeutic options are lacking. Currently, the best treatment for MPNSTs is complete surgical resection with wide negative margins, but this is often complicated by the tumor size and location and/or the presence of metastases. Radiation or chemotherapy may be combined with surgery, but patient responses are poor. Targeted treatments, including small-molecule inhibitors of oncogenic proteins such as mitogen-activated protein kinase kinase (MEK), cyclin-dependent kinases 4 and 6 (CDK4/6), and Src-homology 2 domain-containing phosphatase 2 (SHP2), are promising therapeutics for MPNSTs, especially when combined together, but they have yet to gain approval. Immunotherapeutic approaches have been revolutionary for the treatment of some other cancers, but their utility as single agents in sarcoma is limited and not approved for MPNSTs. The immunosuppressive niche of MPNSTs is thought to confer inherent treatment resistance, particularly to immunotherapies. Remodeling an inherently “cold” tumor microenvironment into a “hot” immune milieu to bolster the anti-tumor activity of immunotherapies is of great interest throughout the cancer community. This review focuses on novel therapeutics that target dysregulated factors and pathways in MPNSTs, as well as different types of immunotherapies currently under investigation for this disease. We also consider how certain therapeutics may be combined to remodel the MPNST immune microenvironment and thereby generate a durable anti-tumor immune response to immunotherapy. Full article
(This article belongs to the Special Issue Next-Generation Cancer Therapies)
Show Figures

Figure 1

17 pages, 8756 KiB  
Article
A Diet Rich in Essential Amino Acids Inhibits the Growth of HCT116 Human Colon Cancer Cell In Vitro and In Vivo
by Giovanni Corsetti, Claudia Romano, Silvia Codenotti, Evasio Pasini, Alessandro Fanzani, Tiziano Scarabelli and Francesco S. Dioguardi
Int. J. Mol. Sci. 2025, 26(14), 7014; https://doi.org/10.3390/ijms26147014 - 21 Jul 2025
Viewed by 337
Abstract
The metabolic hyperactivity of tumor cells demands a substantial amount of energy and molecules to build new cells and expand the tumor, diverting these resources from healthy cells. Amino acids (AAs) are the only totipotent and essential molecules for protein construction. Previous in [...] Read more.
The metabolic hyperactivity of tumor cells demands a substantial amount of energy and molecules to build new cells and expand the tumor, diverting these resources from healthy cells. Amino acids (AAs) are the only totipotent and essential molecules for protein construction. Previous in vitro studies in human and murine cancer cells, along with in vivo studies in mice, have shown that an excess of essential amino acids (EAAs) exerts an inhibitory effect on tumor proliferation by promoting apoptosis and autophagy. In this study, both in vitro and in vivo, we evaluated whether a mixture based on EAA can influence the development of human colon cancer (HCT116). To this end, in vitro, we assessed the proliferation of HCT116 cells treated with a special mix of EAA. In vivo, immunosuppressed athymic nude mice, injected with HCT116 cells subcutaneously (s.c.) or intraperitoneally (i.p.), were given a modified EAAs-rich diet (EAARD) compared to the standard laboratory diet (StD). In vitro data showed that the EAA mix impairs cancer growth by inducing apoptosis and autophagy. In vivo, the results demonstrated that EAARD-fed mice developed s.c. tumors significantly smaller than those of StD-fed mice (total mass 3.24 vs. 6.09 g, respectively). Mice injected i.p. and fed with EAARD showed a smaller and more limited number of intra-peritoneal tumors than StD-fed mice (total mass 0.79 vs. 4.77 g, respectively). EAAs prevents the growth of HCT116 cells by inducing autophagy and apoptosis, increasing endoplasmic reticulum stress, and inhibiting inflammation and neo-vascularization. In addition, the EAARD-fed mice, maintained muscle mass and white and brown adipose tissues. A diet with an excess of EAAs affects the survival and proliferative capacity of human colon cancer cells, maintaining anabolic stimuli in muscular cells. Full article
(This article belongs to the Special Issue Innovative Research on Nutrition and Epigenetics in Cancer)
Show Figures

Graphical abstract

13 pages, 6330 KiB  
Article
Erythroblasts Promote the Development of a Suppressive Lymphocyte Phenotype via Treg Induction and PD1 Upregulation on the Surfaces of B-Cells: A Study on the Subpopulation-Specific Features of Erythroblasts
by Kirill Nazarov, Roman Perik-Zavodskii, Julia Shevchenko and Sergey Sennikov
Curr. Issues Mol. Biol. 2025, 47(7), 550; https://doi.org/10.3390/cimb47070550 - 15 Jul 2025
Viewed by 243
Abstract
This study identifies the novel effects of soluble factors derived from murine erythroblasts on lymphoid cell phenotypes. These effects were observed following the treatment of splenic mononuclear cells with erythroblast-conditioned media received from both healthy mice and mice subjected to hematopoiesis-activating conditions (hypoxia, [...] Read more.
This study identifies the novel effects of soluble factors derived from murine erythroblasts on lymphoid cell phenotypes. These effects were observed following the treatment of splenic mononuclear cells with erythroblast-conditioned media received from both healthy mice and mice subjected to hematopoiesis-activating conditions (hypoxia, blood loss, and hemolytic anemia), suggesting a common mechanism of action. Using flow cytometry, we elucidated that erythroblast-derived soluble products modulate T cell differentiation by promoting Treg development and increasing PD-1 surface expression on B cells. The immunoregulatory potential of erythroblasts is subpopulation-dependent: CD45+ erythroblasts respond to hemolytic stress by upregulating the surface expression of immunosuppressive molecules PDL1 and Galectin-9, while CD45- erythroblasts primarily increase TGFb production. These findings highlight the regulatory role of erythroblasts in modulating immune responses. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

31 pages, 2698 KiB  
Review
Tumor Microenvironment in Melanoma—Characteristic and Clinical Implications
by Hubert Sikorski, Michał Aleksander Żmijewski and Anna Piotrowska
Int. J. Mol. Sci. 2025, 26(14), 6778; https://doi.org/10.3390/ijms26146778 - 15 Jul 2025
Viewed by 828
Abstract
Cutaneous melanoma is an aggressive cancer with an increasing incidence worldwide, highlighting the need for research into its pathogenesis. The tumor microenvironment (TME) plays a critical role in melanoma progression and consists of cellular components and an extracellular matrix (ECM) rich in cytokines [...] Read more.
Cutaneous melanoma is an aggressive cancer with an increasing incidence worldwide, highlighting the need for research into its pathogenesis. The tumor microenvironment (TME) plays a critical role in melanoma progression and consists of cellular components and an extracellular matrix (ECM) rich in cytokines and signaling molecules. The most abundant stromal cells within the TME are cancer-associated fibroblasts (CAFs), which remodel the ECM and modulate immune responses. Among immune cells, tumor-associated macrophages (TAMs) predominate, and their polarization toward the M2 phenotype supports tumor progression. Tumor-infiltrating lymphocytes (TILs) have diverse functions, including cytotoxic T-cells, helper T-cells that modulate immune response, B-cells forming tertiary lymphoid structures (TLS), and regulatory T-cells with immunosuppressive properties. Dendritic cells (DCs) also play a complex role in the TME. A notable subpopulation are mature regulatory dendritic cells (mregDCs), which contribute to immune evasion. All of these TME components may drive tumorigenesis. Advancements in melanoma treatment—including immunotherapy and targeted therapies—have significantly improved outcomes in advanced-stage disease. In parallel, emerging approaches targeting the tumor microenvironment and gut microbiome, as well as personalized strategies such as neoantigen vaccines and cell-based therapies, are under active investigation and may further enhance therapeutic efficacy in the near future. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies for Melanoma)
Show Figures

Figure 1

16 pages, 1236 KiB  
Communication
Chemoradiation-Altered Micromilieu of Glioblastoma Cells Particularly Impacts M1-like Macrophage Activation
by Mona Shojaei, Benjamin Frey, Florian Putz, Rainer Fietkau, Udo S. Gaipl and Anja Derer
Int. J. Mol. Sci. 2025, 26(14), 6574; https://doi.org/10.3390/ijms26146574 - 8 Jul 2025
Viewed by 448
Abstract
Glioblastoma is a highly aggressive brain tumor with an overall poor prognosis due to its immunosuppressive tumor microenvironment (TME). Microglia and tumor-associated macrophages (TAMs) with pro-tumorigenic properties are dominant populations of immune cells in the glioblastoma TME. To date, several studies targeting TAMs [...] Read more.
Glioblastoma is a highly aggressive brain tumor with an overall poor prognosis due to its immunosuppressive tumor microenvironment (TME). Microglia and tumor-associated macrophages (TAMs) with pro-tumorigenic properties are dominant populations of immune cells in the glioblastoma TME. To date, several studies targeting TAMs to fight tumor progression in different tumor entities have been initiated. However, the impact of standard therapy schemes of glioblastoma cells on macrophage polarization, activation, and phagocytosis remains controversial. The same applies to the relevance of PD-1/PD-L1 blockade in the interaction between macrophages and tumor cells. Our study, therefore, investigated patient-oriented treatment of GLIOBLASTOMA by examining the phagocytic capacity of polarized M1- and M2-like macrophages using GL261-luc2 tumor cells as a preclinical model system. Additionally, we analyzed the expression of activation and immune checkpoint markers on these macrophage subtypes following contact with tumor cells and their microenvironment. These factors were also determined after PD-1 blockade was initiated. The analyses revealed that the immunoregulatory M2-like macrophages generally exhibited a higher phagocytosis rate than the pro-inflammatory M1-like macrophages; however, this was not influenced by the pretreatment of glioblastoma cells with chemo- or radiotherapy. This could not be improved by blocking the PD-1 receptor. Furthermore, there were no modulations in the expression of differentiation, activation, or immune checkpoint molecules of M1- and M2-like macrophages after cell-to-cell contact with glioblastoma cells. But the medium conditioned by tumor cells strongly altered M1-like macrophages toward a more activated state, whereas M2-like cells were only mildly influenced. This was further enhanced by tumor cell treatment, with the most prominent effect after irradiation. These results suggest that conventional GLIOBLASTOMA tumor cell treatment affects the immunogenic status of macrophage subtypes, which is relevant for enhancing the anti-tumor immune response in brain tumors. Full article
(This article belongs to the Special Issue The Role of Macrophages in Cancers)
Show Figures

Figure 1

29 pages, 1175 KiB  
Review
Tumor Microenvironment: An Emerging Landscape for Lung Cancer Therapy
by S. M. Sohag, Sharmin Nur Toma, Md. Al-Imran Imon, Maiweilan Maihemuti, Famim Ahmed, Mst. Afsana Mimi, Imran Mahmud and Md. Mahmudul Hasan
Future Pharmacol. 2025, 5(3), 34; https://doi.org/10.3390/futurepharmacol5030034 - 30 Jun 2025
Viewed by 498
Abstract
The tumor microenvironment (TME) is crucial for the onset, development, and resistance to treatment of lung cancer. The tumor microenvironment consisting of a complex array of immune cells, fibroblasts, endothelial cells, extracellular matrix elements, and signaling molecules, facilitates tumor growth and spread while [...] Read more.
The tumor microenvironment (TME) is crucial for the onset, development, and resistance to treatment of lung cancer. The tumor microenvironment consisting of a complex array of immune cells, fibroblasts, endothelial cells, extracellular matrix elements, and signaling molecules, facilitates tumor growth and spread while inhibiting the body’s antitumor immune response. In lung cancer, tumor-associated macrophages, cancer-associated fibroblasts, mast cells, and dendritic cells interact through cytokines, chemokines, growth factors, and matrix metalloproteinases to create an immunosuppressive and proangiogenic milieu. Hypoxic conditions within the TME further enhance cancer cell adaptability through hypoxia-inducible factors (HIFs), promoting epithelial–mesenchymal transition, immune evasion, and metastasis. Moreover, miRNAs have emerged as key regulators of gene expression within the TME, offering novel insights into tumor behavior and potential therapeutic targets. Targeting dynamic interactions within the TME, particularly through the modulation of immune responses, angiogenesis, and stromal remodeling, offers promising avenues for precision pharmacological approaches. This review covers the current understanding of the lung TME, highlighting its impact on cancer pathophysiology and treatment strategies. Understanding and therapeutically reprogramming the TME may pave the way for personalized and more effective interventions for lung cancer treatment. Full article
Show Figures

Figure 1

21 pages, 5329 KiB  
Article
Development of Immune-Regulatory Pseudo-Protein-Coated Iron Oxide Nanoparticles for Enhanced Treatment of Triple-Negative Breast Tumor
by Ying Ji, Juan Li, Li Ma, Zhijie Wang, Bochu Du, Hiu Yee Kwan, Zhaoxiang Bian and Chih-Chang Chu
Nanomaterials 2025, 15(13), 1006; https://doi.org/10.3390/nano15131006 - 30 Jun 2025
Viewed by 460
Abstract
Triple-negative breast cancer (TNBC) frequently evades immune recognition and elimination, resulting in an immunosuppressive microenvironment. The phagocytic activity of tumor-associated macrophages underscores the development of nanomaterials as a promising strategy to target these macrophages and modulate their polarization, thereby advancing immunotherapy against TNBC. [...] Read more.
Triple-negative breast cancer (TNBC) frequently evades immune recognition and elimination, resulting in an immunosuppressive microenvironment. The phagocytic activity of tumor-associated macrophages underscores the development of nanomaterials as a promising strategy to target these macrophages and modulate their polarization, thereby advancing immunotherapy against TNBC. This research developed functional polymers that are complexed with therapeutic molecules as a coating strategy for iron oxide nanoparticles. An arginine-based poly (ester urea urethane) polymer complexed with a macrophage-polarizing molecule (APU-R848) could provide a synergistic effect with iron oxide nanoparticles (IONPs) to stimulate the M1-polarization of macrophages at the tumor site, resulting in a versatile nano-platform for immune regulation of TNBC. In the 4T1 in vivo breast tumor model, the APU-R848-IONPs demonstrated an improved intratumoral biodistribution compared to IONPs without a polymer coating. APU-R848-IONPs significantly reversed the immune-suppressive tumor environment by reducing the M2/M1 macrophage phenotype ratio by 51%, associated with an elevated population of cytotoxic T cells and a significantly enhanced production of tumoricidal cytokines. The activated immune response induced by APU-R848-IONP resulted in a significant anti-tumor effect, demonstrating an efficacy that was more than 3.2-fold more efficient compared to the controls. These immune-regulatory pseudo-protein-coated iron oxide nanoparticles represent an effective nano-strategy for macrophages’ regulation and the activation of anti-tumor immunity, providing a new treatment modality for triple-negative breast cancer. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Figure 1

18 pages, 9359 KiB  
Article
Ovalbumin Peptide–Selenium Nanoparticles Alleviate Immune Suppression in Cyclophosphamide-Induced Mice: A Combined Transcriptomic and Proteomic Approach to Reveal the Mechanism
by Yingnan Zeng, Qi Yang, Zhiyang Du, Xuanting Liu, Xiaomin Shang, Menglei Xu, Jingbo Liu, Siwen Lyu and Ting Zhang
Foods 2025, 14(13), 2295; https://doi.org/10.3390/foods14132295 - 28 Jun 2025
Viewed by 480
Abstract
Immunocompromise is a growing health concern, and food-derived immunomodulators are expected to serve as a valuable supplement to traditional drug therapies. Ovalbumin peptide (OP) was employed as a stabilizer to prepare OP–selenium nanoparticles (OP-SeNPs), which showed immunomodulatory effects in vitro; however, the effects [...] Read more.
Immunocompromise is a growing health concern, and food-derived immunomodulators are expected to serve as a valuable supplement to traditional drug therapies. Ovalbumin peptide (OP) was employed as a stabilizer to prepare OP–selenium nanoparticles (OP-SeNPs), which showed immunomodulatory effects in vitro; however, the effects and underlying mechanisms in vivo were not yet fully understood. This study investigated the immunomodulatory activity of OP-SeNPs in cyclophosphamide (CTX)-induced immunosuppressed mice on immune organs, molecules, and cells, with the underlying mechanism explored by transcriptomic and proteomic studies. The results demonstrated that OP-SeNPs alleviated tissue damage in the spleen and thymus, improved the immunosuppressive state by promoting the secretion of cytokines (IL-1β, IFN-γ, IL-4, and IL-6), immunoglobulins (IgA, IgG, IgM, and sIgA), and promoting the proliferation of splenic lymphocytes. PI3K-Akt, Rap1, p53, PPAR, and Hippo signaling pathways formed an important regulatory network that synergistically influenced immune modulation. OP-SeNPs are potential food-derived immunomodulators, setting the stage for deep exploration of the mechanisms driving their immunomodulatory effects. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

18 pages, 996 KiB  
Review
Future Perspectives and Conclusions from Animal Models of CHI3L1-Related Inflammation-Associated Cancer
by Emiko Mizoguchi and Siyuan Wang
Cells 2025, 14(13), 982; https://doi.org/10.3390/cells14130982 - 26 Jun 2025
Viewed by 659
Abstract
Among the molecules implicated in inflammation-associated tumorigenesis, Chitinase 3-like 1 (CHI3L1/YKL-40/Brp-39) has emerged as a particularly compelling target due to its multifaced roles in immune regulation, tissue remodeling, and cancer progression. Elevated CHI3L1 expression is observed in various human cancers and corresponding animal [...] Read more.
Among the molecules implicated in inflammation-associated tumorigenesis, Chitinase 3-like 1 (CHI3L1/YKL-40/Brp-39) has emerged as a particularly compelling target due to its multifaced roles in immune regulation, tissue remodeling, and cancer progression. Elevated CHI3L1 expression is observed in various human cancers and corresponding animal models. CHI3L1 directly promotes tumor cell proliferation and angiogenesis and also contributes to immune evasion by establishing an immunosuppressive environment in inflamed tissues. Mechanistically, CHI3L1 exerts its effects through the modulation of STAT3, MAPK, and PI3K/Akt signaling pathways and by interacting with cell surface receptors, such as IL-13Rα2 and RAGE. Studies using transgenic and knockout mouse models have revealed a strong association between CHI3L1 expression and cancer progression. In models of colon and lung cancer, CHI3L1 overexpression correlates with increased tumor size and number, whereas CHI3L1 deficiency markedly suppresses tumor formation. However, its involvement appears to be context-dependent and varies among different epithelial tumor types. These findings suggest that CHI3L1 is a potential therapeutic target and diagnostic biomarker for inflammation-associated cancers. Animal studies provide valuable insights into the immunological mechanisms of CHI3L1-mediated tumorigenesis but also highlight the need for cautious interpretation due to inherent technical limitations. Full article
(This article belongs to the Special Issue Pathogenic Mechanisms of Chronic Inflammation-Associated Cancer)
Show Figures

Figure 1

13 pages, 2263 KiB  
Article
Subclassification-Specific Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Implications for Appropriate Pharmacotherapy
by Masahiko Kinoshita, Yasunori Sato, Shoji Kubo, Hiroji Shinkawa, Kenjiro Kimura, Kohei Nishio, Ryota Tanaka, Shigeaki Kurihara and Takeaki Ishizawa
Cancers 2025, 17(13), 2082; https://doi.org/10.3390/cancers17132082 - 21 Jun 2025
Viewed by 353
Abstract
Background/Objectives: Intrahepatic cholangiocarcinoma (iCCA) is subclassified into small- and large-duct types. Small-duct-type iCCAs are associated with a better prognosis, and each subclassification requires different surgical strategies. The efficacy of chemotherapy, including immune checkpoint inhibitors, may vary between subclassifications. However, there are no [...] Read more.
Background/Objectives: Intrahepatic cholangiocarcinoma (iCCA) is subclassified into small- and large-duct types. Small-duct-type iCCAs are associated with a better prognosis, and each subclassification requires different surgical strategies. The efficacy of chemotherapy, including immune checkpoint inhibitors, may vary between subclassifications. However, there are no reports on tumor immune microenvironment (TIME) analyses based on iCCA subclassifications. This study investigated subclassification-specific TIMEs in iCCAs for the purpose of establishing appropriate pharmacotherapy. Methods: A total of 131 resected iCCA cases were analyzed, comprising 73 tumors classified as small-duct-type and 58 as large-duct-type based on pathological evaluation. Immunohistochemical analyses targeting CD8, PD-1, PD-L1, CTLA-4, and S100 protein (a dendritic cell [DC] marker) were performed to investigate the immune-cell status in each subclassification. Results: Large-duct-type iCCA had a significantly higher CD8 expression in tumor-infiltrating cells than small-duct-type ICC. However, the expression of other molecules did not significantly differ between the two tumor types. The proportion of tumors with a high level of S100 protein expression (DC-high group) in tumor-infiltrating cells was significantly higher in small-duct-type ICCs than in large-duct-type iCCAs (30% vs. 1.7%). In small-duct-type iCCAs, the expression levels of CD8, PD-1, PD-L1, and CTLA-4 were significantly higher in the DC-high group than in the DC-low group. Conclusions: We revealed subclassification-specific TIMEs in iCCAs. A subset of small-duct-type iCCAs exhibited strong DC infiltration. In these patients, the tumors may establish an immunosuppressive TIME to evade antitumor immunity triggered by DC-mediated antigen presentation. These findings may contribute to the development of tailored pharmacotherapy for each iCCA subclassification. Full article
(This article belongs to the Special Issue The Pathology of Biliary Tract Carcinoma (BTC))
Show Figures

Figure 1

25 pages, 1626 KiB  
Review
From Fish Oil to Resolution: A Narrative Review on the Potential of SPM-Enriched Marine Oil for Exercise-Induced Muscle Damage Recovery
by Leticia C. de Souza, Jose M. Moris, Paul M. Gordon, Jeffery L. Heileson and LesLee K. Funderburk
Nutrients 2025, 17(12), 2014; https://doi.org/10.3390/nu17122014 - 16 Jun 2025
Viewed by 1593
Abstract
Exercise-induced muscle damage (EIMD) initiates an inflammatory response that is essential for tissue repair. However, when prolonged or excessive, this response can impair recovery and muscular performance. Specialized pro-resolving mediators (SPMs), derived from the metabolism of omega-3 (n-3) polyunsaturated fatty acids [...] Read more.
Exercise-induced muscle damage (EIMD) initiates an inflammatory response that is essential for tissue repair. However, when prolonged or excessive, this response can impair recovery and muscular performance. Specialized pro-resolving mediators (SPMs), derived from the metabolism of omega-3 (n-3) polyunsaturated fatty acids (PUFAs), facilitate the resolution of inflammation without causing immunosuppression. Evidence from preclinical studies indicates that SPM administration accelerates muscle repair and functional recovery by enhancing the clearance of apoptotic cells, suppressing pro-inflammatory signaling and modulating macrophage polarization. However, translation to human applications remains limited as commercially available SPM-enriched marine oils do not contain active SPMs but rather their monohydroxylated precursors, including 14-Hydroxy-Docosahexaenoic Acid (14-HDHA), 17-Hydroxy-Docosahexaenoic Acid (17-HDHA), and 18-Hydroxy-Eicosapentaenoic Acid (18-HEPE) in addition to low doses of the n-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Furthermore, the variable increases in circulating SPM concentrations as a result of dietary intake of EPA and DHA, whether from fish or fish oil supplements, and the wide diversity of SPM molecules (many of which remain under investigation), highlight the complexity of their structural and functional networks. While advances in lipidomics have identified SPMs and their pathway intermediates in human biological samples, further research is needed to determine optimal dosing strategies, delivery mechanisms, and the real impact of SPM-enriched marine oil on athletic performance and recovery. This narrative review examines the biological rationale and current evidence surrounding SPM-enriched marine oil supplementation and its potential to enhance muscle recovery following EIMD. By synthesizing findings from preclinical and human studies, the potential of SPM-enriched supplementation as a novel tool for optimizing performance recovery in athletic populations is reviewed to inform future research directions. Full article
Show Figures

Figure 1

Back to TopTop