bims-flamet Biomed News
on Cytokines and immunometabolism in metastasis
Issue of 2025–10–26
25 papers selected by
Peio Azcoaga, Biodonostia HRI



  1. Cell Rep. 2025 Oct 22. pii: S2211-1247(25)01234-3. [Epub ahead of print]44(11): 116463
      Metabolic-immune crosstalk in the tumor microenvironment (TME) is a critical driver of tumorigenesis, progression, and immune evasion. Tumor cells undergo profound metabolic reprogramming, causing nutrient competition, toxic metabolite accumulation, and the formation of cold niches that gradually exhaust effector immune cells. In contrast, immunosuppressive cells exhibit strong metabolic adaptability, reinforcing the suppressive milieu. Moreover, tertiary lymphoid structures provide nutrient- and oxygen-rich "moats" that sustain the functions of B and T cells. In addition, metabolic-immune interactions establish novel checkpoints through an "enzyme-metabolite-receptor" axis, which synergize with PD-1/CTLA-4 pathways to promote resistance to immune checkpoint inhibitors (ICIs). Although monotherapies with metabolic inhibitors have shown limited efficacy, their combination with ICIs is promising. Therefore, this review discusses the field from three perspectives: metabolic stress in the TME, immune cell adaptation, and targeting metabolic immune checkpoints in combination with immunotherapy.
    Keywords:  CP: Cancer; CP: Metabolism
    DOI:  https://doi.org/10.1016/j.celrep.2025.116463
  2. Curr Pharm Biotechnol. 2025 Oct 16.
      A defining characteristic of tumor cells is their preferential reliance on aerobic glycolysis for lactate production, even under oxygen-sufficient conditions - the well-known Warburg effect. Recent advances have revealed lactate to be far more than a metabolic waste product, establishing its role as a versatile signaling molecule with multiple functions in cancer progression. Acting simultaneously as a pro-inflammatory mediator, hypoxia surrogate, tumor burden indicator, and metastasis predictor, lactate exerts profound and wide-ranging effects on immune cell function within the tumor microenvironment (TME). The immunomodulatory properties of lactate create a profoundly immunosuppressive milieu that facilitates tumor immune evasion. It achieves this through coordinated suppression of antitumor immune effectors, including natural killer cells, dendritic cells, and cytotoxic T lymphocytes, while simultaneously enhancing the immunosuppressive functions of regulatory T cells, tumorassociated macrophages, and endothelial cells. This dual mechanism of action promotes tumor progression and metastasis through multiple pathways. The groundbreaking discovery of lysine lactylation (Kla) has further expanded our understanding of lactate's biological roles, revealing a direct molecular connection between tumor metabolism and epigenetic regulation. This review provides a comprehensive synthesis of current knowledge regarding lactate-mediated immune modulation in the TME, examines recent advances in our understanding of lactate-dependent tumor biology, and evaluates emerging therapeutic strategies that target lactate metabolism. By integrating these perspectives, we aim to offer both fundamental insights and practical guidance for the development of novel anticancer therapies that target metabolic-epigenetic crosstalk.
    Keywords:  Cancer therapy.; Immune cells; Immune evasion; Lactate; Tumor microenvironment; lactylation
    DOI:  https://doi.org/10.2174/0113892010415740251006000648
  3. Clin Cancer Res. 2025 Oct 21.
      Chimeric antigen receptor (CAR)-engineered invariant natural killer T cells (CAR-NKTs) are a novel cell platform for cancer immunotherapy. Unlike conventional T cells, NKTs are characterized by innate antitumor properties, minimal alloreactivity, and a unique ability to modulate the tumor microenvironment (TME). This article provides a comprehensive overview of preclinical and early clinical studies evaluating CAR-NKTs in both autologous and allogeneic clinical settings. We discuss the contributions of CAR signaling domains, cytokine co-expression, and other functional measures that correlate with CAR-NKT persistence, function, and metabolic fitness. We also discuss the critical role of immunocompetent animal models in elucidating the interactions of CAR-NKTs with the TME and other components of the immune system. Finally, we review strategies that combine CAR-NKTs with other therapeutic approaches to promote potential synergistic benefits in cancer patients.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-25-0197
  4. World J Gastrointest Oncol. 2025 Oct 15. 17(10): 109398
      Colorectal cancer (CRC) exhibits profound lipid metabolic reprogramming, a hallmark of malignant transformation that supports tumorigenesis, immune evasion, and therapeutic resistance. Dysregulated lipid metabolism in CRC involves altered fatty acid synthesis, uptake, oxidation, and cholesterol metabolism, which collectively drive cancer cell proliferation, metastasis, and interactions with the tumor microenvironment (TME). This review synthesizes current insights into lipid metabolic rewiring in CRC, its role in shaping immunosuppressive TME dynamics, and emerging therapeutic strategies targeting lipid pathways.
    Keywords:  Colorectal cancer; High-fat diet; Lipid metabolism reprogramming; Targeted drugs; Tumor microenvironment
    DOI:  https://doi.org/10.4251/wjgo.v17.i10.109398
  5. Cureus. 2025 Sep;17(9): e92707
      The tumor microenvironment (TME) plays a crucial role in cancer progression, metastasis, immune evasion, and treatment resistance. However, the current literature often studies its components separately. This review offers an integrated view of the dynamic interactions among fibroblasts, immune and vascular cells, the extracellular matrix, cytokines, exosomes, and microbiota within the TME. It discusses classical mechanisms such as epithelial-mesenchymal transition, stromal remodeling, and metabolic rewiring alongside emerging paradigms like microbiome-driven immunomodulation and exosome-mediated therapy resistance. Spatial heterogeneity and the temporal evolution of the tumor niche are examined using recent advances in single-cell and spatial transcriptomics, 3D bioprinting, and patient-derived organoid models. Key findings emphasize the microbiome's influence on immune responses and the role of exosomes in transferring resistance traits and regulating intercellular signaling. By connecting molecular insights with clinical perspectives, the review explores translational strategies targeting the TME, including checkpoint inhibitors, stromal modulators, anti-angiogenic agents, and engineered CAR-T therapies. This comprehensive view highlights the importance of considering cancer as a complex, evolving ecosystem rather than just a cell-autonomous disease and provides a foundational framework for precision oncology approaches aimed at disrupting harmful TME interactions to improve therapeutic effectiveness and patient outcomes.
    Keywords:  cancer progression; immune evasion; precision oncology; stromal remodeling; tumor microenvironment
    DOI:  https://doi.org/10.7759/cureus.92707
  6. J Neuropathol Exp Neurol. 2025 Oct 21. pii: nlaf119. [Epub ahead of print]
      Tumor-associated macrophages (TAMs) are the predominant immune cells in the tumor microenvironment (TME) of human meningiomas. They exhibit different functional phenotypes that contribute either to an anti-inflammatory and anti-tumor response (M1 phenotype), or an immunosuppressive and pro-tumor response (M2 phenotype). In meningiomas of cats, the specific role of TAMs in determining prognosis and post-surgery outcome remains unclear. This study aimed to characterize the macrophage population in feline meningiomas, differentiate M1 from M2 phenotype, and investigate the relationship of the results to prognosis. Fifty-seven surgically removed feline meningiomas were studied. Immunolabeling was performed on formalin-fixed paraffin embedded samples with primary antibodies anti-MAC387 (M1 macrophage), CD163 and CD204 (M2 macrophage), Iba1 (pan-macrophage), and Ki67. The cells in all cases expressed Iba1 and CD163, while MAC387 and CD204-expression varied. Moreover, macrophages were more numerous in high-grade tumors across both M1 and M2 phenotypes. Although none of the markers were predictive of recurrence on Cox models (P > .05), based on the significant association between M2 CD163-positive macrophages and high-grade tumors, and their shorter post-surgery recurrence-free times, the results suggest that M2 macrophages might play a role in the behavior and prognosis of feline meningiomas.
    Keywords:  M1 phenotype; M2 phenotype; cat; immune cells; macrophage; meningioma; prognosis
    DOI:  https://doi.org/10.1093/jnen/nlaf119
  7. Front Immunol. 2025 ;16 1706172
      
    Keywords:  biosensor; cancer immunotherapy; immune cell tracking; monitoring immune responses; monitoring tumor; theranostics; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1706172
  8. Front Immunol. 2025 ;16 1662238
      Adoptive T cell therapy has transformed cancer treatment, with chimeric antigen receptor (CAR) T cell therapy demonstrating remarkable clinical success in hematological malignancies. By genetically engineering a patient's own T cells to recognize and attack cancer cells, CAR T therapy has achieved durable remissions in several blood cancers. However, its efficacy in solid tumors remains limited, largely due to the immunosuppressive tumor microenvironment (TME), which impairs T cell infiltration, persistence, and function. To address these challenges, innovative strategies are being developed to reprogram T cell signaling within the hostile TME. One promising class involves chimeric non-antigen receptors (CNARs), which modulate T cell activity independently of direct antigen recognition. Among these, chimeric switch receptors (CSRs) convert inhibitory checkpoint signals into activating cues, while inverted cytokine receptors (ICRs) redirect suppressive cytokine signals to promote T cell activation. In this review, we provide a focused overview of the design principles, mechanistic functions, and therapeutic potentials of CSRs and ICRs as adjuncts to CAR T therapy in solid tumors. We also discuss key considerations regarding safety, specificity, and clinical translation to inform future advancements in engineered receptor strategies for cancer immunotherapy.
    Keywords:  CAR-T; chimeric switch receptors; immune suppression; immunotherapy; inverted cytokine receptors; solid cancers; synthetic biology
    DOI:  https://doi.org/10.3389/fimmu.2025.1662238
  9. Biochim Biophys Acta Rev Cancer. 2025 Oct 19. pii: S0304-419X(25)00223-9. [Epub ahead of print] 189481
      Raf-1 Kinase Inhibitor Protein (RKIP) is a well-known metastasis suppressor that regulates key oncogenic signaling pathways. Emerging evidence also points to a dual role for RKIP as an immunomodulatory molecule, influencing the tumor immune microenvironment, a complex niche that promotes immune evasion and overall tumor progression. This review explores RKIP's impact on immune surveillance via its interactions with signaling pathways and cellular components of the tumor microenvironment, as well as its role in recruiting and polarizing immune cells with different dynamics in reshaping an immunocompetent milieu. We further discuss how RKIP downregulation promotes immune evasion, alters cytokine profiles, and reduces effector immune cell infiltration. Current studies suggest that RKIP supports type I interferon signaling and antigen presentation, while its loss contributes to an immunosuppressive, pro-metastatic environment. Therefore, by regulating the expression of RKIP in the pathways that control TME architecture, the TME can be reprogramed towards an immunoprotected milieu. Altogether, this review underscores RKIP's role in tumor immunity, offering new perspectives for therapeutic strategies aimed at overcoming cancer immune evasion and improving immunotherapy efficacy.
    Keywords:  Cancer; Immune evasion; RKIP; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.bbcan.2025.189481
  10. Cancer Cell Int. 2025 Oct 24. 25(1): 372
      Breast cancer (BC) development is significantly affected by the tumor microenvironment (TME), in which macrophage polarization plays a central role. Macrophages can assume pro-inflammatory M1 or anti-inflammatory M2 phenotypes, with M1 macrophages suppressing and M2 macrophages facilitating tumor growth. Accumulating evidence indicates long non-coding RNAs (lncRNAs) as significant regulators of macrophage polarization in BC. LncRNAs can promote either M1 or M2 phenotypes, hence modulating tumor growth and metastasis. Accordingly, this review aims to shed light on the mechanisms by which lncRNAs regulate macrophage polarization and to explore their impact on BC development, offering insights into potential therapeutic strategies targeting the tumor immune microenvironment.
    Keywords:  Breast cancer; LncRNAs; Long non-coding RNAs; M1 macrophage; M2 macrophage; Macrophage polarization
    DOI:  https://doi.org/10.1186/s12935-025-04017-0
  11. Breast Cancer. 2025 Oct 22.
      Breast cancer (BC) is among the most prevalent and immunogenic malignancies in women, characterized by rapid proliferation and significant immune cell infiltration into the tumor microenvironment (TME). The programmed cell death protein 1 (PD-1) and its ligand PD-L1 form a critical immune checkpoint axis exploited by tumors to evade immune detection. Targeting this pathway with immune checkpoint inhibitors (ICIs) has shown clinical promise, particularly in triple-negative breast cancer (TNBC). The IMpassion130 trial demonstrated that atezolizumab plus nab-paclitaxel extended progression-free survival (PFS) to 7.5 months compared to 5.0 months with chemotherapy alone. Similarly, the KEYNOTE-522 trial reported a 64.8% pathological complete response (pCR) rate with pembrolizumab-chemotherapy versus 51.2% in the control group. This review summarizes the PD-1/PD-L1 pathway and highlights the therapeutic impact, clinical advances, and future potential of ICIs in BC treatment.
    Keywords:  Breast cancer; Immune checkpoint inhibitor (ICI); Programmed cell death 1 (PD-1); Programmed cell death ligand1 (PD-L1); Triple-negative breast cancer (TNBC)
    DOI:  https://doi.org/10.1007/s12282-025-01793-5
  12. Cell Oncol (Dordr). 2025 Oct 23.
      
    Keywords:  Cancer-associated fibroblasts; Spatial transcriptomics; Tumor microenvironment
    DOI:  https://doi.org/10.1007/s13402-025-01108-y
  13. Front Immunol. 2025 ;16 1693336
      N6-methyladenosine (m6A) is the most prevalent internal modification of eukaryotic mRNA and has emerged as a pivotal regulator of gene expression at the post-transcriptional level. In the tumor immune microenvironment, tumor-associated macrophages (TAMs) represent a highly plastic and heterogeneous population that profoundly influences cancer progression, immune evasion, and therapeutic response. Recent studies have uncovered that m6A modification, mediated by dynamic "writers," "erasers," and "readers," exerts critical regulatory effects on TAM differentiation, polarization, and functional reprogramming. By modulating the stability, translation, and decay of transcripts involved in inflammatory signaling, metabolic adaptation, and immune checkpoints, m6A shapes the balance between tumor-promoting (M2-like) and tumor-suppressive (M1-like) macrophage phenotypes. Moreover, dysregulation of m6A machinery in TAMs has been linked to the suppression of anti-tumor immunity and resistance to immunotherapy, highlighting its translational potential as a therapeutic target. This review summarizes current advances in understanding the roles and mechanisms of m6A modification in TAM biology, discusses its implications in tumor immunity, and outlines the challenges and opportunities of targeting the m6A-TAM axis for cancer treatment.
    Keywords:  YTHDF2; cancer immunotherapy; m6A RNA modification; tumor microenvironment; tumor-associated macrophages
    DOI:  https://doi.org/10.3389/fimmu.2025.1693336
  14. Cancer Lett. 2025 Oct 22. pii: S0304-3835(25)00669-X. [Epub ahead of print] 218097
      Cancer dormancy refers to a reversible state where cancer cells enter a quiescent phase, allowing them to evade therapeutic interventions and remain undetected. This state can lead to potential reactivation years later, resulting in relapse and metastasis. This phenomenon presents a significant challenge in cancer treatment, as dormant cells often exhibit resistance to conventional therapies. Recent studies emphasize the crucial role of metabolic reprogramming in regulating cancer dormancy, closely interacting with the tumor microenvironment. Dormant cancer cells undergo metabolic adaptations that enable their survival in a hostile tumor microenvironment. These adaptations include a decreased reliance on glycolysis and an increased dependence on oxidative phosphorylation and fatty acid oxidation. Exosomes, extracellular matrix, and cancer-associated fibroblasts dynamically regulate these metabolic states by mediating intercellular communication and modulating the biochemical and mechanical properties of the tumor microenvironment. In parallel, epigenetic regulation fine-tunes metabolic gene expression, reinforcing the dormant phenotype and enabling plastic transitions between dormancy and proliferation. Additionally, these cells utilize autophagy to recover nutrients and manage microenvironmental stress. These metabolic changes help dormant cells maintain a low metabolic state while preserving their ability to reactivate when conditions become favorable. Understanding the relationship between dormancy and metabolism offers new therapeutic opportunities aimed at targeting metabolic pathways to prevent relapse and metastasis. This review explores the mechanisms of metabolic reprogramming in dormancy induction, maintenance, and escape, providing insights into potential therapeutic strategies.
    Keywords:  Autophagy; Cancer dormancy; Fatty acid oxidation; Metabolic reprogramming; Oxidative phosphorylation; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.canlet.2025.218097
  15. Expert Rev Anticancer Ther. 2025 Oct 22.
       INTRODUCTION: The tumor microbiome, a diverse microbial community within the tumor microenvironment (TME), significantly influences cancer progression and immunotherapy outcomes in colorectal cancer (CRC). Understanding its role in modulating immune responses and therapeutic resistance is critical for advancing precision oncology.
    AREAS COVERED: This review examines the tumor microbiome's impact on CRC immunotherapy, focusing on immune checkpoint inhibitors (ICIs) like anti-PD-1/PD-L1 and anti-CTLA-4. It explores microbial composition, their immune-modulatory mechanisms, and metabolite-driven resistance pathways, including short-chain fatty acids and polyamines. Emerging strategies such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and targeted antibiotics are discussed, alongside challenges in personalizing microbiome-based therapies. Literature was sourced from peer-reviewed studies on tumor microbiome dynamics and immunotherapy resistance.
    EXPERT OPINION: The tumor microbiome shapes CRC immunotherapy efficacy by modulating immune evasion and TME dynamics. Targeted interventions like FMT and probiotics show promise in enhancing ICI responses, but challenges include microbial variability, safety concerns, and ethical considerations. Future research should prioritize personalized microbiome profiling and standardized protocols to optimize therapeutic outcomes and overcome resistance in CRC.
    Keywords:  Immune Checkpoint Inhibitors; Immunotherapy Resistance; Microbial Metabolites; Microbiome-Targeted Therapies; Tumor Microbiome
    DOI:  https://doi.org/10.1080/14737140.2025.2579656
  16. J Immunother Cancer. 2025 Oct 23. pii: e011768. [Epub ahead of print]13(10):
       BACKGROUND: Chimeric antigen receptor (CAR) T cells targeting CD19 induce durable responses in B-cell acute lymphoblastic leukemia (B-ALL). However, the contribution of the tumor microenvironment to the therapeutic response after CAR T-cell treatment remains incompletely understood.
    METHODS: We performed single-cell RNA sequencing and spectral flow cytometry-based analyses of bone marrow-resident immune cells from B-ALL patients before and after CAR T-cell treatment.
    RESULTS: We observed profound changes in the microenvironment in response to CAR T-cell-mediated inflammation, including an increase in myeloid cells. Significant induction of the interferon response, hypoxia, and TGF-β signaling was accompanied by expansion of myeloid-derived suppressor cells (MDSCs) and endogenous exhausted CD8+ T cells. PD-1 expression in endogenous T cells post-treatment was associated with a lack of durable response in the cohort of patients analyzed. Further, we revealed that HIF-1α, VEGF, and TGFBR2 are key players in the intercellular communication between CAR T cells and the immune niche, potentially driving widespread T-cell dysfunction. Infusion of anti-CD19 CAR T cells led to increased accumulation of human MDSCs, exacerbation of a hypoxic environment and T-cell exhaustion in hematopoietic stem/progenitor cell-humanized mice bearing a human tumor.
    CONCLUSIONS: In conclusion, CAR T-cell-mediated myeloid activation is associated with pathways of immune dysregulation that may antagonize the effects of therapy.
    Keywords:  Adoptive cell therapy - ACT; Chimeric antigen receptor - CAR; Leukemia; Tumor microenvironment - TME
    DOI:  https://doi.org/10.1136/jitc-2025-011768
  17. Biochem Cell Biol. 2025 Jan 01. 103 1-11
       BACKGROUND: In eukaryotic cells, phospholipid asymmetry is actively maintained, with phosphatidylserine (PS) typically confined to the inner leaflet of the plasma membrane (PM), due to the active performance of the PS flippase ATP11/CDC50A complex. However, in the tumor microenvironment (TME), PS exposure on the outer leaflet occurs from multiple sources, including apoptotic tumor cells, necrotic tissue, viable endothelial cells, and tumor-derived exosomes. Especially, in apoptotic cells, the PS scamblase Xkr8 mediates PS externalization. This exposed PS plays a crucial role in immune suppression within the TME. PS binds to receptors on phagocytes, primarily macrophages and dendritic cells (DCs), triggering efferocytosis (the engulfment of PS-positive cells, usually apoptotic cells) and promoting anti-inflammatory responses.
    METHOD: To understand the immune suppressive role of PS exposure on tumor cells, we deleted CDC50A, the PS flippase, from the tumor. Thus, in this tumor, PS is constantly on the outer leaflet of PM, called PSout tumor model. On the contrary, we knocked out Xkr8 from tumor cells. Thus, even tumor cells undergo apoptosis, PS can still stay in the inner leaflet of PM, called PSin tumor model. Taking the advantage of the PSin and PSout model, we could investigate the anti-tumor immune responses of PS externalization in TME.
    RESULTS: Using PSout model, we found that these PSout tumors exhibited enhanced growth, M2-polarized tumor-associated macrophages (TAMs), and reduced tumor-antigen-specific T cell infiltration. In TME, the PS receptor TIM-3 on TAMs was responsible for PS sensing. Using PSin model, we found that the PSin tumors exhibited increased anti-tumor immunity, featuring suppressed tumor progress, TAM M1 polarization, suppressed IL-10 secretion, and enhanced natural killer (NK) cell cytotoxicity. Thus, blocking PS externalization via targeting Xkr8 could serve as a promising strategy for anti-tumor immunotherapy.
    THERAPEUTIC APPLICATIONS: However, there is no available Xkr8 inhibitor or direct anti-PS blocking antibody for therapeutic use. Thus, we developed our unique "PS all-block" strategy leveraging an engineered protein which binds only to PS, without sending signals to immune receptors, functioning as a dominant negative. The "PS all-block" can neutralize PS molecules from all sources without triggering downstream immune suppression pathways. Our data suggested that the "PS all-block" was a more effective antitumor immunotherapy compared to our successfully developed Xkr8 inhibition, as Xkr8 targeting only neutralized apoptotic PS, while the "PS all-block" approach could neutralize PS from all sources.
    DOI:  https://doi.org/10.1139/bcb-2025-0320
  18. Mol Ther Oncol. 2025 Dec 18. 33(4): 201061
      Although adoptive natural killer cell therapies have been safe and somewhat effective in hematological malignancies, their performance in solid tumors is hindered by the solid tumor microenvironment, which impairs natural killer cell trafficking and function. Strategies that boost natural killer cell infiltration and maintain their activity in the tumor microenvironment have demonstrated enhanced therapeutic benefits. Multiple strategies have been explored to enhance natural killer cell infiltration and sustain their activity within the tumor microenvironment. These encompass tactics like equipping natural killer cells with chemokine receptors or inducing tumor cells to secrete chemokines to enhance homing, arming natural killer cells with chimeric antigen receptors or concomitant use of natural killer cell engagers to direct natural killer cells to tumors, and blocking immunosuppressive factors such as transforming growth factor-β and immune checkpoints or stimulation by cytokines to make the tumor microenvironment more permissive to natural killer cell function, among others. Here we summarize recent advances in the strategies to augment tumor infiltration of active natural killer cells, aiming to improve natural killer cell-based immunotherapies against pediatric and adult solid tumors.
    Keywords:  MT: Special Issue - Advancements in pediatric cancer therapy; anti-TGF-β therapy; chemokine; chemokine receptor; chimeric antigen receptor; cytokine stimulation; immune checkpoint inhibitors; natural killer cell engagers; natural killer cells; solid tumors; tumor microenvironment
    DOI:  https://doi.org/10.1016/j.omton.2025.201061
  19. Front Immunol. 2025 ;16 1679271
      Macrophages, the most abundant immune cells in many solid tumors, are no longer viewed solely as accomplices of cancer but as powerful therapeutic allies. This review charts the rapid rise of macrophage-based immunotherapies, from CD47/SIRPα checkpoint blockade and CAR-macrophages to macrophage-drug conjugates (MDCs). We emphasize emerging frontiers - RNA-based reprogramming, epigenetic modulation, small activating RNA and circRNA approaches, and macrophage-derived extracellular vesicles - that are redefining how tumor-associated macrophages can be targeted or harnessed. Distinct from earlier TAM reviews, we integrate outcomes from ongoing and completed clinical trials, highlight therapeutic platforms beyond classical depletion and polarization, and frame macrophages not only as targets but also as delivery vehicles. By spotlighting both innovative strategies and the challenges of moving them into the clinic, we aim to provide a forward-looking guide for researchers and clinicians shaping the next generation of cancer immunotherapy.
    Keywords:  cancer; cell therapy; immunotherapy; macrophages; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1679271
  20. Adv Sci (Weinh). 2025 Oct 20. e06995
      Tumor-associated macrophages (TAMs) predominantly exert functions that facilitate tumor progression. Triggering receptor expressed on myeloid cell 2 (TREM2) is expressed in TAMs, playing a crucial role in mediating the immunosuppressive function of TAMs. The mechanisms by which TREM2+ TAMs promote tumor growth and inhibit anti-tumor immunity remain unclear. Through single-cell sequencing of tumor tissues derived from wild-type and Trem2 knockout mice bearing subcutaneous lung cancer, it is found that TREM2 deletion hindered tumor growth, with a notable increase in and improved functionality of CD4+ T and natural killer (NK) cells in the tumor microenvironment. TREM2 deficiency led to ATP-binding cassette transporter A1 (ABCA1) downregulation, causing cholesterol accumulation in TAMs and promoting a pro-inflammatory phenotype. This results in increased chemokine (C-X3-C motif) ligand 1 (CX3CL1) secretion of macrophages, recruiting more CD4+ T and NK cells to the tumor site, enhancing the anti-tumor response. After screening food and drug administration (FDA)-approved drugs, bortezomib and ataluren are found to effectively inhibit TREM2 expression in TAMs, indicating a potential therapeutic strategy against TREM2. This study elucidates the mechanism by which TREM2 shapes the immunosuppressive microenvironment and promotes tumorigenesis, highlighting TREM2 as a target for cancer immunotherapy.
    Keywords:  CX3CL1; TREM2; anti‐tumor immunity; cholesterol; tumor microenvironment; tumor‐associated macrophages
    DOI:  https://doi.org/10.1002/advs.202506995
  21. Front Oncol. 2025 ;15 1675537
      Resistance to programmed cell death is a defining hallmark of cancer and a persistent barrier to successful therapy. Dual-function proteins such as p53, Ras, HIF-1α, BNIP3, and NF-κB act as molecular switches that determine cell fate between apoptosis and survival. In tumors, these proteins are deregulated not only by intrinsic mutations but also by extrinsic signals from the tumor microenvironment (TME). This Mini Review critically analyzes previous therapeutic approaches, emphasizing overlooked mechanisms such as Ras-mediated suppression of p53. It proposes a sequential therapeutic strategy: first, dismantling TME adaptations (hypoxia, inflammation, protective autophagy); second, inhibiting oncogenic Ras signaling; and third, restoring p53 activity. The phased approach integrates biomarker-guided patient stratification, recognizes tumor-microenvironment co-evolution, and highlights how resistance evolves over time. Although the concept does not resolve all challenges, it outlines a rational framework for restoring apoptotic competence and provides a pathway for translational and clinical testing.
    Keywords:  BNIP3; HIF-1α; NF-κB; Ras; apoptosis; p53; sequencing; tumor microenvironment
    DOI:  https://doi.org/10.3389/fonc.2025.1675537
  22. Front Immunol. 2025 ;16 1682322
      Neuroimmuno-oncology is an emerging interdisciplinary field that explores the complex interactions among the nervous system, the immune system, and tumor cells within the tumor microenvironment (TME). Recent studies have underscored the critical role of neurons in gliomas, where synaptic signaling and the release of neurotrophic factors contribute not only to tumor progression but also to mechanisms of immune evasion. Neurotransmitters such as glutamate and gamma-aminobutyric acid (GABA), along with neuron-derived factors including brain-derived neurotrophic factor (BDNF) and neuroligin-3 (NLGN3), have been shown to modulate immune cell function and promote the formation of an immunosuppressive TME. In particular, neuronal electrical activity mediated through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) signaling facilitates immune escape in glioma cells, leading to the development of an "immune-excluded" phenotype that compromises the efficacy of immunotherapy. Therapeutic strategies that combine AMPAR antagonists with immune checkpoint inhibitors-alongside neuromodulatory techniques such as repetitive transcranial magnetic stimulation (rTMS) or deep brain stimulation (DBS)-hold potential to reprogram the neuro-immune-tumor axis, remodel the immune landscape, and improve immunotherapy responses in central nervous system malignancies. Advancing our understanding of how neuronal activity regulates the glioma immune microenvironment may open new avenues for precision-targeted therapeutic approaches in neuro-oncology.
    Keywords:  AMPA receptor; glioma; immune suppression; immunotherapy; neuronal activity; neuro–immune–tumor axis; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1682322
  23. Phys Life Rev. 2025 Oct 12. pii: S1571-0645(25)00152-6. [Epub ahead of print]55 142-209
      Protein post-translational modifications (PTMs), which involve the covalent attachment of specific chemical groups to amino acid residues, significantly reshape protein structure and function. These modifications play a crucial role in fundamental physiological processes such as signal transduction, metabolic regulation, and protein homeostasis. In the context of pan-cancer, various types of PTMs, including phosphorylation, acetylation, glycosylation, and ubiquitination, create an intricate crosstalk network that finely tunes the stability and function of immune checkpoint molecules, directly influencing tumor immune evasion and immune cell recognition. Additionally, PTMs exert multilayered regulation over the functional states of key immune cells, such as T cells, macrophages, and dendritic cells (DCs), thereby determining the intensity and nature of immune responses within the tumor microenvironment (TME). Furthermore, PTMs are pivotal in antigen processing and presentation by influencing antigen diversity and epitope display, which facilitates tumor cell escape from immune surveillance. Dynamic analyses reveal that PTM landscapes exhibit spatiotemporal specificity during tumor initiation, progression, and metastasis, closely correlating with tumor stage and the establishment of an immunosuppressive microenvironment. Based on these findings, immunotherapeutic strategies targeting key PTM-modifying enzymes, such as kinases, deacetylases, and deubiquitinases, are rapidly emerging. However, these approaches still face challenges, including drug specificity, resistance, and off-target effects. The exploration of synergistic effects through the combinational targeting of distinct PTM pathways, along with a deeper understanding of the interactive regulatory networks among PTMs, opens promising avenues for the development of next-generation precision immunotherapies. This review aims to systematically elucidate the multifaceted roles and dynamic regulation of PTMs in tumor immunity, providing a theoretical foundation and research direction for identifying novel immunotherapeutic targets and optimizing therapeutic strategies.
    Keywords:  Antigen presentation; Crosstalk mechanisms; Dynamic regulation; Immune checkpoints; Immune evasion; Pan-cancer perspective; Post-translational modifications (PTMs); Tumor immunity
    DOI:  https://doi.org/10.1016/j.plrev.2025.10.001
  24. Acta Biochim Biophys Sin (Shanghai). 2025 Oct 23.
      The T cell receptor (TCR) initiates signaling by specifically recognizing peptide-MHC complexes, triggering the phosphorylation of CD3 chain immunoreceptor tyrosine-based activation motifs (ITAMs). This recruits kinases such as ZAP70, triggering a tightly regulated signaling cascade that governs T cell activation, differentiation, and effector functions. In contrast, the chimeric antigen receptor (CAR) is a synthetic construct that bypasses MHC restriction by fusing an antigen-binding domain with intracellular signaling modules (usually CD3ζ and co-stimulatory domains) from the TCR complex and other receptors. CAR-T cell therapy has revolutionized the treatment of hematologic malignancies, resulting in durable remission of B-cell leukemia, lymphoma, and multiple myeloma. However, its efficacy in solid tumors is limited by intrinsic barriers: poor CAR-T-cell trafficking/infiltration into tumors, the immunosuppressive tumor microenvironment (TME), intratumoral metabolic competition, and tumor antigen heterogeneity/loss. To improve CAR-T-cell function in solid tumors, numerous studies have explored multiple strategies: engineering CARs to boost immune synapse formation via optimized receptor clustering, increasing the ITAM number/strength to amplify downstream signaling, and incorporating novel/multiple co-stimulatory domains to sustain T-cell activation and persistence. Additionally, approaches include the use of CAR-T cells that secrete pro-inflammatory cytokines, epigenetic reprogramming to preserve T-cell stemness and functionality, and the use of synthetic biology tools for tunable/logic-gated CAR activation. Here, we summarize the current understanding of CAR signaling dynamics and highlight recent breakthrough strategies designed to overcome these challenges in solid tumors. These advances narrow the liquid-solid tumor efficacy gap, holding promise for better clinical outcomes in patients with solid malignancies and a new era of personalized immunotherapy.
    Keywords:  CAR-T; TCR; immunotherapy; solid tumor
    DOI:  https://doi.org/10.3724/abbs.2025190
  25. Drug Resist Updat. 2025 Oct 13. pii: S1368-7646(25)00118-9. [Epub ahead of print]84 101315
      Breast cancer stem cells (BCSCs) are recognized as a critical subpopulation involved in cancer recurrence and metastasis, as they are capable of self-renewal and differentiate into various cell types. BCSCs play significant roles in tumor progression, being regulated by key signaling pathways such as Notch, PI3K/AKT/mTOR, and Hedgehog, and their interactions with the tumor microenvironment, which affect tumor growth and resistance to therapeutics. This review focuses on the surface markers of BCSCs, their roles in recurrence and metastasis, and the key signaling pathways. It also discusses the recent progress in understanding how BCSCs contribute to drug resistance and explores potential therapeutic strategies targeting these cells and their microenvironment to improve clinical outcomes and prevent relapse.
    Keywords:  Breast cancer; Cancer stem cells; Drug resistance; Tumor metastasim
    DOI:  https://doi.org/10.1016/j.drup.2025.101315