bims-flamet Biomed News
on Cytokines and immunometabolism in metastasis
Issue of 2025–11–09
thirty-six papers selected by
Peio Azcoaga, Biodonostia HRI



  1. Mol Biol Rep. 2025 Nov 04. 53(1): 41
      Breast cancer is the leading cancer type that is reported in women worldwide, with its 2.3 million annual incidence and 685,000-related deaths (WHO, 2023). Although genetic mutations have proven to be major driving agents of tumorigenesis, the tumor microenvironment (TME) is increasingly being considered as the definitive indicator of disease progression, metastasis, immune evasion and therapeutic resistance. It is an active, diverse cellular webwork of substances cancer-related fibroblasts, (CAFs), tumor-related macrophages, (TAMs), regulatory T cells, (Tregs), myeloid-derived suppressor cells (MDSCs), and cancer-related adipocytes; (CAAs). These cellular processes interact reciprocally with not only malignant cells through paracrine communication but also through the composition of the extracellular matrix, (ECM). Recent research indicates that CAFs produce pro-invasive factors i.e. TGF-B and HGF whereas TAMs tend to be M2-like and secret VEGF and IL-10 to encourage angiogenesis and immune subversion. Cytokines, chemokines, ECM proteins, and tumor-derived EVs are non-cellular factors serving to alter cellular phenotypes and facilitate construction of metastatic niche. Besides oncogenic microRNAs (e.g., miR-21, miR-10b) that is released in exosome, hypoxia-mediated activation of HIF-1a induces both neovascularization and metabolic reprogramming. The repolarization of TAM (e.g. CSF1R inhibitors) and depletion of CAF and inhibition of EV pathway are also being explored as therapeutic mechanisms against TME resistance and are in preclinical and clinical development. In particular, the combination procedures with immunomodulation and TME disruption have demonstrated some success in conquering resistance in TNBC models. Therefore, it is paramount to understand how the surrounding tissue, the immune system, the immune system, and other elements behave over time and space as cofounding partners of tumors, in a way that would create better precision and clinical outcomes in breast cancer patients.
    Keywords:  Breast cancer; Epithelial- to- mesenchymal transition (EMT); Extracellular vesicles (EVs); Metastasis; Targeted therapy; Tumor microenvironment; Tumor-associated macrophages (TAMs)
    DOI:  https://doi.org/10.1007/s11033-025-11231-6
  2. Crit Rev Oncol Hematol. 2025 Nov 05. pii: S1040-8428(25)00393-2. [Epub ahead of print] 105005
      The B-cell lymphoma 2 (BCL-2) family of proteins plays a central role in regulating apoptosis and has emerged as a key therapeutic target in oncology since the FDA approval of venetoclax in 2016. Within the tumor microenvironment (TME), intricate cellular and molecular interactions alter apoptotic signaling by upregulating anti-apoptotic BCL-2 members, thereby promoting tumor cell survival, immune evasion, and therapy resistance. This review critically examines how anti-apoptotic BCL-2 proteins sustain a pro-tumor TME by supporting immunosuppressive populations such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), while suppressing tumor-infiltrating lymphocyte activity and driving metabolic reprogramming that enhances cancer cell fitness. Furthermore, biophysical and biochemical stressors in the TME-including hypoxia, angiogenesis, oxidative imbalance, and osmotic stress-further modulate BCL-2 expression, reinforcing tumor progression and treatment resistance. Recent advances demonstrate that pharmacological inhibition or genetic modulation of BCL-2 family members-particularly targeting BCL-2, BCL-XL, or MCL-1, or activating pro-apoptotic mediators such as NOXA-can reprogram the TME from an immunosuppressive ("cold") to an immune-responsive ("hot") phenotype. This transition enhances anti-tumor immunity, increases cytotoxic infiltration, and potentiates the efficacy of immunotherapeutic approaches, including PD-L1 blockade. Overall, this review provides an integrated perspective on how BCL-2 family signaling dynamically interacts with the TME to sustain malignancy and highlights the therapeutic potential of BCL-2 inhibition in reshaping the immune landscape and overcoming resistance across cancer types.
    Keywords:  BCL-2; BCL-XL; BH3 mimetics; MCL-1; apoptosis; hypoxia; tumor microenvironment; venetoclax
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.105005
  3. Cancer Manag Res. 2025 ;17 2567-2587
      Myeloid-derived suppressor cells (MDSCs) arise from myeloid progenitors in the bone marrow and, under the influence of tumor- and immune-cell-derived cytokines, chemokines, and growth factors, enhance immunosuppressive activity within the tumor microenvironment (TME). Noncoding RNAs (ncRNAs)-including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs)-have emerged as critical regulators of MDSCs biology. Recent evidence has shown that ncRNAs are intimately involved in MDSCs recruitment, differentiation, and suppressive function by modulating key signaling pathways, including STAT3, NF-κB, and PI3K/AKT. Mechanistically, ncRNAs act through epigenetic control (eg, histone modifications and chromatin remodeling), post-transcriptional regulation (eg, miRNA sponging), and fine-tuning of gene networks. These insights highlight RNA-based strategies that target ncRNAs to disrupt MDSCs-mediated immune suppression and potentiate antitumor immunity, while acknowledging ongoing challenges such as delivery specificity, stability, and off-target effects. This review synthesizes current understanding of how ncRNAs regulate MDSCs via major signaling axes and discusses implications for cancer progression and therapeutic development.
    Keywords:  MDSCs; ncRNAs; tumor; tumor immunity; tumor microenvironment
    DOI:  https://doi.org/10.2147/CMAR.S550896
  4. Front Immunol. 2025 ;16 1688342
      The tumor microenvironment (TME) comprises non-cancerous cells, extracellular matrix, and signaling molecules that interact with tumor cells. These dynamic interactions critically influence tumor development, progression, metastasis, and treatment response. Cancer-associated adipocytes (CAAs), as a main component of the tumor-adipose microenvironment (TAME), have various functions, including remodeling the extracellular matrix and interacting with tumor cells or infiltrated leukocytes through a variety of mutual signals. Dysfunctional adipocytes can release different metabolic substrates, adipokines and cytokines to affect the activity and function of immune cells in TME, especially T cells, thus promoting the proliferation, progression, invasion and migration of cancer cells. In this review, we summarize the effects of secretions of adipocytes on the activity and function of different types of T cells in TME, and discuss the possible targets of adipocytes in cancer therapy to provide new ideas for anti-cancer therapy by targeting adipocytes.
    Keywords:  T cell; cancer-associated adipocyte; immunity; inflammatory factors; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1688342
  5. Front Immunol. 2025 ;16 1690068
      Lactylation, a recently discovered post-translational modification (PTM), plays a critical role in cancer biology. Warburg effect induces lactate accumulation, which serves as a metabolic end-product and intercellular signaling mediator within the tumor microenvironment (TME). Beyond fueling tumor growth, elevated lactate levels drive histone and non-histone lactylation, which modulates gene expression and protein function. This epigenetic reprogramming induces immunosuppressive phenotypes in immune cells that are resident in the tumor microenvironment, including impaired effector function, enhanced immunosuppressive cytokine secretion, and altered tumor antigen presentation, collectively facilitating immune escape. This review provides a synthesis of the current understanding of lactate and lactylation in tumor immunosuppression, detailing molecular mechanisms underlying immune cell inhibition (tumor-associated macrophages, T cells, T-reg cells, NK cells and NKT cells, as well as neutrophils) and evaluating emerging therapeutic strategies (e.g., inhibitors of MCTs/LDHA, site-specific antibodies, genetic code expansion technology). We aimed to accelerate the clinical translation of lactylation-targeted anticancer therapies by highlighting recent advances.
    Keywords:  antitumor immunity; histone and non-histone lactylation; immunosuppressive phenotypes; lactate accumulation; lactylation; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1690068
  6. J Inflamm Res. 2025 ;18 15257-15280
      Tumor-associated macrophages (TAMs) are a heterogeneous population of immune cells that play a pivotal role in the tumor microenvironment (TME) of lung cancer. TAMs, which include both monocyte-derived macrophages (MDMs) and tissue-resident macrophages (TRMs), exhibit distinct functions that influence tumor progression, metastasis, and response to therapy. Recent studies have highlighted the spatiotemporal heterogeneity of TAMs, with MDMs primarily promoting tumor growth and immune suppression, while TRMs contribute to tissue homeostasis but can be reprogrammed to support tumor progression. Both subtypes contribute to the formation of an immunosuppressive TME, facilitate tumor metastasis through matrix remodeling, and contribute to therapeutic resistance by modulating the efficacy of chemotherapy, radiation therapy, and immunotherapy. Understanding the specific roles and heterogeneity of MDMs and TRMs as components of TAMs in lung cancer opens avenues for targeted therapies, such as inhibiting their recruitment, reprogramming their polarization, or blocking their pro-tumorigenic functions. This review synthesizes current knowledge on TAMs in lung cancer, highlighting their dual roles and the potential for developing novel therapeutic strategies that target these macrophages to improve patient outcomes.
    Keywords:  immunotherapy; macrophage polarization; therapeutic resistance; tumor microenvironment
    DOI:  https://doi.org/10.2147/JIR.S552449
  7. Adv Clin Exp Med. 2025 Nov 05.
      Today, it is well established that the tumor microenvironment (TME), the tumor niche, along with melanoma cells, plays a crucial role in cancer dissemination and influences the effectiveness of anticancer therapies. Therefore, it may serve as a potential therapeutic target in melanoma treatment. In our research, we focused on the effects exerted by cells within the melanoma microenvironment on cancer progression and the development of therapy resistance. Specifically, we examined stromal cells accompanying melanoma cells in the tumor - cancer-associated fibroblasts (CAFs), cancer-associated keratinocytes (CAKs), and cancer-associated adipocytes (CAAs). Particular attention was given to keratinocytes, as their role in the melanoma microenvironment remains the least understood.
    Keywords:  drug resistance; melanoma; microenvironment; signaling pathways; stromal cells
    DOI:  https://doi.org/10.17219/acem/211897
  8. Front Immunol. 2025 ;16 1689714
      Innate immune cells and pathways are central to shaping the tumor microenvironment (TME), where they influence tumor growth, metastasis, and responsiveness to immunotherapy. Although research on innate immunity in cancer has expanded considerably, the mechanisms driving immune dysfunction remain incompletely understood. This review summarizes current knowledge on the functional states of innate immune cells within the TME and highlights how metabolic reprogramming contributes to immune suppression and tumor progression. We further discuss recent advances in therapeutic strategies targeting innate immune pathways, emphasizing their translational potential. Importantly, we also examine unresolved controversies and knowledge gaps across innate immune cells, metabolic networks, and innate immune factors such as complement and cytokines, outlining key challenges for clinical translation. By linking mechanistic insights with emerging interventions and identifying future directions, this review provides a framework for integrating innate immunity into next-generation cancer treatment.
    Keywords:  immune cells; immune factors; immunotherapy; innate immunity; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1689714
  9. Semin Immunol. 2025 Nov 01. pii: S1044-5323(25)00074-0. [Epub ahead of print]80 102002
      IL-1β belongs to the IL-1 family and has the particularity to need to be cleaved by caspase-1 to be active. Once processed, IL-1β is secreted and binds to IL-1R1 on target cells, leading to the transcription of specific genes. Within a tumor, IL-1β is produced and secreted by various cell types, such as immune cells, fibroblasts or cancer cells and has pleiotropic effects on immune cells, angiogenesis, cancer cell proliferation, migration and metastasis. Thus, depending on the cancer type, the treatments or the tumor microenvironment (TME), IL-1β has opposite effects on cancer progression, thus raising the question of inducing or inhibiting IL-1β. Here, we will analyze the impact of IL-1β on cancer cells and immune cells of the TME in different types of cancers.
    Keywords:  Cancer; IL-1β; Immune cells; Inflammasomes
    DOI:  https://doi.org/10.1016/j.smim.2025.102002
  10. Cell Cycle. 2025 Nov 08. 1-23
      Hepatocellular carcinoma (HCC) is a highly aggressive liver cancer, and its progression is significantly influenced by the tumor microenvironment (TME). Tumor-derived exosomes (TEXs), an important component of the TME, significantly influence tumor growth by regulating immune responses, facilitating metastasis, and enhancing resistance to therapy. These extracellular vesicles (EVs) transport bioactive substances, such as proteins, lipids, and nucleic acids that promote interaction between cells in the TME. Recent research indicates that HCC-derived exosomes can inhibit immune cell activity, specifically in T cells, thus creating an immunosuppressive TME that facilitates tumor immune escape. They also augment metastatic capability by restructuring the extracellular matrix and promoting angiogenesis. Moreover, HCC-derived exosomes have been associated with developing resistance to drug therapy by transferring molecules such as apoptotic signals and oncogenic microRNAs, circRNAs and lncRNA. Understanding how HCC-derived exosomes affect immune modulation, metastasis, and drug resistance could yield innovative therapeutic targets to enhance therapy outcomes. This review focuses on recent research on the diverse functions of TEXs in HCC progression.
    Keywords:  Hepatocellular carcinoma; drug resistance; exosome; immune modulation; metastasis
    DOI:  https://doi.org/10.1080/15384101.2025.2583289
  11. Front Cell Dev Biol. 2025 ;13 1677201
      Myeloid-derived suppressor cells (MDSCs) play critical roles in tumor immune evasion. These heterogeneous cells are broadly classified into granulocytic (G-MDSC), monocytic (M-MDSC), and their immature precursors, early-stage MDSCs (e-MDSCs). Elucidating their differentiation and expansion mechanisms is crucial for advancing cancer immunotherapy. This review examines the key signaling pathways (e.g., JAK/STAT, NF-κB, Notch), regulatory cytokines, metabolic factors, and epigenetic modifications central to MDSC biology. A comprehensive understanding of these intricate networks provides valuable insights into tumor immune evasion and facilitates the identification of novel therapeutic targets designed to overcome MDSC-mediated immunosuppression.
    Keywords:  differentiation; expansion; immunotherapy; myeloid-derived suppressor cells; signal transduction; tumor microenvironment
    DOI:  https://doi.org/10.3389/fcell.2025.1677201
  12. Biomed Pharmacother. 2025 Oct 31. pii: S0753-3322(25)00876-5. [Epub ahead of print]193 118682
      Circular RNAs (circRNAs) are becoming more widely acknowledged for their role in breast cancer (BC) and have emerged as significant regulators of a variety of biological processes. By sponging microRNAs (miRNAs), these covalently closed noncoding RNAs can function as competing endogenous RNAs (ceRNAs), encode peptides, and control the expression of parental genes. CircRNAs influence various cellular behaviors through these mechanisms. In vivo, they affect tumor growth and metastasis, while in vitro, they regulate BC cell proliferation, migration, invasion, and apoptosis. Certain circRNAs regulate fatty acid synthesis and uptake, leading to enhanced oxidative phosphorylation in mitochondria. They also modify glucose metabolism by controlling critical glycolytic enzymes and transporters, and stimulate lipid biosynthesis. Others use miRNA-mediated signaling axes to change the expression of glutamine transporters and metabolic enzymes, which in turn affects glutamine metabolism. Additionally, circRNAs influence important signaling pathways such as PI3K/AKT/mTOR, which promote improved cell survival and resistance to drugs. Certain circRNAs in the tumor microenvironment support the recruitment of myeloid-derived suppressor cells (MDSCs), encourage M2 macrophage polarization, and alter immune checkpoints by influencing T-cell activity and PD-L1 expression. Numerous circRNAs are promising biomarkers because of their strong correlations with clinicopathological parameters, treatment response, and prognosis. The classification, biological roles, immunoregulatory functions, and therapeutic potential of circRNAs in BC are compiled in this review.
    Keywords:  Breast cancer; Circular RNAs (circRNAs); Competing endogenous RNAs (ceRNAs); Drug resistance; Immune modulation; Metabolic reprogramming; MicroRNA (miRNA); PI3K/AKT/mTOR pathway; Tumor metabolism; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.biopha.2025.118682
  13. Int J Biol Macromol. 2025 Nov 05. pii: S0141-8130(25)09348-1. [Epub ahead of print] 148791
      Although the immune checkpoint role of programmed death ligand 1 (PD-L1) is well-established and exploited in cancer immunotherapy, its intrinsic role within tumor biology and therapeutic development remains less understood, largely due to incomplete mechanistic insights. Here, we demonstrate a previously unrecognized mechanism through which PD-L1 drives epithelial-mesenchymal transition (EMT), enhances stemness, and drives metastasis in triple-negative breast cancer (TNBC) cells. Mechanistically, PD-L1 exerts these effects by silencing the anti-metastatic miR-200 family through miR-106b-TET3 axis. Furthermore, PD-L1 knockout is associated with improved tumor outcomes in orthotopic mouse models. Collectively, our findings identify a non-classical function of PD-L1 and TET3 as a critical epigenetic modifier that suppresses PD-L1-mediated EMT and breast cancer stemness during metastatic progression.
    Keywords:  PD-L1; TET3; Triple-negative breast cancer
    DOI:  https://doi.org/10.1016/j.ijbiomac.2025.148791
  14. Front Immunol. 2025 ;16 1656733
      Cancer remains a leading cause of mortality worldwide, with rising incidence and death rates continuing to rise. While conventional treatments such as surgery, radiotherapy, and chemotherapy form the backbone of cancer care, they are often limited by adverse effects, recurrence risk, and incomplete tumor eradication. Tumor immunotherapy-particularly immune checkpoint inhibitors and chimeric antigen receptor (CAR) T cell therapy-has emerged as a transformative approach by activating and reprogramming anti-tumor immune responses. Despite these advances, significant challenges persist, including limited response rates to checkpoint inhibitors, the immunosuppressive nature of the tumor microenvironment (TME), and resistance mechanisms employed by tumor cells. Growing evidence suggests that immune cell senescence is a critical contributor to TME-driven immunosuppression. Senescent immune cells exhibit functional decline, elevated expression of inhibitory immune checkpoint molecules, and increased secretion of pro-inflammatory cytokines, collectively impairing anti-tumor immunity and reducing the efficacy of immunotherapy. This review highlights the role of immune cell senescence in shaping the immunosuppressive TME and driving resistance to immunotherapy. It further discusses emerging therapeutic strategies that combine immunotherapy with senescence-targeting interventions, aiming to provide novel insights into the development of more effective cancer treatment strategies.
    Keywords:  anti-aging therapy; cancer immunotherapy; cancer treatment strategies; immune cell senescence; immunosuppression; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1656733
  15. Crit Rev Oncol Hematol. 2025 Nov 01. pii: S1040-8428(25)00389-0. [Epub ahead of print]216 105001
      Gastrointestinal tumors (GITs) are among the most prevalent and lethal malignancies worldwide, with liver metastasis (LM) being a major contributor to poor patient prognosis, contributing to recurrence in a substantial proportion (approximately 37 %) of patients undergoing radical gastrectomy. Dismally, the prognosis for GI-LM (Gastrointestinal tumors Liver Metastasis) patients remains exceedingly poor, with survival rates beyond five years languishing below 10 %. Recent studies have highlighted the critical role of tumor metabolic reprogramming and its interaction with the tumor microenvironment (TME) in the progression of gastrointestinal tumor liver metastasis. Tumor cells undergo metabolic alterations, such as enhanced glycolysis, increased glutamine metabolism, and altered lipid metabolism, to meet the demands of rapid proliferation. These metabolic changes also reshape the liver microenvironment by secreting metabolites (e.g., lactate, succinate), promoting immune suppression, angiogenesis, and stromal remodeling, thereby facilitating liver metastasis. Conversely, the liver microenvironment, including immune cells, stromal cells, and extracellular matrix components, influences tumor cell metabolism through nutrient competition, cytokine signaling, and hypoxia-driven mechanisms, creating a bidirectional interaction. This review summarizes the molecular mechanisms underlying the interplay between tumor metabolic reprogramming and the microenvironment in gastrointestinal tumor liver metastasis, explores how metabolic changes drive TME remodeling to promote metastasis, and discusses emerging therapeutic strategies targeting these mechanisms. The aim is to provide new insights for precision medicine in treating gastrointestinal tumor liver metastasis.
    Keywords:  Gastrointestinal tumors; Immune evasion; Liver metastasis; Metabolic reprogramming; Stromal interactions; Tumor metabolism; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.105001
  16. J Control Release. 2025 Nov 04. pii: S0168-3659(25)01003-X. [Epub ahead of print] 114389
      High Intensity Focused Ultrasound (HiFUS) combined with cavitation-responsive liposomes (IMP301) offers a therapeutic strategy to enhance immunotherapy efficacy in triple-negative breast cancer (TNBC). This study investigates how HIFUS, a non-invasive modality, remodels the tumor microenvironment (TME), transforming "suppressive" tumors into "responsive" ones and improving susceptibility to immune checkpoint blockade (ICB). We show that HIFUS increases vascular permeability and induces immunogenic cell death (ICD), enabling localized release of doxorubicin (Dox) from IMP301. In preclinical models, treatment improved immune infiltration, with elevated NK cells, CD8 T cells, and M1 macrophages, and reduced myeloid-derived suppressor cells (MDSCs). Antigen-specific T cells increased ~3.1-fold (p < 0.0001), and long-lasting memory T cells prevented metastasis and recurrence. Notably, targeted Dox delivery minimized systemic toxicity, permitting higher dosing with fewer adverse effects. In tumor growth studies, the combination of HIFUS and IMP301 with anti-PD1 achieved complete remission, highlighting potential for durable cancer control. Specifically, remission occurred in 4 of 10 mice, with survival extended to 50 vs 26 days (p < 0.0001). These findings demonstrate that integrating HiFUS with IMP301 effectively modulates the TME and enhances checkpoint blockade therapy, supporting its translational potential in TNBC and other solid tumors.
    Keywords:  Cancer immunotherapy; Focused ultrasound therapy; Immune checkpoint inhibition; Sonoresponsive material; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.jconrel.2025.114389
  17. Cell Rep Med. 2025 Nov 03. pii: S2666-3791(25)00468-9. [Epub ahead of print] 102395
      Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) have garnered significant research attention in the last decade. As key stromal cells of the TME, studies have explored them as a potential target for controlling cancer. Using high-throughput technologies like single-cell RNA sequencing coupled with proteomics, the classification of different CAF subgroups reveals a complex system that varies by cancer type. Unraveling novel big data, potentially through AI platforms, will be key to identifying the role of CAFs in tumor progression and therapy escape mechanisms, enabling new therapies that manipulate CAFs to increase patients' survival. We summarize and discuss new developments in an attempt to target CAFs for cancer control. Furthermore, we explore whether the complex heterogeneity of CAF origins can be targeted by single drugs in the future or require targeted, personalized therapies to combat cancer.
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102395
  18. Cell Biol Toxicol. 2025 Nov 04. 41(1): 149
      Lipolysis, a tightly regulated metabolic process, is hijacked by cancer cells to meet their energy and biosynthetic demands under stress. Central to this process is hormone-sensitive lipase (HSL), a key enzyme that orchestrates lipid mobilization by hydrolyzing diacylglycerols into free fatty acids (FFAs). This review explores the pivotal and multifaceted role of HSL in cancer metabolism, focusing on its dual function acting as both a tumor promoter and suppressor depending on the cancer type and microenvironment. Lipolysis, the breakdown of triglycerides into free fatty acids (FFAs), is essential for maintaining energy homeostasis, and is co-opted by tumor cells to fuel growth. Enzymes such as ATGL, HSL, and MGL synergistically regulate lipolysis, with HSL being the driver in this process. Dysregulation of HSL can either promote or inhibit cancer growth, depending on the tumor type. We examine how deregulated HSL activity contributes to tumor progression, metastasis, and therapy resistance through metabolic reprogramming, particularly in the context of cancer-associated adipocytes (CAAs) and fibroblasts (CAFs). CAAs and CAFs within the tumor microenvironment modulate lipid metabolism, influencing tumor progression. The review also discusses the interplay between HSL and oncogenic signaling pathways, its regulation by hormonal and transcriptional networks, and its impact on immune modulation and cachexia. Finally, we evaluate the therapeutic potential of targeting HSL, emphasizing the need for cancer-type-specific strategies to exploit its vulnerabilities without exacerbating metabolic imbalance. By decoding HSL's role in cancer energetics, this review provides a foundation for novel interventions aimed at disrupting tumor lipid metabolism. Although, therapeutic strategies targeting lipolytic enzymes, such as HSL holds promise, this review also iterates the requisite for context specific considerations for successful application.
    Keywords:  Cancer metabolism; Enzyme regulation; Hormone-sensitive lipase; Lipid droplets; Lipolysis; Tumor progression
    DOI:  https://doi.org/10.1007/s10565-025-10098-4
  19. Front Oncol. 2025 ;15 1697588
       Background: The tumor microenvironment (TME) is characterized by high lactate and proton accumulation resulting from glycolytic metabolism. While acidosis is known to influence immune and stromal cells, its direct effects on erythrocytes-the most abundant circulating cells-remain underexplored.
    Methods: An integrative review of cancer metabolism, erythrocyte physiology, and lactate transport systems was conducted using PubMed and Web of Science. From this synthesis, the CILLO-E hypothesis (Cancer-Induced Lactate Load on Erythrocytes) was formulated.
    Results: The hypothesis proposes that lactate and protons enter erythrocytes via MCT1, leading to intracellular acidification. This process disrupts glycolytic enzymes, reduces ATP production, and impairs Na+/K+-ATPase and Ca²+-ATPase activity. Energy depletion causes Ca²+ overload, which activates scramblase and inhibits flippase, resulting in PS exposure and premature eryptosis. In parallel, reduced 2,3-BPG synthesis alters hemoglobin-oxygen affinity, exacerbating hypoxia. Together, these mechanisms provide a biochemical explanation for the normocytic-normochromic anemia frequently observed in cancer. Importantly, cancer-associated anemia is multifactorial, and CILLO-E should be viewed not as a comprehensive explanation but as a complementary mechanism acting through lactate-induced erythrocyte dysfunction.
    Conclusions: The CILLO-E hypothesis reframes erythrocytes as active metabolic targets in the TME rather than passive oxygen carriers. By linking lactate-driven metabolic stress to erythrocyte dysfunction, anemia, and systemic hypoxia, it suggests a feedback loop that promotes tumor progression and highlights opportunities for erythrocyte-based biomarkers and therapeutic strategies.
    Keywords:  CILLO-E Hypothesis; cancer-associated anemia; eryptosis; erythrocyte metabolism; lactate shuttle; metabolic adaptation; redox regulation; tumor microenvironment
    DOI:  https://doi.org/10.3389/fonc.2025.1697588
  20. Sci Transl Med. 2025 Nov 05. 17(823): eadr6207
      Estrogen receptor-positive (ER+) luminal breast cancer comprises 75% of patients with breast cancer and presents notable treatment challenges because of endocrine resistance. The effectiveness of immunotherapy in endocrine therapy-resistant luminal breast cancer remains unclear. This limitation is due in part to a lack of immunocompetent preclinical models investigating the comprehensive involvement of immune cells in the tumor microenvironment (TME) in the context of endocrine resistance. In this study, we identified a subtype of immunosuppressive (M2-like) programmed death ligand 1-positive (PD-L1+) tumor-associated macrophages (TAMs) critically fostering resistance to tamoxifen (TMX) and fulvestrant (FV) through maintaining cancer stem cell (CSC) activity in new mouse models. These TAMs are recruited by Delta-like ligand 1 (DLL1), a Notch signaling ligand expressed in luminal tumor cells, through the CCR3/CCL7 axis. Combination therapy with anti-DLL1 and anti-PD-L1 antibodies with TMX reduced tumor growth and associated CSCs and reprogrammed the immunosuppressive TME in both preclinical mouse models and patient-derived explants, thus laying the foundation for a future combined immune-endocrine therapy in these patients.
    DOI:  https://doi.org/10.1126/scitranslmed.adr6207
  21. Cell Mol Bioeng. 2025 Oct;18(5): 473-488
       Introduction: Triple negative breast cancer (TNBC) has significantly worse outcomes compared to other subtypes. Strains in the tumor microenvironment (TME) generated by cancer-associated fibroblasts (CAFs) can regulate TNBC progression. Recent studies suggest that expression of VEGFR-2 on TNBC is linked to decreased survival, while our prior studies show strains activate VEGFR-2 to drive angiogenesis. We hypothesized that VEGFR-2 on TNBC can be mechanically activated to alter migration and proliferation.
    Methods: We utilized MDA-MB-231 TNBC cells loaded into the center chamber of a multi-microtissue TME model; opposing side chambers were loaded with CAFs or normal breast fibroblasts (NBFs). A second series of studies utilized magnetic beads to generate strains in the model without secretion of growth factors. Microtissues were analyzed for TNBC migration and proliferation via Ki67 staining.
    Results: TNBC cells migrated significantly more towards CAFs compared to NBFs (5×); TME models with magnetic beads showed a 2× increase in migration compared to no strain controls. TNBC cells treated with shRNA against VEGFR-2 demonstrated decreased overall migration but still significantly more towards CAFs vs. NBFs (2×). Proliferation analyses showed strain significantly increased Ki67 in control cells (10%+ vs. 28%+) but not in shVEGFR-2 TNBC (~ 10% all conditions).
    Discussion: These studies demonstrate that strain in the TME drives increased migration and proliferation of TNBC. Loss of VEGFR-2 suppresses migration and growth, even with mechanical stimulation. Therefore, our results suggest that mechanosignaling via VEGFR-2 on TNBC may regulate disease progression and potentially explain failure of anti-VEGFR-2 drugs in breast cancer patients.
    Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-025-00866-x.
    Keywords:  Microphysiological systems; Triple negative breast cancer; Tumor mechanobiology; Tumor microenvironment
    DOI:  https://doi.org/10.1007/s12195-025-00866-x
  22. Front Cell Dev Biol. 2025 ;13 1676945
      Trogocytosis, a rapid and contact-dependent exchange of plasma membrane fragments and associated molecules between cells, has recently emerged as a critical but underappreciated player in cancer biology. Traditionally studied in the context of immune cell communication, trogocytosis is now recognized for its paradoxical role in modulating tumor progression and therapeutic response across a broad spectrum of malignancies. This review highlights the novel and dynamic functions of trogocytosis in shaping the tumor microenvironment (TME), promoting immune evasion and influencing metastatic potential. Notably, cancer cells exploit trogocytosis to acquire immune regulatory molecules such as CD45, CD4 and checkpoint proteins, effectively dampening anti-tumor responses while enhancing their own survival. Simultaneously, immune effector cells including macrophages, T cells and natural killer (NK) cells leverage trogocytosis to recognize, attack and even kill tumor cells through mechanisms such as trogoptosis. Compelling new evidence also links trogocytosis to therapeutic resistance, particularly in chimeric antigen receptor (CAR-T and CAR-NK) cell therapies, where tumor antigens like CD19 and CD22 are siphoned off by effector cells, leading to T cell fratricide, functional exhaustion and tumor relapse. Beyond its biological significance, trogocytosis is gaining attention as a translational tool in oncology. It offers a novel platform for antigen-specific drug delivery, spatially restricted immune modulation and biomarker discovery through the detection of trogocytosed molecules on circulating immune-cells or extracellular vesicles. These findings redefine trogocytosis as not merely a passive membrane exchange process, but a central mechanism of intercellular communication with profound implications for cancer progression, immunotherapy and precision medicine.
    Keywords:  CAR-immune cell fraticide; anti-tumorigenic trogocytosis; chemoresistance; metastasis; pro-tumorigenic trogocytosis; tumor microenvironment
    DOI:  https://doi.org/10.3389/fcell.2025.1676945
  23. Front Biosci (Landmark Ed). 2025 Oct 30. 30(10): 37633
      Liver cancer, particularly hepatocellular carcinoma (HCC), represents a global health challenge. The tumor microenvironment (TME) plays a pivotal role in the progression and therapeutic resistance of HCC. Interventional therapies have emerged as pivotal modalities in the treatment of liver cancer, especially in cases that are unsuitable for surgical resection. The evolution of these techniques has been markedly enhanced by the integration of artificial intelligence (AI), which has the potential to increase precision, improve outcomes, and personalize patient care. This review covers modern interventional therapies for liver cancer, highlighting recent advances in minimally invasive procedures. It describes the intricate liver TME and emphasizes the importance of characterizing its diversity and identifying therapeutic targets. Additionally, we discuss how AI can decipher TME complexities, predict responses, categorize patients, and personalize treatments. By elucidating connections between the TME, therapeutic interventions, and AI, this review aims to improve the management and care of patients with liver cancer.
    Keywords:  artificial intelligence; hepatocellular carcinoma; interventional therapy; liver cancer; tumor microenvironment
    DOI:  https://doi.org/10.31083/FBL37633
  24. World J Clin Oncol. 2025 Oct 24. 16(10): 108954
      Neurotransmitter-mediated regulation plays a multi-dimensional role in the tumor microenvironment, profoundly influencing key processes such as tumor immune evasion, metabolic reprogramming, and metastasis. However, the upstream regulatory mechanisms linking neural inputs to immune evasion and metabolic reprogramming remain incompletely understood. We systematically summarize current evidence from molecular, cellular, and immunological studies to elucidate how neurotransmitter-dependent mechanisms drive dynamic changes in the tumor microenvironment through the regulation of tumor cells and immune cells, and map the complex interaction networks between the nervous system and tumor progression. We propose a unifying "neuro-metabolic-immune axis" framework that highlights the dual role of neurotransmitters in suppressing anti-tumor immunity and facilitating tumor adaptation. By mapping this axis, we reveal new insights into tumor ecology and identify neural pathways as promising therapeutic targets. Targeting these pathways may enhance immunotherapy and disrupt tumor-supportive metabolism, offering new directions in precision oncology.
    Keywords:  Immune evasion; Immunotherapy; Metabolic reprogramming; Neurotransmitter signaling; Tumor microenvironment
    DOI:  https://doi.org/10.5306/wjco.v16.i10.108954
  25. Int J Surg. 2025 Nov 06.
      Extrahepatic cholangiocarcinoma (eCCA) is a highly malignant tumor with a propensity for metastasis, reflected in its estimated 5-year survival rate of 11%. Metastasis greatly impairs the effectiveness of cancer therapies and increases cancer-related deaths. Therefore, gaining an understanding of the complex metastatic process is essential for development of effective treatments. Metastasis progression involves remodeling of the tumor microenvironment (TME) and the formation of pre-metastatic niches (PMNs), facilitating the migration and survival of tumor cells from the primary site to distant locations. As the eCCA progresses, immune cells, fibroblasts and endothelial cells in the TME are gradually converted to a tumor-supportive phenotype, coordinating tumor metastasis through intercellular interactions. Recent studies have confirmed that bile, a body fluid in close contact with eCCA, is also involved in the formation of supportive TME and tumor metastasis. On the other hand, primary tumors construct PMNs conducive to cancer cell colonization in distant organs prior to metastatic tumor formation through paracrine effects. This review aimed to summarize the mechanisms by which TME and PMNs drive eCCA metastasis, current treatment strategies for metastatic eCCA, and prospects for future research.
    Keywords:  extrahepatic cholangiocarcinoma; metastasis; premetastatic niches; therapies; tumor microenvironment
    DOI:  https://doi.org/10.1097/JS9.0000000000003852
  26. Front Immunol. 2025 ;16 1675807
      Adoptive cell therapy (ACT) employing chimeric antigen receptor (CAR) engineering represents a transformative advancement in cancer immunotherapy. CAR-T cell therapies have demonstrated significant clinical success in hematological malignancies, yet their application to solid tumors faces persistent challenges. Key limitations include the paucity of tumor-specific antigens, poor intratumoral infiltration, immunosuppressive tumor microenvironment (TME), and treatment-related toxicities such as cytokine release syndrome (CRS) and neurotoxicity. In contrast, CAR natural killer (CAR-NK) cells show promise in solid tumors such as ovarian, pancreatic, and glioblastoma, with encouraging preclinical and early clinical evidence, although limited persistence and antigen heterogeneity remain major challenges. Unlike CAR-T cells, CAR-NK therapies mediate tumor clearance through both cytotoxic (e.g., granzyme/perforin release) and cytokine-mediated mechanisms while mitigating toxicity risks. Their lack of human leukocyte antigen (HLA) dependency enables "off-the-shelf" manufacturing from allogeneic donors, circumventing patient-specific production bottlenecks. CAR-macrophage (CAR-M) therapies further address solid tumor barriers by leveraging innate phagocytic clearance, antigen-presenting functions, and TME penetration. Macrophages inherently infiltrate hypoxic tumor regions and remodel stromal barriers, enabling CAR-Ms to synergize with adaptive immunity by cross-priming T cells. Preclinical models highlight CAR-M efficacy in depleting immunosuppressive tumor-associated macrophages (TAMs) and reversing TME-driven immune evasion. Emerging CAR- Gamma-Delta T (CAR-γδ T) cell therapies combine CAR-mediated antigen specificity with the intrinsic tumoricidal activity of γδ T cells, which recognize stress-induced ligands independently of major histocompatibility complex (MHC) presentation. This dual-targeting capability enhances tumor selectivity while reducing on-target/off-tumor toxicity. This review systematically examines cellular sources, mechanistic advantages and clinical progress. By evaluating these platforms' complementary strengths, we propose rational strategies for integrating CAR-NK, CAR-M, and CAR-γδ T cells into tailored therapeutic regimens for solid tumors.
    Keywords:  NK cells; chimeric antigen receptor; macrophage cells; solid tumor; γδT cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1675807
  27. Cureus. 2025 Oct;17(10): e93950
      Found in all human cells, the purine nucleoside adenosine plays various roles in different metabolic pathways. Adenosine is not stored in vesicles but is released continuously, based on metabolic demands. Essential in energy production, adenosine is a full agonist at four known receptors (A1, A2A, A2B, and A3), and is produced either intracellularly or in the extracellular space. Adenosine is the building block for adenosine triphosphate (ATP), a vasodilator that inhibits certain cerebral neurotransmissions. Adenosine diphosphate (ADP) releases energy via its phosphate bonds and can "recharge" by adding phosphate groups later on, unlike ATP. The A3 receptors are most densely expressed in humans in the liver, lungs, immune cells, heart, and brain, and A3 agonists confer cytoprotection, making A3 agonists an intriguing potential anticancer drug. A3 receptors are so highly expressed in cancer cells and tumors that they serve as cancer biomarkers. To date, A3 agonists and A2A antagonists have emerged as potential anticancer drugs. Paradoxically, A3 is upregulated in primary tumors and metastatic disease, and A3 activity correlates with invasive actions of tumor cells. This contradictory effect, paralleled by the pro- and anti-inflammatory effects of A3, may be explicable because the downregulation of A3 receptors may produce different effects than A3 agonism. Adenosine is abundant in the tumor microenvironment (TME), and cancer cells seem adept at adjusting their metabolic processes to attune themselves to their specific TME. The derangement of energy metabolism is characteristic of cancer, and cancer spreads as normal cells in proximity to a tumor become increasingly neoplastic and tumorigenic. This expands our old notion of tumors as discrete, separate bodies and views them now as complex layers of cancer cells of different functions; these cells, which can include recruited normal cells, interact with each other. The role of mast cells is emerging as an important, albeit enigmatic, part of the TME. Mast cells are abundant in tumors and appear to have a pro-inflammatory effect, but it is not entirely clear if they serve to promote or oppose the growth of tumors. Crosstalk between mast cells and some types of cancer cells, involving adenosine, has been observed. The translational impact and clinical implications of these findings remain speculative.
    Keywords:  a3 receptor; adenosine; adenosine receptors; anticancer drugs; cancer cell biology; cytoprotection; mast cells proliferation; tumor microenvironment (tme)
    DOI:  https://doi.org/10.7759/cureus.93950
  28. Cell Death Discov. 2025 Nov 06. 11(1): 506
      The phospholipid scramblases Xkr8 and TMEM16F externalize phosphatidylserine (PS) by distinct mechanisms. Xkr8 is activated by caspase-mediated proteolytic cleavage and, in synergy with the inactivation of P4-ATPase flippases, results in the irreversible externalization of PS on apoptotic cells and an "eat-me" signal for efferocytosis. In contrast, TMEM16F is a calcium-activated scramblase that reversibly externalizes PS on viable cells via the transient increase in intracellular calcium in live cells. The tumor microenvironment (TME) is abundant with exposed PS, resulting from prolonged oncogenic and metabolic stresses and high apoptotic indexes of tumors. Such chronic PS externalization in the TME has been linked to host immune evasion from interactions of PS with inhibitory PS receptors, such as TAM and TIM family receptors. Here, in an effort to better understand the contributions of apoptotic vs live cell PS-externalization to tumorigenesis and immune evasion, we employed an EO771 orthotopic breast cancer model and genetically ablated Xkr8 and TMEM16F using CRISPR/Cas9. While neither the knockout of Xkr8 nor TMEM16F showed defects in cell intrinsic properties related to proliferation, tumor-sphere formation, and growth factor signaling, both knockouts suppressed tumorigenicity in immune-competent mice, but not in NOD/SCID or RAG-knockout immune-deficient strains. Mechanistically, Xkr8-KO tumors suppressed macrophage-mediated efferocytosis, and TMEM16F-KO suppressed ER stress/calcium-induced PS externalization. Our data support an emerging idea in immune-oncology that constitutive PS externalization, mediated by scramblase dysregulation on tumor cells, supports immune evasion in the tumor microenvironment. This links apoptosis/efferocytosis and oncogenic stress involving calcium dysregulation, contributing to PS-mediated immune escape and cancer progression.
    DOI:  https://doi.org/10.1038/s41420-025-02789-y
  29. Cytokine Growth Factor Rev. 2025 Oct 25. pii: S1359-6101(25)00137-6. [Epub ahead of print]86 181-198
      Natural killer (NK) cells serve as critical first responders within the immune system, orchestrating antitumor responses by directly eliminating malignant cells and modulating broader immune functions. Their capacity to recognize stressed targets without MHC-restricted neoantigen presentation, together with a favorable safety profile, underpins growing enthusiasm for NK cell‑based therapeutics. Nonetheless, the clinical efficacy of NK-cell-based therapies in solid tumor has been hindered by tumor microenvironment (TME)-driven immunosuppression, metabolic competition, and cell-intrinsic exhaustion mechanisms. In this review, we outline the journey from the discovery of NK cells to their role as a breakthrough target in cancer immunotherapy. We then provide a comprehensive framework of NK cell biology, from homeostatic regulation to TME-driven epigenetic, transcriptional, and metabolic adaptations. Notably, we discuss recent investigations into NK cell subset heterogeneity and their interactions with tumor cells, immune cells and the gut microbiome. Finally, we critically evaluated clinical progress while highlighting the imperative for rationally designed, mechanism-based combinations that address tumor escape pathways. The review concludes with a forward-looking perspective on engineering solutions to unlock the full therapeutic potential of NK cells across solid tumors and hematologic malignancies.
    Keywords:  CAR-NK cells; Cancer immunotherapy; NK cell metabolism; NK cell subsets; NK cells; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.cytogfr.2025.10.003
  30. Ann Med Surg (Lond). 2025 Nov;87(11): 7112-7127
      Cervical cancer, predominantly caused by high-risk human papillomavirus (HPV) infections, continues to be a major health challenge globally. Recent research has highlighted the significant role of neutrophils, a type of white blood cell integral to the immune system, in the pathogenesis of cervical cancer. This review examines the dualistic role of neutrophils in cervical cancer, emphasizing their contribution to both inflammation and tumor progression. Understanding the intricate relationship between neutrophils and cervical cancer could unveil new therapeutic targets for better disease management. Neutrophils are key mediators of the immune response and inflammation. In the context of cervical cancer, these cells are recruited to the tumor microenvironment where they can adopt tumor-promoting phenotypes. Tumor-associated neutrophils (TANs) facilitate various processes that aid tumor growth and metastasis, such as producing reactive oxygen species (ROS) and proteases that induce DNA damage, releasing cytokines and chemokines that promote angiogenesis, and forming neutrophil extracellular traps (NETs) that enhance metastatic potential. Furthermore, TANs contribute to immune suppression by inhibiting the activity of cytotoxic T lymphocytes and natural killer cells, allowing cancer cells to evade immune surveillance. Given their pivotal role in cervical cancer progression, neutrophils represent a promising target for novel therapeutic strategies. Approaches such as inhibiting neutrophil recruitment to the tumor site, blocking NET formation, and modulating TAN phenotypes from pro-tumor to anti-tumor are being explored. These strategies aim to disrupt the supportive role of neutrophils in tumor development and progression, potentially leading to improved outcomes for patients with cervical cancer.
    Keywords:  cervical cancer; inflammation; metastasis; neutrophils; tumor microenvironment
    DOI:  https://doi.org/10.1097/MS9.0000000000002679
  31. Cell Rep. 2025 Nov 03. pii: S2211-1247(25)01297-5. [Epub ahead of print]44(11): 116526
      Cancer-associated adipocytes (CAAs) reprogram the metabolic status of the tumor microenvironment (TME). The metabolic crosstalk between CAAs and CD8+T cells in the TME remains unclear. Here, we report that CAAs undergo lipolysis, releasing free fatty acids that promote lipid peroxidation and disturb mitochondrial homeostasis in CD8+T cells, leading to their functional exhaustion. Importantly, we uncover that fibroblast growth factor 21 (FGF21) drives CAA lipolysis in an autocrine manner by upregulating adipose triglyceride lipase (ATGL) via FGFR1/KLB-p38 signaling. FGF21 deletion in adipose tissue or ATGL inhibition impedes CAA lipolysis, mitigates lipid peroxidation, normalizes mitochondrial dynamics of CD8+T cells, and restores their effector function, consequently blunting tumor growth. Moreover, FGF21 deficiency or ATGL inhibition enhances the anti-tumor activity of CD8+T cells in response to anti-PD-1 treatment, yielding greater therapeutic efficacy. Our findings highlight the pivotal role of CAA lipolysis in CD8+T cell dysfunction within the TME, suggesting that targeting CAA lipolysis represents a valuable avenue for improving cancer immunotherapy.
    Keywords:  ATGL; CD8(+)T; CP: cancer; CP: immunology; FGF21; T cell exhaustion; cancer-associated adipocytes; lipolysis; tumor immunity
    DOI:  https://doi.org/10.1016/j.celrep.2025.116526
  32. Ann Med Surg (Lond). 2025 Oct;87(10): 6551-6563
      Tumor immunology is a critical area of cancer research that focuses on the interplay between the immune system and tumor cells. The immune system has the ability to recognize and eliminate cancer cells through various immune cells, such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. However, tumors have developed sophisticated mechanisms to evade immune detection, such as the loss of tumor antigen expression, recruitment of immunosuppressive cells, and upregulation of immune checkpoints. These immune evasion strategies allow tumors to grow uncontrollably and metastasize. Immunotherapy has emerged as a promising treatment modality that harnesses the power of the immune system to fight cancer. Key strategies include immune checkpoint inhibitors, which block inhibitory receptors like programmed cell death protein 1 and cytotoxic T-lymphocyte-associated protein 4, and adoptive cell therapies, which involve the transfer of tumor-specific T cells or NK cells into patients. Additionally, monoclonal antibodies and cancer vaccines are being explored to directly target tumor cells and enhance immune responses. Despite the success of these therapies, challenges such as tumor heterogeneity, resistance to treatment, and immune-related adverse effects persist.
    Keywords:  CAR-T cell therapy; cancer immunotherapy; immune checkpoints; immune evasion; tumor microenvironment (TME)
    DOI:  https://doi.org/10.1097/MS9.0000000000003719
  33. Cell Signal. 2025 Nov 05. pii: S0898-6568(25)00628-X. [Epub ahead of print] 112213
      Growing evidence indicates that the complex and heterogeneous tumor microenvironment (TME) beyond the tumor cells plays a pivotal role in cancer development and therapy. Notably, cancer-associated fibroblasts (CAFs)-a key stromal cell population within the TME-are closely associated with malignant tumor progression and therapeutic resistance and have progressively emerged as a research focus in oncology. As a major mesenchymal cell population in thyroid cancer (TC), CAFs exhibit high heterogeneity and dynamic plasticity. They secrete numerous cytokines and non-cytokine factors that abnormally remodel the extracellular matrix, directly influence the biological behavior of tumor cells, and concurrently modulate immune responses, consequently promoting therapeutic resistance. Given the unique and crucial role of CAFs in promoting TC progression, a thorough understanding of their intricate biology and the precise molecular mechanisms that regulate the malignant behavior of TC is essential for developing effective therapeutic strategies to improve outcomes for patients with TC. The present review provides an overview of the latest research advances on CAFs in TC, encompassing their origin, phenotype, and functional heterogeneity; the fundamental mechanisms by which CAFs promote TC progression and therapeutic resistance through bidirectional interactions with other TME components, including immune and tumor cells; and a systematic summary of therapeutic strategies targeting CAFs and their clinical translation outcomes. Furthermore, this review offers a concise perspective on future research directions in the field of CAFs.
    Keywords:  Cancer associate fibroblasts(CAFs); Extracellular matrix(ECM); Fibrosis; Heterogeneity; Thyroid cancer(TC)
    DOI:  https://doi.org/10.1016/j.cellsig.2025.112213
  34. J Immunother Cancer. 2025 Nov 04. pii: e012127. [Epub ahead of print]13(11):
      The proper functioning of the immune system requires an adequate balance between myeloid and lymphoid populations. Tumor growth alters this balance, also through the dramatic expansion of immunosuppressive myeloid populations, which block specific immunity, fueling tumor growth and dissemination and limiting the effectiveness of antitumor therapies, including immunotherapy. Tumors alter the expansion and functions of myeloid cells by acting locally in the tumor microenvironment, as well as on myeloid progenitors, through the manipulation of metabolic traits that govern their functions. The understanding of these metabolic alterations and their clinical translation is expected to offer new valid therapeutic options.
    Keywords:  Immunosuppression; Immunotherapy; Macrophage; Myeloid-derived suppressor cell - MDSC; Tumor microenvironment - TME
    DOI:  https://doi.org/10.1136/jitc-2025-012127
  35. Expert Rev Clin Immunol. 2025 Nov 05. 1-14
       INTRODUCTION: The tumor microenvironment is a dynamic and balanced internal environment that accompanies the whole process of tumor development, invasion, and metastasis. Immune checkpoint therapy, chimeric antigen receptor cell therapy, oncolytic virus therapy, and bispecific antibody therapy are the most anticipated immunotherapy methods. These therapies break the microenvironment conducive to tumor growth by regulating anti-tumor immunity. The underlying characteristics of the tumor immune microenvironment are the key scientific issues to break the bottleneck of solid tumor immunotherapy. With the rapid development and application of single-cell sequencing technology and spatial oncology technology, scientists have gradually recognized more complex details of cell-cell interaction in the tumor microenvironment.
    AREAS COVERED: We review the latest research progress in tumor immune escape, tumor metabolic reprogramming, and neuroimmunity [PubMed database published between 2000 and 21 May 2025]. We analyze the effects of these biological processes on anti-tumor immunity, to seek breakthroughs for the design of combined immune therapy. We also summarize the latest research on major immunotherapies mentioned above.
    EXPERT OPINION: Through the integration of frontier and hot basic scientific issues and the design of clinical trials, we hope to identify the potential combination treatment plan for immunotherapy to overcome the tumor microenvironment of solid tumors.
    Keywords:  CAR-NK; immune checkpoint inhibitor; immune escape; oncolytic virus; tumor microenvironment
    DOI:  https://doi.org/10.1080/1744666X.2025.2585349