bims-flamet Biomed News
on Cytokines and immunometabolism in metastasis
Issue of 2024‒06‒30
thirty papers selected by
Peio Azcoaga, Biodonostia HRI



  1. BMB Rep. 2024 Jun 26. pii: 6203. [Epub ahead of print]
      Immunotherapy represents a promising treatment strategy for targeting various tumor types. However, the overall response rate is low due to the tumor microenvironment (TME). In the TME, numerous distinct factors actively induce immunosuppression, restricting the efficacy of anticancer immune reactions. Recently, metabolic reprogramming of tumors has been recognized for its role in modulating the tumor microenvironment to enhance immune cell responses in the TME. Furthermore, recent elucidations underscore the critical role of metabolic limitations imposed by the tumor microenvironment on the effectiveness of antitumor immune cells, guiding the development of novel immunotherapeutic approaches. Hence, achieving a comprehensive understanding of the metabolic requirements of both cancer and immune cells within the TME is pivotal. This insight not only aids in acknowledging the current limitations of clinical practices but also significantly shapes the trajectory of future research endeavors in the domain of cancer immunotherapy. In addition, therapeutic interventions targeting metabolic limitations have exhibited promising potential as combinatory treatments across diverse cancer types. In this review, we first discuss the metabolic barriers in the TME. Second, we explore how the immune response is regulated by metabolites. Finally, we will review the current strategy for targeting metabolism to not simply inhibit tumor growth but also enhance antitumor immune responses. Thus, we could suggest potent combination therapy for improving immunotherapy with metabolic inhibitors.
  2. Front Immunol. 2024 ;15 1379365
      Metal ions play an essential role in regulating the functions of immune cells by transmitting intracellular and extracellular signals in tumor microenvironment (TME). Among these immune cells, we focused on the impact of metal ions on T cells because they can recognize and kill cancer cells and play an important role in immune-based cancer treatment. Metal ions are often used in nanomedicines for tumor immunotherapy. In this review, we discuss seven metal ions related to anti-tumor immunity, elucidate their roles in immunotherapy, and provide novel insights into tumor immunotherapy and clinical applications.
    Keywords:  T cells; metal ions; nanomedicines; tumor immunotherapy; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2024.1379365
  3. Pathol Int. 2024 Jun 25.
      Paget's "Seed and Soil" theory, proposed in 1889, emphasizes the importance of the microenvironment where cancer cells grow in metastatic sites. Over a century later, this concept remains a cornerstone in comprehending cancer biology and devising treatment strategies. The "Seed and Soil" theory, which initially explained how cancer spreads to distant organs, now also applies to the tumor microenvironment (TME) within primary tumors. This theory emphasizes the critical interaction between cancer cells ("seeds") and their surrounding environment ("soil") and how this interaction affects both tumor progression within the primary site and at metastatic sites. An important point to note is that the characteristics of the TME are not static but dynamic, undergoing substantial changes during tumor progression and after treatment with therapeutic drugs. Cancer-associated fibroblasts (CAFs), recognized as the principal noncancerous cellular component within the TME, play multifaceted roles in tumor progression including promoting angiogenesis, remodeling the extracellular matrix, and regulating immune responses. In this comprehensive review, we focus on the findings regarding how the dynamics of CAFs contribute to cancer progression and drug sensitivity. Understanding the dynamics of CAFs could provide new insights into cancer pathology and lead to important advancements in cancer research and treatment.
    Keywords:  cancer‐associated fibroblasts; drug sensitivity; dynamics; seed and soil; tumor microenvironment; tumor progression
    DOI:  https://doi.org/10.1111/pin.13461
  4. Int J Stem Cells. 2024 Jun 26.
      Glutathione (GSH), the main cellular antioxidant, dynamically influences tumor growth, metastasis, and resistance to therapy in the tumor microenvironment (TME), which comprises cancer cells, immune cells, stromal cells, and non-cellular components, including the extracellular matrix, metabolites, hypoxia, and acidity. Cancer stem cells (CSCs) and T cells are minor but significant cell subsets of the TME. GSH dynamics influences the fate of CSCs and T cells. Here, we explored GSH dynamics in CSCs and T cells within the TME, as well as therapeutic approaches that could target these dynamics.
    Keywords:  Cancer stem cells; Glutathione dynamics; T cells; Tumor microenvironment
    DOI:  https://doi.org/10.15283/ijsc24060
  5. MedComm (2020). 2024 Jul;5(7): e602
      Tumor-driven immune suppression is a critical mechanism by which cancer cells evade the host immune system, leading to tumor growth and metastasis. The tumor immune microenvironment contains a large population of immune-suppressing myeloid cells, which play a key role in tumor development and drug resistance to existing immunotherapy. Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) are important components of the immunosuppressive microenvironment. Uncovering the molecular mechanisms of PMN-MDSCs and finding specific targets for PMN-MDSCs to regulate tumor immune microenvironment is the focus and challenge of current immunotherapy. In a recent issue of Nature, Wang and colleagues revealed that CD300ld on PMN-MDSCs is required for tumor-driven immune suppression(1), this provided a new target for cancer immunotherapy, The study identified CD300ld as a novel, highly conserved tumor immunosuppressive receptor. CD300ld is highly expressed specifically on PMN-MDSCs and is a key receptor in regulating the recruitment and immunosuppressant function of PMN-MDSCs. Targeting CD300ld can reshape the tumor immune microenvironment by inhibiting the recruitment and function of PMN-MDSCs, resulting in broad-spectrum anti-tumor effects. CD300ld target shows good safety, conservation, anti-tumor effectiveness, and synergism with the Programmed death-1 target, which is expected to become a new ideal target for tumor immunotherapy.
    DOI:  https://doi.org/10.1002/mco2.602
  6. Vaccines (Basel). 2024 Jun 07. pii: 633. [Epub ahead of print]12(6):
      We previously reported that nano-pulse treatment (NPT), a pulsed power technology, resulted in 4T1-luc mammary tumor elimination and a strong in situ vaccination, thereby completely protecting tumor-free animals against a second live tumor challenge. The mechanism whereby NPT mounts effective antitumor immune responses in the 4T1 breast cancer predominantly immunosuppressive tumor microenvironment (TME) remains unanswered. In this study, orthotopic 4T1 mouse breast tumors were treated with NPT (100 ns, 50 kV/cm, 1000 pulses, 3 Hz). Blood, spleen, draining lymph nodes, and tumors were harvested at 4-h, 8-h, 1-day, 3-day, 7-day, and 3-month post-treatment intervals for the analysis of frequencies, death, and functional markers of various immune cells in addition to the suppressor function of regulatory T cells (Tregs). NPT was verified to elicit strong in situ vaccination (ISV) against breast cancer and promote both acute and long-term T cell memory. NPT abolished immunosuppressive dominance systemically and in the TME by substantially reducing Tregs, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs). NPT induced apoptosis in Tregs and TAMs. It also functionally diminished the Treg suppression capacity, explained by the downregulation of activation markers, particularly 4-1BB and TGFβ, and a phenotypic shift from predominantly activated (CD44+CD62L-) to naïve (CD44-CD62L+) Tregs. Importantly, NPT selectively induced apoptosis in activated Tregs and spared effector CD4+ and CD8+ T cells. These changes were followed by a concomitant rise in CD8+CD103+ tissue-resident memory T cells and TAM M1 polarization. These findings indicate that NPT effectively switches the TME and secondary lymphatic systems from an immunosuppressive to an immunostimulatory state, allowing cytotoxic T cell function and immune memory formation to eliminate cancer cells and account for the NPT in situ vaccination.
    Keywords:  apoptosis; breast cancer; immunosuppression; in situ vaccination; memory T cells; myeloid-derived suppressor cells; nano-pulse treatment (NPT); regulatory T cells; tumor microenvironment; tumor-associated macrophages
    DOI:  https://doi.org/10.3390/vaccines12060633
  7. MedComm (2020). 2024 Jul;5(7): e607
      In a recent Nature elegant study, Wang et al. identified CD300ld, a novel functionally highly conserved tumor immunosuppressive receptor, highly expressed specifically on polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), as well as a key receptor in the regulation of recruitment and immunosuppressive function of PMN-MDSCs. Targeting CD300ld could remodel the tumor immune microenvironment, resulting in a broad-spectrum anti-tumor effect.
    Keywords:  immunotherapy; neutrophil; tumor
    DOI:  https://doi.org/10.1002/mco2.607
  8. Inflamm Res. 2024 Jun 27.
      Alternatively activated macrophage (M2) polarization can result in one of four subtypes based on cytokines and signaling pathways associated with macrophage activation: M2a, M2b, M2c, and M2d macrophages. The majority of M2 subtypes are anti-inflammatory and pro-angiogenic, secreting growth factors (VEGF, PDGF) and matrix metalloproteinases (MMP2, MMP9) which boost tumor growth, metastasis, and invasion. M2-polarized macrophages are associated with immune suppressor cells harboring Myeloid derived suppressor cells, Regulatory T cells (Tregs), Regulatory B cells as well as alternatively activated (N2) neutrophils. Treg cells selectively support the metabolic stability, mitochondrial integrity, and survival rate of M2-like TAMs in an indirect environment. Also, the contribution of Breg cells influences macrophage polarization towards the M2 direction. TAM is activated when TAN levels in the tumor microenvironment are insufficient or vice versa, suggesting that macrophage and its polarization are fine-tuned. Understanding the functions of immune suppressive cells, mediators, and signaling pathways involved with M2 polarization will allow us to identify potential strategies for targeting the TAM repolarization phenotype for innovative immunotherapy approaches. In this review, we have highlighted the critical factors for M2 macrophage polarization, differential cytokine/chemokine profiles of M1 and M2 macrophage subtypes, and other immune cells' impact on the polarization within the immunosuppressive niche.
    Keywords:  Immunosuppressive niche; Immunotherapy; M2 macrophage; Macrophage polarization; Tumor-associated macrophage
    DOI:  https://doi.org/10.1007/s00011-024-01907-3
  9. Front Cell Dev Biol. 2024 ;12 1416472
      Even with sufficient oxygen, tumor cells use glycolysis to obtain the energy and macromolecules they require to multiply, once thought to be a characteristic of tumor cells known as the "Warburg effect". In fact, throughout the process of carcinogenesis, immune cells and stromal cells, two major cellular constituents of the tumor microenvironment (TME), also undergo thorough metabolic reprogramming, which is typified by increased glycolysis. In this review, we provide a full-scale review of the glycolytic remodeling of several types of TME cells and show how these TME cells behave in the acidic milieu created by glucose shortage and lactate accumulation as a result of increased tumor glycolysis. Notably, we provide an overview of putative targets and inhibitors of glycolysis along with the viability of using glycolysis inhibitors in combination with immunotherapy and chemotherapy. Understanding the glycolytic situations in diverse cells within the tumor immunological milieu will aid in the creation of subsequent treatment plans.
    Keywords:  aerobic glycolysis; cancer progression; metabolic reprogramming; targeted therapy; tumor microenvironment
    DOI:  https://doi.org/10.3389/fcell.2024.1416472
  10. Int J Mol Sci. 2024 Jun 13. pii: 6532. [Epub ahead of print]25(12):
      Chemokines and cytokines represent an emerging field of immunotherapy research. They are responsible for the crosstalk and chemoattraction of immune cells and tumor cells. For instance, CXCL9/10/11 chemoattract effector CD8+ T cells to the tumor microenvironment, making an argument for their promising role as biomarkers for a favorable outcome. The cytokine Interleukin-15 (IL-15) can promote the chemokine expression of CXCR3 ligands but also XCL1, contributing to an important DC-T cell interaction. Recruited cytotoxic T cells can be clonally expanded by IL-2. Delivering or inducing these chemokines and cytokines can result in tumor shrinkage and might synergize with immune checkpoint inhibition. In addition, blocking specific chemokine and cytokine receptors such as CCR2, CCR4 or Il-6R can reduce the recruitment of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs) or regulatory T cells (Tregs). Efforts to target these chemokines and cytokines have the potential to personalize cancer immunotherapy further and address patients that are not yet responsive because of immune cell exclusion. Targeting cytokines such as IL-6 and IL-15 is currently being evaluated in clinical trials in combination with immune checkpoint-blocking antibodies for the treatment of metastatic melanoma. The improved overall survival of melanoma patients might outweigh potential risks such as autoimmunity. However, off-target toxicity needs to be elucidated.
    Keywords:  chemokines; cytokines; immunotherapy; tumor microenvironment
    DOI:  https://doi.org/10.3390/ijms25126532
  11. Microbiol Immunol. 2024 Jun 25.
      The tumor microenvironment of hepatoblastoma (HB), the most common pediatric liver tumor, predominantly exhibits a myeloid immune landscape. in which tumor-associated macrophages (TAMs) are considered the core component. The crosstalk between TAMs and HB cells markedly influences tumor behavior. TAM-derived factors are involved in tumor proliferation and vascular invasion. On the other hand, HB cell secretome attracts, stimulates, and reprograms TAMs to be immunosuppressive in favor of tumor invasion, rather than their innate role in combating tumor growth, such crosstalk sometimes forms bidirectional feedback loops, making the tumor more virulent and resistant to routine therapeutics. Consequently, TAMs are the common denominator of most suggested HB immunotherapeutic strategies. Macrophage immune checkpoint inhibitors, macrophage-mediated antibody-dependent cellular phagocytosis, and the novel chimeric antigen receptor macrophage therapy (CAR Mφ) are currently under trial. In this review, we will summarize the significance of TAMs and their potential role as a therapeutic target in HB.
    Keywords:  chimeric antigen receptor; hepatoblastoma; immune checkpoint inhibitors; tumor microenvironment; tumor‐associated macrophages
    DOI:  https://doi.org/10.1111/1348-0421.13162
  12. Cell Biochem Funct. 2024 Jul;42(5): e4081
      Butyrophilin (BTN) proteins are a type of membrane protein that belongs to the Ig superfamily. They exhibit a high degree of structural similarity to molecules in the B7 family. They fulfill a complex function in regulating immune responses, including immunomodulatory roles, as they influence γδ T cells. The biology of BTN molecules indicates that they are capable of inhibiting the immune system's ability to detect antigens within tumors. A dynamic association between BTN molecules and cellular surfaces is also recognized in specific contexts, influencing their biology. Notably, the dynamism of BTN3A1 is associated with the immunosuppression of T cells or the activation of Vγ9Vδ2 T cells. Cancer immunotherapy relies heavily on T cells to modulate immune function within the intricate interaction of the tumor microenvironment (TME). A significant interaction between the TME and antitumor immunity involves the presence of BTN, which should be taken into account when developing immunotherapy. This review explores potential therapeutic applications of BTN molecules, based on the current understanding of their biology.
    Keywords:  Vγ9Vδ2 + T cells; butyrophilin; cancer immunotherapy; tumor microenvironment
    DOI:  https://doi.org/10.1002/cbf.4081
  13. An Acad Bras Cienc. 2024 ;pii: S0001-37652024000200707. [Epub ahead of print]96(2): e20231212
      The tumor microenvironment (TME) harbors several cell types, such as tumor cells, immune cells, and non-immune cells. These cells communicate through several mechanisms, such as cell-cell contact, cytokines, chemokines, and extracellular vesicles (EVs). Tumor-derived vesicles are known to have the ability to modulate the immune response. Monocytes are a subset of circulating innate immune cells and play a crucial role in immune surveillance, being recruited to tissues where they differentiate into macrophages. In the context of tumors, it has been observed that tumor cells can attract monocytes to the TME and induce their differentiation into tumor-associated macrophages with a pro-tumor phenotype. Tumor-derived EVs have emerged as essential structures mediating this process. Through the transfer of specific molecules and signaling factors, tumor-derived EVs can shape the phenotype and function of monocytes, inducing the expression of cytokines and molecules by these cells, thus modulating the TME towards an immunosuppressive environment.
    DOI:  https://doi.org/10.1590/0001-3765202420231212
  14. Cancers (Basel). 2024 Jun 13. pii: 2211. [Epub ahead of print]16(12):
      Among professional antigen-presenting cells, dendritic cells (DCs) orchestrate innate and adaptive immunity and play a pivotal role in anti-tumor immunity. DCs are a heterogeneous population with varying functions in the tumor microenvironment (TME). Tumor-associated DCs differentiate developmentally and functionally into three main subsets: conventional DCs (cDCs), plasmacytoid DCs (pDCs), and monocyte-derived DCs (MoDCs). There are two major subsets of cDCs in TME, cDC1 and cDC2. cDC1 is critical for cross-presenting tumor antigens to activate cytotoxic CD8+ T cells and is also required for priming earlier CD4+ T cells in certain solid tumors. cDC2 is vital for priming anti-tumor CD4+ T cells in multiple tumor models. pDC is a unique subset of DCs and produces type I IFN through TLR7 and TLR9. Studies have shown that pDCs are related to immunosuppression in the TME through the secretion of immunosuppressive cytokines and by promoting regulatory T cells. MoDCs differentiate separately from monocytes in response to inflammatory cues and infection. Also, MoDCs can cross-prime CD8+ T cells. In this review, we summarize the subsets and functions of DCs. We also discuss the role of different DC subsets in shaping T cell immunity in TME and targeting DCs for potential immunotherapeutic benefits against cancer.
    Keywords:  antigen presentation; cancer; dendritic cells; tumor microenvironment; tumor-infiltrating T cells
    DOI:  https://doi.org/10.3390/cancers16122211
  15. Mol Oncol. 2024 Jun 22.
      Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T cells have demonstrated remarkable success in the treatment of relapsed/refractory melanoma and hematological malignancies, respectively. These treatments have marked a pivotal shift in cancer management. However, as "living drugs," their effectiveness is dependent on their ability to proliferate and persist in patients. Recent studies indicate that the mechanisms regulating these crucial functions, as well as the T cell's differentiation state, are conditioned by metabolic shifts and the distinct utilization of metabolic pathways. These metabolic shifts, conditioned by nutrient availability as well as cell surface expression of metabolite transporters, are coupled to signaling pathways and the epigenetic landscape of the cell, modulating transcriptional, translational, and post-translational profiles. In this review, we discuss the processes underlying the metabolic remodeling of activated T cells, the impact of a tumor metabolic environment on T cell function, and potential metabolic-based strategies to enhance T cell immunotherapy.
    Keywords:  T cells; anti‐tumor immunotherapy; chimeric antigen receptor; immunometabolism; nutrient transporters; tumor microenvironment
    DOI:  https://doi.org/10.1002/1878-0261.13691
  16. Discov Oncol. 2024 Jun 25. 15(1): 244
      The tumor microenvironment includes a complex network of immune T-cell subsets that play important roles in colorectal cancer (CRC) progression and are key elements of CRC immunotherapy. T cells develop and migrate within tumors, recognizing tumor-specific antigens to regulate immune surveillance. Current immunotherapies are divided into the following main categories based on the regulatory role of T-cell subsets in the tumor immune microenvironment (TIME): cytokines, monoclonal antibodies, peptide vaccines, CAR-T cells and more. This review describes the composition of the tumor immune microenvironment in colorectal cancer and the involvement of T cells in the pathogenesis and progression of CRC as well as current T-cell-related immunotherapies. Further studies on CRC-specific tumor antigens, the gene regulation of T cells, and the regulation of immune activity are needed.
    Keywords:  CAR-T; Colorectal cancer; Monoclonal antibody; T cell; Tumor immune microenvironment
    DOI:  https://doi.org/10.1007/s12672-024-01117-7
  17. J Control Release. 2024 Jun 26. pii: S0168-3659(24)00422-X. [Epub ahead of print]
      Tumor-associated macrophages (TAMs) constitute 50-80% of stromal cells in most solid tumors with high mortality and poor prognosis. Tumor-infiltrating dendritic cells (TIDCs) and TAMs are key components mediating immune responses within the tumor microenvironment (TME). Considering their refractory properties, simultaneous remodeling of TAMs and TIDCs is a potential strategy of boosting tumor immunity and restoring immunosurveillance. In this study, mannose-decorated poly(lactic-co-glycolic acid) nanoparticles loading with R848 (Man-pD-PLGA-NP@R848) were prepared to dually target TAMs and TIDCs for efficient tumor immunotherapy. The three-dimensional (3D) cell culture model can simulate tumor growth as influenced by the TME and its 3D structural arrangement. Consequently, cancer spheroids enriched with tumor-associated macrophages (TAMs) were fabricated to assess the therapeutic effectiveness of Man-pD-PLGA-NP@R848. In the TME, Man-pD-PLGA-NP@R848 targeted both TAMs and TIDCs in a mannose receptor-mediated manner. Subsequently, Man-pD-PLGA-NP@R848 released R848 to activate Toll-like receptors 7 and 8, following dual-reprograming of TIDCs and TAMs. Man-pD-PLGA-NP@R848 could uniquely reprogram TAMs into antitumoral phenotypes, decrease angiogenesis, reprogram the immunosuppressive TME from "cold tumor" into "hot tumor", with high CD4+ and CD8+ T cell infiltration, and consequently hinder tumor development in B16F10 tumor-bearing mice. Therefore, dual-reprograming of TIDCs and TAMs with the Man-pD-PLGA-NP@R848 is a promising cancer immunotherapy strategy.
    Keywords:  Hot tumor; Mannose receptor; Nanoparticles; Spheroid; Tumor-associated macrophages; cancer immunotherapy
    DOI:  https://doi.org/10.1016/j.jconrel.2024.06.062
  18. Cell Signal. 2024 Jun 25. pii: S0898-6568(24)00242-0. [Epub ahead of print] 111274
      Tumor-associated macrophages (TAMs) secrete cytokines, chemokines, and growth factors in the tumor microenvironment (TME) to support cancer progression. Higher TAM infiltration in the breast TME is associated with a poor prognosis. Previous studies have demonstrated the role of macrophages in stimulating long-range intercellular bridges referred to as tunneling nanotubes (TNTs) in cancer cells. Intercellular communication between cancer cells via TNTs promotes cancer growth, invasion, metastasis, and therapy resistance. Given the important role of TNTs and macrophages in cancer, the role of macrophage-induced TNTs in chemotherapy drug doxorubicin resistance is not known. Furthermore, the mechanism of macrophage-mediated TNT formation is elusive. In this study, it is shown that the macrophage-conditioned medium (MΦCM) partially mimicked inflammatory TME, induced an EMT phenotype, and increased migration in MCF-7 breast cancer cells. Additionally, secreted proteins in MΦCM induced TNT formation in MCF-7 cells, which led to increased resistance to doxorubicin. Transcriptomic analysis of MΦCM-treated MCF-7 cells showed enrichment of the NF-κB and focal adhesion pathways, as well as upregulation of genes involved in EMT, extracellular remodeling, and actin cytoskeleton reorganization. Interestingly, inhibitors of PKC, Src, NF-κB, and p38 decreased macrophage-induced TNT formation in MCF-7 cells. These results reveal the novel role of PKC and Src in inducing TNT formation in cancer cells and suggest that inhibition of PKC and Src activity may likely contribute to reduced macrophage-breast cancer cell interaction and the potential therapeutic strategy of cancer.
    Keywords:  Breast cancer; Macrophage-conditioned medium; Transcriptomics; Tumor microenvironment; Tunneling nanotubes
    DOI:  https://doi.org/10.1016/j.cellsig.2024.111274
  19. Front Pharmacol. 2024 ;15 1408993
      Ginsenosides, the primary bioactive ingredients derived from the root of Panax ginseng, are eagerly in demand for tumor patients as a complementary and alternative drug. Ginsenosides have increasingly become a "hot topic" in recent years due to their multifunctional role in treating colorectal cancer (CRC) and regulating tumor microenvironment (TME). Emerging experimental research on ginsenosides in the treatment and immune regulation of CRC has been published, while no review sums up its specific role in the CRC microenvironment. Therefore, this paper systematically introduces how ginsenosides affect the TME, specifically by enhancing immune response, inhibiting the activation of stromal cells, and altering the hallmarks of CRC cells. In addition, we discuss their impact on the physicochemical properties of the tumor microenvironment. Furthermore, we discuss the application of ginsenosides in clinical treatment as their efficacy in enhancing tumor patient immunity and prolonging survival. The future perspectives of ginsenoside as a complementary and alternative drug of CRC are also provided. This review hopes to open up a new horizon for the cancer treatment of Traditional Chinese Medicine monomers.
    Keywords:  colorectal cancer; ginsenosides; immunomodulation; panax ginseng; traditional Chinese medicine; tumor microenvironment
    DOI:  https://doi.org/10.3389/fphar.2024.1408993
  20. J Immunother Cancer. 2024 Jun 21. pii: e008917. [Epub ahead of print]12(6):
      BACKGROUND AND AIMS: The immunosuppressive tumor microenvironment (TME) plays an essential role in cancer progression and immunotherapy response. Despite the considerable advancements in cancer immunotherapy, the limited response to immune checkpoint blockade (ICB) therapies in patients with hepatocellular carcinoma (HCC) remains a major challenge for its clinical implications. Here, we investigated the molecular basis of the protein O-fucosyltransferase 1 (POFUT1) that drives HCC immune evasion and explored a potential therapeutic strategy for enhancing ICB efficacy.METHODS: De novo MYC/Trp53-/- liver tumor and the xenograft tumor models were used to evaluate the function of POFUT1 in immune evasion. Biochemical assays were performed to elucidate the underlying mechanism of POFUT1-mediated immune evasion.
    RESULTS: We identified POFUT1 as a crucial promoter of immune evasion in liver cancer. Notably, POFUT1 promoted HCC progression and inhibited T-cell infiltration in the xenograft tumor and de novo MYC/Trp53-/- mouse liver tumor models. Mechanistically, we demonstrated that POFUT1 stabilized programmed death ligand 1 (PD-L1) protein by preventing tripartite motif containing 21-mediated PD-L1 ubiquitination and degradation independently of its protein-O-fucosyltransferase activity. In addition, we further demonstrated that PD-L1 was required for the tumor-promoting and immune evasion effects of POFUT1 in HCC. Importantly, inhibition of POFUT1 could synergize with anti-programmed death receptor 1 therapy by remodeling TME in the xenograft tumor mouse model. Clinically, POFUT1 high expression displayed a lower response rate and worse clinical outcome to ICB therapies.
    CONCLUSIONS: Our findings demonstrate that POFUT1 functions as a novel regulator of tumor immune evasion and inhibition of POFUT1 may be a potential therapeutic strategy to enhance the efficacy of immune therapy in HCC.
    Keywords:  Hepatocellular Carcinoma; Immune Checkpoint Inhibitor; Tumor microenvironment - TME
    DOI:  https://doi.org/10.1136/jitc-2024-008917
  21. Bioconjug Chem. 2024 Jun 26.
      Cancer immunotherapy has yielded remarkable results across a variety of tumor types. Nevertheless, the complex and immunosuppressive microenvironment within solid tumors poses significant challenges to established therapies such as immune checkpoint blockade (ICB) and chimeric antigen receptor T-cell (CAR-T) therapy. Within the milieu, tumor-associated macrophages (TAMs) play a significant role by directly suppressing T-cell functionality and fostering an immunosuppressive environment. Effective regulation of TAMs is, therefore, crucial to enhancing the efficacy of immunotherapies. Various therapeutic strategies targeting TAM modulation have emerged, including blocking TAM recruitment, direct elimination, promoting repolarization toward the M1 phenotype, and enhancing phagocytic capacity against tumor cells. The recently introduced CAR macrophage (CAR-M) therapy opens new possibilities for macrophage-based immunotherapy. Compared with CAR-T, CAR-M may demonstrate superior targeting and infiltration capabilities toward solid tumors. This review predominantly delves into the origin and development process of TAMs, their role in promoting tumor growth, and provides a comprehensive overview of immunotherapies targeting TAMs. It underscores the significance of regulating TAMs in bolstering antitumor therapies while discussing the potential and challenges of developing TAMs as targets for immunotherapy.
    DOI:  https://doi.org/10.1021/acs.bioconjchem.4c00242
  22. Int Immunopharmacol. 2024 Jun 26. pii: S1567-5769(24)00802-6. [Epub ahead of print]138 112282
      Hypoxia is a hallmark of solid tumors. Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment, and CAF-derived exosomes are involved in cancer genesis and progression. Here, this work investigated the role and mechanism of exosomal circHIF1A derived from hypoxia-induced CAFs in hepatocellular carcinoma (HCC) tumorigenesis. CAFs isolated from fresh HCC tissues were incubated in normoxia or hypoxia condition (N/CAFs or H/CAFs), and then the exosomes from N/CAFs or H/CAFs were isolated for functional analysis. Cell proliferation, migration and invasion were analyzed by cell counting kit-8, colony formation, and transwell assays. Immune evasion was evaluated by measuring the cytotoxicity and viability of CD8+T cells. qRT-PCR and western blotting analyses were used for the level measurement of genes and proteins. The binding between Hu antigen R (HuR) and circHIF1A or Programmed death ligand 1 (PD-L1) was analyzed by RNA immunoprecipitation assay. Functionally, we found that CAFs, especially CAFs under hypoxic stress (H/CAFs), promoted the proliferation, migration, invasion and EMT progression in HCC cells, as well as induced immune escape by suppressing CD8+T cell cytotoxicity and activity in an exosome-dependent manner. H/CAFs-derived exosomes showed highly expressed circHIF1A, and could secrete circHIF1A into HCC cells via exosomes. The oncogenic effects of H/CAFs-secreted exosomes were abolished by circHIF1A knockdown. Mechanistically, circHIF1A interacted with HuR to stabilize PD-L1 expression in HCC cells. Meanwhile, circHIF1A silencing suppressed HCC cell proliferation, mobility and immune escape by regulating PD-L1 expression. In all, exosomal circHIF1A derived from hypoxic-induced CAFs promoted the proliferation, migration, invasion, EMT progression and immune escape in HCC cells by up-regulating PD-L1 expression in a HuR-dependent manner.
    Keywords:  CD8(+)T cells; Exosomes; HCC; HuR; PD-L1; Proliferation
    DOI:  https://doi.org/10.1016/j.intimp.2024.112282
  23. Int J Mol Sci. 2024 Jun 13. pii: 6501. [Epub ahead of print]25(12):
      Breast cancer poses a global health challenge, yet the influence of ethnicity on the tumor microenvironment (TME) remains understudied. In this investigation, we examined immune cell infiltration in 230 breast cancer samples, emphasizing diverse ethnic populations. Leveraging tissue microarrays (TMAs) and core samples, we applied multiplex immunofluorescence (mIF) to dissect immune cell subtypes across TME regions. Our analysis revealed distinct immune cell distribution patterns, particularly enriched in aggressive molecular subtypes triple-negative and HER2-positive tumors. We observed significant correlations between immune cell abundance and key clinicopathological parameters, including tumor size, lymph node involvement, and patient overall survival. Notably, immune cell location within different TME regions showed varying correlations with clinicopathologic parameters. Additionally, ethnicities exhibited diverse distributions of cells, with certain ethnicities showing higher abundance compared to others. In TMA samples, patients of Chinese and Caribbean origin displayed significantly lower numbers of B cells, TAMs, and FOXP3-positive cells. These findings highlight the intricate interplay between immune cells and breast cancer progression, with implications for personalized treatment strategies. Moving forward, integrating advanced imaging techniques, and exploring immune cell heterogeneity in diverse ethnic cohorts can uncover novel immune signatures and guide tailored immunotherapeutic interventions, ultimately improving breast cancer management.
    Keywords:  B cells; FOXP3; T-helper cells; TAMs; TILs; Treg cells; ethnic breast cancer; multiplex immunofluorescence; multispectral imaging; tumor microenvironment
    DOI:  https://doi.org/10.3390/ijms25126501
  24. Expert Opin Biol Ther. 2024 Jun 24. 1-13
      INTRODUCTION: Growing attention has been drawn to urologic tumors due to their rising incidence and suboptimal clinical treatment outcomes. Cancer therapy resistance poses a significant challenge in clinical oncology, limiting the efficacy of conventional treatments and contributing to disease progression. Recent research has unveiled a complex interplay between the host microbiota and cancer cells, highlighting the role of the microbiota in modulating therapeutic responses.AREAS COVERED: We used the PubMed and Web of Science search engines to identify key publications in the fields of tumor progression and urologic tumor treatment, specifically focusing on the role of the microbiota. In this review, we summarize the current literature on how microbiota influence the tumor microenvironment and anti-tumor immunity, as well as their impact on treatments for urinary system malignancies, highlighting promising future applications.
    EXPERT OPINION: We explore how the composition and function of the gut microbiota influence the tumor microenvironment and immune response, ultimately impacting treatment outcomes. Additionally, we discuss emerging strategies targeting the microbiota to enhance therapeutic efficacy and overcome resistance. The application of antibiotics, fecal microbiota transplantation, and oncolytic bacteria has improved tumor treatment outcomes, which provides a novel insight into developing therapeutic strategies for urologic cancer.
    Keywords:  Gut microbiota; intratumoral microbiota; therapy resistance; tumor microenvironment; urologic cancer
    DOI:  https://doi.org/10.1080/14712598.2024.2371543
  25. J Hepatocell Carcinoma. 2024 ;11 1143-1156
      Hepatocellular carcinoma (HCC) stands as the prevailing form of primary liver cancer, characterized by a poor prognosis and high mortality rate. A pivotal factor in HCC tumorigenesis is epigenetics, specifically the regulation of gene expression through methylation. This process relies significantly on the action of proteins that modify methylation, including methyltransferases, their associated binding proteins, and demethylases. These proteins are crucial regulators, orchestrating the methylation process by regulating enzymes and their corresponding binding proteins. This orchestration facilitates the reading, binding, detection, and catalysis of gene methylation sites. Methylation ences the development, prolisignificantly influferation, invasion, and prognosis of HCC. Furthermore, methylation modification and its regulatory mechanisms activate distinct biological characteristics in HCC cancer stem cells, such as inducing cancer-like differentiation of stem cells. They also influence the tumor microenvironment (TME) in HCC, modulate immune responses, affect chemotherapy resistance in HCC patients, and contribute to HCC progression through signaling pathway feedback. Given the essential role of methylation in genetic information, it holds promise as a potential tool for the early detection of HCC and as a target to improve drug resistance and promote apoptosis in HCC cells.
    Keywords:  HCC tumorigenesis; biomarkers; methylation modifying proteins; methyltransferases
    DOI:  https://doi.org/10.2147/JHC.S458734
  26. Int J Mol Sci. 2024 Jun 09. pii: 6383. [Epub ahead of print]25(12):
      T lymphocytes play a critical role in antitumor immunity, but their exhaustion poses a significant challenge for immune evasion by malignant cells. Circular RNAs (circRNAs), characterized by their covalently closed looped structure, have emerged as pivotal regulators within the neoplastic landscape. Recent studies have highlighted their multifaceted roles in cellular processes, including gene expression modulation and protein function regulation, which are often disrupted in cancer. In this review, we systematically explore the intricate interplay between circRNAs and T cell modulation within the tumor microenvironment. By dissecting the regulatory mechanisms through which circRNAs impact T cell exhaustion, we aim to uncover pathways crucial for immune evasion and T cell dysfunction. These insights can inform innovative immunotherapeutic strategies targeting circRNA-mediated molecular pathways. Additionally, we discuss the translational potential of circRNAs as biomarkers for therapeutic response prediction and as intervention targets. Our comprehensive analysis aims to enhance the understanding of immune evasion dynamics in the tumor microenvironment by facilitating the development of precision immunotherapy.
    Keywords:  T cells; cancer; circular RNAs; immune checkpoint inhibitors; tumor microenvironment
    DOI:  https://doi.org/10.3390/ijms25126383
  27. Cancers (Basel). 2024 Jun 12. pii: 2201. [Epub ahead of print]16(12):
      The tumor immune microenvironment is pivotal in cancer initiation, advancement, and regulation. Its molecular and cellular composition is critical throughout the disease, as it can influence the balance between suppressive and cytotoxic immune responses within the tumor's vicinity. Studies on the tumor immune microenvironment have enriched our understanding of the intricate interplay between tumors and their immunological surroundings in various human cancers. These studies illuminate the role of significant components of the immune microenvironment, which have not been extensively explored in pediatric tumors before and may influence the responsiveness or resistance to therapeutic agents. Our deepening understanding of the pediatric tumor immune microenvironment is helping to overcome challenges related to the effectiveness of existing therapeutic strategies, including immunotherapies. Although in the early stages, targeted therapies that modulate the tumor immune microenvironment of pediatric solid tumors hold promise for improved outcomes. Focusing on various aspects of tumor immune biology in pediatric patients presents a therapeutic opportunity that could improve treatment outcomes. This review offers a comprehensive examination of recent literature concerning profiling the immune microenvironment in various pediatric tumors. It seeks to condense research findings on characterizing the immune microenvironment in pediatric tumors and its impact on tumor development, metastasis, and response to therapeutic modalities. It covers the immune microenvironment's role in tumor development, interactions with tumor cells, and its impact on the tumor's response to immunotherapy. The review also discusses challenges targeting the immune microenvironment for pediatric cancer therapies.
    Keywords:  immune microenvironment; immune suppression; immunotherapy; metastasis; pediatric cancers; tumor-immune biology
    DOI:  https://doi.org/10.3390/cancers16122201
  28. Integr Med (Encinitas). 2024 May;23(2): 24-35
      Tumor microenvironment infiltration by cells of the T helper cell type 1 (TH1) system, including TH1 cells, M1 macrophages, natural killer cells, and CD8+ T cells, is associated with better cancer prognosis. In contrast, tumor microenvironment infiltration by cells of the TH2 system, including TH2 cells, M2 macrophages, and innate lymphoid cells type 2, as well as immune suppressive myeloid-derived suppressor cells and regulatory T cells, is associated with poorer cancer prognosis. Beyond the tumor itself and a myriad of other modifying factors, such as genetic and epigenetic influences on tumorigenesis, the overall immune state of the patient, termed the macroenvironment, has also been shown to significantly influence cancer outcomes. Alterations in the tricarboxylic acid (TCA) cycle (TCA cycle breaks) involving loss of function of succinate dehydrogenase, isocitrate dehydrogenase, and fumarate hydratase have been shown to be associated with an intracellular metabolic shift away from oxidative phosphorylation and into glycolysis in cells that are transforming into cancer cells. The same loss of function of succinate dehydrogenase and isocitrate dehydrogenase has also been identified as inducing a shift in macrophages toward glycolysis that is associated with M1 macrophage polarization. M1 macrophages make interleukin 12, which stimulates TH1 cells and natural killer cells to produce interferon gamma (IFN-γ), which in turn stimulates M1 macrophage activity, forming an activation loop. IFN-γ also drives activation of CD8+ T cells. Thus, M1 macrophage activation initiates and sustains activation of the TH1 system of cells. In this fashion, TCA cycle breaks at succinate dehydrogenase and isocitrate dehydrogenase that promote cellular transformation into cancer cells are also associated with upregulation of the TH1 system that provides anti-cancer immune surveillance. The TH1 and TH2 systems are known to inhibit each other's activation. It is this author's hypothesis that, in patients whose macroenvironment is sufficiently TH2-dominant, the metabolic shift toward glycolysis induced by TCA cycle breaks that gives rise to mutagenic changes in tissue parenchymal cells is not counterbalanced by adequate activation of M1 macrophages, thus giving rise to cancer cell development. For instance, the atopic TH2-high asthma phenotype, a TH2 dominance-based comorbidity, is associated with a more than doubled incidence of colon, breast, lung, and prostate cancer, compared with non-asthmatics. Failure of TCA cycle breaks to induce M1 polarization of tissue-resident macrophages yields a tissue environment in which the tissue-resident macrophages fail to routinely perform M1-associated functions such as phagocytizing newly developing cancer cells. Failure of M1 phenotypic expression in both tissue-resident macrophages and monocyte-derived macrophages recruited to the tumor microenvironment yields both a loss of direct antitumor M1 macrophage actions and failure of TH1 system activation in general, including failure of CD8+ T cell activation, yielding a cancer-permissive tumor microenvironment and a poorer prognosis in patients with existing cancers. This paper proposes a conceptual framework that connects established elements in the existing research and points to the utility of a patient profiling process, aimed at personalization of treatment through identification and targeting of elements in each patient's tumor microenvironment and macroenvironment that contribute to unfavorable prognosis.
  29. Int J Oncol. 2024 Aug;pii: 79. [Epub ahead of print]65(2):
      Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
    Keywords:  cancer; chronic inflammation; leptin; myeloid‑derived suppressor cells; obesity; tumor
    DOI:  https://doi.org/10.3892/ijo.2024.5667
  30. Cancer Lett. 2024 Jun 25. pii: S0304-3835(24)00474-9. [Epub ahead of print] 217079
      Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
    DOI:  https://doi.org/10.1016/j.canlet.2024.217079