bims-stacyt Biomed News
on Paracrine crosstalk between cancer and the organism
Issue of 2021‒10‒31
nine papers selected by
Cristina Muñoz Pinedo
L’Institut d’Investigació Biomèdica de Bellvitge


  1. Vaccines (Basel). 2021 Oct 14. pii: 1178. [Epub ahead of print]9(10):
      Leukocytes often undergo rapid changes in cell phenotype, for example, from a resting to an activated state, which places significant metabolic demands on the cell. These rapid changes in metabolic demand need to be tightly regulated to support immune cell effector functions during the initiation and downregulation of an immune response. Prospects for implementing cancer immunotherapy also rest on the idea of optimizing the metabolic profile of immune cell effectors. Here, we examine this issue by focusing on neutrophils and NK cells as cells of increasing interest in cancer immunology and tumor immunometabolism, because they can be targeted or, in the case of NK, used as effectors in immunotherapy. In addition, neutrophils and NK cells have been shown to functionally interact. In the case of neutrophils, we also extended our interest to polymorphonuclear MDSC (PMN-MDSCs), since the granulocytic subset of MDSCs share many phenotypes and are functionally similar to pro-tumor neutrophils. Finally, we reviewed relevant strategies to target tumor metabolism, focusing on neutrophils and NK cells.
    Keywords:  PMN-MDSCs; natural killer cells; neutrophils; tumor metabolism; tumor microenvironment
    DOI:  https://doi.org/10.3390/vaccines9101178
  2. Cancer Biol Ther. 2021 Oct 25. 1-20
      Tumor metastasis is the leading cause of cancer mortality, often characterized by abnormal cell growth and invasion to distant organs. The cancer invasion due to epithelial to mesenchymal transition is affected by metabolic and oxygen availability in the tumor-associated micro-environment. A precise alteration in oxygen and metabolic signaling between healthy and metastatic cells is a substantial probe for understanding tumor progression and metastasis. Molecular heterogeneity in the tumor microenvironment help to sustain the metastatic cell growth during their survival shift from low to high metabolic-oxygen-rich sites and reinforces the metastatic events. This review highlighted the crucial role of oxygen and metabolites in metastatic progression and exemplified the role of metabolic rewiring and oxygen availability in cancer cell adaptation. Furthermore, we have also addressed potential applications of altered oxygen and metabolic networking with tumor type that could be a signature pattern to assess tumor growth and chemotherapeutics efficacy in managing cancer metastasis.
    Keywords:  Cancer metabolism; angeogenesis; cancer recurrence; dormancy; lymphogenesis; metabolic reprogramming; metastasis
    DOI:  https://doi.org/10.1080/15384047.2021.1992233
  3. Nat Commun. 2021 Oct 28. 12(1): 6218
      Cytotoxic chemotherapeutics primarily function through DNA damage-induced tumor cell apoptosis, although the inflammation provoked by these agents can stimulate anti-cancer immune responses. The mechanisms that control these distinct effects and limit immunogenic responses to DNA-damage mediated cell death in vivo are currently unclear. Using a mouse model of BCR-ABL+ B-cell acute lymphoblastic leukemia, we show that chemotherapy-induced anti-cancer immunity is suppressed by the tumor microenvironment through production of the cytokine IL-6. The chemotherapeutic doxorubicin is curative in IL-6-deficient mice through the induction of CD8+ T-cell-mediated anti-cancer responses, while moderately extending lifespan in wild type tumor-bearing mice. We also show that IL-6 suppresses the effectiveness of immune-checkpoint inhibition with anti-PD-L1 blockade. Our results suggest that IL-6 is a key regulator of anti-cancer immune responses induced by genotoxic stress and that its inhibition can switch cancer cell clearance from primarily apoptotic to immunogenic, promoting and maintaining durable anti-tumor immune responses.
    DOI:  https://doi.org/10.1038/s41467-021-26407-4
  4. Annu Rev Med. 2021 Oct 26.
      In order to fuel their relentless expansion, cancers must expand their vasculature to augment delivery of oxygen and essential nutrients. The disordered web of irregular vessels that results, however, leaves gaps in oxygen delivery that foster tumor hypoxia. At the same time, tumor cells increase their oxidative metabolism to cope with the energetic demands of proliferation, which further worsens hypoxia due to heightened oxygen consumption. In these hypoxic, nutrient-deprived environments, tumors and suppressive stroma evolve to flourish while antitumor immunity collapses due to a combination of energetic deprivation, toxic metabolites, acidification, and other suppressive signals. Reversal of cancer hypoxia thus has the potential to increase the survival and effector function of tumor-infiltrating T cells, as well as to resensitize tumors to immunotherapy. Early clinical trials combining hypoxia reduction with immune checkpoint blockade have shown promising results in treating patients with advanced, metastatic, and therapeutically refractory cancers. Expected final online publication date for the Annual Review of Medicine, Volume 73 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
    DOI:  https://doi.org/10.1146/annurev-med-060619-022830
  5. Front Physiol. 2021 ;12 722528
      Little is known about the effects of training load on exercise-induced plasma increase of interleukin-6 (IL-6) and soluble IL-6 receptor (sIL-6R) and their relationship with vascular remodeling. We sought to evaluate the role of sIL 6R as a regulator of IL-6-induced vascular remodeling. Forty-four male marathon runners were recruited and allocated into two groups: low-training (LT, <100 km/week) and high-training (HT, ≥100 km/week), 22 athletes per group. Twenty-one sedentary participants were used as reference. IL-6, sIL-6R and sgp130 levels were measured in plasma samples obtained before and immediately after finishing a marathon (42.2-km). Aortic diameter was measured by echocardiography. The inhibitory effect of sIL-6R on IL-6-induced VSMC migration was assessed using cultured A7r5 VSMCs. Basal plasma IL-6 and sIL-6R levels were similar among sedentary and athlete groups. Plasma IL-6 and sIL-6R levels were elevated after the marathon, and HT athletes had higher post-race plasma sIL-6R, but not IL-6, level than LT athletes. No changes in sgp130 plasma levels were found in LT and HT groups before and after running the marathon. Athletes had a more dilated ascending aorta and aortic root than sedentary participants with no differences between HT and LT athletes. However, a positive correlation between ascending aorta diameter and plasma IL-6 levels corrected by training load and years of training was observed. IL-6 could be responsible for aorta dilation because IL-6 stimulated VSMC migration in vitro, an effect that is inhibited by sIL-6R. However, IL-6 did not modify cell proliferation, collagen type I and contractile protein of VSMC. Our results suggest that exercise induces vascular remodeling. A possible association with IL-6 is proposed. Because sIL-6R inhibits IL-6-induced VSMC migration, a possible mechanism to regulate IL-6-dependent VSMC migration is also proposed.
    Keywords:  exercise; interleukin-6; runners; soluble interleukin-6 receptor; vascular smooth muscle cell
    DOI:  https://doi.org/10.3389/fphys.2021.722528
  6. Pharm Biol. 2021 Dec;59(1): 1473-1479
      CONTEXT: Icariin (ICA), a flavonol glycoside extracted from Epimedium brevicornum Maxim (Berberidaceae), has been proven to inhibit inflammatory response in ischaemic rats in our laboratory's previous work. However, its underlying mechanism is still unclear.OBJECTIVE: This study investigates the effects of ICA on endoplasmic reticulum (ER) stress mediated inflammation induced by cerebral ischaemia-reperfusion (I/R) injury in vitro.
    MATERIALS AND METHODS: The primary cultured microglia were treated with oxygen-glucose deprivation (OGD) for 2 h followed by a 24 h reoxygenation. ICA (0.37, 0.74 and 1.48 μmol/L) administration was performed 1 h prior OGD and acting through 2 h OGD. The control group was cultured in normal conditions. At 24 h after reoxygenation, the expression of IRE1α, XBP1u, XBP1s, NLRP3 and caspase-1 was detected by western blotting (WB) and quantitative real-time (qRT) PCR; the expression of p-IRE1α was examined by WB; the expression of IL-1β, IL-6 and TNF-α was measured by WB and enzyme-linked immunosorbent assay (ELISA).
    RESULTS: ICA (0.37, 0.74 and 1.48 μmol/L) reduced the ratio of p-IRE1α/IRE1α, the mRNA level of IRE1α, the expression of XBP1u, XBP1s, NLRP3, caspase-1 at both the mRNA and protein level expression of IL-1β, IL-6 and TNF-α in OGD/R injured microglia. Overexpression of IRE1 significantly reversed the effects of ICA.
    DISCUSSION AND CONCLUSIONS: These results suggested that ICA might decrease the expression of IL-1β, IL-6 and TNF-α by inhibiting IRE1/XBP1s pathway. The anti-inflammatory effect of ICA may provide a potential therapeutic strategy for the treatment of brain injury after stroke.
    Keywords:  IRE1; Microglia; inflammation; oxygen-glucose deprivation
    DOI:  https://doi.org/10.1080/13880209.2021.1991959
  7. Cancer Immunol Res. 2021 Oct 29.
      T cells are the key players in eliminating malignant tumors. Adoptive transfer of tumor antigen-specific T cells and immune checkpoint blockade has yielded durable antitumor responses in the clinic, but not all patients respond initially and some that do respond eventually have tumor progression. Thus, new approaches to enhance the utility of immunotherapy are needed. T-cell activation and differentiation status are tightly controlled at the transcriptional, epigenetic, and metabolic levels. Amino acids are involved in multiple steps of T-cell antitumor immunity, including T-cell activation, proliferation, effector function, memory formation as well as functional exhaustion. In this review, we briefly discuss how amino acid metabolism is linked to T-cell fate decisions and summarize how amino acid deprivation or accumulation of certain amino acid metabolites within the tumor microenvironment diminishes T-cell functionality. Furthermore, we discuss potential strategies for immunotherapy via modulating amino acid metabolism either in T cells intrinsically or extrinsically to achieve therapeutic efficacy.
    DOI:  https://doi.org/10.1158/2326-6066.CIR-21-0459
  8. EMBO Rep. 2021 Oct 26. e52509
      Aneuploidy is a chromosomal abnormality associated with poor prognosis in many cancer types. Here, we tested the hypothesis that the unfolded protein response (UPR) mechanistically links aneuploidy and local immune dysregulation. Using a single somatic copy number alteration (SCNA) score inclusive of whole-chromosome, chromosome arm, and focal alterations in a pan-cancer analysis of 9,375 samples in The Cancer Genome Atlas (TCGA) database, we found an inverse correlation with a cytotoxicity (CYT) score across disease stages. Co-expression patterns of UPR genes changed substantially between SCNAlow and SCNAhigh groups. Pathway activity scores showed increased activity of multiple branches of the UPR in response to aneuploidy. The PERK branch showed the strongest association with a reduced CYT score. The conditioned medium of aneuploid cells transmitted XBP1 splicing and caused IL-6 and arginase 1 transcription in receiver bone marrow-derived macrophages and markedly diminished the production of IFN-γ and granzyme B in activated human T cells. We propose the UPR as a mechanistic link between aneuploidy and immune dysregulation in the tumor microenvironment.
    Keywords:  T cells; aneuploidy; macrophages; tumor immune microenvironment; unfolded protein response
    DOI:  https://doi.org/10.15252/embr.202152509
  9. Int J Radiat Oncol Biol Phys. 2021 Nov 01. pii: S0360-3016(21)01080-4. [Epub ahead of print]111(3S): S88-S89
      PURPOSE/OBJECTIVE(S): Tumor irradiation causes pro-inflammatory changes in the tumor microenvironment (TME). We have shown that these changes may be countered by the induction of immunosuppressive changes that drive therapy resistance. Here, we test the hypothesis that radiation activates adenosine signaling and induces immunosuppressive myeloid cells in the TME.MATERIALS/METHODS: Using the 4T1 orthotopic breast cancer mouse model, we tested immunophenotypic changes to the TME by spectral flow cytometry and by bulk RNASeq of tumor infiltrating leukocytes (CD45+, TILs) 72-hours after tumor irradiation (8 Gy x 1) using the small animal radiation research platform (SARRP, Xstrahl; 220 kV). Human peripheral blood mononuclear cells (PBMCs) were cultured in vitro with 0.1, 1, and 10uM of adenosine receptor agonist, 5'-N-ethylcarboxamindoadenosine (NECA), in the presence of CD3/CD28 beads. Cytokine quantification of cell-free supernatant was performed 48-hours later using the Meso Scale Discovery platform.
    RESULTS: Radiation increases the abundance of CD11b+ myeloid cells (77.4% vs. 84.4%) and decreases TCRb+ lymphocytes (6.3% vs. 3.6%) in the TME. The reduction of lymphocytes was driven by CD4+ T-cells (5.2% vs 2.7%), including a proportional reduction in Tregs (1.4% vs 0.63%), with no change in abundance of CD8+ T-cells (0.53% vs 0.43%). The increase in myeloid cells was predominantly influenced by granulocytic- and monocytic-myeloid-derived suppressor cells (PMN-MDSCs: 22% vs 34.5%; M-MSCSs: 3.0% vs 10.0%). By bulk RNASeq of CD45+ TILs, radiation increased the expression of adenosine 2A receptor (A2AR, log2 fold change 1.13) and A2BR (log2 fold change 0.92), but not CD73 - an ecto-nucleotidase that metabolizes AMP to adenosine. By flow cytometry, radiation increased expression of both CD73 and A2AR in the myeloid population, with CD73 expression significantly increased in the monocytic compartment (Ly6C+, 63% vs 70%) and A2AR expression increased in the granulocytic compartment (Ly6G, 89% vs 97%). The addition of adenosine analog, NECA, to human PBMCs resulted in significant reduction in effector T-cell cytokines (IL-2, IFN- γ, granzyme B) across doses, an increase in IL-6 and MCP-1/CCL2 at the intermediate dose tested, and increased IFN-β and TGF- β while decreasing IL-1 β, IL-8 only at the highest dose.
    CONCLUSION: Radiation induces an immunosuppressive TME, increasing the abundance of PMN-MDSCs and M-MDSCs and activating adenosine signaling. Radiation increases the expression of adenosine receptors on CD45+ TILs at the transcriptomic level and cell surface expression of CD73 on monocytes and A2AR on granulocytes. Treatment of human PBMCs with adenosine receptor agonist resulted in loss of effector T-cell function and suppressive changes to cytokine milieu. These data suggest a mechanistic link between radiation, adenosine signaling and the myeloid compartment.
    DOI:  https://doi.org/10.1016/j.ijrobp.2021.07.210