[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Tumor Microenvironment and Molecular Aberrations Convey Immune Evasion

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Immunology and Immunotherapy".

Deadline for manuscript submissions: closed (31 May 2023) | Viewed by 25283

Special Issue Editor


E-Mail Website
Guest Editor
Department of Medical and Molecular Sciences, College of Health Sciences, University of Delaware, Newark, DE 19716, USA
Interests: DNA methylation; cancer invasion and metastasis in breast and pancreatic cancers; carcinoma-associated fibroblasts; epithelial–mesenchymal transition; AKT signaling pathway; tumor microenvironment and immune evasion
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

It was widely recognized that the complex interplay between immunity and cancer determines whether cancer cells will survive or be destroyed. The battle between tumoricidal and tumor promoting activity relies on the extent to which the antitumor immune response is exerted. In general, immune evasive mechanisms adapted by cancers encompass downregulation of antigen presentations or recognition, lack of immune effector cells, obstruction of antitumor immune cell maturation, accumulation of immunosuppressive cells, production of inhibitory cytokines, chemokines or ligands/receptors, establishment of a hypoxic tumor microenvironment, development of cancer-promoting metabolisms, and upregulation of immune checkpoint modulators. As such, restoring or stimulating tumoricidal effects, in conjunction with surgical resection, as well as chemo- or radiation-mediated, hormone-based, kinase-targeted, DNA repair-disrupted, small molecule inhibitor-mediated, signal transduction pathway-modified, aberrant epigenome-reverted, and cytokine-involved treatments, may ignite promising therapeutic regiments to eradicate fatal cancers. This Special Issue welcomes papers outlining mechanisms of immune escape or depicting strategies for boosting immune surveillance, in synergy with additional non-immune interventions to eradicate human cancers.

Dr. Huey-Jen Lin
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • CD8+ tumor-infiltrating lymphocytes
  • cytotoxic T lymphocytes -associated protein 4
  • dendritic cells
  • immune evasion
  • hypoxia-inducible factors
  • myeloid-derived suppressor cells
  • natural killer
  • programmed death receptor and ligand
  • regulatory T cells
  • tumor-associated macrophages
  • tumor microenvironment

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

11 pages, 975 KiB  
Article
In Patients Treated by Selective Internal Radiotherapy, Cellular In Vitro Immune Function Is Predictive of Survival
by Aglaia Domouchtsidou, Ferdinand Beckmann, Beate Marenbach, Stefan P. Mueller, Jan Best, Ken Herrmann, Peter A. Horn, Vahé Barsegian and Monika Lindemann
Cancers 2023, 15(16), 4055; https://doi.org/10.3390/cancers15164055 - 11 Aug 2023
Viewed by 1376
Abstract
In patients with liver malignancies, the cellular immune function was impaired in vitro after selective internal radiotherapy (SIRT). Because immunosuppression varied substantially, in the current study, we investigated in 25 SIRT patients followed up for ten years whether the lymphocyte function was correlated [...] Read more.
In patients with liver malignancies, the cellular immune function was impaired in vitro after selective internal radiotherapy (SIRT). Because immunosuppression varied substantially, in the current study, we investigated in 25 SIRT patients followed up for ten years whether the lymphocyte function was correlated with survival. Peripheral blood mononuclear cells were stimulated with four microbial antigens (tuberculin, tetanus toxoid, Candida albicans and CMV) before therapy and at four time points thereafter, and lymphocyte proliferation was determined by H3-thymidine uptake. The median sum of the responses to these four antigens decreased from 39,464 counts per minute (CPM) increment (range 1080–204,512) before therapy to a minimum of 700 CPM increment on day 7 after therapy (0–93,187, p < 0.0001). At all five time points, the median survival in patients with weaker responses was 2- to 3.5-fold shorter (p < 0.05). On day 7, the median survival in patients with responses below and above the cutoff of a 2 CPM increment was 185 and 523 days, respectively (χ2 = 9.4, p = 0.002). In conclusion, lymphocyte function could be a new predictor of treatment outcome after SIRT. Full article
Show Figures

Figure 1

Figure 1
<p><b>Cellular response towards four microbial antigens in 25 patients treated by selective internal radiotherapy (SIRT).</b> Panel (<b>a</b>) shows individual data prior to SIRT and one hour and on day 2, 7 and 28 thereafter, together with median and interquartile range. Red dots indicate values for patients with a short survival (&lt;median of 369 days), and green dots those for patients with a long survival (≥median). Data were compared by 1-way ANOVA, and the comparison of data from all test points yielded a <span class="html-italic">p</span> value of &lt;0.0001. <span class="html-italic">p</span> values in panel (<b>a</b>) indicate the significance of pairwise comparisons, corrected for multiple comparisons. Panel (<b>b</b>–<b>f</b>) indicate the results of a receiver operating characteristic (ROC) curve analysis of the cellular response towards four microbial antigens and of survival (&lt;median vs. ≥median). Each of these panels provides information on area under the curve (AUC), cutoff, sensitivity, specificity and likelihood ratio. On day 7 (panel (<b>e</b>)), we considered two different cutoffs for the cumulative (cum.) antigen response, as we observed two maxima for the likelihood ratio. CPM incr., counts-per-minute increment, i.e., antigen-specific response minus the negative control.</p>
Full article ">Figure 2
<p><b>Impact of the cumulative antigen response on the survival of 25 patients treated by selective internal radiotherapy (SIRT).</b> Panel (<b>a</b>) shows the survival curves with respect to the cumulative (cum.) antigen response (towards four microbial antigens) prior to SIRT, and panel (<b>b</b>–<b>f</b>) show curves after SIRT. Please note that the analysis on day 7 after SIRT was performed with two different cutoff values, as explained in the legend to <a href="#cancers-15-04055-f001" class="html-fig">Figure 1</a><b>e</b>. Survival curves were compared by the Log Rank (Mantel–Cox) test. CPM increment, counts-per-minute increment, i.e., antigen-specific response minus the negative control.</p>
Full article ">
17 pages, 2321 KiB  
Article
Nigericin Boosts Anti-Tumor Immune Response via Inducing Pyroptosis in Triple-Negative Breast Cancer
by Lisha Wu, Shoumin Bai, Jing Huang, Guohui Cui, Qingjian Li, Jingshu Wang, Xin Du, Wenkui Fu, Chuping Li, Wei Wei, Huan Lin and Man-Li Luo
Cancers 2023, 15(12), 3221; https://doi.org/10.3390/cancers15123221 - 16 Jun 2023
Cited by 7 | Viewed by 3114
Abstract
Although immune checkpoint inhibitors improved the clinical outcomes of advanced triple negative breast cancer (TBNC) patients, the response rate remains relatively low. Nigericin is an antibiotic derived from Streptomyces hydrophobicus. We found that nigericin caused cell death in TNBC cell lines MDA-MB-231 [...] Read more.
Although immune checkpoint inhibitors improved the clinical outcomes of advanced triple negative breast cancer (TBNC) patients, the response rate remains relatively low. Nigericin is an antibiotic derived from Streptomyces hydrophobicus. We found that nigericin caused cell death in TNBC cell lines MDA-MB-231 and 4T1 by inducing concurrent pyroptosis and apoptosis. As nigericin facilitated cellular potassium efflux, we discovered that it caused mitochondrial dysfunction, leading to mitochondrial ROS production, as well as activation of Caspase-1/GSDMD-mediated pyroptosis and Caspase-3-mediated apoptosis in TNBC cells. Notably, nigericin-induced pyroptosis could amplify the anti-tumor immune response by enhancing the infiltration and anti-tumor effect of CD4+ and CD8+ T cells. Moreover, nigericin showed a synergistic therapeutic effect when combined with anti-PD-1 antibody in TNBC treatment. Our study reveals that nigericin may be a promising anti-tumor agent, especially in combination with immune checkpoint inhibitors for advanced TNBC treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Nigericin-treated TNBC cells exhibit both pyroptotic and apoptotic features. (<b>A</b>) Human TNBC cell line MDA-MB-231 and mouse TNBC cell line 4T1 were exposed to increasing concentrations of nigericin (0, 0.25, 0.5, 1, 2, 5, 10 and 20 μg/mL) for 24 h. Cell viability was measured using MTS assays, and IC50 value was calculated based on cell viability curve. Displayed are mean ± SD relative to control. (<b>B</b>) Flow cytometry analysis of PI and Annexin V stained TNBC cells before and after nigericin treatment (nigericin 2 μg/mL for 12 h, Annexin V+PI−: early apoptotic cells, Annexin V+PI+: late apoptotic/necrotic cells). Right panel shows cell percentages in left flow cytometry plots. Data are shown as mean ± SD (<span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>) LDH released from TNBC cells upon nigericin treatment (2 μg/mL) at different time points was detected using the LDH release assay. Each bar represents mean ± SD of experimental triplicates (one-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). (<b>D</b>) IL-1β released from TNBC cells upon nigericin treatment (2 μg/mL for 24 h) was detected with the IL-1β ELISA. Each bar represents mean ± SD of experimental triplicates (<span class="html-italic">t</span>-test, *** <span class="html-italic">p</span> &lt; 0.001). Level of cleaved Caspase-1 or cleaved Caspase-3 was detected via western blots. (<b>E</b>) Nigericin treatment induced the cleavage of Caspase-1 and Caspase-3, as detected by western blots. (<b>F</b>) Representative SEM pictures of MDA-MB-231 treated with nigericin (2 μg/mL for 24 h). White arrows indicate membrane pores. Black arrows indicate pyroptotic bubbles. Yellow arrows indicate apoptotic bodies. TNBC, triple-negative breast cancer; PI, propidium iodide; LDH, lactate dehydrogenase; IL, interleukin; SEM, scanning electron microscope.</p>
Full article ">Figure 2
<p>Nigericin-induced TNBC pyroptosis is GSDMD-dependent. (<b>A</b>) Western blots detected GSDMD or GSDME in TNBC cells upon nigericin treatment (2 μg/mL or 4 μg/mL for 12 h). (<b>B</b>) Confocal microscopy detected the N-terminal GSDMD migration from cytoplasm to membrane in MDA-MB-231 cells upon nigericin treatment (2 μg/mL for 12 h). (<b>C</b>) Flow cytometry detected PI- and Annexin V-stained TNBC cells with and without GSDMD knockdown, which occurred upon nigericin treatment (nigericin 2 μg/mL for 12 h, Annexin V+PI−: early apoptotic cells; Annexin V+PI+: late apoptotic/necrotic cells). (<b>D</b>) LDH released from TNBC cells with or without GSDMD knockdown, treated with nigericin (2 μg/mL for 12 h). Bar graphs represent mean ± SD of experimental triplicates (<span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>E</b>) Western blots detected supernatant HMGB1 and cytoplasm GSDMD in control and GSDMD knockdown TNBC cells treated with nigericin (2 μg/mL for 12 h). (<b>F</b>) Representative phase-contrast images of control and GSDMD knockdown TNBC cells treated with nigericin (2 μg/mL for 24 h). Red arrows indicate pyroptotic cells, and yellow arrows indicate apoptotic cells. TNBC, triple-negative breast cancer; FL-GSDMD, full-length gasdermin-D; N-GSDMD, N-terminal fragments of gasdermin-D; FL-GSDME, full-length gasdermin-E; N-GSDME, N-terminal fragments of gasdermin-E; DAPI, 4′,6-diamidino-2-phenylindole; PI, propidium iodide; LDH, lactate dehydrogenase; HMGB1, high mobility group box-1.</p>
Full article ">Figure 3
<p>Nigericin treatment leads to mitochondrial dysfunction. (<b>A</b>) Changes in intracellular potassium level in TNBC cells before and after nigericin treatment (2 μg/mL for 6 h), as detected via ICP-MS. Bar graphs stand for mean ± SD of experimental triplicates (<span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05). (<b>B</b>) Mitochondrial membrane potential changes in TNBC cells (nigericin 2 μg/mL for 6 h), as detected via JC-1 probes. At high mitochondrial membrane potential, JC-1 aggregated and yielded red-colored emission (590 nm). At low mitochondrial membrane potential, JC-1 was predominantly a monomer that yielded green-colored emission (530 nm). (<b>C</b>) Metabolites screened via mass spectrometry in 4T1 cells with or without nigericin treatment. Metabolic pathway enrichment of differential metabolites was performed based on Metaboanalyst (<a href="https://www.metaboanalyst.ca" target="_blank">https://www.metaboanalyst.ca</a> (accessed on 31 October 2020)). (<b>D</b>,<b>E</b>) Nigericin significantly impacted mitochondrial metabolism. (<b>D</b>) Metabolites of TCA cycle and (<b>E</b>) oxidative phosphorylation upon nigericin treatment detected via mass spectrometry. Bar graphs stand for mean ± SD of experimental quadruplicates (<span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). TNBC, triple-negative breast cancer; JC-1, tetraethylbenzimidazolylcarbocyanine iodide; ICP-MS, inductively coupled plasma mass spectrometry; TCA cycle, tricarboxylic acid cycle; FADH2, flavin adenine dinucleotide; NADH, nicotinamide adenine dinucleotide.</p>
Full article ">Figure 4
<p>Nigericin-mediated mitochondria dysfunction leads to Caspase-1-induced pyroptosis. (<b>A</b>) Changes in ROS level in TNBC cells treated with indicated reagents, as detected via flow cytometry. (<b>B</b>,<b>C</b>) Western blots detected expression of Caspase-1 and GSDMD in TNBC cells treated with indicated reagents. (<b>D</b>) LDH released from TNBC cells treated in indicated reagents was assessed using LDH assay kits. Bar graphs are shown as mean ± SD of experimental triplicates (one-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). TNBC, triple-negative breast cancer; ROS, reactive oxygen species; Nig, nigericin; NAC, N-acetylcysteine; GSDMD, gasdermin-D; LDH, lactate dehydrogenase.</p>
Full article ">Figure 5
<p>Nigericin-mediated mitochondria dysfunction leads to Caspase-3 dependent apoptosis. (<b>A</b>–<b>C</b>) Changes in Parp-1 and Caspase-3 in TNBC cells treated with indicated reagents, as analyzed via western blots. (<b>D</b>) LDH released from TNBC cells treated with indicated reagents was assessed using LDH assay kits. Each bar represent means± SD of experimental triplicates (one-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). TNBC, triple-negative breast cancer; NAC, N-acetylcysteine; Nig, nigericin; GSDMD, gasdermin-D; LDH, lactate dehydrogenase.</p>
Full article ">Figure 6
<p>Nigericin combined with anti-PD-1 is an effective anti-tumor strategy. (<b>A</b>) Flow cytometry analysis of TNF-α secreted by CD4+ and CD8+ T cells from human PBMCs co-cultured with MDA-MB-231 and treated with indicated reagents. Each bar represents mean ± SD (one-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). (<b>B</b>) 4T1 cells were injected orthotopically into left inguinal mammary fat pad of BALB/c mice, which were treated with either nigericin (subcutaneous) or anti-PD-1 antibody (intraperitoneal) alone or together. Tumor volumes were monitored every 3–4 days. Displayed are means ± SD of different groups (two-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). (<b>C</b>,<b>D</b>) Representative image (<b>C</b>) and tumor weights (<b>D</b>) of 4T1 xenografts. Each bar represents mean ± SD (one-way ANOVA, *** <span class="html-italic">p</span> &lt; 0.001). (<b>E</b>) Proportions of infiltrated CD4 and CD8 in tumors were detected via flow cytometry. Each bar represents mean ± SD (one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). (<b>F</b>) Flow cytometry analysis of IFN-γ or TNF-α secreted by CD4+ and CD8+ T cells. Displayed are means ± SD of different groups (one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.001). (<b>G</b>) Expression of cleaved Caspase-1 or cleaved Caspase-3 of tumors from each group, as detected via IHC. Each bar represents mean ± SD (one-way ANOVA, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). PD-1, programmed death-1; Nig, nigericin; PBMCs, human peripheral blood mononuclear cells; TNF, tumor necrosis factor; IHC, immuno-histochemistry.</p>
Full article ">Figure 7
<p>Schematic summary of mechanism of Nigericin in inducing anti-tumor immune response. Nigericin caused cellular potassium efflux and mitochondrial dysfunction, leading to mitochondrial ROS accumulation, as well as activation of Caspase-1/GSDMD-mediated pyroptosis and Caspase-3-dependent apoptosis in TNBC cells. Finally, nigericin-induced pyroptosis could amplify anti-tumor immune response by enhancing infiltration and anti-tumor effects of CD4+ and CD8+ T cells.</p>
Full article ">
26 pages, 60053 KiB  
Article
Pan-Cancer Landscape of NEIL3 in Tumor Microenvironment: A Promising Predictor for Chemotherapy and Immunotherapy
by Weixin Liao, Shaozhuo Huang, Lin Li, Jialiang Wang, Jing Li, Yongjian Chen, Lubiao Chen, Yifan Lian and Yuehua Huang
Cancers 2023, 15(1), 109; https://doi.org/10.3390/cancers15010109 - 24 Dec 2022
Cited by 1 | Viewed by 1949
Abstract
With the aim of enhancing the understanding of NEIL3 in prognosis prediction and therapy administration, we conducted a pan-cancer landscape analysis on NEIL3. The mutation characteristics, survival patterns, and immune features of NEIL3 across cancers were analyzed. Western blotting, qPCR, and immunohistochemistry were [...] Read more.
With the aim of enhancing the understanding of NEIL3 in prognosis prediction and therapy administration, we conducted a pan-cancer landscape analysis on NEIL3. The mutation characteristics, survival patterns, and immune features of NEIL3 across cancers were analyzed. Western blotting, qPCR, and immunohistochemistry were conducted to validate the bioinformatics results. The correlation between NEIL3 and chemotherapeutic drugs, as well as immunotherapies, was estimated. NEIL3 was identified as an oncogene with prognostic value in predicting clinical outcomes in multiple cancers. Combined with the neoantigen, tumor mutational burden (TMB), and microsatellite instability (MSI) results, a strong relationship between NEIL3 and the TME was observed. NEIL3 was demonstrated to be closely associated with multiple immune parameters, including infiltrating immunocytes and pro-inflammatory chemokines, which was verified by experiments. More importantly, patients with a higher expression of NEIL3 were revealed to be more sensitive to chemotherapeutic regimens and immune checkpoint inhibitors in selected cancers, implying that NEIL3 may be an indicator for therapeutic administration. Our study indicated NEIL3 has a strong association with the immune microenvironment and phenotypic changes in certain types of cancers, which facilitated the improved understanding of NEIL3 across cancers and highlighted the potential for clinical application of NEIL3 in precision medical stratification. Full article
Show Figures

Figure 1

Figure 1
<p>The mRNA expression level of NEIL3 across human cancers. The mRNA expression of NEIL3 in (<b>A</b>) 31 types of normal tissues (N, number of samples); (<b>B</b>) 21 types of cancer cell lines in CCLE database (N, number of samples); (<b>C</b>) 20 types of cancers and normal tissues in TCGA database (N, normal; T, tumor); (<b>D</b>) tumor and normal tissues in TCGA combined with GTEx database (N, normal; T, tumor); and (<b>E</b>) tumor and paired adjacent normal tissue in TCGA database. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Experimental validation of the expression and prognosis of NEIL3 in cancers. (A) mRNA expression of NEIL3 in LIHC, COAD, and KIRC cell lines. (B) Protein expression of NEIL3 in LIHC, COAD, and KIRC cell lines, as well as their quantitative results. (<b>C</b>) mRNA expression of NEIL3 in 12 patients’ LIHC and paired adjacent liver tissues. (<b>D</b>) Representative images of IHC staining of HCC patient samples with low and high NEIL3 expression. (<b>E</b>) Difference in OS between high and low NEIL3 expression groups. ns indicates no significance, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Mutation features of NEIL3 among cancers. (<b>A</b>) Alteration frequency of NEIL3 among 33 types of cancers. (<b>B</b>) Three-dimensional structure diagram of the NEIL3 protein. The green part represents the missense mutation area, black represents the truncating mutation area, and orange represents the inframe mutation area. (<b>C</b>) Mutation diagram of NEIL3. Light green circles represent missense mutations, light orange circles represent splice mutations, and gray circles represent truncating mutations. In the case of different mutation types at a single position, the color of the circle represents the most frequent mutation type. (<b>D</b>) Fractional genomic alteration of NEIL3 across 32 cancers. Each dot represents a sample. (<b>E</b>) Mutation types of NEIL3 at mRNA expression level.</p>
Full article ">Figure 4
<p>Prediction of NEIL3 expression and affected genes related to NEIL3 mutation. (<b>A</b>) The expression of NEIL3 between NEIL3 mutation and non-mutation groups. (<b>B</b>) Expression of the genes affected by NEIL3 mutation. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Potential role of altered NEIL3 in cancers. (<b>A</b>–<b>D</b>) GSVA of enriched pathways and OS analysis of the NEIL3-altered and -unaltered groups in BLCA (<b>A</b>), LUAD (<b>B</b>), LUSC (<b>C</b>), and UCEC (<b>D</b>).</p>
Full article ">Figure 6
<p>NEIL3 expression level is correlated with immune characteristics in cancers. Correlation between the expression level of NEIL3 and (<b>A</b>) lymphocyte infiltration in KIPAN, KIRC, and LIHC; (<b>B</b>) stromal score, immune score, and ESTIMATE score of the top 3 cancers; and (<b>C</b>) 40 common immune checkpoint genes across cancers. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Experimental validation of the association of NEIL3 and immunity in cancers. (<b>A</b>) mRNA expression of NEIL3 and immune biomarkers in 24 LIHC samples. (<b>B</b>) Immune biomarkers positively correlated with NEIL3 expression with statistical significance. (<b>C</b>) Representative images of IHC staining for NEIL3 and CCR4 in 9 types of cancers.</p>
Full article ">Figure 8
<p>NEIL3 expression is correlated with neoantigen number, TMB, and MSI in various cancers. Correlation between NEIL3 expression level and (<b>A</b>) neoantigen number; (<b>B</b>) TMB; and (<b>C</b>) MSI across cancers.</p>
Full article ">Figure 9
<p>NEIL3 expression is correlated with MMR and DNA methyltransferase genes in multiple cancers. Correlation between NEIL3 expression level and (<b>A</b>) mutation of five MMR genes, namely, MLH1, MSH2, MSH6, PMS2, and EPCAM; (<b>B</b>) expression of four DNA methyltransferase genes, namely, DNMT1 (red), DNMT2 (blue), DNMT3A (green), and DNMT3B (purple), in various cancers. The innermost brown and blue circles represent the R and <span class="html-italic">p</span> values of the correlation analysis, respectively. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>NEIL3 was correlated with chemotherapy sensitivity and ICB response. The association of NEIL3 expression level with chemotherapy sensitivity in (<b>A</b>) CGP; (<b>B</b>) CTRP; and (<b>C</b>) CCLE databases. (<b>D</b>) The CARE score of the relationship of NEIL3 and chemotherapy resistance in three databases. (<b>E</b>) The NEIL3 expression ratio between ICI responders and non-responders in the listed cohorts. Cohort A: Willy Hugo (PMID:26997480); Cohort B: Alexandra Snyder (PMID:28552987); Cohort C: Nadeem Riaz (PMID:29033130); Cohort D: Diana Miao (PMID:29301960); Cohort E: Sanjeev Mariathasan (PMID:29443960). (<b>F</b>) The difference in NEIL3 non-silent mutation rate between ICI responders and non-responders in the following cohorts. Cohort A: Naiyer A Rizvi (PMID:25765070); Cohort B: Eliezer M Van Allen (PMID: 26359337); Cohort C: Willy Hugo (PMID:26997480); Cohort D: Nadeem Riaz (PMID:29033130). 1256580-46-7, also known as Alectinib.</p>
Full article ">
25 pages, 9862 KiB  
Article
ACAP1 Deficiency Predicts Inferior Immunotherapy Response in Solid Tumors
by Qiyi Yi, Youguang Pu, Fengmei Chao, Po Bian and Lei Lv
Cancers 2022, 14(23), 5951; https://doi.org/10.3390/cancers14235951 - 1 Dec 2022
Cited by 6 | Viewed by 2492
Abstract
Background: ACAP1 plays a key role in endocytic recycling, which is essential for the normal function of lymphocytes. However, the expression and function of ACAP1 in lymphocytes have rarely been studied. Methods: Large-scale genomic data, including multiple bulk RNA-sequencing datasets, single-cell sequencing datasets, [...] Read more.
Background: ACAP1 plays a key role in endocytic recycling, which is essential for the normal function of lymphocytes. However, the expression and function of ACAP1 in lymphocytes have rarely been studied. Methods: Large-scale genomic data, including multiple bulk RNA-sequencing datasets, single-cell sequencing datasets, and immunotherapy cohorts, were exploited to comprehensively characterize ACAP1 expression, regulation, and function. Gene set enrichment analysis (GSEA) was used to uncover the pathways associated with ACAP1 expression. Eight algorithms, including TIMER, CIBERSORT, CIBERSORT-ABS, QUANTISEQ, xCELL, MCPCOUNTER, EPIC, and TIDE, were applied to estimate the infiltrating level of immune cells. Western blotting, qPCR, and ChIP-PCR were used to validate the findings from bioinformatic analyses. A T-cell co-culture killing assay was used to investigate the function of ACAP1 in lymphocytes. Results: ACAP1 was highly expressed in immune-related tissues and cells and minimally in other tissues. Moreover, single-cell sequencing analysis in tumor samples revealed that ACAP1 is expressed primarily in tumor-infiltrating lymphocytes (TILs), including T, B, and NK cells. ACAP1 expression is negatively regulated by promoter DNA methylation, with its promoter hypo-methylated in immune cells but hyper-methylated in other cells. Furthermore, SPI1 binds to the ACAP1 promoter and positively regulates its expression in immune cells. ACAP1 levels positively correlate with the infiltrating levels of TILs, especially CD8+ T cells, across a broad range of solid cancer types. ACAP1 deficiency is associated with poor prognosis and immunotherapeutic response in multiple cancer types treated with checkpoint blockade therapy (ICT). Functionally, the depletion of ACAP1 by RNA interference significantly impairs the T cell-mediated killing of tumor cells. Conclusions: Our study demonstrates that ACAP1 is essential for the normal function of TILs, and its deficiency indicates an immunologically “cold” status of tumors that are resistant to ICT. Full article
Show Figures

Figure 1

Figure 1
<p>ACAP1 mRNA expression across different tissues and cell lines. (<b>A</b>) Violin plots of ACAP1 expression levels across all available tissues ordered by ACAP1 expression in the GTEx Portal. (<b>B</b>) ACAP1 expression levels in human tissues and cell lines were visualized by BioGPS. Red: tissues and cells with relatively high ACAP1 expression. (<b>C</b>) Violin plots of ACAP1 expression levels across different types of cancer cell lines in the CCLE dataset. (<b>D</b>) The protein levels of ACAP1 from indicated cell lines were determined by Western blotting.</p>
Full article ">Figure 2
<p>Single-cell gene expression analysis of ACAP1. (<b>A</b>,<b>B</b>) Single-cell RNA sequencing analyses of ACAP1 mRNA expression across various cell types in melanoma datasets GSE72096 and GSE115978. (<b>C</b>) Single-cell expression patterns of ACAP1 in the glioblastoma dataset “Neftel 2019” are shown with tSNE plots. (<b>D</b>,<b>E</b>) Single-cell expression patterns of ACAP1 in prostate cancer datasets, including “He 2021” and “Wu 2021 (PC-P1)”, are shown with UMAP plots. (<b>F</b>) Single-cell expression patterns of ACAP1 in the cutaneous melanoma dataset “Wu 2021 (M-P1)” are shown with UMAP plots. (<b>G</b>–<b>I</b>) Single-cell expression patterns of ACAP1 in breast cancer datasets, including “Wu 2021 (BC-P1)”, “Wu 2021 (BC-P2)”, and “Wu 2021 (BC-P3)”, are shown with UMAP plots.</p>
Full article ">Figure 3
<p>Pan-cancer analysis of ACAP1 expression in human cancer. (<b>A</b>) Comparing of ACAP1 mRNA levels in tumor vs. normal samples across TCGA cancer types by combing the TCGA and GTEx data. (<b>B</b>) Paired comparison of ACAP1 mRNA levels in tumor vs. normal samples in TCGA. Green: decreased ACAP1 expression in tumors; Red: elevated gene expression in tumors. (<b>C</b>) Comparison of ACAP1 protein levels in tumor vs. normal samples across all cancer types available in CPTAC using UALCAN webtool. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05, ns (non-significant).</p>
Full article ">Figure 4
<p>Implications of ACAP1 expression on overall survival of cancer patients across multiple cancer types. (<b>A</b>–<b>U</b>) Overall survival analyses of cancer patients stratified by ACAP1 mRNA level with the Kaplan–Meier method in TCGA datasets. (<b>V</b>–<b>Y</b>) Overall survival analyses of cancer patients stratified by ACAP1 mRNA level in ICGC-LIRI-JP(LIHC), GSE68465(LUAD), GSE22153(SKCM), and CGGA325(GBM) datasets.</p>
Full article ">Figure 5
<p>Transcriptional regulation of ACAP1. (<b>A</b>) Heatmap of TCGA samples (I), ACAP1 mRNA expression (II), β-value (methylation level) of 4 CpG sites, including cg13295242, cg13670306, cg11807006, and cg25671438, in the ACAP1 promoter region (III), copy number variation (IV), and SPI1 mRNA expression (V) in TCGA pan-cancer dataset. The samples were ordered by ACAP1 expression. Blue: low level; Red: high level. (<b>B</b>) Violin plots showing the β-value of cg25671438 in indicated cancers of TCGA. (<b>C</b>,<b>D</b>) Violin plots showing the β-value of cg25671438 and the average β-value of 4 CpG sites in indicated cancer cell lines of GSE68379. (<b>E</b>) Heatmap of the Spearman correlation coefficient of ACAP1 mRNA levels with β value of CpG sites in ACAP1 promoter and copy number across 33 cancer types in TCGA. (<b>F</b>) The β-value of 4 CpG sites in the ACAP1 promoter region of Huh-7 and SK-HEP-1 cells in GSE68379. (<b>G</b>) The impact of 5-aza on ACAP1 mRNA level in Huh-7 and SK-HEP-1 cells. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Transcriptional regulation of ACAP1 by SPI1. (<b>A</b>) Heatmap of the correlation coefficient of ACAP1 mRNA levels and SPI1 levels across 33 cancer types in TCGA. (<b>B</b>) Scatter plot displays the SPI1 and ACAP1 mRNA expression in EBV-transformed lymphocytes of GTEx dataset. (<b>C</b>) Scatter plot displays the SPI1 and ACAP1 mRNA expression in whole blood of GTEx dataset. (<b>D</b>) ChIP-sequencing peaks of SPI1 in macrophage, B lymphocyte, and lymphoma; H3K4me3 in Ramos B-lymphocytes, Jurkat T-cell, A549 lung cancer cells, Capan-1 pancreatic ductal cancer cells, Hela-S3 cervix cancer cells, HCT116 colon cancer cells, DU145 prostate cancer cells, esophagus cells, brain cells, and MDA-MB-231 breast cancer cells. The binding region of SPI1 on ACAP1 promoter is highlighted in cyan shaded box. The SPI1 binding sites on ACAP1 promoter predicted by JASPAR are shown. (<b>E</b>) SPI1 binding motif MA0080.1 and MA0080.2 from JASPAR curated motif database. (<b>F</b>) ChIP-PCR showed SPI1 binds to the promoter of ACAP1 in Jurkat cells. (<b>G</b>) Analysis of SPI1 overexpression on ACAP1 protein expression in Jurkat by Western blotting. (<b>H</b>) Effects of hypoxia-mimicking CoCl<sub>2</sub> treatment on HIF1α, SPI1, and ACAP1 expression in Jurkat cells were determined by Western blotting, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>The Spearman correlations of ACAP1 expression with immune cell infiltration across 32 cancer types in TCGA.</p>
Full article ">Figure 8
<p>ACAP1 knockdown impairs the cytotoxicity of T cells against tumor cells. (<b>A</b>) Western blotting of lysates from TALL-104 cells infected with control or two different ACAP1-targeting shRNA lentiviruses. (<b>B</b>) Representative images of live/dead A549 cells co-cultured with different TALL-104 cells at a 1:2 cell ratio for 24 h were shown. Red-fluorescent PI (propidium iodide) was used to detect dead cells. Green-fluorescent CMFDA (5-chloromethylfluorescein diacetate) was used to detect live cells. Scale bars, 100 μm. (<b>C</b>) Three random fields were analyzed, and live/dead cell ratios were quantified. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>ACAP1 deficiency correlates with inferior ICT response and prognosis in multiple cancer types. (<b>A</b>) ACAP1 expression in different response groups; Kaplan–Meier OS and PFS estimates according to ACAP1 expression in “VanAllen2015” cohort, of which melanoma patients were treated with anti-CTLA-4 antibody (ipilimumab). (<b>B</b>) ACAP1 expression in different response groups; Kaplan–Meier OS estimates according to ACAP1 expression in “Snyder 2014” cohort, in which melanoma patients were treated with anti-CTLA-4 antibody (tremelimumab or ipilimumab). (<b>C</b>) ACAP1 expression in different response groups; Kaplan–Meier OS and PFS estimates according to ACAP1 expression in the “Gide 2019” cohort, in which melanoma patients were treated with anti-PD1 antibody (nivolumab or pembrolizumab) or anti-CTLA-4/PD-1 antibody (ipilimumab + pembrolizumab) (one patient with the extreme value of ACAP1 expression was excluded). (<b>D</b>) ACAP1 expression in different response groups; Kaplan–Meier OS estimates according to ACAP1 expression in both “prior to therapy” and “during therapy” groups of “Riaz 2017” cohort, in which melanoma patients were treated with anti-PD1 antibody (nivolumab). (<b>E</b>) Kaplan–Meier OS and PFS estimate according to ACAP1 expression in the “Miao 2018” cohort, in which RCC patients were treated with anti-PD-1 and/or anti-CTLA-4 antibodies (nivolumab or atezolizumab or nivolumab + ipilimumab). (<b>F</b>) ACAP1 expression in different response groups in the “Ruppin 2021” cohort, of which LUAD (lung adenocarcinoma) patients were treated with anti-PD-1 antibody (pembrolizumab). (<b>G</b>) ACAP1 expression in different response groups; Kaplan–Meier OS and PFS estimates according to ACAP1 expression in GSE126044, in which NSCLC patients were treated with anti-PD-1 antibody (nivolumab). (<b>H</b>) ACAP1 expression in different response groups; Kaplan–Meier OS estimates according to ACAP1 expression in the “IMvigor210” cohort, in which mUC patients were treated with anti-PD-L1 antibody (atezolizumab). OS: overall survival. PFS: progression-free survival. Kaplan–Meier survival curves with <span class="html-italic">p</span>-values derived by log-rank test were shown. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">

Review

Jump to: Research

28 pages, 1578 KiB  
Review
Polarization of Cancer-Associated Macrophages Maneuver Neoplastic Attributes of Pancreatic Ductal Adenocarcinoma
by Huey-Jen Lin, Yingguang Liu, Kailey Caroland and Jiayuh Lin
Cancers 2023, 15(13), 3507; https://doi.org/10.3390/cancers15133507 - 5 Jul 2023
Cited by 4 | Viewed by 3121
Abstract
Mounting evidence links the phenomenon of enhanced recruitment of tumor-associated macrophages towards cancer bulks to neoplastic growth, invasion, metastasis, immune escape, matrix remodeling, and therapeutic resistance. In the context of cancer progression, naïve macrophages are polarized into M1 or M2 subtypes according to [...] Read more.
Mounting evidence links the phenomenon of enhanced recruitment of tumor-associated macrophages towards cancer bulks to neoplastic growth, invasion, metastasis, immune escape, matrix remodeling, and therapeutic resistance. In the context of cancer progression, naïve macrophages are polarized into M1 or M2 subtypes according to their differentiation status, gene signatures, and functional roles. While the former render proinflammatory and anticancer effects, the latter subpopulation elicits an opposite impact on pancreatic ductal adenocarcinoma. M2 macrophages have gained increasing attention as they are largely responsible for molding an immune-suppressive landscape. Through positive feedback circuits involving a paracrine manner, M2 macrophages can be amplified by and synergized with neighboring neoplastic cells, fibroblasts, endothelial cells, and non-cell autonomous constituents in the microenvironmental niche to promote an advanced disease state. This review delineates the molecular cues expanding M2 populations that subsequently convey notorious clinical outcomes. Future therapeutic regimens shall comprise protocols attempting to abolish environmental niches favoring M2 polarization; weaken cancer growth typically assisted by M2; promote the recruitment of tumoricidal CD8+ T lymphocytes and dendritic cells; and boost susceptibility towards gemcitabine as well as other chemotherapeutic agents. Full article
Show Figures

Figure 1

Figure 1
<p>Pancreatic ductal adenocarcinoma cells synergize with the tumor microenvironment to provoke polarization of M2 macrophages. Arrows with pointed or with blocked ends indicate activation or inhibition between regulators, respectively, while a fading effect at the start of arrows represents secretion of modulators. Plain straight lines depict interaction between ligands and receptors. Cell surface proteins are noted in rectangular boxes on cell membranes. The circular lipid bilayer depicts an extracellular vesicle. Abbreviations used include aryl hydrocarbon receptor (AhR), protein kinases B (AKT), Bcl-2-associated athanogene 3 (BAG3), cancer-associated fibroblast (CAF), CC-chemokine ligand (CCL), cluster of differentiated (CD), CXC chemokine ligand (CXCL), colony-stimulating factor (CSF) and receptor (CSF1R), dendritic cell (DC), double cortin-like kinase 1 (Dclk1), endothelial cell (Endo), epidermal growth factor receptor (EGFR), epithelial cell (Epi), ezrin (EZR), galectin (gal), granulocyte-macrophage colony-stimulating factor (GM-CSF), hypoxia-inducible factor (HIF), interleukin (IL), insulin-like growth factor binding protein 2 (IGFBP2), interferon-induced transmembrane protein 2 (IFITM-2), interferon regulatory factor 4 (Irf4), Kirsten rat sarcoma (KRAS), mammalian target of rapamycin (mTOR), microRNA (miR), monocyte (M), nucleotide-binding and leucine-rich repeat receptor containing pyrin domain 3 (NLRP3), pancreatic ductal adenocarcinoma (PDAC), phosphatidylinositol 3-kinase (PI3K), reactive oxygen species (ROS), regenerating gene family member 4 (REG4), sialic-acid-binding immunoglobulin-like lectin 15 (SIGLEC15), signal transducer and activator of transcription (STAT), spleen tyrosine kinase (SYK), suppression of tumorigenicity 2 (ST2), tumor-associated macrophage (TAM), transforming growth factor β (TGF-β), and T helper-2 (T<sub>H</sub>2).</p>
Full article ">Figure 2
<p>M2 macrophage promotes a plethora of neoplastic features of pancreatic ductal adenocarcinoma and suppresses tumoricidal effects exerted from cytotoxic T lymphocyte. Arrows and straight plain lines, as well as overlapping abbreviations used in both figures, are described in the legend of <a href="#cancers-15-03507-f001" class="html-fig">Figure 1</a>. Additional abbreviations that are used in <a href="#cancers-15-03507-f002" class="html-fig">Figure 2</a> include apolipoprotein E (ApoE), arginine (Arg), adenylyl cyclase-associated protein 1 (CAP-1), CC-chemokine ligand (CCL), CC-chemokine receptor (CCR), cytidine deaminase (CDA), chitinase 3-like-1 (CHI3L1), CXC chemokine ligand (CXCL), endothelial cell (EC), epithelial–mesenchymal transition (EMT), extracellular signal-regulated kinase (ERK), fibronectin-1 (FN1), gemcitabine (GEM), hypoxia-inducible factor 1α (HIF-1α), interferon (IFN), insulin-like growth factor (IGF) and receptor (IGFR), IFN-stimulated gene 15 (ISG15), immunomodulatory cationic antimicrobial peptide 18/LL-37 (hCAP-18/LL-37), low-density-lipoprotein receptor (LDLR), lysyl oxidase-like protein 2 (LOXL2), macrophage inflammatory protein-3α(MIP3α), microRNA (miR), matrix metalloproteinase 9 (MMP-9), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), oncostatin M (OSM), pancreatic cancer stem-like cells (PCSCs), programmed death receptor-1 (PD-1) and ligand (PD-L1), pyruvate kinase isoform M2 (PKM2), Toll-like receptor 4 (TLR4), tryptophan (Trp), tumor necrosis factor α (TNF-α), vascular cell adhesion molecule 1 (VCAM-1), vascular endothelial growth factor (VEGF) and receptor (VEGFR), and YWHAZ/14-3-3 protein zeta/delta (14-3-3ζ).</p>
Full article ">
22 pages, 1413 KiB  
Review
The Monocyte, a Maestro in the Tumor Microenvironment (TME) of Breast Cancer
by Hoda T. Amer, Ulrike Stein and Hend M. El Tayebi
Cancers 2022, 14(21), 5460; https://doi.org/10.3390/cancers14215460 - 7 Nov 2022
Cited by 21 | Viewed by 4707
Abstract
Breast cancer (BC) is well-known for being a leading cause of death worldwide. It is classified molecularly into luminal A, luminal B HER2−, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These subtypes differ in their prognosis; thus, understanding the tumor microenvironment [...] Read more.
Breast cancer (BC) is well-known for being a leading cause of death worldwide. It is classified molecularly into luminal A, luminal B HER2−, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These subtypes differ in their prognosis; thus, understanding the tumor microenvironment (TME) makes new treatment strategies possible. The TME contains populations that exhibit anti-tumorigenic actions such as tumor-associated eosinophils. Moreover, it contains pro-tumorigenic populations such as tumor-associated neutrophils (TANs), or monocyte-derived populations. The monocyte-derived populations are tumor-associated macrophages (TAMs) and MDSCs. Thus, a monocyte can be considered a maestro within the TME. Moreover, the expansion of monocytes in the TME depends on many factors such as the BC stage, the presence of macrophage colony-stimulating factor (M-CSF), and the presence of some chemoattractants. After expansion, monocytes can differentiate into pro-inflammatory populations such as M1 macrophages or anti-inflammatory populations such as M2 macrophages according to the nature of cytokines present in the TME. Differentiation to TAMs depends on various factors such as the BC subtype, the presence of anti-inflammatory cytokines, and epigenetic factors. Furthermore, TAMs and MDSCs not only have a role in tumor progression but also are key players in metastasis. Thus, understanding the monocytes further can introduce new target therapies. Full article
Show Figures

Figure 1

Figure 1
<p>TAMs in the breast cancer TME. 1. The effect of TAMs on T cells: 1.a. TAMs can suppress T-cell proliferation via the programmed death-ligand 1 (PDL-1) [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 1.b. TGF-beta affects T-cell function by upregulation of PDL-1 on TAMs [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 1.c. Tregs are induced by IL-10, TGF-B, and PDGF-2, thus suppressing T cells [<a href="#B45-cancers-14-05460" class="html-bibr">45</a>]. 1.d. Treg recruitment happens through CCL7/8/22 [<a href="#B45-cancers-14-05460" class="html-bibr">45</a>]. 1.e. Increased activity of arginase enzyme and iNOS result in the increased level of NO and RNOS (ONOO<sup>−</sup>) leading to nitrosylation and thus impairing T-cell self-stimulation by IL-2 in addition to nitration of TCR signaling complex altering T-cell function [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 2. The effect of TAMs on NK cells: 2.a. The inhibitory effect of TAMs can be due to the expression of PDL-1 (which is highly expressed on TAMs) or 2.b. it can be due to TGF-B secretion [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 2.c. TAMs also suppress NK IFN-Y production [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 3. TAMs induce angiogenesis by releasing VEGF, PDGF, and IL-8 [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 4. Interaction between TAMs and cancer cells: 4.a. It was observed that VCAM1+ tumor cells have increased survival in a leukocyte-rich environment due to the adhesion of leukocyte receptors on BC cells (VCAM1) to TAM α 4 integrin [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 4.b. TGF-B was shown to upregulate PDL-1 on cancer cells, thus inducing an inhibitory effect on immune cells [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 4.c. There is a paracrine loop between cancer cells and TAMs. TAMs secrete epidermal growth factor (EGF) that binds to EGFRs on the cancer cells [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 4.d. TAMs express M-CSFR, which is a monocyte colony-stimulating factor receptor also known as colony-stimulating factor 1 receptor (CSF-1R or cFMS). The M-CSFR binds to the M-CSF (CSF-1) that is produced by cancer cells [<a href="#B41-cancers-14-05460" class="html-bibr">41</a>]. The binding of cancer cells with TAMs allows the co-migration of two different cell types, thus enhancing invasion, motility, and intravasation [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 5. CD163 and C206 are considered the commonly used self-markers for TAMs. 6. MCP-1 is a monocyte recruiter that is produced by TAMs; monocytes respond to TME and differentiate into TAMs [<a href="#B46-cancers-14-05460" class="html-bibr">46</a>]. 7.a. TAMs coordinate in the extracellular proteolysis through the secretion of tissue-remodeling cysteine cathepsin proteases that contribute to ECM and collagen degradation [<a href="#B47-cancers-14-05460" class="html-bibr">47</a>]. 7.b. Moreover, MMPs contribute to collagen degradation. Types I, III, IV, and VI are the major collagens that play an important role in tumors [<a href="#B47-cancers-14-05460" class="html-bibr">47</a>]. Myeloid cells remodel ECM by degrading the collagen through matrix metalloproteinases (MMPs) [<a href="#B34-cancers-14-05460" class="html-bibr">34</a>].</p>
Full article ">Figure 2
<p>IL-10: key inducers and key anti-inflammatory mechanisms. a. Inducing agents increase the release of IL-10 from different immune cells [<a href="#B25-cancers-14-05460" class="html-bibr">25</a>,<a href="#B54-cancers-14-05460" class="html-bibr">54</a>,<a href="#B55-cancers-14-05460" class="html-bibr">55</a>] b. IL-10 acts as an inhibitor for a number of pro-inflammatory cytokines released by macrophages/monocytes. c. IL-10 induces a small subset of genes in human monocytes [<a href="#B56-cancers-14-05460" class="html-bibr">56</a>]. d. IL-10 is an inhibitor for NF-kB (nuclear localization of nuclear factor kB), a transcriptional factor responsible for the expression of inflammatory genes; however, other transcriptional factors such as NF-IL6, AP-1, and AP-2 were not affected by IL-10 [<a href="#B57-cancers-14-05460" class="html-bibr">57</a>].</p>
Full article ">Figure 3
<p>Role of TAMs and MDSCs in metastasis: 1. Tumor growth and invasion. 1.a. TAMs produce MMPs (MMP1/2/3/7/9) and cathepsin. 1.b. Chemokines such as acidic and rich in cysteine (SPARC), chemokine (C-C motif) ligand 18 (CCL18), αvβ5 integrins, phosphatidylinositol transfer protein 3 (PITPNM3), epidermal growth factor (EGF), EGF receptor (EGFR), colony-stimulating factor 1 (CSF-1), and CSF-1 receptor (CSF-1R) allow the interaction between tumor cells, thus facilitating the invasion step [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>].1.c. MDSCs facilitate the invasion step by producing MMPs and TGF-beta [<a href="#B73-cancers-14-05460" class="html-bibr">73</a>]. 2. Intravasation. 2.a. TAMs and MDSCs release VEGF-A [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>,<a href="#B73-cancers-14-05460" class="html-bibr">73</a>]. 3. Extravasation. 3.a. Metastasis-associated macrophages (MAMs) accumulate at the metastatic site and also release VEGF-A. 3.b. The CCL2-CCR2 signaling pathway activates the CCL3-CCR1 signaling pathway in MAMs, leading to their accumulation in the metastatic site and thus attracting more tumor cells and prolonging the process of seeding. 4. Seeding [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>]. 5. The metastatic niche in the lungs is induced by MDSCs through the production of CXCL-17 which recruits more MDSCs in the lungs, leading to an increase in the levels of platelet-derived growth factor-beta (PDGF-beta) that will induce angiogenesis, thus creating favorable conditions for metastatic cells [<a href="#B74-cancers-14-05460" class="html-bibr">74</a>].</p>
Full article ">
25 pages, 1213 KiB  
Review
Oral Immune-Related Adverse Events Caused by Immune Checkpoint Inhibitors: Salivary Gland Dysfunction and Mucosal Diseases
by Yoshiaki Yura and Masakazu Hamada
Cancers 2022, 14(3), 792; https://doi.org/10.3390/cancers14030792 - 4 Feb 2022
Cited by 17 | Viewed by 3291
Abstract
Conventional chemotherapy and targeted therapies have limited efficacy against advanced head and neck squamous cell carcinoma (HNSCC). The immune checkpoint inhibitors (ICIs) such as antibodies against CTLA-4, PD-1, and PD-L1 interrupt the co-inhibitory pathway of T cells and enhance the ability of CD8 [...] Read more.
Conventional chemotherapy and targeted therapies have limited efficacy against advanced head and neck squamous cell carcinoma (HNSCC). The immune checkpoint inhibitors (ICIs) such as antibodies against CTLA-4, PD-1, and PD-L1 interrupt the co-inhibitory pathway of T cells and enhance the ability of CD8+ T cells to destroy tumors. Even in advanced HNSCC patients with recurrent diseases and distant metastasis, ICI therapy shows efficiency and become an effective alternative to conventional chemotherapy. However, as this therapy releases the immune tolerance state, cytotoxic CD8+ T cells can also attack organs and tissues expressing self-antigens that cross-react with tumor antigens and induce immune-related adverse events (irAEs). When patients with HNSCC are treated with ICIs, autoimmune diseases occur in multiple organs including the skin, digestive tract, endocrine system, liver, and respiratory tract. Treatment of various malignancies, including HNSCC, with ICIs may result in the appearance of oral irAEs. In the oral cavity, an oral lichenoid reaction (OLR) and pemphigoid develop. Sicca syndrome also occurs in association with ICIs, affecting the salivary glands to induce xerostomia. It is necessary to elucidate the pathogenic mechanisms of these intractable diseases that are not seen with conventional therapy. Early diagnosis and appropriate approaches to irAEs are needed for efficient treatment of advanced HNSCC by ICIs. Full article
Show Figures

Figure 1

Figure 1
<p>CTLA-4 and PD-1/PD-L1 in cellular (<b>A</b>) and humoral (<b>B</b>) tumor immunity. (<b>A</b>) In the CD8<sup>+</sup> T cell-based pathway, when tumor antigens are released into the tumor microenvironment from dying tumor cells, DCs take up the protein antigens and decomposes them to peptide antigens. In regional lymph nodes, the peptide antigens are cross-presented via MHC class I on the cell surface and recognized by the TCR of CD8<sup>+</sup> T cells. In addition, the co-stimulatory binding of CD80/86 of DCs with CD28 of T cells is required for naïve CD8<sup>+</sup> T cells to differentiate into cytotoxic CD8<sup>+</sup> T cells. Activated CD8<sup>+</sup> T cells express the co-inhibitory molecule CTLA-4 to prevent excess activation of CD8<sup>+</sup> T cells. CD8<sup>+</sup> T cells move to a peripheral tumor site and recognize tumor peptide antigens presented via MHC class I and exert antitumor activity. However, PD-I on CD8<sup>+</sup> T cells binds PD-L1 on tumor cells and suppresses the activity of CD8<sup>+</sup> T cells. Treg cells locally suppress the activity of CD8<sup>+</sup> T cells and CD4<sup>+</sup> helper cells. (<b>B</b>) The germinal center (GC) plays an important role in the proliferation and differentiation of B cells. Tfh cells physically bind to GC B cells by co-stimulatory/co-inhibitory pairs such as CD28–CD80/86, CD40L–CD40, ICOS–ICOSL, PD-1, and PD-L1. BCR expressed on B cells detects the protein antigens, and take them into the cells. Peptide antigens processed in B cells are then presented via MHC class II and recognized by Tfh cells. Interaction between Tfh and B cells promotes the differentiation of GC B cells into plasma cells and memory B cells. Tfr cells suppress Tfh and B cells. LN, lymph node; Treg, regulatory T; TCR, T cell receptor; irAEs, immune-related adverse events; GC, germinal center; Tfh, T follicular helper; Tfr, T follicular regulatory; PC, plasma cell; BCR, B cell receptor.</p>
Full article ">Figure 2
<p>Characteristics of 76 previously reported patients with ICI-induced Sicca syndrome. (<b>A</b>) Sex. (<b>B</b>) Types of underlying malignancies. (<b>C</b>) Time to onset, months. (<b>D</b>) Prevalence of Sicca syndrome-related serum autoantibodies. (<b>E</b>) Other irAEs. (<b>F</b>) Degree of improvement of irAEs.</p>
Full article ">
25 pages, 861 KiB  
Review
Breast Cancer Tumor Microenvironment and Molecular Aberrations Hijack Tumoricidal Immunity
by Huey-Jen Lin, Yingguang Liu, Denene Lofland and Jiayuh Lin
Cancers 2022, 14(2), 285; https://doi.org/10.3390/cancers14020285 - 7 Jan 2022
Cited by 16 | Viewed by 4362
Abstract
Breast cancer is the most common malignancy among females in western countries, where women have an overall lifetime risk of >10% for developing invasive breast carcinomas. It is not a single disease but is composed of distinct subtypes associated with different clinical outcomes [...] Read more.
Breast cancer is the most common malignancy among females in western countries, where women have an overall lifetime risk of >10% for developing invasive breast carcinomas. It is not a single disease but is composed of distinct subtypes associated with different clinical outcomes and is highly heterogeneous in both the molecular and clinical aspects. Although tumor initiation is largely driven by acquired genetic alterations, recent data suggest microenvironment-mediated immune evasion may play an important role in neoplastic progression. Beyond surgical resection, radiation, and chemotherapy, additional therapeutic options include hormonal deactivation, targeted-signaling pathway treatment, DNA repair inhibition, and aberrant epigenetic reversion. Yet, the fatality rate of metastatic breast cancer remains unacceptably high, largely due to treatment resistance and metastases to brain, lung, or bone marrow where tumor bed penetration of therapeutic agents is limited. Recent studies indicate the development of immune-oncological therapy could potentially eradicate this devastating malignancy. Evidence suggests tumors express immunogenic neoantigens but the immunity towards these antigens is frequently muted. Established tumors exhibit immunological tolerance. This tolerance reflects a process of immune suppression elicited by the tumor, and it represents a critical obstacle towards successful antitumor immunotherapy. In general, immune evasive mechanisms adapted by breast cancer encompasses down-regulation of antigen presentations or recognition, lack of immune effector cells, obstruction of anti-tumor immune cell maturation, accumulation of immunosuppressive cells, production of inhibitory cytokines, chemokines or ligands/receptors, and up-regulation of immune checkpoint modulators. Together with altered metabolism and hypoxic conditions, they constitute a permissive tumor microenvironment. This article intends to discern representative incidents and to provide potential innovative therapeutic regimens to reinstate tumoricidal immunity. Full article
Show Figures

Figure 1

Figure 1
<p>Representative aberrations in breast cancer cell (<b>A</b>), tumoricidal cells (<b>B</b>), cancer-promoting cells and metabolites (<b>C</b>) lead to immune evasion. This crosstalk map indicates the tumoricidal cells and factors (in green) and the pro-tumorigenic counterparts and factors (in red). Rectangular boxes represent cell surface molecules; plain lines indicate binding; solid arrows represent activation; and dashed lines show inhibition between modulators. Abbreviations used include cluster of differentiated (CD), CTL-associated protein 4 (CTLA-4), cyclooxygenase-2 (COX2), cytotoxic T lymphocyte (CTL), dendritic cell (DC), human leukocyte antigen G (HLA-G), hypoxia-inducible factor (HIF), indoleamine 2,3-dioxygenase (IDO), inducible nitric oxide synthase (iNOS), interleukin (IL), lymphocyte activation gene-3 (LAG-3), lectin-like transcript-1 (LLT1), M2 macrophage (M2), myelin and lymphocyte protein 2 (MAL2), major histocompatibility complex class I (MHC-I), myeloid-derived suppressor cells (MDSC), macrophage c-mer tyrosine kinase (Mertk), major histocompatibility complex (MHC), MHC-I related chain A (MICA), mucin 1 C-terminal (MUC1-C), natural killer cell (NK), nitric oxide (NO), reactive oxygen species (ROS), regulatory T cell (Treg), programmed death receptor-1 (PD-1), programmed death-ligand 1 (PD-L1), sialic acid binding Ig like lectin-10 (Siglec-10), signal regulatory protein α (SIRPA), T-cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3), transforming growth factor β (TGF-β), and UL16 binding protein 2 (ULBP2). It is worth mentioning the sizes of various cells may be disproportional to their actual dimensions.</p>
Full article ">
Back to TopTop