33 research outputs found

    Dynamical System Modeling of Immune Reconstitution after Allogeneic Stem Cell Transplantation Identifies Patients at Risk for Adverse Outcomes

    Get PDF
    AbstractSystems that evolve over time and follow mathematical laws as they evolve are called dynamical systems. Lymphocyte recovery and clinical outcomes in 41 allograft recipients conditioned using antithymocyte globulin (ATG) and 4.5-Gy total body irradiation were studied to determine if immune reconstitution could be described as a dynamical system. Survival, relapse, and graft-versus-host disease (GVHD) were not significantly different in 2 cohorts of patients receiving different doses of ATG. However, donor-derived CD3+ cell reconstitution was superior in the lower ATG dose cohort, and there were fewer instances of donor lymphocyte infusion (DLI). Lymphoid recovery was plotted in each individual over time and demonstrated 1 of 3 sigmoid growth patterns: Pattern A (n = 15) had rapid growth with high lymphocyte counts, pattern B (n = 14) had slower growth with intermediate recovery, and pattern C (n = 10) had poor lymphocyte reconstitution. There was a significant association between lymphocyte recovery patterns and both the rate of change of donor-derived CD3+ at day 30 after stem cell transplantation (SCT) and clinical outcomes. GVHD was observed more frequently with pattern A, relapse and DLI more so with pattern C, with a consequent survival advantage in patients with patterns A and B. We conclude that evaluating immune reconstitution after SCT as a dynamical system may differentiate patients at risk of adverse outcomes and allow early intervention to modulate that risk

    Comparison of Myeloid Cells in Circulation and in the Tumor Microenvironment of Patients with Colorectal and Breast Cancers

    Get PDF
    We have previously reported levels of myeloid cells in the periphery and in the tumor microenvironment (TME) of patients with primary breast cancer (PBC) and colorectal cancer (CRC). We found that both PBC and CRC patients have significantly higher levels of granulocytic and immature myeloid cells in the TME. Additionally, we reported an expansion of circulating granulocytic myeloid cells in CRC patients, but not in PBC patients. In this report, we compared levels of myeloid cells between these two common cancers and have added data from more cancer patients. We also investigated associations between clinical stage/histological grade of tumors and levels of myeloid cells in cancer patients. We found that although granulocytic myeloid cells were expanded in the TME of both PBC and CRC patients, the levels of these cells were significantly higher in the TME of CRC patients. Moreover, our results indicate that increased levels of circulating granulocytic myeloid cells are associated with poorly differentiated tumors in CRC patients. Taken together, this work suggests that CRC patients may benefit more from the development of therapeutic agents to promote myeloid cell differentiation or inhibition for the reversal of immune suppression

    Editorial of Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine

    No full text
    The global burden of influenza-associated respiratory mortality is higher than previous estimates, with over 0 [...

    Breast Cancer Cells and PD-1/PD-L1 Blockade Upregulate the Expression of PD-1, CTLA-4, TIM-3 and LAG-3 Immune Checkpoints in CD4+ T Cells

    No full text
    Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype, and it exhibits resistance to common breast cancer therapies. Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1 (PD-1) and its ligand, PD-L1, have been approved to treat various cancers. However, the therapeutic efficacy of targeting PD-1/PD-L1 axis in breast cancer is under clinical investigation. In addition, the mechanisms of action of drugs targeting PD-1 and PD-L1 have not been fully elucidated. In this study, we investigated the effect of human TNBC cell lines, MDA-MB-231 and MDA-MB-468, and the non-TNBC cell line, MCF-7, on the expression of immune checkpoints (ICs) on CD4+ T cell subsets, including regulatory T cells (Tregs), using a co-culture system. We also examined the effect of blocking PD-1 or PD-L1 separately and in combination on IC expression by CD4+ T cell subsets. We found that breast cancer cells upregulate the expression of ICs including PD-1, cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and lymphocyte activation gene-3 (LAG-3) in CD4+ T cell subsets. We also found that the co-blockade of PD-1 and PD-L1 further upregulates the co-expression of TIM-3 and LAG-3 on CD4+CD25+ T cells and CD4+CD25+FoxP3+Helios+ Tregs in the presence of TNBC cells, but not in non-TNBC cells. Our results indicate the emergence of compensatory inhibitory mechanisms, most likely mediated by Tregs and activated non-Tregs, which could lead to the development of TNBC resistance against PD-1/PD-L1 blockade

    Intrinsic and acquired cancer immunotherapy resistance

    No full text
    Cancer immunotherapies, such as immune checkpoint inhibitors (ICIs), have revolutionized the treatment of various cancers and have shown a great efficacy in inducing antitumor immunity. Cancer immunotherapy in the form of adoptive cell transfer (ACT) have also been developed to eradicate tumor cells in a specific and effective manner, and it includes the administration of autologous tumor-infiltrating T-cells (TILs), T-cell receptor (TCR)-modified T-cells, or genetically engineered chimeric antigen receptor (CAR)-specific T-cells (CARTs) in cancer patients. Additionally, cancer vaccines and recombinant cytokines can be used as monotherapy or adjuvant therapy. Despite the success of immunotherapies in treating various solid tumors and hematologic malignancies, a significant number of patients do not benefit from these therapies and exhibit limited or no response. Some cancer patients do not respond to immunotherapies as a result of primary or intrinsic tumor resistance, while others respond to immunotherapies but develop resistance over time, referred to as adaptive or acquired tumor resistance. Tumor intrinsic- and extrinsic-mediated mechanisms, including genetic and epigenetic alterations, tumor-mutational loads, overexpression of co-inhibitory immune checkpoints, and elevated levels of suppressive immune cells and cytokines, can lead to a compromised antitumor immunity favoring tumorigenesis and cancer progression. This chapter outlines mechanisms of intrinsic tumor resistance and the emergence of acquired tumor resistance to cancer immunotherapies. Moreover, this chapter describes combined cancer immunotherapies, which may offer a great therapeutic potential to overcome tumor resistance against therapy and improve clinical outcomes in cancer patients. +é(c) 2022 Elsevier Inc. All rights reservedCancer immunotherapies, such as immune checkpoint inhibitors (ICIs), have revolutionized the treatment of various cancers and have shown a great efficacy in inducing antitumor immunity. Cancer immunotherapy in the form of adoptive cell transfer (ACT) have also been developed to eradicate tumor cells in a specific and effective manner, and it includes the administration of autologous tumor-infiltrating T-cells (TILs), T-cell receptor (TCR)-modified T-cells, or genetically engineered chimeric antigen receptor (CAR)-specific T-cells (CARTs) in cancer patients. Additionally, cancer vaccines and recombinant cytokines can be used as monotherapy or adjuvant therapy. Despite the success of immunotherapies in treating various solid tumors and hematologic malignancies, a significant number of patients do not benefit from these therapies and exhibit limited or no response. Some cancer patients do not respond to immunotherapies as a result of primary or intrinsic tumor resistance, while others respond to immunotherapies but develop resistance over time, referred to as adaptive or acquired tumor resistance. Tumor intrinsic- and extrinsic-mediated mechanisms, including genetic and epigenetic alterations, tumor-mutational loads, overexpression of co-inhibitory immune checkpoints, and elevated levels of suppressive immune cells and cytokines, can lead to a compromised antitumor immunity favoring tumorigenesis and cancer progression. This chapter outlines mechanisms of intrinsic tumor resistance and the emergence of acquired tumor resistance to cancer immunotherapies. Moreover, this chapter describes combined cancer immunotherapies, which may offer a great therapeutic potential to overcome tumor resistance against therapy and improve clinical outcomes in cancer patients. +é(c) 2022 Elsevier Inc. All rights reserve

    Intratumoral FoxP3+Helios+ Regulatory T Cells Upregulating Immunosuppressive Molecules Are Expanded in Human Colorectal Cancer

    No full text
    Regulatory T cells (Tregs) can be antitumorigenic or pro-tumorigenic in colorectal cancer (CRC) depending on the presence of different Treg subsets with various immunosuppressive molecules. Some studies reported the phenotypic characteristics of tumor-infiltrating immune cells in CRC, but limited studies have focused on the co-expression of suppressive molecules on immune cells. The aim of this study was to characterize immune cells in the tumor microenvironment (TME), compared to paired adjacent non-tumor colon tissue of CRC patients. Additionally, we investigated co-expression of immunosuppressive molecules on different Treg subsets in the TME, normal colon tissue, and peripheral blood of CRC patients and healthy donors. In this preliminary study, we report that the majority of CD3+ T cells in the TME are CD4+ T cells with high co-expression of programmed death 1 (PD-1)/cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and PD-1/CD39 molecules. Levels of CD4+FoxP3+Helios+ Tregs were significantly increased in the TME. Furthermore, we observed increased levels of PD-1/CTLA-4 and PD-1/CD39 co-expressing cells within FoxP3+Helios+ and FoxP3+Helios− Treg subsets, indicative of their potent immunosuppressive potential. These results suggest synergistic associations between PD-1/CTLA-4 and PD-1/CD39 in dampening T-cell activation and function along with suppressing tumor-specific immune responses, suggesting that dual blockade of these molecules could be a more effective strategy for inducing antitumor immune responses in CRC

    Correction to: Metabolic reprogramming of T regulatory cells in the hypoxic tumor microenvironment (Cancer Immunology, Immunotherapy, (2021), 70, 8, (2103-2121), 10.1007/s00262-020-02842-y)

    No full text
    in the hypoxic tumor microenvironment, written by Varun Sasidharan Nair, Reem Saleh, Salman M. Toor, Farhan S. Cyprian and Eyad Elkord, was originally published electronically on the publisher’s internet portal on 03 February 2021 without open access. With the author(s)’ decision to opt for Open Choice the copyright of the article changed on 25 May 2021 to © The Author(s) 2021 and this article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The original article has been corrected

    Blockade of PD-1, PD-L1, and TIM-3 Altered Distinct Immune- and Cancer-Related Signaling Pathways in the Transcriptome of Human Breast Cancer Explants

    Get PDF
    Immune checkpoint inhibitors (ICIs) are yet to have a major advantage over conventional therapies, as only a fraction of patients benefit from the currently approved ICIs and their response rates remain low. We investigated the effects of different ICIs—anti-programmed cell death protein 1 (PD-1), anti-programmed death ligand-1 (PD-L1), and anti-T cell immunoglobulin and mucin-domain containing-3 (TIM-3)—on human primary breast cancer explant cultures using RNA-Seq. Transcriptomic data revealed that PD-1, PD-L1, and TIM-3 blockade follow unique mechanisms by upregulating or downregulating distinct pathways, but they collectively enhance immune responses and suppress cancer-related pathways to exert anti-tumorigenic effects. We also found that these ICIs upregulated the expression of other IC genes, suggesting that blocking one IC can upregulate alternative ICs, potentially giving rise to compensatory mechanisms by which tumor cells evade anti-tumor immunity. Overall, the transcriptomic data revealed some unique mechanisms of the action of monoclonal antibodies (mAbs) targeting PD-1, PD-L1, and TIM-3 in human breast cancer explants. However, further investigations and functional studies are warranted to validate these findings

    DNA methylation and repressive histones in the promoters of PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, PD-L1, and galectin-9 genes in human colorectal cancer

    No full text
    Abstract Background Colorectal cancer (CRC) is the third most commonly diagnosed human malignancy worldwide. Upregulation of inhibitory immune checkpoints by tumor-infiltrating immune cells (TIICs) or their ligands by tumor cells leads to tumor evasion from host immunosurveillance. Changes in DNA methylation pattern and enrichment of methylated histone marks in the promoter regions could be major contributors to the upregulation of immune checkpoints (ICs) in the tumor microenvironment (TME). Methods Relative expressions of various immune checkpoints and ligands in colon normal tissues (NT) and colorectal tumor tissues (TT) were assessed by qRT-PCR. The epigenetic modifications behind this upregulation were determined by investigating the CpG methylation status of their promoter regions using bisulfite sequencing. Distributions of histone 3 lysine 9 trimethylation (H3K9me3) and histone 3 lysine 27 trimethylation (H3K27me3) in promoter regions of these genes were assessed by chromatin immunoprecipitation (ChIP) assay. Results We found that the expression levels of PD-1, CTLA-4, TIM-3, TIGIT, PD-L1, and galectin-9 were significantly higher in colorectal tumor tissues, compared with colon normal tissues. To study the role of DNA methylation, we checked the promoter CpG methylation of ICs and ligands and found that only CTLA-4 and TIGIT, among other genes, were significantly hypomethylated in TT compared with NT. Next, we checked the abundance of repressive histones (H3K9me3 and H3K27me3) in the promoter regions of ICs/ligands. We found that bindings of H3K9me3 in PD-1 and TIGIT promoters and H3K27me3 in CTLA-4 promotor were significantly lower in TT compared with NT. Additionally, bindings of both H3K9me3 and H3K27me3 in the TIM-3 promoter were significantly lower in TT compared with NT. Conclusion This study shows that both DNA hypomethylation and H3K9me3 and H3K27me3 repressive histones are involved in upregulation of CTLA-4 and TIGIT genes. However, repressive histones, but not DNA hypomethylation, are involved in upregulation of PD-1 and TIM-3 genes in CRC tumor tissue. These epigenetic modifications could be utilized as diagnostic biomarkers for CRC
    corecore