14 research outputs found

    Targeting Amino Acid Metabolic Vulnerabilities in Myeloid Malignancies

    Get PDF
    Tumor cells require a higher supply of nutrients for growth and proliferation than normal cells. It is well established that metabolic reprograming in cancers for increased nutrient supply exposes a host of targetable vulnerabilities. In this article we review the documented changes in expression patterns of amino acid metabolic enzymes and transporters in myeloid malignancies and the growing list of small molecules and therapeutic strategies used to disrupt amino acid metabolic circuits within the cell. Pharmacological inhibition of amino acid metabolism is effective in inducing cell death in leukemic stem cells and primary blasts, as well as in reducing tumor burden in in vivo murine models of human disease. Thus targeting amino acid metabolism provides a host of potential translational opportunities for exploitation to improve the outcomes for patients with myeloid malignancies

    Stress induced transcriptional regulation of the glycine transporter type 1A (GlyT-1A/SLC6A9) in human intestinal epithelia

    Get PDF
    Phd ThesisThere is mounting experimental evidence demonstrating protection by free glycine against stress in several cell types. The glycine transporter type 1 (GlyT-1) mediates the high affinity supply of glycine, which together with cysteine is required for the synthesis of the antioxidant glutathione. Previous work in this laboratory has established that GlyT-1 is expressed on the apical and basal membranes of intestinal epithelial cells and that its mRNA levels are regulated by stress. In the present study exactly how stress signals to transcriptional induction of GlyT-1 was investigated. Caco-2 cells transfected with reporter constructs of sequences of the GlyT-1a proximal promoter and 5’UTR cloned upstream of a ÎČ-galactosidase coding sequence, showed increased reporter activity following treatment with thapsigargin (Tg), tunicamycin (Tu), amino acid (AA) starvation, tert-Butylhydroquinone (tBHQ) or Diethyl maleate (DEM). Despite no changes in Nrf-2 mRNA levels, a significant increase in total Nrf-2 protein abundance was evident on western-blots following DEM treatment of Caco-2 cells. However, gel shift showed no protein-DNA complexes between Nrf-2 protein and a DNA probe sequence of the putative antioxidant response element (ARE) identified in the GlyT-1a 5’ flank. Despite a significant siRNA mediated knock-down of Nrf-2 mRNA and protein, there was no further effect on GlyT-1a expression. Unlike Nrf-2, the knock-down of Atf-4 diminished the basal and stressed induced expression of GlyT-1a. Atf-4 was detected bound to DNA probes containing a potential amino acid response element (AARE) located in the first exon of the GlyT-1a gene by gel shift and super shift assays. QPCR assays performed on DNA isolated from Caco-2 cells by chromatin immunoprecipitation (ChIP) using antibodies against Atf-4, demonstrated 9, 5 and 2-fold enrichment of the GlyT-1a AARE following Tu, AA starvation and DEM treatment respectively. Site directed mutation of the GlyT-1a AARE showed a 75% reduction in reporter activity as well as attenuated protein-DNA interaction with a representative probe. It is evident from the data presented in this thesis that the direct interaction of Atf-4 at the proposed GlyT-1a AARE contributes to its transcriptional up-regulation following endoplasmic reticulum stress, nutrient stress and oxidative stress

    MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers

    Get PDF
    Background: Targeting of MDSCs is a major clinical challenge in the era of immunotherapy. Antibodies which deplete MDSCs in murine models can reactivate T cell responses. In humans such approaches have not developed due to difficulties in identifying targets amenable to clinical translation. Methods: RNA-sequencing of M-MDSCs and G-MDSCs from cancer patients was undertaken. Flow cytometry and immunohistochemistry of blood and tumours determined MDSC CD33 expression. MDSCs were treated with Gemtuzumab ozogamicin and internalisation kinetics, and cell death mechanisms determined by flow cytometry, confocal microscopy and electron microscopy. Effects on T cell proliferation and CAR-T cell anti-tumour cytotoxicity were identified in the presence of Gemtuzumab ozogamicin. Findings: RNA-sequencing of human M-MDSCs and G-MDSCs identified transcriptomic differences, but that CD33 is a common surface marker. Flow cytometry indicated CD33 expression is higher on M-MDSCs, and CD33+ MDSCs are found in the blood and tumours regardless of cancer subtype. Treatment of human MDSCs leads to Gemtuzumab ozogamicin internalisation, increased p-ATM, and cell death; restoring T cell proliferation. Anti-GD2-/mesothelin-/EGFRvIII-CAR-T cell activity is enhanced in combination with the anti-MDSC effects of Gemtuzumab ozogamicin. Interpretation: The study identifies that M-MDSCs and G-MDSCs are transcriptomically different but CD33 is a therapeutic target on peripheral and infiltrating MDSCs across cancer subtypes. The immunotoxin Gemtuzumab ozogamicin can deplete MDSCs providing a translational approach to reactivate T cell and CAR-T cell responses against multiple cancers. In the rare conditions of HLH/MAS gemtuzumab ozogamicin provides a novel anti-myeloid strategy. Fund: This work was supported by Cancer Research UK, CCLG, Treating Children with Cancer, and the alumni and donors to the University of Birmingham. (c) 2019 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/)

    G-CSF induces CD15(+) CD14(+) cells from granulocytes early in the physiological environment of pregnancy and the cancer immunosuppressive microenvironment

    Get PDF
    OBJECTIVES: Recombinant granulocyte colony‐stimulating factor (G‐CSF) is frequently administered to patients with cancer to enhance granulocyte recovery post‐chemotherapy. Clinical trials have also used G‐CSF to modulate myeloid cell function in pregnancy and inflammatory diseases. Although the contribution of G‐CSF to expanding normal granulocytes is well known, the effect of this cytokine on the phenotype and function of immunosuppressive granulocytic cells remains unclear. Here, we investigate the impact of physiological and iatrogenic G‐CSF on an as yet undescribed granulocyte phenotype and ensuing outcome on T cells in the settings of cancer and pregnancy. METHODS: Granulocytes from patients treated with recombinant G‐CSF, patients with late‐stage cancer and women enrolled on a trial of recombinant G‐CSF were phenotyped by flow cytometry. The ability and mechanism of polarised granulocytes to suppress T‐cell proliferation were assessed by cell proliferation assays, flow cytometry and ELISA. RESULTS: We observed that G‐CSF leads to a significant upregulation of CD14 expression on CD15(+) granulocytes. These CD15(+)CD14(+) cells are identified in the blood of patients with patients undergoing neutrophil mobilisation with recombinant G‐CSF, and physiologically in women early in pregnancy or in those treated as a part of a clinical trial. Immunohistochemistry of tumor tissue or placental tissue identified the expression of G‐CSF. The G‐CSF upregulates the release of reactive oxygen species (ROS) in CD15(+)CD14(+) cells leading to the suppression of T‐cell proliferation. CONCLUSIONS: G‐CSF induces a population of ROS(+) immunosuppressive CD15(+)CD14(+) granulocytes. Strategies for how recombinant G‐CSF can be scheduled to reduce effects on T‐cell therapies should be developed in future clinical studies

    Targeting the arginine metabolic brake enhances immunotherapy for leukaemia

    Get PDF
    Therapeutic approaches which aim to target Acute Myeloid Leukaemia through enhancement of patients’ immune responses have demonstrated limited efficacy to date, despite encouraging preclinical data. Examination of AML patients treated with azacitidine (AZA) and vorinostat (VOR) in a Phase II trial, demonstrated an increase in the expression of Cancer‐Testis Antigens (MAGE, RAGE, LAGE, SSX2 and TRAG3) on blasts and that these can be recognised by circulating antigen‐specific T cells. Although the T cells have the potential to be activated by these unmasked antigens, the low arginine microenvironment created by AML blast Arginase II activity acts a metabolic brake leading to T cell exhaustion. T cells exhibit impaired proliferation, reduced IFN‐γ release and PD‐1 up‐regulation in response to antigen stimulation under low arginine conditions. Inhibition of arginine metabolism enhanced the proliferation and cytotoxicity of anti‐NY‐ESO T cells against AZA/VOR treated AML blasts, and can boost anti‐CD33 Chimeric Antigen Receptor‐T cell cytotoxicity. Therefore, measurement of plasma arginine concentrations in combination with therapeutic targeting of arginase activity in AML blasts could be a key adjunct to immunotherapy

    PARC:a phase I/II study evaluating the safety and activity of pegylated recombinant human arginase BCT-100 in relapsed/refractory cancers of children and young adults

    Get PDF
    Background: The survival for many children with relapsed/refractory cancers remains poor despite advances in therapies. Arginine metabolism plays a key role in the pathophysiology of a number of pediatric cancers. We report the first in child study of a recombinant human arginase, BCT-100, in children with relapsed/refractory hematological, solid or CNS cancers.Procedure: PARC was a single arm, Phase I/II, international, open label study. BCT-100 was given intravenously over one hour at weekly intervals. The Phase I section utilized a modified 3 + 3 design where escalation/de-escalation was based on both the safety profile and the complete depletion of arginine (defined as adequate arginine depletion; AAD <8ÎŒM arginine in the blood after 4 doses of BCT-100). The Phase II section was designed to further evaluate the clinical activity of BCT-100 at the pediatric RP2D determined in the Phase I section, by recruitment of patients with pediatric cancers into 4 individual groups. A primary evaluation of response was conducted at eight weeks with patients continuing to receive treatment until disease progression or unacceptable toxicity.Results: 49 children were recruited globally. The Phase I cohort of the trial established the Recommended Phase II Dose of 1600U/kg iv weekly in children, matching that of adults. BCT-100 was very well tolerated. No responses defined as a CR, CRi or PR were seen in any cohort within the defined 8 week primary evaluation period. However a number of these relapsed/refractory patients experienced prolonged radiological SD.Conclusion: Arginine depletion is a clinically safe and achievable strategy in children with cancer. The RP2D of BCT-100 in children with relapsed/refractory cancers is established at 1600U/kg intravenously weekly and can lead to sustained disease stability in this hard to treat population.Clinical trial registration: EudraCT, 2017-002762-44; ISRCTN, 21727048; and ClinicalTrials.gov, NCT03455140

    Macrophage-derived IL-1ÎČ and TNF-α regulate arginine metabolism in neuroblastoma

    Get PDF
    © 2018 American Association for Cancer Research. Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1b and TNFa in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1b and TNFa established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1b and TNFa in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited

    Invariant NKT cells metabolically adapt to the acute myeloid leukaemia environment

    Get PDF
    Acute myeloid leukaemia (AML) creates an immunosuppressive environment to conventional T cells through Arginase 2 (ARG2)-induced arginine depletion. We identify that AML blasts release the acute phase protein serum amyloid A (SAA), which acts in an autocrine manner to upregulate ARG2 expression and activity, and promote AML blast viability. Following in vitro cross-talk invariant natural killer T (iNKT) cells become activated, upregulate mitochondrial capacity, and release IFN-Îł. iNKT retain their ability to proliferate and be activated despite the low arginine AML environment, due to the upregulation of Large Neutral Amino Acid Transporter-1 (LAT-1) and Argininosuccinate Synthetase 1 (ASS)-dependent amino acid pathways, resulting in AML cell death. T cell proliferation is restored in vitro and in vivo. The capacity of iNKT cells to restore antigen-specific T cell immunity was similarly demonstrated against myeloid-derived suppressor cells (MDSCs) in wild-type and Jα18−/− syngeneic lymphoma-bearing models in vivo. Thus, stimulation of iNKT cell activity has the potential as an immunotherapy against AML or as an adjunct to boost antigen-specific T cell immunotherapies in haematological or solid cancers
    corecore