11 research outputs found

    A blinded in vitro analysis of the intrinsic immunogenicity of hepatotoxic drugs: implications for preclinical risk assessment

    Get PDF
    In vitro preclinical drug-induced liver injury (DILI) risk assessment relies largely on use of hepatocytes to measure drug-specific changes in cell function or viability. Unfortunately, this does not provide indications towards the immunogenicity of drugs and/or the likelihood for idiosyncratic reactions in the clinic. This is because the molecular initiating event in immune DILI is an interaction of the drug-derived antigen with MHC proteins and the T-cell receptor. This study utilised immune cells from drug-naïve donors, recently established immune cell co-culture systems and blinded compounds with and without DILI liabilities to determine whether these new methods offer an improvement over established assessment methods for the prediction of immune-mediated DILI. Ten blinded test compounds (6 with known DILI liabilities; 4 with lower DILI liabilities) and five training compounds, with known T-cell-mediated immune reactions in patients, were investigated. Naïve T-cells were activated with 4/5 of the training compounds (nitroso sulfamethoxazole, vancomycin, Bandrowski's base and carbamazepine) and clones derived from the priming assays were activated with drug in a dose-dependent manner. The test compounds with DILI liabilities did not stimulate T-cell proliferative responses during dendritic cell-T-cell co-culture; however, CD4+ clones displaying reactivity were detected towards 2 compounds (ciprofloxacin and erythromycin) with known liabilities. Drug-responsive T-cells were not detected with the compounds with lower DILI liabilities. This study provides compelling evidence that assessment of intrinsic drug immunogenicity, although complex, can provide valuable information regarding immune liabilities of some compounds prior to clinical studies or when immune reactions are observed in patients

    Detection of Hepatic Drug Metabolite-Specific T-Cell Responses Using a Human Hepatocyte, Immune Cell Coculture System

    Get PDF
    Drug-responsive T-cells are activated with the parent compound or metabolites, often via different pathways (pharmacological interaction and hapten). An obstacle to the investigation of drug hypersensitivity is the scarcity of reactive metabolites for functional studies and the absence of coculture systems to generate metabolites in situ. Thus, the aim of this study was to utilize dapsone metabolite-responsive T-cells from hypersensitive patients, alongside primary human hepatocytes to drive metabolite formation, and subsequent drug-specific T-cell responses. Nitroso dapsone-responsive T-cell clones were generated from hypersensitive patients and characterized in terms of cross-reactivity and pathways of T-cell activation. Primary human hepatocytes, antigen-presenting cells, and T-cell cocultures were established in various formats with the liver and immune cells separated to avoid cell contact. Cultures were exposed to dapsone, and metabolite formation and T-cell activation were measured by LC-MS and proliferation assessment, respectively. Nitroso dapsone-responsive CD4+ T-cell clones from hypersensitive patients were found to proliferate and secrete cytokines in a dose-dependent manner when exposed to the drug metabolite. Clones were activated with nitroso dapsone-pulsed antigen-presenting cells, while fixation of antigen-presenting cells or omission of antigen-presenting cells from the assay abrogated the nitroso dapsone-specific T-cell response. Importantly, clones displayed no cross-reactivity with the parent drug. Nitroso dapsone glutathione conjugates were detected in the supernatant of hepatocyte immune cell cocultures, indicating that hepatocyte-derived metabolites are formed and transferred to the immune cell compartment. Similarly, nitroso dapsone-responsive clones were stimulated to proliferate with dapsone, when hepatocytes were added to the coculture system. Collectively, our study demonstrates the use of hepatocyte immune cell coculture systems to detect in situ metabolite formation and metabolite-specific T-cell responses. Similar systems should be used in future diagnostic and predictive assays to detect metabolite-specific T-cell responses when synthetic metabolites are not available

    T cell assays differentiate clinical and subclinical SARS-CoV-2 infections from cross-reactive antiviral responses

    Get PDF
    Identification of protective T cell responses against SARS-CoV-2 requires distinguishing people infected with SARS-CoV-2 from those with cross-reactive immunity to other coronaviruses. Here we show a range of T cell assays that differentially capture immune function to characterise SARS-CoV-2 responses. Strong ex vivo ELISpot and proliferation responses to multiple antigens (including M, NP and ORF3) are found in 168 PCR-confirmed SARS-CoV-2 infected volunteers, but are rare in 119 uninfected volunteers. Highly exposed seronegative healthcare workers with recent COVID-19-compatible illness show T cell response patterns characteristic of infection. By contrast, >90% of convalescent or unexposed people show proliferation and cellular lactate responses to spike subunits S1/S2, indicating pre-existing cross-reactive T cell populations. The detection of T cell responses to SARS-CoV-2 is therefore critically dependent on assay and antigen selection. Memory responses to specific non-spike proteins provide a method to distinguish recent infection from pre-existing immunity in exposed populations

    Investigation of SARS-CoV-2 faecal shedding in the community: a prospective household cohort study (COVID-LIV) in the UK

    Get PDF
    Background SARS-CoV-2 is frequently shed in the stool of patients hospitalised with COVID-19. The extent of faecal shedding of SARS-CoV-2 among individuals in the community, and its potential to contribute to spread of disease, is unknown. Methods In this prospective, observational cohort study among households in Liverpool, UK, participants underwent weekly nasal/throat swabbing to detect SARS-CoV-2 virus, over a 12-week period from enrolment starting July 2020. Participants that tested positive for SARS-CoV-2 were asked to provide a stool sample three and 14 days later. In addition, in October and November 2020, during a period of high community transmission, stool sampling was undertaken to determine the prevalence of SARS-CoV-2 faecal shedding among all study participants. SARS-CoV-2 RNA was detected using Real-Time PCR. Results A total of 434 participants from 176 households were enrolled. Eighteen participants (4.2%: 95% confidence interval [CI] 2.5–6.5%) tested positive for SARS-CoV-2 virus on nasal/throat swabs and of these, 3/17 (18%: 95% CI 4–43%) had SARS-CoV-2 detected in stool. Two of three participants demonstrated ongoing faecal shedding of SARS-CoV-2, without gastrointestinal symptoms, after testing negative for SARS-CoV-2 in respiratory samples. Among 165/434 participants without SARS-CoV-2 infection and who took part in the prevalence study, none had SARS-CoV-2 in stool. There was no demonstrable household transmission of SARS-CoV-2 among households containing a participant with faecal shedding. Conclusions Faecal shedding of SARS-CoV-2 occurred among community participants with confirmed SARS-CoV-2 infection. However, during a period of high community transmission, faecal shedding of SARS-CoV-2 was not detected among participants without SARS-CoV-2 infection. It is unlikely that the faecal-oral route plays a significant role in household and community transmission of SARS-CoV-2

    DEVELOPMENT OF IN VITRO CELL-BASED SYSTEMS TO INVESTIGATE DRUG IMMUNOGENICITY

    Get PDF
    Typically, drugs are too small to elicit an immune response and therefore the occurrence of drug-mediated reactions is extremely rare. Characteristically these reactions are inherently difficult to predict from only the pharmacology of a drug or from the genotype and phenotype of the patient. Current models such as the lymphocyte transformation cells and drug challenge tests fall short of providing a panacea due to their invasive nature and inaccurate predictive capabilities. The ability to model hypersensitivity reactions in vitro would therefore prove useful in the clinic and during design, preventing morbidity and mortality. An ideal model would mimic the immune microenvironment through incorporating drug, patient, and disease specific factors. In the studies described in this thesis, we aimed to develop tools for future hypersensitivity modelling, encompassing drug, patient and disease specific factors alongside the immune microenvironment. We focused on models which may help predict chemically reactive metabolite formation resulting in T-cell activation, incorporating drug and patient factors. We utilised drug-metabolite specific T-cell clones, donor specific antigen presenting cells and a metabolising system to demonstrate chemically reactive metabolite formation and subsequent T-cell activation. Dapsone-nitroso specific T-cell clones demonstrated strong proliferative responses when cultured in situ dapsone and a metabolising system. Furthermore, through mass spectrometry analysis, we demonstrate the formation of multiple chemically reactive metabolites within the cell culture medium. We next investigated how the tissue microenvironment is related to flucloxacillin accumulation and novel adduct formation in flucloxacillin-mediated drug induced liver injury. Utilising the HepaRG cell line and primary human hepatocytes, evidence suggests that MRP2 and P-gp expression and function may play a role in flucloxacillin accumulation in liver-like cells. Furthermore, utilising membrane transporter inhibitors, we demonstrated how disruption of transporters increases the formation of flucloxacillin modified proteins and peptides for possible T-cell presentation and activation, contributing to the DILI mechanism. We next aimed to incorporate the HLA-predisposition of flucloxacillin in drug induced liver injury alongside the target tissue, creating an autologous T-cell mediated cytotoxicity model. Through use of donor specific induced pluripotent stem cells, T-cell clones and antigen presenting cells, we were able to demonstrate flucloxacillin-specific T-cell mediated killing of HLA-B*57:01 hepatocyte-like cells. Finally, utilising traditional and well-defined T-cell models, we demonstrated how the use of a conventionally safe therapeutic moiety, green tea, may result in a hypersensivity reaction, given the right patient and disease factors. T-cell isolation from HLA-B*35:01 donors demonstrated a strong dose-proliferation relationship upon exposure to epigallocatechin gallate and green tea extract

    Investigating the Immune Basis of Green Tea Extract Induced Liver Injury in Healthy Donors Expressing HLA-B*35:01.

    No full text
    Epigallocatechin-3-O-gallate (EGCG) is the major component of green tea extract, commonly found in dietary supplements, and has been associated with immune-mediated liver injury. The purpose of this study was to investigate the immunogenicity of EGCG in healthy donors expressing HLA-B*35:01, and characterize EGCG responsive T-cell clones. We have shown that EGCG can prime peripheral blood mononuclear cells and T-cells from donors with and without the HLA-B*35:01 allele. T-cell clones were CD4+ve and capable of secreting Th1, Th2, and cytolytic molecules. These data demonstrate that EGCG can activate T-cells in vitro, suggesting a significant role in the pathogenesis of green tea extract induced liver injury

    T cell mediated hypersensitivity to previously tolerated iodinated contrast media precipitated by introduction of atezolizumab

    No full text
    Many adverse reactions associated with immune checkpoint inhibitor (ICI) treatments are immunologically driven and may necessitate discontinuation of the ICI. Herein, we present a patient who had been administered the radio contrast media amidotrizoate multiple times without issue but who then developed a Stevens-Johnson syndrome reaction after coadministration of atezolizumab. Causality was confirmed by a positive re-challenge with amidotrizoate and laboratory investigations that implicated T cells. Importantly, the introduction of atezolizumab appears to have altered the immunologic response to amidotrizoate in terms of the tolerance–elicitation continuum. Proof of concept studies demonstrated enhancement of recall responses to a surrogate antigen panel following in-vitro (healthy donors) and in-vivo (ICI patients) administrations of ICIs. Our findings highlight the importance of considering all concomitant medications in patients on ICIs who develop immune-mediated adverse reactions. In the event of some immune-related adverse reactions, it may be critical to identify the culprit antigen-forming entity that the ICIs have altered the perception of rather than simply attribute causality to the ICI itself in order to optimize both patient safety and treatment of malignancies
    corecore