口头摘要

IF 2.3 4区 医学 Q3 GENETICS & HEREDITY
{"title":"口头摘要","authors":"","doi":"10.1111/iji.12630","DOIUrl":null,"url":null,"abstract":"<p><b><span>Alison Cleaton</span></b>, <i>Emma Burrows, Kimberley Robinson, Michael Richardson, Deborah Pritchard, Tracey Rees</i></p><p><i>Welsh Blood Service, Ely Valley Road, Talbot Green, UK</i></p><p>Regular HLA antibody testing is undertaken for patients awaiting renal transplantation, using LABScreen™ HLA antibody assays. During the COVID-19 pandemic, we observed unexplained changes to some HLA antibody profiles. Investigation revealed that several patients had COVID-19 prior to the changes, therefore, a review of all patients on the transplant waiting list with known COVID-19 infection was undertaken. Sixty-six out of two hundred thirty-seven patients on the transplant waiting list had COVID-19 (March 2020–July 2022). The HLA antibody results from samples prior to and following COVID-19 infection were analysed for changes in existing HLA antibody levels (increased Luminex Median Fluorescent Intensity (MFI) values), or expanded antibody profiles (increased cRF). Fifty-two (78.8%) patients had no detectable change in cRF or MFI; five (7.6%) had changes in MFI (but no change in cRF); nine (13.6%) had changes in MFI and cRF. Two out of nine patients had no recorded prior sensitisation event; four had a previous transplant, four blood transfusions, four pregnancy; and three multiple sensitising events. All nine patients had sustained cRF changes in subsequent samples (follow up to December 2022). Three out of nine patients were consequently identified as having altered immunosuppression due to the COVID-19 infection; these patients had a 20%–76% rise in cRF and now all have a cRF 98%–100%. While the majority of patients awaiting kidney, transplantation had no change to their HLA antibody profile following COVID-19 infection, nine patients had an increase in cRF, which has not been transient. Reduction or withdrawal of immunosuppression to aid recovery from COVID-19 was identified as the cause for three patients.</p><p><b><span>Adrienne Seitz</span></b>, <i>Clive Carter, Brendan Clark, Richard Baker</i></p><p><i>Leeds Teaching Hospitals NHS Trust, Leeds, UK</i></p><p>The level of pre-transplant immune risk is assessed through measuring serum IgG HLA antibodies which can be produced by long lived plasma cells and memory B-cells. Memory B-cells can circulate without producing antibodies, therefore their contribution to the antibody pool may not be fully appreciated. We describe an in vitro method for improving the assessment of pretransplant risk through the non-specific stimulation of peripheral memory B-cells. Peripheral blood mononuclear cells from three unsensitised volunteers and six sensitised patients were cultured for 9 days with the toll-like receptor agonist R848 and interleukin-2. Cell culture supernatant was tested for IgG HLA antibodies using single antigen beads. This was compared with a matched serum sample. Resting Day-0 and stimulated Day-9 B-cell phenotypes were assessed using flow cytometry, confirming the switch to antibody secreting (CD24-CD38hi), class-switched memory (CD27+IgD-) and plasma (CD38+CD138+) cells. HLA Class I and II antibodies were found in the cell supernatant, and 65% were present in the matched serum sample. When the supernatant demonstrated additional HLA antibodies, these could be attributed to a previous transplant, or had been present in the patient's historic serum profile. We demonstrate a method that can uncover peripheral memory using technology accessible to most H&amp;I laboratories. This assay could be useful when assessing live donor pairs where the donor may repeat mismatches associated with pregnancy, and in re-grafts, prior to removal of ‘other unacceptable antigens’. Finally, it could be considered alongside delisting strategies in the context of novel peri-transplant agents.</p><p><b><span>Sophie Chambers</span></b>, <i>Robert Whittle, John Goodwin, Tim Key</i></p><p><i>NHS Blood and Transplant, Barnsley, UK</i></p><p>Differences in amino acids (aa) at positions 76–83 of exon II of HLA-B and a subset of HLA-A primarily account for the highly immunogenic public epitopes Bw4 and Bw6. Bw4-specific antibodies are commonplace in alloimmunised Bw6 homozygotes, whilst Bw6-specific antibodies are encountered in Bw4 homozygotes. As variation exists within the Bw4 complex it has been reported previously that in a proportion of Bw6 homozygous individuals with Bw4 specific antibodies, reactivity can be restricted to HLA Bw4 subtype epitopes. We report three individuals with a Bw4 phenotype who demonstrate Luminex Single Antigen Bead determined alloantibodies to Bw4 epitopes distinct from their own. For renal patient L bearing B13, reactivity was present to all Bw4-positive beads other than B13. Reactivity was consistent with Epitope 249 defined by aa at positions 82L + 145R/ 83R + 145R. Platelet refractory patient G bearing A*24:02 demonstrated reactivity to all Bw4 beads other than A24, consistent with Epitope 423 defined by 144Q. For allogeneic-HSCT patient F bearing B*27:05, reactivity was present to all Bw4 beads except B*27:05 with reactivity defined by aa 77N + 81A + 82L. Our observations highlight that Bw4 subtype epitope profiles should not be overlooked in Bw4-positive individuals, that single amino acid differences in the Bw4 complex appear sufficient to generate alloantibodies and differences outside aa positions 76–83 may contribute to antibody binding. Bw4 subtype antibodies can impact organ allocation in the renal transplant setting, restrict appropriate platelet support for immunological refractoriness and complicate donor selection in HLA-mismatched allogeneic-HSCT.</p><p><b><span>Ryan Stevens</span></b>, <i>Felicity May</i></p><p><i>Welsh Transplantation and Immunogenetics Laboratory, Pontyclun, UK</i></p><p>The Welsh Transplantation and Immunogenetics Laboratory maintain a register of local patients active on the national renal/pancreas waiting list. Previously, data pulled from local IT systems was distributed monthly to service users in PDF format and a printed copy was held locally. This system had multiple drawbacks, including lack of ability to easily interrogate/update the data. In collaboration with the NHS Wales Microsoft 365 Centre of Excellence, a digital solution was created using Power BI. This allows up-to-date data to be instantly available, which gives users a more accurate overview to support decision making, and eliminates the requirement for PDF/paper distribution. The Dashboard can only be accessed by approved users, and access can be restricted to specific datasets (e.g., dialysis unit staff can only view patients at that unit). The Dashboard consists of two pages. A ‘Statistics’ page graphically displays the Register by organ type, blood group, dialysis unit, and so forth. The ‘demographics’ page tabulates data within the Register, which can be ordered or filtered in a variety of ways to aid in patient selection for donor offers. Each patient also has a colour coded ‘Sample Status’ to highlight when routine antibody screening samples are due. Comments and/or attachments can be added to each patient entry. Since successful go live December 2022, the dashboard has been praised by staff and service users for ease of access of data to assist delivery of safe and efficient patient care, as well as reduced incidence of overdue samples.</p><p><b><span>Kirsty Clark</span></b>, <i>Jane Matthews, Claire Romaines, Ruth Chisman, Arash Akbarzad-Yousefi</i></p><p><i>NHSBT Newcastle, Newcastle upon Tyne, UK</i></p><p>Despite advancements in organ allocation and immunosuppression, cardiothoracic transplantation continues to have the lowest 5-year survival when compared to all other forms of solid organ transplantation. A key factor in allograft loss is the formation of <i>de novo</i> Donor Specific HLA Antibodies (dnDSA). Recently, research has been driven away from the conventional method of defining mismatch at the antigenic level and has instead focused on HLA eplet mismatches. This study aimed to evaluate the role of eplet mismatching within our local cardiothoracic patient cohort. A retrospective analysis of 2 years of cardiothoracic transplant data was performed. The antibody data from all eligible patients were reanalysed to investigate the role of eplet mismatch load and to identify possible high risk eplets. This study found no evidence to directly support the theory of eplet load mismatches. However, four previously identified high risk eplets (McCaughan et al., <i>American Journal of Transplantation</i> 2018) namely, 55PP, 52LR, 55R and 75S were confirmed within our patient cohort. Findings from this single centre study provide potential evidence of high-risk HLA eplet mismatches; all of which are present within the HLA-DQ locus, indicating possible high immunogenicity for mismatches at this locus. Particular attention to minimise HLA-DQ mismatches may reduce the incidence of dnDSA and subsequently, allograft loss. Whilst avoiding mismatches in cardiothoracic transplantation is not always possible, further understanding of this area could lead to refinements in post-transplant monitoring and immunosuppression regimens.</p><p><b><span>Charlotte A. Cambridge</span></b><sup>1</sup>, <i>Jonathan A.M. Lucas<sup>1</sup>, Xenia Georgiou<sup>1</sup>, Gabriel J. Benitez<sup>1</sup>, Neema P. Mayor<sup>1,2</sup>, Steven G.E. Marsh<sup>1,2</sup></i></p><p><i><sup>1</sup>Anthony Nolan Research Institute, London, UK; <sup>2</sup>UCL Cancer Institute, Royal Free Hospital, UK</i></p><p>Submission of novel sequences to the IPD-IMGT/HLA Database from patients with haematological malignancies is not permitted, unless confirmed in the germline. To investigate if sequencing blood and buccal DNA yields different results, we typed 47 patients in remission from malignant disease and 58 healthy donors for HLA-A, -B, -C and -E using PacBio SMRT sequencing. Higher Phred scores (33.2 vs. 32.7, <i>p</i> &lt; 0.05) and lower cluster diversity (0.06 vs. 0.11, <i>p</i> &lt; 0.0001) were observed in sequences from patient buccal DNA versus blood, indicating sequences of higher quality with fewer background errors. No differences were observed in donor blood or buccal material. Blood DNA generated better HLA typing results that were automatically accepted (80.7% vs. 79.8%) with less allele dropout (2.0% vs. 7.2%). HLA typing results were concordant between blood and buccal derived DNA, including four novel sequences observed in patient samples, confirming these as germline mutations. Next, we analysed sequences from blood DNA for patients in remission from malignant (<i>n</i> = 406) versus non-malignant (<i>n</i> = 46) disease across nine libraries containing ≥3 of HLA-A, -B, -C, -E, -F and -G. No significant differences in Phred score (33.60 vs. 33.56) or cluster diversity (0.05 vs. 0.06) were observed. Overall, there were no differences in final HLA typing results for blood and buccal DNA samples for the same individual, and no difference in sequence quality between malignant and non-malignant patient samples. All sample types, if taken at the point of remission, are reliable sources for HLA typing and identification of novel sequence variation.</p><p><b><span>Ravneet Kaur Bola</span></b>, <i>Madalina Pinzaru, Marlowe Macadangdang, Ufot Udoffia, Sandra Frater, Franco Tavarozzi</i></p><p><i>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>Buccal epithelial cells are used for germline HLA testing, particularly useful in patients where loss of heterozygosity (LOH) is suspected. Anthony Nolan's (AN) patient buccal collection method utilises CytoSoft® brushes, however, we are always looking for ways to optimise our processes and realise that the design of the brushes may lead to an increased chance of blood contamination with some patients, potentially confounding HLA typing in LOH cases.</p><p>AN uses a gentler collection tool for our registry donor recruitment, FLOQSwabs®, therefore a trial was devised to identify the best buccal collection method for our patients, collaborating with the Royal Marsden Hospital. Each consented patient was swabbed using three scenarios: (1) FLOQSwabs®, cheek; (2) FLOQSwabs®, left gutter; (3) CytoSoft® brushes, right gutter. Samples from 54 patients were processed. DNA was extracted from each swab and one extraction per patient was selected for Next Generation Sequencing (NGS) in a pre-determined sequence, producing equal amounts of NGS data per scenario. Assessment looked at DNA extraction and NGS failure rates, together with DNA quantity produced.</p><p>DNA extraction failure rates were 9.3%, 17.6%, 29.9% for the three scenarios respectively.</p><p>On average, the DNA concentration from FLOQSwabs® were at least two times higher than with CytoSoft® brushes. Due to NGS robustness, all extractions selected for NGS were successfully typed. In summary, FLOQSwabs® swabbed from the cheek are the preferred collection method. These produced the lowest number of failed DNA extractions, while providing a softer and gentler swabbing experience for the patient.</p><p><b><span>Claire Lenehan</span></b><i>, James Kelleher, David Keegan, Mary Keogan, Khairin Khalib</i></p><p><i>NHISSOT Beaumont Hospital, Dublin, Ireland</i></p><p>Antibodies to human leukocyte antigens (HLA) are a complication for transplantation. The introduction of Luminex® technology has allowed for precise characterization of these antibodies with high sensitivity. The clinical significance of these antibodies remains controversial due to the detection of biologically irrelevant antibodies directed against denatured HLA molecules (dHLA). We aimed to determine the correlation of donor specific antibodies (DSA) detected by Luminex single antigen bead assay with flow cross match (FXCM) results, using a previously published protocol, with clinically validated cut-offs.</p><p>The capability of HLA antibodies in 170 non-classically sensitised patients to result in a positive crossmatch was investigated. Forty-nine FCXM against incompatible donor cells were completed. Fifty-six percent of T cell and 61% of B cell FCXM were positive. The rate of positive FCXM results was significantly higher for HLA-A and -B antibodies (<i>p</i> = 0.013). MFI value was a poor predictor of FCXM results. Conversely, the rate of negative FCXM results for DSA with MFI values of 5000 to 10,000 was not significantly different to that of MFI values &lt;5000. The rate of positive FCXM was higher for patients with DSA MFI &gt; 10,000. A significant proportion of the FCXM results were positive regardless of MFI strength indicating that DSA in non-classically sensitised patients are not limited to reactivity to dHLA. FXCM facilitates risk assessment. Based on the data in this study, we now offer a FCXM in living donors, or highly sensitised patients when DSA MFI is less than 10,000.</p><p><b><span>Kay Poulton</span>\n <span>,</span></b><sup>1,2</sup> <i>Madeleine Harris<sup>1</sup>, Andrew Canterbury<sup>2</sup>, Marie Hampson<sup>1</sup>, Judith Worthington<sup>1</sup>, Marcus Russell-Lowe<sup>1</sup></i></p><p><i><sup>1</sup>Manchester Royal Infirmary, Oxford Road, UK, <sup>2</sup>MC Diagnostics Limited, St Asaph, Wales, <sup>3</sup>University of Manchester, Manchester, UK</i></p><p>Single Antigen Bead assays have revolutionised the identification and definition of HLA-specific antibodies. They have enabled the widespread use of virtual crossmatching and almost eliminated hyperacute rejection due to HLA incompatibility during the 20 years since their introduction. But for highly sensitised patients, there is a low likelihood of finding a compatible donor. The only option for some patients is strategic de-listing of specificities which may impact the lowest immunological risk to a new graft. In this study, 257 serum samples from 82 potential recipients were crossmatched against cells from 84 potential donors. Only results where serum samples have been tested by LABScreen, HISTO SPOT® HLA AB, Complement Dependent Cytotoxicity (CDC) and Flow Cytometry were included in the analysis. The results were analysed to assess the ability of HISTO SPOT® HLA AB to predict the crossmatch result. Of the 136 samples analysed, 17 (12.5%) were CDC positive, and 82 (60.3%) were positive by flow cytometry. Twenty-eight sera tested negative using HISTO SPOT. All of these (100%) were also negative by CDC and 25 (89.3%) were also negative by Flow Cytometry. Three sera which tested negative by HISTO SPOT with a positive flow cytometry crossmatch were CDC negative, each had cumulative MFIs of &lt;3000 against Donor Specific Antigens. In this early study, HISTO SPOT® HLA AB has 100% negative predictive value for CDC crossmatch negativity and 89.3% by Flow Cytometry. It may therefore prove a useful additional tool to inform de-listing strategies used to facilitate transplantation in highly sensitised patients.</p><p><b><span>David Wimbury</span></b></p><p><i>Transplant Laboratory, University Hospitals of Leicester NHS Trust, Leicester, UK</i></p><p>Previous work has shown that variations in temperature, incubation time and operator methodology can have a drastic effect on results in solid phase assays. In HLA antibody screening, differences in overall MFI levels could impact the number of antibodies detected as positive and/or their risk level, thereby potentially affecting transplantation opportunities. Normalisation of MFI in each sample to a baseline could create more consistent results between samples at different time points, reducing the measurement uncertainty. This method normalises overall sample MFIs to a baseline. Positive control samples and three patients with extensive sample histories were tested with this methodology. CVs of MFIs before and after normalisation were analysed with paired <i>t</i>-tests.</p><p>Overall, MFI data became more comparable between samples post-normalisation. MFI ranges on a bead-by-bead basis initially varied between runs from around 500 to as much as 20,000. This was decreased after normalisation; the average MFI range between runs decreased by 1337 for Class I and by 2402 for Class II. All samples showed a statistically significant decrease in coefficient variation (CV) of MFIs between runs after normalisation, both Class I and Class II (<i>p</i> &lt; 0.0001). Inconsistencies in HLA antibody screening results between samples from the same patient can make for troublesome clinical interpretations, especially when there is no sensitising event accounting for reactivity changes. The procedure shown here provides a novel method for reducing the variability caused by differences in assay conditions and has the potential to give more consistent results therefore providing a clearer clinical picture.</p><p><b><span>Deborah Pritchard</span></b></p><p><i>Welsh Transplantation Laboratory, Pontyclun, UK</i></p><p>Immunological compatibility testing for potential live kidney donors and patients requires multiple tests on patient and donor samples (HLA typing, HLA antibody testing, crossmatch assessment). Only on completion of all tests can a report be produced. The local KPI is 90% of cases reported in a turnaround time (TAT) of 10 working days, but performance since Q1 2021/22 has been &lt;75%. This quality improvement project (May–August 2022) identified delays from process maps and selected two areas as targets for improvement; antibody testing and report production. Two plan-do-study-act (PDSA) cycles were performed testing six change ideas generated from staff workshops. Changes included identification of case samples within the testing process to allow more efficient workflow; a dedicated individual with responsibility for reporting rather than shared responsibility; E-mail notification when tests have been completed; coordination of HLA antibody batch testing with sample arrival to reduce wait time, and introduction of visualisation boards to track cases more efficiently. One ‘just do it’ change was also introduced; an interface to import antibody results from HLA Fusion software to the laboratory information management system (LIMS). The mean TAT was reduced from 11.9 to 8.8 days. Improvements were seen in the antibody testing mean TAT (9.1 days→6 days) and report production TAT (3.6 days→2.2 days). This resulted in compliance with the KPI: 95% of cases were reported in 10 days (October–December 2022). The improvement was due to removing manual data entry processes, reducing wait times between procedures and eliminating duplication of work.</p><p><b><span>Evelien Little</span></b>, <i>Nicola Brosnan, Jade Kally, Adrian Silk, Lisa Walsh, Franco Tavarozzi</i></p><p><i>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>With the onset of Next Generation Sequencing (NGS), HLA typing has become more streamlined with the ability to use either singleplex or multiplex primer strategies. The Anthony Nolan Histocompatibility Laboratory utilises GenDx NGSgo® singleplex primers for NGS to achieve high-allelic resolution typing for HLA-A, -B, -C, -DRB1, -DRB3,4,5, -DQB1, -DQA1, -DPA1, and -DPB1. Prior to this work, all samples received for HLA typing were tested for 11 loci, but the number of loci requested may vary. The use of singleplex, as opposed to multiplex, primers has allowed us to implement a flexible approach to provide a tailored and bespoke service. The aim of this work was to develop a process to type samples that need various combinations of testing on the same run. Modifications were needed to our Laboratory Information Management System (LIMS) to allow significant changes throughout the NGS testing process. As a result of this project and as our ongoing flexibility improvement process, we also implemented the ability to repeat PCR failures within the same run, allowing the process to proceed without delay, and merge multiple NGS libraries.</p><p>Since we no longer perform unnecessary testing, this generated cost savings by increased capacity on our Illumina® flow cells, allowing more samples per run, potentially reducing our turnaround times, as well as meeting our customer needs. To conclude, singleplex primers have proved extremely useful in allowing flexibility with no discernible increase in run time, using LIMS to simplify and track the process, end to end.</p><p><b><span>Victoria Wood</span></b><sup>1,2</sup>, <i>Brendan Clark<sup>1</sup>, Eric Hewitt<sup>2</sup>, Sunil Daga<sup>1</sup></i></p><p><i><sup>1</sup>Leeds Teaching Hospitals NHS Trust, Leeds, UK; <sup>2</sup>University of Leeds, Leeds, UK</i></p><p>A core aspect of a renal Transplant Immunology service is the detection of HLA antibodies in the context of prospective donors. The presence of HLA-antibodies is detrimental to chances of transplant and, as donor-specific antibodies (DSA), to graft outcomes. With growing numbers of highly sensitised patients requiring renal transplant across HLA-antibody barriers, an increased understanding of antibody functional characteristics could lead to more informed donor choices. The avidity of an antibody-antigen interaction provides insight into the antibody's capability to induce antibody-mediated changes, such as intracellular signalling, leading to tissue remodelling and graft damage. Chaotropic agents reduce protein stability and have been previously used within ELISA protocols to estimate avidity of antibody-antigen interactions through a process called chaotropic disruption.</p><p>Modification of our standard One Lambda LABScreen Single Antigen bead (SAB) protocol to include a chaotropic agent has demonstrated technical viability of applying chaotropic disruption to a solid-phase assay and that this manipulation can be confidently interpreted. A maximum molarity of chaotropic agent was established which is not detrimental to bead surface antigen integrity - 1 molar. This was verified using flow cytometric analysis with HCA2 monoclonal, which binds to both native and denatured HLA-Class I, in comparison with HLA-A, -B, -C monoclonal which binds to native HLA-Class I only. Initial testing using sera with known HLA antibody profiles showed antibody-specific patterns of binding and disassociation. This novel method represents a potential accessible method of testing HLA antibody avidity in an NHS laboratory setting.</p><p><b><span>Mr Steven Jervis</span></b><sup>1,2</sup>, <i>Dr Antony Payton<sup>3</sup>, Dr Marcus Lowe<sup>1,2</sup>, Dr Altug Didikoglu<sup>4</sup>, Professor Arpana Verma<sup>2</sup>, Professor Kay Poulton<sup>1,2</sup></i></p><p><i><sup>1</sup>Manchester Transplantation Laboratory, Oxford Road, United Kingdom</i>; \n<i><sup>2</sup>Faculty of Biology, Medicine and Health, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, University of Manchester, Manchester, United Kingdom; <sup>3</sup>Division of Informatics, Imaging &amp; Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; <sup>4</sup>The Centre for Biological Timing, Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, United Kingdom</i></p><p>Studies have hypothesised that the combination of pre-existing genetic traits and specific environmental triggers determine the onset of narcolepsy. The most impactful genetic risk factor is the presence of Human Leukocyte Antigen (HLA) DQB1*06:02 encoded on the Major Histocompatibility Complex (MHC), however, the presence of HLA-DQB1*06:02 is not ubiquitous in all narcolepsy cases. The most poignant genetic risk factors outside the MHC are predominantly located in genes associated with the immune system. In addition to the traditional symptoms of narcolepsy, the co-morbidities can vary with a cohort of sufferers complaining of cognitive dysfunction, particularly memory and attention. These self-reports are not substantiated by consistent scientific evidence whereas there is significant evidence outlining the genetic contribution underpinning variation in cognitive abilities in the general population. In this study we impute targeted non-MHC narcolepsy associated single nucleotide polymorphisms (SNPs) from 1,558 non-pathological elderly volunteers who have been followed for change in cognitive function for up to a 24-year period. Specifically, we investigate 13 previously documented narcolepsy associated SNPs with a odds ratio greater than or equal to 1.00 combined with a minor allele frequency of greater than 0.05. We observed an association between rs306336, rs4290173 and rs2834168 and a faster decline in long term memory. Similarly, we observed a protective effect of rs10995245 against the decline of long-term memory loss. This investigation suggests that the cognitive problems reported by cohorts of narcoleptic patients may be due to genetic predispositions and supports the variation seen in the co-morbidities associated with narcolepsy.</p><p><b><span>Raji Patel</span></b>, <i>Aliyye Karasu, Liezelle Pagala, Jyoti Bhatt, Edgar Correa, Gloria Adeyemo, Maame-Esi Yeboah, Abigail Sarkodie, Andrew Joahill, Suzette Cavanna, Carla Rosser, Colin Brown</i></p><p><i>NHS Blood and Transplant-Colindale, London, UK</i></p><p>The British Bone Marrow Registry (BBMR) utilises Oragene® saliva collection kits to facilitate extended HLA typing of potential donors by Next Generation Sequencing (NGS). Saliva is a good alternative source of DNA as its collection is non-invasive, allows for self-collection and is stable at ambient temperature. In January 2023, the Qiagen EZ2 robot was introduced into our laboratory to provide DNA extraction for clinical samples. However, Qiagen were unable to provide a validated protocol for the extraction of DNA from saliva samples using this instrument. In this study, we assessed the capability of Qiagen EZ2 tissue kits to obtain DNA from saliva samples using the Qiagen EZ2 Connect robot. Using the protocol, we developed, DNA was successfully extracted from 10 saliva samples, with an average DNA concentration of 33 ng/μl and a 260/280 ratio of 1.72. The quality of DNA was comparable to DNA extracted from saliva samples using the validated Roche Magnapure method. The suitability of saliva DNA for high resolution HLA typing was assessed using the One Lambda FASTplex kit. Using TypeStream Visual (TSV) NGS analysis software, we achieved average mapped reads of 334,691 and the results met our documented acceptance criteria for all quality metrics. In conclusion, we have demonstrated that the Qiagen EZ2 Connect robot can be used for successful extraction of DNA from saliva samples to a comparable quality of blood. Validation of this process has allowed our department to avoid unnecessary delays in BBMR extended HLA typing requests and thereby supports patient care.</p><p><b>Sebastian Fernando</b>, <i><span>Jennifer Lord</span>, Nicola Martin, Alison Logan, Kay Poulton</i></p><p><i>University of Manchester NHS Foundation Trust, Manchester, UK</i></p><p>Our current routine HLA typing methodology to support disease association testing is LABType™ SSO (One Lambda). An alternative HLA typing methodology EUROArray (EUROIMMUN), was evaluated for concordance and efficiency. This system combines polymerase chain reaction and microarray technologies. Amplified target DNA labelled with a fluorescent dye hybridises to complimentary DNA probes using BIOCHIP technology. Fluorescence signals are evaluated automatically using the EUROIMMUN Microarray Scanner and EUROArrayScan software. Routine and external proficiency scheme peripheral blood and DNA samples previously HLA typed by LABType™ SSO were tested using the appropriate EUROArray assay. Forty-six samples were tested with the HLA-B27 Direct assay, 41 samples with the HLA-B*57:01 Direct assay and 42 samples with the HLA-DQ2/DQ8-h Direct assay. A range of relevant HLA-B and HLA-DQ alleles were selected to ensure the EUROArray system could differentiate between alleles of interest. We found 100% concordance when compared to the LABType™ SSO results for all tests. The workflow was simple and straightforward with a time saving when compared to LABType™ SSO, however the cost of the EUROArray assay was higher. This study has shown the EUROArray Direct assays for HLA-B27, -B57 and -DQ2/8 detection are valid alternative methodologies to support disease association testing. Advantages of the EuroArray system include no additional DNA isolation, the inclusion of numerous integrated controls for high reliability of results and fully automated standardised evaluation and result generation. Limitations of this assay include the use of blood samples within 14 days of venepuncture, although samples can be frozen to mitigate this.</p><p><b><span>Amy De'Ath</span></b>, <i>Deborah Pritchard, Tracey Rees</i></p><p><i>UK NEQAS for H&amp;I, Talbot Green, UK</i></p><p>Scheme 2A and 2B assesses participants’ ability to correctly determine the cytotoxic and flow cytometry crossmatch status across 40 cell/serum combinations per year, respectively. An analysis of performance by UK and Ireland laboratories from 2018–2022 was performed.</p><p>There were between 15–22 participants in 2A and 19–22 in 2B. Unsatisfactory performance (UP) in 2A ranged between 0% (2020/21)–38.9% (2018), latest 20%. UP in 2B ranged from 0% (2020/21)–10.5% (2022). Over the 5 years, four laboratories had UP in 2B (one lab in two consecutive years) and 11 in 2A. 2 labs had UP in both schemes. Two out of four labs with UP in 2B were due to performance in the T-cell crossmatch, 1/4 in B-cell and 1/4 in T and B cell crossmatch performance. Six out of eleven labs with UP in 2A were in the B-cell without DTT category, 1/11 in B-cell with DTT, 3/11 in both B-cell with and without DTT and 1/11 in T-cell with and without DTT and B-cell with DTT. In 2B, an average of 36/40 crossmatch combinations per year were assessed. An average of 3% assignments per year were incorrect with a 50:50 split of false negatives and false positives. The use of equivocal reporting was low, average 0.5%. Four percent of samples were reported as not tested. Performance in flow cytometry crossmatching is better than cytotoxic crossmatching during the 5 years. These schemes offer a technical assessment of crossmatching, NEQAS encourage laboratories to participate in our educational schemes which more closely mimic clinical practice.</p><p><b><span>Amy Bedford</span></b>, <i>Kathryn Howson, Graham Knighton, Jacqueline Pires, Sarah Maxfield</i></p><p><i>Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK</i></p><p>HLA-B*27 testing is supportive in the diagnosis of autoimmune diseases including Ankylosing Spondylitis due to their strong association with the HLA-B*27 positive genotype. Testing an average of 383 samples per month, Cambridge Tissue Typing laboratory aimed to introduce a direct-from-blood HLA-B*27 detection assay to eradicate DNA extraction required by current polymerase chain reaction (PCR) sequence specific primer (SSP) based methods, and the associated workload. Utilising the BAG Diagnostics FastQ® B*27 Direct assay, DNA is amplified directly from EDTA whole blood by PCR using SSP. Fluorescent-labelled TaqMan® hydrolysis probes are utilised to enable detection of the amplified product by real-time PCR on the QuantStudioTM 3. Implementation of the FastQ® B*27 Direct assay reduced DNA extraction workload by an average of 64%. Increased batch size capacity (93, vs. 20 using the previous technique) streamlined workflows. Risks associated with exposure to carcinogens ethidium bromide and ultraviolet light and handling boiling agarose gel were eradicated. The absence of DNA extraction and adjustment leads to variance in the volume of nucleated cells utilised, which can result in delayed amplification/false negative results. This highlights the importance of sufficient sample mixing prior to aspiration and stringent result acceptance criteria during analysis using PlexTyper® software, with an average repeat rate of 4%. In summary, implementation of robust mixing procedures and stringent quality metrics have facilitated transition to BAG Diagnostics FastQ® B*27 Direct assay leading to a decrease in workload and demands on staff time, and improvement in the health and safety within the laboratory.</p><p><b><span>Kelly Spence</span></b>, <i>Sylvia McConnell, David Turner</i></p><p><i>H&amp;I Department, SNBTS, Edinburgh, UK</i></p><p>H&amp;I supports the Clinical Apheresis Unit (CAU) and Tissues, Cells and Advanced Therapeutics (TCAT) departments by performing CD34+ stem cell enumeration in acute myeloid leukaemia and multiple myeloma patients receiving autologous stem cell transplantation. The enumeration of CD34+ cells using flow cytometry and the International Society of Hematotherapy and Graft Engineering (ISHAGE) protocol is an established method for the evaluation of stem cell numbers in peripheral blood and apheresis products. Whilst a single (flow cytometry) platform is the recommended approach, locally a dual platform method is in operation, using both flow cytometry and Haematology analyser data. The BD Stem Cell Enumeration assay has been validated on the BD FACSLyric flow cytometer. A comparison between the single and dual platform methodologies using the ISHAGE protocol was undertaken. Analysis was performed on 30 stem cell harvest samples. Strong correlation between the single and dual platform methods was observed for CD34+ absolute count (cells/μl) for stem cell harvest samples (<i>r</i> = 0.99, <i>p</i> &lt; 0.05). There was also a strong correlation between the White Blood Cell (WBC) count from the Haematology analyser and the CD45+ count from the flow cytometer (<i>r</i> = 0.94, <i>p</i> &lt; 0.05). In this validation both the single and dual platform methods for calculating CD34+ cell counts were similar, as were the WBC counts and CD45+ counts from the Haematology analyser and the flow cytometer. This data supports using the recommended single platform method as part of the CD34+ enumeration testing.</p><p><b><span>Rebecca McGuire</span></b><sup>1,2</sup>, <i>Paul Wright<sup>3</sup>, Steven Jervis<sup>1</sup>, Stephine Whiteside<sup>1</sup>, Malcolm Guiver<sup>4</sup>, Kay Poulton<sup>1,2</sup></i></p><p><i><sup>1</sup>Transplantation Laboratory, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK; <sup>2</sup>Faculty of Biology, Medicine and Health, Division of Medical Education, School of Medical Sciences, University of Manchester, Manchester, UK; <sup>3</sup>H&amp;I Laboratory, Liverpool Clinical Laboratories, Liverpool, UK; <sup>4</sup>Department of Clinical Virology, Manchester University NHS Foundation Trust, Manchester, UK</i></p><p>Acute myeloid leukaemia is an aggressive haematological malignancy with a poor prognosis. Allogeneic haematopoietic progenitor cell transplantation is the only available curative treatment, but it does not eliminate the risk of relapse. Hypotheses for improving relapse rates include utilising donors with advantageous KIR2DL1 allele groups. However, an inexpensive, rapid, and reliable method to discern KIR2DL1 groups does not currently exist. Here, we show that homology between several Killer-cell Immunoglobulin-like Receptor (KIR) loci restricts accurate genotyping of KIR2DL1 allele groups by traditional TaqMan™- based real-time PCR methods. Using multiple sequence analysis, we found that the single nucleotide polymorphisms within codons 114 (rs11673144) and 245 (rs34721508), used to discern KIR2DL1 allele groups, are shared between other KIR loci. In a traditional TaqMan™ real-time PCR assay, this results in non-specific binding and incorrect allele group assignment. Contrary to existing literature, our analysis demonstrated that specificity could not be achieved through the introduction of 3´ terminal mismatches in the forward primers. The results described illustrate the challenges in designing an efficient real-time PCR assay for functional group typing of KIR2DL1. We anticipate our assay to provide the foundation for a more sophisticated real-time PCR assay. Developing a test capable of defining KIR2DL1 allele groups remains of interest to research groups focused on reducing the incidence of acute myeloid leukaemia relapse post-transplant. With further improvements, the described assay could fulfil this goal.</p><p><b><span>Abigail Levy</span></b>, <i>Arthi Anand, John Wintour-Pittom</i></p><p><i>Hammersmith H&amp;I NWLP, London, UK</i></p><p>The Histocompatibility &amp; Immunogenetics (H&amp;I) department at NWLP support renal transplantation at West London Renal Transplant centres. A 24/7 on-call service operates for crossmatching of local patients for transplantation. Timely and effective communication between the renal team receiving deceased donor offers and H&amp;I on-call team is critical for turnaround of compatibility assessment and testing. Historically H&amp;I on-call team have been contactable via ICHT switchboard with provision of a weekly on-call rota. A number of challenges were experienced using this method including calls being directed to the wrong team member interrupting rest and the potential for GDPR breach with patient identifiable information recorded in home environment. ICHT implementation of ALERTIVE app in June 2022 provided alternative on-call communication. H&amp;I were the first pathology service to adopt the app and collaborated with Trust Telecoms &amp; ThamesNet Services to set up the App to meet H&amp;I on-call needs, with Go-Live in February 2023. ALERTIVE improves the speed and quality of communication, between clinical staff and H&amp;I on-call team, in time critical deceased donor transplantation pathway. The ALERTIVE app has been in routine use for three months and has had a very favourable response from H&amp;I on-call team. The app has simplified the process of communicating between Renal and H&amp;I on-call teams, enabling more efficient decision-making. The clinical messaging app is giving us a wealth of new data that was either very hard to get or was unavailable including number of offer related call outs. There are exciting new uses for this data including workforce planning.</p><p><b><span>Dayna Badaro</span></b><sup>1,2</sup><i>, Sarah Maxfield<sup>1</sup></i></p><p><i><sup>1</sup>Cambridge University Hospital NHS Foundation Trust, Cambridge, UK; <sup>2</sup>University of Manchester, Manchester, UK; <sup>3</sup>Organ and Tissue Donation and Transplantation, NHS Blood and Transplant, Stoke Gifford, UK</i></p><p>In the United Kingdom, prior to September 2019, deceased donor kidneys were allocated following the National Health Service Blood and Transplant-Organ Donation and Transplantation (NHSBT-ODT) 2006 Kidney Allocation Scheme (KAS). A review by the Kidney Advisory Group prompted significant changes to national allocation with the aim of improving fairness in kidney offering, reducing transplant waiting times and improving longevity of matched transplants. A retrospective clinical audit aimed to review the impact for patients on the Cambridge deceased donor kidney transplant waiting list (TWL), and determine whether the objectives of the 2019 KAS have been met. Transplant data was obtained from NHSBT-ODT spanning 24 months pre-(<i>n</i> = 298) and post-(<i>n</i> = 303) implementation. Data was reviewed taking into consideration the impact of the COVID-19 pandemic. Results show reduced average wait time (days) for highly sensitised patients with a calculated reaction frequency (cRF) ≥85%. In accordance with simulation data, donor/recipient index matching has been achieved, with the majority of ‘low risk’ donors allocated to ‘low risk’ recipients, and the same being true for ‘high risk’ donors/recipients. Divergent donor/recipient age matching was accounted for by difficult to match patients prioritised according to Tier A (cRF 100%/wait time &gt;7 years/matchability score of 10). Contrary to simulation data, Cambridge did not observe a reduction in donation after cardiac death (DCD) transplants following changes to national allocation (<i>n</i> = 174 pre, <i>n</i> = 192 post). In summary data indicates a reduction in average wait time for highly sensitised patients on the Cambridge TWL and suggests objectives of the 2019 KAS have been met.</p><p><b><span>Michelle Carr</span></b><sup>1</sup>, <i>Shelley Harris<sup>1</sup>, Judith Worthington<sup>1</sup>, Alex Woywodt<sup>2</sup>, Kay Poulton<sup>1</sup></i></p><p><i><sup>1</sup>Transplant Laboratory MFT, Manchester, UK; <sup>2</sup>Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK</i></p><p>In January 2020 a 24-year-old male with chronic renal failure secondary to reflux nephropathy received his third kidney transplant from a fully HLA matched DBD donor. In March 2020 his kidney function declined with a concurrent rise in creatinine and proteinuria. Tacrolimus levels were variable throughout the course of the patient's transplant history and non-adherence was suspected. A retrospective analysis of non-HLA antibodies using LABScreen™ Autoantibody kits identified that the patient developed antibodies to Glutathione S-Transferase Theta 1 (GSTT1). After his second transplant tacrolimus levels were below target range (&lt;2.5 μg/L), which was followed by the first appearance of the GSTT1 antibody (6000 MFI = 95% percentile). This coincided with a biopsy showing chronic transplant glomerulopathy. Following his third transplant tacrolimus levels remained variable with some levels above and below target range. His transplant function deteriorated further in Spring 2023. A biopsy at this time showed borderline T cell mediated rejection with moderate interstitial fibrosis and tubular atrophy. There was a rapid increase in GSTT1 antibody levels with a peak of 11,000 MFI (&gt;95% percentile). We propose that immune-mediated inflammatory processes triggered by non-adherence episodes caused cellular damage. In response to such damage, intracellular components such as GSTT1 would be released, enabling recognition and the subsequent immune response leading to GSTT1 antibody production. We propose that the detection of antibodies to GSTT1 should be regarded a marker of nephrotoxicity and ongoing trauma to the transplanted kidney.</p><p><b><span>Adrian Handley</span></b>, <i>David Briggs, Clare Collins</i></p><p><i>NHS Blood and Transplant, Birmingham, UK</i></p><p>All measurements come with an element of uncertainty and are only truly useful when that degree of uncertainty is understood. The use of semi-quantitative data from Luminex HLA antibody testing is essential in enabling cardiothoracic transplant compatibility assessment; however, the level of uncertainty was not understood. BSHI/BTS CTAG guidelines set risk levels based on MFI values produced in Luminex Single Antigen Bead HLA antibody assays. Additionally, ISO15189:2012 states that laboratories should calculate measurement uncertainty (MU) for all accredited tests. In this study we used simple statistical tools to identify degrees of MU and some of the contributing factors. These allowed us to quickly assess the effectiveness of changes subsequently made. We identified significant difference between results obtained by individual testing personnel and were able to reduce this through our interventions. Reed et al. (<i>Am J Transplant</i>. 2013;13(7):1859–1870) suggested MU as %CV was in the region of 20%–62%. Our study found this to range from 8% at 20,000 MFI up to 18% at 1000 MFI, the threshold for positivity.</p><p>The tools we developed allow ongoing monitoring of assay performance and provide the basis for future quality initiatives, contributing to sustainable, evidence-based quality improvement. MU calculations are an effective quality assurance and improvement tool and are easily implemented. By combining the tools used in this study, levels and sources of variation can be identified, and the effect of any changes to process can be easily assessed for effectiveness.</p><p><b><span>Saima Azhar Salim</span></b>, <i>Louise Walsh, Geraldine Donnelly, David Keegan, Joseph Kelly, Mary Keogan</i></p><p><i>H&amp;I Lab, Beaumont Hospital, Dublin, Ireland</i></p><p>Flow cytometric crossmatch (FXM) is performed during pre-transplant histocompatibility workup. False positive results may prevent a potential recipient receiving a suitable transplant. Many labs use a three-colour FXM assay established using a dual-laser flow cytometer, which includes phycoerythrin (PE), fluorescein isothiocyanate (FITC) and peridinin-chlorophyll proteins (PerCP). There is significant spectral overlap between PE and FITC potentially leading to high background fluorescence, and possible false positive B cell FXM. This study was undertaken to determine optimum fluorochrome combinations to minimise spectral overlap and maximise consistency with the current method for which clinically validated cut-offs are available. Fluorochromes Brilliant Violet (BV421) and Allophycocyanin (APC) were selected for labelling CD3+T cells and CD19+B cells respectively. Thirty-four FXM (total 63 samples for T cell and 61 samples for B cell FXM) were performed with fluorochromes CD3BV421, CD19APC and anti-human IgG FITC. Fourteen FXM (total 35 samples for T and B cell FXM) were performed with fluorochromes CD3PerCP, CD19APC and anti-human IgG FITC. FXM assays were performed on Becton Dickenson (BD) FACSLyric™ and results compared to the standard FXM assay. In comparison to the standard FXM, a statistically significant difference in sample T cell ratio and B cell ratios (<i>p</i> &lt; 0.005) were observed for CD3BV421-CD19APC FXM. Our pilot data suggest no significant difference in T and B cell ratios for CD3PerCP-CD19APC (<i>p</i> &gt; 0.05). Data suggest that substitution of CD19PE with CD19APC minimises spectral overlap, reducing the risk of false positive FXM results, without significantly altering T and B cell ratios.</p><p><b><span>Ana Bultitude</span></b><sup>1</sup>, <i>Anthony Poles<sup>2</sup>, Sue Jordan<sup>1</sup>, Anthony Calvert<sup>2</sup>, Deborah Sage<sup>1</sup></i></p><p><i><sup>1</sup>NHS Blood and Transplant, Tooting</i>, <i>UK; <sup>2</sup>NHS Blood and Transplant, Filton, UK</i></p><p>HNA-3 is a bi-allelic antigen, -3a/-3b, with 95% of the UK population encoding at least one HNA-3a allele. HNA-3 is expressed on a variety of cell types including lymphocytes and renal endothelial cells. Accordingly, HNA-3 specific antibodies, which can develop against the non-self-variant in homozygous individuals, have been implicated in rejection episodes following renal transplantation. Here, we report a case of a renal transplant recipient displaying anti-HNA-3a antibodies that prevented transplantation. In August 2022, the patient was offered an altruistic kidney with a 2,1,1 mismatch grade, yielding a negative virtual crossmatch result. However, final wet laboratory flow cytometric crossmatch (FCXM) results presented an unexplained strong positive T and B cell result with a negative auto FCXM. The patient had no previous transplant history and no detectable HLA antibodies since their initial referral. HNA genotyping was performed on both patient and donor, determining HNA-3b3b and HNA-3a3a genotypes respectively. Patient sera was screened for anti-HNA antibodies, confirming the presence of HNA-3a-specific antibodies. The patient was removed from the virtual crossmatching programme as they no longer met the eligibility criteria but remained on the deceased donor waiting list. A further nine deceased donor kidneys were accepted and crossmatched, all of which were T and B cell positive. In March 2023, the patient received a deceased donor kidney which produced a T and B cell negative FCXM. The donor was retrospectively genotyped as HNA-3b3b. The patient continues to do well with no antibody-mediated rejection detected thus far.</p><p><b><span>Hawzhin Jabar</span></b>, <i>Luke Foster</i></p><p><i>NHS Blood and Transplant, Birmingham, UK</i></p><p>Accurate HLA typing is essential to facilitate safe solid organ transplantation. Within the UK, H&amp;I laboratories are required to meet the minimum typing requirements set by NHSBT-OTDT, which includes reporting results that allow for the assessment of any donor specific antibodies (DSA) in a particular donor-recipient combination. Failure to do so can lead to inappropriate allocation or an increased risk of transplant rejection. Here we report the identification of the DRB1*14:15 allele in a deceased solid organ donor who was HLA typed on-call using LinkSeq real-time PCR (One Lambda). Ordinarily, it would not be a requirement to report HLA-DRB1*14 to the second field under the minimum typing requirements, however, interestingly, HLA-DRB1*14:15 does not encode the DR14 antigen, but codes for the DR8 antigen. Therefore, in this instance there was a requirement to report HLA-DRB1*14 to the second field to allow accurate allocation and assessment of any potential DSA. Despite being locally rare, HLA-DRB1*14:15 is listed as well-documented in European populations within the CIWD 3.0.0 catalogue, and common in Asian/Pacific Islands and Native American populations. In this case, donation proceeded with both kidneys being transplanted in two patients, one locally, and out of region. Although the patient transplanted locally was cRF 0% and therefore had no DSA, elucidation of the DR8 antigen may be important for any future post-transplant DSA monitoring.</p><p><b><span>Richard Battle</span></b>, <i>Emma Ross, Sylvia McConnell, David Turner</i></p><p><i>H&amp;I Department, SNBTS, Edinburgh, UK</i></p><p>The SNBTS H&amp;I laboratory provides ∼2300 apheresis donor platelet units annually to HLA sensitised refractory patients across Scotland from a panel of ∼800 typed donors. Data is recorded for each transfusion, including match grade (A = matched for HLA-A and B antigens, B1 = one HLA antigen mismatch, B2 = two antigen mismatch etc.) and cumulative HLA antibody MFI (cMFI) against mismatches. Platelet transfusions were grouped by match and cMFI and assessed against post counts relative to pre-counts that is, a measure of transfusion increment. Pre- and post-platelet counts were available on 1486 cases between 2015–2022. Recipient and donor HLA-A and B types at first field defined A, B1–B4 matching. cMFI was calculated following One Lambda SAB I testing. Analyses used ANOVA or <i>t</i>-test between groups. In 1166 HLA compatible transfusions (cMFI &lt; 2000) no differences were seen in mean increment between A, B1, B2, B3 and B4 matches; 22.5, 22.9, 22.1, 24.1, 30.2 respectively (ANOVA <i>p</i> = 0.14). In 1486 patients grouped according to cMFI, cases with &lt; 2000 had a mean increment = 23.0, with &gt; 2000 &lt; 10,000 mean = 18.0 (<i>p</i> &lt; 0.001) and &gt; 10,000 mean = 8.0 (ANOVA <i>p</i> = 1.55 × 10–15). This analysis of platelet counts after HLA selected platelet provision shows that, as expected, the match grade of the platelets does not impact on the immediate increment. The level of HLA antibody, as defined by cMFI, affects the post transfusion count, especially when cMFI &gt; 10,000. This data will help locally in selection of optimal platelet units for patients.</p><p><b><span>Patrick Flynn</span></b><sup>1</sup>, <i>Sebastian Fernando<sup>2</sup>, Judith Worthington<sup>1</sup>, Kay Poulton<sup>1</sup></i></p><p><i><sup>1</sup>Transplantation Laboratory, Manchester Royal Infirmary, Manchester, UK; <sup>2</sup>School of Health Education and Public Health Sciences, University of Manchester, Manchester, UK</i></p><p>The aim of this study was to devise an algorithm that would predict Flow Cytometry crossmatch results using SAB Median Fluorescent Intensity (MFI) levels and to test this correlation using samples tested from a NEQAS Scheme 2B cohort.159 NEQAS 2B serum samples were screened using LABScreen™ SAB and 40 NEQAS 2B peripheral blood samples were HLA typed with LABType™ SSO. Donor-Specific Antibodies (DSA) were identified for each cell-serum combination tested and cumulative MFI values calculated for each test. HLA Class I MFIs were combined to predict the T cell crossmatch. For the B cell crossmatch prediction, two options were considered: (i) HLA Class II MFI values alone and (ii) HLA Class I + Class II MFIs. Receiver Operating Characteristic analysis was carried out to identify the combined MFI cut off that predicted NEQAS consensus results with the greatest sensitivity and specificity value. HLA Class I combined MFI &gt; 5000 predicted T cell crossmatch results with 96% sensitivity, 100% specificity, 100% Positive Predictive Value (PPV) and 92% Negative Predictive Value (NPV). For B cell results, HLA Class I + Class II combined MFIs &gt; 11,000 gave the best model showing 97% sensitivity, 79% specificity, 95% PPV and 85% NPV. However, for samples with only HLA Class II sensitisation, combined MFIs &gt; 13,000 improved the B cell crossmatch predictions: 92% Sensitivity, 91% specificity, 92% PPV and 91% NPV. Using this model, combined MFI values can be used to predict the immunological risk posed by DSA when it is not possible to carry out a crossmatch test.</p><p><b><span>Jonathan A.M. Lucas</span></b><sup>1</sup>, <i>Richard M. Szydlo<sup>1,2</sup>, Shelley Hewerdine<sup>1</sup>, Steven G.E. Marsh<sup>1,3</sup>, Neema P. Mayor<sup>1,3</sup></i></p><p><i><sup>1</sup>Anthony Nolan Research Institute, Royal Free Hospital, UK; <sup>2</sup>Department of Medicine, Imperial College, UK; <sup>3</sup>UCL Cancer Institute, Royal Free Hospital, UK</i></p><p>The effect of matching for HLA-E on the outcome of hematopoietic cell transplantation (HCT) has thus far been inconsistent and has not been studied in a UK cohort where there is an abundant use of alemtuzumab for T-cell depletion. We analysed HLA-E genotypes in 1513 UK HCT patients with a haematological malignancy and their unrelated donors using full-length PacBio Single Molecule Real-Time DNA sequencing. After adjusting for clinical factors that affected outcome prognoses including classical HLA matching out of 12, the presence of mismatches at both HLA-E loci (HLA-Emm; <i>n</i> = 30) was significantly associated with a reduced risk of relapse (HR 0.44; 95% CI 0.20–0.98, <i>p</i> = 0.04) in comparison to being HLA-E matched (HLA-Em; <i>n</i> = 788). A non-significant but beneficial effect on Overall Survival (OS) and Event-Free Survival (EFS) was also observed for two HLA-Emm loci compared to HLA-Em (OS: HR 0.73, <i>p</i> = 0.30; EFS: HR 0.77, <i>p</i> = 0.38). There were no significant differences in HCT outcomes correlated with a single HLA-Emm (<i>n</i> = 450). Assessing directionality of mismatches showed that a bi-directional HLA-Emm (<i>n</i> = 45) was significantly associated with reduced risks of relapse (HR 0.40; 95% CI 0.20–0.83, <i>p</i> = 0.01), increase in EFS (HR 0.55; 95% CI 0.32–0.93, <i>p</i> = 0.02) and a non-significant increase in OS (HR 0.72, <i>p</i> = 0.16) compared to HLA-Em. We hypothesise that mismatching alleles at the HLA-E locus results in sufficient genetic disparity to provide a stronger Graft-versus-Leukemia effect, without eliciting detrimental Graft-versus-Host responses, hence no significant differences in acute Graft-versus-Host Disease or Transplant Related Mortality, as observed in this study.</p><p><b><span>Sajadhossein</span>\n <span>Bazrafshani</span></b><sup>1</sup>, <i>Mohammadreza Bazrafshani<sup>2</sup></i></p><p><i><sup>1</sup>ENT Department, Worcester, UK; <sup>2</sup>Kerman University of Medical Sciences, Kerman, Iran</i></p><p>Cyclosporine, a calcineurin inhibitor, has a narrow therapeutic index and shows considerable inter-individual variability in pharmacokinetics. Cyclosporine is a P-glycoprotein (P-gp) substrate, a multidrug resistance gene (MDR1) product. Some of the single nucleotide polymorphisms (SNPs) of MDR-1 correlate with the variable activity of P-gp in vivo, and it is thought that these polymorphisms are associated with pharmacokinetic variations in cyclosporine therapy. Genotyping assays (PCR-SSP and PCR-RFLP) were performed for detection of frequency within two functional MDR-1 SNPs (C1236T in exon 12 and C3435T in exon 26) in 60 patients and the correlation between genotyping and concentration/dose ratio of cyclosporine was investigated. Data analysis revealed that C3435T polymorphism correlated with the concentration/dose ratio significantly. The concentration/dose ratios were 59.36 μg/L/kg/12 h (<i>p</i> &lt; 0.001) and 30 μg/L/kg/12 h (<i>p</i> = 0.049) lower in homozygous wild-type patients (CC) rather than homozygous mutant-type (TT) and heterozygous patients (CT) respectively. Moreover, heterozygous patients (CT) had 29.36 μg/L/kg/12 h (<i>p</i> = 0.001) which was less than homozygous mutant-type patients (TT). These findings suggest that, for the given dose, the blood concentration is lower in homozygous wild-type individuals (CC type in 3435 position). Our results revealed that MDR-1 genotype appears to influence cyclosporine drug levels and MDR-1 genotyping may provide a useful clinical guide in predicting the required dose after renal transplantation.</p><p><b><span>Mazen Mabrok</span></b><sup>1</sup>, <i>Renuka Palanicawander<sup>2</sup>, Betia Nouri<sup>1</sup>, Rachel Smith<sup>1</sup>, Arthi Anand<sup>1</sup>, Natalia Brodaczewska<sup>3</sup></i></p><p><i><sup>1</sup>Histocompatibility &amp; Immunogenetics Laboratory, North West London Pathology, Imperial College Healthcare NHS Trust, London, UK; <sup>2</sup>Centre for Haematology, Imperial College Healthcare NHS trust, London, UK; <sup>3</sup>Specialist Integrated Haematological Malignancy Diagnostic service (SIHMDS), Imperial College Healthcare NHS Trust, London, UK</i></p><p>Monitoring of donor chimerism after haematopoietic stem cells transplantation (HSCT) is vital for early effective therapeutic interventions. Short tandem repeat (STR) assays are the current gold standard for chimerism monitoring after allogeneic HSCT. The emergence of Next Generation Sequencing (NGS) CE-IVD approved assays with improved limit of detection of around 0.05% offer a promising alternative. In this evaluation exercise, we explored the technical capacity of NGS in post-transplant chimerism monitoring, by retrospectively analysing samples from 13 patients from our centre using the Dvysr® NGS chimerism assay (Sweden, Stockholm). All patients enrolled in this evaluation were tested at three time points (whole blood &amp; T- Cell), selected in collaboration with the clinical team based on clinical signs of relapse, increase in MRD markers and the profile obtained using the PowerPlex 16 Multiplex STR system (Promega). Our results showed very strong correlation between the NGS and the STR assays (Pearson score 0.998 &amp; 0.999) with a shift of 1%–1.5% at the pre-relapse time point observed in 55% of the patients enrolled in this evaluation. The NGS assay needed 50% less time to analyse and in 70% of the patients tested, the NGS assay yielded more informative markers than the STR assay. NGS chimerism assay promises improved diagnostic performance and usability in our evaluation. Increased sample size and inclusion of lineage specific cell separation will further strengthen validity of the improved diagnostic performance and usability of NGS chimerism assay as alternative to STR assays including correlation with clinical presentation.</p><p><b><span>Emma Holmes</span></b><sup>1</sup>, <i>Jasmaine Lee<sup>2</sup>, Winnie Chong<sup>2</sup>, Deborah Sage<sup>1</sup>, Martin Howell<sup>2</sup></i></p><p><i><sup>1</sup>NHSBT Tooting Centre, Tooting, UK; <sup>2</sup>NHSBT Service Development, Colindale, UK</i></p><p>Nanopore sequencing presents a new technology for high resolution HLA typing that is, considered faster and potentially cheaper than existing methods used for next generation sequencing (NGS). NanoTYPE (Omixon) can be used for batches of up to 24 samples or for single samples, which has the potential to be used for deceased donor HLA typing. The aim of this study was to evaluate the NanoTYPE assay in two NHSBT H&amp;I laboratories; Colindale Service Development and Tooting, using the same batch of 96 samples that had previously been typed by current rapid HLA typing or NGS methods. All NanoTYPE reagents, R9.4 flow cells and a MinION mk1B device were provided by Omixon and Oxford Nanopore Technologies for this study. Eight samples were set-up using the single-sample protocol. The remaining samples were set-up in multi-batches of varying sizes. There was 100% concordance at second field resolution between both sites, and with previous typing data, for HLA-A, -B, -C, DRB4, DRB5 and DPA1. Non-concordance with previous typing data was seen for either HLA-DRB1, DRB3, DQA1, DQB1 or DPB1 loci in 10/96 (10%) samples at Tooting and 9/96 (9%) samples at Colindale, which was attributed to allele imbalance, known low amplification of certain genotypes such as DQB1*03, allele dropout and detection of potential novel alleles. After manual review of the sequencing data, 95/96 samples at Colindale and 93/96 samples at Tooting were concordant with previous results. Our study has proven that the NanoTYPE assay is simple to use and enables high resolution HLA typing.</p><p><b><span>Emma Ross</span></b><sup>1</sup>, <i>Sylvia McConnell<sup>1</sup>, Richard Battle<sup>1</sup>, Nicole Priddee<sup>2</sup>, David Turner<sup>1</sup></i></p><p><i><sup>1</sup>H&amp;I Department, SNBTS, Edinburgh, UK; <sup>2</sup>Donor Medicine, SNBTS, Edinburgh, UK</i></p><p>The SNBTS H&amp;I laboratory is responsible for the provision of HLA selected platelets in Scotland. In 2022, 96 patients were supported with a total of 2317 platelet units. A key performance indicator (KPI) for the service is &gt;60% of allocated donor platelets should be an A grade or B1 grade match with the patient (A grade = no HLA-A or HLA-B mismatches at first field; B1 = only one HLA-A or HLA-B mismatch). This target has averaged 57.7% (range 46%−72%) between January 2016 and September 2022, with an overall downward trend observed. In contrast, the number of individual donor platelets issued as HLA selected increased from 1288 units in 2016 to 2317 in 2022 (+55.5%). To make the best use of a limited donor panel, H&amp;I have worked with SNBTS Donor Services (DS) to increase availability of best matched donor platelets. Utilising a Quality Improvement approach, business analytic tools and specialist donor communications, we concentrated on the national identification and (re)engagement of HLA-A and -B homozygous donors which constitute 3.5% of the panel but have the potential to provide A grade matches for ∼68% of patients (Jan–June 2023). Since the project initiation we have recorded an upward trend in the percentage of A and B1 platelet allocations over the period October 2022 (49.5%)–May 2023 (72.4%). The target of &gt;60% has been achieved consistently from Jan 2023. Collaborative working with DS has enhanced the donor panel, improving ‘off the shelf’ access to best matched HLA selected platelets.</p><p><b><span>Felicity May</span></b><sup>1</sup>, <i>Sian Griffin<sup>2</sup>, Madhvi Menon<sup>3</sup>, Tracy Hussell<sup>3</sup>, Tracey Rees<sup>1</sup></i></p><p><i><sup>1</sup>Welsh Transplantation and Immunogenetics Laboratory, Welsh Blood Service, Pontyclun, UK; <sup>2</sup>Department of Nephrology and Transplantation, University Hospital of Wales, Cardiff, UK; <sup>3</sup>Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK</i></p><p>Human leukocyte antigen incompatible (HLAi) transplantation remains an important option for very highly sensitised patients. Crossing the HLA barrier is associated with increased risk of antibody-mediated rejection (AMR) and graft failure. Desensitisation, induction and maintenance immunosuppression aim to minimise this risk. There is poor consensus on optimal treatment protocols, and patient response varies. In this single centre, retrospective study, we assessed incidence of rejection and graft survival in a clinical cohort of 27 HLAi transplant recipients desensitised with rituximab and cycles of double filtration plasmapheresis. There was poor association between established risk factors and incidence of AMR, suggesting the involvement of other, currently unknown, factors. HLA antibody response and AMR are influenced by a complex cytokine network supporting the generation and survival of antibody producing cells. We conducted a literature review and graded 459 serum protein analytes (cytokines, chemokines, hormones etc.) based on the quality of evidence and proximity of involvement in humoral memory response. We evaluated serum concentrations of the 40 highest graded analytes in longitudinal samples derived from our clinical cohort using a bespoke multiplexed Luminex assay. Due to low samples numbers and heterogeneity of the analyte profiles, we were unable to establish clear association with patient clinical outcomes. However, we observed significant and sustained changes to key signalling molecules known to influence germinal centres, B-cells and plasma cells. We noted a reduction in levels post-desensitisation/immunosuppression for the majority of our panel, but increased or unchanged levels of several key supportive signals and suppression of several immune regulators.</p><p><i>Agnieszka Ojrzynska<sup>1</sup>, Kylara Hassall<sup>1</sup>, Katie Butler<sup>1</sup>, Graham Shirling<sup>1</sup>, Sharon Vivers<sup>2</sup>, Raymond Fernando<sup>1</sup></i></p><p><i><sup>1</sup>Solid Organ Group, Royal Free Hospital, London, UK; <sup>2</sup>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>Increasing numbers of multiple myeloma patients, treated with Daratumumab (Dara), are being listed for kidney transplantation. Dara is a human monoclonal anti-CD38 IgG antibody that interferes with both pre-transplant allogeneic lymphocyte crossmatches and pre-transfusion compatibility testing, as it binds to CD38 molecules expressed on many blood cells including lymphocytes. This can simulate the presence of donor-specific antibodies (DSA) and lead to false positive crossmatch results. The aim of this study was to investigate methods to mitigate Dara interference with flow cytometry crossmatches (FCXM). Our centre has transplanted four patients undergoing Dara treatment. Three of these patients were HLA antibody negative and one patient was HLA antibody positive but HLA DSA negative. Six third party peripheral blood FCXMs, and one retrospective transplant FCXM with spleen cells, were performed using our standard procedure, as well as an amended protocol using donor cells pre-incubated with dithiothreitol (DTT) (0.05–0.1 M) which can cleave the stabilising disulphide bonds of the CD38 molecule. All third-party crossmatches using our standard procedure were T cell positive, two were also B cell positive. The retrospective crossmatch performed with spleen cells was T and B cell positive. All crossmatches performed with DTT were T and B cell negative. Our results show that DTT eliminates the false positivity observed when sera from Dara treated patients are used in FCXM. HLA positive control results were unaffected suggesting that HLA molecules are not affected by DTT treatment. Therefore, DTT can be used to mitigate DARA mediated false positive results in FCXM.</p><p><b><span>Daniel Eggleston</span></b>, <i>Helena Lee</i></p><p><i>Manchester Royal Infirmary, Manchester, UK</i></p><p>Haematopoietic progenitor cell transplantation (HPCT) is well-established as a curative treatment for malignant and non-malignant haematological disorders. However, patient monitoring is still essential following transplantation. Post-transplant donor chimerism testing can identify complications including relapse or graft failure. Relapse is a particular area of concern for patients diagnosed with Myelodysplastic syndromes (MDS) or high-risk leukaemias who have undergone HPCT. Most laboratories perform chimerism monitoring by PCR-Short Tandem Repeats (PCR-STR). This technique uses differences in short repeated sequences of DNA and capillary electrophoresis to determine percentage donor chimerism. However, other methods are emerging that offer improved sensitivity and more informative markers. One such technique is digital PCR (dPCR). During dPCR, a PCR reaction mixture is partitioned into individual droplets. Each function as an individual assay, with the presence of a reaction indicating donor or recipient DNA, increasing technique sensitivity. Using dPCR, we investigated eleven patients diagnosed with MDS or Myeloid Leukaemias whose percentage donor chimerism reached 100% by PCR-STR but dropped in subsequent samples. We tested our cohort using JETA DigitalTRACE™ dPCR technology with the Qiagen QIAcuity machine at two or more timepoints preceding this reduction in donor chimerism using three markers. In 10 of our 11 patients, a reduction in donor chimerism was identifiable by dPCR whereas analysis by PCR-STR had suggested samples were 100% donor chimerism. This raises the possibility of an earlier detection of changes in donor chimerism and an improved follow up process. Chimerism by dPCR is therefore a viable replacement for PCR-STR and may identify events earlier post-HPCT.</p>","PeriodicalId":14003,"journal":{"name":"International Journal of Immunogenetics","volume":null,"pages":null},"PeriodicalIF":2.3000,"publicationDate":"2023-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/iji.12630","citationCount":"0","resultStr":"{\"title\":\"Oral Abstracts\",\"authors\":\"\",\"doi\":\"10.1111/iji.12630\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><b><span>Alison Cleaton</span></b>, <i>Emma Burrows, Kimberley Robinson, Michael Richardson, Deborah Pritchard, Tracey Rees</i></p><p><i>Welsh Blood Service, Ely Valley Road, Talbot Green, UK</i></p><p>Regular HLA antibody testing is undertaken for patients awaiting renal transplantation, using LABScreen™ HLA antibody assays. During the COVID-19 pandemic, we observed unexplained changes to some HLA antibody profiles. Investigation revealed that several patients had COVID-19 prior to the changes, therefore, a review of all patients on the transplant waiting list with known COVID-19 infection was undertaken. Sixty-six out of two hundred thirty-seven patients on the transplant waiting list had COVID-19 (March 2020–July 2022). The HLA antibody results from samples prior to and following COVID-19 infection were analysed for changes in existing HLA antibody levels (increased Luminex Median Fluorescent Intensity (MFI) values), or expanded antibody profiles (increased cRF). Fifty-two (78.8%) patients had no detectable change in cRF or MFI; five (7.6%) had changes in MFI (but no change in cRF); nine (13.6%) had changes in MFI and cRF. Two out of nine patients had no recorded prior sensitisation event; four had a previous transplant, four blood transfusions, four pregnancy; and three multiple sensitising events. All nine patients had sustained cRF changes in subsequent samples (follow up to December 2022). Three out of nine patients were consequently identified as having altered immunosuppression due to the COVID-19 infection; these patients had a 20%–76% rise in cRF and now all have a cRF 98%–100%. While the majority of patients awaiting kidney, transplantation had no change to their HLA antibody profile following COVID-19 infection, nine patients had an increase in cRF, which has not been transient. Reduction or withdrawal of immunosuppression to aid recovery from COVID-19 was identified as the cause for three patients.</p><p><b><span>Adrienne Seitz</span></b>, <i>Clive Carter, Brendan Clark, Richard Baker</i></p><p><i>Leeds Teaching Hospitals NHS Trust, Leeds, UK</i></p><p>The level of pre-transplant immune risk is assessed through measuring serum IgG HLA antibodies which can be produced by long lived plasma cells and memory B-cells. Memory B-cells can circulate without producing antibodies, therefore their contribution to the antibody pool may not be fully appreciated. We describe an in vitro method for improving the assessment of pretransplant risk through the non-specific stimulation of peripheral memory B-cells. Peripheral blood mononuclear cells from three unsensitised volunteers and six sensitised patients were cultured for 9 days with the toll-like receptor agonist R848 and interleukin-2. Cell culture supernatant was tested for IgG HLA antibodies using single antigen beads. This was compared with a matched serum sample. Resting Day-0 and stimulated Day-9 B-cell phenotypes were assessed using flow cytometry, confirming the switch to antibody secreting (CD24-CD38hi), class-switched memory (CD27+IgD-) and plasma (CD38+CD138+) cells. HLA Class I and II antibodies were found in the cell supernatant, and 65% were present in the matched serum sample. When the supernatant demonstrated additional HLA antibodies, these could be attributed to a previous transplant, or had been present in the patient's historic serum profile. We demonstrate a method that can uncover peripheral memory using technology accessible to most H&amp;I laboratories. This assay could be useful when assessing live donor pairs where the donor may repeat mismatches associated with pregnancy, and in re-grafts, prior to removal of ‘other unacceptable antigens’. Finally, it could be considered alongside delisting strategies in the context of novel peri-transplant agents.</p><p><b><span>Sophie Chambers</span></b>, <i>Robert Whittle, John Goodwin, Tim Key</i></p><p><i>NHS Blood and Transplant, Barnsley, UK</i></p><p>Differences in amino acids (aa) at positions 76–83 of exon II of HLA-B and a subset of HLA-A primarily account for the highly immunogenic public epitopes Bw4 and Bw6. Bw4-specific antibodies are commonplace in alloimmunised Bw6 homozygotes, whilst Bw6-specific antibodies are encountered in Bw4 homozygotes. As variation exists within the Bw4 complex it has been reported previously that in a proportion of Bw6 homozygous individuals with Bw4 specific antibodies, reactivity can be restricted to HLA Bw4 subtype epitopes. We report three individuals with a Bw4 phenotype who demonstrate Luminex Single Antigen Bead determined alloantibodies to Bw4 epitopes distinct from their own. For renal patient L bearing B13, reactivity was present to all Bw4-positive beads other than B13. Reactivity was consistent with Epitope 249 defined by aa at positions 82L + 145R/ 83R + 145R. Platelet refractory patient G bearing A*24:02 demonstrated reactivity to all Bw4 beads other than A24, consistent with Epitope 423 defined by 144Q. For allogeneic-HSCT patient F bearing B*27:05, reactivity was present to all Bw4 beads except B*27:05 with reactivity defined by aa 77N + 81A + 82L. Our observations highlight that Bw4 subtype epitope profiles should not be overlooked in Bw4-positive individuals, that single amino acid differences in the Bw4 complex appear sufficient to generate alloantibodies and differences outside aa positions 76–83 may contribute to antibody binding. Bw4 subtype antibodies can impact organ allocation in the renal transplant setting, restrict appropriate platelet support for immunological refractoriness and complicate donor selection in HLA-mismatched allogeneic-HSCT.</p><p><b><span>Ryan Stevens</span></b>, <i>Felicity May</i></p><p><i>Welsh Transplantation and Immunogenetics Laboratory, Pontyclun, UK</i></p><p>The Welsh Transplantation and Immunogenetics Laboratory maintain a register of local patients active on the national renal/pancreas waiting list. Previously, data pulled from local IT systems was distributed monthly to service users in PDF format and a printed copy was held locally. This system had multiple drawbacks, including lack of ability to easily interrogate/update the data. In collaboration with the NHS Wales Microsoft 365 Centre of Excellence, a digital solution was created using Power BI. This allows up-to-date data to be instantly available, which gives users a more accurate overview to support decision making, and eliminates the requirement for PDF/paper distribution. The Dashboard can only be accessed by approved users, and access can be restricted to specific datasets (e.g., dialysis unit staff can only view patients at that unit). The Dashboard consists of two pages. A ‘Statistics’ page graphically displays the Register by organ type, blood group, dialysis unit, and so forth. The ‘demographics’ page tabulates data within the Register, which can be ordered or filtered in a variety of ways to aid in patient selection for donor offers. Each patient also has a colour coded ‘Sample Status’ to highlight when routine antibody screening samples are due. Comments and/or attachments can be added to each patient entry. Since successful go live December 2022, the dashboard has been praised by staff and service users for ease of access of data to assist delivery of safe and efficient patient care, as well as reduced incidence of overdue samples.</p><p><b><span>Kirsty Clark</span></b>, <i>Jane Matthews, Claire Romaines, Ruth Chisman, Arash Akbarzad-Yousefi</i></p><p><i>NHSBT Newcastle, Newcastle upon Tyne, UK</i></p><p>Despite advancements in organ allocation and immunosuppression, cardiothoracic transplantation continues to have the lowest 5-year survival when compared to all other forms of solid organ transplantation. A key factor in allograft loss is the formation of <i>de novo</i> Donor Specific HLA Antibodies (dnDSA). Recently, research has been driven away from the conventional method of defining mismatch at the antigenic level and has instead focused on HLA eplet mismatches. This study aimed to evaluate the role of eplet mismatching within our local cardiothoracic patient cohort. A retrospective analysis of 2 years of cardiothoracic transplant data was performed. The antibody data from all eligible patients were reanalysed to investigate the role of eplet mismatch load and to identify possible high risk eplets. This study found no evidence to directly support the theory of eplet load mismatches. However, four previously identified high risk eplets (McCaughan et al., <i>American Journal of Transplantation</i> 2018) namely, 55PP, 52LR, 55R and 75S were confirmed within our patient cohort. Findings from this single centre study provide potential evidence of high-risk HLA eplet mismatches; all of which are present within the HLA-DQ locus, indicating possible high immunogenicity for mismatches at this locus. Particular attention to minimise HLA-DQ mismatches may reduce the incidence of dnDSA and subsequently, allograft loss. Whilst avoiding mismatches in cardiothoracic transplantation is not always possible, further understanding of this area could lead to refinements in post-transplant monitoring and immunosuppression regimens.</p><p><b><span>Charlotte A. Cambridge</span></b><sup>1</sup>, <i>Jonathan A.M. Lucas<sup>1</sup>, Xenia Georgiou<sup>1</sup>, Gabriel J. Benitez<sup>1</sup>, Neema P. Mayor<sup>1,2</sup>, Steven G.E. Marsh<sup>1,2</sup></i></p><p><i><sup>1</sup>Anthony Nolan Research Institute, London, UK; <sup>2</sup>UCL Cancer Institute, Royal Free Hospital, UK</i></p><p>Submission of novel sequences to the IPD-IMGT/HLA Database from patients with haematological malignancies is not permitted, unless confirmed in the germline. To investigate if sequencing blood and buccal DNA yields different results, we typed 47 patients in remission from malignant disease and 58 healthy donors for HLA-A, -B, -C and -E using PacBio SMRT sequencing. Higher Phred scores (33.2 vs. 32.7, <i>p</i> &lt; 0.05) and lower cluster diversity (0.06 vs. 0.11, <i>p</i> &lt; 0.0001) were observed in sequences from patient buccal DNA versus blood, indicating sequences of higher quality with fewer background errors. No differences were observed in donor blood or buccal material. Blood DNA generated better HLA typing results that were automatically accepted (80.7% vs. 79.8%) with less allele dropout (2.0% vs. 7.2%). HLA typing results were concordant between blood and buccal derived DNA, including four novel sequences observed in patient samples, confirming these as germline mutations. Next, we analysed sequences from blood DNA for patients in remission from malignant (<i>n</i> = 406) versus non-malignant (<i>n</i> = 46) disease across nine libraries containing ≥3 of HLA-A, -B, -C, -E, -F and -G. No significant differences in Phred score (33.60 vs. 33.56) or cluster diversity (0.05 vs. 0.06) were observed. Overall, there were no differences in final HLA typing results for blood and buccal DNA samples for the same individual, and no difference in sequence quality between malignant and non-malignant patient samples. All sample types, if taken at the point of remission, are reliable sources for HLA typing and identification of novel sequence variation.</p><p><b><span>Ravneet Kaur Bola</span></b>, <i>Madalina Pinzaru, Marlowe Macadangdang, Ufot Udoffia, Sandra Frater, Franco Tavarozzi</i></p><p><i>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>Buccal epithelial cells are used for germline HLA testing, particularly useful in patients where loss of heterozygosity (LOH) is suspected. Anthony Nolan's (AN) patient buccal collection method utilises CytoSoft® brushes, however, we are always looking for ways to optimise our processes and realise that the design of the brushes may lead to an increased chance of blood contamination with some patients, potentially confounding HLA typing in LOH cases.</p><p>AN uses a gentler collection tool for our registry donor recruitment, FLOQSwabs®, therefore a trial was devised to identify the best buccal collection method for our patients, collaborating with the Royal Marsden Hospital. Each consented patient was swabbed using three scenarios: (1) FLOQSwabs®, cheek; (2) FLOQSwabs®, left gutter; (3) CytoSoft® brushes, right gutter. Samples from 54 patients were processed. DNA was extracted from each swab and one extraction per patient was selected for Next Generation Sequencing (NGS) in a pre-determined sequence, producing equal amounts of NGS data per scenario. Assessment looked at DNA extraction and NGS failure rates, together with DNA quantity produced.</p><p>DNA extraction failure rates were 9.3%, 17.6%, 29.9% for the three scenarios respectively.</p><p>On average, the DNA concentration from FLOQSwabs® were at least two times higher than with CytoSoft® brushes. Due to NGS robustness, all extractions selected for NGS were successfully typed. In summary, FLOQSwabs® swabbed from the cheek are the preferred collection method. These produced the lowest number of failed DNA extractions, while providing a softer and gentler swabbing experience for the patient.</p><p><b><span>Claire Lenehan</span></b><i>, James Kelleher, David Keegan, Mary Keogan, Khairin Khalib</i></p><p><i>NHISSOT Beaumont Hospital, Dublin, Ireland</i></p><p>Antibodies to human leukocyte antigens (HLA) are a complication for transplantation. The introduction of Luminex® technology has allowed for precise characterization of these antibodies with high sensitivity. The clinical significance of these antibodies remains controversial due to the detection of biologically irrelevant antibodies directed against denatured HLA molecules (dHLA). We aimed to determine the correlation of donor specific antibodies (DSA) detected by Luminex single antigen bead assay with flow cross match (FXCM) results, using a previously published protocol, with clinically validated cut-offs.</p><p>The capability of HLA antibodies in 170 non-classically sensitised patients to result in a positive crossmatch was investigated. Forty-nine FCXM against incompatible donor cells were completed. Fifty-six percent of T cell and 61% of B cell FCXM were positive. The rate of positive FCXM results was significantly higher for HLA-A and -B antibodies (<i>p</i> = 0.013). MFI value was a poor predictor of FCXM results. Conversely, the rate of negative FCXM results for DSA with MFI values of 5000 to 10,000 was not significantly different to that of MFI values &lt;5000. The rate of positive FCXM was higher for patients with DSA MFI &gt; 10,000. A significant proportion of the FCXM results were positive regardless of MFI strength indicating that DSA in non-classically sensitised patients are not limited to reactivity to dHLA. FXCM facilitates risk assessment. Based on the data in this study, we now offer a FCXM in living donors, or highly sensitised patients when DSA MFI is less than 10,000.</p><p><b><span>Kay Poulton</span>\\n <span>,</span></b><sup>1,2</sup> <i>Madeleine Harris<sup>1</sup>, Andrew Canterbury<sup>2</sup>, Marie Hampson<sup>1</sup>, Judith Worthington<sup>1</sup>, Marcus Russell-Lowe<sup>1</sup></i></p><p><i><sup>1</sup>Manchester Royal Infirmary, Oxford Road, UK, <sup>2</sup>MC Diagnostics Limited, St Asaph, Wales, <sup>3</sup>University of Manchester, Manchester, UK</i></p><p>Single Antigen Bead assays have revolutionised the identification and definition of HLA-specific antibodies. They have enabled the widespread use of virtual crossmatching and almost eliminated hyperacute rejection due to HLA incompatibility during the 20 years since their introduction. But for highly sensitised patients, there is a low likelihood of finding a compatible donor. The only option for some patients is strategic de-listing of specificities which may impact the lowest immunological risk to a new graft. In this study, 257 serum samples from 82 potential recipients were crossmatched against cells from 84 potential donors. Only results where serum samples have been tested by LABScreen, HISTO SPOT® HLA AB, Complement Dependent Cytotoxicity (CDC) and Flow Cytometry were included in the analysis. The results were analysed to assess the ability of HISTO SPOT® HLA AB to predict the crossmatch result. Of the 136 samples analysed, 17 (12.5%) were CDC positive, and 82 (60.3%) were positive by flow cytometry. Twenty-eight sera tested negative using HISTO SPOT. All of these (100%) were also negative by CDC and 25 (89.3%) were also negative by Flow Cytometry. Three sera which tested negative by HISTO SPOT with a positive flow cytometry crossmatch were CDC negative, each had cumulative MFIs of &lt;3000 against Donor Specific Antigens. In this early study, HISTO SPOT® HLA AB has 100% negative predictive value for CDC crossmatch negativity and 89.3% by Flow Cytometry. It may therefore prove a useful additional tool to inform de-listing strategies used to facilitate transplantation in highly sensitised patients.</p><p><b><span>David Wimbury</span></b></p><p><i>Transplant Laboratory, University Hospitals of Leicester NHS Trust, Leicester, UK</i></p><p>Previous work has shown that variations in temperature, incubation time and operator methodology can have a drastic effect on results in solid phase assays. In HLA antibody screening, differences in overall MFI levels could impact the number of antibodies detected as positive and/or their risk level, thereby potentially affecting transplantation opportunities. Normalisation of MFI in each sample to a baseline could create more consistent results between samples at different time points, reducing the measurement uncertainty. This method normalises overall sample MFIs to a baseline. Positive control samples and three patients with extensive sample histories were tested with this methodology. CVs of MFIs before and after normalisation were analysed with paired <i>t</i>-tests.</p><p>Overall, MFI data became more comparable between samples post-normalisation. MFI ranges on a bead-by-bead basis initially varied between runs from around 500 to as much as 20,000. This was decreased after normalisation; the average MFI range between runs decreased by 1337 for Class I and by 2402 for Class II. All samples showed a statistically significant decrease in coefficient variation (CV) of MFIs between runs after normalisation, both Class I and Class II (<i>p</i> &lt; 0.0001). Inconsistencies in HLA antibody screening results between samples from the same patient can make for troublesome clinical interpretations, especially when there is no sensitising event accounting for reactivity changes. The procedure shown here provides a novel method for reducing the variability caused by differences in assay conditions and has the potential to give more consistent results therefore providing a clearer clinical picture.</p><p><b><span>Deborah Pritchard</span></b></p><p><i>Welsh Transplantation Laboratory, Pontyclun, UK</i></p><p>Immunological compatibility testing for potential live kidney donors and patients requires multiple tests on patient and donor samples (HLA typing, HLA antibody testing, crossmatch assessment). Only on completion of all tests can a report be produced. The local KPI is 90% of cases reported in a turnaround time (TAT) of 10 working days, but performance since Q1 2021/22 has been &lt;75%. This quality improvement project (May–August 2022) identified delays from process maps and selected two areas as targets for improvement; antibody testing and report production. Two plan-do-study-act (PDSA) cycles were performed testing six change ideas generated from staff workshops. Changes included identification of case samples within the testing process to allow more efficient workflow; a dedicated individual with responsibility for reporting rather than shared responsibility; E-mail notification when tests have been completed; coordination of HLA antibody batch testing with sample arrival to reduce wait time, and introduction of visualisation boards to track cases more efficiently. One ‘just do it’ change was also introduced; an interface to import antibody results from HLA Fusion software to the laboratory information management system (LIMS). The mean TAT was reduced from 11.9 to 8.8 days. Improvements were seen in the antibody testing mean TAT (9.1 days→6 days) and report production TAT (3.6 days→2.2 days). This resulted in compliance with the KPI: 95% of cases were reported in 10 days (October–December 2022). The improvement was due to removing manual data entry processes, reducing wait times between procedures and eliminating duplication of work.</p><p><b><span>Evelien Little</span></b>, <i>Nicola Brosnan, Jade Kally, Adrian Silk, Lisa Walsh, Franco Tavarozzi</i></p><p><i>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>With the onset of Next Generation Sequencing (NGS), HLA typing has become more streamlined with the ability to use either singleplex or multiplex primer strategies. The Anthony Nolan Histocompatibility Laboratory utilises GenDx NGSgo® singleplex primers for NGS to achieve high-allelic resolution typing for HLA-A, -B, -C, -DRB1, -DRB3,4,5, -DQB1, -DQA1, -DPA1, and -DPB1. Prior to this work, all samples received for HLA typing were tested for 11 loci, but the number of loci requested may vary. The use of singleplex, as opposed to multiplex, primers has allowed us to implement a flexible approach to provide a tailored and bespoke service. The aim of this work was to develop a process to type samples that need various combinations of testing on the same run. Modifications were needed to our Laboratory Information Management System (LIMS) to allow significant changes throughout the NGS testing process. As a result of this project and as our ongoing flexibility improvement process, we also implemented the ability to repeat PCR failures within the same run, allowing the process to proceed without delay, and merge multiple NGS libraries.</p><p>Since we no longer perform unnecessary testing, this generated cost savings by increased capacity on our Illumina® flow cells, allowing more samples per run, potentially reducing our turnaround times, as well as meeting our customer needs. To conclude, singleplex primers have proved extremely useful in allowing flexibility with no discernible increase in run time, using LIMS to simplify and track the process, end to end.</p><p><b><span>Victoria Wood</span></b><sup>1,2</sup>, <i>Brendan Clark<sup>1</sup>, Eric Hewitt<sup>2</sup>, Sunil Daga<sup>1</sup></i></p><p><i><sup>1</sup>Leeds Teaching Hospitals NHS Trust, Leeds, UK; <sup>2</sup>University of Leeds, Leeds, UK</i></p><p>A core aspect of a renal Transplant Immunology service is the detection of HLA antibodies in the context of prospective donors. The presence of HLA-antibodies is detrimental to chances of transplant and, as donor-specific antibodies (DSA), to graft outcomes. With growing numbers of highly sensitised patients requiring renal transplant across HLA-antibody barriers, an increased understanding of antibody functional characteristics could lead to more informed donor choices. The avidity of an antibody-antigen interaction provides insight into the antibody's capability to induce antibody-mediated changes, such as intracellular signalling, leading to tissue remodelling and graft damage. Chaotropic agents reduce protein stability and have been previously used within ELISA protocols to estimate avidity of antibody-antigen interactions through a process called chaotropic disruption.</p><p>Modification of our standard One Lambda LABScreen Single Antigen bead (SAB) protocol to include a chaotropic agent has demonstrated technical viability of applying chaotropic disruption to a solid-phase assay and that this manipulation can be confidently interpreted. A maximum molarity of chaotropic agent was established which is not detrimental to bead surface antigen integrity - 1 molar. This was verified using flow cytometric analysis with HCA2 monoclonal, which binds to both native and denatured HLA-Class I, in comparison with HLA-A, -B, -C monoclonal which binds to native HLA-Class I only. Initial testing using sera with known HLA antibody profiles showed antibody-specific patterns of binding and disassociation. This novel method represents a potential accessible method of testing HLA antibody avidity in an NHS laboratory setting.</p><p><b><span>Mr Steven Jervis</span></b><sup>1,2</sup>, <i>Dr Antony Payton<sup>3</sup>, Dr Marcus Lowe<sup>1,2</sup>, Dr Altug Didikoglu<sup>4</sup>, Professor Arpana Verma<sup>2</sup>, Professor Kay Poulton<sup>1,2</sup></i></p><p><i><sup>1</sup>Manchester Transplantation Laboratory, Oxford Road, United Kingdom</i>; \\n<i><sup>2</sup>Faculty of Biology, Medicine and Health, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, University of Manchester, Manchester, United Kingdom; <sup>3</sup>Division of Informatics, Imaging &amp; Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; <sup>4</sup>The Centre for Biological Timing, Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, United Kingdom</i></p><p>Studies have hypothesised that the combination of pre-existing genetic traits and specific environmental triggers determine the onset of narcolepsy. The most impactful genetic risk factor is the presence of Human Leukocyte Antigen (HLA) DQB1*06:02 encoded on the Major Histocompatibility Complex (MHC), however, the presence of HLA-DQB1*06:02 is not ubiquitous in all narcolepsy cases. The most poignant genetic risk factors outside the MHC are predominantly located in genes associated with the immune system. In addition to the traditional symptoms of narcolepsy, the co-morbidities can vary with a cohort of sufferers complaining of cognitive dysfunction, particularly memory and attention. These self-reports are not substantiated by consistent scientific evidence whereas there is significant evidence outlining the genetic contribution underpinning variation in cognitive abilities in the general population. In this study we impute targeted non-MHC narcolepsy associated single nucleotide polymorphisms (SNPs) from 1,558 non-pathological elderly volunteers who have been followed for change in cognitive function for up to a 24-year period. Specifically, we investigate 13 previously documented narcolepsy associated SNPs with a odds ratio greater than or equal to 1.00 combined with a minor allele frequency of greater than 0.05. We observed an association between rs306336, rs4290173 and rs2834168 and a faster decline in long term memory. Similarly, we observed a protective effect of rs10995245 against the decline of long-term memory loss. This investigation suggests that the cognitive problems reported by cohorts of narcoleptic patients may be due to genetic predispositions and supports the variation seen in the co-morbidities associated with narcolepsy.</p><p><b><span>Raji Patel</span></b>, <i>Aliyye Karasu, Liezelle Pagala, Jyoti Bhatt, Edgar Correa, Gloria Adeyemo, Maame-Esi Yeboah, Abigail Sarkodie, Andrew Joahill, Suzette Cavanna, Carla Rosser, Colin Brown</i></p><p><i>NHS Blood and Transplant-Colindale, London, UK</i></p><p>The British Bone Marrow Registry (BBMR) utilises Oragene® saliva collection kits to facilitate extended HLA typing of potential donors by Next Generation Sequencing (NGS). Saliva is a good alternative source of DNA as its collection is non-invasive, allows for self-collection and is stable at ambient temperature. In January 2023, the Qiagen EZ2 robot was introduced into our laboratory to provide DNA extraction for clinical samples. However, Qiagen were unable to provide a validated protocol for the extraction of DNA from saliva samples using this instrument. In this study, we assessed the capability of Qiagen EZ2 tissue kits to obtain DNA from saliva samples using the Qiagen EZ2 Connect robot. Using the protocol, we developed, DNA was successfully extracted from 10 saliva samples, with an average DNA concentration of 33 ng/μl and a 260/280 ratio of 1.72. The quality of DNA was comparable to DNA extracted from saliva samples using the validated Roche Magnapure method. The suitability of saliva DNA for high resolution HLA typing was assessed using the One Lambda FASTplex kit. Using TypeStream Visual (TSV) NGS analysis software, we achieved average mapped reads of 334,691 and the results met our documented acceptance criteria for all quality metrics. In conclusion, we have demonstrated that the Qiagen EZ2 Connect robot can be used for successful extraction of DNA from saliva samples to a comparable quality of blood. Validation of this process has allowed our department to avoid unnecessary delays in BBMR extended HLA typing requests and thereby supports patient care.</p><p><b>Sebastian Fernando</b>, <i><span>Jennifer Lord</span>, Nicola Martin, Alison Logan, Kay Poulton</i></p><p><i>University of Manchester NHS Foundation Trust, Manchester, UK</i></p><p>Our current routine HLA typing methodology to support disease association testing is LABType™ SSO (One Lambda). An alternative HLA typing methodology EUROArray (EUROIMMUN), was evaluated for concordance and efficiency. This system combines polymerase chain reaction and microarray technologies. Amplified target DNA labelled with a fluorescent dye hybridises to complimentary DNA probes using BIOCHIP technology. Fluorescence signals are evaluated automatically using the EUROIMMUN Microarray Scanner and EUROArrayScan software. Routine and external proficiency scheme peripheral blood and DNA samples previously HLA typed by LABType™ SSO were tested using the appropriate EUROArray assay. Forty-six samples were tested with the HLA-B27 Direct assay, 41 samples with the HLA-B*57:01 Direct assay and 42 samples with the HLA-DQ2/DQ8-h Direct assay. A range of relevant HLA-B and HLA-DQ alleles were selected to ensure the EUROArray system could differentiate between alleles of interest. We found 100% concordance when compared to the LABType™ SSO results for all tests. The workflow was simple and straightforward with a time saving when compared to LABType™ SSO, however the cost of the EUROArray assay was higher. This study has shown the EUROArray Direct assays for HLA-B27, -B57 and -DQ2/8 detection are valid alternative methodologies to support disease association testing. Advantages of the EuroArray system include no additional DNA isolation, the inclusion of numerous integrated controls for high reliability of results and fully automated standardised evaluation and result generation. Limitations of this assay include the use of blood samples within 14 days of venepuncture, although samples can be frozen to mitigate this.</p><p><b><span>Amy De'Ath</span></b>, <i>Deborah Pritchard, Tracey Rees</i></p><p><i>UK NEQAS for H&amp;I, Talbot Green, UK</i></p><p>Scheme 2A and 2B assesses participants’ ability to correctly determine the cytotoxic and flow cytometry crossmatch status across 40 cell/serum combinations per year, respectively. An analysis of performance by UK and Ireland laboratories from 2018–2022 was performed.</p><p>There were between 15–22 participants in 2A and 19–22 in 2B. Unsatisfactory performance (UP) in 2A ranged between 0% (2020/21)–38.9% (2018), latest 20%. UP in 2B ranged from 0% (2020/21)–10.5% (2022). Over the 5 years, four laboratories had UP in 2B (one lab in two consecutive years) and 11 in 2A. 2 labs had UP in both schemes. Two out of four labs with UP in 2B were due to performance in the T-cell crossmatch, 1/4 in B-cell and 1/4 in T and B cell crossmatch performance. Six out of eleven labs with UP in 2A were in the B-cell without DTT category, 1/11 in B-cell with DTT, 3/11 in both B-cell with and without DTT and 1/11 in T-cell with and without DTT and B-cell with DTT. In 2B, an average of 36/40 crossmatch combinations per year were assessed. An average of 3% assignments per year were incorrect with a 50:50 split of false negatives and false positives. The use of equivocal reporting was low, average 0.5%. Four percent of samples were reported as not tested. Performance in flow cytometry crossmatching is better than cytotoxic crossmatching during the 5 years. These schemes offer a technical assessment of crossmatching, NEQAS encourage laboratories to participate in our educational schemes which more closely mimic clinical practice.</p><p><b><span>Amy Bedford</span></b>, <i>Kathryn Howson, Graham Knighton, Jacqueline Pires, Sarah Maxfield</i></p><p><i>Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK</i></p><p>HLA-B*27 testing is supportive in the diagnosis of autoimmune diseases including Ankylosing Spondylitis due to their strong association with the HLA-B*27 positive genotype. Testing an average of 383 samples per month, Cambridge Tissue Typing laboratory aimed to introduce a direct-from-blood HLA-B*27 detection assay to eradicate DNA extraction required by current polymerase chain reaction (PCR) sequence specific primer (SSP) based methods, and the associated workload. Utilising the BAG Diagnostics FastQ® B*27 Direct assay, DNA is amplified directly from EDTA whole blood by PCR using SSP. Fluorescent-labelled TaqMan® hydrolysis probes are utilised to enable detection of the amplified product by real-time PCR on the QuantStudioTM 3. Implementation of the FastQ® B*27 Direct assay reduced DNA extraction workload by an average of 64%. Increased batch size capacity (93, vs. 20 using the previous technique) streamlined workflows. Risks associated with exposure to carcinogens ethidium bromide and ultraviolet light and handling boiling agarose gel were eradicated. The absence of DNA extraction and adjustment leads to variance in the volume of nucleated cells utilised, which can result in delayed amplification/false negative results. This highlights the importance of sufficient sample mixing prior to aspiration and stringent result acceptance criteria during analysis using PlexTyper® software, with an average repeat rate of 4%. In summary, implementation of robust mixing procedures and stringent quality metrics have facilitated transition to BAG Diagnostics FastQ® B*27 Direct assay leading to a decrease in workload and demands on staff time, and improvement in the health and safety within the laboratory.</p><p><b><span>Kelly Spence</span></b>, <i>Sylvia McConnell, David Turner</i></p><p><i>H&amp;I Department, SNBTS, Edinburgh, UK</i></p><p>H&amp;I supports the Clinical Apheresis Unit (CAU) and Tissues, Cells and Advanced Therapeutics (TCAT) departments by performing CD34+ stem cell enumeration in acute myeloid leukaemia and multiple myeloma patients receiving autologous stem cell transplantation. The enumeration of CD34+ cells using flow cytometry and the International Society of Hematotherapy and Graft Engineering (ISHAGE) protocol is an established method for the evaluation of stem cell numbers in peripheral blood and apheresis products. Whilst a single (flow cytometry) platform is the recommended approach, locally a dual platform method is in operation, using both flow cytometry and Haematology analyser data. The BD Stem Cell Enumeration assay has been validated on the BD FACSLyric flow cytometer. A comparison between the single and dual platform methodologies using the ISHAGE protocol was undertaken. Analysis was performed on 30 stem cell harvest samples. Strong correlation between the single and dual platform methods was observed for CD34+ absolute count (cells/μl) for stem cell harvest samples (<i>r</i> = 0.99, <i>p</i> &lt; 0.05). There was also a strong correlation between the White Blood Cell (WBC) count from the Haematology analyser and the CD45+ count from the flow cytometer (<i>r</i> = 0.94, <i>p</i> &lt; 0.05). In this validation both the single and dual platform methods for calculating CD34+ cell counts were similar, as were the WBC counts and CD45+ counts from the Haematology analyser and the flow cytometer. This data supports using the recommended single platform method as part of the CD34+ enumeration testing.</p><p><b><span>Rebecca McGuire</span></b><sup>1,2</sup>, <i>Paul Wright<sup>3</sup>, Steven Jervis<sup>1</sup>, Stephine Whiteside<sup>1</sup>, Malcolm Guiver<sup>4</sup>, Kay Poulton<sup>1,2</sup></i></p><p><i><sup>1</sup>Transplantation Laboratory, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK; <sup>2</sup>Faculty of Biology, Medicine and Health, Division of Medical Education, School of Medical Sciences, University of Manchester, Manchester, UK; <sup>3</sup>H&amp;I Laboratory, Liverpool Clinical Laboratories, Liverpool, UK; <sup>4</sup>Department of Clinical Virology, Manchester University NHS Foundation Trust, Manchester, UK</i></p><p>Acute myeloid leukaemia is an aggressive haematological malignancy with a poor prognosis. Allogeneic haematopoietic progenitor cell transplantation is the only available curative treatment, but it does not eliminate the risk of relapse. Hypotheses for improving relapse rates include utilising donors with advantageous KIR2DL1 allele groups. However, an inexpensive, rapid, and reliable method to discern KIR2DL1 groups does not currently exist. Here, we show that homology between several Killer-cell Immunoglobulin-like Receptor (KIR) loci restricts accurate genotyping of KIR2DL1 allele groups by traditional TaqMan™- based real-time PCR methods. Using multiple sequence analysis, we found that the single nucleotide polymorphisms within codons 114 (rs11673144) and 245 (rs34721508), used to discern KIR2DL1 allele groups, are shared between other KIR loci. In a traditional TaqMan™ real-time PCR assay, this results in non-specific binding and incorrect allele group assignment. Contrary to existing literature, our analysis demonstrated that specificity could not be achieved through the introduction of 3´ terminal mismatches in the forward primers. The results described illustrate the challenges in designing an efficient real-time PCR assay for functional group typing of KIR2DL1. We anticipate our assay to provide the foundation for a more sophisticated real-time PCR assay. Developing a test capable of defining KIR2DL1 allele groups remains of interest to research groups focused on reducing the incidence of acute myeloid leukaemia relapse post-transplant. With further improvements, the described assay could fulfil this goal.</p><p><b><span>Abigail Levy</span></b>, <i>Arthi Anand, John Wintour-Pittom</i></p><p><i>Hammersmith H&amp;I NWLP, London, UK</i></p><p>The Histocompatibility &amp; Immunogenetics (H&amp;I) department at NWLP support renal transplantation at West London Renal Transplant centres. A 24/7 on-call service operates for crossmatching of local patients for transplantation. Timely and effective communication between the renal team receiving deceased donor offers and H&amp;I on-call team is critical for turnaround of compatibility assessment and testing. Historically H&amp;I on-call team have been contactable via ICHT switchboard with provision of a weekly on-call rota. A number of challenges were experienced using this method including calls being directed to the wrong team member interrupting rest and the potential for GDPR breach with patient identifiable information recorded in home environment. ICHT implementation of ALERTIVE app in June 2022 provided alternative on-call communication. H&amp;I were the first pathology service to adopt the app and collaborated with Trust Telecoms &amp; ThamesNet Services to set up the App to meet H&amp;I on-call needs, with Go-Live in February 2023. ALERTIVE improves the speed and quality of communication, between clinical staff and H&amp;I on-call team, in time critical deceased donor transplantation pathway. The ALERTIVE app has been in routine use for three months and has had a very favourable response from H&amp;I on-call team. The app has simplified the process of communicating between Renal and H&amp;I on-call teams, enabling more efficient decision-making. The clinical messaging app is giving us a wealth of new data that was either very hard to get or was unavailable including number of offer related call outs. There are exciting new uses for this data including workforce planning.</p><p><b><span>Dayna Badaro</span></b><sup>1,2</sup><i>, Sarah Maxfield<sup>1</sup></i></p><p><i><sup>1</sup>Cambridge University Hospital NHS Foundation Trust, Cambridge, UK; <sup>2</sup>University of Manchester, Manchester, UK; <sup>3</sup>Organ and Tissue Donation and Transplantation, NHS Blood and Transplant, Stoke Gifford, UK</i></p><p>In the United Kingdom, prior to September 2019, deceased donor kidneys were allocated following the National Health Service Blood and Transplant-Organ Donation and Transplantation (NHSBT-ODT) 2006 Kidney Allocation Scheme (KAS). A review by the Kidney Advisory Group prompted significant changes to national allocation with the aim of improving fairness in kidney offering, reducing transplant waiting times and improving longevity of matched transplants. A retrospective clinical audit aimed to review the impact for patients on the Cambridge deceased donor kidney transplant waiting list (TWL), and determine whether the objectives of the 2019 KAS have been met. Transplant data was obtained from NHSBT-ODT spanning 24 months pre-(<i>n</i> = 298) and post-(<i>n</i> = 303) implementation. Data was reviewed taking into consideration the impact of the COVID-19 pandemic. Results show reduced average wait time (days) for highly sensitised patients with a calculated reaction frequency (cRF) ≥85%. In accordance with simulation data, donor/recipient index matching has been achieved, with the majority of ‘low risk’ donors allocated to ‘low risk’ recipients, and the same being true for ‘high risk’ donors/recipients. Divergent donor/recipient age matching was accounted for by difficult to match patients prioritised according to Tier A (cRF 100%/wait time &gt;7 years/matchability score of 10). Contrary to simulation data, Cambridge did not observe a reduction in donation after cardiac death (DCD) transplants following changes to national allocation (<i>n</i> = 174 pre, <i>n</i> = 192 post). In summary data indicates a reduction in average wait time for highly sensitised patients on the Cambridge TWL and suggests objectives of the 2019 KAS have been met.</p><p><b><span>Michelle Carr</span></b><sup>1</sup>, <i>Shelley Harris<sup>1</sup>, Judith Worthington<sup>1</sup>, Alex Woywodt<sup>2</sup>, Kay Poulton<sup>1</sup></i></p><p><i><sup>1</sup>Transplant Laboratory MFT, Manchester, UK; <sup>2</sup>Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK</i></p><p>In January 2020 a 24-year-old male with chronic renal failure secondary to reflux nephropathy received his third kidney transplant from a fully HLA matched DBD donor. In March 2020 his kidney function declined with a concurrent rise in creatinine and proteinuria. Tacrolimus levels were variable throughout the course of the patient's transplant history and non-adherence was suspected. A retrospective analysis of non-HLA antibodies using LABScreen™ Autoantibody kits identified that the patient developed antibodies to Glutathione S-Transferase Theta 1 (GSTT1). After his second transplant tacrolimus levels were below target range (&lt;2.5 μg/L), which was followed by the first appearance of the GSTT1 antibody (6000 MFI = 95% percentile). This coincided with a biopsy showing chronic transplant glomerulopathy. Following his third transplant tacrolimus levels remained variable with some levels above and below target range. His transplant function deteriorated further in Spring 2023. A biopsy at this time showed borderline T cell mediated rejection with moderate interstitial fibrosis and tubular atrophy. There was a rapid increase in GSTT1 antibody levels with a peak of 11,000 MFI (&gt;95% percentile). We propose that immune-mediated inflammatory processes triggered by non-adherence episodes caused cellular damage. In response to such damage, intracellular components such as GSTT1 would be released, enabling recognition and the subsequent immune response leading to GSTT1 antibody production. We propose that the detection of antibodies to GSTT1 should be regarded a marker of nephrotoxicity and ongoing trauma to the transplanted kidney.</p><p><b><span>Adrian Handley</span></b>, <i>David Briggs, Clare Collins</i></p><p><i>NHS Blood and Transplant, Birmingham, UK</i></p><p>All measurements come with an element of uncertainty and are only truly useful when that degree of uncertainty is understood. The use of semi-quantitative data from Luminex HLA antibody testing is essential in enabling cardiothoracic transplant compatibility assessment; however, the level of uncertainty was not understood. BSHI/BTS CTAG guidelines set risk levels based on MFI values produced in Luminex Single Antigen Bead HLA antibody assays. Additionally, ISO15189:2012 states that laboratories should calculate measurement uncertainty (MU) for all accredited tests. In this study we used simple statistical tools to identify degrees of MU and some of the contributing factors. These allowed us to quickly assess the effectiveness of changes subsequently made. We identified significant difference between results obtained by individual testing personnel and were able to reduce this through our interventions. Reed et al. (<i>Am J Transplant</i>. 2013;13(7):1859–1870) suggested MU as %CV was in the region of 20%–62%. Our study found this to range from 8% at 20,000 MFI up to 18% at 1000 MFI, the threshold for positivity.</p><p>The tools we developed allow ongoing monitoring of assay performance and provide the basis for future quality initiatives, contributing to sustainable, evidence-based quality improvement. MU calculations are an effective quality assurance and improvement tool and are easily implemented. By combining the tools used in this study, levels and sources of variation can be identified, and the effect of any changes to process can be easily assessed for effectiveness.</p><p><b><span>Saima Azhar Salim</span></b>, <i>Louise Walsh, Geraldine Donnelly, David Keegan, Joseph Kelly, Mary Keogan</i></p><p><i>H&amp;I Lab, Beaumont Hospital, Dublin, Ireland</i></p><p>Flow cytometric crossmatch (FXM) is performed during pre-transplant histocompatibility workup. False positive results may prevent a potential recipient receiving a suitable transplant. Many labs use a three-colour FXM assay established using a dual-laser flow cytometer, which includes phycoerythrin (PE), fluorescein isothiocyanate (FITC) and peridinin-chlorophyll proteins (PerCP). There is significant spectral overlap between PE and FITC potentially leading to high background fluorescence, and possible false positive B cell FXM. This study was undertaken to determine optimum fluorochrome combinations to minimise spectral overlap and maximise consistency with the current method for which clinically validated cut-offs are available. Fluorochromes Brilliant Violet (BV421) and Allophycocyanin (APC) were selected for labelling CD3+T cells and CD19+B cells respectively. Thirty-four FXM (total 63 samples for T cell and 61 samples for B cell FXM) were performed with fluorochromes CD3BV421, CD19APC and anti-human IgG FITC. Fourteen FXM (total 35 samples for T and B cell FXM) were performed with fluorochromes CD3PerCP, CD19APC and anti-human IgG FITC. FXM assays were performed on Becton Dickenson (BD) FACSLyric™ and results compared to the standard FXM assay. In comparison to the standard FXM, a statistically significant difference in sample T cell ratio and B cell ratios (<i>p</i> &lt; 0.005) were observed for CD3BV421-CD19APC FXM. Our pilot data suggest no significant difference in T and B cell ratios for CD3PerCP-CD19APC (<i>p</i> &gt; 0.05). Data suggest that substitution of CD19PE with CD19APC minimises spectral overlap, reducing the risk of false positive FXM results, without significantly altering T and B cell ratios.</p><p><b><span>Ana Bultitude</span></b><sup>1</sup>, <i>Anthony Poles<sup>2</sup>, Sue Jordan<sup>1</sup>, Anthony Calvert<sup>2</sup>, Deborah Sage<sup>1</sup></i></p><p><i><sup>1</sup>NHS Blood and Transplant, Tooting</i>, <i>UK; <sup>2</sup>NHS Blood and Transplant, Filton, UK</i></p><p>HNA-3 is a bi-allelic antigen, -3a/-3b, with 95% of the UK population encoding at least one HNA-3a allele. HNA-3 is expressed on a variety of cell types including lymphocytes and renal endothelial cells. Accordingly, HNA-3 specific antibodies, which can develop against the non-self-variant in homozygous individuals, have been implicated in rejection episodes following renal transplantation. Here, we report a case of a renal transplant recipient displaying anti-HNA-3a antibodies that prevented transplantation. In August 2022, the patient was offered an altruistic kidney with a 2,1,1 mismatch grade, yielding a negative virtual crossmatch result. However, final wet laboratory flow cytometric crossmatch (FCXM) results presented an unexplained strong positive T and B cell result with a negative auto FCXM. The patient had no previous transplant history and no detectable HLA antibodies since their initial referral. HNA genotyping was performed on both patient and donor, determining HNA-3b3b and HNA-3a3a genotypes respectively. Patient sera was screened for anti-HNA antibodies, confirming the presence of HNA-3a-specific antibodies. The patient was removed from the virtual crossmatching programme as they no longer met the eligibility criteria but remained on the deceased donor waiting list. A further nine deceased donor kidneys were accepted and crossmatched, all of which were T and B cell positive. In March 2023, the patient received a deceased donor kidney which produced a T and B cell negative FCXM. The donor was retrospectively genotyped as HNA-3b3b. The patient continues to do well with no antibody-mediated rejection detected thus far.</p><p><b><span>Hawzhin Jabar</span></b>, <i>Luke Foster</i></p><p><i>NHS Blood and Transplant, Birmingham, UK</i></p><p>Accurate HLA typing is essential to facilitate safe solid organ transplantation. Within the UK, H&amp;I laboratories are required to meet the minimum typing requirements set by NHSBT-OTDT, which includes reporting results that allow for the assessment of any donor specific antibodies (DSA) in a particular donor-recipient combination. Failure to do so can lead to inappropriate allocation or an increased risk of transplant rejection. Here we report the identification of the DRB1*14:15 allele in a deceased solid organ donor who was HLA typed on-call using LinkSeq real-time PCR (One Lambda). Ordinarily, it would not be a requirement to report HLA-DRB1*14 to the second field under the minimum typing requirements, however, interestingly, HLA-DRB1*14:15 does not encode the DR14 antigen, but codes for the DR8 antigen. Therefore, in this instance there was a requirement to report HLA-DRB1*14 to the second field to allow accurate allocation and assessment of any potential DSA. Despite being locally rare, HLA-DRB1*14:15 is listed as well-documented in European populations within the CIWD 3.0.0 catalogue, and common in Asian/Pacific Islands and Native American populations. In this case, donation proceeded with both kidneys being transplanted in two patients, one locally, and out of region. Although the patient transplanted locally was cRF 0% and therefore had no DSA, elucidation of the DR8 antigen may be important for any future post-transplant DSA monitoring.</p><p><b><span>Richard Battle</span></b>, <i>Emma Ross, Sylvia McConnell, David Turner</i></p><p><i>H&amp;I Department, SNBTS, Edinburgh, UK</i></p><p>The SNBTS H&amp;I laboratory provides ∼2300 apheresis donor platelet units annually to HLA sensitised refractory patients across Scotland from a panel of ∼800 typed donors. Data is recorded for each transfusion, including match grade (A = matched for HLA-A and B antigens, B1 = one HLA antigen mismatch, B2 = two antigen mismatch etc.) and cumulative HLA antibody MFI (cMFI) against mismatches. Platelet transfusions were grouped by match and cMFI and assessed against post counts relative to pre-counts that is, a measure of transfusion increment. Pre- and post-platelet counts were available on 1486 cases between 2015–2022. Recipient and donor HLA-A and B types at first field defined A, B1–B4 matching. cMFI was calculated following One Lambda SAB I testing. Analyses used ANOVA or <i>t</i>-test between groups. In 1166 HLA compatible transfusions (cMFI &lt; 2000) no differences were seen in mean increment between A, B1, B2, B3 and B4 matches; 22.5, 22.9, 22.1, 24.1, 30.2 respectively (ANOVA <i>p</i> = 0.14). In 1486 patients grouped according to cMFI, cases with &lt; 2000 had a mean increment = 23.0, with &gt; 2000 &lt; 10,000 mean = 18.0 (<i>p</i> &lt; 0.001) and &gt; 10,000 mean = 8.0 (ANOVA <i>p</i> = 1.55 × 10–15). This analysis of platelet counts after HLA selected platelet provision shows that, as expected, the match grade of the platelets does not impact on the immediate increment. The level of HLA antibody, as defined by cMFI, affects the post transfusion count, especially when cMFI &gt; 10,000. This data will help locally in selection of optimal platelet units for patients.</p><p><b><span>Patrick Flynn</span></b><sup>1</sup>, <i>Sebastian Fernando<sup>2</sup>, Judith Worthington<sup>1</sup>, Kay Poulton<sup>1</sup></i></p><p><i><sup>1</sup>Transplantation Laboratory, Manchester Royal Infirmary, Manchester, UK; <sup>2</sup>School of Health Education and Public Health Sciences, University of Manchester, Manchester, UK</i></p><p>The aim of this study was to devise an algorithm that would predict Flow Cytometry crossmatch results using SAB Median Fluorescent Intensity (MFI) levels and to test this correlation using samples tested from a NEQAS Scheme 2B cohort.159 NEQAS 2B serum samples were screened using LABScreen™ SAB and 40 NEQAS 2B peripheral blood samples were HLA typed with LABType™ SSO. Donor-Specific Antibodies (DSA) were identified for each cell-serum combination tested and cumulative MFI values calculated for each test. HLA Class I MFIs were combined to predict the T cell crossmatch. For the B cell crossmatch prediction, two options were considered: (i) HLA Class II MFI values alone and (ii) HLA Class I + Class II MFIs. Receiver Operating Characteristic analysis was carried out to identify the combined MFI cut off that predicted NEQAS consensus results with the greatest sensitivity and specificity value. HLA Class I combined MFI &gt; 5000 predicted T cell crossmatch results with 96% sensitivity, 100% specificity, 100% Positive Predictive Value (PPV) and 92% Negative Predictive Value (NPV). For B cell results, HLA Class I + Class II combined MFIs &gt; 11,000 gave the best model showing 97% sensitivity, 79% specificity, 95% PPV and 85% NPV. However, for samples with only HLA Class II sensitisation, combined MFIs &gt; 13,000 improved the B cell crossmatch predictions: 92% Sensitivity, 91% specificity, 92% PPV and 91% NPV. Using this model, combined MFI values can be used to predict the immunological risk posed by DSA when it is not possible to carry out a crossmatch test.</p><p><b><span>Jonathan A.M. Lucas</span></b><sup>1</sup>, <i>Richard M. Szydlo<sup>1,2</sup>, Shelley Hewerdine<sup>1</sup>, Steven G.E. Marsh<sup>1,3</sup>, Neema P. Mayor<sup>1,3</sup></i></p><p><i><sup>1</sup>Anthony Nolan Research Institute, Royal Free Hospital, UK; <sup>2</sup>Department of Medicine, Imperial College, UK; <sup>3</sup>UCL Cancer Institute, Royal Free Hospital, UK</i></p><p>The effect of matching for HLA-E on the outcome of hematopoietic cell transplantation (HCT) has thus far been inconsistent and has not been studied in a UK cohort where there is an abundant use of alemtuzumab for T-cell depletion. We analysed HLA-E genotypes in 1513 UK HCT patients with a haematological malignancy and their unrelated donors using full-length PacBio Single Molecule Real-Time DNA sequencing. After adjusting for clinical factors that affected outcome prognoses including classical HLA matching out of 12, the presence of mismatches at both HLA-E loci (HLA-Emm; <i>n</i> = 30) was significantly associated with a reduced risk of relapse (HR 0.44; 95% CI 0.20–0.98, <i>p</i> = 0.04) in comparison to being HLA-E matched (HLA-Em; <i>n</i> = 788). A non-significant but beneficial effect on Overall Survival (OS) and Event-Free Survival (EFS) was also observed for two HLA-Emm loci compared to HLA-Em (OS: HR 0.73, <i>p</i> = 0.30; EFS: HR 0.77, <i>p</i> = 0.38). There were no significant differences in HCT outcomes correlated with a single HLA-Emm (<i>n</i> = 450). Assessing directionality of mismatches showed that a bi-directional HLA-Emm (<i>n</i> = 45) was significantly associated with reduced risks of relapse (HR 0.40; 95% CI 0.20–0.83, <i>p</i> = 0.01), increase in EFS (HR 0.55; 95% CI 0.32–0.93, <i>p</i> = 0.02) and a non-significant increase in OS (HR 0.72, <i>p</i> = 0.16) compared to HLA-Em. We hypothesise that mismatching alleles at the HLA-E locus results in sufficient genetic disparity to provide a stronger Graft-versus-Leukemia effect, without eliciting detrimental Graft-versus-Host responses, hence no significant differences in acute Graft-versus-Host Disease or Transplant Related Mortality, as observed in this study.</p><p><b><span>Sajadhossein</span>\\n <span>Bazrafshani</span></b><sup>1</sup>, <i>Mohammadreza Bazrafshani<sup>2</sup></i></p><p><i><sup>1</sup>ENT Department, Worcester, UK; <sup>2</sup>Kerman University of Medical Sciences, Kerman, Iran</i></p><p>Cyclosporine, a calcineurin inhibitor, has a narrow therapeutic index and shows considerable inter-individual variability in pharmacokinetics. Cyclosporine is a P-glycoprotein (P-gp) substrate, a multidrug resistance gene (MDR1) product. Some of the single nucleotide polymorphisms (SNPs) of MDR-1 correlate with the variable activity of P-gp in vivo, and it is thought that these polymorphisms are associated with pharmacokinetic variations in cyclosporine therapy. Genotyping assays (PCR-SSP and PCR-RFLP) were performed for detection of frequency within two functional MDR-1 SNPs (C1236T in exon 12 and C3435T in exon 26) in 60 patients and the correlation between genotyping and concentration/dose ratio of cyclosporine was investigated. Data analysis revealed that C3435T polymorphism correlated with the concentration/dose ratio significantly. The concentration/dose ratios were 59.36 μg/L/kg/12 h (<i>p</i> &lt; 0.001) and 30 μg/L/kg/12 h (<i>p</i> = 0.049) lower in homozygous wild-type patients (CC) rather than homozygous mutant-type (TT) and heterozygous patients (CT) respectively. Moreover, heterozygous patients (CT) had 29.36 μg/L/kg/12 h (<i>p</i> = 0.001) which was less than homozygous mutant-type patients (TT). These findings suggest that, for the given dose, the blood concentration is lower in homozygous wild-type individuals (CC type in 3435 position). Our results revealed that MDR-1 genotype appears to influence cyclosporine drug levels and MDR-1 genotyping may provide a useful clinical guide in predicting the required dose after renal transplantation.</p><p><b><span>Mazen Mabrok</span></b><sup>1</sup>, <i>Renuka Palanicawander<sup>2</sup>, Betia Nouri<sup>1</sup>, Rachel Smith<sup>1</sup>, Arthi Anand<sup>1</sup>, Natalia Brodaczewska<sup>3</sup></i></p><p><i><sup>1</sup>Histocompatibility &amp; Immunogenetics Laboratory, North West London Pathology, Imperial College Healthcare NHS Trust, London, UK; <sup>2</sup>Centre for Haematology, Imperial College Healthcare NHS trust, London, UK; <sup>3</sup>Specialist Integrated Haematological Malignancy Diagnostic service (SIHMDS), Imperial College Healthcare NHS Trust, London, UK</i></p><p>Monitoring of donor chimerism after haematopoietic stem cells transplantation (HSCT) is vital for early effective therapeutic interventions. Short tandem repeat (STR) assays are the current gold standard for chimerism monitoring after allogeneic HSCT. The emergence of Next Generation Sequencing (NGS) CE-IVD approved assays with improved limit of detection of around 0.05% offer a promising alternative. In this evaluation exercise, we explored the technical capacity of NGS in post-transplant chimerism monitoring, by retrospectively analysing samples from 13 patients from our centre using the Dvysr® NGS chimerism assay (Sweden, Stockholm). All patients enrolled in this evaluation were tested at three time points (whole blood &amp; T- Cell), selected in collaboration with the clinical team based on clinical signs of relapse, increase in MRD markers and the profile obtained using the PowerPlex 16 Multiplex STR system (Promega). Our results showed very strong correlation between the NGS and the STR assays (Pearson score 0.998 &amp; 0.999) with a shift of 1%–1.5% at the pre-relapse time point observed in 55% of the patients enrolled in this evaluation. The NGS assay needed 50% less time to analyse and in 70% of the patients tested, the NGS assay yielded more informative markers than the STR assay. NGS chimerism assay promises improved diagnostic performance and usability in our evaluation. Increased sample size and inclusion of lineage specific cell separation will further strengthen validity of the improved diagnostic performance and usability of NGS chimerism assay as alternative to STR assays including correlation with clinical presentation.</p><p><b><span>Emma Holmes</span></b><sup>1</sup>, <i>Jasmaine Lee<sup>2</sup>, Winnie Chong<sup>2</sup>, Deborah Sage<sup>1</sup>, Martin Howell<sup>2</sup></i></p><p><i><sup>1</sup>NHSBT Tooting Centre, Tooting, UK; <sup>2</sup>NHSBT Service Development, Colindale, UK</i></p><p>Nanopore sequencing presents a new technology for high resolution HLA typing that is, considered faster and potentially cheaper than existing methods used for next generation sequencing (NGS). NanoTYPE (Omixon) can be used for batches of up to 24 samples or for single samples, which has the potential to be used for deceased donor HLA typing. The aim of this study was to evaluate the NanoTYPE assay in two NHSBT H&amp;I laboratories; Colindale Service Development and Tooting, using the same batch of 96 samples that had previously been typed by current rapid HLA typing or NGS methods. All NanoTYPE reagents, R9.4 flow cells and a MinION mk1B device were provided by Omixon and Oxford Nanopore Technologies for this study. Eight samples were set-up using the single-sample protocol. The remaining samples were set-up in multi-batches of varying sizes. There was 100% concordance at second field resolution between both sites, and with previous typing data, for HLA-A, -B, -C, DRB4, DRB5 and DPA1. Non-concordance with previous typing data was seen for either HLA-DRB1, DRB3, DQA1, DQB1 or DPB1 loci in 10/96 (10%) samples at Tooting and 9/96 (9%) samples at Colindale, which was attributed to allele imbalance, known low amplification of certain genotypes such as DQB1*03, allele dropout and detection of potential novel alleles. After manual review of the sequencing data, 95/96 samples at Colindale and 93/96 samples at Tooting were concordant with previous results. Our study has proven that the NanoTYPE assay is simple to use and enables high resolution HLA typing.</p><p><b><span>Emma Ross</span></b><sup>1</sup>, <i>Sylvia McConnell<sup>1</sup>, Richard Battle<sup>1</sup>, Nicole Priddee<sup>2</sup>, David Turner<sup>1</sup></i></p><p><i><sup>1</sup>H&amp;I Department, SNBTS, Edinburgh, UK; <sup>2</sup>Donor Medicine, SNBTS, Edinburgh, UK</i></p><p>The SNBTS H&amp;I laboratory is responsible for the provision of HLA selected platelets in Scotland. In 2022, 96 patients were supported with a total of 2317 platelet units. A key performance indicator (KPI) for the service is &gt;60% of allocated donor platelets should be an A grade or B1 grade match with the patient (A grade = no HLA-A or HLA-B mismatches at first field; B1 = only one HLA-A or HLA-B mismatch). This target has averaged 57.7% (range 46%−72%) between January 2016 and September 2022, with an overall downward trend observed. In contrast, the number of individual donor platelets issued as HLA selected increased from 1288 units in 2016 to 2317 in 2022 (+55.5%). To make the best use of a limited donor panel, H&amp;I have worked with SNBTS Donor Services (DS) to increase availability of best matched donor platelets. Utilising a Quality Improvement approach, business analytic tools and specialist donor communications, we concentrated on the national identification and (re)engagement of HLA-A and -B homozygous donors which constitute 3.5% of the panel but have the potential to provide A grade matches for ∼68% of patients (Jan–June 2023). Since the project initiation we have recorded an upward trend in the percentage of A and B1 platelet allocations over the period October 2022 (49.5%)–May 2023 (72.4%). The target of &gt;60% has been achieved consistently from Jan 2023. Collaborative working with DS has enhanced the donor panel, improving ‘off the shelf’ access to best matched HLA selected platelets.</p><p><b><span>Felicity May</span></b><sup>1</sup>, <i>Sian Griffin<sup>2</sup>, Madhvi Menon<sup>3</sup>, Tracy Hussell<sup>3</sup>, Tracey Rees<sup>1</sup></i></p><p><i><sup>1</sup>Welsh Transplantation and Immunogenetics Laboratory, Welsh Blood Service, Pontyclun, UK; <sup>2</sup>Department of Nephrology and Transplantation, University Hospital of Wales, Cardiff, UK; <sup>3</sup>Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK</i></p><p>Human leukocyte antigen incompatible (HLAi) transplantation remains an important option for very highly sensitised patients. Crossing the HLA barrier is associated with increased risk of antibody-mediated rejection (AMR) and graft failure. Desensitisation, induction and maintenance immunosuppression aim to minimise this risk. There is poor consensus on optimal treatment protocols, and patient response varies. In this single centre, retrospective study, we assessed incidence of rejection and graft survival in a clinical cohort of 27 HLAi transplant recipients desensitised with rituximab and cycles of double filtration plasmapheresis. There was poor association between established risk factors and incidence of AMR, suggesting the involvement of other, currently unknown, factors. HLA antibody response and AMR are influenced by a complex cytokine network supporting the generation and survival of antibody producing cells. We conducted a literature review and graded 459 serum protein analytes (cytokines, chemokines, hormones etc.) based on the quality of evidence and proximity of involvement in humoral memory response. We evaluated serum concentrations of the 40 highest graded analytes in longitudinal samples derived from our clinical cohort using a bespoke multiplexed Luminex assay. Due to low samples numbers and heterogeneity of the analyte profiles, we were unable to establish clear association with patient clinical outcomes. However, we observed significant and sustained changes to key signalling molecules known to influence germinal centres, B-cells and plasma cells. We noted a reduction in levels post-desensitisation/immunosuppression for the majority of our panel, but increased or unchanged levels of several key supportive signals and suppression of several immune regulators.</p><p><i>Agnieszka Ojrzynska<sup>1</sup>, Kylara Hassall<sup>1</sup>, Katie Butler<sup>1</sup>, Graham Shirling<sup>1</sup>, Sharon Vivers<sup>2</sup>, Raymond Fernando<sup>1</sup></i></p><p><i><sup>1</sup>Solid Organ Group, Royal Free Hospital, London, UK; <sup>2</sup>Anthony Nolan Histocompatibility Laboratories, London, UK</i></p><p>Increasing numbers of multiple myeloma patients, treated with Daratumumab (Dara), are being listed for kidney transplantation. Dara is a human monoclonal anti-CD38 IgG antibody that interferes with both pre-transplant allogeneic lymphocyte crossmatches and pre-transfusion compatibility testing, as it binds to CD38 molecules expressed on many blood cells including lymphocytes. This can simulate the presence of donor-specific antibodies (DSA) and lead to false positive crossmatch results. The aim of this study was to investigate methods to mitigate Dara interference with flow cytometry crossmatches (FCXM). Our centre has transplanted four patients undergoing Dara treatment. Three of these patients were HLA antibody negative and one patient was HLA antibody positive but HLA DSA negative. Six third party peripheral blood FCXMs, and one retrospective transplant FCXM with spleen cells, were performed using our standard procedure, as well as an amended protocol using donor cells pre-incubated with dithiothreitol (DTT) (0.05–0.1 M) which can cleave the stabilising disulphide bonds of the CD38 molecule. All third-party crossmatches using our standard procedure were T cell positive, two were also B cell positive. The retrospective crossmatch performed with spleen cells was T and B cell positive. All crossmatches performed with DTT were T and B cell negative. Our results show that DTT eliminates the false positivity observed when sera from Dara treated patients are used in FCXM. HLA positive control results were unaffected suggesting that HLA molecules are not affected by DTT treatment. Therefore, DTT can be used to mitigate DARA mediated false positive results in FCXM.</p><p><b><span>Daniel Eggleston</span></b>, <i>Helena Lee</i></p><p><i>Manchester Royal Infirmary, Manchester, UK</i></p><p>Haematopoietic progenitor cell transplantation (HPCT) is well-established as a curative treatment for malignant and non-malignant haematological disorders. However, patient monitoring is still essential following transplantation. Post-transplant donor chimerism testing can identify complications including relapse or graft failure. Relapse is a particular area of concern for patients diagnosed with Myelodysplastic syndromes (MDS) or high-risk leukaemias who have undergone HPCT. Most laboratories perform chimerism monitoring by PCR-Short Tandem Repeats (PCR-STR). This technique uses differences in short repeated sequences of DNA and capillary electrophoresis to determine percentage donor chimerism. However, other methods are emerging that offer improved sensitivity and more informative markers. One such technique is digital PCR (dPCR). During dPCR, a PCR reaction mixture is partitioned into individual droplets. Each function as an individual assay, with the presence of a reaction indicating donor or recipient DNA, increasing technique sensitivity. Using dPCR, we investigated eleven patients diagnosed with MDS or Myeloid Leukaemias whose percentage donor chimerism reached 100% by PCR-STR but dropped in subsequent samples. We tested our cohort using JETA DigitalTRACE™ dPCR technology with the Qiagen QIAcuity machine at two or more timepoints preceding this reduction in donor chimerism using three markers. In 10 of our 11 patients, a reduction in donor chimerism was identifiable by dPCR whereas analysis by PCR-STR had suggested samples were 100% donor chimerism. This raises the possibility of an earlier detection of changes in donor chimerism and an improved follow up process. Chimerism by dPCR is therefore a viable replacement for PCR-STR and may identify events earlier post-HPCT.</p>\",\"PeriodicalId\":14003,\"journal\":{\"name\":\"International Journal of Immunogenetics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.3000,\"publicationDate\":\"2023-09-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1111/iji.12630\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"International Journal of Immunogenetics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1111/iji.12630\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"GENETICS & HEREDITY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"International Journal of Immunogenetics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/iji.12630","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

摘要

Alison Cleaton、Emma Burrows、Kimberley Robinson、Michael Richardson、Deborah Pritchard、Tracey ReesWelsh血液服务中心、Ely Valley Road、Talbot Green、UK使用LABScreen对等待肾移植的患者进行定期HLA抗体检测™ HLA抗体测定。在新冠肺炎大流行期间,我们观察到一些HLA抗体谱发生了无法解释的变化。调查显示,几名患者在变化前患有新冠肺炎,因此,对移植等待名单上所有已知感染新冠肺炎的患者进行了审查。移植等待名单上的237名患者中有六十六名患有新冠肺炎(2020年3月至2022年7月)。分析新冠肺炎感染前后样本的HLA抗体结果,以了解现有HLA抗体水平的变化(增加的Luminex中值荧光强度(MFI)值)或扩大的抗体谱(增加的cRF)。52名(78.8%)患者的cRF或MFI没有可检测的变化;5例(7.6%)MFI发生变化(但cRF无变化);9例(13.6%)MFI和cRF发生变化。九分之二的患者之前没有记录过致敏事件;其中4人曾做过移植手术,4次输血,4次怀孕;以及三个多重致敏事件。所有9名患者在随后的样本中均出现持续的cRF变化(随访至2022年12月)。九分之三的患者因此被确定为因新冠肺炎感染而免疫抑制改变;这些患者的cRF增加了20%-76%,现在所有患者的cRF98%-100%。尽管大多数等待肾脏移植的患者在新冠肺炎感染后HLA抗体谱没有变化,但有9名患者的cRF增加,这并不是暂时的。减少或停止免疫抑制以帮助从新冠肺炎中恢复被确定为三名患者的原因。Adrienne Seitz,Clive Carter,Brendan Clark,Richard BakerLeeds教学医院NHS Trust,Leeds,UK通过测量血清IgG HLA抗体来评估移植前的免疫风险水平,该抗体可由长寿浆细胞和记忆B细胞产生。记忆B细胞可以在不产生抗体的情况下循环,因此它们对抗体库的贡献可能没有得到充分的重视。我们描述了一种通过非特异性刺激外周记忆B细胞来改善移植前风险评估的体外方法。用toll样受体激动剂R848和白细胞介素-2培养来自三名未致敏志愿者和六名致敏患者的外周血单核细胞9天。使用单个抗原珠测试细胞培养上清液中的IgG HLA抗体。将其与匹配的血清样本进行比较。使用流式细胞术评估静息的第0天和刺激的第9天B细胞表型,确认转换为分泌抗体(CD24-CD38hi)、类别转换记忆(CD27+IgD-)和血浆(CD38+CD138+)细胞。在细胞上清液中发现HLA I和II类抗体,65%存在于匹配的血清样品中。当上清液显示出额外的HLA抗体时,这些抗体可能归因于先前的移植,或者已经存在于患者的历史血清档案中。我们展示了一种可以使用大多数H&amp;I实验室。当评估活体供体对时,这种测定可能是有用的,其中供体可能重复与妊娠相关的错配,以及在去除“其他不可接受的抗原”之前的再移植物中。最后,在新型围移植药物的背景下,它可以与除名策略一起考虑。Sophie Chambers,Robert Whittle,John Goodwin,Tim KeyNHS Blood and Transplant,Barnsley,UK HLA-B外显子II 76–83位氨基酸(aa)和HLA-a亚群的差异主要是高免疫原性公共表位Bw4和Bw6的原因。Bw4特异性抗体在同种免疫的Bw6纯合子中常见,而Bw6特异性抗体则在Bw4纯合子中出现。由于Bw4复合物中存在变异,先前已经报道,在具有Bw4特异性抗体的一定比例的Bw6纯合个体中,反应性可以局限于HLA Bw4亚型表位。我们报告了三名具有Bw4表型的个体,他们证明了Luminex单抗原珠确定的Bw4表位的同种抗体与他们自己的不同。对于携带B13的肾脏患者L,对除B13以外的所有Bw4阳性珠都存在反应性。反应性与氨基酸在82L+145R/83R+145R位置定义的表位249一致。携带A*24:02的血小板难治性患者G表现出对除A24以外的所有Bw4珠的反应性,与144Q定义的表位423一致。对于携带B*27:05的异基因HSCT患者F,除B*27:005外,所有Bw4珠都存在反应性,反应性由aa 77N+81A+82L定义。 Saima Azhar Salim,Louise Walsh,Geraldine Donnelly,David Keegan,Joseph Kelly,Mary KeoganH&amp;爱尔兰都柏林博蒙特医院I实验室在移植前组织相容性检查期间进行流式细胞术交叉配型(FXM)。假阳性结果可能会阻止潜在的接受者接受合适的移植。许多实验室使用使用双激光流式细胞仪建立的三色FXM测定法,其中包括藻红蛋白(PE)、异硫氰酸荧光素(FITC)和周青素叶绿素蛋白(PerCP)。PE和FITC之间存在显著的光谱重叠,可能导致高背景荧光和可能的假阳性B细胞FXM。本研究旨在确定最佳荧光染料组合,以最大限度地减少光谱重叠,并最大限度地与现有方法保持一致,目前已有临床验证的截止值。选用荧光染料亮紫(BV421)和别藻蓝蛋白(APC)分别标记CD3+T细胞和CD19+B细胞。用荧光染料CD3BV421、CD19APC和抗人IgG-FITC进行34个FXM(共63个T细胞样品和61个B细胞FXM样品)。用荧光染料CD3PerCP、CD19APC和抗人IgG FITC进行14个FXM(总共35个T细胞和B细胞FXM样品)。在Becton Dickenson(BD)FACSLyric上进行FXM测定™ 以及与标准FXM测定相比较的结果。与标准FXM相比,对于CD3BV421-CD19APC FXM,观察到样品T细胞比率和B细胞比率的统计学显著差异(p<0.005)。我们的初步数据表明,CD3PerCP-CD19APC的T细胞和B细胞比率没有显著差异(p>0.05)。数据表明,用CD19APC取代CD19PE可以最大限度地减少光谱重叠,降低FXM结果假阳性的风险,而不会显著改变T细胞和B细胞比率。Ana Bultitude1、Anthony Poles2、Sue Jordan1、AntAnthony Calvert2、Deborah Sage11NHS血液与移植,英国Tooting;2NHS血液与移植,菲尔顿,UKHNA-3是一种双等位基因抗原,-3a/-3b,95%的英国人群编码至少一个HNA-3a等位基因。HNA-3在多种细胞类型上表达,包括淋巴细胞和肾内皮细胞。因此,可以在纯合个体中产生针对非自身变体的HNA-3特异性抗体与肾移植后的排斥反应有关。在此,我们报告了一例肾移植受者显示出阻止移植的抗HNA-3a抗体的病例。2022年8月,患者获得了一个2,1不匹配等级的无私肾脏,产生了阴性的虚拟交叉配型结果。然而,最终的湿实验室流式细胞仪交叉配型(FCXM)结果显示,无法解释的强阳性T和B细胞结果为阴性。该患者自首次转诊以来没有移植史,也没有可检测的HLA抗体。对患者和供体进行HNA基因分型,分别测定HNA-3b3b和HNA-3a3a基因型。对患者血清进行抗HNA抗体筛选,确认存在HNA-3a特异性抗体。该患者被从虚拟交叉配型计划中除名,因为他们不再符合资格标准,但仍在已故捐赠者等待名单上。另外9个死亡的供肾被接受并交叉配型,均为T细胞和B细胞阳性。2023年3月,患者接受了一个死亡的供体肾脏,该肾脏产生了T和B细胞阴性的FCXM。供体的基因分型为HNA-3b3b。患者继续表现良好,迄今为止未检测到抗体介导的排斥反应。Hawzhin Jabar,Luke FosterNHS血液与移植,伯明翰,英国准确的HLA分型对于促进安全的实体器官移植至关重要。在英国,H&amp;I实验室需要满足NHSBT-OTDT设定的最低分型要求,其中包括报告结果,以便评估特定供体-受体组合中的任何供体特异性抗体(DSA)。不这样做可能会导致不适当的分配或增加移植排斥反应的风险。在这里,我们报道了在一名已故实体器官捐献者中使用LinkSeq实时PCR(One Lambda)进行HLA分型的DRB1*14:15等位基因的鉴定。通常,在最低分型要求下,不需要向第二字段报告HLA-DRB1*14,然而,有趣的是,HLA-DRB1x14:15不编码DR14抗原,而是编码DR8抗原。因此,在这种情况下,需要向第二个字段报告HLA-DRB1*14,以便准确分配和评估任何潜在的DSA。尽管HLA-DRB1*14:15在当地很罕见,但在CIWD 3.0.0目录中,它在欧洲人群中被列为有充分记录的,在亚洲/太平洋岛屿和美洲原住民中很常见。 NanoTYPE(Omixon)可用于多达24个样本的批次或单个样本,有可能用于已故供体HLA分型。本研究的目的是在两个NHSBT H&amp;I实验室;Colindale Service Development和Tooting,使用了同一批96个样本,这些样本之前已经通过当前的快速HLA分型或NGS方法进行了分型。所有NanoTYPE试剂、R9.4流动池和MinION mk1B装置均由Omixon和Oxford Nanopore Technologies提供用于本研究。使用单样本方案设置了八个样本。剩余的样品被分为不同大小的多个批次。对于HLA-A、-B、-C、DRB4、DRB5和DPA1,两个位点之间的第二视野分辨率与先前的分型数据100%一致。Tooting的10/96(10%)样本和Colindale的9/96(9%)样本中的HLA-DRB1、DRB3、DQA1、DQB1或DPB1基因座与先前的分型数据不一致,这归因于等位基因失衡、某些基因型如DQB1*03的已知低扩增、等位基因缺失和潜在新等位基因的检测。在对测序数据进行手动审查后,Colindale的95/96个样本和Tooting的93/96个样本与之前的结果一致。我们的研究已经证明,NanoTYPE检测方法使用简单,能够实现高分辨率HLA分型。Emma Ross1,Sylvia McConnell1,Richard Battle1,Nicole Pridde2,David Turner11H&amp;英国爱丁堡SNBTS I部门;2Donor Medicine,英国爱丁堡SNBTS;I实验室负责在苏格兰提供HLA选择的血小板。2022年,96名患者获得了总计2317个血小板单位的支持。服务的关键性能指标(KPI)是&gt;60%的分配供体血小板应为与患者匹配的A级或B1级(A级=第一字段没有HLA-A或HLA-B错配;B1=只有一个HLA-A或HLA B错配)。在2016年1月至2022年9月期间,这一目标平均为57.7%(范围为46%-72%),总体呈下降趋势。相比之下,作为HLA选择的个体供体血小板数量从2016年的1288个单位增加到2022年的2317个(+55.5%);我曾与SNBTS捐献者服务中心(DS)合作,以增加最佳匹配捐献者血小板的可用性。利用质量改进方法、商业分析工具和专家捐赠者沟通,我们专注于HLA-a和-B纯合子捐赠者的国家识别和(重新)参与,这些捐赠者占小组的3.5%,但有可能为约68%的患者提供a级匹配(2023年1月至6月)。自项目启动以来,我们记录到在2022年10月(49.5%)至2023年5月(72.4%)期间,A和B1血小板分配的百分比呈上升趋势;自2023年1月以来,已持续实现60%。与DS的合作增强了供体群体,改善了获得HLA最佳匹配血小板的“现成”途径。Felicity May1、Sian Griffin2、Madhvi Menon3、Tracy Hussell3、Tracey Rees11威尔士血液服务局威尔士移植和免疫遗传学实验室,英国庞蒂克伦;2英国加的夫威尔士大学医院肾脏病与移植科;3英国曼彻斯特大学免疫学、感染免疫和呼吸医学系人类白细胞抗原不相容(HLAi)移植仍然是高度敏感患者的重要选择。跨越HLA屏障与抗体介导的排斥反应(AMR)和移植物衰竭的风险增加有关。去敏、诱导和维持免疫抑制旨在将这种风险降至最低。在最佳治疗方案上没有达成一致意见,患者的反应也各不相同。在这项单中心回顾性研究中,我们评估了27名对利妥昔单抗和双重过滤血浆置换周期不敏感的HLAi移植受者的临床队列中的排斥反应发生率和移植物存活率。已确定的风险因素与AMR发病率之间的相关性较差,这表明存在其他目前未知的因素。HLA抗体反应和AMR受到支持抗体产生细胞的产生和存活的复杂细胞因子网络的影响。我们进行了一项文献综述,并根据证据质量和体液记忆反应的接近程度对459种血清蛋白分析物(细胞因子、趋化因子、激素等)进行了分级。我们使用定制的多重Luminex测定法评估了来自我们临床队列的纵向样本中40种最高等级分析物的血清浓度。由于样本数量少和分析物图谱的异质性,我们无法建立与患者临床结果的明确关联。 然而,我们观察到影响生发中心、B细胞和浆细胞的关键信号分子发生了显著和持续的变化。我们注意到,我们小组的大多数人在脱敏/免疫抑制后的水平有所下降,但一些关键支持信号的水平增加或不变,一些免疫调节因子受到抑制。Agnieszka Ojrzynska1、Kylara Hassall1、Katie Butler1、Graham Shirling1、Sharon Vivers2、Raymond Fernando 11英国伦敦皇家免费医院实体器官小组;2英国伦敦的Anthony Nolan组织相容性实验室越来越多接受Daratumumab(Dara)治疗的多发性骨髓瘤患者被列入肾移植名单。Dara是一种人单克隆抗CD38 IgG抗体,它与包括淋巴细胞在内的许多血细胞上表达的CD38分子结合,从而干扰移植前异基因淋巴细胞交叉配型和输血前兼容性测试。这可以模拟供体特异性抗体(DSA)的存在,并导致假阳性交叉配型结果。本研究的目的是研究用流式细胞术交叉配型(FCXM)减轻Dara干扰的方法。我们的中心已经移植了四名接受达拉治疗的患者。其中3例HLA抗体阴性,1例HLA抗体阳性但HLA DSA阴性。使用我们的标准程序以及使用二硫苏糖醇(DTT)(0.05–0.1M)预孵育的供体细胞的修订方案,进行了六个第三方外周血FCXM和一个脾细胞的回顾性移植FCXM,DTT可以切割CD38分子的稳定二硫键。使用我们的标准程序的所有第三方交叉配型均为T细胞阳性,其中两人也是B细胞阳性。对脾脏细胞进行的回顾性交叉配型为T细胞和B细胞阳性。用DTT进行的所有交叉配型均为T细胞和B细胞阴性。我们的结果表明,当Dara治疗患者的血清用于FCXM时,DTT消除了观察到的假阳性。HLA阳性对照结果未受影响,表明HLA分子不受DTT治疗的影响。因此,DTT可用于减轻FCXM中DARA介导的假阳性结果。Daniel Eggleston,英国曼彻斯特Helena LeeManchester皇家医院造血祖细胞移植(HPCT)已被公认为恶性和非恶性血液病的治疗方法。然而,移植后的患者监测仍然至关重要。移植后供体嵌合试验可以确定并发症,包括复发或移植失败。复发是经HPCT诊断为骨髓增生异常综合征(MDS)或高危白血病的患者特别关注的领域。大多数实验室通过PCR短串联重复序列(PCR-STR)进行嵌合监测。该技术利用DNA短重复序列的差异和毛细管电泳来确定供体嵌合的百分比。然而,其他方法正在出现,它们提供了更高的灵敏度和更多的信息标记。一种这样的技术是数字PCR(dPCR)。在dPCR过程中,PCR反应混合物被分配成单独的液滴。每种方法都作为一种单独的检测方法,存在表明供体或受体DNA的反应,提高了技术灵敏度。使用dPCR,我们调查了11名被诊断为MDS或髓系白血病的患者,这些患者的供体嵌合百分比通过PCR-STR达到100%,但在随后的样本中有所下降。我们使用JETA DigitalTRACE测试了我们的团队™ 在使用三种标记物减少供体嵌合之前的两个或多个时间点使用Qiagen-QIAcity机器的dPCR技术。在我们的11名患者中,有10名患者的供体嵌合减少可通过dPCR进行鉴定,而PCR-STR的分析表明样本是100%的供体嵌合。这提高了早期检测供体嵌合变化和改进随访过程的可能性。因此,通过dPCR的嵌合体是PCR-STR的可行替代品,并且可以在HPCT后更早地识别事件。 HLA分型结果在血液和口腔来源的DNA之间是一致的,包括在患者样本中观察到的四个新序列,证实这些是种系突变。接下来,我们分析了9个含有≥3个HLA-A、-B、-C、-E、-F和-G的文库中恶性(n=406)和非恶性(n=46)疾病缓解患者的血液DNA序列。Phred评分(33.60 vs.33.56)或聚类多样性(0.05 vs.0.06)没有观察到显著差异。总体而言,同一个体的血液和口腔DNA样本的最终HLA分型结果没有差异,恶性和非恶性患者样本的序列质量也没有差异。所有样本类型,如果在病情缓解时采集,都是HLA分型和鉴定新序列变异的可靠来源。Ravneet Kaur Bola、Madalina Pinzaru、Marlowe Macadangang、Ufot Udofia、Sandra Frater、Franco Tavarozzi Anthony Nolan英国伦敦组织相容性实验室口腔上皮细胞用于种系HLA检测,特别适用于怀疑杂合性缺失(LOH)的患者。Anthony Nolan(AN)的患者口腔采集方法使用CytoSoft®刷,然而,我们一直在寻找优化流程的方法,并意识到刷的设计可能会增加一些患者血液污染的机会,从而可能混淆LOH病例的HLA分型。AN在我们的注册捐赠者招募中使用了一种更温和的采集工具FLOQSwabs®,因此,与皇家马斯登医院合作,设计了一项试验,为我们的患者确定最佳的口腔采集方法。使用三种情况对每个同意的患者进行拭子:(1)FLOQSwabs®,脸颊;(2) FLOQSwabs®,左侧排水沟;(3) CytoSoft®刷子,右侧水槽。对54名患者的样本进行了处理。从每个拭子中提取DNA,并按照预先确定的序列为每个患者选择一次提取用于下一代测序(NGS),每个场景产生等量的NGS数据。评估着眼于DNA提取和NGS失败率,以及产生的DNA量。三种情况下DNA提取失败率分别为9.3%、17.6%和29.9%。平均而言,FLOQSwabs®的DNA浓度至少是CytoSoft®刷子的两倍。由于NGS的稳健性,为NGS选择的所有提取都被成功地分型。总之,从脸颊擦拭FLOQSwabs®是首选的采集方法。这些DNA提取失败的次数最少,同时为患者提供了更柔软、更温和的拭子体验。Claire Lenehan、James Kelleher、David Keegan、Mary Keogan、爱尔兰都柏林Khairin KhalibNHISSOT Beaumont医院和人类白细胞抗原抗体是移植的并发症。Luminex®技术的引入使这些抗体能够以高灵敏度进行精确表征。由于检测到针对变性HLA分子(dHLA)的生物学无关抗体,这些抗体的临床意义仍然存在争议。我们的目的是确定通过Luminex单抗原珠粒分析检测到的供体特异性抗体(DSA)与流动交叉匹配(FXCM)结果的相关性,使用先前发表的方案,并使用临床验证的截止值。研究了170名非经典致敏患者的HLA抗体导致阳性交叉配型的能力。完成了49个针对不相容供体细胞的FCXM。56%的T细胞和61%的B细胞FCXM阳性。HLA-A和-B抗体的FCXM结果阳性率显著较高(p=0.013)。MFI值是FCXM结果的较差预测因素。相反,MFI值为5000至10000的DSA的FCXM阴性结果率与MFI值&lt;对于DSA MFI&gt;无论MFI强度如何,FCXM结果的显著比例都是阳性的,这表明非经典致敏患者的DSA并不局限于对dHLA的反应性。FXCM促进风险评估。根据这项研究中的数据,当DSA MFI低于10000时,我们现在为活体捐赠者或高敏感患者提供FCXM。Kay Poulton,1,2 Madeleine Harris1,Andrew Canterbury2,Marie Hampson1,Judith Worthington1,Marcus Russell-Lowe11英国牛津路曼彻斯特皇家医院,2MC Diagnostics Limited,St Asaph,Wales,3曼彻斯特大学,曼彻斯特,UKSingle抗原珠分析已经彻底改变了HLA特异性抗体的鉴定和定义。在引入后的20年里,它们使虚拟交叉配型得以广泛使用,并几乎消除了由于HLA不兼容而引起的超急性排斥反应。 Eveline Little、Nicola Brosnan、Jade Kally、Adrian Silk、Lisa Walsh、Franco Tavarozzi Anthony Nolan组织相容性实验室,英国伦敦。Anthony Nolan组织相容性实验室利用GenDx NGSgo®NGS单引物实现HLA-A、-B、-C、-DRB1、-DRB3、4、5、-DQB1、-DQA1、-DPA1和-DPB1的高等位基因分辨率分型。在这项工作之前,所有接受HLA分型的样本都测试了11个基因座,但要求的基因座数量可能有所不同。与多重引物相比,使用单一引物使我们能够实施灵活的方法来提供量身定制的服务。这项工作的目的是开发一种对需要在同一次运行中进行各种测试组合的样本进行打字的过程。需要对我们的实验室信息管理系统(LIMS)进行修改,以便在整个NGS测试过程中进行重大更改。由于该项目以及我们正在进行的灵活性改进过程,我们还实现了在同一次运行中重复PCR失败的能力,使该过程能够毫不拖延地进行,并合并多个NGS库。由于我们不再进行不必要的测试,这通过增加Illumina®流动池的容量节省了成本,每次运行允许更多的样本,有可能缩短我们的周转时间,并满足我们的客户需求。总之,单引物已被证明在运行时间没有明显增加的情况下具有极大的灵活性,使用LIMS端到端地简化和跟踪过程。Victoria Wood1,2,Brendan Clark1,Eric Hewitt2,Sunil Daga11利兹教学医院NHS信托,英国利兹;2英国利兹大学肾移植免疫学服务的核心是在潜在捐赠者的背景下检测HLA抗体。HLA抗体的存在对移植的机会不利,并且作为供体特异性抗体(DSA),对移植结果不利。随着越来越多的高度敏感患者需要跨越HLA抗体屏障进行肾移植,对抗体功能特征的了解增加可能会导致更明智的供体选择。抗体-抗原相互作用的亲和力提供了对抗体诱导抗体介导的变化的能力的深入了解,例如细胞内信号传导,导致组织重塑和移植物损伤。嗜潮剂降低了蛋白质的稳定性,并且以前已经在ELISA方案中用于通过一种称为嗜潮破坏的过程来估计抗体-抗原相互作用的亲和力。将我们的标准One Lambda LABScreen单抗原珠(SAB)方案修改为包括离液剂,已经证明了将离液破坏应用于固相测定的技术可行性,并且可以自信地解释这种操作。建立了离液剂的最大摩尔浓度,该最大摩尔浓度不损害珠表面抗原的完整性-1摩尔。与仅结合天然HLA I类的HLA-A、-B、-C单克隆相比,HCA2单克隆结合天然和变性HLA I类,使用流式细胞术分析验证了这一点。使用具有已知HLA抗体谱的血清进行的初步测试显示了抗体特异性的结合和解离模式。这种新方法代表了一种在NHS实验室环境中测试HLA抗体亲和力的潜在可行方法。Steven Jervis1,2先生,Antony Payton3博士,Marcus Lowe1,2博士,Altug Didikoglu4博士,Arpana Verma2教授,Kay Poult1,21Manchester移植实验室,英国牛津路;2英国曼彻斯特大学健康科学学院人口健康、卫生服务研究和初级保健处生物、医学和健康学院;3信息学、影像学和;英国曼彻斯特大学生物、医学和健康学院健康科学学院数据科学;4英国曼彻斯特大学生物科学学院神经科学系生物计时中心的研究假设,预先存在的遗传特征和特定的环境触发因素的结合决定了嗜睡症的发作。最具影响力的遗传风险因素是主要组织相容性复合体(MHC)上编码的人类白细胞抗原(HLA)DQB1*06:02的存在,然而,并非所有发作性睡病病例中都普遍存在HLA-DQB1*06:02。MHC之外最尖锐的遗传风险因素主要位于与免疫系统相关的基因中。除了发作性睡病的传统症状外,合并症还可能因一批患者抱怨认知功能障碍,特别是记忆和注意力功能障碍而有所不同。 这些自我报告没有得到一致的科学证据的证实,而有重要证据概述了普通人群认知能力变异的基因贡献。在这项研究中,我们对1558名非病理性老年志愿者进行了长达24年的认知功能变化跟踪,对其进行了靶向非MHC发作性睡病相关单核苷酸多态性(SNPs)的估算。具体而言,我们研究了13个先前记录的发作性睡病相关SNPs,其比值比大于或等于1.00,次要等位基因频率大于0.05。我们观察到rs306336、rs4290173和rs2834168与长期记忆的快速下降之间存在关联。同样,我们观察到rs10995245对长期记忆丧失的下降具有保护作用。这项研究表明,发作性睡病患者队列报告的认知问题可能是由于遗传易感性,并支持与发作性睡症相关的合并症的变化。Raji Patel、Aliyye Karasu、Liezelle Pagala、Jyoti Bhatt、Edgar Correa、Gloria Adeyemo、Maame Esi Yeboah、Abigail Sarkodie、Andrew Joahill、Suzette Cavanna、Carla Rosser、Colin BrownNHS Blood and Transplant Colindale,伦敦,UK英国骨髓登记处(BBMR)利用Oragene®唾液采集试剂盒,通过下一代测序(NGS)促进潜在捐献者的HLA扩展分型。唾液是一种很好的DNA替代来源,因为它的采集是非侵入性的,可以自我采集,并且在环境温度下稳定。2023年1月,Qiagen EZ2机器人被引入我们的实验室,为临床样本提供DNA提取。然而,Qiagen无法提供使用该仪器从唾液样本中提取DNA的有效方案。在这项研究中,我们评估了Qiagen EZ2组织试剂盒使用Qiagen EZ2 Connect机器人从唾液样本中获取DNA的能力。使用我们开发的方案,成功地从10个唾液样本中提取了DNA,平均DNA浓度为33纳克/μl,260/280比例为1.72。DNA的质量与使用经验证的Roche Magnapure方法从唾液样本中提取的DNA相当。唾液DNA用于高分辨率HLA分型的适用性使用One Lambda FASTplex试剂盒进行评估。使用TypeStream Visual(TSV)NGS分析软件,我们获得了334691的平均映射读数,结果符合我们记录的所有质量指标的验收标准。总之,我们已经证明,Qiagen EZ2 Connect机器人可以成功地从唾液样本中提取DNA,其血液质量相当。该过程的验证使我们的部门避免了BBMR扩展HLA分型请求的不必要延迟,从而支持患者护理。Sebastian Fernando、Jennifer Lord、Nicola Martin、Alison Logan、Kay Poulton曼彻斯特大学NHS基金会信托基金会,英国曼彻斯特我们目前支持疾病关联测试的常规HLA分型方法是LABType™ SSO(One Lambda)。评估了另一种HLA分型方法EUROArray(EUROIMMIN)的一致性和有效性。该系统结合了聚合酶链式反应和微阵列技术。用荧光染料标记的扩增的靶DNA使用BIOCHIP技术与互补的DNA探针杂交。荧光信号使用EUROIMINM微阵列扫描仪和EUROArrayScan软件自动评估。常规和外部熟练方案外周血和DNA样本先前通过LABType进行HLA分型™ SSO使用适当的EUROArray测定法进行测试。46个样本用HLA-B27直接测定法进行了测试,41个样本用HLA B*57:01直接测定法和42个样本用HLA-DQ2/DQ8-h直接测定法。选择了一系列相关的HLA-B和HLA-DQ等位基因,以确保EUROArray系统能够区分感兴趣的等位基因。与LABType相比,我们发现100%一致™ 所有测试的SSO结果。与LABType相比,工作流程简单明了,节省了时间™ SSO,然而EUROArray测定的成本更高。这项研究表明,EUROArray直接检测HLA-B27、-B57和-DQ2/8是支持疾病关联检测的有效替代方法。EuroArray系统的优点包括无需额外的DNA分离,包含大量集成控制,可实现结果的高可靠性,以及完全自动化的标准化评估和结果生成。这种检测的局限性包括在静脉穿刺后14天内使用血液样本,尽管可以冷冻样本以缓解这种情况。 一项回顾性临床审计旨在审查剑桥已故捐赠者肾移植等待名单(TWL)上患者的影响,并确定是否达到了2019年KAS的目标。移植数据来自NHSBT-ODT,实施前(n=298)和实施后(n=303)24个月。审查数据时考虑到了新冠肺炎大流行的影响。结果显示,计算出的反应频率(cRF)≥85%的高度敏感患者的平均等待时间(天)减少。根据模拟数据,已经实现了捐赠者/接受者指数匹配,大多数“低风险”捐赠者被分配给“低风险的”接受者,“高风险”捐赠者/接受者也是如此。根据A级(cRF 100%/等待时间&gt;7年/匹配性得分为10),难以匹配的患者优先考虑了不同的供体/受体年龄匹配。与模拟数据相反,在国家分配发生变化后,剑桥大学没有观察到心脏死亡(DCD)移植后的捐赠减少(前174例,后192例)。总之,数据表明剑桥TWL高敏感患者的平均等待时间减少,并表明2019年KAS的目标已经实现。Michelle Carr1、Shelley Harris1、Judith Worthington1、Alex Woywodt2、Kay Poult11英国曼彻斯特移植实验室MFT;2英国普雷斯顿兰卡希尔教学医院NHS基金会信托2020年1月,一名24岁的男性因反流性肾病继发慢性肾衰竭,接受了HLA完全匹配的DBD捐赠者的第三次肾移植。2020年3月,他的肾功能下降,同时肌酐和蛋白尿升高。在患者的整个移植史过程中,他克莫司的水平是可变的,怀疑存在不依从性。LABScreen对非HLA抗体的回顾性分析™ 自身抗体试剂盒鉴定出患者产生了谷胱甘肽S-转移酶Theta 1(GSTT1)抗体。第二次移植后,他克莫司水平低于目标范围(&lt;2.5μg/L),随后首次出现GSTT1抗体(6000MFI=95%百分位)。这与活检显示慢性移植肾小球病变相吻合。第三次移植后,他克莫司的水平仍不稳定,有些水平高于或低于目标范围。他的移植功能在2023年春季进一步恶化。此时的活检显示交界性T细胞介导的排斥反应伴有中度间质纤维化和肾小管萎缩。GSTT1抗体水平迅速增加,峰值为11000 MFI(>95%百分位)。我们提出,由不粘附事件引发的免疫介导的炎症过程会导致细胞损伤。作为对这种损伤的反应,细胞内成分如GSTT1将被释放,从而实现识别和随后的免疫反应,从而产生GSTT1抗体。我们建议,GSTT1抗体的检测应被视为肾毒性和移植肾持续创伤的标志。阿德里安·汉德利(Adrian Handley)、大卫·布里格斯(David Briggs)、克莱尔·科林斯(Clare CollinsNHS)英国伯明翰血液与移植中心(Blood and Transplant)所有测量都带有不确定性因素,只有在了解了不确定性程度时才真正有用。使用来自Luminex HLA抗体测试的半定量数据对于进行心胸移植兼容性评估至关重要;然而,人们并不了解这种不确定性的程度。BSHI/BTS CTAG指南根据Luminex单抗原珠HLA抗体测定中产生的MFI值设定风险水平。此外,ISO15189:2012规定,实验室应计算所有认证测试的测量不确定度(MU)。在这项研究中,我们使用了简单的统计工具来确定MU的程度和一些促成因素。这些使我们能够迅速评估随后所做的改变的有效性。我们发现了个体检测人员获得的结果之间的显著差异,并能够通过我们的干预措施来减少这种差异。Reed等人。(Am J Transplant.2013;13(7):1859-1870)表明MU的%CV在20%-62%的范围内。我们的研究发现,这一比例从20000 MFI时的8%到1000 MFI(阳性阈值)时的18%不等。我们开发的工具允许对检测性能进行持续监测,并为未来的质量举措提供基础,有助于可持续、循证的质量改进。MU计算是一种有效的质量保证和改进工具,并且易于实施。通过结合本研究中使用的工具,可以确定变化的水平和来源,并且可以很容易地评估过程中任何变化的影响的有效性。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Oral Abstracts

Alison Cleaton, Emma Burrows, Kimberley Robinson, Michael Richardson, Deborah Pritchard, Tracey Rees

Welsh Blood Service, Ely Valley Road, Talbot Green, UK

Regular HLA antibody testing is undertaken for patients awaiting renal transplantation, using LABScreen™ HLA antibody assays. During the COVID-19 pandemic, we observed unexplained changes to some HLA antibody profiles. Investigation revealed that several patients had COVID-19 prior to the changes, therefore, a review of all patients on the transplant waiting list with known COVID-19 infection was undertaken. Sixty-six out of two hundred thirty-seven patients on the transplant waiting list had COVID-19 (March 2020–July 2022). The HLA antibody results from samples prior to and following COVID-19 infection were analysed for changes in existing HLA antibody levels (increased Luminex Median Fluorescent Intensity (MFI) values), or expanded antibody profiles (increased cRF). Fifty-two (78.8%) patients had no detectable change in cRF or MFI; five (7.6%) had changes in MFI (but no change in cRF); nine (13.6%) had changes in MFI and cRF. Two out of nine patients had no recorded prior sensitisation event; four had a previous transplant, four blood transfusions, four pregnancy; and three multiple sensitising events. All nine patients had sustained cRF changes in subsequent samples (follow up to December 2022). Three out of nine patients were consequently identified as having altered immunosuppression due to the COVID-19 infection; these patients had a 20%–76% rise in cRF and now all have a cRF 98%–100%. While the majority of patients awaiting kidney, transplantation had no change to their HLA antibody profile following COVID-19 infection, nine patients had an increase in cRF, which has not been transient. Reduction or withdrawal of immunosuppression to aid recovery from COVID-19 was identified as the cause for three patients.

Adrienne Seitz, Clive Carter, Brendan Clark, Richard Baker

Leeds Teaching Hospitals NHS Trust, Leeds, UK

The level of pre-transplant immune risk is assessed through measuring serum IgG HLA antibodies which can be produced by long lived plasma cells and memory B-cells. Memory B-cells can circulate without producing antibodies, therefore their contribution to the antibody pool may not be fully appreciated. We describe an in vitro method for improving the assessment of pretransplant risk through the non-specific stimulation of peripheral memory B-cells. Peripheral blood mononuclear cells from three unsensitised volunteers and six sensitised patients were cultured for 9 days with the toll-like receptor agonist R848 and interleukin-2. Cell culture supernatant was tested for IgG HLA antibodies using single antigen beads. This was compared with a matched serum sample. Resting Day-0 and stimulated Day-9 B-cell phenotypes were assessed using flow cytometry, confirming the switch to antibody secreting (CD24-CD38hi), class-switched memory (CD27+IgD-) and plasma (CD38+CD138+) cells. HLA Class I and II antibodies were found in the cell supernatant, and 65% were present in the matched serum sample. When the supernatant demonstrated additional HLA antibodies, these could be attributed to a previous transplant, or had been present in the patient's historic serum profile. We demonstrate a method that can uncover peripheral memory using technology accessible to most H&I laboratories. This assay could be useful when assessing live donor pairs where the donor may repeat mismatches associated with pregnancy, and in re-grafts, prior to removal of ‘other unacceptable antigens’. Finally, it could be considered alongside delisting strategies in the context of novel peri-transplant agents.

Sophie Chambers, Robert Whittle, John Goodwin, Tim Key

NHS Blood and Transplant, Barnsley, UK

Differences in amino acids (aa) at positions 76–83 of exon II of HLA-B and a subset of HLA-A primarily account for the highly immunogenic public epitopes Bw4 and Bw6. Bw4-specific antibodies are commonplace in alloimmunised Bw6 homozygotes, whilst Bw6-specific antibodies are encountered in Bw4 homozygotes. As variation exists within the Bw4 complex it has been reported previously that in a proportion of Bw6 homozygous individuals with Bw4 specific antibodies, reactivity can be restricted to HLA Bw4 subtype epitopes. We report three individuals with a Bw4 phenotype who demonstrate Luminex Single Antigen Bead determined alloantibodies to Bw4 epitopes distinct from their own. For renal patient L bearing B13, reactivity was present to all Bw4-positive beads other than B13. Reactivity was consistent with Epitope 249 defined by aa at positions 82L + 145R/ 83R + 145R. Platelet refractory patient G bearing A*24:02 demonstrated reactivity to all Bw4 beads other than A24, consistent with Epitope 423 defined by 144Q. For allogeneic-HSCT patient F bearing B*27:05, reactivity was present to all Bw4 beads except B*27:05 with reactivity defined by aa 77N + 81A + 82L. Our observations highlight that Bw4 subtype epitope profiles should not be overlooked in Bw4-positive individuals, that single amino acid differences in the Bw4 complex appear sufficient to generate alloantibodies and differences outside aa positions 76–83 may contribute to antibody binding. Bw4 subtype antibodies can impact organ allocation in the renal transplant setting, restrict appropriate platelet support for immunological refractoriness and complicate donor selection in HLA-mismatched allogeneic-HSCT.

Ryan Stevens, Felicity May

Welsh Transplantation and Immunogenetics Laboratory, Pontyclun, UK

The Welsh Transplantation and Immunogenetics Laboratory maintain a register of local patients active on the national renal/pancreas waiting list. Previously, data pulled from local IT systems was distributed monthly to service users in PDF format and a printed copy was held locally. This system had multiple drawbacks, including lack of ability to easily interrogate/update the data. In collaboration with the NHS Wales Microsoft 365 Centre of Excellence, a digital solution was created using Power BI. This allows up-to-date data to be instantly available, which gives users a more accurate overview to support decision making, and eliminates the requirement for PDF/paper distribution. The Dashboard can only be accessed by approved users, and access can be restricted to specific datasets (e.g., dialysis unit staff can only view patients at that unit). The Dashboard consists of two pages. A ‘Statistics’ page graphically displays the Register by organ type, blood group, dialysis unit, and so forth. The ‘demographics’ page tabulates data within the Register, which can be ordered or filtered in a variety of ways to aid in patient selection for donor offers. Each patient also has a colour coded ‘Sample Status’ to highlight when routine antibody screening samples are due. Comments and/or attachments can be added to each patient entry. Since successful go live December 2022, the dashboard has been praised by staff and service users for ease of access of data to assist delivery of safe and efficient patient care, as well as reduced incidence of overdue samples.

Kirsty Clark, Jane Matthews, Claire Romaines, Ruth Chisman, Arash Akbarzad-Yousefi

NHSBT Newcastle, Newcastle upon Tyne, UK

Despite advancements in organ allocation and immunosuppression, cardiothoracic transplantation continues to have the lowest 5-year survival when compared to all other forms of solid organ transplantation. A key factor in allograft loss is the formation of de novo Donor Specific HLA Antibodies (dnDSA). Recently, research has been driven away from the conventional method of defining mismatch at the antigenic level and has instead focused on HLA eplet mismatches. This study aimed to evaluate the role of eplet mismatching within our local cardiothoracic patient cohort. A retrospective analysis of 2 years of cardiothoracic transplant data was performed. The antibody data from all eligible patients were reanalysed to investigate the role of eplet mismatch load and to identify possible high risk eplets. This study found no evidence to directly support the theory of eplet load mismatches. However, four previously identified high risk eplets (McCaughan et al., American Journal of Transplantation 2018) namely, 55PP, 52LR, 55R and 75S were confirmed within our patient cohort. Findings from this single centre study provide potential evidence of high-risk HLA eplet mismatches; all of which are present within the HLA-DQ locus, indicating possible high immunogenicity for mismatches at this locus. Particular attention to minimise HLA-DQ mismatches may reduce the incidence of dnDSA and subsequently, allograft loss. Whilst avoiding mismatches in cardiothoracic transplantation is not always possible, further understanding of this area could lead to refinements in post-transplant monitoring and immunosuppression regimens.

Charlotte A. Cambridge1, Jonathan A.M. Lucas1, Xenia Georgiou1, Gabriel J. Benitez1, Neema P. Mayor1,2, Steven G.E. Marsh1,2

1Anthony Nolan Research Institute, London, UK; 2UCL Cancer Institute, Royal Free Hospital, UK

Submission of novel sequences to the IPD-IMGT/HLA Database from patients with haematological malignancies is not permitted, unless confirmed in the germline. To investigate if sequencing blood and buccal DNA yields different results, we typed 47 patients in remission from malignant disease and 58 healthy donors for HLA-A, -B, -C and -E using PacBio SMRT sequencing. Higher Phred scores (33.2 vs. 32.7, p < 0.05) and lower cluster diversity (0.06 vs. 0.11, p < 0.0001) were observed in sequences from patient buccal DNA versus blood, indicating sequences of higher quality with fewer background errors. No differences were observed in donor blood or buccal material. Blood DNA generated better HLA typing results that were automatically accepted (80.7% vs. 79.8%) with less allele dropout (2.0% vs. 7.2%). HLA typing results were concordant between blood and buccal derived DNA, including four novel sequences observed in patient samples, confirming these as germline mutations. Next, we analysed sequences from blood DNA for patients in remission from malignant (n = 406) versus non-malignant (n = 46) disease across nine libraries containing ≥3 of HLA-A, -B, -C, -E, -F and -G. No significant differences in Phred score (33.60 vs. 33.56) or cluster diversity (0.05 vs. 0.06) were observed. Overall, there were no differences in final HLA typing results for blood and buccal DNA samples for the same individual, and no difference in sequence quality between malignant and non-malignant patient samples. All sample types, if taken at the point of remission, are reliable sources for HLA typing and identification of novel sequence variation.

Ravneet Kaur Bola, Madalina Pinzaru, Marlowe Macadangdang, Ufot Udoffia, Sandra Frater, Franco Tavarozzi

Anthony Nolan Histocompatibility Laboratories, London, UK

Buccal epithelial cells are used for germline HLA testing, particularly useful in patients where loss of heterozygosity (LOH) is suspected. Anthony Nolan's (AN) patient buccal collection method utilises CytoSoft® brushes, however, we are always looking for ways to optimise our processes and realise that the design of the brushes may lead to an increased chance of blood contamination with some patients, potentially confounding HLA typing in LOH cases.

AN uses a gentler collection tool for our registry donor recruitment, FLOQSwabs®, therefore a trial was devised to identify the best buccal collection method for our patients, collaborating with the Royal Marsden Hospital. Each consented patient was swabbed using three scenarios: (1) FLOQSwabs®, cheek; (2) FLOQSwabs®, left gutter; (3) CytoSoft® brushes, right gutter. Samples from 54 patients were processed. DNA was extracted from each swab and one extraction per patient was selected for Next Generation Sequencing (NGS) in a pre-determined sequence, producing equal amounts of NGS data per scenario. Assessment looked at DNA extraction and NGS failure rates, together with DNA quantity produced.

DNA extraction failure rates were 9.3%, 17.6%, 29.9% for the three scenarios respectively.

On average, the DNA concentration from FLOQSwabs® were at least two times higher than with CytoSoft® brushes. Due to NGS robustness, all extractions selected for NGS were successfully typed. In summary, FLOQSwabs® swabbed from the cheek are the preferred collection method. These produced the lowest number of failed DNA extractions, while providing a softer and gentler swabbing experience for the patient.

Claire Lenehan, James Kelleher, David Keegan, Mary Keogan, Khairin Khalib

NHISSOT Beaumont Hospital, Dublin, Ireland

Antibodies to human leukocyte antigens (HLA) are a complication for transplantation. The introduction of Luminex® technology has allowed for precise characterization of these antibodies with high sensitivity. The clinical significance of these antibodies remains controversial due to the detection of biologically irrelevant antibodies directed against denatured HLA molecules (dHLA). We aimed to determine the correlation of donor specific antibodies (DSA) detected by Luminex single antigen bead assay with flow cross match (FXCM) results, using a previously published protocol, with clinically validated cut-offs.

The capability of HLA antibodies in 170 non-classically sensitised patients to result in a positive crossmatch was investigated. Forty-nine FCXM against incompatible donor cells were completed. Fifty-six percent of T cell and 61% of B cell FCXM were positive. The rate of positive FCXM results was significantly higher for HLA-A and -B antibodies (p = 0.013). MFI value was a poor predictor of FCXM results. Conversely, the rate of negative FCXM results for DSA with MFI values of 5000 to 10,000 was not significantly different to that of MFI values <5000. The rate of positive FCXM was higher for patients with DSA MFI > 10,000. A significant proportion of the FCXM results were positive regardless of MFI strength indicating that DSA in non-classically sensitised patients are not limited to reactivity to dHLA. FXCM facilitates risk assessment. Based on the data in this study, we now offer a FCXM in living donors, or highly sensitised patients when DSA MFI is less than 10,000.

Kay Poulton ,1,2 Madeleine Harris1, Andrew Canterbury2, Marie Hampson1, Judith Worthington1, Marcus Russell-Lowe1

1Manchester Royal Infirmary, Oxford Road, UK, 2MC Diagnostics Limited, St Asaph, Wales, 3University of Manchester, Manchester, UK

Single Antigen Bead assays have revolutionised the identification and definition of HLA-specific antibodies. They have enabled the widespread use of virtual crossmatching and almost eliminated hyperacute rejection due to HLA incompatibility during the 20 years since their introduction. But for highly sensitised patients, there is a low likelihood of finding a compatible donor. The only option for some patients is strategic de-listing of specificities which may impact the lowest immunological risk to a new graft. In this study, 257 serum samples from 82 potential recipients were crossmatched against cells from 84 potential donors. Only results where serum samples have been tested by LABScreen, HISTO SPOT® HLA AB, Complement Dependent Cytotoxicity (CDC) and Flow Cytometry were included in the analysis. The results were analysed to assess the ability of HISTO SPOT® HLA AB to predict the crossmatch result. Of the 136 samples analysed, 17 (12.5%) were CDC positive, and 82 (60.3%) were positive by flow cytometry. Twenty-eight sera tested negative using HISTO SPOT. All of these (100%) were also negative by CDC and 25 (89.3%) were also negative by Flow Cytometry. Three sera which tested negative by HISTO SPOT with a positive flow cytometry crossmatch were CDC negative, each had cumulative MFIs of <3000 against Donor Specific Antigens. In this early study, HISTO SPOT® HLA AB has 100% negative predictive value for CDC crossmatch negativity and 89.3% by Flow Cytometry. It may therefore prove a useful additional tool to inform de-listing strategies used to facilitate transplantation in highly sensitised patients.

David Wimbury

Transplant Laboratory, University Hospitals of Leicester NHS Trust, Leicester, UK

Previous work has shown that variations in temperature, incubation time and operator methodology can have a drastic effect on results in solid phase assays. In HLA antibody screening, differences in overall MFI levels could impact the number of antibodies detected as positive and/or their risk level, thereby potentially affecting transplantation opportunities. Normalisation of MFI in each sample to a baseline could create more consistent results between samples at different time points, reducing the measurement uncertainty. This method normalises overall sample MFIs to a baseline. Positive control samples and three patients with extensive sample histories were tested with this methodology. CVs of MFIs before and after normalisation were analysed with paired t-tests.

Overall, MFI data became more comparable between samples post-normalisation. MFI ranges on a bead-by-bead basis initially varied between runs from around 500 to as much as 20,000. This was decreased after normalisation; the average MFI range between runs decreased by 1337 for Class I and by 2402 for Class II. All samples showed a statistically significant decrease in coefficient variation (CV) of MFIs between runs after normalisation, both Class I and Class II (p < 0.0001). Inconsistencies in HLA antibody screening results between samples from the same patient can make for troublesome clinical interpretations, especially when there is no sensitising event accounting for reactivity changes. The procedure shown here provides a novel method for reducing the variability caused by differences in assay conditions and has the potential to give more consistent results therefore providing a clearer clinical picture.

Deborah Pritchard

Welsh Transplantation Laboratory, Pontyclun, UK

Immunological compatibility testing for potential live kidney donors and patients requires multiple tests on patient and donor samples (HLA typing, HLA antibody testing, crossmatch assessment). Only on completion of all tests can a report be produced. The local KPI is 90% of cases reported in a turnaround time (TAT) of 10 working days, but performance since Q1 2021/22 has been <75%. This quality improvement project (May–August 2022) identified delays from process maps and selected two areas as targets for improvement; antibody testing and report production. Two plan-do-study-act (PDSA) cycles were performed testing six change ideas generated from staff workshops. Changes included identification of case samples within the testing process to allow more efficient workflow; a dedicated individual with responsibility for reporting rather than shared responsibility; E-mail notification when tests have been completed; coordination of HLA antibody batch testing with sample arrival to reduce wait time, and introduction of visualisation boards to track cases more efficiently. One ‘just do it’ change was also introduced; an interface to import antibody results from HLA Fusion software to the laboratory information management system (LIMS). The mean TAT was reduced from 11.9 to 8.8 days. Improvements were seen in the antibody testing mean TAT (9.1 days→6 days) and report production TAT (3.6 days→2.2 days). This resulted in compliance with the KPI: 95% of cases were reported in 10 days (October–December 2022). The improvement was due to removing manual data entry processes, reducing wait times between procedures and eliminating duplication of work.

Evelien Little, Nicola Brosnan, Jade Kally, Adrian Silk, Lisa Walsh, Franco Tavarozzi

Anthony Nolan Histocompatibility Laboratories, London, UK

With the onset of Next Generation Sequencing (NGS), HLA typing has become more streamlined with the ability to use either singleplex or multiplex primer strategies. The Anthony Nolan Histocompatibility Laboratory utilises GenDx NGSgo® singleplex primers for NGS to achieve high-allelic resolution typing for HLA-A, -B, -C, -DRB1, -DRB3,4,5, -DQB1, -DQA1, -DPA1, and -DPB1. Prior to this work, all samples received for HLA typing were tested for 11 loci, but the number of loci requested may vary. The use of singleplex, as opposed to multiplex, primers has allowed us to implement a flexible approach to provide a tailored and bespoke service. The aim of this work was to develop a process to type samples that need various combinations of testing on the same run. Modifications were needed to our Laboratory Information Management System (LIMS) to allow significant changes throughout the NGS testing process. As a result of this project and as our ongoing flexibility improvement process, we also implemented the ability to repeat PCR failures within the same run, allowing the process to proceed without delay, and merge multiple NGS libraries.

Since we no longer perform unnecessary testing, this generated cost savings by increased capacity on our Illumina® flow cells, allowing more samples per run, potentially reducing our turnaround times, as well as meeting our customer needs. To conclude, singleplex primers have proved extremely useful in allowing flexibility with no discernible increase in run time, using LIMS to simplify and track the process, end to end.

Victoria Wood1,2, Brendan Clark1, Eric Hewitt2, Sunil Daga1

1Leeds Teaching Hospitals NHS Trust, Leeds, UK; 2University of Leeds, Leeds, UK

A core aspect of a renal Transplant Immunology service is the detection of HLA antibodies in the context of prospective donors. The presence of HLA-antibodies is detrimental to chances of transplant and, as donor-specific antibodies (DSA), to graft outcomes. With growing numbers of highly sensitised patients requiring renal transplant across HLA-antibody barriers, an increased understanding of antibody functional characteristics could lead to more informed donor choices. The avidity of an antibody-antigen interaction provides insight into the antibody's capability to induce antibody-mediated changes, such as intracellular signalling, leading to tissue remodelling and graft damage. Chaotropic agents reduce protein stability and have been previously used within ELISA protocols to estimate avidity of antibody-antigen interactions through a process called chaotropic disruption.

Modification of our standard One Lambda LABScreen Single Antigen bead (SAB) protocol to include a chaotropic agent has demonstrated technical viability of applying chaotropic disruption to a solid-phase assay and that this manipulation can be confidently interpreted. A maximum molarity of chaotropic agent was established which is not detrimental to bead surface antigen integrity - 1 molar. This was verified using flow cytometric analysis with HCA2 monoclonal, which binds to both native and denatured HLA-Class I, in comparison with HLA-A, -B, -C monoclonal which binds to native HLA-Class I only. Initial testing using sera with known HLA antibody profiles showed antibody-specific patterns of binding and disassociation. This novel method represents a potential accessible method of testing HLA antibody avidity in an NHS laboratory setting.

Mr Steven Jervis1,2, Dr Antony Payton3, Dr Marcus Lowe1,2, Dr Altug Didikoglu4, Professor Arpana Verma2, Professor Kay Poulton1,2

1Manchester Transplantation Laboratory, Oxford Road, United Kingdom; 2Faculty of Biology, Medicine and Health, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, University of Manchester, Manchester, United Kingdom; 3Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; 4The Centre for Biological Timing, Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, United Kingdom

Studies have hypothesised that the combination of pre-existing genetic traits and specific environmental triggers determine the onset of narcolepsy. The most impactful genetic risk factor is the presence of Human Leukocyte Antigen (HLA) DQB1*06:02 encoded on the Major Histocompatibility Complex (MHC), however, the presence of HLA-DQB1*06:02 is not ubiquitous in all narcolepsy cases. The most poignant genetic risk factors outside the MHC are predominantly located in genes associated with the immune system. In addition to the traditional symptoms of narcolepsy, the co-morbidities can vary with a cohort of sufferers complaining of cognitive dysfunction, particularly memory and attention. These self-reports are not substantiated by consistent scientific evidence whereas there is significant evidence outlining the genetic contribution underpinning variation in cognitive abilities in the general population. In this study we impute targeted non-MHC narcolepsy associated single nucleotide polymorphisms (SNPs) from 1,558 non-pathological elderly volunteers who have been followed for change in cognitive function for up to a 24-year period. Specifically, we investigate 13 previously documented narcolepsy associated SNPs with a odds ratio greater than or equal to 1.00 combined with a minor allele frequency of greater than 0.05. We observed an association between rs306336, rs4290173 and rs2834168 and a faster decline in long term memory. Similarly, we observed a protective effect of rs10995245 against the decline of long-term memory loss. This investigation suggests that the cognitive problems reported by cohorts of narcoleptic patients may be due to genetic predispositions and supports the variation seen in the co-morbidities associated with narcolepsy.

Raji Patel, Aliyye Karasu, Liezelle Pagala, Jyoti Bhatt, Edgar Correa, Gloria Adeyemo, Maame-Esi Yeboah, Abigail Sarkodie, Andrew Joahill, Suzette Cavanna, Carla Rosser, Colin Brown

NHS Blood and Transplant-Colindale, London, UK

The British Bone Marrow Registry (BBMR) utilises Oragene® saliva collection kits to facilitate extended HLA typing of potential donors by Next Generation Sequencing (NGS). Saliva is a good alternative source of DNA as its collection is non-invasive, allows for self-collection and is stable at ambient temperature. In January 2023, the Qiagen EZ2 robot was introduced into our laboratory to provide DNA extraction for clinical samples. However, Qiagen were unable to provide a validated protocol for the extraction of DNA from saliva samples using this instrument. In this study, we assessed the capability of Qiagen EZ2 tissue kits to obtain DNA from saliva samples using the Qiagen EZ2 Connect robot. Using the protocol, we developed, DNA was successfully extracted from 10 saliva samples, with an average DNA concentration of 33 ng/μl and a 260/280 ratio of 1.72. The quality of DNA was comparable to DNA extracted from saliva samples using the validated Roche Magnapure method. The suitability of saliva DNA for high resolution HLA typing was assessed using the One Lambda FASTplex kit. Using TypeStream Visual (TSV) NGS analysis software, we achieved average mapped reads of 334,691 and the results met our documented acceptance criteria for all quality metrics. In conclusion, we have demonstrated that the Qiagen EZ2 Connect robot can be used for successful extraction of DNA from saliva samples to a comparable quality of blood. Validation of this process has allowed our department to avoid unnecessary delays in BBMR extended HLA typing requests and thereby supports patient care.

Sebastian Fernando, Jennifer Lord, Nicola Martin, Alison Logan, Kay Poulton

University of Manchester NHS Foundation Trust, Manchester, UK

Our current routine HLA typing methodology to support disease association testing is LABType™ SSO (One Lambda). An alternative HLA typing methodology EUROArray (EUROIMMUN), was evaluated for concordance and efficiency. This system combines polymerase chain reaction and microarray technologies. Amplified target DNA labelled with a fluorescent dye hybridises to complimentary DNA probes using BIOCHIP technology. Fluorescence signals are evaluated automatically using the EUROIMMUN Microarray Scanner and EUROArrayScan software. Routine and external proficiency scheme peripheral blood and DNA samples previously HLA typed by LABType™ SSO were tested using the appropriate EUROArray assay. Forty-six samples were tested with the HLA-B27 Direct assay, 41 samples with the HLA-B*57:01 Direct assay and 42 samples with the HLA-DQ2/DQ8-h Direct assay. A range of relevant HLA-B and HLA-DQ alleles were selected to ensure the EUROArray system could differentiate between alleles of interest. We found 100% concordance when compared to the LABType™ SSO results for all tests. The workflow was simple and straightforward with a time saving when compared to LABType™ SSO, however the cost of the EUROArray assay was higher. This study has shown the EUROArray Direct assays for HLA-B27, -B57 and -DQ2/8 detection are valid alternative methodologies to support disease association testing. Advantages of the EuroArray system include no additional DNA isolation, the inclusion of numerous integrated controls for high reliability of results and fully automated standardised evaluation and result generation. Limitations of this assay include the use of blood samples within 14 days of venepuncture, although samples can be frozen to mitigate this.

Amy De'Ath, Deborah Pritchard, Tracey Rees

UK NEQAS for H&I, Talbot Green, UK

Scheme 2A and 2B assesses participants’ ability to correctly determine the cytotoxic and flow cytometry crossmatch status across 40 cell/serum combinations per year, respectively. An analysis of performance by UK and Ireland laboratories from 2018–2022 was performed.

There were between 15–22 participants in 2A and 19–22 in 2B. Unsatisfactory performance (UP) in 2A ranged between 0% (2020/21)–38.9% (2018), latest 20%. UP in 2B ranged from 0% (2020/21)–10.5% (2022). Over the 5 years, four laboratories had UP in 2B (one lab in two consecutive years) and 11 in 2A. 2 labs had UP in both schemes. Two out of four labs with UP in 2B were due to performance in the T-cell crossmatch, 1/4 in B-cell and 1/4 in T and B cell crossmatch performance. Six out of eleven labs with UP in 2A were in the B-cell without DTT category, 1/11 in B-cell with DTT, 3/11 in both B-cell with and without DTT and 1/11 in T-cell with and without DTT and B-cell with DTT. In 2B, an average of 36/40 crossmatch combinations per year were assessed. An average of 3% assignments per year were incorrect with a 50:50 split of false negatives and false positives. The use of equivocal reporting was low, average 0.5%. Four percent of samples were reported as not tested. Performance in flow cytometry crossmatching is better than cytotoxic crossmatching during the 5 years. These schemes offer a technical assessment of crossmatching, NEQAS encourage laboratories to participate in our educational schemes which more closely mimic clinical practice.

Amy Bedford, Kathryn Howson, Graham Knighton, Jacqueline Pires, Sarah Maxfield

Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK

HLA-B*27 testing is supportive in the diagnosis of autoimmune diseases including Ankylosing Spondylitis due to their strong association with the HLA-B*27 positive genotype. Testing an average of 383 samples per month, Cambridge Tissue Typing laboratory aimed to introduce a direct-from-blood HLA-B*27 detection assay to eradicate DNA extraction required by current polymerase chain reaction (PCR) sequence specific primer (SSP) based methods, and the associated workload. Utilising the BAG Diagnostics FastQ® B*27 Direct assay, DNA is amplified directly from EDTA whole blood by PCR using SSP. Fluorescent-labelled TaqMan® hydrolysis probes are utilised to enable detection of the amplified product by real-time PCR on the QuantStudioTM 3. Implementation of the FastQ® B*27 Direct assay reduced DNA extraction workload by an average of 64%. Increased batch size capacity (93, vs. 20 using the previous technique) streamlined workflows. Risks associated with exposure to carcinogens ethidium bromide and ultraviolet light and handling boiling agarose gel were eradicated. The absence of DNA extraction and adjustment leads to variance in the volume of nucleated cells utilised, which can result in delayed amplification/false negative results. This highlights the importance of sufficient sample mixing prior to aspiration and stringent result acceptance criteria during analysis using PlexTyper® software, with an average repeat rate of 4%. In summary, implementation of robust mixing procedures and stringent quality metrics have facilitated transition to BAG Diagnostics FastQ® B*27 Direct assay leading to a decrease in workload and demands on staff time, and improvement in the health and safety within the laboratory.

Kelly Spence, Sylvia McConnell, David Turner

H&I Department, SNBTS, Edinburgh, UK

H&I supports the Clinical Apheresis Unit (CAU) and Tissues, Cells and Advanced Therapeutics (TCAT) departments by performing CD34+ stem cell enumeration in acute myeloid leukaemia and multiple myeloma patients receiving autologous stem cell transplantation. The enumeration of CD34+ cells using flow cytometry and the International Society of Hematotherapy and Graft Engineering (ISHAGE) protocol is an established method for the evaluation of stem cell numbers in peripheral blood and apheresis products. Whilst a single (flow cytometry) platform is the recommended approach, locally a dual platform method is in operation, using both flow cytometry and Haematology analyser data. The BD Stem Cell Enumeration assay has been validated on the BD FACSLyric flow cytometer. A comparison between the single and dual platform methodologies using the ISHAGE protocol was undertaken. Analysis was performed on 30 stem cell harvest samples. Strong correlation between the single and dual platform methods was observed for CD34+ absolute count (cells/μl) for stem cell harvest samples (r = 0.99, p < 0.05). There was also a strong correlation between the White Blood Cell (WBC) count from the Haematology analyser and the CD45+ count from the flow cytometer (r = 0.94, p < 0.05). In this validation both the single and dual platform methods for calculating CD34+ cell counts were similar, as were the WBC counts and CD45+ counts from the Haematology analyser and the flow cytometer. This data supports using the recommended single platform method as part of the CD34+ enumeration testing.

Rebecca McGuire1,2, Paul Wright3, Steven Jervis1, Stephine Whiteside1, Malcolm Guiver4, Kay Poulton1,2

1Transplantation Laboratory, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK; 2Faculty of Biology, Medicine and Health, Division of Medical Education, School of Medical Sciences, University of Manchester, Manchester, UK; 3H&I Laboratory, Liverpool Clinical Laboratories, Liverpool, UK; 4Department of Clinical Virology, Manchester University NHS Foundation Trust, Manchester, UK

Acute myeloid leukaemia is an aggressive haematological malignancy with a poor prognosis. Allogeneic haematopoietic progenitor cell transplantation is the only available curative treatment, but it does not eliminate the risk of relapse. Hypotheses for improving relapse rates include utilising donors with advantageous KIR2DL1 allele groups. However, an inexpensive, rapid, and reliable method to discern KIR2DL1 groups does not currently exist. Here, we show that homology between several Killer-cell Immunoglobulin-like Receptor (KIR) loci restricts accurate genotyping of KIR2DL1 allele groups by traditional TaqMan™- based real-time PCR methods. Using multiple sequence analysis, we found that the single nucleotide polymorphisms within codons 114 (rs11673144) and 245 (rs34721508), used to discern KIR2DL1 allele groups, are shared between other KIR loci. In a traditional TaqMan™ real-time PCR assay, this results in non-specific binding and incorrect allele group assignment. Contrary to existing literature, our analysis demonstrated that specificity could not be achieved through the introduction of 3´ terminal mismatches in the forward primers. The results described illustrate the challenges in designing an efficient real-time PCR assay for functional group typing of KIR2DL1. We anticipate our assay to provide the foundation for a more sophisticated real-time PCR assay. Developing a test capable of defining KIR2DL1 allele groups remains of interest to research groups focused on reducing the incidence of acute myeloid leukaemia relapse post-transplant. With further improvements, the described assay could fulfil this goal.

Abigail Levy, Arthi Anand, John Wintour-Pittom

Hammersmith H&I NWLP, London, UK

The Histocompatibility & Immunogenetics (H&I) department at NWLP support renal transplantation at West London Renal Transplant centres. A 24/7 on-call service operates for crossmatching of local patients for transplantation. Timely and effective communication between the renal team receiving deceased donor offers and H&I on-call team is critical for turnaround of compatibility assessment and testing. Historically H&I on-call team have been contactable via ICHT switchboard with provision of a weekly on-call rota. A number of challenges were experienced using this method including calls being directed to the wrong team member interrupting rest and the potential for GDPR breach with patient identifiable information recorded in home environment. ICHT implementation of ALERTIVE app in June 2022 provided alternative on-call communication. H&I were the first pathology service to adopt the app and collaborated with Trust Telecoms & ThamesNet Services to set up the App to meet H&I on-call needs, with Go-Live in February 2023. ALERTIVE improves the speed and quality of communication, between clinical staff and H&I on-call team, in time critical deceased donor transplantation pathway. The ALERTIVE app has been in routine use for three months and has had a very favourable response from H&I on-call team. The app has simplified the process of communicating between Renal and H&I on-call teams, enabling more efficient decision-making. The clinical messaging app is giving us a wealth of new data that was either very hard to get or was unavailable including number of offer related call outs. There are exciting new uses for this data including workforce planning.

Dayna Badaro1,2, Sarah Maxfield1

1Cambridge University Hospital NHS Foundation Trust, Cambridge, UK; 2University of Manchester, Manchester, UK; 3Organ and Tissue Donation and Transplantation, NHS Blood and Transplant, Stoke Gifford, UK

In the United Kingdom, prior to September 2019, deceased donor kidneys were allocated following the National Health Service Blood and Transplant-Organ Donation and Transplantation (NHSBT-ODT) 2006 Kidney Allocation Scheme (KAS). A review by the Kidney Advisory Group prompted significant changes to national allocation with the aim of improving fairness in kidney offering, reducing transplant waiting times and improving longevity of matched transplants. A retrospective clinical audit aimed to review the impact for patients on the Cambridge deceased donor kidney transplant waiting list (TWL), and determine whether the objectives of the 2019 KAS have been met. Transplant data was obtained from NHSBT-ODT spanning 24 months pre-(n = 298) and post-(n = 303) implementation. Data was reviewed taking into consideration the impact of the COVID-19 pandemic. Results show reduced average wait time (days) for highly sensitised patients with a calculated reaction frequency (cRF) ≥85%. In accordance with simulation data, donor/recipient index matching has been achieved, with the majority of ‘low risk’ donors allocated to ‘low risk’ recipients, and the same being true for ‘high risk’ donors/recipients. Divergent donor/recipient age matching was accounted for by difficult to match patients prioritised according to Tier A (cRF 100%/wait time >7 years/matchability score of 10). Contrary to simulation data, Cambridge did not observe a reduction in donation after cardiac death (DCD) transplants following changes to national allocation (n = 174 pre, n = 192 post). In summary data indicates a reduction in average wait time for highly sensitised patients on the Cambridge TWL and suggests objectives of the 2019 KAS have been met.

Michelle Carr1, Shelley Harris1, Judith Worthington1, Alex Woywodt2, Kay Poulton1

1Transplant Laboratory MFT, Manchester, UK; 2Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK

In January 2020 a 24-year-old male with chronic renal failure secondary to reflux nephropathy received his third kidney transplant from a fully HLA matched DBD donor. In March 2020 his kidney function declined with a concurrent rise in creatinine and proteinuria. Tacrolimus levels were variable throughout the course of the patient's transplant history and non-adherence was suspected. A retrospective analysis of non-HLA antibodies using LABScreen™ Autoantibody kits identified that the patient developed antibodies to Glutathione S-Transferase Theta 1 (GSTT1). After his second transplant tacrolimus levels were below target range (<2.5 μg/L), which was followed by the first appearance of the GSTT1 antibody (6000 MFI = 95% percentile). This coincided with a biopsy showing chronic transplant glomerulopathy. Following his third transplant tacrolimus levels remained variable with some levels above and below target range. His transplant function deteriorated further in Spring 2023. A biopsy at this time showed borderline T cell mediated rejection with moderate interstitial fibrosis and tubular atrophy. There was a rapid increase in GSTT1 antibody levels with a peak of 11,000 MFI (>95% percentile). We propose that immune-mediated inflammatory processes triggered by non-adherence episodes caused cellular damage. In response to such damage, intracellular components such as GSTT1 would be released, enabling recognition and the subsequent immune response leading to GSTT1 antibody production. We propose that the detection of antibodies to GSTT1 should be regarded a marker of nephrotoxicity and ongoing trauma to the transplanted kidney.

Adrian Handley, David Briggs, Clare Collins

NHS Blood and Transplant, Birmingham, UK

All measurements come with an element of uncertainty and are only truly useful when that degree of uncertainty is understood. The use of semi-quantitative data from Luminex HLA antibody testing is essential in enabling cardiothoracic transplant compatibility assessment; however, the level of uncertainty was not understood. BSHI/BTS CTAG guidelines set risk levels based on MFI values produced in Luminex Single Antigen Bead HLA antibody assays. Additionally, ISO15189:2012 states that laboratories should calculate measurement uncertainty (MU) for all accredited tests. In this study we used simple statistical tools to identify degrees of MU and some of the contributing factors. These allowed us to quickly assess the effectiveness of changes subsequently made. We identified significant difference between results obtained by individual testing personnel and were able to reduce this through our interventions. Reed et al. (Am J Transplant. 2013;13(7):1859–1870) suggested MU as %CV was in the region of 20%–62%. Our study found this to range from 8% at 20,000 MFI up to 18% at 1000 MFI, the threshold for positivity.

The tools we developed allow ongoing monitoring of assay performance and provide the basis for future quality initiatives, contributing to sustainable, evidence-based quality improvement. MU calculations are an effective quality assurance and improvement tool and are easily implemented. By combining the tools used in this study, levels and sources of variation can be identified, and the effect of any changes to process can be easily assessed for effectiveness.

Saima Azhar Salim, Louise Walsh, Geraldine Donnelly, David Keegan, Joseph Kelly, Mary Keogan

H&I Lab, Beaumont Hospital, Dublin, Ireland

Flow cytometric crossmatch (FXM) is performed during pre-transplant histocompatibility workup. False positive results may prevent a potential recipient receiving a suitable transplant. Many labs use a three-colour FXM assay established using a dual-laser flow cytometer, which includes phycoerythrin (PE), fluorescein isothiocyanate (FITC) and peridinin-chlorophyll proteins (PerCP). There is significant spectral overlap between PE and FITC potentially leading to high background fluorescence, and possible false positive B cell FXM. This study was undertaken to determine optimum fluorochrome combinations to minimise spectral overlap and maximise consistency with the current method for which clinically validated cut-offs are available. Fluorochromes Brilliant Violet (BV421) and Allophycocyanin (APC) were selected for labelling CD3+T cells and CD19+B cells respectively. Thirty-four FXM (total 63 samples for T cell and 61 samples for B cell FXM) were performed with fluorochromes CD3BV421, CD19APC and anti-human IgG FITC. Fourteen FXM (total 35 samples for T and B cell FXM) were performed with fluorochromes CD3PerCP, CD19APC and anti-human IgG FITC. FXM assays were performed on Becton Dickenson (BD) FACSLyric™ and results compared to the standard FXM assay. In comparison to the standard FXM, a statistically significant difference in sample T cell ratio and B cell ratios (p < 0.005) were observed for CD3BV421-CD19APC FXM. Our pilot data suggest no significant difference in T and B cell ratios for CD3PerCP-CD19APC (p > 0.05). Data suggest that substitution of CD19PE with CD19APC minimises spectral overlap, reducing the risk of false positive FXM results, without significantly altering T and B cell ratios.

Ana Bultitude1, Anthony Poles2, Sue Jordan1, Anthony Calvert2, Deborah Sage1

1NHS Blood and Transplant, Tooting, UK; 2NHS Blood and Transplant, Filton, UK

HNA-3 is a bi-allelic antigen, -3a/-3b, with 95% of the UK population encoding at least one HNA-3a allele. HNA-3 is expressed on a variety of cell types including lymphocytes and renal endothelial cells. Accordingly, HNA-3 specific antibodies, which can develop against the non-self-variant in homozygous individuals, have been implicated in rejection episodes following renal transplantation. Here, we report a case of a renal transplant recipient displaying anti-HNA-3a antibodies that prevented transplantation. In August 2022, the patient was offered an altruistic kidney with a 2,1,1 mismatch grade, yielding a negative virtual crossmatch result. However, final wet laboratory flow cytometric crossmatch (FCXM) results presented an unexplained strong positive T and B cell result with a negative auto FCXM. The patient had no previous transplant history and no detectable HLA antibodies since their initial referral. HNA genotyping was performed on both patient and donor, determining HNA-3b3b and HNA-3a3a genotypes respectively. Patient sera was screened for anti-HNA antibodies, confirming the presence of HNA-3a-specific antibodies. The patient was removed from the virtual crossmatching programme as they no longer met the eligibility criteria but remained on the deceased donor waiting list. A further nine deceased donor kidneys were accepted and crossmatched, all of which were T and B cell positive. In March 2023, the patient received a deceased donor kidney which produced a T and B cell negative FCXM. The donor was retrospectively genotyped as HNA-3b3b. The patient continues to do well with no antibody-mediated rejection detected thus far.

Hawzhin Jabar, Luke Foster

NHS Blood and Transplant, Birmingham, UK

Accurate HLA typing is essential to facilitate safe solid organ transplantation. Within the UK, H&I laboratories are required to meet the minimum typing requirements set by NHSBT-OTDT, which includes reporting results that allow for the assessment of any donor specific antibodies (DSA) in a particular donor-recipient combination. Failure to do so can lead to inappropriate allocation or an increased risk of transplant rejection. Here we report the identification of the DRB1*14:15 allele in a deceased solid organ donor who was HLA typed on-call using LinkSeq real-time PCR (One Lambda). Ordinarily, it would not be a requirement to report HLA-DRB1*14 to the second field under the minimum typing requirements, however, interestingly, HLA-DRB1*14:15 does not encode the DR14 antigen, but codes for the DR8 antigen. Therefore, in this instance there was a requirement to report HLA-DRB1*14 to the second field to allow accurate allocation and assessment of any potential DSA. Despite being locally rare, HLA-DRB1*14:15 is listed as well-documented in European populations within the CIWD 3.0.0 catalogue, and common in Asian/Pacific Islands and Native American populations. In this case, donation proceeded with both kidneys being transplanted in two patients, one locally, and out of region. Although the patient transplanted locally was cRF 0% and therefore had no DSA, elucidation of the DR8 antigen may be important for any future post-transplant DSA monitoring.

Richard Battle, Emma Ross, Sylvia McConnell, David Turner

H&I Department, SNBTS, Edinburgh, UK

The SNBTS H&I laboratory provides ∼2300 apheresis donor platelet units annually to HLA sensitised refractory patients across Scotland from a panel of ∼800 typed donors. Data is recorded for each transfusion, including match grade (A = matched for HLA-A and B antigens, B1 = one HLA antigen mismatch, B2 = two antigen mismatch etc.) and cumulative HLA antibody MFI (cMFI) against mismatches. Platelet transfusions were grouped by match and cMFI and assessed against post counts relative to pre-counts that is, a measure of transfusion increment. Pre- and post-platelet counts were available on 1486 cases between 2015–2022. Recipient and donor HLA-A and B types at first field defined A, B1–B4 matching. cMFI was calculated following One Lambda SAB I testing. Analyses used ANOVA or t-test between groups. In 1166 HLA compatible transfusions (cMFI < 2000) no differences were seen in mean increment between A, B1, B2, B3 and B4 matches; 22.5, 22.9, 22.1, 24.1, 30.2 respectively (ANOVA p = 0.14). In 1486 patients grouped according to cMFI, cases with < 2000 had a mean increment = 23.0, with > 2000 < 10,000 mean = 18.0 (p < 0.001) and > 10,000 mean = 8.0 (ANOVA p = 1.55 × 10–15). This analysis of platelet counts after HLA selected platelet provision shows that, as expected, the match grade of the platelets does not impact on the immediate increment. The level of HLA antibody, as defined by cMFI, affects the post transfusion count, especially when cMFI > 10,000. This data will help locally in selection of optimal platelet units for patients.

Patrick Flynn1, Sebastian Fernando2, Judith Worthington1, Kay Poulton1

1Transplantation Laboratory, Manchester Royal Infirmary, Manchester, UK; 2School of Health Education and Public Health Sciences, University of Manchester, Manchester, UK

The aim of this study was to devise an algorithm that would predict Flow Cytometry crossmatch results using SAB Median Fluorescent Intensity (MFI) levels and to test this correlation using samples tested from a NEQAS Scheme 2B cohort.159 NEQAS 2B serum samples were screened using LABScreen™ SAB and 40 NEQAS 2B peripheral blood samples were HLA typed with LABType™ SSO. Donor-Specific Antibodies (DSA) were identified for each cell-serum combination tested and cumulative MFI values calculated for each test. HLA Class I MFIs were combined to predict the T cell crossmatch. For the B cell crossmatch prediction, two options were considered: (i) HLA Class II MFI values alone and (ii) HLA Class I + Class II MFIs. Receiver Operating Characteristic analysis was carried out to identify the combined MFI cut off that predicted NEQAS consensus results with the greatest sensitivity and specificity value. HLA Class I combined MFI > 5000 predicted T cell crossmatch results with 96% sensitivity, 100% specificity, 100% Positive Predictive Value (PPV) and 92% Negative Predictive Value (NPV). For B cell results, HLA Class I + Class II combined MFIs > 11,000 gave the best model showing 97% sensitivity, 79% specificity, 95% PPV and 85% NPV. However, for samples with only HLA Class II sensitisation, combined MFIs > 13,000 improved the B cell crossmatch predictions: 92% Sensitivity, 91% specificity, 92% PPV and 91% NPV. Using this model, combined MFI values can be used to predict the immunological risk posed by DSA when it is not possible to carry out a crossmatch test.

Jonathan A.M. Lucas1, Richard M. Szydlo1,2, Shelley Hewerdine1, Steven G.E. Marsh1,3, Neema P. Mayor1,3

1Anthony Nolan Research Institute, Royal Free Hospital, UK; 2Department of Medicine, Imperial College, UK; 3UCL Cancer Institute, Royal Free Hospital, UK

The effect of matching for HLA-E on the outcome of hematopoietic cell transplantation (HCT) has thus far been inconsistent and has not been studied in a UK cohort where there is an abundant use of alemtuzumab for T-cell depletion. We analysed HLA-E genotypes in 1513 UK HCT patients with a haematological malignancy and their unrelated donors using full-length PacBio Single Molecule Real-Time DNA sequencing. After adjusting for clinical factors that affected outcome prognoses including classical HLA matching out of 12, the presence of mismatches at both HLA-E loci (HLA-Emm; n = 30) was significantly associated with a reduced risk of relapse (HR 0.44; 95% CI 0.20–0.98, p = 0.04) in comparison to being HLA-E matched (HLA-Em; n = 788). A non-significant but beneficial effect on Overall Survival (OS) and Event-Free Survival (EFS) was also observed for two HLA-Emm loci compared to HLA-Em (OS: HR 0.73, p = 0.30; EFS: HR 0.77, p = 0.38). There were no significant differences in HCT outcomes correlated with a single HLA-Emm (n = 450). Assessing directionality of mismatches showed that a bi-directional HLA-Emm (n = 45) was significantly associated with reduced risks of relapse (HR 0.40; 95% CI 0.20–0.83, p = 0.01), increase in EFS (HR 0.55; 95% CI 0.32–0.93, p = 0.02) and a non-significant increase in OS (HR 0.72, p = 0.16) compared to HLA-Em. We hypothesise that mismatching alleles at the HLA-E locus results in sufficient genetic disparity to provide a stronger Graft-versus-Leukemia effect, without eliciting detrimental Graft-versus-Host responses, hence no significant differences in acute Graft-versus-Host Disease or Transplant Related Mortality, as observed in this study.

Sajadhossein Bazrafshani1, Mohammadreza Bazrafshani2

1ENT Department, Worcester, UK; 2Kerman University of Medical Sciences, Kerman, Iran

Cyclosporine, a calcineurin inhibitor, has a narrow therapeutic index and shows considerable inter-individual variability in pharmacokinetics. Cyclosporine is a P-glycoprotein (P-gp) substrate, a multidrug resistance gene (MDR1) product. Some of the single nucleotide polymorphisms (SNPs) of MDR-1 correlate with the variable activity of P-gp in vivo, and it is thought that these polymorphisms are associated with pharmacokinetic variations in cyclosporine therapy. Genotyping assays (PCR-SSP and PCR-RFLP) were performed for detection of frequency within two functional MDR-1 SNPs (C1236T in exon 12 and C3435T in exon 26) in 60 patients and the correlation between genotyping and concentration/dose ratio of cyclosporine was investigated. Data analysis revealed that C3435T polymorphism correlated with the concentration/dose ratio significantly. The concentration/dose ratios were 59.36 μg/L/kg/12 h (p < 0.001) and 30 μg/L/kg/12 h (p = 0.049) lower in homozygous wild-type patients (CC) rather than homozygous mutant-type (TT) and heterozygous patients (CT) respectively. Moreover, heterozygous patients (CT) had 29.36 μg/L/kg/12 h (p = 0.001) which was less than homozygous mutant-type patients (TT). These findings suggest that, for the given dose, the blood concentration is lower in homozygous wild-type individuals (CC type in 3435 position). Our results revealed that MDR-1 genotype appears to influence cyclosporine drug levels and MDR-1 genotyping may provide a useful clinical guide in predicting the required dose after renal transplantation.

Mazen Mabrok1, Renuka Palanicawander2, Betia Nouri1, Rachel Smith1, Arthi Anand1, Natalia Brodaczewska3

1Histocompatibility & Immunogenetics Laboratory, North West London Pathology, Imperial College Healthcare NHS Trust, London, UK; 2Centre for Haematology, Imperial College Healthcare NHS trust, London, UK; 3Specialist Integrated Haematological Malignancy Diagnostic service (SIHMDS), Imperial College Healthcare NHS Trust, London, UK

Monitoring of donor chimerism after haematopoietic stem cells transplantation (HSCT) is vital for early effective therapeutic interventions. Short tandem repeat (STR) assays are the current gold standard for chimerism monitoring after allogeneic HSCT. The emergence of Next Generation Sequencing (NGS) CE-IVD approved assays with improved limit of detection of around 0.05% offer a promising alternative. In this evaluation exercise, we explored the technical capacity of NGS in post-transplant chimerism monitoring, by retrospectively analysing samples from 13 patients from our centre using the Dvysr® NGS chimerism assay (Sweden, Stockholm). All patients enrolled in this evaluation were tested at three time points (whole blood & T- Cell), selected in collaboration with the clinical team based on clinical signs of relapse, increase in MRD markers and the profile obtained using the PowerPlex 16 Multiplex STR system (Promega). Our results showed very strong correlation between the NGS and the STR assays (Pearson score 0.998 & 0.999) with a shift of 1%–1.5% at the pre-relapse time point observed in 55% of the patients enrolled in this evaluation. The NGS assay needed 50% less time to analyse and in 70% of the patients tested, the NGS assay yielded more informative markers than the STR assay. NGS chimerism assay promises improved diagnostic performance and usability in our evaluation. Increased sample size and inclusion of lineage specific cell separation will further strengthen validity of the improved diagnostic performance and usability of NGS chimerism assay as alternative to STR assays including correlation with clinical presentation.

Emma Holmes1, Jasmaine Lee2, Winnie Chong2, Deborah Sage1, Martin Howell2

1NHSBT Tooting Centre, Tooting, UK; 2NHSBT Service Development, Colindale, UK

Nanopore sequencing presents a new technology for high resolution HLA typing that is, considered faster and potentially cheaper than existing methods used for next generation sequencing (NGS). NanoTYPE (Omixon) can be used for batches of up to 24 samples or for single samples, which has the potential to be used for deceased donor HLA typing. The aim of this study was to evaluate the NanoTYPE assay in two NHSBT H&I laboratories; Colindale Service Development and Tooting, using the same batch of 96 samples that had previously been typed by current rapid HLA typing or NGS methods. All NanoTYPE reagents, R9.4 flow cells and a MinION mk1B device were provided by Omixon and Oxford Nanopore Technologies for this study. Eight samples were set-up using the single-sample protocol. The remaining samples were set-up in multi-batches of varying sizes. There was 100% concordance at second field resolution between both sites, and with previous typing data, for HLA-A, -B, -C, DRB4, DRB5 and DPA1. Non-concordance with previous typing data was seen for either HLA-DRB1, DRB3, DQA1, DQB1 or DPB1 loci in 10/96 (10%) samples at Tooting and 9/96 (9%) samples at Colindale, which was attributed to allele imbalance, known low amplification of certain genotypes such as DQB1*03, allele dropout and detection of potential novel alleles. After manual review of the sequencing data, 95/96 samples at Colindale and 93/96 samples at Tooting were concordant with previous results. Our study has proven that the NanoTYPE assay is simple to use and enables high resolution HLA typing.

Emma Ross1, Sylvia McConnell1, Richard Battle1, Nicole Priddee2, David Turner1

1H&I Department, SNBTS, Edinburgh, UK; 2Donor Medicine, SNBTS, Edinburgh, UK

The SNBTS H&I laboratory is responsible for the provision of HLA selected platelets in Scotland. In 2022, 96 patients were supported with a total of 2317 platelet units. A key performance indicator (KPI) for the service is >60% of allocated donor platelets should be an A grade or B1 grade match with the patient (A grade = no HLA-A or HLA-B mismatches at first field; B1 = only one HLA-A or HLA-B mismatch). This target has averaged 57.7% (range 46%−72%) between January 2016 and September 2022, with an overall downward trend observed. In contrast, the number of individual donor platelets issued as HLA selected increased from 1288 units in 2016 to 2317 in 2022 (+55.5%). To make the best use of a limited donor panel, H&I have worked with SNBTS Donor Services (DS) to increase availability of best matched donor platelets. Utilising a Quality Improvement approach, business analytic tools and specialist donor communications, we concentrated on the national identification and (re)engagement of HLA-A and -B homozygous donors which constitute 3.5% of the panel but have the potential to provide A grade matches for ∼68% of patients (Jan–June 2023). Since the project initiation we have recorded an upward trend in the percentage of A and B1 platelet allocations over the period October 2022 (49.5%)–May 2023 (72.4%). The target of >60% has been achieved consistently from Jan 2023. Collaborative working with DS has enhanced the donor panel, improving ‘off the shelf’ access to best matched HLA selected platelets.

Felicity May1, Sian Griffin2, Madhvi Menon3, Tracy Hussell3, Tracey Rees1

1Welsh Transplantation and Immunogenetics Laboratory, Welsh Blood Service, Pontyclun, UK; 2Department of Nephrology and Transplantation, University Hospital of Wales, Cardiff, UK; 3Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK

Human leukocyte antigen incompatible (HLAi) transplantation remains an important option for very highly sensitised patients. Crossing the HLA barrier is associated with increased risk of antibody-mediated rejection (AMR) and graft failure. Desensitisation, induction and maintenance immunosuppression aim to minimise this risk. There is poor consensus on optimal treatment protocols, and patient response varies. In this single centre, retrospective study, we assessed incidence of rejection and graft survival in a clinical cohort of 27 HLAi transplant recipients desensitised with rituximab and cycles of double filtration plasmapheresis. There was poor association between established risk factors and incidence of AMR, suggesting the involvement of other, currently unknown, factors. HLA antibody response and AMR are influenced by a complex cytokine network supporting the generation and survival of antibody producing cells. We conducted a literature review and graded 459 serum protein analytes (cytokines, chemokines, hormones etc.) based on the quality of evidence and proximity of involvement in humoral memory response. We evaluated serum concentrations of the 40 highest graded analytes in longitudinal samples derived from our clinical cohort using a bespoke multiplexed Luminex assay. Due to low samples numbers and heterogeneity of the analyte profiles, we were unable to establish clear association with patient clinical outcomes. However, we observed significant and sustained changes to key signalling molecules known to influence germinal centres, B-cells and plasma cells. We noted a reduction in levels post-desensitisation/immunosuppression for the majority of our panel, but increased or unchanged levels of several key supportive signals and suppression of several immune regulators.

Agnieszka Ojrzynska1, Kylara Hassall1, Katie Butler1, Graham Shirling1, Sharon Vivers2, Raymond Fernando1

1Solid Organ Group, Royal Free Hospital, London, UK; 2Anthony Nolan Histocompatibility Laboratories, London, UK

Increasing numbers of multiple myeloma patients, treated with Daratumumab (Dara), are being listed for kidney transplantation. Dara is a human monoclonal anti-CD38 IgG antibody that interferes with both pre-transplant allogeneic lymphocyte crossmatches and pre-transfusion compatibility testing, as it binds to CD38 molecules expressed on many blood cells including lymphocytes. This can simulate the presence of donor-specific antibodies (DSA) and lead to false positive crossmatch results. The aim of this study was to investigate methods to mitigate Dara interference with flow cytometry crossmatches (FCXM). Our centre has transplanted four patients undergoing Dara treatment. Three of these patients were HLA antibody negative and one patient was HLA antibody positive but HLA DSA negative. Six third party peripheral blood FCXMs, and one retrospective transplant FCXM with spleen cells, were performed using our standard procedure, as well as an amended protocol using donor cells pre-incubated with dithiothreitol (DTT) (0.05–0.1 M) which can cleave the stabilising disulphide bonds of the CD38 molecule. All third-party crossmatches using our standard procedure were T cell positive, two were also B cell positive. The retrospective crossmatch performed with spleen cells was T and B cell positive. All crossmatches performed with DTT were T and B cell negative. Our results show that DTT eliminates the false positivity observed when sera from Dara treated patients are used in FCXM. HLA positive control results were unaffected suggesting that HLA molecules are not affected by DTT treatment. Therefore, DTT can be used to mitigate DARA mediated false positive results in FCXM.

Daniel Eggleston, Helena Lee

Manchester Royal Infirmary, Manchester, UK

Haematopoietic progenitor cell transplantation (HPCT) is well-established as a curative treatment for malignant and non-malignant haematological disorders. However, patient monitoring is still essential following transplantation. Post-transplant donor chimerism testing can identify complications including relapse or graft failure. Relapse is a particular area of concern for patients diagnosed with Myelodysplastic syndromes (MDS) or high-risk leukaemias who have undergone HPCT. Most laboratories perform chimerism monitoring by PCR-Short Tandem Repeats (PCR-STR). This technique uses differences in short repeated sequences of DNA and capillary electrophoresis to determine percentage donor chimerism. However, other methods are emerging that offer improved sensitivity and more informative markers. One such technique is digital PCR (dPCR). During dPCR, a PCR reaction mixture is partitioned into individual droplets. Each function as an individual assay, with the presence of a reaction indicating donor or recipient DNA, increasing technique sensitivity. Using dPCR, we investigated eleven patients diagnosed with MDS or Myeloid Leukaemias whose percentage donor chimerism reached 100% by PCR-STR but dropped in subsequent samples. We tested our cohort using JETA DigitalTRACE™ dPCR technology with the Qiagen QIAcuity machine at two or more timepoints preceding this reduction in donor chimerism using three markers. In 10 of our 11 patients, a reduction in donor chimerism was identifiable by dPCR whereas analysis by PCR-STR had suggested samples were 100% donor chimerism. This raises the possibility of an earlier detection of changes in donor chimerism and an improved follow up process. Chimerism by dPCR is therefore a viable replacement for PCR-STR and may identify events earlier post-HPCT.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
4.70
自引率
0.00%
发文量
48
审稿时长
6-12 weeks
期刊介绍: The International Journal of Immunogenetics (formerly European Journal of Immunogenetics) publishes original contributions on the genetic control of components of the immune system and their interactions in both humans and experimental animals. The term ''genetic'' is taken in its broadest sense to include studies at the evolutionary, molecular, chromosomal functional and population levels in both health and disease. Examples are: -studies of blood groups and other surface antigens- cell interactions and immune response- receptors, antibodies, complement components and cytokines- polymorphism- evolution of the organisation, control and function of immune system components- anthropology and disease associations- the genetics of immune-related disease: allergy, autoimmunity, immunodeficiency and other immune pathologies- All papers are seen by at least two independent referees and only papers of the highest quality are accepted.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信