peri-operative third party red blood cell transfusion in renal transplantation and the risk of...

6
Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss Samantha Fidler a,b,1 , Ramyasuda Swaminathan c,d,2 , Wai Lim e,3 , Paolo Ferrari f,4 , Campbell Witt a,b,5 , Frank T. Christiansen a,b,6 , Lloyd J. D'Orsogna a,b,7 , Ashley B. Irish c,d, ,8 a Department of Clinical Immunology, PathWest, Royal Perth Hospital, Western Australia, Australia b School of Pathology and Laboratory Medicine, University of Western Australia, Australia c Medical Renal Transplant Unit, Royal Perth Hospital, Western Australia, Australia d School of Medicine and Pharmacology, University of Western Australia, Australia e Department of Nephrology Sir Charles Gairdner Hospital, Perth, Western Australia, Australia f Department of Nephrology Fremantle Hospital, Fremantle, Western Australia, Australia abstract article info Article history: Received 12 August 2013 Received in revised form 23 September 2013 Accepted 23 September 2013 Keywords: Donor-specic antibody (DSA) Peri-operative transfusion Kidney transplantation Antibody mediated rejection Historic red blood cell transfusion (RBCT) may induce anti-HLA antibody which, if donor specic (DSA), is associated with increased antibody-mediated rejection (AMR). Whether post-operative RBCT inuences this risk is unknown. We examined the RBCT history in 258 renal transplant recipients stratied according to prevalent recipient HLA antibody (DSA, Non-DSA or No Antibody). AMR occurred more frequently in patients who received RBCT both pre and post transplant compared with all other groups (Pre + Post-RBCT 21%, Pre-RBCT 4%, Post-RBCT 6%, No-RBCT 6%, HR 4.1 p = 0.004). In the 63 pa- tients who received Pre + Post-RBCT, 65% (13/20) with DSA developed AMR compared with 0/6 in the Non-DSA group and 2/37 (5%) in the No-Antibody group (HR 13.9 p b 0.001). In patients who received No-RBCT, Pre-RBCT or Post-RBCT there was no difference in AMR between patients with DSA, Non-DSA or No-Antibody. Graft loss was independently associated with Pre + Post-RBCT (HR 6.5, p = 0.001) AMR (HR 23.9 p b 0.001) and Non- AMR (6.0 p = 0.003) after adjusting for DSA and delayed graft function. Re-exposure to RBCT at the time of transplant is associated with increased AMR only in patients with preformed DSA, suggesting that RBCT provides additional allostimulation. Patients receiving Pre + Post-RBCT also had an increased risk of graft loss independently of AMR or DSA. Both pre and post procedural RBCT in renal transplan- tation is associated with modication of immunological risk and warrants additional study. Crown Copyright © 2013 Published by Elsevier B.V. All rights reserved. 1. Introduction The presence of pre-transplant anti-HLA antibody directed against the donor antigens (DSA) in the presence of a negative CDC crossmatch is associated with increased risk of antibody mediated rejection (AMR) and graft failure [13]. HLA antibodies are formed as a consequence of prior transplantation, pregnancy and blood transfusion due to exposure to foreign HLA antigens [49]. However blood transfusion prior to transplant is immunomodulatory and appears to reduce the risk of acute allograft rejection and graft loss despite an increased risk of sensi- tisation [1012]. Historically it had been observed that large volumes of third-party red blood cell transfusion (RBCT) (up to 20 units) over a prolonged period are required to induce enduring antibodies, especially in males or nulliparous females [4,1315]. However in the presence of another immune stimulating process such as pregnancy or transplanta- tion, co-administration of third party RBCT results in broad HLA anti- body production which is more potent and enduring [6,16,17]. In the Transplant Immunology 29 (2013) 2227 Abbreviations: AMR, antibody-mediated rejection; RBCT, red blood cell transfusion; cPRA, calculated panel reactive antibody; DSA, donor specic antibodies; DGF, delayed graft function; BPAR, biopsy proven acute rejection; MFI, mean uorescence intensity; SAB, single antigen bead. This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike License, which permits non-commercial use, dis- tribution, and reproduction in any medium, provided the original author and source are credited. Corresponding author at: Medical Renal Transplant Unit, Royal Perth Hospital, GPO Box X2213, Perth, Western Australia 6847, Australia. Tel.: +61 8 9224 2162; fax: +61 8 9224 2160. E-mail address: [email protected] (A.B. Irish). 1 Participated in the performance of research laboratory methods, research design and writing the paper. 2 Participated with data collection and analysis. 3 Participated with data collection and writing the paper. 4 Participated with data collection and writing the paper. 5 Participated in research design, data analysis and writing the paper. 6 Participated in research design and writing the paper. 7 Participated in data analysis and writing the paper. 8 Participated in research design, data collection and analysis and writing the paper. 0966-3274/$ see front matter. Crown Copyright © 2013 Published by Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.trim.2013.09.008 Contents lists available at ScienceDirect Transplant Immunology journal homepage: www.elsevier.com/locate/trim

Upload: ashley-b

Post on 26-Dec-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

Transplant Immunology 29 (2013) 22–27

Contents lists available at ScienceDirect

Transplant Immunology

j ourna l homepage: www.e lsev ie r .com/ locate / t r im

Peri-operative third party red blood cell transfusion in renal transplantation and therisk of antibody-mediated rejection and graft loss☆

Samantha Fidler a,b,1, Ramyasuda Swaminathan c,d,2, Wai Lim e,3, Paolo Ferrari f,4, Campbell Witt a,b,5,Frank T. Christiansen a,b,6, Lloyd J. D'Orsogna a,b,7, Ashley B. Irish c,d,⁎,8

a Department of Clinical Immunology, PathWest, Royal Perth Hospital, Western Australia, Australiab School of Pathology and Laboratory Medicine, University of Western Australia, Australiac Medical Renal Transplant Unit, Royal Perth Hospital, Western Australia, Australiad School of Medicine and Pharmacology, University of Western Australia, Australiae Department of Nephrology Sir Charles Gairdner Hospital, Perth, Western Australia, Australiaf Department of Nephrology Fremantle Hospital, Fremantle, Western Australia, Australia

Abbreviations: AMR, antibody-mediated rejection; RBcPRA, calculated panel reactive antibody; DSA, donor spgraft function; BPAR, biopsy proven acute rejection; MFSAB, single antigen bead.☆ This is an open-access article distributed under the tAttribution-NonCommercial-ShareAlike License, which petribution, and reproduction in any medium, provided thecredited.⁎ Corresponding author at: Medical Renal Transplant U

Box X2213, Perth, Western Australia 6847, Australia. Tel.:9224 2160.

E-mail address: [email protected] (A.B. Ir1 Participated in the performance of research laborator

writing the paper.2 Participated with data collection and analysis.3 Participated with data collection and writing the pape4 Participated with data collection and writing the pape5 Participated in research design, data analysis and wri6 Participated in research design and writing the paper7 Participated in data analysis and writing the paper.8 Participated in research design, data collection and an

0966-3274/$ – see front matter. Crown Copyright © 2013http://dx.doi.org/10.1016/j.trim.2013.09.008

a b s t r a c t

a r t i c l e i n f o

Article history:Received 12 August 2013Received in revised form 23 September 2013Accepted 23 September 2013

Keywords:Donor-specific antibody (DSA)Peri-operative transfusionKidney transplantationAntibody mediated rejection

Historic red blood cell transfusion (RBCT)may induce anti-HLA antibodywhich, if donor specific (DSA), is associatedwith increased antibody-mediated rejection (AMR).Whether post-operative RBCT influences this risk is unknown.We examined the RBCT history in 258 renal transplant recipients stratified according to prevalent recipient HLAantibody (DSA, Non-DSA or No Antibody).AMR occurred more frequently in patients who received RBCT both pre and post transplant compared with allother groups (Pre + Post-RBCT 21%, Pre-RBCT 4%, Post-RBCT 6%, No-RBCT 6%, HR 4.1 p = 0.004). In the 63 pa-tients who received Pre + Post-RBCT, 65% (13/20)with DSA developed AMR comparedwith 0/6 in the Non-DSAgroup and 2/37 (5%) in the No-Antibody group (HR 13.9 p b 0.001). In patientswho receivedNo-RBCT, Pre-RBCTor Post-RBCT there was no difference in AMR between patients with DSA, Non-DSA or No-Antibody. Graft losswas independently associated with Pre + Post-RBCT (HR 6.5, p = 0.001) AMR (HR 23.9 p b 0.001) and Non-AMR (6.0 p = 0.003) after adjusting for DSA and delayed graft function.Re-exposure to RBCT at the time of transplant is associated with increased AMR only in patients with preformedDSA, suggesting that RBCT provides additional allostimulation. Patients receiving Pre + Post-RBCT also had anincreased risk of graft loss independently of AMR or DSA. Both pre and post procedural RBCT in renal transplan-tation is associated with modification of immunological risk and warrants additional study.

Crown Copyright © 2013 Published by Elsevier B.V. All rights reserved.

CT, red blood cell transfusion;ecific antibodies; DGF, delayedI, mean fluorescence intensity;

erms of the Creative Commonsrmits non-commercial use, dis-original author and source are

nit, Royal Perth Hospital, GPO+61 8 9224 2162; fax: +61 8

ish).y methods, research design and

r.r.ting the paper..

alysis and writing the paper.

Published by Elsevier B.V. All rights

1. Introduction

The presence of pre-transplant anti-HLA antibody directed againstthe donor antigens (DSA) in the presence of a negative CDC crossmatchis associated with increased risk of antibody mediated rejection (AMR)and graft failure [1–3]. HLA antibodies are formed as a consequence ofprior transplantation, pregnancy and blood transfusion due to exposureto foreign HLA antigens [4–9]. However blood transfusion prior totransplant is immunomodulatory and appears to reduce the risk ofacute allograft rejection and graft loss despite an increased risk of sensi-tisation [10–12]. Historically it had been observed that large volumes ofthird-party red blood cell transfusion (RBCT) (up to 20 units) over aprolonged period are required to induce enduring antibodies, especiallyin males or nulliparous females [4,13–15]. However in the presence ofanother immune stimulating process such as pregnancy or transplanta-tion, co-administration of third party RBCT results in broad HLA anti-body production which is more potent and enduring [6,16,17]. In the

reserved.

Page 2: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

23S. Fidler et al. / Transplant Immunology 29 (2013) 22–27

current transplant era, transfusion in patientswith end stage kidney dis-ease is less frequent due to the widespread use of epoetins. Howeverduring acute illness or surgery patients may still be exposed to bloodproducts, although specifically transfusing patients for immunologicalbenefit is no longer routine [18–20]. Leucodepletion of blood productshas also been shown not to prevent the risk of allosensitisation associat-ed with RBCT [14,21–23]. The majority of studies on the role of bloodtransfusion was performed in the period before the use of sensitiveand specific solid phase antibody detection assays were available andcell-dependent cytotoxicity assays were utilised. Although it isestablished that DSA detected at the time of transplant is associatedwith an increased risk of AMR why some patients with DSA developAMR and others do not is unclear andmay relate to variability in the an-tibody sub-type, complement binding ability, or the amount or breadthof antibody [1,24–26]. Transfusion in the peri-operative and early post-transplant period depends on individualised patient managementfactors and is commonly thought not to be an immunological stimulusbecause it is assumed that the concomitant use of immunosuppressionmitigates this risk. We hypothesised that post-transplant transfusionin patients with preformed HLA antibody may provide additionalallostimulation or immunological recall and increase the risk of AMR.We therefore investigated the relationship of pre-transplant and peri-operative transfusion in renal transplant recipients with and withoutpre-transplant HLA antibody determined by Luminex single antigenbead (SAB) assay.

2. Subject and methods

2.1. Patients

We studied 258 transplant recipients of which 246 patients receiveda kidney transplant and 12 patients received a simultaneous pancreas–kidney transplant between June 2003 and October 2007. Patients weretransplanted at 3 tertiary centres and peri-operative care and decisionfor transfusionswas individualised, clinically indicated and notmandat-ed by protocol. No donor-specific transfusions occurred. Leucocyte de-pleted packed red cells were used. All patients received a calcineurininhibitor (CNI) (tacrolimus or cyclosporine) at the time of transplanta-tion in combination with mycophenolate mofetil or mycophenolatesodium and corticosteroids and the Interleukin-2 receptor antibodybasiliximab was commonly used for induction. The need for biopsy,medication adjustments and transfusion was determined by thecaring clinical teams and was not protocol driven. Transfusion historywas obtained from the West Australian Red Cross Blood Bank, theWestmead Hospital Transfusion Laboratory, patient medical recordsand direct patient interrogation. Patient follow-up was a median of67 months (IQR 54–77). Patients provided written consent for partici-pation in this study.

2.2. Laboratory methods

These are reported in detail elsewhere however stored donor DNAwas typed by sequence based typing at HLA-A, -B, -C, -DRB1, DQB1,DPB1 loci and DRB3, 4, 5 and DQA1 where required [27]. All recipientswere transplanted with a negative T cell CDC crossmatch. B cell cross-matching was performed for 80% of the patients; however a positive Bcell crossmatch was not considered an absolute contraindication totransplantation. Sera collected at the time of transplant were screenedretrospectively for anti-HLA class I and/or class II antibodies usingthe Luminex Mixed Screen assay (OneLambda Inc.) and those with apositive screen were characterised for HLA class I and/or class II anti-bodies specificity using single antigen beads (LABScreen Single Antigenbeads, OneLambda Inc.). Antibodies were considered to be positive ifthe normalised mean fluorescence intensity (MFI) value for a particularbead was greater than 500. HLA antibodies with an MFI N500 directedagainst a donor HLA antigen were considered to be DSA.

2.3. Clinical outcome parameters

Transfusion history was recorded as never transfused (No-RBCT),transfused at any time prior to renal transplant but not after renaltransplant surgery (Pre-RBCT), not transfused prior to transplant buttransfused at the time of, or within 30 days of transplant surgery(Post-RBCT) and transfused both prior to and within 30 days of thetransplant (Pre + Post-RBCT). Delayed graft function (DGF) was de-fined as the need for dialysis within the first 48 h of transplantation.Graft loss was defined as the return to dialysis (i.e. death-censored) un-less otherwise indicated. All rejection episodes were proven by biopsy(BPAR) and the first BPAR was used to construct time to event analysisandwheremultiple rejections occurred, the highest reported gradewasrecorded. Time to AMR was recorded as a separate event to allow anal-ysis by rejection type (AMR vs Non-AMR). Treatment of rejectionwas atthe treating clinician's discretion and was not mandated by protocol.Histological reporting of renal biopsies was undertaken by the local his-topathologists as part of routine clinical care and was initially madewithout information as to the presence or absence of DSA (due to vary-ing laboratory testing and reporting changes over the period of study).The biopsy findings were graded according to the Banff classification2003. AMR was defined as C4d positivity in PTC alone or in conjunctionwith transplant glomerulitis and/or peri-tubular capillaritis and/or ar-teritis, and also in the absence of C4d when transplant glomerulitisand peri-tubular capillaritis were detected.

2.4. Statistical analysis

Statistical analyses were performed by using SPSSv18 (SPSS Inc.,Chicago IL, USA). For categorical data Fisher's exact test or Pearson'schi-square tests were used. Parametric data were compared byANOVA or t-test, and for non parametric data Mann–Whitney U testor Kruskal–Wallis one-way ANOVA was used. Comparisons of withingroup differences by z-test were made with Bonferroni adjustmentreported at the p b 0.05 level. Time to event of interest (AMR, graftand patient survival) was estimated by the method of Kaplan–Meierand Cox proportional hazard regression analysis with the predictorsatisfying the proportional hazard assumption. Covariates examinedwere HLA-antibody at entry, rejection, gender, re-transplantation, anddelayed graft function. Results were expressed as hazard ratios (HR)with 95% CI.

3. Results

Sixty-five patients had pre-transplant HLA-antibody: DSA group n = 37 (14%) andNon-DSA group n = 28 (11%) while the remaining 193 (75%) patients had no HLA anti-body defined using the MFI cut-off of b500 or with a negative antibody screen. Baselineclinical and demographic data of these groups is reported in detail elsewhere andsummarised in Table 1. [27]As expected, patients with any HLA antibodyweremore com-monly female (41/65 vs 53/193 p = 0.003) and more likely to have undergone prior kid-ney transplant (20/65 vs 7/193 p b 0.001) and to have received Pre-RBCT (39/65 vs70/193 P = 0.011). There was no difference in haemoglobin between the groupseither at time of transplant (DSA 124 ± 19, Non-DSA 124 ± 18, No-Antibody124 ± 15 g/L p = 0.99) or at 30 days post transplant (DSA 109 ± 17, Non-DSA113 ± 13, No-Antibody 114 ± 17 g/L p = 0.19). Patients with pre-transplant DSAwere significantly more likely to have been transfused within the first 30 peri-operativedays (DSA 70%) than those with Non-DSA (43%) or no HLA antibody (38% p b 0.001)although the amount of RBCT was not different [DSA 4 (2–4), Non-DSA 2 (2–4) and No-Antibody 2 (2–4) units median and IQR p = 0.17] and N90% of all post-transplant RBCTgiven within the first 2 peri-operative days.

In order to explore further the relationship between transfusion and pre-transplantDSA we divided the patients into four groups according to their transfusion status— No-RBCT, Pre-RBCT, Post-RBCT and Pre + Post-RBCT groups as previously defined (Table 2).Overall 109/258 (42%) received Pre-RBCT and 111/258 (43%) of patients received Post-RBCT. The prevalence of HLA antibody amongst these groups varied significantly asexpected. The No-RBCT group were much more likely to have no HLA antibody (86%)than the other groups (p b 0.05). Conversely however, the Pre + Post-RBCT group weremore likely to have DSA (p b 0.05), receive a repeat transplant and less likely to receivea pre-emptive or living donor transplant, although time on dialysis was similar tothose with Pre- and Post-RBCT. Patients with Pre-RBCT only were significantly less likelyto have Non-AMR rejection than all other groups (p b 0.05 Table 3). Patients in the

Page 3: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

Table 3Univariate and multivariate adjusted Cox Proportional Hazards models for the effect ofDelayed Graft Function (DGF), entry HLA antibody, Antibody and Non-AntibodyMediatedRejection (AMR and Non-AMR) and Transfusion status on death censored and combinedpatient and graft loss.

Death censored graft lossN = 27

Combined patient and graft lossN = 51

Univariate Multivariate Univariate Multivariate

DGF 2.1 (0.92–4.8)0.078

0.9 (0.4–2.1)0.73

2.5 (1.5–4.5)0.002

1.5 (0.8–2.8)0.23

Non-DSA 1.4 (0.47–4.2)0.54

1.3 (0.4–4)0.73

1.0 (0.5–2.0)0.93

0.9 (0.3–2.1)0.74

DSA 2.6 (1.1–6.4)0.034

0.56 (0.2–1.6)0.28

2.2 (1.1–4.2)0.024

0.93 (0.4–2.0)0.86

Non-AMR 7.0 (2.3–21.5)0.001

6.0 (1.9–19.3)0.003

2.3 (1.2–4.2)0.012

2.0 (1.0–3.8)0.048

AMR 25.8 (8–83.2)b0.001

23.9 (6.9–83.2)b0.001

6.0 (2.9–12.5)b0.001

4.8 (2.1–10.8)b0.001

Pre-RBCT 0.5 (0.1–4.6)0.55

0.64 (0.1–6.0)0.70

1.8 (0.7–4.7)0.22

2.2 (0.8–5.8)0.12

Post-RBCT 2.7 (0.8–9.7)0.13

2.6 (0.7–9.6)0.15

1.6 (0.6–4.2)0.32

1.6 (0.6–4.1)0.37

Pre + Post-RBCT 7.1 (2.4–7.3)b0.001

6.5 (2.0–21.6)0.002

5.1 (2.9–10.9)b0.001

3.9 (1.7–8.9)0.001

Table 1Demographic and clinical features at time of transplant by entry HLA-antibody.

NoHLA antibodyn = 193

HLA antibody n = 65

Non-DSAn = 28

DSAn = 37

P value

Age at transplant (years) 47 ± 13 46 ± 11 44 ± 11 0.45Recipient female N (%) 53 (28)a 19 (68) 22 (60) b0.001Cadaveric N (%) 118 (61) 21 (75) 28 (76) 0.11Repeat transplant N (%) 7 (4)a 9 (32) 11 (30) b0.001Pre-emptive transplant N (%) 22 (11) 2 (7) 3 (8) 0.70Months on dialysis 25 (9–51) 38 (12–54) 50 (13–82) 0.042HLA mismatch 3 (3–4) 2 (1–4) 3 (2–4) 0.11DGF N (%) 34 (18) 3 (11) 10 (27) 0.22IL2R-Ab induction N (%) 87 (47) 15 (54) 25 (68) 0.65Anti-T cell induction N (%) 3 (2) 1 (4) 2 (5)Initial CNI tacrolimus N (%) 123 (64) 16 (57) 28 (76) 0.26Pre-RBCT N (%) 70 (36)a 16 (57) 23 (62) 0.003Units of RBCT 5 (2–9) 5 (2–9) 5 (4–13) 0.69Post-RBCT N (%) 73 (38) 12 (43) 26 (70)# 0.001Units of RBCT 2 (2–4) 2 (2–4) 4 (2–4) 0.17Haemoglobin at transplant g/L 124 ± 15 124 ± 18 124 ± 19 0.99Haemoglobin at 1 month g/L 114 ± 17 113 ± 13 109 ± 17 0.19

a Significantly different from Non-DSA and DSA at the p b 0.05 level.

24 S. Fidler et al. / Transplant Immunology 29 (2013) 22–27

Pre + Post-RBCT group were more likely to have DGF than all other groups, and had a4 fold increased risk of AMR (Table 3 and Fig. 1 p = 0.004) with a median time to AMRof 2 months.

Given the association of Pre + Post RBCT with AMR we next examined the impor-tance of pre-transplant DSA, a known risk for AMR, in the various transfusion groups.When stratifiedby thepresenceof DSA therewasnodifference in the risk of AMRbetweenthose with or without DSA in either of the No-RBCT, Pre-RBCT or Post-RBCT groups indi-vidually (data not shown) or when these 3 groups were combined (Fig. 2a). In thePre + Post RBCT group the risk of AMR was 13.9 times greater in those patients withDSA than in patients with Non-DSA or No-Antibody (Fig. 2b p = 0.001). Indeed all 13episodes of AMR in the DSA group occurred exclusively in patients who had receivedPre + Post-RBCT. On the other hand, 0/6 patients with Non-DSA and 2/37 in the No-antibody group who had received Pre + Post-RBCT developed AMR. The median timebetween post-operative transfusion and AMR in the DSA patients was 25 days (IQR5–761 days).

Univariate predictors of AMR were pre-transplant DSA (HR 6.6 95%CI 2.9–14.7,p b 0.001), DGF (HR 2.6. 1.1–6.1, p = 0.039), re-transplant (HR 3.3 1.3–8.3 p = 0.024)and Pre + Post-RBCT (HR 4.1, 1.6–10.8 p = 0.005) but not females (HR 0.9 0.38–2.1).There was a significant interaction between Pre-RBCT and Post-RBCT (HR 4.4 2.0–9.8p = 0.001) and between DSA and Post-RBCT (HR 10.6 4.7–23.8 p b 0.0001) on the riskof AMR. In amultivariate Coxmodel incorporating the above univariate factors and adding

Table 2Patient demographics, HLA antibody, rejection and outcomes by transfusion status.

No-RBCTN = 101

Pre-RBCTN = 46

Age at transplant (years) 43 ± 13 48 ± 14a

Female recipients N (%) 24 (24)b 17 (37)Living donors N (%) 42 (42) 16 (35)Repeat transplant N (%) 3 (3) 10 (22)c

Pre-emptive transplant N (%) 12 (12) 2 (4)Time on dialysis (months) 28 (12–48) 48 (13–74)HLA mismatch (IQR) 3 (2–4) 3 (2–4)DGF N (%) 9 (9) 5 (11)Hb at transplant g/L 127 ± 14 130 ± 15Hb at 1 month g/L 115 ± 15 116 ± 16No HLA-antibody N (%) 87 (86)d 33 (72)Non-DSA N (%) 6 (6) 10 (22)a

DSA N (%) 8 (8) 3 (7)No BPAR N (%) 65 (64) 35 (76)Non-AMR N (%) 30 (30) 9 (20)d

AMR N (%) 6(6) 2 (4)Time to AMR (months) 50 (40–60) 55 (49, 71)Time to Non-AMR (months) 7 (1–19) 1 (0–8)

Results shown as mean ± SD, median and (IQR) or number (percentage).a Significantly different from No-RBCT (p b 0.05).b Significantly different from Post-RBCT and Pre + Post-RBCT (p b 0.05).c Significantly different from No-RBCT and Post-RBCT (p b 0.05).d Significantly different from all 3 other groups (p b 0.05).

interaction terms, only the interaction between DSA and Post-RBCT (HR 7.2, 2.9–18.0,p = 0.001) remained a significant predictor of AMR.

Death-censored graft loss (Fig. 3 and Table 3) and combined patient and graft loss(Table 3) was significantly increased in the Pre + Post-RBCT group (HR 7.1 p b 0.001)compared with all other transfusion groups. This difference persisted even after exclusionof the 37 patients with preformed DSA (HR 4.9 95% CI 1.5–15.8 p = 0.007 and 3.9 1.7–7.8p = 0.001 respectively). Neither gender nor retransplant were associated with graft orpatient loss. We used significant univariate predictors of graft loss including DGF, HLAantibody (DSA and Non-DSA), AMR and Non-AMR rejection and transfusion history in amultivariate Cox Proportional Hazards model (Table 3). AMR, Non-AMR rejection andPre + Post RBCT were independently associated with death censored and all cause graftloss. However DGF and DSA were no longer predictive by multivariate analysis.

4. Discussion

We show that the risk of AMR associatedwith the presence of DSA atthe time of transplant is modulated by exposure to RBCT. In our cohort,AMR was predominantly observed only in sensitised patients withDSA all of whom had received RBCT prior to transplant and were then

Post-RBCTN = 48

Pre + Post-RBCTN = 63

P value

51 ± 12a 47 ± 13 0.00427 (56) 26 (41) 0.00121 (44) 12 (19)c 0.0140 (0) 14 (22)c 0.00112 (25)d 1 (2)a b0.00133 (13–70) 40 (17–79) 0.0613 (3–4) 3 (2–4) 0.438 (17) 25 (40)d b0.001116 ± 13 123 ± 13 b0.001113 ± 16 109 ± 15 0.04536 (75) 37 (59) Χ2 29.7 p = 0.0016 (13) 6 (10)6 (13) 20 (32) a

28 (58) 26 (41) a Χ2 21.0 p = 0.00217 (35) 24 (38)3(6) 13 (21)d

31 (2, 31, 60) 2 (0–30)2 (0.5–15) 2 (1–7)

Page 4: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

Fig. 1. Kaplan–Meier plot of time to AMR by transfusion status in all patients. Patientswith Pre + Post-RBCT had HR 4.1 (95%CI 1.6–10.8 p = 0.004) for AMR compared withNo-RBCT. Note Y-axis is truncated.

Fig. 2. a&b: Time to antibody mediated rejection (AMR) by antibody status at the time oftransplant in the combined 195 patients receiving No-RBCT, Pre-RBCT and Post-RBCT(p = 0.56, panel a) and the 63 patients with Pre + Post-RBCT (p = 0.001, HR 13.9 DSAvs No-antibody, panel b). Note different Y axis origin.

25S. Fidler et al. / Transplant Immunology 29 (2013) 22–27

transfused within the first 30 days (usually 48 h). Pre-transplant DSAand Pre + Post-RBCT were each independent predictors of AMR. How-ever, there was a strong interaction between pre-transplant DSA andPost-RBCT, which eliminated all other predictors (DGF, re-transplant,gender), and conferred a 7.2 fold increase in the risk of AMR. In contrast,patients with DSAwho received only Pre-RBCT or Post-RBCT, orwho re-ceived no transfusion did not experience AMR.

Overall, patients with DSA had the highest rate of post-operativetransfusion. In our centres, the decision to give peri-operative transfu-sion was solely determined by clinical need, authorised by differentclinical teams and, in all cases,madewithout knowledge of the patient'santi-HLA antibody status. Our reported post-operative transfusion rateof 43% is very similar to those reported elsewhere [28,29]. The reasonfor the higher rate of peri-operative transfusion in patients with DSAcompared with Non-DSA is therefore uncertain. It may be due to thegreater medical and surgical complexities of this patient group andtheir greaterwaiting time on dialysis for example. However, it is notablethat there was no difference in gender, re-transplantation, deceaseddonors or Pre-RBCT between the DSA and Non-DSA groups at time ofsurgery, or between the haemoglobin at surgery or at 1 month post-surgery. Although residual confounding by indication remains possible,it is not possible to either entirely adjust or explain and this differencerequires further testing.

The immunological interaction of blood transfusion and transplanta-tion is complex. Pre-RBCT is associated with better graft outcomes andless acute rejection, and this is suggested to be due to immunomodulationwith down-regulation of an immune response and the induction of regu-latory T-cells [11,30]. Indeed our study confirms the continued benefit ofPre-RBCT alone with this group having the lowest rate of Non-AMR. Wealso confirm that Pre-RBCT is still associated with an increased risk ofHLA-antibody sensitisation. Several recent reports [28,29] raise someconcern that post-operative transfusion is associated with poor graft out-come. However these studies did not consider sensitisation or prior trans-fusion as potential modifiers and these factors may account for theirconflicting conclusions. Herewe report that peri-operative blood transfu-sion is associated with an increased risk of AMR, but only in recipientswith pre-transplant DSA detected using solid phase assays, all of whomhad been previously exposed to RBCT and other sensitising events. This

effect of peri-operative transfusion was not found in recipients withoutDSA, suggesting that the combination of DSA and peri-operative bloodtransfusionmay be particularly detrimental to the transplanted graft. Im-portantly, adverse events after peri-operative blood transfusion includednot only antibodymediated rejection, but also poorer long term graft out-come and recipient death, independent of the risk of AMR and Non-AMR,consistent with the findings of O'Brien et al. [28].

In light of our findings it is worth considering the immunologicalmechanisms whereby blood transfusion could increase the pathogenic-ity of pre-existing DSA. This might be through direct quantitative orqualitative alterations in antibody or indirectly via specific transfusionfactors. Scornik et al. [16] have previously identified that re-exposureto blood in those with prior sensitising events such as transplant orpregnancy elicits a broad antibody response;findings that are consistent

Page 5: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

Fig. 3. Time to death censored graft loss by transfusion status HR 7.1 (2.4–21.3) p b 0.001for Pre + Post-RBCT compared with No-RBCT.

26 S. Fidler et al. / Transplant Immunology 29 (2013) 22–27

with our study. Interestingly, broad recall of an antibody response topreviously exposed transplant antigens, rather than to the transfusionantigens, was induced by third party transfusions. [31] Qian et al. [32]have shown in a cardiac rodent model that pre-sensitisation withallogeneic RBCT causes accelerated graft rejection in the presence ofcomplement and antibody binding to graft endothelium. Complementactivation products and quantitative changes in cytokines may bepresent within stored blood products [33,34]. Norda and colleaguesfound that stored plasma components may differ significantly inthe amount and timing of complement activation products, particularlyC3a, which could specifically trigger pathological changes if pre-existingeffector DSA is present. Theoretically cytokines andother factors presentin blood products could also induce non-complement activating DSAto class switch to IgG1 and/or IgG3 complement activating antibodies.Mice models of transfusion related acute lung injury suggest thatMHC class I specific antibody binding to nonhematopoietic cells drivescomplement activation and production of reactive oxygen species[35]. T cell allorecognition of allogeneic HLA molecules, present evenin leucodepleted blood products, may be associated with specificand non-specific immune activation including increased cytokineproduction and cytotoxicity function. Whether exposure to RBCT insensitised patients stimulates an increase in the absolute amount orbreadth of DSA and/or class switching and complement bindingwas be-yond the scope of this study. Serially monitoring these changes wouldbe informative however the frequency of sampling and interventionsfor management of rejection which alter antibody measurement con-found interpretation. These putative adverse effects of peri-operativeblood transfusion could be investigated further using in-vivo animalmodels.

This data suggests that avoidance of peri-operative blood transfu-sion or, given the impossibility of eliminating all transfusion risk,establishing a newparadigm for RBCT in sensitised transplant recipientsshould be considered. It is established that leucodepleted unmatchedRBCT does not reduce sensitisation [23] and therefore selecting HLAmatched blood with its significantly reduced risk of HLA specific anti-body production may be particularly suited to patients with DSA [36].The use of HLAmatched blood transfusion products for renal transplan-tation patients is feasible and may be effective within a clinical setting[36,37]. Furthermore recipients with high levels of sensitization may

even benefit from pre-operative storage of autologous blood productsfor use in the event of requiring a peri-operative blood transfusion[38]. The beneficial effects of blood transfusion on graft survival havebeen reported to be associated with HLA-DR matching, and while HLAtyping of the blood donors was not available for this study it is highlylikely that the observed adverse effect is associated with specific HLAmismatches between the blood donor and organ recipient [36].

In conclusion, we have shown that Pre-RBCT alone is still associatedwith a lower rate of Non-AMR rejection and an increased risk of HLA an-tibody.However, peri-operative blood transfusion in sensitised renal re-cipients with DSA and prior transfusion is associated with AMR. Post-RBCT may therefore be an additional factor modifying the risk of AMRin patients with HLA-antibody. We also confirm and expand upon theprevious findings that perioperative blood transfusion is associatedwith poorer graft and patient survival, and show that this is mostevident in those with previous exposure to RBCT, independent ofacute rejection episodes. These findings suggest that RBCT remains apotent and complex modifiable immunomodulator of renal transplantoutcomes and additional studies to further define mechanisms forthese effects are warranted.

Disclosure

This is an original work and the manuscript or parts of it have notbeen submitted elsewhere for publication. All authors have read andapproved submission of the manuscript, and that material in themanuscript has not been published and is not being considered forpublication elsewhere in whole or part in any language except asan abstract.

Acknowledgements

The authors wish to acknowledge the clinicians and transplantnurses at Royal Perth Hospital, Sir Charles Gairdner Hospital and Fre-mantle Hospital in Western Australia involved in the collection ofthis data. We wish to thank the Department of Clinical Immunology,Royal Perth Hospital for compatibility testing of the patients in thisstudy.

References

[1] Lefaucheur C, Loupy A, Hill GS, Andrade J, Nochy D, Antoine C, et al. Preexistingdonor-specific HLA antibodies predict outcome in kidney transplantation. J Am SocNephrol 2010;21:1398–406.

[2] Otten HG, Verhaar MC, Borst HP, Hene RJ, van Zuilen AD. Pretransplant donor-specific HLA class-I and -II antibodies are associated with an increased risk forkidney graft failure. Am J Transplant 2012;12:1618–23.

[3] Song EY, Lee YJ, Hyun J, Kim YS, Ahn C, Ha J, et al. Clinical relevance of pretransplantHLA class II donor-specific antibodies in renal transplantation patients with negativeT-cell cytotoxicity crossmatches. Ann Lab Med 2012;32:139–44.

[4] Opelz G, Graver B, Mickey MR, Terasaki PI. Lymphocytotoxic antibody responses totransfusions in potential kidney transplant recipients. Transplantation 1981;32:177–83.

[5] Sanfilippo F, Vaughn WK, Bollinger RR, Spees EK. The influence of pretransplanttransfusions, using different blood products, on patient sensitization and renal allo-graft survival. Transplantation 1984;37:350–6.

[6] Scornik JC, Ireland JE, Howard RJ, Fennell III RS, Pfaff WW. Role of regular andleukocyte-free blood transfusions in the generation of broad sensitization. Trans-plantation 1984;38:594–8.

[7] Pfaff WW, Howard RJ, Scornik JC, Day C, Renderer J, Scott J, et al. Incidental andpurposeful random donor blood transfusion. Sensitization and transplantation.Transplantation 1989;47:130–3.

[8] ChowMP, Hu HY, Lyou JY, Lin JS, Yung CH, Lee A, et al. Red cells, HLA and plateletantibody formation in patients with multiple transfusions. Acta Haematol1994;92:57–60.

[9] Novotny VM, van Doorn R, Witvliet MD, Claas FH, Brand A. Occurrence of allogeneicHLA and non-HLA antibodies after transfusion of prestorage filtered platelets andred blood cells: a prospective study. Blood 1995;85:1736–41.

[10] Blajchman MA. Immunomodulatory effects of allogeneic blood transfusions: clinicalmanifestations and mechanisms. Vox Sang 1998;74(Suppl. 2):315–9.

[11] Claas FH, Roelen DL, van Rood JJ, Brand A. Modulation of the alloimmune responseby blood transfusions. Transfus Clin Biol 2001;8:315–7.

[12] Opelz G, Graver B, Terasaki PI. Induction of high kidney graft survival rate by multi-ple transfusion. Lancet 1981;1:1223–5.

Page 6: Peri-operative third party red blood cell transfusion in renal transplantation and the risk of antibody-mediated rejection and graft loss

27S. Fidler et al. / Transplant Immunology 29 (2013) 22–27

[13] Scornik JC, Meier-Kriesche HU. Blood transfusions in organ transplant patients:mechanisms of sensitization and implications for prevention. Am J Transplant2011;11:1785–91.

[14] Karpinski M, Pochinco D, Dembinski I, Laidlaw W, Zacharias J, Nickerson P. Leuko-cyte reduction of red blood cell transfusions does not decrease allosensitizationrates in potential kidney transplant candidates. J Am Soc Nephrol 2004;15:818–24.

[15] Scornik JC, Pfaff WW, Howard RJ, Fennell III RS, Ramos E, Peterson JC, et al. Increasedantibody responsiveness to blood transfusions in pediatric patients. Transplantation1994;58:1361–5.

[16] Scornik JC, Ireland JE, Howard RJ, Pfaff WW. Assessment of the risk for broad sensi-tization by blood transfusions. Transplantation 1984;37:249–53.

[17] Sanfilippo F, Vaughn WK, Bollinger RR, Spees EK. Comparative effects of pregnancy,transfusion, and prior graft rejection on sensitization and renal transplant results.Transplantation 1982;34:360–6.

[18] Fox KM, Yee J, Cong Z, Brooks JM, Petersen J, Lamerato L, et al. Transfusion burden innon-dialysis chronic kidney disease patients with persistent anemia treated in rou-tine clinical practice: a retrospective observational study. BMC Nephrol 2012;13:5.

[19] Ibrahim HN, Skeans MA, Li Q, Ishani A, Snyder JJ. Blood transfusions in kidney trans-plant candidates are common and associated with adverse outcomes. Clin Transpl2011;25:653–9.

[20] Aalten J, Bemelman FJ, van den Berg-Loonen EM, Claas FH, Christiaans MH, de FijterJW, et al. Pre-kidney-transplant blood transfusions do not improve transplantationoutcome: a Dutch national study. Nephrology, dialysis, transplantation. Off PublEur Dial Transplant Assoc Eur Ren Assoc 2009;24:2559–66.

[21] Balasubramaniam GS, Morris M, Gupta A, Mesa IR, Thuraisingham R, Ashman N.Allosensitization rate of male patients awaiting first kidney grafts after leuko-depleted blood transfusion. Transplantation 2012;93:418–22.

[22] Scornik JC. Leukoreduced blood and sensitization of organ transplant candidates.Transfusion 2012;52:1838–9 [author reply 9].

[23] van de Watering L, Hermans J, Witvliet M, Versteegh M, Brand A. HLA and RBCimmunization after filtered and buffy coat-depleted blood transfusion in cardiacsurgery: a randomized controlled trial. Transfusion 2003;43:765–71.

[24] AlMahri A, Holgersson J, Alheim M. Detection of complement-fixing and non-fixingantibodies specific for endothelial precursor cells and lymphocytes using flowcytometry. Tissue Antigens 2012;80:404–15.

[25] Chen G, Sequeira F, Tyan DB. Novel C1q assay reveals a clinically relevant subset ofhuman leukocyte antigen antibodies independent of immunoglobulin G strengthon single antigen beads. Hum Immunol 2011;72:849–58.

[26] MujtabaMA, GogginsW, Lobashevsky A, Sharfuddin AA, YaqubMS,Mishler DP, et al.The strength of donor-specific antibody is a more reliable predictor of antibody-mediated rejection than flow cytometry crossmatch analysis in desensitized kidneyrecipients. Clin Transplant 2011;25:E96-102.

[27] Fidler SJ, Irish AB, Lim W, Ferrari P, Witt CS, Christiansen FT. Pre-transplant donorspecific anti-HLA antibody is associated with antibody-mediated rejection, progres-sive graft dysfunction and patient death. Transpl Immunol Jun. 2013;28(4):148–53.

[28] O'Brien FJ, Lineen J, Kennedy CM, Phelan PJ, Kelly PO, Denton MD, et al. Effect ofperioperative blood transfusions on long term graft outcomes in renal transplantpatients. Clin Nephrol 2012;77:432–7.

[29] Scornik JC, Schold JD, Bucci M, Meier-Kriesche HU. Effects of blood transfusionsgiven after renal transplantation. Transplantation 2009;87:1381–6.

[30] Bushell A, Karim M, Kingsley CI, Wood KJ. Pretransplant blood transfusion with-out additional immunotherapy generates CD25 + CD4+ regulatory T cells: apotential explanation for the blood-transfusion effect. Transplantation 2003;76:449–55.

[31] Scornik JC, Ireland JE, Howard RJ, Pfaff WW, Fennell III RS. Sensitization by bloodtransfusions in previously transplanted patients. Transplantation 1983;35:505–6.

[32] Qian Z, Lee CY, Murata K, Liu J, Fox-Talbot K, Wasowska BA, et al. Antibody and com-plement mediated injury in transplants following sensitization by allogeneic bloodtransfusion. Transplantation 2006;82:857–64.

[33] Norda R, Schott U, Berseus O, Akerblom O, Nilsson B, Ekdahl KN, et al. Complementactivation products in liquid stored plasma and C3a kinetics after transfusion of au-tologous plasma. Vox Sang 2012;102:125–33.

[34] Pandey P, Chaudhary R, Aggarwal A, Kumar R, Khetan D, Verma A. Transfusion-associated immunomodulation: quantitative changes in cytokines as a measure ofimmune responsiveness after one time blood transfusion in neurosurgery patients.Asian J Transf Sci 2010;4:78–85.

[35] Strait RT, HicksW, Barasa N, Mahler A, KhodounM, Kohl J, et al. MHC class I-specificantibody binding to nonhematopoietic cells drives complement activation to inducetransfusion-related acute lung injury in mice. J Exp Med 2011;208:2525–44.

[36] Magee BA, Martin J, Cole MP, Morris KG, Courtney AE. Effects of HLA-matchedblood transfusion for patients awaiting renal transplantation. Transplantation2012;94:1111–6.

[37] Scornik JC. HLA-matched blood transfusions? Transplantation 2012;94:1085.[38] Sasaki H, Chikaraishi T, Furuhata S, Tsutsumi H, Miyano S, Nakazawa R, et al.

Autologous blood transfusion for kidney transplant recipients. Transplant Proc2008;40:1371–2.