• Aucun résultat trouvé

4. General discussion

N/A
N/A
Protected

Academic year: 2021

Partager "4. General discussion"

Copied!
64
0
0

Texte intégral

(1)

4. General discussion

4.1 Minor antigens in renal transplantation

Several years ago, a possible role of non-HLA immunity in allograft injury has been suggested after data from The Collaborative Transplant Study showed an unexpected negative effect of PRA on graft survival in HLA-identical sibling transplants (182). The effect of PRA was apparent only beyond one year post- transplantation and progressed slowly over time in that population in comparison with deceased donor patients who were subjected to earlier graft loss. A possible explanation was that antibodies to minor histocompatibility antigens occurred frequently with anti-HLA antibodies or cross-reacted with epitopes shared by HLA antibodies. However, the finding that a substantial proportion of sibling transplants failed even in the absence of any detectable antibody reactivity suggested that non-HLA immunity could contribute significantly to graft loss.

Antibodies to non-HLA antigens have been pointed out as likely to be involved in renal allograft rejection and graft loss throughout associative studies. The most abundant literature in that field undoubtedly relates to MICA antibodies followed by antibodies to AT1R and antibodies to vimentin or agrin. However, no experimental model was able to demonstrate at the present time a real pathogenicity of those antibodies, especially as a substantial proportion of them has been reported to be auto-antibodies and therefore, might be simple artefacts due to cross-reactivity with epitopes from infectious agents for example.

Since MICA had appeared in recent years as a non-HLA antigenic target in renal

transplantation, we decided to start a research in 2008 because little was actually

(2)

known in that field at that time, especially with regard to epidemiology, risk factors for sensitization and the autologuous or allogeneic nature of MICA antibodies. By a happy coincidence, we received support from a firm that aimed to market a new kit for identifying MICA antibodies by Luminex technology.

4.2 Epidemiology of MICA antibodies and risk factors for MICA sensitization Our work showed that MICA sensitization was 4-fold higher among CKD patients than in healthy volunteers. Unexpectedly, the prevalence of MICA antibodies was also found to be 2 fold higher among males as compared to females while we could identify pregnancy as an independent risk factor for MICA sensitization.

The origin of the influence of male gender on the presence of MICA antibodies has not been investigated yet but mechanisms controlling MICA expression might be related to sex. The other routes for MICA allo-immunisation were mainly those involved in HLA immunization: previous graft and blood transfusions. In addition, uremia “per se” also appeared as an independent risk factor for the development of MICA antibodies. Nevertheless, the presence of MICA antibodies among controls suggested that other possible routes for MICA sensitization might exist. A possible explanation might be cross-reactivity against epitopes shared by for example, both microbial agents and MICA antigens.

Since MICA is a cell-surface stress-inducible glycoprotein expressed on ECs, the high prevalence of MICA antibodies among CKD patients might be related to the

“chronic microinflammatory state” of such patients. This “microinflammatory

state” relates to an increased percentage of CD14+/CD16+ monocytes in

peripheral blood that promote endothelial injury through the release of pro-

(3)

inflammatory cytokines and to patient-related factors, such as underlying disease, comorbidity, oxidative stress, infectious, genetic and immunologic factors, as well as those arising from dialysis treatment itself, mainly membrane and dialysate biocompatibility (317). Indeed, chronically haemodialyzed patients were recently shown to exhibit a profound decrease in NKG2D bearing cells within both the CD8+ T cell and NK cell populations, and conversely, both increased membrane-bound MICA on monocytes and soluble MICA levels, possibly leading to the induction of MICA antibodies (318).

4.3 Impact of MICA antibodies on graft survival

Since 5 years approximately, several groups reported an association of MICA

antibodies with lower graft survival or rejection (290, 291, 300, 304). However,

those previous publications showed a crude univariate analysis of the

relationship between MICA antibodies and graft outcomes without investigating

the impact of possible confounding factors likely to influence graft loss. Until now,

no study in renal transplantation gave the formal proof of neither the

pathogenicity of such antibodies nor that those possibly deleterious antibodies

were DSA. Moreover, positive MICA reactivity had not been confirmed on MICA

expressing cell lines. Up to now, 2 studies tried to demonstrate the donor

specificity of MICA antibodies. The first was performed in heart transplant

recipients and it concluded to a lack of a deleterious impact of MICA antibodies

whether they were DSA or NDSA (294). The second aimed to address the effect

of MICA mismatching on the development of MICA antibodies but did not focus

on graft loss (306).

(4)

To date, how MICA antigens might be deleterious to the graft remains unclearly understood. Especially, the possibility that MICA and HLA antibodies could act in concert to allograft injury is not determined.

Unlike Zou’s and Terasaki’s studies, our findings showed a clear lack of effect of either pre- or post-transplant MICA antibodies on renal graft outcomes. This discrepancy might be explained by differences in immunosuppressive regimen given to patients. In our 2 studies, patients received a higher overall amount of immunosuppression, probably modulating the allo-immune response to the graft and therefore, benefiting graft outcome. Indeed, our patients received more frequently tacrolimus and MPA as maintenance therapy rather than cyclosporine and azathioprine in Zou’s and Terasaki’s reports. Likewise, induction therapy was also more prevalent among our patients. Moreover, authors do not give information about the possible discontinuation of the corticoids therapy in the post-transplant setting. In our cohort, 70% of patients still had corticoids at 1y post-transplantation. No data about the incidence, the severity of AR and recovery of renal graft function were reported in Zou’s and Terasaki’s studies while they may affect the graft survival (319).

4.4 How to provide evidence of a possible pathogenicity of MICA antibodies towards the renal graft?

Undoubtedly, to design a study enable to answer to the question whether MICA

antibodies are pathogenic will face the difficulty to clearly isolate the effect of

MICA antibodies on graft outcome as MICA and HLA sensitization are closely

linked. This link has been reflected in all the previous publications related on

(5)

MICA (see annexes). As reported in our second article, preliminary statistical calculations were performed in order to determine the sample size needed to detect a difference of death-censored graft survival of at least 5% 3 years after MICA testing considering a prevalence of MICA antibodies of about 10%, as showed by Terasaki (291). These analyses revealed that to avoid the possible confounding impact of anti-HLA antibodies on survival, at least 3500 patients should have been included to reach ± 170 patients who were HLA-/MICA+.

However, the prevalence of post-transplant MICA antibodies reached about 5%

in our cohort making it should have required 7000 patients to test our hypothesis.

Therefore, we considered such a study unrealistic because it would have involved huge logistics and financial support.

To design an animal model would also not be devoid of some pitfalls. A major one would be that mice lack MICA expression. Nevertheless, some might imagine creating a transgenic mice model since the coding regions for the human MICA gene are known and could be cloned. Kidneys from transgenic mice could be transplanted in wild type mice previously immunized with recombinant MICA antigens and parameters such as albuminuria/creatininuria ratio in daily urine samples could be screened until sacrifice.

Another mean, but expensive, to study the relationship between MICA antibodies

and graft failure could be the use of a non-human primate model. For example,

monkey donor and recipient pairs should be selected for ABO and HLA

compatibility but MICA mismatching. Recipients should be submitted to

chimerism protocols such as nonlethal total body irradiation, local thymic

(6)

irradiation followed by the infusion of ATG and intravenous donor bone marrow transplantation before heterotopic renal transplantation and bilateral native binephrectomy. For example, cyclosprorine administered at day 0 would be progressively tapered to finally be discontinued. Serum samples would be tested for anti-donor MICA antibodies, anti-HLA antibodies and creatinine. Renal graft biopsies would be performed at regular intervals when creatinine is stable and in addition, when creatinine is rising. We might follow the development of MICA antibodies in monkeys that remained negative for anti-HLA antibodies and the appearance of allograft injury.

Just imagine money is not an obstacle and we have huge logistic means such as a large staff and platforms dedicated to research and highly sophisticated equipment, we could set up a prospective study through a large collaborative network that would allow for the recruitment of a huge amount of patients with available clinical and histological data. Patients should be donor HLA full- matched and mismatched for MICA with no pre-transplant HLA or MICA antibodies detected by single antigen flow beads assays. They should receive a standardized induction and immunosuppressive therapy with clear protocols for reduction of immunosuppression, especially in cases of infection or cancer.

Patients should undergo routine blood tests every 3 months to assess HLA and

MICA status and renal graft biopsies at Day 0, Month 3, Month 6 and yearly

thereafter, until 10 yr post-transplantation. Only the patients who never

developed HLA antibodies would be kept for analysis. Sera positive for MICA

(7)

antibodies should be controlled for cytotoxicity on cell lines expressing a single MICA antigen.

To increase the level of evidence for the pathogenicity of MICA antibodies, the following criteria should be present:

i) Good function over time should be associated with patients who do not develop MICA antibodies

ii) The appearance of MICA antibodies should precede immune-mediated graft injury

iii) MICA antibodies should be confirmed as the strongest independent risk factor for immunological graft loss in an appropriate logistic regression model

iv) MICA+ patients whose graft failed from immune-mediated injury compared with MICA+ patients who lost their graft from a non-immunological cause or kept a functioning graft should be confirmed to be DSA

v) MICA+ patients whose graft failed from immune-mediated injury compared with MICA+ patients who lost their graft from a non-immunological cause or kept a functioning graft should have high MICA antibodies titers (dose response effect)

vi) MICA+ patients whose graft failed from immune-mediated injury compared with MICA+ patients who lost their graft from a non-immunological cause should have intra-graft MICA antibodies

The ultimate evidence of the pathogenicity of MICA antibodies should be the

demonstration that eliminating de novo antibodies would result in long-term

survival.

(8)

In addition, we may assume that probably MICA antibodies do not act alone but rather exert their effect in concert with antibodies to other minor antigens unexplored so far.

Therefore, another way that should be explored to better understand the possible pathogenic effect of MICA antibodies should be the use of proteogenomics. This approach combines proteomics and genomics. Proteomics is the systematic analysis by chromatography and mass spectrometry of all proteins encoded by the genome in any defined biologic compartment and genomics involves microarrays, which determines gene expression profile (mRNA transcripts) correlated to a corresponding level of protein or other available genomic techniques evolved in recent years such as SNP arrays detecting SNPs of human genome, alterations in gene copy number or alternative RNA splicing.

The proteomic approach has already been used to screen non-HLA antibody response in patients undergoing chronic haemodialysis and awaiting a kidney transplant, to identify compartment-specific non-HLA targets after renal transplantation and recently, to predict the development of chronic allograft injury (320, 321).

The proteogenomic approach might be used in order to better understand the

possible role of minor antigens in rejection. A large multi-centre study could be

set up to compare patterns of proteins and antibodies in both serum and graft

between patients with histological features of humoral AR or CR and patients

with normal biopsy. In the event that a specific pattern emerged, it should be

firstly confirmed in a validation sample set and the second step would be to

(9)

identify polymorphisms or aberrations in the genes that encode the selected proteins or alterations in the translation of RNA transcripts. However, the study design would require that patients are matched for donor and recipient demographic characteristics and immunosuppression. Such studies would be very expensive and time consuming requiring high throughput technology and advanced bioinformatics analyses for an uncertain expected benefit.

Until now, proteomics has been used in small patient cohort size with most of the time, the lack of data validation from an independent patient sample set.

Moreover, none of candidate biomarkers identified by proteomics has emerged so far as a reliable clinical tool for the diagnosis of AR or CR (322-324). This seems to be related to the inconsistency in sample handling, and the lack of standardization of experimental design leading to the identification of different discriminatory proteins by different groups. Current proteomic studies suggest that a single biomarker does not provide sufficient specificity to distinguish between diseased and healthy status. Therefore, robust study design with appropriate statistical power, blinding and validation set is crucial to improve the reliability of proteogenomic-driven results, making possible the identification of a panel of candidate markers that will form a signature pattern with sufficient diagnostic performance and good predictive value.

Although our findings suggest that MICA antibodies do not affect renal graft

outcomes, we cannot exclude that high titer and/or cytotoxic MICA antibodies

might be deleterious in rare patients.

(10)

Therefore, the question is whether it would be reasonable to incur significant financial and human resources to resolve an issue that concerns finally a low number of patients for an uncertain expected benefit.

Of interest, it is also unclear how long MICA antibodies persist over time.

Terasaki and colleagues reported that the frequency of post-transplant MICA antibodies was stable along with time (291). However, nearly two thirds of patients who were sensitized against MICA before transplantation had no more MICA antibodies at 1yr post-transplantation in our cohort (data not shown). This might suggest that MICA antibodies could be absorbed by the graft or MICA sensitization might be a fluctuating phenomenon. Likewise, in our paper dealing with the safety of Influenza A/H1N1 vaccine, MICA antibodies were seen to disappear one month after a positive screening in several kidney transplant patients and in one haemodialysis patient, advocating for the transitory nature of these antibodies. We strongly believe that MICA immunisation is highly influenced by environmental stress and also that immunosuppression modulates the development of MICA antibodies.

4.5 Influenza A/H1N1 vaccination and MICA sensitization

Our research showed that Influenza A/H1N1-adjuvanted vaccine did not trigger either an increased production of anti-HLA or MICA antibodies nor induced AR or a deterioration of renal graft function up to 6 months after vaccination.

Possible MICA sensitization after Influenza A/H1N1 vaccination has also been

recently addressed in cystic fibrosis patients and in lung transplant recipients in

our institution. Analysis of the data confirmed that again transplanted patients

(11)

reached lower antibody titres against Influenza A/H1N and had an inferior seroconversion rate compared with cystic fibrosis patients and controls.

Only one lung transplant recipient developed de novo HLA antibodies. No patient produced de novo MICA antibodies or experienced accelerated graft loss.

5. Summary

MICA antibodies are not rare in renal transplantation. Their prevalence is reaching 14% in stage V CKD patients and falls to 5% 1 year after transplantation, possibly due to the modulation of antibody production by immunosuppression.

In the present work, we identified uremia, previous graft, blood transfusion and pregnancy as independent risk factors for MICA allo-sensitization. However, up to 20% of MICA antibodies may also be auto-antibodies, possibly because of cross-reactivity against epitopes shared by both microbial agents and MICA antigens.

Either MICA antibodies had been detected before or after transplantation, our findings suggest a lack of meaningful clinical relevance in renal transplantation.

Rather than pathogenic, MICA antibodies might be simply stress-induced

surrogate markers of high immunological risk, making interest for pre-transplant

screening or post-transplant monitoring by current flow beads assays highly

questionable.

(12)

6. Perspectives

The big challenge of renal transplantation is to identify and to validate new pre- and post-transplant biomarkers using patient serum, urines or DNA in order to facilitate personalized transplantation medicine and customization of immunosuppression.

Indeed, the better risk assessment of graft loss in kidney transplant recipients might allow for tailored immunosuppression. This strategy might decrease the risk of AR or CR in patients labelled “at high risk” on the one hand and to avoid immunosuppression-related complications such as renal toxicity, infections and cancers in low risk patients on the other hand. Moreover, the identification of biomarkers might aid the understanding of underlying mechanisms by indicating damage early after transplantation when pathological changes are taking place at the molecular level. This will enable physicians to better predict the likelihood of an individual’s allograft survival and to assist the development of new target drugs. Finally, biomarkers might also allow better matching of donor and recipient and the assessment of an individual’s risk for graft injury. Indeed, while current methods for diagnosing allograft injury require invasive allograft biopsies and detect pathological changes at advanced and often irreversible stages of allograft damage, the use of more sensitive and specific methodologies based on recipient and donor genotyping or transcriptional and proteomic profiling might bridge this gap.

Nowadays, the ongoing biomarker studies increasingly integrate data from

multiple platforms, such as genotype analyses of single nucleotide

(13)

polymorphisms, epigenetic studies and analyses of mRNA, miRNA, as well as protein, peptide, antibody and metabolite profiling.

However, the path from discovery and validation of a biomarker towards its approval by clinicians is tortuous. Indeed, the phase of discovery and validation requires high-throughput technologies on multiple molecular platforms and thus, large cohort, large infrastructure, huge logistics means and financial support.

Moreover, replication cohorts are often needed to increase the sensitivity and specificity of the biomarker. Quality control tools have also to be implemented to ensure the comparability of data from different laboratories. Another important step towards confirming the clinic usefulness of a biomarker is to identify and control for experimental confounders including technology bias or patient bias.

At the present time, there is still a long way to go before the identification of biomarkers with high specificity for a defined “disease status” and good predictive value for diagnosis enabling clinicians safely to tailor immunosuppression.

Therefore, it looks that promoting translational research in transplantation units

should be a central concern for clinicians not only to address this issue but also

simply to have a feedback on their own daily practice and to ensure high quality

patient care.

(14)

7. References

1. Ojo AO, Hanson JA, Meier-Kriesche H, Okechukwu CN, Wolfe RA, Leichtman AB, Agodoa LY, Kaplan B, Port FK. Survival in recipients of marginal cadaveric donor kidneys compared with other recipients and wait-listed transplant candidates. J Am Soc Nephrol 2001; 12(3):589-97.

2. Habwe VQ. Posttransplantation quality of life: more than graft function. Am J Kidney Dis 2006; 47(4 Suppl 2):S98-110.

3. Meier-Kriesche HU, Schold JD, Kaplan B. Long-term renal allograft survival:have we made significant progress or is it time to rethink our analytic and therapeutic strategies?

Am J Transplant 2004; 4(8):1289-95.

4. Collins AJ, Foley R, Herzog C, Chavers B, Gilbertson D, Ishani A, Kasiske B, Liu J, Mau LW, McBean M, Murray A, St Peter W, Xue J, Fan Q, Guo H, Li Q, Li S, Li S, Peng Y, Qiu Y, Roberts T, Skeans M, Snyder J, Solid C, Wang C, Weinhandl E, Zaun D, Zhang R, Arko C, Chen SC, Dalleska F, Daniels F, Dunning S, Ebben J, Frazier E, Hanzlik C, Johnson R, Sheets D, Wang X, Forrest B, Constantini E, Everson S, Eggers P, Agodoa L. Excerpts from the United States Renal Data System 2007 annual data report. Am J Kidney Dis 2008; 51(1 Suppl 1):S1-320.

5. Opelz G, Döhler B; Collaborative Transplant Study. Influence of immunosuppressive regimens on graft survival and secondary outcomes after kidney transplantation.

Transplantation 2009; 87(6):795-802

6. Gill JS, Rose C, Pereira BJ, Tonelli M. The importance of transitions between dialysis and transplantation in the care of end-stage renal disease patients. Kidney Int 2007;

71(5):442-7.

7. Cosio FG, Hickson LJ, Griffin MD, Stegall MD, Kudva Y. Patient survival and cardiovascular risk after kidney transplantation: the challenge of diabetes. Am J Transplant 2008; 8(3):593-9.

(15)

8. Meier-Kriesche HU, Baliga R, Kaplan B. Decreased renal function is a strong risk factor for cardiovascular death after renal transplantation. Transplantation 2003;

75(8):1291-5.

9. El-Zoghby ZM, Stegall MD, Lager DJ, Kremers WK, Amer H, Gloor JM, Cosio FG.

Identifying specific causes of kidney allograft loss. Am J Transplant 2009; 9(3):527-35.

10. Segoloni GP, Messina M, Giraudi R, Leonardi G, Torta E, Gabrielli D, Ferrari A, Pellu V, Tattoli F, Fop F. Renal transplantation in patients over 65 years of age: no more a contraindication but a growing indication. Transplant Proc 2005; 37(2):721-5.

11. Briganti EM, Russ GR, McNeil JJ, Atkins RC, Chadban SJ. Risk of renal allograft loss from recurrent glomerulonephritis. N Engl J Med 2002; 347(2):103-9.

12. Matas AJ. Recurrent disease after kidney transplantation--it is time to unite to address this problem! Am J Transplant 2006; 6(11):2527-8.

13. Hariharan S, Adams MB, Brennan DC, Davis CL, First MR, Johnson CP, Ouseph R, Peddi VR, Pelz CJ, Roza AM, Vincenti F, George V. Recurrent and de novo glomerular disease after renal transplantation: a report from Renal Allograft Disease Registry (RADR). Transplantation 1999; 68(5):635-41.

14. Schweitzer EJ, Matas AJ, Gillingham KJ, Payne WD, Gores PF, Dunn DL, Sutherland DE, Najarian JS. Causes of renal allograft loss. Progress in the 1980s, challenges for the 1990s. Ann Surg 1991; 214(6):679-88.

15. Matas AJ, Humar A, Gillingham KJ, Payne WD, Gruessner RW, Kandaswamy R, Dunn DL, Najarian JS, Sutherland DE. Five preventable causes of kidney graft loss in the 1990s: a single-center analysis. Kidney Int 2002; 62(2):704-14.

16. Solez K, Axelsen RA, Benediktsson H, Burdick JF, Cohen AH, Colvin RB, Croker BP, Droz D, Dunnill MS, Halloran PF, et al. International standardization of criteria for the histologic diagnosis of renal allograft rejection: the Banff working classification of kidney transplant pathology. Kidney Int 1993; 44(2):411-22.

(16)

17. Solez K, Colvin RB, Racusen LC, Sis B, Halloran PF, Birk PE, Campbell PM, Cascalho M, Collins AB, Demetris AJ, Drachenberg CB, Gibson IW, Grimm PC, Haas M, Lerut E, Liapis H, Mannon RB, Marcus PB, Mengel M, Mihatsch MJ, Nankivell BJ, Nickeleit V, Papadimitriou JC, Platt JL, Randhawa P, Roberts I, Salinas-Madriga L, Salomon DR, Seron D, Sheaff M, Weening JJ. Banff '05 Meeting Report: differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy ('CAN'). Am J Transplant 2007; 7(3):518-26.

18. Sellarés J, de Freitas DG, Mengel M, Reeve J, Einecke G, Sis B, Hidalgo LG, Famulski K, Matas A, Halloran PF. Understanding the Causes of Kidney Transplant Failure: The Dominant Role of Antibody-Mediated Rejection and Nonadherence. Am J Transplant 2011 [Epub ahead of print]

19. Einecke G, Sis B, Reeve J, Mengel M, Campbell PM, Hidalgo LG, Kaplan B, Halloran PF. Antibody-mediated microcirculation injury is the major cause of late kidney transplant failure. Am J Transplant 2009; 9(11):2520-31.

20. Safinia N, Afzali B, Atalar K, Lombardi G, Lechler RI. T-cell alloimmunity and chronic allograft dysfunction. Kidney Int Suppl 2010; (119):S2-12.

21. Pietra BA, Wiseman A, Bolwerk A, Rizeq M, Gill RG. CD4 T cell-mediated cardiac allograft rejection requires donor but not host MHC class II. J Clin Invest 2000;

106(8):1003- 10.

22. Baker RJ, Hernandez-Fuentes MP, Brookes PA, Chaudhry AN, Lechler RI. The role of the allograft in the induction of donor-specific T cell hyporesponsiveness.

Transplantation 2001; 72(3):480-5.

23. Rogers NJ, Lechler RI. Allorecognition. Am J Transplant 2001; 1(2):97-102.

24. Crispe IN, Husmann LA, Bevan MJ. T cell receptor expression and receptor- mediated induction of clonal growth in the developing mouse thymus. High surface beta-

(17)

chain density is a requirement for functional maturity. Eur J Immunol 1986; 16(10):1283- 8.

25. Matzinger P, Bevan MJ. Hypothesis: why do so many lymphocytes respond to major histocompatibility antigens? Cell Immunol 1977; 29(1):1-5.

26. Valujskikh A, Lantz O, Celli S, Matzinger P, Heeger PS. Cross-primed CD8(+) T cells mediate graft rejection via a distinct effector pathway. Nat Immunol 2002; 3(9):844-51.

27. Steele DJ, Laufer TM, Smiley ST, Ando Y, Grusby MJ, Glimcher LH, Auchincloss H Jr. Two levels of help for B cell alloantibody production. J Exp Med 1996; 183(2):699- 703.

28. Taylor AL, Negus SL, Negus M, Bolton EM, Bradley JA, Pettigrew GJ. Pathways of helper CD4 T cell allorecognition in generating alloantibody and CD8 T cell alloimmunity.

Transplantation 2007; 83(7):931-7.

29. Sayegh MH, Perico N, Gallon L, Imberti O, Hancock WW, Remuzzi G, Carpenter CB. Mechanisms of acquired thymic unresponsiveness to renal allografts. Thymic recognition of immunodominant allo-MHC peptides induces peripheral T cell anergy.

Transplantation 1994; 58(2):125-32.

30. Herrera OB, Golshayan D, Tibbott R, Salcido Ochoa F, James MJ, Marelli-Berg FM, Lechler RI. A novel pathway of alloantigen presentation by dendritic cells. J Immunol 2004; 173(8):4828-37.

31. Game DS, Rogers NJ, Lechler RI. Acquisition of HLA-DR and costimulatory molecules by T cells from allogeneic antigen presenting cells. Am J Transplant 2005;

5(7):1614-25.

32. Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Papworth GD, Zahorchak AF, Logar AJ, Wang Z, Watkins SC, Falo LD Jr, Thomson AW. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 2004;

104(10):3257-66.

(18)

33. Mandelbrot DA, Oosterwegel MA, Shimizu K, Yamada A, Freeman GJ, Mitchell RN, Sayegh MH, Sharpe AH. B7-dependent T-cell costimulation in mice lacking CD28 and CTLA4. J Clin Invest 2001; 107(7):881-7.

34. Sandner SE, Salama AD, Houser SL, Palmer E, Turka LA, Sayegh MH. New TCR transgenic model for tracking allospecific CD4 T-cell activation and tolerance in vivo. Am J Transplant 2003; 3(10):1242-50.

35. Reed AJ, Noorchashm H, Rostami SY, Zarrabi Y, Perate AR, Jeganathan AN, Caton AJ, Naji A. Alloreactive CD4 T cell activation in vivo: an autonomous function of the indirect pathway of alloantigen presentation. J Immunol. 2003; 171(12):6502-9.

36. Li XC, Zheng XX, Strom TB. T-cell growth factors in allograft rejection and tolerance.

Transplant Proc 1999; 31(1-2):342-3.

37. Li XC, Ima A, Li Y, Zheng XX, Malek TR, Strom TB. Blocking the common gamma- chain of cytokine receptors induces T cell apoptosis and long-term islet allograft survival.

J Immunol 2000; 164(3):1193-9.

38. Smith XG, Bolton EM, Ruchatz H, Wei X, Liew FY, Bradley JA. Selective blockade of IL-15 by soluble IL-15 receptor alpha-chain enhances cardiac allograft survival. J Immunol 2000; 165(6):3444-50.

39. Krieger NR, Yin DP, Fathman CG. CD4+ but not CD8+ cells are essential for allorejection. J Exp Med 1996; 184(5):2013-8.

40. Amsen D, Spilianakis CG, Flavell RA. How are T(H)1 and T(H)2 effector cells made?

Curr Opin Immunol 2009; 21(2):153-60.

41. O'Connell PJ, Pacheco-Silva A, Nickerson PW, Muggia RA, Bastos M, Kelley VR, Strom TB. Unmodified pancreatic islet allograft rejection results in the preferential expression of certain T cell activation transcripts. J Immunol 1993; 150(3):1093-104.

42. Nickerson P, Steurer W, Steiger J, Zheng X, Steele AW, Strom TB. Cytokines and the Th1/Th2 paradigm in transplantation. Curr Opin Immunol1994; 6(5):757-64.

(19)

43. Miura M, El-Sawy T, Fairchild RL. Neutrophils mediate parenchymal tissue necrosis and accelerate the rejection of complete major histocompatibility complex-disparate cardiac allografts in the absence of interferon-gamma. Am J Pathol 2003; 162(2):509-19.

44. Gras J, Wieërs G, Vaerman JL, Truong DQ, Sokal E, Otte JB, Délépaut B, Cornet A, de Ville de Goyet J, Latinne D, Reding R. Early immunological monitoring after pediatric liver transplantation: cytokine immune deviation and graft acceptance in 40 recipients.

Liver Transpl 2007; 13(3):426-33.

45. Sadeghi M, Daniel V, Naujokat C, Schmidt J, Mehrabi A, Zeier M, Opelz G. Evidence for IFN-gamma up- and IL-4 downregulation late post-transplant in patients with good kidney graft outcome. Clin Transplant 2007; 21(4):449-59.

46. Tay SS, Plain KM, Bishop GA. Role of IL-4 and Th2 responses in allograft rejection and tolerance. Curr Opin Organ Transplant 2009; 14(1):16-22.

47. Hochrein H, O'Keeffe M, Luft T, Vandenabeele S, Grumont RJ, Maraskovsky E, Shortman K. Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 production by mouse and human dendritic cells. J Exp Med. 2000; 192(6):823-33.

48. Kaneko K, Wang Z, Kim SH, Morelli AE, Robbins PD, Thomson AW. Dendritic cells genetically engineered to express IL-4 exhibit enhanced IL-12p70 production in response to CD40 ligation and accelerate organ allograft rejection. Gene Ther 2003;

10(2):143-52.

49. Le Moine A, Flamand V, Demoor FX, Noël JC, Surquin M, Kiss R, Nahori MA, Pretolani M, Goldman M, Abramowicz D. Critical roles for IL-4, IL-5, and eosinophils in chronic skin allograft rejection. J Clin Invest 1999; 103(12):1659-67.

50. Braun MY, Desalle F, Le Moine A, Pretolani M, Matthys P, Kiss R, Goldman M. IL-5 and eosinophils mediate the rejection of fully histoincompatible vascularized cardiac allografts: regulatory role of alloreactive CD8(+) T lymphocytes and IFN-gamma. Eur J Immunol 2000; 30(5):1290-6.

(20)

51. Poulin LF, Richard M, Le Moine A, Kiss R, McKenzie AN, Goldman M, Renauld JC, Van Snick J, Braun MY. Interleukin-9 promotes eosinophilic rejection of mouse heart allografts. Transplantation 2003; 76(3):572-7.

52. Bettelli E, Korn T, Kuchroo VK. Th17: the third member of the effector T cell trilogy.

Curr Opin Immunol 2007; 19(6):652-7.

53. Mitsdoerffer M, Lee Y, Jäger A, Kim HJ, Korn T, Kolls JK, Cantor H, Bettelli E, Kuchroo VK. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci U S A 2010; 107(32):14292-7.

54. Yuan X, Paez-Cortez J, Schmitt-Knosalla I, D'Addio F, Mfarrej B, Donnarumma M, Habicht A, Clarkson MR, Iacomini J, Glimcher LH, Sayegh MH, Ansari MJ. A novel role of CD4 Th17 cells in mediating cardiac allograft rejection and vasculopathy. J Exp Med 2008; 205(13):3133-44.

55. Yuan X, Ansari MJ, D'Addio F, Paez-Cortez J, Schmitt I, Donnarumma M, Boenisch O, Zhao X, Popoola J, Clarkson MR, Yagita H, Akiba H, Freeman GJ, Iacomini J, Turka LA, Glimcher LH, Sayegh MH. Targeting Tim-1 to overcome resistance to transplantation tolerance mediated by CD8 T17 cells. Proc Natl Acad Sci U S A 2009;

106(26):10734-9.

56. Vokaer B, Van Rompaey N, Lemaître PH, Lhommé F, Kubjak C, Benghiat FS, Iwakura Y, Petein M, Field KA, Goldman M, Le Moine A, Charbonnier LM. Critical role of regulatory T cells in Th17-mediated minor antigen-disparate rejection. J Immunol 2010;

185(6): 3417-25.

57. Min SI, Ha J, Park CG, Won JK, Park YJ, Min SK, Kim SJ. Sequential evolution of IL- 17 responses in the early period of allograft rejection. Exp Mol Med 2009; 41(10):707-16.

58. el-Sawy T, Fahmy NM, Fairchild RL. Chemokines: directing leukocyte infiltration into allografts. Curr Opin Immunol 2002; 14(5):562-8.

(21)

59. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration:

the multistep paradigm. Cell 1994; 76(2):301-14.

60. Le Moine A, Goldman M, Abramowicz D. Multiple pathways to allograft rejection.

Transplantation 2002; 73(9):1373-81.

61. Ito A, Shimura H, Nitahara A, Tomiyama K, Ito M, Kanekura T, Okumura K, Yagita H, Kawai K. NK cells contribute to the skin graft rejection promoted by CD4+ T cells activated through the indirect allorecognition pathway. Int Immunol 2008; 20(10):1343-9.

62. Hidalgo LG, Sis B, Sellares J, Campbell PM, Mengel M, Einecke G, Chang J, Halloran PF. NK cell transcripts and NK cells in kidney biopsies from patients with donor- specific antibodies: evidence for NK cell involvement in antibody-mediated rejection. Am J Transplant 2010; 10(8):1812-22.

63. Sun K, Alvarez M, Ames E, Barao I, Chen M, Longo DL, Redelman D, Murphy WJ.

Mouse NK cell-mediated rejection of bone marrow allografts exhibits patterns consistent with Ly49 subset licensing. Blood 2011 Dec 19. [Epub ahead of print]

64. Kroemer A, Edtinger K, Li XC. The innate natural killer cells in transplant rejection and tolerance induction. Curr Opin Organ Transplant 2008; 13(4):339-43.

65. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol 2008; 9(5):503-10.

66. Hirohashi T, Chase CM, Della Pelle P, Sebastian D, Alessandrini A, Madsen JC, Russell PS, Colvin RB. A novel pathway of chronic allograft rejection mediated by NK cells and alloantibody. Am J Transplant 2012; 12(2):313-21.

67. Lanier LL. NK cell recognition. Annu Rev Immunol 2005; 23:225-74.

68. Walzer T, Jaeger S, Chaix J, Vivier E. Natural killer cells: from CD3(-)NKp46(+) to post-genomics meta-analyses. Curr Opin Immunol 2007; 19(3):365-72.

69. Rabinovich BA, Shannon J, Su RC, Miller RG. Stress renders T cell blasts sensitive to killing by activated syngeneic NK cells. J Immunol 2000; 165(5):2390-7.

(22)

70. Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, Caligiuri MA.

Evidence for discrete stages of human natural killer cell differentiation in vivo. J Exp Med 2006; 203(4):1033-43.

71. Kroemer A, Xiao X, Degauque N, Edtinger K, Wei H, Demirci G, Li XC. The innate NK cells, allograft rejection, and a key role for IL-15. J Immunol 2008; 180(12):7818-26.

72. Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease.

Blood 2001; 97(1):14-32.

73. Gill RG. NK cells: elusive participants in transplantation immunity and tolerance. Curr Opin Immunol 2010; 22(5):649-54.

74. Kopf M, Brombacher F, Hodgkin PD, Ramsay AJ, Milbourne EA, Dai WJ, Ovington KS, Behm CA, Köhler G, Young IG, Matthaei KI. IL-5-deficient mice have a developmental defect in CD5+ B-1 cells and lack eosinophilia but have normal antibody and cytotoxic T cell responses. Immunity 1996; 4(1):15-24.

75. Mattes J, Foster PS. Regulation of eosinophil migration and Th2 cell function by IL-5 and eotaxin. Curr Drug Targets Inflamm Allergy 2003; 2(2):169-74.

76. Fukushi J, Ono M, Morikawa W, Iwamoto Y, Kuwano M. The activity of soluble VCAM-1 in angiogenesis stimulated by IL-4 and IL-13. J Immunol 2000; 165(5):2818-23.

77. Doucet C, Brouty-Boyé D, Pottin-Clemenceau C, Jasmin C, Canonica GW, Azzarone B. IL-4 and IL-13 specifically increase adhesion molecule and inflammatory cytokine expression in human lung fibroblasts. Int Immunol 1998; 10(10):1421-33.

78. Pretolani M, Ruffié C, Lapa e Silva JR, Joseph D, Lobb RR, Vargaftig BB. Antibody to very late activation antigen 4 prevents antigen-induced bronchial hyperreactivity and cellular infiltration in the guinea pig airways. J Exp Med 1994; 180(3):795-805.

79. Odaka M, Matsukura S, Kuga H, Kokubu F, Kasama T, Kurokawa M, Kawaguchi M, Ieki K, Suzuki S, Watanabe S, Homma T, Takeuchi H, Nohtomi K, Schleimer RP, Adachi M. Differential regulation of chemokine expression by Th1 and Th2 cytokines and

(23)

mechanisms of eotaxin/CCL-11 expression in human airway smooth muscle cells. Int Arch Allergy Immunol 2007; 143 Suppl 1: 84-8.

80. Matsukura S, Stellato C, Plitt JR, Bickel C, Miura K, Georas SN, Casolaro V, Schleimer RP. Activation of eotaxin gene transcription by NF-kappa B and STAT6 in human airway epithelial cells. J Immunol 1999; 163(12):6876-83.

81. Gutierrez-Ramos JC, Lloyd C, Gonzalo JA. Eotaxin: from an eosinophilic chemokine to a major regulator of allergic reactions. Immunol Today 1999; 20(11):500-4.

82. Li L, Xia Y, Nguyen A, Lai YH, Feng L, Mosmann TR, Lo D. Effects of Th2 cytokines on chemokine expression in the lung: IL-13 potently induces eotaxin expression by airway epithelial cells. J Immunol 1999; 162(5):2477-87.

83. Collins PD, Marleau S, Griffiths-Johnson DA, Jose PJ, Williams TJ. Cooperation between interleukin-5 and the chemokine eotaxin to induce eosinophil accumulation in vivo. J Exp Med 1995; 182(4):1169-74.

84. Stein ML, Villanueva JM, Buckmeier BK, Yamada Y, Filipovich AH, Assa'ad AH, Rothenberg ME. Anti-IL-5 (mepolizumab) therapy reduces eosinophil activation ex vivo and increases IL-5 and IL-5 receptor levels. J Allergy Clin Immunol 2008; 121(6):1473- 83.

85. Assa'ad AH, Spicer RL, Nelson DP, Zimmermann N, Rothenberg ME.

Hypereosinophilic syndromes. Chem Immunol 2000; 76:208-29.

86. Wu W, Samoszuk MK, Comhair SA, Thomassen MJ, Farver CF, Dweik RA, Kavuru MS, Erzurum SC, Hazen SL. Eosinophils generate brominating oxidants in allergen- induced asthma. J Clin Invest 2000; 105(10):1455-63

87. Liles WC, Kiener PA, Ledbetter JA, Aruffo A, Klebanoff SJ. Differential expression of Fas (CD95) and Fas ligand on normal human phagocytes: implications for the regulation of apoptosis in neutrophils. J Exp Med 1996; 184(2):429-40.

(24)

88. Wagner C, Iking-Konert C, Denefleh B, Stegmaier S, Hug F, Hänsch GM. Granzyme B and perforin: constitutive expression in human polymorphonuclear neutrophils. Blood 2004; 103(3):1099-104.

89. Yapici Ü, Kers J, Bemelman FJ, Roelofs JJ, Groothoff JW, van der Loos CM, van Donselaar-van der Pant KA, Idu MM, Claessen N, ten Berge IJ, Florquin S. Interleukin- 17 positive cells accumulate in renal allografts during acute rejection and are independent predictors of worse graft outcome. Transpl Int 2011; 24(10):1008-17.

90. Van Kooten C, Boonstra JG, Paape ME, Fossiez F, Banchereau J, Lebecque S, Bruijn JA, De Fijter JW, Van Es LA, Daha MR. Interleukin-17 activates human renal epithelial cells in vitro and is expressed during renal allograft rejection. J Am Soc Nephrol 1998; 9(8):1526-34.

91. Bennouna S, Bliss SK, Curiel TJ, Denkers EY. Cross-talk in the innate immune system: neutrophils instruct recruitment and activation of dendritic cells during microbial infection. J Immunol 2003; 171(11):6052-8.

92. Potter NS, Harding CV. Neutrophils process exogenous bacteria via an alternate class I MHC processing pathway for presentation of peptides to T lymphocytes. J Immunol 2001; 167(5):2538-46.

93. Buonocore S, Paulart F, Le Moine A, Braun M, Salmon I, Van Meirvenne S, Thielemans K, Goldman M, Flamand V. Dendritic cells overexpressing CD95 (Fas) ligand elicit vigorous allospecific T-cell responses in vivo. Blood 2003; 101(4):1469-76.

94. Wehner J, Morrell CN, Reynolds T, Rodriguez ER, Baldwin WM 3rd. Antibody and complement in transplant vasculopathy. Circ Res 2007; 100(2):191-203.

95. Zhang X, Reed EF. Effect of antibodies on endothelium. Am J Transplant 2009;

9(11):2459-65.

(25)

96. Baldwin WM 3rd, Valujskikh A, Fairchild RL. Antibody-mediated rejection:

emergence of animal models to answer clinical questions. Am J Transplant 2010;

10(5):1135-42.

97. Benzaquen LR, Nicholson-Weller A, Halperin JA. Terminal complement proteins C5b-9 release basic fibroblast growth factor and platelet-derived growth factor from endothelial cells. J Exp Med 1994; 179(3):985-92.

98. Saadi S, Platt JL. Humoral rejection and endothelial cell activation, 2001.

Xenotransplantation 2002; 9(4):239-41.

99. Albrecht EA, Chinnaiyan AM, Varambally S, Kumar-Sinha C, Barrette TR, Sarma JV, Ward PA. C5a-induced gene expression in human umbilical vein endothelial cells. Am J Pathol 2004; 164(3):849-59.

100. Ikeda K, Nagasawa K, Horiuchi T, Tsuru T, Nishizaka H, Niho Y. C5a induces tissue factor activity on endothelial cells. Thromb Haemost 1997; 77(2):394-8.

101. Saadi S, Holzknecht RA, Patte CP, Stern DM, Platt JL. Complement-mediated regulation of tissue factor activity in endothelium. J Exp Med 1995; 182(6):1807-14.

102. Nakashima S, Qian Z, Rahimi S, Wasowska BA, Baldwin WM 3rd. Membrane attack complex contributes to destruction of vascular integrity in acute lung allograft rejection. J Immunol 2002; 169(8):4620-7.

103. Baldwin WM 3rd, Larsen CP, Fairchild RL. Innate immune responses to transplants: a significant variable with cadaver donors. Immunity 2001; 14(4):369-76.

104. Jindra PT, Zhang X, Mulder A, Claas F, Veale J, Jin YP, Reed EF. Anti-HLA antibodies can induce endothelial cell survival or proliferation depending on their concentration. Transplantation 2006; 82(1 Suppl):S33-5.

105. Jin YP, Korin Y, Zhang X, Jindra PT, Rozengurt E, Reed EF. RNA interference elucidates the role of focal adhesion kinase in HLA class I-mediated focal adhesion

(26)

complex formation and proliferation in human endothelial cells. J Immunol 2007;178(12):7911-22.

106. Jindra PT, Jin YP, Rozengurt E, Reed EF. HLA class I antibody-mediated endothelial cell proliferation via the mTOR pathway. J Immunol 2008; 180(4):2357-66.

107. Fukami N, Ramachandran S, Saini D, Walter M, Chapman W, Patterson GA, Mohanakumar T. Antibodies to MHC class I induce autoimmunity: role in the pathogenesis of chronic rejection. J Immunol 2009; 182(1):309-18.

108. Yamakuchi M, Kirkiles-Smith NC, Ferlito M, Cameron SJ, Bao C, Fox-Talbot K, Wasowska BA, Baldwin WM 3rd, Pober JS, Lowenstein CJ. Antibody to human leukocyte antigen triggers endothelial exocytosis. Proc Natl Acad Sci U S A 2007;

104(4):1301-6.

109. Lee CY, Lotfi-Emran S, Erdinc M, Murata K, Velidedeoglu E, Fox-Talbot K, Liu J, Garyu J, Baldwin WM 3rd, Wasowska BA. The involvement of FcR mechanisms in antibody-mediated rejection. Transplantation 2007; 84(10):1324-34.

110. Jurcevic S, Ainsworth ME, Pomerance A, Smith JD, Robinson DR, Dunn MJ, Yacoub MH, Rose ML. Antivimentin antibodies are an independent predictor of transplant-associated coronary artery disease after cardiac transplantation.

Transplantation 2001; 71(7):886-92.

111. Dragun D, Müller DN, Bräsen JH, Fritsche L, Nieminen-Kelhä M, Dechend R, Kintscher U, Rudolph B, Hoebeke J, Eckert D, Mazak I, Plehm R, Schönemann C, Unger T, Budde K, Neumayer HH, Luft FC, Wallukat G. Angiotensin II type1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 2005; 352(6):558-69.

112. Chopek MW, Simmons RL, Platt JL. ABO-incompatible kidney transplantation:

initial immunopathologic evaluation. Transplant Proc. 1987; 19(6):4553-7.

113. Delikouras A, Hayes M, Malde P, Lechler RI, Dorling A. Nitric oxide-mediated expression of Bcl-2 and Bcl-xl and protection from tumor necrosis factor-alpha-mediated

(27)

apoptosis in porcine endothelial cells after exposure to low concentrations of xenoreactive natural antibody. Transplantation 2001; 71(5):599-605.

114. Wang H, Arp J, Liu W, Faas SJ, Jiang J, Gies DR, Ramcharran S, Garcia B, Zhong R, Rother RP. Inhibition of terminal complement components in presensitized transplant recipients prevents antibody-mediated rejection leading to long-term graft survival and accommodation. J Immunol 2007; 179(7):4451-63.

115. Chen Song S, Zhong S, Xiang Y, Li JH, Guo H, Wang WY, Xiong YL, Li XC, Chen Shi S, Chen XP, Chen G. Complement inhibition enables renal allograft accommodation and long-term engraftment in presensitized nonhuman primates. Am J Transplant 2011;

11(10):2057-66.

116. Ding JW, Zhou T, Ma L, Yin D, Shen J, Ding CP, Tang IY, Byrne GW, Chong AS.

Expression of complement regulatory proteins in accommodated xenografts induced by anti-alpha-Gal IgG1 in a rat-to-mouse model. Am J Transplant 2008; 8(1):32-40.

117. Uehara S, Chase CM, Cornell LD, Madsen JC, Russell PS, Colvin RB. Chronic cardiac transplant arteriopathy in mice: relationship of alloantibody, C4d deposition and neointimal fibrosis. Am J Transplant 2007; 7(1):57-65.

118. Smith RN, Kawai T, Boskovic S, Nadazdin O, Sachs DH, Cosimi AB, Colvin RB.

Four stages and lack of stable accommodation in chronic alloantibody-mediated renal allograft rejection in Cynomolgus monkeys. Am J Transplant 2008; 8(8):1662-72.

119. Patel R, Terasaki PI. Significance of the positive crossmatch test in kidney transplantation. N Engl J Med 1969; 280(14):735-9.

120. Gebel HM, Bray RA, Nickerson P. Pre-transplant assessment of donor-reactive, HLA-specific antibodies in renal transplantation: contraindication vs. risk. Am J Transplant 2003; 3(12):1488-500.

(28)

121. Vaidya S, Partlow D, Susskind B, Noor M, Barnes T, Gugliuzza K. Prediction of crossmatch outcome of highly sensitized patients by single and/or multiple antigen bead luminex assay. Transplantation 2006; 82(11):1524-8.

122. Monien S, Salama A, Schönemann C. ELISA methods detect HLA antibodies with variable sensitivity. Int J Immunogenet 2006; 33(3):163-6.

123. Bryan CF, Martinez J, Muruve N, Nelson PW, Pierce GE, Ross G, Shield CF, Warady BA, Aeder MI, Harrell KM, Helling TS, Luger AM. IgM antibodies identified by a DTT-ameliorated positive crossmatch do not influence renal graft outcome but the strength of the IgM lymphocytotoxicity is associated with DR phenotype. Clin Transplant 2001; 15 Suppl 6:28-35.

124. Stastny P, Ring S, Lu C, Arenas J, Han M, Lavingia B. Role of immunoglobulin (Ig)- G and IgM antibodies against donor human leukocyte antigens in organ transplant recipients. Hum Immunol 2009; 70(8):600-4.

125. McCalmon RT Jr, Tardif GN, Sheehan MA, Fitting K, Kortz W, Kam I. IgM antibodies in renal transplantation. Clin Transplant 1997; 11(6):558-64.

126. Heidt S, Roelen DL, de Vaal YJ, Kester MG, Eijsink C, Thomas S, van Besouw NM, Volk HD, Weimar W, Claas FH, Mulder A. A Novel ELISPOT Assay to Quantify HLA- Specific B Cells in HLA-Immunized Individuals. Am J Transplant 2012; 12(6):1469-1478.

127. Claas FH, Doxiadis II. Management of the highly sensitized patient. Curr Opin Immunol 2009; 21(5):569-72.

128. Claas FH. Towards selective HLA mismatching in clinical transplantation.

Transplantation 2009; 88(6):760-1.

129. Vo AA, Lukovsky M, Toyoda M, Wang J, Reinsmoen NL, Lai CH, Peng A, Villicana R, Jordan SC. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med 2008; 359(3):242-51.

(29)

130. Anglicheau D, Loupy A, Suberbielle C, Zuber J, Patey N, Noël LH, Cavalcanti R, Le Quintrec M, Audat F, Méjean A, Martinez F, Mamzer-Bruneel MF, Thervet E, Legendre C. Posttransplant prophylactic intravenous immunoglobulin in kidney transplant patients at high immunological risk: a pilot study. Am J Transplant 2007; 7(5):1185-92.

131. Gloor JM, DeGoey SR, Pineda AA, Moore SB, Prieto M, Nyberg SL, Larson TS, Griffin MD, Textor SC, Velosa JA, Schwab TR, Fix LA, Stegall MD. Overcoming a positive crossmatch in living-donor kidney transplantation. Am J Transplant 2003;

3(8):1017-23.

132. Stegall MD, Gloor J, Winters JL, Moore SB, Degoey S. A comparison of plasmapheresis versus high-dose IVIG desensitization in renal allograft recipients with high levels of donor specific alloantibody. Am J Transplant 2006; 6(2):346-51.

133. Jordan SC, Tyan D, Stablein D, McIntosh M, Rose S, Vo A, Toyoda M, Davis C, Shapiro R, Adey D, Milliner D, Graff R, Steiner R, Ciancio G, Sahney S, Light J.

Evaluation of intravenous immunoglobulin as an agent to lower allosensitization and improve transplantation in highly sensitized adult patients with end-stage renal disease:

report of the NIH IG02 trial. J Am Soc Nephrol 2004; 15(12):3256-62.

134. Glotz D, Antoine C, Julia P, Suberbielle-Boissel C, Boudjeltia S, Fraoui R, Hacen C, Duboust A, Bariety J. Desensitization and subsequent kidney transplantation of patients using intravenous immunoglobulins (IVIg). Am J Transplant 2002; 2(8):758- 60.

135. Montgomery RA, Zachary AA, Racusen LC, Leffell MS, King KE, Burdick J, Maley WR, Ratner LE. Plasmapheresis and intravenous immune globulin provides effective rescue therapy for refractory humoral rejection and allows kidneys to be successfully transplanted into cross-match-positive recipients. Transplantation 2000; 70(6):887-95.

136. Sonnenday CJ, Ratner LE, Zachary AA, Burdick JF, Samaniego MD, Kraus E, Warren DS, Montgomery RA. Preemptive therapy with plasmapheresis/intravenous

(30)

immunoglobulin allows successful live donor renal transplantation in patients with a positive cross-match. Transplant Proc 2002; 34(5):1614-6.

137. Böhmig GA, Regele H, Exner M, Derhartunian V, Kletzmayr J, Säemann MD, Hörl WH, Druml W, Watschinger B. C4d-positive acute humoral renal allograft rejection:

effective treatment by immunoadsorption. J Am Soc Nephrol 2001; 12(11):2482-9.

138. Cosio FG, Gloor JM, Sethi S, Stegall MD. Transplant glomerulopathy. Am J Transplant 2008; 8(3):492-6.

139. Gloor JM, Cosio FG, Rea DJ, Wadei HM, Winters JL, Moore SB, DeGoey SR, Lager DJ, Grande JP, Stegall MD. Histologic findings one year after positive crossmatch or ABO blood group incompatible living donor kidney transplantation. Am J Transplant 2006; 6(8):1841-7.

140. Gloor JM, Winters JL, Cornell LD, Fix LA, DeGoey SR, Knauer RM, Cosio FG, Gandhi MJ, Kremers W, Stegall MD. Baseline donor-specific antibody levels and outcomes in positive crossmatch kidney transplantation. Am J Transplant 2010;

10(3):582-9.

141. Loupy A, Suberbielle-Boissel C, Hill GS, Lefaucheur C, Anglicheau D, Zuber J, Martinez F, Thervet E, Méjean A, Charron D, Duong van Huyen JP, Bruneval P, Legendre C, Nochy D. Outcome of subclinical antibody-mediated rejection in kidney transplant recipients with preformed donor-specific antibodies. Am J Transplant 2009;

9(11):2561-70.

142. Marfo K, Lu A, Ling M, Akalin E. Desensitization protocols and their outcome. Clin J Am Soc Nephrol. 2011 Apr;6(4):922-36.

143. Porter KA. The effects of antibodies on human renal allografts. Transplant Proc 1976; 8(2):189-97.

(31)

144. Kissmeyer-Nielsen F, Olsen S, Petersen VP, Fjeldborg O. Hyperacute rejection of kidney allografts, associated with pre-existing humoral antibodies against donor cells.

Lancet 1966; 2(7465):662-5.

145. Taylor CJ, Kosmoliaptsis V, Summers DM, Bradley JA. Back to the future:

application of contemporary technology to long-standing questions about the clinical relevance of human leukocyte antigen-specific alloantibodies in renal transplantation.

Hum Immunol 2009; 70(8):563-8.

146. Burns JM, Cornell LD, Perry DK, Pollinger HS, Gloor JM, Kremers WK, Gandhi MJ, Dean PG, Stegall MD. Alloantibody levels and acute humoral rejection early after positive crossmatch kidney transplantation. Am J Transplant 2008; 8(12):2684-94.

147. Kerman RH, Susskind B, Buyse I, Pryzbylowski P, Ruth J, Warnell S, Gruber SA, Katz S, Van Buren CT, Kahan BD. Flow cytometry-detected IgG is not a contraindication to renal transplantation: IgM may be beneficial to outcome. Transplantation 1999;

68(12):1855-8.

148. Ogura K, Terasaki PI, Johnson C, Mendez R, Rosenthal JT, Ettenger R, Martin DC, Dainko E, Cohen L, Mackett T, et al. The significance of a positive flow cytometry crossmatch test in primary kidney transplantation. Transplantation 1993; 56(2):294-8.

149. Lefaucheur C, Suberbielle-Boissel C, Hill GS, Nochy D, Andrade J, Antoine C, Gautreau C, Charron D, Glotz D. Clinical relevance of preformed HLA donor-specific antibodies in kidney transplantation. Am J Transplant 2008; 8(2):324-31.

150. Singh N, Djamali A, Lorentzen D, Pirsch JD, Leverson G, Neidlinger N, Voss B, Torrealba JR, Hofmann RM, Odorico J, Fernandez LA, Sollinger HW, Samaniego M.

Pretransplant donor-specific antibodies detected by single-antigen bead flow cytometry are associated with inferior kidney transplant outcomes.Transplantation 2010;90(10):1079-84.

(32)

151. Riethmüller S, Ferrari-Lacraz S, Müller MK, Raptis DA, Hadaya K, Rüsi B, Laube G, Schneiter G, Fehr T, Villard J. Donor-specific antibody levels and three generations of crossmatches to predict antibody-mediated rejection in kidney transplantation.

Transplantation 2010; 90(2):160-7.

152. Lefaucheur C, Loupy A, Hill GS, Andrade J, Nochy D, Antoine C, Gautreau C, Charron D, Glotz D, Suberbielle-Boissel C. Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol 2010; 21(8):1398-406.

153. Amico P, Hönger G, Mayr M, Steiger J, Hopfer H, Schaub S. Clinical relevance of pretransplant donor-specific HLA antibodies detected by single-antigen flow-beads.

Transplantation 2009; 87(11):1681-8.

154. Caro-Oleas JL, González-Escribano MF, González-Roncero FM, Acevedo-Calado MJ, Cabello-Chaves V, Gentil-Govantes MA, Núñez-Roldán A. Clinical relevance of HLA donor-specific antibodies detected by single antigen assay in kidney transplantation.

Nephrol Dial Transplant 2011 [Epub ahead of print].

155. van den Berg-Loonen EM, Billen EV, Voorter CE, van Heurn LW, Claas FH, van Hooff JP, Christiaans MH. Clinical relevance of pretransplant donor-directed antibodies detected by single antigen beads in highly sensitized renal transplant patients.

Transplantation 2008; 85(8):1086-90.

156. Couzi L, Araujo C, Guidicelli G, Bachelet T, Moreau K, Morel D, Robert G, Wallerand H, Moreau JF, Taupin JL, Merville P. Interpretation of positive flow cytometric crossmatch in the era of the single-antigen bead assay. Transplantation 2011;

91(5):527-35.

157. Süsal C, Ovens J, Mahmoud K, Döhler B, Scherer S, Ruhenstroth A, Tran TH, Heinold A, Opelz G. No association of kidney graft loss with human leukocyte antigen antibodies detected exclusively by sensitive Luminex single-antigen testing: a Collaborative Transplant Study report. Transplantation 2011; 91(8):883-7.

(33)

158. Hönger G, Hopfer H, Arnold ML, Spriewald BM, Schaub S, Amico P. Pretransplant IgG subclasses of donor-specific human leukocyte antigen antibodies and development of antibody-mediated rejection. Transplantation 2011; 92(1):41-7.

159. Yabu JM, Higgins JP, Chen G, Sequeira F, Busque S, Tyan DB. C1q-fixing human leukocyte antigen antibodies are specific for predicting transplant glomerulopathy and late graft failure after kidney transplantation.Transplantation 2011; 91(3):342-7.

160.Otten HG, Verhaar MC, Borst HP, Hené RJ, Zuilen AD. Pretransplant Donor- Specific HLA Class-I and -II Antibodies Are Associated with an Increased Risk for Kidney Graft Failure. Am J Transplant 2012 Mar 8 [Epub ahead of print].

161. Morales-Buenrostro LE, Terasaki PI, Marino-Vázquez LA, Lee JH, El-Awar N, Alberú J. "Natural" human leukocyte antigen antibodies found in nonalloimmunized healthy males. Transplantation 2008; 86(8):1111-5.

162. Zoet YM, Brand-Schaaf SH, Roelen DL, Mulder A, Claas FH, Doxiadis II.

Challenging the golden standard in defining donor-specific antibodies: does the solid phase assay meet the expectations? Tissue Antigens 2011; 77(3):225-8.

163. Colvin RB. Antibody-mediated renal allograft rejection: diagnosis and pathogenesis.

J Am Soc Nephrol 2007; 18(4):1046-56.

164. Crespo M, Pascual M, Tolkoff-Rubin N, Mauiyyedi S, Collins AB, Fitzpatrick D, Farrell ML, Williams WW, Delmonico FL, Cosimi AB, Colvin RB, Saidman SL. Acute humoral rejection in renal allograft recipients: I. Incidence, serology and clinical characteristics. Transplantation 2001; 71(5):652-8.

165. Mauiyyedi S, Crespo M, Collins AB, Schneeberger EE, Pascual MA, Saidman SL, Tolkoff-Rubin NE, Williams WW, Delmonico FL, Cosimi AB, Colvin RB. Acute humoral rejection in kidney transplantation: II. Morphology, immunopathology and pathologic classification. J Am Soc Nephrol 2002; 13(3):779-87.

(34)

166. Solez K, Colvin RB, Racusen LC, Haas M, Sis B, Mengel M, Halloran PF, Baldwin W, Banfi G, Collins AB, Cosio F, David DS, Drachenberg C, Einecke G, Fogo AB, Gibson IW, Glotz D, Iskandar SS, Kraus E, Lerut E, Mannon RB, Mihatsch M, Nankivell BJ, Nickeleit V, Papadimitriou JC, Randhawa P, Regele H, Renaudin K, Roberts I, Seron D, Smith RN, Valente M. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant 2008; 8(4):753-60.

167. Everly MJ, Everly JJ, Arend LJ, Brailey P, Susskind B, Govil A, Rike A, Roy- Chaudhury P, Mogilishetty G, Alloway RR, Tevar A, Woodle ES. Reducing de novo donor-specific antibody levels during acute rejection diminishes renal allograft loss. Am J Transplant 2009; 9(5):1063-71.

168. Everly MJ, Rebellato LM, Ozawa M, Briley KP, Catrou PG, Haisch CE, Terasaki PI.

Beyond histology: lowering human leukocyte antigen antibody to improve renal allograft survival in acute rejection. Transplantation 2010; 89(8):962-7.

169. Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. Endothelial gene expression in kidney transplants with alloantibody indicates antibody-mediated damage despite lack of C4d staining. Am J Transplant 2009; 9(10):2312-23.

170. Worthington JE, Martin S, Al-Husseini DM, Dyer PA, Johnson RW.

Posttransplantation production of donor HLA-specific antibodies as a predictor of renal transplant outcome. Transplantation 2003; 75(7):1034-40.

171. Mao Q, Terasaki PI, Cai J, Briley K, Catrou P, Haisch C, Rebellato L. Extremely high association between appearance of HLA antibodies and failure of kidney grafts in a five-year longitudinal study. Am J Transplant 2007; 7(4):864-71.

172. Lachmann N, Terasaki PI, Budde K, Liefeldt L, Kahl A, Reinke P, Pratschke J, Rudolph B, Schmidt D, Salama A, Schönemann C. Anti-human leukocyte antigen and donor-specific antibodies detected by luminex posttransplant serve as biomarkers for chronic rejection of renal allografts. Transplantation 2009; 87(10):1505-13.

(35)

173. Lee PC, Zhu L, Terasaki PI, Everly MJ. HLA-specific antibodies developed in the first year posttransplant are predictive of chronic rejection and renal graft loss.

Transplantation 2009; 88(4):568-74.

174. Terasaki PI, Ozawa M. Predicting kidney graft failure by HLA antibodies:

aprospective trial. Am J Transplant 2004 Mar; 4(3):438-43.

175. Mizutani K, Terasaki P, Rosen A, Esquenazi V, Miller J, Shih RN, Pei R, Ozawa M, Lee J. Serial ten-year follow-up of HLA and MICA antibody production prior to kidney graft failure. Am J Transplant 2005; 5(9):2265-72.

176. Cooper JE, Gralla J, Cagle L, Goldberg R, Chan L, Wiseman AC. Inferior kidney allograft outcomes in patients with de novo donor-specific antibodies are due to acute rejection episodes. Transplantation 2011; 91(10):1103-9.

177. Bartel G, Regele H, Wahrmann M, Huttary N, Exner M, Hörl WH, Böhmig GA.

Posttransplant HLA alloreactivity in stable kidney transplant recipients-incidences and impact on long-term allograft outcomes. Am J Transplant 2008; 8(12):2652-60.

178. Akalin E, Pascual M. Sensitization after kidney transplantation. Clin J Am Soc Nephrol 2006; 1(3):433-40.

179. Dinavahi R, George A, Tretin A, Akalin E, Ames S, Bromberg JS, Deboccardo G, Dipaola N, Lerner SM, Mehrotra A, Murphy BT, Nadasdy T, Paz-Artal E, Salomon DR, Schröppel B, Sehgal V, Sachidanandam R, Heeger PS. Antibodies reactive to non-HLA antigens in transplant glomerulopathy. J Am Soc Nephrol 2011; 22(6):1168-78.

180. Ahern AT, Artruc SB, DellaPelle P, Cosimi AB, Russell PS, Colvin RB, Fuller TC.

Hyperacute rejection of HLA-AB-identical renal allografts associated with B lymphocyte and endothelial reactive antibodies. Transplantation1982; 33(1):103-6.

181. Grafft CA, Cornell LD, Gloor JM, Cosio FG, Gandhi MJ, Dean PG, Stegall MD, Amer H. Antibody-mediated rejection following transplantation from an HLA-identical sibling. Nephrol Dial Transplant 2010; 25(1):307-10.

(36)

182. Opelz G; Collaborative Transplant Study. Non-HLA transplantation immunity revealed by lymphocytotoxic antibodies. Lancet 2005; 365(9470):1570-6.

183. Amico P, Hönger G, Bielmann D, Lutz D, Garzoni D, Steiger J, Mihatsch MJ, Dragun D, Schaub S. Incidence and prediction of early antibody-mediated rejection due to non-human leukocyte antigen-antibodies. Transplantation 2008; 85(11):1557-63.

184. Brasile L, Rodman E, Shield CF 3rd, Clarke J, Cerilli J. The association of antivascular endothelial cell antibody with hyperacute rejection: a case report. Surgery 1986; 99(5):637-40.

185. Sumitran-Karuppan S, Tyden G, Reinholt F, Berg U, Moller E. Hyperacute rejections of two consecutive renal allografts and early loss of the third transplant caused by non-HLA antibodies specific for endothelial cells. Transpl Immunol 1997; 5(4):321-7.

186. Zhang Q, Cecka JM, Gjertson DW, Ge P, Rose ML, Patel JK, Ardehali A, Kobashigawa JA, Fishbein MC, Reed EF. HLA and MICA: targets of antibody-mediated rejection in heart transplantation. Transplantation 2011; 91(10):1153-8.

187. Andree H, Nickel P, Nasiadko C, Hammer MH, Schönemann C, Pruss A, Volk HD, Reinke P. Identification of dialysis patients with panel-reactive memory T cells before kidney transplantation using an allogeneic cell bank. J Am Soc Nephrol 2006; 17(2):573- 80.

188. Augustine JJ, Poggio ED, Heeger PS, Hricik DE. Preferential benefit of antibody induction therapy in kidney recipients with high pretransplant frequencies of donor- reactive interferon-gamma enzyme-linked immunosorbent spots. Transplantation 2008;

86(4):529-34.

189. Bordron A, Révélen R, D'Arbonneau F, Dueymes M, Renaudineau Y, Jamin C, Youinou P. Functional heterogeneity of anti-endothelial cell antibodies. Clin Exp Immunol 2001; 124(3):492-501.

(37)

190. Carvalho D, Savage CO, Black CM, Pearson JD. IgG antiendothelial cell autoantibodies from scleroderma patients induce leukocyte adhesion to human vascular endothelial cells in vitro. Induction of adhesion molecule expression and involvement of endothelium-derived cytokines. J Clin Invest 1996; 97(1):111-9.

191. Le Bas-Bernardet S, Hourmant M, Coupel S, Bignon JD, Soulillou JP, Charreau B.

Non-HLA-type endothelial cell reactive alloantibodies in pre-transplant sera of kidney recipients trigger apoptosis. Am J Transplant 2003; 3(2):167-77.

192. Lucchiari N, Panajotopoulos N, Xu C, Rodrigues H, Ianhez LE, Kalil J, Glotz D.

Antibodies eluted from acutely rejected renal allografts bind to and activate human endothelial cells. Hum Immunol. 2000; 61(5):518-27.

193. Fredrich R, Toyoda M, Czer LS, Galfayan K, Galera O, Trento A, Freimark D, Young S, Jordan SC. The clinical significance of antibodies to human vascular endothelial cells after cardiac transplantation. Transplantation 1999; 67(3):385-91.

194. Smith JD, Hamour IM, Burke MM, Mahesh B, Stanford RE, Haj-Yahia S, Robinson DR, Kaul P, Yacoub MH, Banner NR, Rose ML. A reevaluation of the role of IgM non- HLA antibodies in cardiac transplantation. Transplantation 2009; 87(6):864-71.

195. Derhaag JG, Duijvestijn AM, Damoiseaux JG, van Breda Vriesman PJ. Effects of antibody reactivity to major histocompatibility complex (MHC) and non-MHC alloantigens on graft endothelial cells in heart allograft rejection.Transplantation 2000; 69(9):1899- 906.

196. Wu GD, Jin YS, Salazar R, Dai WD, Barteneva N, Barr ML, Barsky LW, Starnes VA, Cramer DV. Vascular endothelial cell apoptosis induced by anti-donor non-MHC antibodies: a possible injury pathway contributing to chronic allograft rejection. J Heart Lung Transplant 2002; 21(11):1174-87.

Références

Documents relatifs

This software checksum, in addition to the hardware checksum of the circuit, checks the modem interfaces and memories at each IMP, and protects the IMPs from erroneous routing

Section 4.1 provides an essential analysis of how address configuration is performed in IPv6, identifying patterns in IPv6 addresses that can be leveraged to reduce the

A Lyndon word is a word which is the strict minimum (for ⪯ lex , hence the alphabet must have been totally ordered) of its conjugacy class. In an equivalent way we set

Au total il y a donc un seuil de perceptibilité des structures de petite taille et à faible contraste propre, et ce seuil est directement lié à la dose d'exposition ... 2-Un

1998 Châteauneuf-du-Pape, Cuvée Mourre des Perdrix, Domaine de la Charbonniere 450 2005 Châteauneuf-du-Pape, Château de Beaucastel, Vignobles Perrin 575 2015

A  50-year-old  woman  presented  to  her  GP  follow-

If Japan is, historically speaking, a special case in its relationship between budo and sports, what is striking at the global level, it is our inability to think the physical

transformed, not only in the direct practical way of becoming cultivated, or shaped into objects of use in the embodied artifacts we call tools…it becomes transformed as an object