Immunosuppression in Sensitized Patients: Comparison
Please note this is a comparison between Version 2 by Lindsay Dong and Version 1 by Jean Kwun.

Following organ transplantation, sensitized patients have higher rates of antibody-mediated rejection (AMR) compared to those who are non-sensitized. More stringent donor matching is required for these patients. Current approaches for sensitized patients focus on reducing preformed antibodies that preclude transplantation; however, this type of desensitization does not modulate the primed immune response in sensitized patients. Thus, an optimized maintenance immunosuppressive regimen is necessary for highly sensitized patients, which may be distinct from non-sensitized patients.

  • sensitization
  • immunosuppression
  • induction
  • B cell
  • plasma cell

1. Introduction

Immunosuppressive strategies for sensitized patients are largely borrowed from those used in non-sensitized patients. However, variability in outcomes reveals the insufficiency of current immunosuppressive regimens in sensitized patients. Sensitized patients with a negative crossmatch (no donor-specific antibody) showed comparable graft survival to non-sensitized patients in the current organ allocation system [4][1] even though these patients might have individual center-driven immunosuppressive regimens which are different from non-sensitized patients (i.e., thymoglobulin with higher Tac trough level, etc.). However, immunologically high-risk transplants occurring in sensitized patients, particularly for crossmatch positive, incompatible transplants, require enhanced immunosuppression. Innovation in this field has largely focused on ‘desensitization’ prior to transplantation, or early post-transplant therapies to reduce the risks of acute antibody-mediated rejection (AMR) [5,6,7,8,9,10,11,12,13,14][2][3][4][5][6][7][8][9][10][11]; however, there has been little examination of the optimal maintenance regimen post-transplant. Furthermore, even with currently available desensitization therapies, both acute AMR and acute cellular rejection (ACR) rates were significantly higher in sensitized/desensitized patients compared to non-sensitized patients [15,16,17][12][13][14]. Recently, changes in deceased donor allocation in the US in particular [18][15], as well as improvements to living kidney donor sharing schemes [19][16], have demonstrated that fewer sensitized patients require the need for cross-match positive living transplantation [20][17]. Nonetheless, patients with pretransplant or de novo donor-specific antibody (DSA) are at greater risk of graft rejection.

2. Choice of Induction Therapy in Sensitized Kidney Transplant Recipients

Induction therapy reduces rates of acute rejection, delayed graft function (DGF), and death after kidney transplantation, and there is a wide variety of induction agents available and used in clinical practice today [21][18]. Rabbit antithymocyte (rATG) polyclonal antibody or interleukin-2 receptor monoclonal antibodies are the most common agents used for induction in non-sensitized patients. Sensitized patients with preformed HLA antibodies are at greater risk of cellular and humoral rejection, and outcomes can be optimized by using polyclonal induction agents, such as ATG or alemtuzumab, that are associated with a lower risk of rejection and better graft survival [22,23,24,25][19][20][21][22]. However, the impact of different induction approaches on sensitized patients has not been fully elucidated and the variability in induction therapy can be largely attributed to transplant center choice and clinician preference rather than patient or donor characteristics [23,24,25,26][20][21][22][23].

2.1. Basiliximab

Basiliximab (Simulect) is a non-depleting chimeric anti-CD25 monoclonal antibody against the interleukin-2 (IL-2) receptor on activated T lymphocytes [27][24]. It is comparable to rATG in patients with low risk of acute rejection, though less effective in high-risk kidney transplant patients, defined as being at risk of DGF or having panel reactive antibody (PRA) > 20% [27,28,29][24][25][26]. Even though activated B cells express CD25 and IL-2 mediated signaling has a critical role for its further differentiation into plasma cells [30][27], our data in a highly sensitized nonhuman primate model demonstrated a clear limitation of basilliximab in controlling robust memory T and B cell immune responses [31][28]. Additionally, basiliximab was associated with a greater risk of biopsy-proven acute rejection (BPAR) than rATG in sensitized (HLA class I and II mismatch) kidney transplant recipients without pre-existing DSA [32][29].

2.2. Thymoglobulin

Thymoglobulin, or rATG, is a polyclonal gamma immunoglobulin and the preferred choice in sensitized patients at high risk for acute rejection or delayed graft function [21,22][18][19]. rATG targets T cells via antibody dependent cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC), but it also depletes B cells and plasma cells since thymus also contains these cell populations. In moderately sensitized patients (positive DSA and negative flow crossmatch), induction with ATG resulted in reduced occurrence of de novo DSA (dnDSA) and AMR compared to basiliximab [38][30]. In simultaneous heart and kidney transplants, sensitized patients (with PRA > 10%) treated with rATG induction had lower mortality [39][31]

2.3. Alemtuzumab

Alemtuzumab is a depleting anti-CD52 antibody that targets T and B cells resulting in lymphocyte depletion and prolonged immunosuppression [46][32]. Low-dose alemtuzumab is used as an induction agent in sensitized patients undergoing kidney transplantation and, when combined with triple maintenance immunosuppression, is well tolerated and has shown favorable patient and allograft outcomes (death-censored graft survival: 79.2%) [46][32]. A prospective study found that the rate of biopsy-confirmed acute rejection in low-risk patients was lower with alemtuzumab when compared with basiliximab, but among high-risk patients, there was no significant difference between alemtuzumab and rATG [50][33]. However, alemtuzumab is associated with prolonged lymphocyte depletion [46,51][32][34] and increased rates of infection [46,52][32][35]

2.4. Rituximab

Rituximab is an anti-CD20 monoclonal antibody that targets B cells, suppresses preformed alloantibodies and reduces peripheral B lymphocytes prior to transplantation [53,54][36][37]. A retrospective study of highly sensitized kidney transplant recipients (XM-positive or DSA positive) treated with IVIG and rituximab induction therapy found increased rates of AMR in sensitized recipients compared to low-risk recipients, but similar long-term patient or graft survival at 6 year follow-up [55][38].

3. Choice of Maintenance Immunosuppression in Sensitized Patients

3.1. Triple Immunosuppression

Maintenance immunosuppressive therapy prevents acute rejection and increases allograft survival following kidney transplantation. The standard maintenance regimen varies by center or clinician preference, but the KDIGO Transplant Work Group guideline recommendations include triple therapy immunosuppression consisting of a calcineurin-inhibitor (CNI), such as tacrolimus, an antimetabolite, such as mycophenolate mofetil (MMF), and a glucocorticoid, such as oral prednisone, in kidney transplant recipients [66][39]. Tacrolimus inhibits activation of T lymphocytes by binding to an intracellular protein, FKBP-12, and inhibiting calcineurin while MMF inhibits T and B cell proliferation. It has been shown that CNIs (cyclosporine and tacrolimus) inhibit antibody production in T and B cell cultures but fail to inhibit immunoglobulin (Ig) production when B cells are cultured with primed T cells [67][40]. According to the most recent Scientific Registry of Transplant Recipients (SRTR) registry data, over 60% of patients are discharged from the hospital on tacrolimus, MMF, and prednisone triple therapy due to its success as a maintenance immunosuppressive regimen [1,68][41][42].

3.2. Limited Efficacy of Standard Maintenance Immunosuppression in Sensitized Patients

Graft function was similar between groups at 1 year follow-up with no graft loss, and both groups were treated with thymoglobulin induction and standard maintenance triple therapy [70][43]. A study in which all patients were maintained on standard triple therapy found an increased incidence of acute AMR in patients with pretransplant DSA than those without (41.7% vs. 1.6%, p < 0.001) and that higher levels of pretransplant DSA had a detrimental effect on 5 year graft survival [74][44]. These studies highlight the limitations of standard triple maintenance immunosuppressive therapy and the need for different therapeutic regimens in the sensitized, DSA positive, CDC-crossmatch negative patient population, particularly in light of the experimental evidence highlighted regarding the inability of CNI to prevent antibody production during cognate T–B cell interactions [67][40].

3.3. Replacing Tacrolimus

The mammalian target of rapamycin (mTOR) controls the T cell response (activation and proliferation) and is a valuable immunosuppressant in clinical transplantation. mTOR inhibitors, such as rapamycin (sirolimus) and everolimus, promote the differentiation and function of various helper T cells and suppress the differentiation of memory CD8+ T cells [75][45]. Furthermore, unlike CNIs, mTOR inhibitors are able to prevent Ig production from B cells when cultured with primed T cells, which suggests their direct impact on B cells [67,76][40][46]. Rapamycin has also shown its superiority over tacrolimus with respect to inhibiting B cell to plasma-cell differentiation [77][47]. In mice sensitized by prior skin graft, preoperative rapamycin increased the expression of regulatory T cells, but did not prolong the survival of mice after cardiac allotransplantation [78][48]. In donor skin-sensitized mice, those with mTOR deletion in T cells had longer mean survival time (MST 19.5 days) versus wild-type recipients (MST 5.4 days) [75][45]. Mice sensitized by skin transplant and treated with rapamycin induction therapy were found to have altered frequencies of splenic and intragraft neutrophils, macrophages, and natural killer (NK) cells [79][49].

4. Newly Available Agents for Sensitized Patients

4.1. Costimulation Blockade

4.1.1. Belatacept

Belatacept, a CTLA4-Ig fusion protein that binds to cluster of differentiation (CD) 80 and CD86 receptors on antigen presenting cells (APC), prevents binding to CD28 on T cells, thereby reducing the T cell–dependent immune response [89][50]. Belatacept has been shown to selectively reduce the humoral response in sensitized, maximally HLA-mismatched non-human primates (NHPs) by suppressing the peripheral and germinal center follicular helper T cell (Tfh) response [90][51]. Translational studies in highly sensitized NHPs found that desensitization with belatacept in combination with bortezomib or carfilzomib therapy led to significantly reduced AMR, DSA, and plasma cells leading to prolonged graft survival, although it should be noted that these animals received tacrolimus-based triple therapy as maintenance [64,91][52][53]. Preliminary studies in animals receiving belatacept in addition to triple therapy indicate a further prolongation of survival, even in a highly sensitized NHP model [92][54].

4.1.2. Anti-CD40mAb

The CD40/CD154 pathway is important for activating T cell differentiation and B cell isotype switching and was found to be important in both the humoral and cell-mediated immunologic response pathways [110][55]. CD4+ helper T cells are mandatory for generating both naïve and memory DSA responses [111][56]. Thus, targeting helper T cells in maintenance therapy may lead to decreased AMR and prolonged allograft survival in kidney transplant recipients. Much of the existing evidence is in large animal models as clinical studies blocking the CD40/CD154 pathway have been halted due to the development of thromboembolic complications and direct platelet activation [112,113][57][58]. Thromboembolic complications were found to be primarily due to blocking interactions with CD154, which is important for thrombus stability [114][59]. However, similar events were not observed in antibodies targeting CD40 [115[60][61][62],116,117], so this may be a more promising therapeutic target. A novel blocking, non-depleting Fc-silent anti-CD40 mAb, iscalimab (CFZ533), has been found to prolong renal allograft survival in NHP in the absence of B cell depletion with no evidence of thromboembolic events [118][63].

4.1.3. Anti-CD154mAb

An initial study found that anti-CD154 monoclonal antibodies prevent acute renal allograft rejection in non-sensitized NHPs [121][64]. A study in donor skin-graft-sensitized mouse recipients of cardiac allografts found that naïve CD8+ T cells depend on CD154 signaling to differentiate into effector T cells, while primed/memory CD8+ T cells remain intact [122][65].

4.2. Adjuvant Therapies

4.2.1. Targeting IL-6 or IL-6R

Interleukin (IL)-6 is a pleiotropic cytokine involved in a variety of pathways regulating immune responses, with an important role in the induction of follicular helper T cells which stimulate B cells to differentiate into memory B cells and IgG-secreting plasma cells [126][66]. The IL-6 receptor (IL-6R) exists as a membrane-bound protein, expressed mostly on hepatocytes and immune cells, and a soluble protein that can bind IL-6 and transmembrane gp130, termed trans-signaling, on nearly all cell types [127,128][67][68]. Interactions between IL-6 and IL-6R lead to the activation of transmembrane protein gp130, eliciting signals to downstream JAK and MAPK pathways and the subsequent activation of inflammatory genes [127][67]. IL-6 is a proinflammatory cytokine that plays a pathologic role in chronic immune disorders, cancer, and transplant rejection [128][68]. IL-6 also promotes Th17 differentiation and inhibits Treg differentiation, suggesting targeting IL-6/IL-6R may have clinical applications in treating autoimmune disease and organ rejection [127,129][67][69].

4.2.2. Anti-BAFF

B cell activating factor (BAFF, also known as BLyS) and a proliferating inducing ligand (APRIL) are cytokines that belong to the tumor necrosis factor family whose primary function is to enhance B cell survival and differentiation into plasma cells [133][70]. Both are currently used in treating autoimmune diseases such as systemic lupus erythematosus (SLE) and Sjogren’s, but several studies have found that high levels of serum BAFF are associated with the formation of anti-HLA DSA, increased risk of AMR, and poor renal graft survival [134,135,136,137,138][71][72][73][74][75]. BAFF is highly expressed in the membrane and renal tubule epithelial cells of transplanted kidneys with acute rejection, and high pretransplant BAFF has been found to predict risk of graft rejection [139][76].

4.2.3. Targeting PC

Conventional PI

Bortezomib was the first proteasome inhibitor (PI) to be FDA-approved for the treatment of malignant plasma cell diseases [148][77]. Proteasome inhibitors, including bortezomib, carfilzomib, oprozomib (ONX 0912), and ONX 0914 (immunoproteasome inhibitor), reduce proliferating B cells and antibody production conceivably by inducing apoptosis of activated B cells [149][78]. Bortezomib has been shown to be effective in preventing AMR and ACR, as well as reducing DSA in kidney transplant recipients [150][79]. Since then, bortezomib has been studied in combination therapy with plasmapheresis (PP) and IVIG with or without rituximab or steroid as plasma cell–targeted therapy in sensitized kidney transplant recipients, showing success in reducing DSA, treating acute or late AMR [151,152][80][81] as rescue or primary treatment [153[82][83][84],154,155], and reducing plasma cell rich acute rejection [156,157][85][86]. Recently, six patients who developed acute AMR received bortezomib and belatacept combination therapy, which led to the reversal of AMR and reduction in circulating DSA [158][87].

5. Concluding Remarks

Conventional maintenance immunosuppression with tacrolimus, MMF, and steroids after lymphocytic depletion has been widely used for managing sensitized patients with incompatible organ transplants considered to be at increased immunological risk of rejection. Despite the fact that these T cell centric approaches are effective in non-sensitized patients, they fail to control the post-transplant humoral response of sensitized patients. Given the known challenges of the primed immune system of the sensitized patient, targeting B cell or T cell interactions with B cells should be considered as part of an optimal maintenance immunosuppression for this patient population. Due to the rapid evolution of agents targeting individual steps of humoral responses, as well as advances in our understanding of AMR, it is possible to design a mechanistically rational approach for the sensitized patient.

References

  1. Sethi, S.; Najjar, R.; Peng, A.; Mirocha, J.; Vo, A.; Bunnapradist, S.; Jordan, S.C.; Huang, E. Allocation of the Highest Quality Kidneys and Transplant Outcomes Under the New Kidney Allocation System. Am. J. Kidney Dis. 2019, 73, 605–614.
  2. Higgins, R.; Lowe, D.; Hathaway, M.; Lam, F.T.; Kashi, H.; Tan, L.C.; Imray, C.; Fletcher, S.; Chen, K.; Krishnan, N.; et al. Double filtration plasmapheresis in antibody-incompatible kidney transplantation. Ther. Apher. Dial. 2010, 14, 392–399.
  3. Montgomery, R.A.; Zachary, A.A.; Racusen, L.C.; Leffell, M.S.; King, K.E.; Burdick, J.; Maley, W.R.; Ratner, L.E. 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, 887–895.
  4. Thielke, J.; DeChristopher, P.J.; Sankary, H.; Oberholzer, J.; Testa, G.; Benedetti, E. Highly successful living donor kidney transplantation after conversion to negative of a previously positive flow-cytometry cross-match by pretransplant plasmapheresis. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2005; Volume 37, pp. 643–644.
  5. Warren, D.S.; Simpkins, C.E.; Cooper, M.; Montgomery, R.A. Modulating alloimmune responses with plasmapheresis and IVIG. Curr. Drug Targets Cardiovasc. Haematol. Disord. 2005, 5, 215–222.
  6. Lorenz, M.; Regele, H.; Schillinger, M.; Kletzmayr, J.; Haidbauer, B.; Derfler, K.; Druml, W.; Bohmig, G.A. Peritransplant immunoadsorption: A strategy enabling transplantation in highly sensitized crossmatch-positive cadaveric kidney allograft recipients. Transplantation 2005, 79, 696–701.
  7. Bartel, G.; Wahrmann, M.; Regele, H.; Kikic, Z.; Fischer, G.; Druml, W.; Muhlbacher, F.; Bohmig, G.A. Peritransplant immunoadsorption for positive crossmatch deceased donor kidney transplantation. Am. J. Transplant. 2010, 10, 2033–2042.
  8. Morath, C.; Beimler, J.; Opelz, G.; Scherer, S.; Schmidt, J.; Macher-Goeppinger, S.; Klein, K.; Sommerer, C.; Schwenger, V.; Zeier, M.; et al. Living donor kidney transplantation in crossmatch-positive patients enabled by peritransplant immunoadsorption and anti-CD20 therapy. Transpl. Int. 2012, 25, 506–517.
  9. Clatworthy, M.R. Targeting B cells and antibody in transplantation. Am. J. Transplant. 2011, 11, 1359–1367.
  10. Loupy, A.; Suberbielle-Boissel, C.; Zuber, J.; Anglicheau, D.; Timsit, M.O.; Martinez, F.; Thervet, E.; Bruneval, P.; Charron, D.; Hill, G.S.; et al. Combined posttransplant prophylactic IVIg/anti-CD 20/plasmapheresis in kidney recipients with preformed donor-specific antibodies: A pilot study. Transplantation 2010, 89, 1403–1410.
  11. Vo, A.A.; Lukovsky, M.; Toyoda, M.; Wang, J.; Reinsmoen, N.L.; Lai, C.H.; Peng, A.; Villicana, R.; Jordan, S.C. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N. Engl. J. Med. 2008, 359, 242–251.
  12. Loupy, A.; Suberbielle-Boissel, C.; Hill, G.S.; Lefaucheur, C.; Anglicheau, D.; Zuber, J.; Martinez, F.; Thervet, E.; Mejean, A.; Charron, D.; et al. Outcome of subclinical antibody-mediated rejection in kidney transplant recipients with preformed donor-specific antibodies. Am. J. Transplant. 2009, 9, 2561–2570.
  13. Gloor, J.M.; Winters, J.L.; Cornell, L.D.; Fix, L.A.; DeGoey, S.R.; Knauer, R.M.; Cosio, F.G.; Gandhi, M.J.; Kremers, W.; Stegall, M.D. Baseline donor-specific antibody levels and outcomes in positive crossmatch kidney transplantation. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2010, 10, 582–589.
  14. Haririan, A.; Nogueira, J.; Kukuruga, D.; Schweitzer, E.; Hess, J.; Gurk-Turner, C.; Jacobs, S.; Drachenberg, C.; Bartlett, S.; Cooper, M. Positive cross-match living donor kidney transplantation: Longer-term outcomes. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2009, 9, 536–542.
  15. Israni, A.K.; Salkowski, N.; Gustafson, S.; Snyder, J.J.; Friedewald, J.J.; Formica, R.N.; Wang, X.; Shteyn, E.; Cherikh, W.; Stewart, D.; et al. New national allocation policy for deceased donor kidneys in the United States and possible effect on patient outcomes. J. Am. Soc. Nephrol. JASN 2014, 25, 1842–1848.
  16. Flechner, S.M.; Thomas, A.G.; Ronin, M.; Veale, J.L.; Leeser, D.B.; Kapur, S.; Peipert, J.D.; Segev, D.L.; Henderson, M.L.; Shaffer, A.A.; et al. The first 9 years of kidney paired donation through the National Kidney Registry: Characteristics of donors and recipients compared with National Live Donor Transplant Registries. Am. J. Transplant. 2018, 18, 2730–2738.
  17. Schinstock, C.A.; Smith, B.H.; Montgomery, R.A.; Jordan, S.C.; Bentall, A.J.; Mai, M.; Khamash, H.A.; Stegall, M.D. Managing highly sensitized renal transplant candidates in the era of kidney paired donation and the new kidney allocation system: Is there still a role for desensitization? Clin. Transplant. 2019, 33, e13751.
  18. Brennan, D.C.; Daller, J.A.; Lake, K.D.; Cibrik, D.; Del Castillo, D. Rabbit antithymocyte globulin versus basiliximab in renal transplantation. N. Engl. J. Med. 2006, 355, 1967–1977.
  19. Bunnapradist, S.; Takemoto, S.K. Multivariate analysis of antibody induction therapy and their associated outcomes in deceased donor transplants. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2005; Volume 37, pp. 889–891.
  20. Gabardi, S.; Martin, S.T.; Roberts, K.L.; Grafals, M. Induction immunosuppressive therapies in renal transplantation. Am. J. Health Syst. Pharm. 2011, 68, 211–218.
  21. Laftavi, M.R.; Pankewycz, O.; Feng, L.; Said, M.; Patel, S. Combined induction therapy with rabbit antithymocyte globulin and rituximab in highly sensitized renal recipients. Immunol. Investig. 2015, 44, 373–384.
  22. Lasmar, M.F.; Dutra, R.S.; Nogueira-Machado, J.A.; Fabreti-Oliveira, R.A.; Siqueira, R.G.; Nascimento, E. Effects of immunotherapy induction on outcome and graft survival of kidney-transplanted patients with different immunological risk of rejection. BMC Nephrol. 2019, 20, 314.
  23. Dharnidharka, V.R.; Naik, A.S.; Axelrod, D.A.; Schnitzler, M.A.; Zhang, Z.; Bae, S.; Segev, D.L.; Brennan, D.C.; Alhamad, T.; Ouseph, R.; et al. Center practice drives variation in choice of US kidney transplant induction therapy: A retrospective analysis of contemporary practice. Transplant. Int. 2018, 31, 198–211.
  24. Chapman, T.M.; Keating, G.M. Basiliximab: A review of its use as induction therapy in renal transplantation. Drugs 2003, 63, 2803–2835.
  25. Mariat, C.; Afiani, A.; Alamartine, E.; Thibaudin, D.; de Filippis, J.P.; Berthoux, F. A pilot study comparing basiliximab and anti-thymocyte globulin as induction therapy in sensitized renal allograft recipients. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2001; Volume 33, pp. 3192–3193.
  26. Dedinská, I.; Graňák, K.; Vnučák, M.; Skálová, P.; Laca, Ĺ.; Krivuš, J.; Galajda, P.; Mokáň, M. Induction Therapy With ATG Compared With Anti-IL2 Basiliximab in Low-Immunologic Risk Kidney Transplant Recipients. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2019; Volume 51, pp. 3259–3264.
  27. Le Gallou, S.; Caron, G.; Delaloy, C.; Rossille, D.; Tarte, K.; Fest, T. IL-2 requirement for human plasma cell generation: Coupling differentiation and proliferation by enhancing MAPK-ERK signaling. J. Immunol. 2012, 189, 161–173.
  28. Burghuber, C.K.; Kwun, J.; Page, E.J.; Manook, M.; Gibby, A.C.; Leopardi, F.V.; Song, M.; Farris, A.B., 3rd; Hong, J.J.; Villinger, F.; et al. Antibody-Mediated Rejection in Sensitized Nonhuman Primates: Modeling Human Biology. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2016, 16, 1726–1738.
  29. Goumard, A.; Sautenet, B.; Bailly, E.; Miquelestorena-Standley, E.; Proust, B.; Longuet, H.; Binet, L.; Baron, C.; Halimi, J.M.; Büchler, M.; et al. Increased risk of rejection after basiliximab induction in sensitized kidney transplant recipients without pre-existing donor-specific antibodies—A retrospective study. Transpl. Int. 2019, 32, 820–830.
  30. Brokhof, M.M.; Sollinger, H.W.; Hager, D.R.; Muth, B.L.; Pirsch, J.D.; Fernandez, L.A.; Bellingham, J.M.; Mezrich, J.D.; Foley, D.P.; D’Alessandro, A.M.; et al. Antithymocyte globulin is associated with a lower incidence of de novo donor-specific antibodies in moderately sensitized renal transplant recipients. Transplantation 2014, 97, 612–617.
  31. Ariyamuthu, V.K.; Amin, A.A.; Drazner, M.H.; Araj, F.; Mammen, P.P.A.; Ayvaci, M.; Mete, M.; Ozay, F.; Ghanta, M.; Mohan, S.; et al. Induction regimen and survival in simultaneous heart-kidney transplant recipients. J. Heart Lung Transplant. 2018, 37, 587–595.
  32. Guthoff, M.; Berger, K.; Althaus, K.; Mühlbacher, T.; Bakchoul, T.; Steurer, W.; Nadalin, S.; Königsrainer, A.; Heyne, N. Low-dose alemtuzumab induction in a tailored immunosuppression protocol for sensitized kidney transplant recipients. BMC Nephrol. 2020, 21, 178.
  33. Hanaway, M.J.; Woodle, E.S.; Mulgaonkar, S.; Peddi, V.R.; Kaufman, D.B.; First, M.R.; Croy, R.; Holman, J. Alemtuzumab Induction in Renal Transplantation. N. Engl. J. Med. 2011, 364, 1909–1919.
  34. Sageshima, J.; Ciancio, G.; Guerra, G.; Gaynor, J.J.; Cova, D.; Zarak, A.; Chen, L.; Mattiazzi, A.; Kupin, W.; Roth, D.; et al. Prolonged lymphocyte depletion by single-dose rabbit anti-thymocyte globulin and alemtuzumab in kidney transplantation. Transpl. Immunol. 2011, 25, 104–111.
  35. Clatworthy, M.R.; Friend, P.J.; Calne, R.Y.; Rebello, P.R.; Hale, G.; Waldmann, H.; Watson, C.J. Alemtuzumab (CAMPATH-1H) for the treatment of acute rejection in kidney transplant recipients: Long-term follow-up. Transplantation 2009, 87, 1092–1095.
  36. Halloran, P.F. Immunosuppressive drugs for kidney transplantation. N. Engl. J. Med. 2004, 351, 2715–2729.
  37. Genberg, H.; Hansson, A.; Wernerson, A.; Wennberg, L.; Tydén, G. Pharmacodynamics of rituximab in kidney allotransplantation. Am. J. Transplant. 2006, 6, 2418–2428.
  38. Kahwaji, J.; Jordan, S.C.; Najjar, R.; Wongsaroj, P.; Choi, J.; Peng, A.; Villicana, R.; Vo, A. Six-year outcomes in broadly HLA-sensitized living donor transplant recipients desensitized with intravenous immunoglobulin and rituximab. Transpl. Int. 2016, 29, 1276–1285.
  39. Kasiske, B.; Zeier, M.; Craig, J.; Ekberg, H.; Garvey, C.; Green, M.; Abariga, S. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am. J. Transplant. 2009, 9 (Suppl. S3), S1–S155.
  40. Heidt, S.; Roelen, D.L.; Eijsink, C.; Eikmans, M.; van Kooten, C.; Claas, F.H.; Mulder, A. Calcineurin inhibitors affect B cell antibody responses indirectly by interfering with T cell help. Clin. Exp. Immunol. 2010, 159, 199–207.
  41. Hart, A.; Lentine, K.L.; Smith, J.M.; Miller, J.M.; Skeans, M.A.; Prentice, M.; Robinson, A.; Foutz, J.; Booker, S.E.; Israni, A.K.; et al. OPTN/SRTR 2019 Annual Data Report: Kidney. Am. J. Transplant. 2021, 21 (Suppl. S2), 21–137.
  42. Enderby, C.; Keller, C.A. An overview of immunosuppression in solid organ transplantation. Am. J. Manag. Care 2015, 21, s12–s23.
  43. Ixtlapale-Carmona, X.; Arvizu, A.; De-Santiago, A.; González-Tableros, N.; López, M.; Castelán, N.; Marino, L.A.; Uribe-Uribe, N.O.; Contreras, A.G.; Vilatobá, M.; et al. Graft immunologic events in deceased donor kidney transplant recipients with preformed HLA-donor specific antibodies. Transpl. Immunol. 2018, 46, 8–13.
  44. Salvadé, I.; Aubert, V.; Venetz, J.P.; Golshayan, D.; Saouli, A.C.; Matter, M.; Rotman, S.; Pantaleo, G.; Pascual, M. Clinically-relevant threshold of preformed donor-specific anti-HLA antibodies in kidney transplantation. Hum. Immunol. 2016, 77, 483–489.
  45. Xie, A.; Yan, H.; Fu, J.; He, A.; Xiao, X.; Li, X.C.; Chen, W. T follicular helper and memory cell responses and the mTOR pathway in murine heart transplantation. J. Heart Lung Transplant. 2020, 39, 134–144.
  46. Matz, M.; Lehnert, M.; Lorkowski, C.; Fabritius, K.; Unterwalder, N.; Doueiri, S.; Weber, U.A.; Mashreghi, M.F.; Neumayer, H.H.; Budde, K. Effects of sotrastaurin, mycophenolic acid and everolimus on human B-lymphocyte function and activation. Transpl. Int. 2012, 25, 1106–1116.
  47. Traitanon, O.; Mathew, J.M.; La Monica, G.; Xu, L.; Mas, V.; Gallon, L. Differential Effects of Tacrolimus versus Sirolimus on the Proliferation, Activation and Differentiation of Human B Cells. PLoS ONE 2015, 10, e0129658.
  48. Gong, W.L.; Sha, C.; Du, G.; Shan, Z.G.; Qi, Z.Q.; Zhou, S.F.; Yang, N.; Zhao, Y.X. Preoperative application of combination of portal venous injection of donor spleen cells and intraperitoneal injection of rapamycin prolongs the survival of cardiac allografts in mice. Asian Pac. J. Trop. Med. 2017, 10, 454–460.
  49. Gong, W.; Liu, B.; Chen, J.; Liu, C.; Shen, Z. Impact of Regulatory T Cells on Innate Immune Cells in a Pre-Sensitized Heart Transplant Model. Ann. Transplant. 2018, 23, 246–251.
  50. Larsen, C.P.; Pearson, T.C.; Adams, A.B.; Tso, P.; Shirasugi, N.; Strobert, E.; Anderson, D.; Cowan, S.; Price, K.; Naemura, J.; et al. Rational development of LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent immunosuppressive properties. Am. J. Transplant. 2005, 5, 443–453.
  51. Kim, E.J.; Kwun, J.; Gibby, A.C.; Hong, J.J.; Farris, A.B., 3rd; Iwakoshi, N.N.; Villinger, F.; Kirk, A.D.; Knechtle, S.J. Costimulation blockade alters germinal center responses and prevents antibody-mediated rejection. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2014, 14, 59–69.
  52. Burghuber, C.K.; Manook, M.; Ezekian, B.; Gibby, A.C.; Leopardi, F.V.; Song, M.; Jenks, J.; Saccoccio, F.; Permar, S.; Farris, A.B.; et al. Dual targeting: Combining costimulation blockade and bortezomib to permit kidney transplantation in sensitized recipients. Am. J. Transplant. 2019, 19, 724–736.
  53. Ezekian, B.; Schroder, P.M.; Mulvihill, M.S.; Barbas, A.; Collins, B.; Freischlag, K.; Yoon, J.; Yi, J.S.; Smith, F.; Olaso, D.; et al. Pretransplant Desensitization with Costimulation Blockade and Proteasome Inhibitor Reduces DSA and Delays Antibody-Mediated Rejection in Highly Sensitized Nonhuman Primate Kidney Transplant Recipients. J. Am. Soc. Nephrol. 2019, 30, 2399–2411.
  54. Schmitz, R.; Fitch, Z.W.; Schroder, P.M.; Choi, A.Y.; Manook, M.; Yoon, J.; Kwun, J.; Knechtle, S.J. Costimulation Blockade-Based Maintenance Immunosuppression in a Sensitized Nonhuman Primate Kidney Allotransplantation Model. Am. J. Transplant. 2020, 20, 341–342.
  55. Elgueta, R.; Benson, M.J.; de Vries, V.C.; Wasiuk, A.; Guo, Y.; Noelle, R.J. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol. Rev. 2009, 229, 152–172.
  56. Chen, C.C.; Koenig, A.; Saison, C.; Dahdal, S.; Rigault, G.; Barba, T.; Taillardet, M.; Chartoire, D.; Ovize, M.; Morelon, E.; et al. CD4+ T Cell Help Is Mandatory for Naive and Memory Donor-Specific Antibody Responses: Impact of Therapeutic Immunosuppression. Front. Immunol. 2018, 9, 275.
  57. Koyama, I.; Kawai, T.; Andrews, D.; Boskovic, S.; Nadazdin, O.; Wee, S.L.; Sogawa, H.; Wu, D.L.; Smith, R.N.; Colvin, R.B.; et al. Thrombophilia associated with anti-CD154 monoclonal antibody treatment and its prophylaxis in nonhuman primates. Transplantation 2004, 77, 460–462.
  58. Xu, H.; Zhang, X.; Mannon, R.B.; Kirk, A.D. Platelet-derived or soluble CD154 induces vascularized allograft rejection independent of cell-bound CD154. J. Clin. Investig. 2006, 116, 769–774.
  59. André, P.; Prasad, K.S.; Denis, C.V.; He, M.; Papalia, J.M.; Hynes, R.O.; Phillips, D.R.; Wagner, D.D. CD40L stabilizes arterial thrombi by a beta3 integrin–dependent mechanism. Nat. Med. 2002, 8, 247–252.
  60. Albach, F.N.; Wagner, F.; Hüser, A.; Igel, J.; Joseph, D.; Hilbert, J.; Schoelch, C.; Padula, S.J.; Steffgen, J. Safety, pharmacokinetics and pharmacodynamics of single rising doses of BI 655064, an antagonistic anti-CD40 antibody in healthy subjects: A potential novel treatment for autoimmune diseases. Eur. J. Clin. Pharm. 2018, 74, 161–169.
  61. Goldwater, R.; Keirns, J.; Blahunka, P.; First, R.; Sawamoto, T.; Zhang, W.; Kowalski, D.; Kaibara, A.; Holman, J. A phase 1, randomized ascending single-dose study of antagonist anti-human CD40 ASKP1240 in healthy subjects. Am. J. Transplant. 2013, 13, 1040–1046.
  62. Espié, P.; He, Y.; Koo, P.; Sickert, D.; Dupuy, C.; Chokoté, E.; Schuler, R.; Mergentaler, H.; Ristov, J.; Milojevic, J.; et al. First-in-human clinical trial to assess pharmacokinetics, pharmacodynamics, safety, and tolerability of iscalimab, an anti-CD40 monoclonal antibody. Am. J. Transplant. 2020, 20, 463–473.
  63. Cordoba, F.; Wieczorek, G.; Audet, M.; Roth, L.; Schneider, M.A.; Kunkler, A.; Stuber, N.; Erard, M.; Ceci, M.; Baumgartner, R.; et al. A novel, blocking, Fc-silent anti-CD40 monoclonal antibody prolongs nonhuman primate renal allograft survival in the absence of B cell depletion. Am. J. Transplant. 2015, 15, 2825–2836.
  64. Kirk, A.D.; Burkly, L.C.; Batty, D.S.; Baumgartner, R.E.; Berning, J.D.; Buchanan, K.; Fechner, J.H., Jr.; Germond, R.L.; Kampen, R.L.; Patterson, N.B.; et al. Treatment with humanized monoclonal antibody against CD154 prevents acute renal allograft rejection in nonhuman primates. Nat. Med. 1999, 5, 686–693.
  65. Zhai, Y.; Meng, L.; Gao, F.; Busuttil, R.W.; Kupiec-Weglinski, J.W. Allograft rejection by primed/memory CD8+ T cells is CD154 blockade resistant: Therapeutic implications for sensitized transplant recipients. J. Immunol. 2002, 169, 4667–4673.
  66. Chavele, K.M.; Merry, E.; Ehrenstein, M.R. Cutting edge: Circulating plasmablasts induce the differentiation of human T follicular helper cells via IL-6 production. J. Immunol. 2015, 194, 2482–2485.
  67. Jordan, S.C.; Choi, J.; Kim, I.; Wu, G.; Toyoda, M.; Shin, B.; Vo, A. Interleukin-6, A Cytokine Critical to Mediation of Inflammation, Autoimmunity and Allograft Rejection: Therapeutic Implications of IL-6 Receptor Blockade. Transplantation 2017, 101, 32–44.
  68. Garbers, C.; Aparicio-Siegmund, S.; Rose-John, S. The IL-6/gp130/STAT3 signaling axis: Recent advances towards specific inhibition. Curr. Opin. Immunol. 2015, 34, 75–82.
  69. Kimura, A.; Kishimoto, T. IL-6: Regulator of Treg/Th17 balance. Eur. J. Immunol. 2010, 40, 1830–1835.
  70. Moore, P.A.; Belvedere, O.; Orr, A.; Pieri, K.; LaFleur, D.W.; Feng, P.; Soppet, D.; Charters, M.; Gentz, R.; Parmelee, D.; et al. BLyS: Member of the tumor necrosis factor family and B lymphocyte stimulator. Science 1999, 285, 260–263.
  71. Friebus-Kardash, J.; Wilde, B.; Keles, D.; Heinold, A.; Kribben, A.; Witzke, O.; Heinemann, F.M.; Eisenberger, U. Pretransplant serum BAFF levels are associated with pretransplant HLA immunization and renal allograft survival. Transpl. Immunol. 2018, 47, 10–17.
  72. Banham, G.; Prezzi, D.; Harford, S.; Taylor, C.J.; Hamer, R.; Higgins, R.; Bradley, J.A.; Clatworthy, M.R. Elevated pretransplantation soluble BAFF is associated with an increased risk of acute antibody-mediated rejection. Transplantation 2013, 96, 413–420.
  73. Pongpirul, W.; Chancharoenthana, W.; Pongpirul, K.; Leelahavanichkul, A.; Kittikowit, W.; Jutivorakool, K.; Nonthasoot, B.; Avihingsanon, Y.; Eiam-Ong, S.; Praditpornsilpa, K.; et al. B-cell activating factor, a predictor of antibody mediated rejection in kidney transplantation recipients. Nephrology 2018, 23, 169–174.
  74. Schuster, A.; Jung, B.; Hofbauer, J.; Kühne, L.; Zecher, D.; Banas, B.; Bergler, T. B-cell activating factor BAFF reflects patients’ immunological risk profile after kidney transplantation. Transpl. Immunol. 2017, 45, 35–41.
  75. Snanoudj, R.; Candon, S.; Roelen, D.L.; Jais, J.P.; Claas, F.H.; Legendre, C.; Chatenoud, L. Peripheral B-cell phenotype and BAFF levels are associated with HLA immunization in patients awaiting kidney transplantation. Transplantation 2014, 97, 917–924.
  76. Wang, X.Z.; Wan, Z.; Xue, W.J.; Zheng, J.; Li, Y.; Ding, C.G. B-Cell Activating Factor Predicts Acute Rejection Risk in Kidney Transplant Recipients: A 6-Month Follow-Up Study. Front. Immunol. 2019, 10, 1046.
  77. Woodle, E.S.; Alloway, R.R.; Girnita, A. Proteasome inhibitor treatment of antibody-mediated allograft rejection. Curr. Opin. Organ Transplant. 2011, 16, 434–438.
  78. Mulder, A.; Heidt, S.; Vergunst, M.; Roelen, D.L.; Claas, F.H. Proteasome inhibition profoundly affects activated human B cells. Transplantation 2013, 95, 1331–1337.
  79. Everly, M.J.; Everly, J.J.; Susskind, B.; Brailey, P.; Arend, L.J.; Alloway, R.R.; Roy-Chaudhury, P.; Govil, A.; Mogilishetty, G.; Rike, A.H.; et al. Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection. Transplantation 2008, 86, 1754–1761.
  80. Walsh, R.C.; Everly, J.J.; Brailey, P.; Rike, A.H.; Arend, L.J.; Mogilishetty, G.; Govil, A.; Roy-Chaudhury, P.; Alloway, R.R.; Woodle, E.S. Proteasome inhibitor-based primary therapy for antibody-mediated renal allograft rejection. Transplantation 2010, 89, 277–284.
  81. Ding, Y.; Francis, J.; Gautam, A.; Pelletier, L.; Sanchorawala, V.; Quillen, K. Durable renal response after combination of bortezomib, corticosteroids, rituximab, and plasmapheresis for late antibody-mediated renal transplant rejection. Clin. Nephrol. 2018, 89, 252–259.
  82. Tzvetanov, I.; Spaggiari, M.; Joseph, J.; Jeon, H.; Thielke, J.; Oberholzer, J.; Benedetti, E. The use of bortezomib as a rescue treatment for acute antibody-mediated rejection: Report of three cases and review of literature. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2012; Volume 44, pp. 2971–2975.
  83. Sureshkumar, K.K.; Hussain, S.M.; Marcus, R.J.; Ko, T.Y.; Khan, A.S.; Tom, K.; Vivas, C.A.; Parris, G.; Jasnosz, K.M.; Thai, N.L. Proteasome inhibition with bortezomib: An effective therapy for severe antibody mediated rejection after renal transplantation. Clin. Nephrol. 2012, 77, 246–253.
  84. Kolonko, A.; Słabiak-Błaż, N.; Karkoszka, H.; Więcek, A.; Piecha, G. The Preliminary Results of Bortezomib Used as A Primary Treatment for An Early Acute Antibody-Mediated Rejection after Kidney Transplantation-A Single-Center Case Series. J. Clin. Med. 2020, 9, 529.
  85. Abbas, K.; Mubarak, M.; Zafar, M.N.; Musharraf, W.; Imam, M.; Aziz, T.; Rizvi, A.H. Management of Plasma Cell-Rich Acute Rejection in Living-Related Kidney Transplant: Role of Proteasome Inhibitor. Exp. Clin. Transpl. 2019, 17, 42–46.
  86. Tasaki, M.; Saito, K.; Nakagawa, Y.; Ikeda, M.; Imai, N.; Ito, Y.; Sudo, M.; Ikezumi, Y.; Yamada, T.; Hasegawa, H.; et al. Bortezomib Eliminates Plasma Cells From a Renal Graft in Plasma Cell-Rich Acute Rejection. In Transplantation Proceedings; Elsevier: Amsterdam, The Netherlands, 2019; Volume 51, pp. 1732–1738.
  87. Jain, D.; Rajab, A.; Young, J.S.; Yin, D.; Nadasdy, T.; Chong, A.S.; Pelletier, R.P. Reversing donor-specific antibody responses and antibody-mediated rejection with bortezomib and belatacept in mice and kidney transplant recipients. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2020, 20, 2675–2685.
More