3. Complement activation plays an important role in graft damage. Reflect on strategies attempted at inactivation of the complement pathway emphasizing on the outcome of these trials.
Dear All Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement? Give examples
Riham Marzouk
2 years ago
Complement pathway is activated upon renal transplantation through the following: Ischemic reperfusion injury which produces ROS reactive oxidative stress factors which initiate complement pathway producing C5a and C5b Also, C3a and C3b are produced during complement activation pathway. Renal (tubules) and hepatic synthesis of C3. Collectin -11 which is soluble lectin expressed in kidney tissue and responsible for complement activation in the kidney, increased with ischemic stress and trigger complement activation. The C5a receptor antagonist has therapeutic effects in any autoimmune disease , C5a is a major inflammatory peptide in antibody mediated rejection so its block may be another hope against transplant associated inflammatory responses.
Christopher L. Nauser, Conrad A. Farrar and Steven H. Sacks. Complement Recognition Pathways in Renal Transplantation. JASN September 2017, 28 (9) 2571-2578.
C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi. The role of complement inhibition in kidney transplantation. British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17.
Abdulrahman Ishag
3 years ago
The role of complement in graft damage;
Complement is implicated in the following events ,that influence graft damage;
1-Ischemia reperfusion injury ;
2- Anti body mediated rejection
3- Cell mediated rejection
4- Post transplant TMA
Therapeutics targeting complement:
1-Human intravenous immunoglobulins (IV IG)
They inhibit complement activation by blocking the receptor for the Fc fragment of IG or the C1q component.
2-Eculizumab:
Inhibitor of C5 complement component, monoclonal antibody,
3-C1 esterase inhibitor
inhibits the activation of both the classical and lectin complement pathways
used in treatment of recurrent TMA post transplant .
Reference ;
1-Schinstock, C.A.; Mannon, R.B.; Budde, K.; Chong, A.S.; Haas, M.; Knechtle, S.; Lefaucheur, C.; Montgomery, R.A.; Nickerson, P.; Tullius, S.G.; et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation. Transplantation 2020, 104, 911–922.
3- Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant (2015) 15(5):1293–302.
4- Gonzalez Suarez, M.L.; Thongprayoon, C.; Mao, M.A.; Leeaphorn, N.; Bathini, T.; Cheungpasitporn, W. Outcomes of Kidney Transplant Patients with Atypical Hemolytic Uremic Syndrome Treated with Eculizumab: A Systematic Review and MetaAnalysis. J. Clin. Med. 2019, 8, 919.
5-Vo, A.A.; Zeevi, A.; Choi, J.; Cisneros, K.; Toyoda, M.; Kahwaji, J.; Peng, A.; Villicana, R.; Puliyanda, D.; Reinsmoen, N.; et al. A Phase I/II Placebo-Controlled Trial of C1-Inhibitor for Prevention of Antibody-Mediated Rejection in HLA Sensitized Patients. Transplantation 2015, 99, 299–308.
Ahmed Omran
3 years ago
Complement system has 3 pathways for activation; Classic (Ag-Ab complex), Lectin pathway(microorganism)and alternative pathway.
ABMR: plasma cells produce antibodies which attach to antigen and initiate the classic pathway leaving its effect in the graft (C4d deposition in peritubular capillaries).
TMA post-transplant produce effect through complement pathway.
T-cell mediated rejection occurs as APC produce c3a and c5a in the activation of cellular immunity. Drugs interfering with complement system
IVIG which produces its effect through binding to fc region on IgG and inhibit complement activation and usually used in ABMR.
Eculizumab: C5 inhibitor as being very expensive , few studies were done and showed no beneficial impact in preventing rejection.
C1estrase inhibitor which acts against both Lectin and classic pathways.
Under trials: BIVV009,Rituximab, Ides. Compstatin family inhibitors.
s CR., TT30., C5aR1 antagonist., Cobra venom factor. Can graft injury happen without activation of complement?
Graft injury can occur away from complement activation.
Anti-HLA-class I antibodies can produce pathological effects ending with changes in the vascular intima and chronic rejection.
Without complement activation, C4d-negative AMR together with high anti-AT1R result in severe vascular rejection.
Reference
Loupy, A; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
. American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions,
Haas .M et al, 28 January 2014
saja Mohammed
3 years ago
antiHLA class 1,11 antigens DSA antibodies can lead to garft damage through the complement activation but still about 40% to 50% of graft rejection that associated with severe vascular changes, such as fibrinoid necrosis, are C4d negative,due to the non–complement fixing antibodies. like AT1R-antibodies (AT1R-Ab) can promote antibody-mediated rejection (AMR) either alone or together with HLA-DSA(1).
reference:
1-Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation Aurelie Philippe1,2:Kidney Int Rep (2019) 4, 510–512; https://doi.org/10.1016/j.ekir.2019.01.012.
Hamdy Hegazy
3 years ago
Complement system is activated by three pathways; Classic (Ag-Ab complex), Lactin pathway(microorganism)and alternative pathway. ABMR: plasma cells form antibodies which attach to antigen and start the classic pathway leaving its fingerprint in the graft (C4d deposition in peritubular capillaries).
TMA post-transplant takes effect via complement pathway.
T-cell mediated rejection happens when APC produce c3a and c5a in the activation of cellular immunity.
Drugs which interfere with complement system IVIG…which bind to fc region on IgG and inhibit complement activation and widely used in ABMR Eculizumab: C5 inhibitor because of its very expensive drug few studies were addressed and show no benefits in preventing rejection C1estrase inhibitor which inhibits both Lactin and classic pathways. Under trials: Rituximab, BIVV009, Ides., Mirococept., Compstatin family inhibitors. sCR., TT30., C5aR1 antagonist., cobra venom factor. Can graft injury happen without activation of complement? Yes, it can happen without complement activation. Anti-HLA-class I antibodies can produce pathological effects ending with changes in the vascular intima and chronic rejection. C4d-negative AMR with high anti-AT1R produces a severe vascular rejection without complement activation.
Reference 1- Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226. 2- Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
3- Valenzuela NM, McNamara JT, Reed EF. Antibody-mediated graft injury: complement-dependent and complement-independent mechanisms. Curr Opin Organ Transplant 2014;19:33-40. 4- American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions, Haas.M et al, 28 January 2014
Dalia Ali
3 years ago
The complement system is a series of protein kinases that are sequentially activated and culminate in the formation of the membrane attack complex (MAC).
The MAC comprises complement components C5 to C9, which are inserted into the cell membrane (pathogen or host), disrupt- ing integrity and causing cell lysis .
In addition, many proximal complement components may augment the immune response to the allograft. The complement system may be activated by three path- ways: the classical pathway, the alternative pathway, and the MBL pathway. IgM or IgG immune complexes activate the classical pathway, and hence this pathway may become acti- vated during antibody-mediated rejection (see section on B Cell Activation). The alternative pathway is constitutively active and must be controlled by a series of regulatory protein
Reference
Kidney
Transplantation Principles and Practice
EIGHTH EDITION
Stuart J. Knechtle, MD, FACS
Mahmoud Hamada
3 years ago
the detection of C4d in kidney allograft biopsies usually indicate acute ABMR.
C4d is the byproduct of classic complement pathway activation.
Eculizumab, a monoclonal human antibody (targeting C5), may be used for prevention of complement mediated acute rejection, however, it is still expensive drug.
T cell may cause graft injury without complement activation.
Wael Hassan
3 years ago
Complement :protein sensitized in liver and activated by many ways as (AG-AB complex)classic pathway or alternative pathway by bacterial endotoxin .
In kidney transplantation it play a role in innate immunity by activate macrophages
Or also by make cell lysis by AG-AB complex.
Eclizumab anti c5( used in ttt of atypical HUS) will corrupt the c5 so stop classical & alternative pathway of its activation .also c1 inhibitor .
Graft injury can happen without complement activation by AG AB complex it self ( deposition in kidney) also nephrotoxicity of CNI on graft kidney (TMA)
Infection by BK virus or CMV
Abdullah Raoof
3 years ago
complement systemis important in Ab mediated rejection . therefore the component of the complement system has become apotential therapuetic targets .
C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking not only C1 complex components , as the name suggests, but also MASP-1 and MASP-2 . It is, therefore, an effective inhibitor of both classical and lectin pathway activation . Earlier study on non-human primate model showed a potential for inhibition of acute antibody-mediated rejection with C1-inhibitor . Very recently, a double-blind study assessed the safety of C1-inhibitor in kidney transplant recipients and suggested the positive effect of this treatment regarding the C1q+HLA antibody levels reduction and antibody-mediated rejection occurrence . The possible use of complement inhibitor based on viral coat proteins is also a subject of ongoing research .
Eculizumab is a humanized monoclonal antibody capable of blocking the cleavage of C5 complement component, thus preventing the release of anaphylatoxin C5a and the assembly of MAC . The C5 component participates in all three main pathways of complement activation; therefore, the blockade of its cleavage can have a positive influence on various medical conditions regardless of the active pathway. Eculizumab was shown to be an effective treatment for paroxysmal nocturnal hemoglobinuria and aHUS including its recurrence in kidney transplantation . Since the complement system is a prominent factor during antibody-mediated rejection, the effects of eculizumab were studied with regard to this condition as well. Stegall et al. studied the effects of terminal complement inhibition on patients with a positive crossmatch and concluded that eculizumab is capable of decreasing the incidence of acute antibody-mediated rejection. Some success in treating the acute antibody-mediated rejection in renal allograft was also reported in several case reports . Extended follow-up study, however, showed that eculizumab treatment has no effect on chronic antibody-mediated rejection in recipients with significant amount of DSA. Moreover, there are known cases of C4d-negative antibody-mediated rejection where the use of eculizumab was ineffective . Similarly, the existence of IgM-mediated acute antibody-mediated rejection was described in patients treated with eculizumab . Therefore, C5 inhibition might be efficacious in the treatment of acute antibody-mediated rejection at least in some cases; however, these effects are lost in preventing chronic rejection development.
references
. Davis AE, III, Mejia P, Lu F. Biological activities of C1 inhibitor. Mol Immunol (2008) 45(16):4057–63.10.1016/j.molimm.2008.06.028
Paréj K, Dobó J, Závodszky P, Gál P. The control of the complement lectin pathway activation revisited: both C1-inhibitor and antithrombin are likely physiological inhibitors, while α2-macroglobulin is not. Mol Immunol (2013) 54(3–4):415–22.10.1016/j.molimm.2013.01.009
Tillou X, Poirier N, Le Bas-Bernardet S, Hervouet J, Minault D, Renaudin K, et al. Recombinant human C1-inhibitor prevents acute antibody-mediated rejection in alloimmunized baboons. Kidney Int (2010) 78(2):152–9.10.1038/ki.2010.75
Vo AA, Zeevi A, Choi J, Cisneros K, Toyoda M, Kahwaji J, et al. A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation (2015) 99(2):299–308.10.1097
. Mauriello CT, Pallera HK, Sharp JA, Woltmann JL, Jr, Qian S, Hair PS, et al. A novel peptide inhibitor of classical and lectin complement activation including ABO incompatibility. Mol Immunol (2013) 53(1–2):132–9.10.1016/j.molimm.2012.07.012
Sharp JA, Hair PS, Pallera HK, Kumar PS, Mauriello CT, Nyalwidhe JO, et al. Peptide inhibitor of complement C1 (PIC1) rapidly inhibits complement activation after intravascular injection in rats. PLoS One (2015) 10(7):e0132446.10.1371/journal.pone.0132446
Fakhouri F, Hourmant M, Campistol JM, Cataland SR, Espinosa M, Gaber AO, et al. Terminal complement inhibitor eculizumab in adult patients with atypical hemolytic uremic syndrome: a single-arm, open-label trial. Am J Kidney Dis (2016) 68(1):84–93.10.1053/j.ajkd.2015.12.034
Stewart ZA, Collins TE, Schlueter AJ, Raife TI, Holanda DG, Nair R, et al. Case report: eculizumab rescue of severe accelerated antibody-mediated rejection after ABO-incompatible kidney transplant. Transplant Proc (2012) 44(10):3033–6.10.1016/j.transproceed.2012.03.053
Chehade H, Rotman S, Matter M, Girardin E, Aubert V, Pascual M. Eculizumab to treat antibody-mediated rejection in a 7-year-old kidney transplant recipient. Pediatrics (2015) 135(2):e551–5.10.1542/peds.2014-2275
Tran D, Boucher A, Collette S, Payette A, Royal V, Senécal L. Eculizumab for the treatment of severe antibody-mediated rejection: a case report and review of the literature. Case Rep Transplant (2016) 2016:9874261.10.1155/2016/9874261
Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant (2015) 15(5):1293–302.
Burbach M, Suberbielle C, Brochériou I, Ridel C, Mesnard L, Dahan K, et al. Report of the inefficacy of eculizumab in two cases of severe antibody-mediated rejection of renal grafts. Transplantation (2014) 98(10):1056–9.10.1097/
Bentall A, Tyan DB, Sequeira F, Everly MJ, Gandhi MJ, Cornell LD, et al. Antibody-mediated rejection despite inhibition of terminal complement. Transpl Int (2014) 27(12):1235–43.
Ben Lomatayo
3 years ago
Complement pathways plays important role in transplantation e.g. CDCXM is complement based test.
some strategies attempted to inactivate complement include the use of C5 inhibitor known as Eculizumab.
You can still have graft injury without complement involvement e.g. mechanism of AMR it self, part involve complement, and the other part involve direct antibody injury, and injury caused by cells like macrophages. We of also have non- complement fixing antibodies. T cell mediated rejection is another examples of graft injury without complement involvement.
Ahmed mehlis
3 years ago
The complement cascade is comprised of more than 30 soluble and cell-bound proteins .
●The complement system is activated through three distinct pathways: the classical pathway (CP), lectin pathway (LP), and alternative pathway (AP).
●complement activation
Complement activation in antibody-mediated rejection. Antibody-mediated rejection is caused by binding of antibodies to human leukocyte antigens (HLA) expressed on endothelial cells of the transplanted organ. The antibodies (referred to as donor specific antibodies, or DSA) activate the classical pathway of complement. Classical pathway activation causes the cleavage of C4, and one of the resultant C4 fragments (C4b) is covalently attached to target surfaces. C4b comprises part of the classical pathway C3 convertase, C4b2a. C3b can become covalently attached to target cells, similar to C4b. A protease called factor I (FI) controls complement activation by cleaving the C4b and C3b molecules, thereby stopping convertase activity. Although they are no longer catalytically active, the C4d and C3dg fragments remain bound to the target cells and can be detected by immunostaining of tissue biopsies
●complement inhibitors
In spite of promising pre-clinical data, a trial that enrolled 27 kidney transplant patients at high risk of DGF who were randomized to treatment with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benefit with treatment . Another clinical trial is ongoing, however, in which kidneys treated with an agent that attaches a complement regulator to cell membranes . This approach was previously shown to be beneficial in a rat kidney transplant model .
●eculizumab
treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in kidney transplant recipients with AMR.
●C1-INH is a substrate-like serine protease inhibitor that blocks several proteases, including C1r, C1s, and the MASPs. Originally used as a replacement therapy for patients with hereditary angioedema who have deficiency of C1-INH protein, it has also been tested as a treatment of AMR . C1-INH appeared to be beneficial in a small trial of six AMR patients who were refractory to conventional therapy ,
this approach holds promise as a novel treatment for transplant patients.
Although eculizumab and C1-INH have shown promise in case reports and small series, their role in transplant medicine requires further study. Many additional anti-complement therapeutics are in clinical development, and some of these new drugs block individual activation pathways or specific components of the complement cascade. This could potentially allow clinicians to block the parts of the complement cascade involved in allograft injury while leaving other parts of the cascade active.
However, testing these new agents in the transplant setting poses several challenges. First, all transplant recipients are treated with multiple immunosuppressive drugs. Thus, new drugs will need to be tested as add-on treatments to these other agents. Second, even within a single diagnosis, such as AMR, there is patient heterogeneity. Complement activation may not be an important part of C4d-negative AMR, for example. The development of new complement biomarkers may therefore be critical for selecting patients most likely to benefit from complement inhibitors, and for discerning a response to treatment.
●references
.Zhuang Q, Lakkis FG. Dendritic cells and innate immunity in kidney transplantation. Kidney Int. (2015)
.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6788431/#!po=7.00935
Last edited 3 years ago by Ahmed mehlis
fakhriya Alalawi
3 years ago
The complement cascade is an important part of the innate immune system. Studies have shown that complement activation contributes to allograft injury in several clinical settings, including ischemia/reperfusion injury and antibody-mediated rejection. Furthermore, the complement system plays a critical role in modulating the responses of T cells and B cells to antigens. Therapeutic complement inhibitors, therefore, may be effective for protecting transplanted organs from several causes of inflammatory injury. Although several anti-complement drugs have shown promise in selected patients, the role of these drugs in transplantation medicine requires further study. Treatment with the C5 inhibitor led to long-term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft.
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR. However, larger series in transplant patients have not shown a consistent benefit, however, and the role of eculizumab for preventing or treatment AMR is not yet clear. C1-INH is a substrate-like serine protease inhibitor that blocks several proteases, including C1r, C1s, and the MASPs. Originally used as replacement therapy for patients with hereditary angioedema who have a deficiency of C1-INH protein, it has also been tested as a treatment of AMR. C1-INH appeared to be beneficial in a small trial of six AMR patients who were refractory to conventional therapy.
Pre-clinical work has shown that other complement inhibitory agents may be useful in the transplant setting, including an AP inhibitor, an LP inhibitor, and C5a blockade. Many new complement inhibitory drugs are in clinical development, some of which will likely soon become available for clinical use.
Reference:
1. Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019 Oct 4;10:2380.
2. Viglietti D, Gosset C, Loupy A, Deville L, Verine J, Zeevi A, et al. C1-inhibitor in acute antibody-mediated rejection non-responsive to conventional therapy in kidney transplant recipients: a pilot study. Am J Transplant. (2015) 16:1596–603.
Shereen Yousef
3 years ago
complement activation contributes to allograft injury in several ways including
ischemia/reperfusion injury.And antibody mediated rejection.
Furthermore, the complement system plays critical roles in modulating the responses of T cells and B cells to antigens.
Therapeutic complement inhibitors, therefore, may be effective for protecting transplanted organs from several causes of inflammatory injury.
Although the complement system is an important effector mechanism for antibody-mediated cytotoxicity, that is only one of its functions. The complement system can be activated in an antibody-independent fashion
The transplanted organ is exposed to recipient complement proteins as soon as it is reperfused. Conversely, complement proteins and fragments generated within the allograft enter the systemic circulation.
Complement fragments also modulate T cell differentiation and the B cell response to antigens.
Consequently, this system modulates the adaptive immune response, mediates many of the downstream effects of B and T cell immunity, and can function independently of the adaptive immune response. Furthermore, the complement cascade interacts with other biologic systems, including toll-like receptors, the inflammasome, and the clotting cascade .
The complement system is activated through three distinct pathways: the classical pathway (CP), lectin pathway (LP), and alternative pathway (AP). These activation pathways can be engaged by different pathologic processes in the allograft, including donor brain death, I/R injury, and antibody mediated rejection.
The CP is activated by antibodies bound to their target ligands. This may be particularly important in those transplant recipients with donor specific antibodies (DSA) reactive to polymorphic human leukocyte antigens (HLA) expressed on endothelial cells of the allograft.
Complement activation through all three pathways leads to cleavage of the C3 protein, generating the C3a and C3b fragments.
C3b binds to nearby surfaces, thereby marking, or “opsonizing,” target cells and surfaces.
Full complement activation also leads to cleavage of C5, generating soluble C5a and the larger C5b fragment. C5b seeds the formation of the membrane attack complex (MAC, or C5b-9), a multimeric complex that forms a pore in target cells and can cause target cell activation or lysis .
Complement Receptors
Although the MAC directly affects target cells, most of the biologic effects of complement system are mediated by receptors for the various fragments. The C3a receptor (C3aR) and C5a receptors (C5aR1 and C5aR2) ,C5aR1 expression, increases in rejecting murine renal allografts .
Once complement is activated within a transplanted organ it can have direct and indirect pathologic effects. multiple different biologically active complement fragments are generated. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation.
In kidney ischemia, activation primarily occurs in the tubulointerstitium whereas in AMR activation occurs in the peritubular capillaries .
Patients with DSA that bind to C1q or to which C3d is bound are at greater risk of developing AMR and they have a worse overall prognosis .
Interestingly, treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
Complement Therapeutics
Eculizumab
IT been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR .
It has also been used in transplant recipients with post-transplant aHUS recurrences .
C5aR1 antagonist :there is pre-clinical data showing that C5aR1 blockade may be a beneficial treatment for rejection , this approach holds promise as a novel treatment for transplant patients.
C1-INH :has also been used to prevent AMR, and it is being tested for treatment of the disease in an ongoing clinical trial.
Pre-clinical work has shown that other complement inhibitory agents may be useful in the transplant setting, including an AP inhibitor (63), a LP inhibitor (56), and C5a blockade . Many new complement inhibitory drugs are in clinical development, some of which will likely soon become available for clinical use .
Monica Grafals and Joshua M. Thurman.The Role of Complement in Organ Transplantation.Front Immunol. (2019); 10: 2380.
Mahmoud Rabie
3 years ago
The complement system has an important role in kidney transplantation especially in ABMR, ischemia reperfusion injury, and recurrence of the original disease as in case of C3 glomerulopathy, atypical HUS, and antiphospholipid syndrome.
The risk of complement mediated ischemia reperfusion injury increases in case of deceased donor due to the presence of soluble C5b-9 in deceased donor graft, and increased expression of complement genes in the deceased donor, and also due to brain death induced complement activation.
The presence of C4d on peritubular capillaries using immunofluorescence is a positive marker for diagnosis of ABMR and also for grading of its severity.
Graft outcome could be predicted by the detection of C3d binding DSA.
There are 3 available ways to inhibit the complement activation cascade:
A monoclonal antibody directed against C5 ( eculizumab ), and 2 plasma derived C1 inhibition.
In renal transplantation, eculizumab can be used to prevent ABMR and for treatment and prevention of aHUS, and it showed a promising results.
Complement is part of innate immunity ,there are three pathways of complement activation:
* the classical pathway, which is triggered directly by pathogen or indirectly by antibody binding to the pathogen surface .
*the MB-lectin pathway.
*and the alternative pathway, which also provides an amplification loop for the other two pathways.
If complement is activated within a transplanted organ it can have direct and indirect pathologic effects. Multiple different biologically active complement fragments are generated. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation. The location of complement activation will vary in different settings. In kidney ischemia, for example, activation primarily occurs in the tubulointerstitium , whereas in AMR activation occurs in the peritubular capillaries . The location of activation determines which cells will be directly affected by MAC or opsonization with C3b. Soluble fragments such as C3a and C5a can have more distant effects, but the site of activation may affect their access to the circulation and peripheral blood cells. It is useful to understand the contribution of the individual complement fragments to injury, as drugs that target specific fragments are in development.
~Complement in Ischemia/Reperfusion Injury:
Several studies have shown that complement is activated after I/R, although the mechanisms may vary between different organs.
Complement activation in the ischemic heart and intestine may be initiated by immunoglobulin, but it primarily involves the LP . In the kidney, complement activation primarily involves the AP and does not require immunoglobulin . Studies in which kidneys from C3 deficient mice were transplanted into wild-type recipients revealed that the allograft itself may be an important source of complement proteins involved in tubulointerstitial activation .
~Complement inhibitory drugs have proven effective in several pre-clinical models of I/R injury. An inhibitory antibody to C5 and a small molecule C5a receptor antagonist were each protective in models of cardiac and kidney I/R injury. LP blockade was protective in models of kidney and cardiac I/R injury .A monoclonal antibody that inhibits the AP was protective in a model of warm I/R injury of the kidney . This same drug also prevented I/R injury in a mouse kidney transplant model, and it also reduced T cell mediated rejection of the organs .
~Antibody Mediated Rejection
In patients with acute and chronic AMR, DSA binds to donor HLA expressed on endothelial cells and activates the CP. Complement activation on the endothelial cell surface is believed to be an important cause of injury to the capillaries .The diagnosis of AMR is based on detecting DSA in the plasma, microvascular inflammation on a biopsy (e.g., glomerulitis or peritubular capillaritis in allografts), and C4d deposition in the peritubular capillaries . However, the diagnostic criteria have been modified to account for C4d-negative cases . It is not known whether the C4d-negative cases of AMR are caused by non-complement-mediated injurious effects of the DSA, or whether it simply reflects variability in the ability to detect the C4d.
Because complement activation by the DSA is such an important component of AMR, assays have been developed to distinguish the complement activating potential of DSA in the circulation. These assays identify immunoglobulin that binds to specific HLA types, and also tests whether the detected antibodies bind to C1q or carry a C3d molecule . Patients with DSA that bind to C1q or to which C3d is bound are at greater risk of developing AMR and they have a worse overall prognosis . These findings highlight the importance of the complement system in AMR, and potentially provide a test for identifying patients at risk of AMR. An inhibitory antibody to C5 was protective in a model of heart transplantation in highly sensitized mice, supporting the importance of complement activation in the pathogenesis of microvascular injury .Interestingly, treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
References:
1. Janeway CA, Jr. How the immune system works to protect the host from infection: a personal view. Proc Natl Acad Sci USA. (2001) 98:7461–8. 10.1073/pnas.131202998 .
2. Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. (2007) 449:819–26. 10.1038/nature06246.
3. Palm NW, Medzhitov R. Pattern recognition receptors and control of adaptive immunity. Immunol Rev. (2009) 227:221–33. 10.1111/j.1600-065X.2008.00731.
Nasrin Esfandiar
3 years ago
Complement has three activation pathways: Classic, alternative and lectin. All are contributing in kidney transplantation. Role of complement begins before transplantation in primary kidney disease such as; C3 glomerulopathy or aHUS and hemodialysis and continues with inflammation associated with brain death, stress of surgery and ischemic-reperfusion injury. In addition its activation can occur during and after organ donation especially in deceased donor. Ischemia- reperfusion induces heparanase and metalloproteinase and causes damage to endothelial cells and loss of complement regulators. Presence of DSA against endothelial cells may activate complement. Dysregulation of alternative complement system may cause damage or sometimes recurrence of primary kidney disease in allograft. Complement activation has important role in humoral or cellular rejection and chronic AMR. Some complement fragments have chemotactic or thrombogenic and induce apoptosis and necrosis.
Anti-ABO or DSAs can trigger complement and cause allograft injury. C4d positive staining shows classic pathway activation of complement system in ABMR. DSAs with C3 and C1q binding capacities can be more harmful to graft. Strategies for complement inhibition in graft:
-Better quality of graft by sCR1 and C1INH and improvement in graft preserving solution
-Treatment of complement-dependent ABMR with eculizumab (anti-C5 monoclonal antibody), C1INH (Berinert), C1 monoclonal antibody B1VV009 or C3 inhibitor (CP40) as an addition to standard therapy (plasmapheresis, IVIG and rituximab). A recombinant endopeptidase (IdeS) can cleave hinge region of IgG subclasses. So, IgG cannot bind to C1q. Clinical trials with APT070 (Mirococept) and Compstatin family inhibitors show benefits with complement inhibition.
1. Biglarnia, AR., Huber-Lang, M., Mohlin, C. et al. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol14, 767–781 (2018)
Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019;10:2380
Alyaa Ali
3 years ago
Complement activation have three pathways which differ according their recognition targets
antibodies are triggers of the classical pathway , carbohydrates for the lectin pathway and permanent low level of activation for the alternative pathway role of complement in transplantation:
Ischaemia -reperfusion injury with activation of lectin pathway due to endothelial injury and amplification of alternative pathway.
Antibody mediated rejection with activation of the classical pathway against antigens of MHC and ABO group and donor specific antigens.
Recurrence of native kidney disease through activation of alternative pathway :
recurrence of haemolytic uraemic disease, C3 glomerulopathy. strategies attempted at inactivation of the complement pathway Currently, there are three complement inhibitors: a monoclonal antibody directed against C5 (eculizumab) and two plasma-derived C1 inhibitors. Compounds blocking the terminal pathway: Eculizumab a monoclonal antibody directed against C5: it inhibits C5 cleavage by the
C5 convertase with stopping of synthesis of C5a and C5b-9 which are the pro-inflammatory, prothrombotic and lytic functions of complement this drug has shown efficacy in treatment and prevention of atypical haemolytic uraemic syndrome and antiphospholipid syndrome, some efficacy in the prevention of antibody-mediated rejection and so far no efficacy in the prevention of delayed graft function
in issue of preventing of delayed graft function : study done to compare patients used two doses of eculizumab and another group on placebo and there is no significant difference between two groups in prevention of delayed graft function.
in issue of prevention of antibody mediated rejection at 1 year, the incidence of acute AMR was 7.7% in the eculizumab group compared to 41.2% in the historical control group Graft and patient survival were excellent in both groups. Compounds blocking the classical activation pathway
C1 inhibition reduces the release of chemotactic microvesicles, which are increasingly
recognized in the settings of transplantation . studies demonstrated the efficacy of blocking C1 with a recombinant C1 inhibitor used to prevent acute ABMR
study done to compare patients receive either placebo or plasma-derived human C1 inhibitor (Berinert®, 20 IU/kg/dose) twice weekly for a total seven doses.No patient developed rejection during the study in the C1-inhibitor group, but one developed rejection in the control group.Tolerance of the drug was acceptable.
Another study used another plasma-derived C1 inhibitor to treat acute
antibody-mediated rejection following kidney transplantation in a randomized double-blind placebo controlled pilot study. The treatment was well tolerated. With regard to efficacy, no patient developed transplant glomerulopathy in the treated group compared with 42% in the control group.
Finally, plasma-derived C1 inhibitor (Berinert®) was used in a pilot study in patients with acute ABMR who were resistant to the standard-of-care treatment. there was a trend towards better renal function at the end of the follow-up period. References
Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019;10:2380. Legendre C, Sberro-Soussan R, Zuber J, Frémeaux-Bacchi V. The role of complement inhibition in kidney transplantation. Br Med Bull. 2017 Dec 1;124(1):5-17.
Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients Transplantation 2016;99:299–308
Montgomery RA, Orandi BJ, Racusen L, et al. Plasma derived C1 esterase inhibitor for acute antibody mediated rejection following kidney transplantation: results of a randomized double-blind placebo controlled pilot study. Am J Transplant 2016;16:3468–78.
Viglietti D, Gosset C , et al. C1 inhibitor in acute antibody-mediated rejection non responsive to conventional therapy in kidney transplant recipients:a pilot study. Am J Transplant 2016;16:1596–603
Assafi Mohammed
3 years ago
Role of Complement in Graft Damage:
The complement system can be activated in an antibody-independent fashion . Complement fragments also modulate T cell differentiation and the B cell response to antigens. Consequently, this system modulates the adaptive immune response, mediates many of the downstream effects of B and T cell immunity, and can function independently of the adaptive immune response.
The complement cascade interacts with other biologic systems, including toll-like receptors, the inflammasome, and the clotting cascade.
Complement activation through all three pathways leads to cleavage of the C3 protein, generating the C3a and C3b fragments. C3b has a reactive thioester bond that can bind covalently to hydroxyl and amine groups on nearby surfaces, thereby marking, or “opsonizing,” target cells and surfaces. Full complement activation also leads to cleavage of C5, generating soluble C5a and the larger C5b fragment. C5b seeds the formation of the membrane attack complex (MAC, or C5b-9), a multimeric complex that forms a pore in target cells and can cause target cell activation or lysis.
Once complement is activated within a transplanted organ it can have direct and indirect pathologic effects. As outlined above, multiple different biologically active complement fragments are generated. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation. The location of complement activation will vary in different settings. In kidney ischemia, for example, activation primarily occurs in the tubulointerstitium , whereas in AMR activation occurs in the peritubular capillaries . The location of activation determines which cells will be directly affected by MAC or opsonization with C3b. Soluble fragments such as C3a and C5a can have more distant effects, but the site of activation may affect their access to the circulation and peripheral blood cells. It is useful to understand the contribution of the individual complement fragments to injury, as drugs that target specific fragments are in development.
\
Although all transplants might cause DSAs to be produced (unless inhibited by immunosuppression), only organ grafts (and not cellular grafts) are susceptible to antibody-mediated damage. Antibodies and complement mainly attack endothelial cells of organ transplants because antibodies and complement are found predominantly inside blood vessels.
Although antibodies and complement can penetrate blood vessel walls, they do so only slowly, and parenchymal cells and cellular transplants are relatively unaffected by DSAs.
The impact of complement on living cells has also been found to depend on the kinetics of activation, rather than on the amount activated or “fixed,” because, over time, cells acquire resistance to complement-mediated injury. Therefore, when antibody and complement are fixed acutely, they might induce cell death and rejection, but, over time, they might induce resistance to cell death and accommodation.Bound antibody can facilitate cellular immunity and cellular rejection, and it can block or suppress cellular immunity and rejection in tumor transplants (i.e., enhancement)
Complement activation in the donor, the graft and the recipient before, during and after transplantation is a major cause of damage to the kidney transplant.
Ischaemia and subsequent reperfusion of the graft is the most important mechanism that triggers complement activation; reperfusion is generally regarded as the most detrimental phase of the transplantation process.
Following transplantation, complement has a role in innate immunological and inflammatory processes that further damage the graft and result in a gradual decrease in its functional mass.
Complement-targeted strategies might have a role in optimizing graft quality as well as in the treatment of antibody-mediated rejection, induction of accommodation and modulation of the adaptive immune response
In patients with acute and chronic AMR, DSA binds to donor HLA expressed on endothelial cells and activates the CP. Complement activation on the endothelial cell surface is believed to be an important cause of injury to the capillaries . The diagnosis of AMR is based on detecting DSA in the plasma, microvascular inflammation on a biopsy (e.g., glomerulitis or peritubular capillaritis in allografts), and C4d deposition in the peritubular capillaries . However, the diagnostic criteria have been modified to account for C4d-negative cases . It is not known whether the C4d-negative cases of AMR are caused by non-complement-mediated injurious effects of the DSA, or whether it simply reflects variability in the ability to detect the C4d.
local production of complement proteins increases after ischemia, and that expression of these proteins by the allograft is associated with T cell mediated rejection.
The standard immunosuppressive medications used to prevent transplant rejection do not directly block the complement cascade. Thus, complement inhibitory drugs may be useful adjuncts to the currently available anti-rejection drugs in several different clinical settings.
Ramy Elshahat
3 years ago
Complement system is part of innate immunity which is activated by three pathways
Classic(antigen antibody complex)lactin pathway(microorganism)and alternative pathway
All the 3 pathways end by common pathway including c3 , c4 and c5 cleavage and formation of MAC complex and cell lysis
The role of complement in graft failure is appear in 3 main situations
1)in humoral rejection…plasma cells form antibodies which attach to antigen and start the classic pathway giving its fingerprint over the graft by c4d deposition in peritubular capillaries
2)in TMA post transplant which need to be differentiate what is its cause as it may be recurrence of 1ry kidney disease,CNI induced or ABMR
3)in Tcell mediated..some studies showed that APC produce c3a and c5a in the activation of cellular immunity
Drugs which interfere with complement system
IVIG…which bind to fc region on IgG and inhibit complement activation and widely used in ABMR
eculizumab…c5 inhibitor because of its very expensive drug few studies were addressed and show no benefits in preventing rejection
And finally c1estrase inhibitor which inhibits both lactin and classic pathways
Reference
(1) Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
(2) Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
Mohammed Sobair
3 years ago
Complement in kidney transplantation:
The role of complement in various aspects of kidney transplantation has been outlined,
including the following conditions (1, 3) :
Ischemia-reperfusion injury,
Antibody-mediated rejection,
Recurrence of native kidney disease such as atypical haemolytic uremic syndrome,
C3 glomerulopathy,
Anti-phospholipid syndrome.
ABO-incompatible transplantation.
The complement system is primarily a host defense mechanism, but it causes
injury to tissue cells through the formation of the membrane attack complex (C5b–
C9), which leads to cell lysis and cell death, and through the stimulation of
inflammation and antigen-specific immunity..
In most vital organs, ischemia–reperfusion injury is mediated by complement
components C5a and C5b–C9 and, as shown in a mouse kidney transplant model,
this is strongly dependent on tissue-mediated production of C3.
The lectin pathway of complement activation is a common trigger of ischemia–
reperfusion injury in several organ models (initiated either by direct binding of the
lectin to ischemic tissue or via natural IgM), and the alternative pathway may
significantly increase the deposition of complement at the site of activation.
Effective priming of CD4+ T cells that mediate graft rejection requires C3a and
C5a, which are produced by dendritic cells and stimulate the presentation of
alloantigen’s and the differentiation of naive CD4+ T helper (TH) cells to TH1 cells.
The reactivity of antigen-primed CD4+ and CD8+ T cells against donor cells is
enhanced by the peripheral synthesis of complement and the generation of the
effectors C3a, C3b and C5a.
Antibody-mediated rejection requires complement (specifically C3) not only for the
efficient priming of B cells against alloantigen’s but also for the induction of
inflammation and thrombosis (by C5a and C5b–C9) at the site of antibody binding
in the graft.(2)
Emerging therapeutic approaches include targeting the donor organ with
complement regulators to prevent stress-induced injury and sensitization of the
recipient, and treatment of the recipient to prevent complement-mediated vascular
injury during antibody-mediated rejection and the recurrence of haemolytic–
uremic syndrome.
Complement inhibition after kidney transplantation:
By blocking:
(a) The activation pathways.
(b) The amplification loop.
(c) The anaphylatoxins .
(d) The terminal pathway.
Currently, there are three available complement inhibitors: a monoclonal antibody
directed against C5 (Eculizumab) and two plasma-derived C1 inhibitors.
Eculizumab is a recombinant fully humanized hybrid IgG2/IgG4 monoclonal antibody that
is directed against the human complement component C5.
Eculizumab has been used mainly to prevent delayed graft function, in acute antibody-
mediated rejection and for treatment/prevention of aHUS recurrence and
antiphospholipid syndrome.(3).
a multi-center trial that began enrollment in 2013 evaluated the role of Eculizumab in the
treatment of biopsy-proven AMR and acute graft dysfunction, study drug’s failure to
improve allograft function assessed by estimated glomerular filtration rate (eGFR) 3
months after transplantation.(4).
Role of C1-INH in the treatment of AMR was explored by two recent studies, Viglietti et
al. treated six patients who had AMR that was not responsive to SOC therapy with C1
-INH ,At the end of the 6-month follow-up period, the authors reported significant
improvement in mean eGFR. Fewer patients demonstrated C4d deposition on allograft
biopsies and circulating C1q fixing DSAs. However, other histologic features, like
chronic glomerulopathy, were unchanged (5).
Reference:
1- Ch. Legendre, R. Sberro-Soussan et al. The role of complement inhibition in kidney
transplantation, British Medical Bulletin, 2017, 124:5–17.
2-Małgorzata Kielar , Agnieszka Gala-Bł ˛adzi ´nska ET AL .Complement Components in
the Diagnosis and Treatment after Kidney Transplantation—Is There a Missing Link?
The role of complement in graft damage: Ischemia reperfusion injury:
the 3 complement pathway activation during cold ischemia affect the function of graft after transplantation (1) Humoral rejection of kidney allograft:
Immune complex binds C1q complement activating the classical complement pathway (2) which lead to inflammation and thrombosis of graft microcirculation causing ischemia, apoptosis, necrosis and graft failure. (3) T cell mediated rejection:
Recent studies showed that local production of C3a & C5a by APCs is necessary for T cell costimulation, (4) Post transplant TMA:
It may be due to complement activation by concomitant autoimmune disease, malignant HTN, infection & AMR (5)
Therapeutics targeting complement: Human intravenous immunoglobulins (IV IG)
used in treatment of AMR, they inhibit complement activation by blocking the receptor for the Fc fragment of IG or the C1q component. (6) Eculizumab:
Inhibitor of C5 complement component, monoclonal antibody,
studies showed the efficacy of eculizumab to decrease incidence of early AMR in sensitized recipients (7)
An extended follow up study showed that eculizumab treatment has no effect on chronic AMR in recipients with significant amount of DSA (8) C1 esterase inhibitor
inhibits the activation of both the classical and lectin complement pathways
used in treatment of recurrent TMA post transplant (9)
A study showed that C1 esterase inhibitor resulted in reduction of C1q antibodies and reduced occurrence of humoral rejection. (10)
The results were encouraging with no serious toxicity
(1) Damman, J.; Nijboer, W.N.; Schuurs, T.A.; Leuvenink, H.G.; Morariu, A.M.; Tullius, S.G.; Van Goor, H.; Ploeg, R.J.; Seelen, M.A. Local renal complement C3 induction by donor brain death is associated with reduced renal allograft function after transplantation. Nephrol. Dial. Transplant. 2011, 26, 2345–2354
(2) Haas, M.; Loupy, A.; Lefaucheur, C.; Roufosse, C.; Glotz, D.; Seron, D.; Nankivell, B.J.; Halloran, P.F.; Colvin, R.B.; Akalin, E.; et al. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell–mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am. J. Transplant. 2018, 18, 293–307
(3) Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
(4) Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naive CD4+ T Cells. Immunity 2008, 28, 425–435.
(5) Benz, K.; Amann, K. Thrombotic microangiopathy: New insights. Curr. Opin. Nephrol. Hypertens. 2010, 19, 242–247.
(6) Schinstock, C.A.; Mannon, R.B.; Budde, K.; Chong, A.S.; Haas, M.; Knechtle, S.; Lefaucheur, C.; Montgomery, R.A.; Nickerson, P.; Tullius, S.G.; et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation. Transplantation 2020, 104, 911–922.
(7) Stegall, M.D.; Diwan, T.; Raghavaiah, S.; Cornell, L.D.; Burns, J.; Dean, P.G.; Cosio, F.G.; Gandhi, M.J.; Kremers, W.; Gloor, J.M. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am. J. Transplant. 2011, 11, 2405–2413.
(8) Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant (2015) 15(5):1293–302.
(9) Gonzalez Suarez, M.L.; Thongprayoon, C.; Mao, M.A.; Leeaphorn, N.; Bathini, T.; Cheungpasitporn, W. Outcomes of Kidney Transplant Patients with Atypical Hemolytic Uremic Syndrome Treated with Eculizumab: A Systematic Review and MetaAnalysis. J. Clin. Med. 2019, 8, 919.
(10) Vo, A.A.; Zeevi, A.; Choi, J.; Cisneros, K.; Toyoda, M.; Kahwaji, J.; Peng, A.; Villicana, R.; Puliyanda, D.; Reinsmoen, N.; et al. A Phase I/II Placebo-Controlled Trial of C1-Inhibitor for Prevention of Antibody-Mediated Rejection in HLA Sensitized Patients. Transplantation 2015, 99, 299–308.
Ban Mezher
3 years ago
Complement system is an important part in immune system integrity. It activated through 3 pathways, each pathway need different stimuli to be activated, but the product of C5 cleavage &formation of MAC are found in all 3 pathways. C1q bind to DSA associated with significant poor graft survival when compared to non-complement fixing DSA.
The complement had different roles in renal transplantation including ischemic-perfusion injury, AMR, Recurrence of original renal disease ( aHUS, C3 glomerulopathy, anti phpspholipid syndrome) & ABO incompatible transplant.
Complement inhibition cab be done by :
blocking of activation pathway.
blocking amplification loop.
blocking anaphylatoxin .
blocking terminal pathway.
C5 complement inhibitor( anti C5 monoclonal Ab) may decrease the frequency of AMR in highly sensitized patients. Eculizumab originally licensed for treatment of PNH & aHUS. Stegall et al & Marks et al studies show a benefit of eculizumab in prevention of early AMR among sensitized recipients.
C1 estrase inhibitor (C1-INH) block classical pathway of complement activation by C1s &C1r inhibition. It licensed for treatment of angioedema type I & type II. C1-INH safety studied in highly sensitized renal recipient which show that there is decrease in C1q+ HLA Ab & decrease in occurance of AMR.
CR1; CD35 is a new inhibitor show promising therapeutic effects. Recompenent soluble CR1 ( APT070, mirocept) has been shown ti decrease ischemic-perfusion injury & improve graft survival.
some DSA can cause graft loss without activation of complement & this damage occurs as a result to antibody dependent cellular cytotoxicity.
References:
Legendre C., Sberro-Soussan R., Zuber J. et al. The Role of Complement Inhibition in Kidney Transplantation. British Medical Bulletin,2017; 124: 5-17.
Malgorzata K., Gala-Bladzinska A., Dumnicka P., et al. Complement Components in the Diagnosis and Treatment after Kidney Transplantation – Is There a Missing Link?. Biomolecules. 2021, 11:773.
Zhang R. Donor-Specific Antibodies in Kidney Transplant Recipients. chase. 2018, 12:182-192.
Tahani Hadi
3 years ago
Complement activation is one of the important part of innate immune system and it’s play an important role in inflammatory processe it’s activation occurs by 3 pathways classical,alternate or lectin all lead to anaphylatoxin c3a and c5a formation leading to direct cell lysis.
Complement cascad activation plays a role in renal transplantation also during ischemia / perfusion process that affect on early graft function.
It has major role in both acute cellular and antibody mediated rejection also in case of thrombotic microangiopathy ,also it has role in many kidney diseases like SLE ,a typical HUS ,IgA nephropathy and even in renal complications related to DM .
Eculizumab is a humanized monoclonal antibody cause blocking of c5 cleavage thus preventing the release of anaphylatoxin which participate in all 3 major pathways although alot of follow up studies showed that it’s usage is ineffective in chronic antibody mediated rejection in case of high DSA in recipient also in c4d negative ABMR.
Amer Hussein
3 years ago
The complement system is considered to be an important part of innate immune system with a significant role in inflammation processes. The activation can occur through classical, alternative, or lectin pathway, resulting in the creation of anaphylatoxins C3a and C5a, possessing a vast spectrum of immune functions, and the assembly of terminal complement cascade, capable of direct cell lysis. The activation processes are tightly regulated; inappropriate activation of the complement cascade plays a significant role in many renal diseases including organ transplantation. Moreover, complement cascade is activated during ischemia/reperfusion injury processes and influences delayed graft function of kidney allografts. Interestingly, complement system has been found to play a role in both acute cellular and antibody-mediated rejections and thrombotic microangiopathy. Therefore, complement system may represent an interesting therapeutical target in kidney transplant pathologies
Wessam Moustafa
3 years ago
Immune system consists of 2 integrated arms
The adaptive and the innate immune systems
Adaptive with consist mainly of B and T cell responses, which contains specific receptors , capable of clonal expansion , able to form memory cells and provide long term immunity
Innate immune system comprised of dendrites ,monocytes, macrophages
They have no specific receptors , can’t clonally expand , and don’t form memory cells ,
Their main function is antigen recognition and presentation ( through APCs ) to lymphoid organs to initiate adaptive immune response
Beside cellular component of innate system, there is non cellular components , which are the complement system , which plays a role in antigens recognition .
Studies have shown that complement activation contributes to allograft injury in several clinical settings, including ischemia/reperfusion injury and antibody mediated rejection.
Besides , the complement system plays critical roles in modulating the responses of T cells and B cells to antigens.
Once complement is activated , various active complement fragments are generated which can directly and indirectly affect the graft
Directly , as these active complement fragments act as chemotaxins
Indirectly as they provide an important signal for B and T cell stimulation
Site of complement activation differs according to the situation, in case of renal ischaemia, activation affect tubulointerstitium ,
In ABMR , activation occur in peritubular capillaries
**In ischemia reperfusion injury
Complement activation occur through alternative pathway .
Complement inhibitory drugs have proven effective in several pre-clinical models of I/R injury.
In spite of promising pre-clinical data, a atudy that included 27 kidney transplant patients at high risk of DGF ,were treated with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benifical effect 2 .
Another clinical trial is ongoing, however, in which kidneys treated with an agent that attaches a complement regulator to cell membranes 3 .
This approach was previously shown to be beneficial in a rat kidney transplant model 4.
**ABMR :
In cases of acute and chronic ABMR , DSAs bind to to Donor HLA which is expressed on surface of endothelial cells activating the complement system .
This activation causes damage to the capillaries
Diagnosis of ABMR depend on detection of circulating DdSAs , micro vascular inflammation in the biopsy , and detection of complement fragments C4d in PTCs
However , cases of C4d negative ABMR have been reported , and this may reflect either non complement mediated DSA effect , or inability to detect C4d
Because complement activation is an important step in ABMR , assays have been developed to detect ability of DSAs to activate the complement .
Complement inhibitory drugs have shown benifical effects in animal models , they induce a state of accommodation which occurs due to upregulation of regulatory complement proteins 5
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment with Positive results have been reported in lung and kidney transplant recipients 6
Larger series in transplant patients have not shown a consistent benefit.
Concluding that the role of eculizumab for preventing or treatment AMR is not yet clear
**References:
1. Janeway CA Jr. How the immune system works to protect the host from infection: a personal view. Proc Natl Acad Sci USA. (2001) 98:7461–8. doi: 10.1073/pnas.131202998
2) Heeger P, Akalin E, Baweja M, Bloom R, Florman S, Haydel B, et al. Lack of efficacy of eculizumab for prevention of delayed graft function (DGF) in deceased donor kidney transplant recipients. Am J Transplant. (2017) 17(Suppl 3):B131.
3). Kassimatis T, Qasem A, Douiri A, Ryan EG, Rebollo-Mesa I, Nichols LL, et al. A double-blind randomised controlled investigation into the efficacy of Mirococept (APT070) for preventing ischaemia reperfusion injury in the kidney allograft (EMPIRIKAL): study protocol for a randomised controlled trial. Trials. (2017) 18:255. doi: 10.1186/s13063-017-1972-x
4). Pratt JR, Jones ME, Dong J, Zhou W, Chowdhury P, Smith RA, et al. Nontransgenic hyperexpression of a complement regulator in donor kidney modulates transplant
5) Dorling A. Transplant accommodation–are the lessons learned from xenotransplantation pertinent for clinical allotransplantation? Am J Transplant. (2012) 12:545–53. doi: 10.1111/j.1600-6143.2011.03821.x
6. Locke JE, Magro CM, Singer AL, Segev DL, Haas M, Hillel AT, et al. The use of antibody to complement protein C5 for salvage treatment of severe antibody-mediated rejection. Am J Transplant. (2009) 9:231–5. doi: 10.1111/j.1600-6143.2008.02451.x
7. Dawson KL, Parulekar A, Seethamraju H. Treatment of hyperacute antibody-mediated lung allograft rejection with eculizumab. J Heart Lung Transplant. (2012) 31:1325–6. doi: 10.1016/j.healun.2012.09.016
MICHAEL Farag
3 years ago
During complement activation, complement protein fragments are released into the plasma, and C3 and C4 fragments are covalently fixed to target tissues. Native kidney biopsies are routinely stained for C3 deposits, and in some centers they are also stained for C4 fragments. Because C4 is covalently attached to target tissues, C4 deposits provide a durable marker of CP activation. Allograft biopsies are now routinely stained for C4d, and detection of C4d in the peritubular capillaries is interpreted as a marker of classical pathway activation in patients with AMR. CP activation on the capillary would also be expected to result in C3 fragment deposition, although C3d deposition seems to be a less sensitive indicator of AMR. It is possible that C3d deposition signifies more complete activation of the complement cascade, and one study found that deposition of C3d on the peritubular capillaries was associated with a worse prognosis Soluble complement fragments can be measured in body fluids by enzyme linked immunosorbent assays (ELISAs). The half-life of these fragments is short, so elevated levels of complement fragments indicates that there is ongoing activation. There are assays that can measure many different complement fragments, including C4a, C3a, Ba, Bb, C5a, and soluble sC5b-9. Other than staining allograft biopsies for C4d, complement biomarkers are not routinely analyzed in transplant recipients. As the use of complement inhibitory drugs expands, however, there will be an increasing need to develop accurate biomarkers. Although eculizumab and C1-INH have shown promise in case reports and small series, their role in transplant medicine requires further study. Many additional anti-complement therapeutics are in clinical development, and some of these new drugs block individual activation pathways or specific components of the complement cascade.
Professor Ahmed Halawa
Admin
3 years ago
Dear All Please answer the question below. We need to know that complement activation is an important part of tissue injury, but graft damage can happen without complement activation. Please give examples!!
One example of complement independent Antibody medicated allograft dysfunction is the C4d-negative ABMR. It was noted that some patients will have histologic evidence of ABMR (microcirculatory inflammation) and positive donor-specific antibodies (DSA) but with little or no C4d staining in the PTCs, a situation that was named as C4d-negative ABMR (1).
The clinical implication of this subtype of ABMR is that the early initiation of treatment for ABMR even in absence of C4d staining can result in a better long term graft outcome (2). if untreated, C4d-negative ABMR may lead to the development of scarring within the graft, transplant glomerulopathy and even graft loss (1).
One study involved 98 renal allograft biopsies performed for clinical indications within the first three months post-transplant, 17 showed C4d-positive ABMR and 16 had histologic changes of ABMR and DSA, but no C4d could be detected in the PTCs (2). Patients with C4d-negative ABMR who received treatment for ABMR had a lower risk for progression to transplant glomerulopathy compared to those who were left untreated (2).
References:
1) Haas M. C4d-negative antibody-mediated rejection in renal allografts: evidence for its existence and effect on graft survival. Clin Nephrol. 2011;75(4):271.
2) Haas M, Mirocha J. Early ultrastructural changes in renal allografts: correlation with antibody-mediated rejection and transplant glomerulopathy. Am J Transplant. 2011; 11(10): 2123.
After alloantigen recognized by naive t cell and its transformation it effector tcell which lead to b cell activation and antibodies formation against donor HLA phenotyping
They cause allograft damage in two principal ways: they activate the classical pathway of the complement system and stimulate macrophages and other immune cells by binding to Fc receptors
(FcR) that recognize the constant regions of specific antibody classes
Reference
Handbook of kidney tranplantation by G.danovitch
DSA may also cause injury through complement-independent mechanisms, such as signaling through FcR receptors on natural killer (NK) cells or other cell types. C4d-negative AMR was included in the most recent revision of the Banff criteria for the diagnosis of AMR .(Erik Stites.The Complement System and Antibody Mediated Transplant Rejection.)
C4d deposition in ABO-incompatible allografts can actually be associated with accommodation and improved outcomes. Thus, detection of C4d without histologic or functional evidence of allograft injury is not sufficient to predict AMR or to guide treatment. On the other side some clinically evident ABMR were found to be without C4d deposition(C4d negative ABMR) …. the question then will be is there any failure to detect C4d or it means inflammation without complement involvement .
Consistently, for several years, the C4d deposition has been considered the gold standard for ABMR diagnosis; however, today, it is recognized that up to 55% of patients can develop ABMR without detectable capillary C4d deposits. Indeed, a C4d-negative ABMR phenotype has been included in Banff 2013 classification (1).
C4d-negative ABMR was associated with glomerulopathy and graft loss.
This can occure directly by binding of Ab to specific FcR receptors on natural killer and macrophages or other cell types.
Biglarnia AR, Huber-Lang M, Mohlin C, Ekdahl KN, Nilsson B. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol. (2018) 14:767–81.
ABMR without C4d deposition can occur in many cases and Banff classification criteria included a category of C4d-negative ABMR. Several mechanisms have been proposed for complement-independent tissue injury in the presence of DSA. It is well known that activated monocytes and macrophages infiltrate into graft tissues. The inflammatory environment triggered by the binding of DSA to endothelial cells alone can induce an allo-reaction of CD4 T-cells via graft endothelial cell HLA-class II.
Kenta I, Takaaki K. Molecular Mechanisms of Antibody-Mediated Rejection and Accommodation in Organ Transplantation. Nephron. 2020;144(1):2-6.
Complement activation is important but antigens recognized by non-HLA antibodies have been reported. Examples such as endothelial cell, angiotensin II type 1 receptor antibodies (AT1R-Ab), endothelin-1 type A receptor, collagen type IV, fibronectin, perlecan, vimentin, agrin, tubulin, laminin, myosin and major histocompatibility complex class I chain-related gene A and B has been reported but AT1R-Ab has gain popularity among those non HLA antibodies.
AT1R-Ab recognised to have mediate graft injury in a complement independent and antibody dependent cell mediated manner. Pre- and post-KT AT1R-Ab were associated with AMR development. studies have shown that pre and post KT AT1R-Ab level more than > 17 U/mL were a risk factor for AMR development.
References
Sorohan, B., Ismail, G., Leca, N., Tacu, D., Obrișcă, B., Constantinescu, I., Baston, C. and Sinescu, I., 2020. Angiotensin II type 1 receptor antibodies in kidney transplantation: An evidence-based comprehensive review. Transplantation Reviews, 34(4), p.100573.
Last edited 3 years ago by Theepa Mariamutu
Mahmud Islam
3 years ago
complement as part of the innate immune system plays role in each stage causing damage to the transplanted kidney. apart from details of this fantastic and complex system, we see damage differently in each stage. for example at early stages and in case of reperfusion ischemia damage is mainly in tubulointerstitial space while in AMR (antibody-mediated rejection) we see capillaritis (peritubular capillaries). https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6788431/
Both c3 and c4 play a role but c4 is stuck to tissue and it became a routine stain in many centers to show AMR
data about eculizumab are conflicting some were compared to historical transplants.
regarding c1 esterase inhibitor some promising results were found (Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney Transplantation: Results of a Randomized Double-Blind Placebo-Controlled Pilot Study. (https://pubmed.ncbi.nlm.nih.gov/27184779/)
Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement? Give examples
Yes.
Anti-HLA-class I antibodies can promote focal adhesion kinase, matrix metalloproteinase-2, and neutral sphingomyelinase-2-dependent proliferation and migration of smooth muscle cells producing changes in the intima leading to chronic rejection. (1)
Reference:
1) Valenzuela NM, McNamara JT, Reed EF. Antibody-mediated graft injury: complement-dependent and complement-independent mechanisms. Curr Opin Organ Transplant 2014;19:33-40.
Ahmed Fouad Omar
3 years ago
Can graft injury happen without activation of complement? Give examples
Yes rejection can occur in absence of complement activation as in C4d-negative antibody-mediated rejection with high Anti-angiotensin II type 1-receptor-activating antibodies (anti-AT1R) have been mentioned to stimulate a severe vascular rejection described as IIB or AMR in Banff classification.
American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions,
Haas.M et al, 28 January 2014
Following transplantation, complement has a role in innate immunological and inflammatory processes that further damage the graft and result in a gradual decrease in its functional mass. Additionally, it modulates the adaptive immune response, mediates many of the downstream effects of B and T cell immunity, and can function independently of the adaptive immune response
Activation of the complement system in the donor, the graft and the recipient before, during and after transplantation is a major cause of damage to the kidney transplant through different ways like antibody-mediated rejection, recurrence of native kidney disease such as atypical haemolytic uraemic syndrome, C3 glomerulopathy, anti-phospholipid syndrome and the accommodation phenomenon in ABO-incompatible transplantation.
The complement system is activated through three distinct pathways: classical pathway Lectin pathway, Alternative pathway
Ischaemia and subsequent reperfusion of the graft is the most important mechanism that triggers complement activation
Complement-targeted strategies might have a role in optimizing graft quality as well as in the treatment of antibody-mediated rejection, induction of accommodation and modulation of the adaptive immune response.
Promising data from preclinical and clinical studies suggest that complement targeted therapies could potentially become part of the standard of care for kidney transplantation.
Complement-targeted agents for the prevention of rejection in transplantation include:
A) Standard of care therapies:
Intravenous immunoglobulin :Inhibits activation of complement, blocks FcR and C1q 2
Rituximab Humanized anti-CD20 expressed on B cells. It Blocks complement-induced enhancement of antibody generation
B) complementary therapies: initial results from early clinical trials are promising but there is still inconsistencies on the effect of complement inhibitors on delayed graft function and ABMR which clearly reflect the complexity of allograft injury and the involvement of complement-independent mechanisms
A number of therapies are being developed but only two categories of complement-inhibiting drugs are in clinical use currently; the humanized anti-C5 mAb eculizumab and preparations of C1INH
Eculizumab: Humanized mAb C5a Inhibits C5 cleavage to form C5a and C5b; blocks the terminal pathway
Benefits: Terminal Complement blockage at the level of C5 preserves the early components of complement which are essential for opsonization of microorganisms and clearance of immune complexes
Risks: An acquired functional C5 deficiency, and in patients with genetically determined C5 deficiency, increase the risk of Neisseria meningitis so it is mandatory to give prophylactic vaccination and antibiotics
Trials and outcome:
In delayed graft function: a randomized, parallel-group, double-blind, placebo-controlled, multicentre study was performed with two doses of Eculizumab administered in an acute setting for the prevention of DGF in adult recipients of deceased-donor kidney transplants who were at increased risk of DGF. 288 pts recruited in North America, South America, Europe and Australia. Unfortunately, DGF rate was not significantly different when comparing Eculizumab to placebo
In preventing antibody-mediated rejection: a single-centre open-label study by Stegall et al. included 26 sensitized patients with preformed HLA-DSAs undergoing living donor renal transplantation showed a reduction in ABMR at 3 months in comparison to control group who received post-transplantation plasmapheresis only (7.7% versus 41.2%),but a 2 years follow-up showed no difference in the histopathological occurrence of ABMR and graft survival between both groups.
On The other hand, A prospective multi-centre study compared 9 weeks of prophylactic eculizumab treatment to plasmapheresis and IVIG .Eculizumab showed a benefit to lower ABMR incidence early and after 3 months only in the patients with C1q-binding HLA-DSAs.
Thus it appears that only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti-complement treatment or that Complement inhibition at C5 did not prevent upstream complement activity due to the generation of immune modulators such as C4a or C3a.
In kidney transplantation to treat or prevent recurrences of atypical HUS: The efficacy of Eculizumab efficacy was found in 4 prospective, multi-centre, single-arm, non-randomized studies involving 100 patients followed at least 2 years. Additionally Zuber et al gathered 21 patients from both an extensive literature search and a French national survey and showed that Eculizumab was efficient for treating and preventing aHUS recurrence after kidney transplantation
Serine protease inhibitors:Purified or recombinant proteins that inactivates complement serine proteases, blocks the classical and lectin pathways
Benefits:
Theoretically elicit a more potent effect than Eculizumab on both T-cell and B-cell components of the allo-immune response. Blocking the generation of complement activation products of C3 that contribute to neutrophil and monocyte recruitment and tissue injury in AMR.
Risks:
The lack of specificity of serine protease inhibitors implies a broad inhibitory effect on both the classical and lectin pathways that may dramatically reduce C3-dependent opsonization of pathogens and increase susceptibility to infections.
Trials and outcome:
In a pilot trial, C1INH was given as a supplement to high-dose IVIg in 6 patients with nonresponsive ABMR. At 6 months, these patients showed improvements in eGFR compared with levels at enrolment and had less C1q-binding DSAs than controls.
In a randomized, placebo-controlled trial that compared add-on treatment with C1INH to placebo in 18 patients with ABMR who were receiving plasmapheresis, IVIg and rituximab, There was no difference in ABMR histopathology or kidney function in both groups at at 20 days However, a subgroup analysis of 14 patients with 6-month protocol biopsy samples identified no transplant glomerulopathy in the C1INH group, whereas transplant glomerulopathy was present in 43% of patients in the placebo group.
Accordingly, Future well designed clinical trials with long surveillance are warranted to further evaluate complement interventions in transplantation
The multifaceted role of complement in kidney transplantation.
Ali-Reza Biglarnia, Markus Huber-Lang, Camilla Mohlin, Kristina N. Ekdahl,and Bo Nilsson
Nature Reviews | Nephrology ,volume 14 .DECEMBER 2018 The role of complement inhibition in kidney transplantation C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17
Amit Sharma
3 years ago
Complement system activation has an important role in renal graft injury, especially in antibody mediated rejection and ischemia/reperfusion injury. Complement activation through classical pathway, lectin pathway and alternative pathway leads to formation of C3b and C5b which causes damage/ lysis to the target cell. (1)
Hence complement inhibitors can be utilized to prevent complement mediated injury in transplant recipients. These include:
(1) C5 inhibitor Eculizumab: It binds to C5 and inhibits C5a and subsequent membrane attack complex formation
It has been used in kidney transplant patients for:
a) Prevention of delayed graft function (DGF): PROTECT study by Alexion was done at multiple centers, but 2 doses of eculizumab failed to show any significant benefit in prevention of DGF. (2)
b) Preventing AMR: Few studies demonstrated that Eculizumab use in desensitized highly sensitized patients reduced incidence of acute AMR at 1 year. (3,4). Another study did not find any difference. (5) Subclinical AMR lesions were seen in patients with persistently high DSA levels.
c) For treatment/prevention of atypical HUS recurrence: Multiple studies have shown that eculizumab causes significant improvement in graft function of patients with recurrence of atypical HUS. (6,7)
d) For prevention/ treatment of C3 glomerulopathy and antiphospholipid syndrome: Some studies have shown promising results. (8,9)
(2) C1 esterase inhibitor: Blocks classical pathway. It has been used to prevent AMR in transplant recipients. A study showed HLA sensitized patients receiving C1-inhibitor did not develop AMR. (10) Another study used C1-inhibitor in treatment of acute AMR and showed that there was no transplant glomerulopathy at 6 months post transplant (versus 42% in the placebo group). (11)
(3) Ides: A recombinant Streptococcus pyogenes- derived endopeptidase which cleaves IgG has been shown to prevent AMR in highly sensitized transplant recipients. (12)
References:
1) Grafals M, Thurman JM. The role of complement in organ transplantation. Front Immunol 2019;10:2380
2) Legendre Ch, Sberro-Soussan R, Zuber J, et al. The role of complement inhibition in kidney transplantation. British Medical Bulletin 2017;124:5-17.
3) Stegall MD, Diwan T, Raghavaiah S, et al. Terminal complement inhibition decreaases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant 2011;11:2405-2413.
4) Glotz D, Legendre C, Lefaucheur C, et al. Eculizumab in prevention of acute antibody mediated rejection in sensitized deceased donor kidney transplant recipients: 1 year outcomes. Transplant Int 2015;28:1-19.
5) Cornell LD, Schinstock CA, Gandhi MJ, et al. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant 2015;15:1293-1302.
6) Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic uremic syndrome. N Eng J Med 2013;368:2169-2181.
7) Zuber J, Le Quintrec M, Krid S, et al. Eculizumab for atypical hemolytic uremic syndrome recurrence in renal transplantation. Am J Transplant 2012;12:3337-3354.
8) Le Quintrec M, Lionet A, Kandel C, et al. Eculizumab for treatment of rapidly progressive C3 glomerulopathy. Am J kidney Dis 2015;65:484-489.
9) Canaud G, Kumar N, Anglicheay D, et al. Eculizumab improves posttransplant thrombotic microangiopathy due to antiphospholipid syndrome recurrence but fails to prevent chronic vascular changes. Am J Transplant 2013;13:2179-2185.
10) Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo controlled trial of C1-inhibitor for prevention of antibody mediated rejection in HLA sensitized patients. Transplantation 2016;99:299-308.
11) Montgomery RA, Orandi BJ, Racusen L, et al. Plasma derived C1 esterase inhibitor for acute antibody mediated rejection following kidney transplantation: results of a randomized double-blind placebo controlled pilot study. Am J Transplant 2016;16:3468-3478.
12) Jordan SC, Lorant T, Choi J. IgG endopeptidase in highly sensitized patients undergoing transplant. N Eng J Med 107;377-1693-1694.
Doaa Elwasly
3 years ago
Inflammatory effect of complemt on allograft
-C3a
C3a/C3aR signaling participates in glomerular and tubular injury , also stimulates epithelial cells to produce chemokines which causes tissue inflammation .There are not currently any specific antagonists of C3a available .(1)
–C5a
C5a has pro-inflammatory effects and is a potent myeloid cell chemoattractant.
C5aR deficiency or blockade is protective against of Ischemia/Reprfusion injury , tubulointerstitial injury, and anti-neutrophil cytoplasmic antibody (ANCA) vasculitis.
C5aR antagonists have been developed (2).
–Membrane Attack Complex
MAC formation on endothelial cells leads to NF- κb activation within the cells leading to production of IL-1α and IL-8 .
A trial that enrolled 27 kidney transplant patients at high risk of DGF treated with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benefit with treatment .Another trial ongoing in which kidneys treated with an agent which attaches a complement regulator to cell membranes ,it was previously shown to be beneficial in a rat kidney transplant model (3)
Eculizumab has been used in transplant patients at high risk of developing AMR, and patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR . Larger studies in transplant patients have not shown a long term benefit, meanwhile eculizumab rolein preventing AMR is not clear.(4)
There is pre-clinical data showing that C5aR1 blockade may be a beneficial treatment for rejection.
Complement inhibitory drugs can have a role in xenotransplanation, but they probably will need to be administered for long time due to the persistence of pathogenic natural antibodies in spite of immunosuppression.
C1-INH has been used in prevention of AMR, other complement inhibitory agents are useful in the transplant setting, as an AP inhibitor , a LP inhibitor , and C5a blockade (2).
The diagnosis of AMR is based on detecting DSA in the plasma, microvascular inflammation on a biopsy , and C4d deposition in the peritubular capillaries ,this criteria have been changed to account for C4d-negative cases . It is un known whether those negative case caused by non-complement-mediated injurious effects of the DSA, or it reflects variability in detection of C4d.(2)
1- Peng Q, Li K, Smyth LA, Xing G, Wang N, Meader L, et al. . C3a and C5a promote renal ischemia-reperfusion injury. J Am Soc Nephrol. (2012) 23:1474–85
2- Grafals M et al, The role of complement in organ transplantation. Frontiers in Immunology2019; 10: 2380.
3-Jane-Wit D, Manes TD, Yi T, Qin L, Clark P, Kirkiles-Smith NC, et al. . Alloantibody and complement promote T cell-mediated cardiac allograft vasculopathy through noncanonical nuclear factor-kappaB signaling in endothelial cells. Circulation. (2013) 128:2504–16.
3- Heeger P, Akalin E, Baweja M, Bloom R, Florman S, Haydel B, et al. Lack of efficacy of eculizumab for prevention of delayed graft function (DGF) in deceased donor kidney transplant recipients. Am J Transplant. (2017) 17(Suppl 3):B131
4-Marks WH, Mamode N, Montgomery R, Stegall MD, Ratner LE, Cornell LD, et al. . Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: a randomized trial. Am J Transplant. (2019) 19:2876–88.
Riham Marzouk
3 years ago
Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement? Give examples
antibody against G protein coupled receptor and angiotensin receptors type I and II were found in the patient with severe rejection with vascular affection without c4d deposition ( without complement fixation). Aurelie Philippe. Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation. Kidney International Reports. 2019 vol (4) issue 4, P 510-12.
Ahmed Faisal
3 years ago
Since the activation of complement in the setting of high levels of DSA plays a major role in the pathogenesis of acute AMR, it makes sense that complement blockade would be an important strategy for prevention and treatment of AMR. Agents targeting C5 and C1 esterase have been evaluated in clinical trials.
C5 inhibition [Eculizumab]
*In a single-arm phase I/II trial conducted at the Mayo Clinic that included 26 highly sensitized recipients of living-donor renal transplants (NCT006707), eculizumab in combination with plasmapheresis resulted in a statistically significant reduction in AMR in the first 3 months compared with 51 historic controls receiving plasmapheresis alone.
*In a phase II randomized, open-label, multicenter trial of eculizumab vs placebo as an add-on to SOC desensitization protocols in living kidney donor recipients (NCT01399593), the eculizumab group failed to achieve a statistically significant reduction in the primary composite endpoint (occurrence of biopsy-proven AMR, graft loss, patient death, or loss to follow-up at the end of the 9-week treatment period posttransplant) when compared with placebo
*In a randomized, open-label, multicenter phase II study in sensitized deceased donor kidney transplant recipients (NCT01567085), 1-year data show that eculizumab was effective in reducing the incidence of acute AMR.
*Eculizumab failures have been reported in cases of C4d-negative AMR.
C1 esterase inhibition
Two C1-INH products that are approved for use by the FDA in the treatment of hereditary angioedema have been evaluated in small pilot studies for AMR: Berinert® (CSL Behring, Kankakee, IL, USA) and Cinryze® (Shire ViroPharma Inc., Lexington, MA, USA)
*A recent double-blind, placebo-controlled, randomized phase I/II trial (NCT01134510) evaluated C1-INH (Berinert) in sensitized renal transplant patients for the prevention of acute AMR. Twenty percent of C1-INH patients developed AMR outside of the study period, whereas 30% of placebo patients developed AMR, 10% during the study period. Delayed graft function developed in 1 patient receiving C1-INH and 4 patients receiving placebo. C1-INH treatment also reduced C1q-positive HLA antibodies in all 5 patients with these antibodies at time of transplantation.
*In a single-arm pilot study, the investigators found that CI-INH (Berinert) in combination with high-dose IVIg improved allograft function in kidney recipients with acute AMR that was nonresponsive to conventional therapy
*A randomized, double-blind, placebo-controlled, multicenter phase IIb study (NCT01147302) evaluated the safety and effect of C1-INH (Cinryze) for the treatment of acute AMR in 18 recipients of donor-sensitized kidney transplants.
Histopathological resolution of AMR was seen in 7 out of 7 C1-INH patients analyzed and 4 out of 6 placebo patients analyzed.
Surveillance 6-month biopsies performed in 14 patients showed no transplant glomerulopathy (ptc + cg ≥ 1b) in the 7 patients in the C1-INH group, whereas 3 out of 7 patients in the placebo group showed transplant glomerulopathy.
In addition, transplant glomerulopathy tended to develop in those patients in the placebo group who had very low levels of C1-INH.
IgG-degrading enzyme of Streptococcus pyogenes (IdeS)
Phase I/II testing (NCT02790437) has begun for a new compound called IgG-degrading enzyme of Streptococcus pyogenes (IdeS). This enzyme cleaves at a very specific amino acid sequence in the hinge region of human IgG and essentially neutralizes all of the IgG in the body within 4 hours of administration.
Reference
Montgomery, RA, Loupy, A, Segev, DL. Antibody-mediated rejection: New approaches in prevention and management. Am J Transplant. 2018; 18(Suppl. 3): 3– 17. https://doi.org/10.1111/ajt.14584
The complement-targeted agents for the prevention of rejection in transplantation.
Ahmed Faisal
3 years ago
-The terminal complement inhibitor, Eculizumab, and C1 esterase inhibitor (C1-INH) have been the subjects of recent clinical trials exploring the potential of targeting the complement cascade in the prevention and treatment of AMR.
-Eculizumab (Soliris, Alexion) is a recombinant humanized anti-C5 monoclonal antibody that prevents the cleavage of C5 by C5 convertase. In a phase 3 clinical trial published in 2006, eculizumab stabilized hemoglobin levels in transfusion-dependent patients with PNH. Following this trial, the FDA approved the use of eculizumab for PNH in March 2007.
-C1-INH is a serine protease inhibitor (SPI) that removes activated C1r and C1r from C1q. Two nano-filtered C1-INH products derived from human serum, cinryze (Takeda/Shire Pharmaceuticals) and berinert (CSL Behring), were approved for the treatment and prophylaxis of attacks of Hereditary angioedema (HAE) after randomized trials demonstrated their efficacy.
-Multi-center trial that began enrollment in 2013 evaluated the role of eculizumab in the treatment of biopsy-proven AMR and acute graft dysfunction.
Patients in the standard of care (SOC) arm received three sessions of plasmapheresis with IVIg administered after each session. Those in the treatment arm received a first dose of 1,200 mg of eculizumab followed by 4 weekly doses of 900 mg and a dose of 1,200 mg at week 5. Patients received additional doses of 1,200 mg at weeks 7 and 9 if DSA levels at week 6 remained >50% of baseline. However, recruitment for this trial was terminated in 2017 due to the study drug’s failure to improve allograft function assessed by estimated glomerular filtration rate (eGFR) 3 months after transplantation [1].
-The role of C1-INH in the treatment of AMR was explored by two recent studies [2,3].
Viglietti et al. treated six patients who had AMR that was not responsive to SOC therapy with C1-INH (berinert, CSL Behring) and high-dose IVIg for a duration of 6 months [2]. At the end of the 6-month follow-up period, the authors reported significant improvement in mean eGFR. Fewer patients demonstrated C4d deposition on allograft biopsies and circulating C1q fixing DSAs. However, other histologic features, like chronic glomerulopathy, were unchanged [2].
Montgomery et al. conducted a randomized phase 2 trial to evaluate the role of a 2-week course of C1-INH (Cinryze, Shire Pharmaceuticals) as an add-on therapy to SOC plasmapheresis and low-dose IVIg, compared to SOC alone. Nine patients were randomized to each arm.While there was no significant difference in graft survival and histologic findings of AMR after 20 days, 6-month allograft biopsies showed an absence of transplant glomerulopathy in the C1-INH treated patients, whereas three out of seven patients in the placebo arm were found to have this histologic feature that is associated with chronic antibody mediated injury and premature allograft loss
-Reference
Tatapudi, Vasishta S, and Robert A Montgomery. “Therapeutic Modulation of the Complement System in Kidney Transplantation: Clinical Indications and Emerging Drug Leads.” Frontiers in immunology vol. 10 2306. 1 Oct. 2019, doi:10.3389/fimmu.2019.02306
[1] Randomized-Controlled Trial Examining the Safety and Efficacy of Eculizumab in the Treatment of AMR NCT01895127. Available online at: https://clinicaltrials.gov/ct2/show/record/NCT01895127?term=eculizumab+antibody+mediated+rejection&rank=3&view=record (accessed September 19, 2019)
[2] Viglietti D, Gosset C, Loupy A, Deville L, Verine J, Zeevi A, et al. . C1 Inhibitor in acute antibody-mediated rejection nonresponsive to conventional therapy in kidney transplant recipients: a pilot study. Am J Transpl. (2016) 16:1596–603. 10.1111/ajt.13663.
[3] Montgomery RA, Orandi BJ, Racusen L, Jackson AM, Garonzik-Wang JM, Shah T, et al. . Plasma-derived C1 esterase inhibitor for acute antibody-mediated rejection following kidney transplantation: results of a randomized double-blind placebo-controlled pilot study. Am J Transpl. (2016) 16:3468–78.
Last edited 3 years ago by Ahmed Faisal
Mohamed Essmat
3 years ago
· complement system is a vital column of the innate immunity. It may lead to graft injury and affection by many ways including AMR , cellular rejection , and TMA. · As soon as the graft is exposed to the recipient’s blood , the complement system may be activated and lead to active events. Once the complement cascade is activated it can lead to direct , through cell lysis by the MAC system , and indirect effects. The activation can occur through classical, alternative, or lectin pathway, resulting eventually in direct cell lysis. · C3a and C5a provide important signals for T and B cell activation in the tubule-interstitium , whereas in Antibodies activation occurs in the peritubular capillaries. The inhibition of some complement components has shown results in preventing AMR · C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking C1 complex components. Still udner trials . · Eculizumab is MAB capable of blocking the cleavage of C5 complement component, thus preventing the release of and the assembly of MAC. Eculizumab is an effective treatment for paroxysmal nocturnal hemoglobinuria and as well as recent uses in TTP . · In one study reported , they study the use of eculizumab in highly sensitized patients with live donor transplantation . They found that the eculizumab treated patients had lower incidence of AMR at one year, but with high rates of subclinical rejection. · Critics regarding eculizumab from effective to ineffect in patients with ABMR. Biglarnia A.R. Huber-Lang M., Mohlin C., Ekdahl K.N., et al. The multifaceted role of complement in kidney transplantation. Nature Reviews Nephrology. 2018, Dec., Vol.14. 767-781. Long-term outcomes of eculizumab-treated positive crossmatch recipients: Allograft survival, histologic findings, and natural history of the donor-specific antibodies Carrie A. Schinstock et al Positive Crossmatch Kidney Transplant Recipien Treated With Eculizumab: Outcomes Beyond 1 Year L. D. Cornell et al
Ahmed Faisal
3 years ago
Can graft injury happen without activation of complement?
YES
-Even in absence of complement activation, some DSAs can cause graft damage through antibody-dependent cellular cytotoxicity.
The innate immune cells, including neutrophils, macrophages, and natural killer cells, can bind to Fc fragments of DSAs, trigger degranulation, and release lytic enzymes, which cause tissue injury and cell death.
This process can mediate smoldering damages to the endothelial cells and is proposed as an important pathogenesis in subclinical and chronic antibody-mediated rejection phenotypes.
-Furthermore, DSAs can cause graft injury by direct activation of endothelial proliferation through increasing vascular endothelial growth factor production, upregulating fibroblast growth factor receptor, and increasing its ligand binding as well as other signaling
pathways for cellular recruitment.
This pathogenesis may contribute to transplant glomerulopathy and vasculopathy that feature vascular intima thickness with smooth muscle cell invasion.
-The latter two complement-independent mechanisms can explain the clinical phenotypes of antibody mediated rejection with negative C4d staining in peritubular capillaries.
-IgG4iDSA–associated subacute and chronic phenotypes of antibody-mediated rejection are consistent with complement independent pathogeneses.
-Reference
Zhang R. Donor-Specific Antibodies in Kidney Transplant Recipients. Clin J Am Soc Nephrol. 2018 Jan 6;13(1):182-192. doi: 10.2215/CJN.00700117. Epub 2017 Apr 26. PMID: 28446536; PMCID: PMC5753302.
The complement system is a part of innate immunity and has an important role in kidney transplant rejection. Although adaptive immunity is highly effective in long term protection against foreign antigens, it is relatively sluggish and, an innate immunity that comprises inflammatory cells and soluble mediators (such as complement) response instantly to foreign instruction.
A growing body of evidence suggested that innate immunity has a principal role in priming of allogenic-specific adaptive immunity and rejection. In addition, the communication between innate and adaptive immune response in kidney transplant rejection is bidirectional.
Also, there is a cross-talk between the complement system and coagulation cascade in kidney transplantation.
DBD can induce activation of complement cascade. In addition, complements have an important role in ischemia reperfusion injury that is a principal cause of DGF. The inflammatory response after cold and warm ischemia time is characterized by graft cell injury and death, activation of the complement system, and filtration of graft parenchyma with monocytes and neutrophils. Ongoing studies assess the role of inhibition of complement activity in reducing ischemia-reperfusion injury.
Preclinical animal studies have shown that treatment of DBD donors with complement inhibitors is associated with improved graft function after transplantation.
Only two categories of complement- inhibiting drugs are in the clinic: thehumanized anti- C5 monoclonal Ab eculizumab (Soliris,Alexion) and preparations of C1INH (for example, Berinert (CSL Behring), Cinryze).
Eculizumab, a humanized monoclonal antibody which inhibits the activity of C5 convertase, thus blocking C5a and C5b formation and, as a result, inhibits the formation of membrane attack complex and was used in transplant recipients to prevent complement mediated microvascular damage that is associated with ongoing antibody mediated rejection. In progress studies have been established for evaluation of its usefulness in antibody mediated rejection, desensitization protocols, and prevention of ischemia reperfusion injuries.
A variety of reports show a range of responses to eculizumab from very effective to no effect in patients with ABMR. Few studies demonstrate that eculizumab is not useful in patients with non- C1q binding HLA- DSAs, only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti- complement treatment. On the other hand, complement inhibition at the level of C5 does not prevent upstream complement activity, more over residual terminal pathway activity via the alternative pathway can occur despite eculizumab treatment.
Based on these observations, the concept of upstream complement inhibition is gaining increasing interest. C1INH, either in its recombinant form or as an enriched preparation from human plasma, has been successfully used to prevent allogeneic and xenogeneic humoral immune responses in preclinical models. Finally, a clinically applicable C3 inhibitor, the latest generation of compstatin analogues (Cp40), has been evaluated in clinical trial.
It would help if you were up to a point in your answers. Don’t distract your reader with an unstructured writing style. Give an example and a reference for blocking a complement component that is directly related to the coagulation cascade.
Ali- Reza Biglarnia1, Markus Huber- Lang2, Camilla Mohlin3, Kristina N. Ekdahl3,4 and Bo Nilsson4, The multifaceted role of complement in kidney transplantation, nature reviews, nephrology, vol 14, December 2018.
Paola Pontrelli 1, Giuseppe Grandaliano and Cees Van Kooten, editorial, kidney transplantation and innate immunity. Frontiers in immunology, vol 11, octobor 2020
antibody against G protein coupled receptor and angiotensin receptors type I and II were found in the patient with severe rejection with vascular affection without c4d deposition ( without complement fixation).
Aurelie Philippe. Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation. Kidney International Reports. 2019 vol (4) issue 4, P 510-12.
Reem Younis
3 years ago
The complement system is part of the innate immune system which enhances the ability of antibodies and phagocytic cells to clear microbes and damaged cells, promote inflammation, and attack pathogen.It consists of a small number of proteins synthesized by the liver circulate in the blood as inactive precursors when activated generate bioactive components including C3b, C3a, C5a, C5b-9 with different proinflammatory, chemoattractant, and cell-damaging functions.
It activates through 3 pathways:
1. classical pathway which triggered by recognition of subclasses of surface-bound IgG and IgM antibodies by C1q
2. The lectin pathway which triggered by recognition of bacterial surface sugar by mannose-binding lectin(MBL).
3. Alternative pathway which activates by spontaneous hydrolysis ofC3.
These 3 pathways lead to the formation of C3 convertases which lead to the generation of cell-killing membrane attack complex (MAC).
Late –onest antibody-mediated rejection(AMR) is the leading cause of allograft loss and poor long-term graft survival. In AMR donor-specific antibodies (DSAs) bind to mismatch HLA molecules and can activate the classical complement pathway leading to allograft vascular injury.
Eculizumab,anti-C5 antibody, C1 esterase inhibitor have been the subjects of recent clinical trials in the prevention and treatment of AMR.
Eculizumab is direct against C5, it is used for the prevention of AMR and delayed graft function but according to the study no difference between eculizumab and placebo in the prevention of delayed graft function. While studies comparing eculizumab and plasma exchanges with plasma exchanges only in preventing AMR showed that graft and patient survival were excellent in both groups and incidence of acute AMR was 7.7% in the eculizumab and plasma exchange group compared to 41.2% in the plasma exchanges group.1
Studies of C1 –inhibitor in preventing AMR showed a reduced incidence of AMR in the C1 inhibitor group compared with the control group.2
References
1.Stegall MD, Diwan T, Raghavaiah S, et al. . Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant 2011;11:2405
2.Vo AA, Zeevi A, Choi J, et al. . A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation 2016;99:299–308.
Mohamad Habli
3 years ago
Answer to Professor’s Halawa question: Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement? Give examples Graft injury could be immune response dependent or independent . For the immune-mediated graft injury, immune response could be complement dependent or independent. Several mechanisms have been proposed for complement-independent tissue damage in the settings of positive DSAs. It is known that activated monocytes and macrophages infiltrate into graft tissues.
Immune- dependent complement independent graft damaged could be explained by the binding of DSA to endothelial cells which alone, can induce an alloreaction of CD4 T-cells via graft endothelial cell HLA-class II.
Recent evidences indicated histological findings of ABMR without C4d deposition in many cases which indicated complement independent mechanisms of injury, at which recent Banff classification criteria included a category of C4d-negative ABMR.
References:
Nephron 2020;144(suppl 1):2–6, DOI: 10.1159/000510747
Haas M. The significance of C4d staining with minimal histologic abnormalities. Curr Opin Organ Transplant. 2010;15(1):21–7
The complement system is divided into 3 major pathways the classical, lectin, and alternative pathways, the end result of these pathways is cleavage of C3 into C3a and C3b, C3b activate cleavage of C5 to C5 a, and C5b.
1- C5b initiate formation of membrane attack complex (MAC) with subsequent lysis of target cell
2- C3b is deposited on target cells (opsonization) then phagocytosis by cells that have complement receptor
3- C3 a, C5a are potent anaphylatoxin that initiate inflammation, vasodilatation
⦁ In the classical pathway, the initiating step is formation of Ag-Ab complexes.(1)
⦁ In the lectin pathway the initiating step is binding of lectins to sugar on the surface of microorganism. (2)
⦁ In alternative pathway (AP) no antigen exposure and no antibody needed for activation of this system, it acts independently (3)
Complement system has important rule in graft dysfunction which is very evident in the following two situations :
1- Antibody mediatied rejection
ABMR is the most common cause of graft failure
Active ABMR occurs due to binding of circulating antibodies to donor alloantigens located on graft endothelium (HLA class I, HLA class II, ABO – antigens or non HLA antigens on endothelial cells)
Ag-Ab complexes activate classical complement pathway with final production of c5b, c3b, c3a, c5a leading to inflammation, cell damage and graft dysfunction
One of degradation products of classical pathway is C4b which is converted to C4d, the later binds covalently to endothelium of peritubular capillaries (PTCS) and acts as a foot print for complement activation and ABMR.
C4d normally can be detected in mesangium, but deposition in PTCS occurs only in ABMR, deposition of C4d may be defuse if > 50% of PTCS are involved or may be focal if < 50 % of PTCS are involved (4)
Diagnosis of ABMR:
⦁ Histologic evidence of acute tissue injury (capillaritis and/or glomerulitis)
⦁ Evidence of antibody interaction with vascular endothelium (C4d staining in peritubular capillaries [PTCs])
⦁ Serologic evidence of circulating DSAs
If the patient has first criteria and only one of the other 2 criteria, the patient is considered to have ABMR, this means C4d staining can replace DSA, and C4d negative ABMR exists (ABMR without activation of complement cascade) (5)
Patients with DSA who are C4d positive has lower graft survival than those who are C4d negative, this means that it is a marker for severity and has prognostic implication (6). but C4d negative patients have higher rate of developing transplant glomerulopathy if untreated
In one study it was found that C4d positive staining was present in all patients with ABMR and no patients with TCMR (7), moreover it was found that C4d was 95 % sensitive and 96 % specific for the presence of DSA
in ABO -incompatible kidney transplants some times C4d staining occur in the absence of histologic features of injury this is called graft accommodation [8].
Current treatment is directed against depleting B cells (rituximab and ATG added if mixed ABMR+ TCMR) , decreasing production of antibodies (IVIG) and removing DSA (plasmapheresis- used in early ABMR < 1 y) and decreasing inflamation produced by rejection (high dose corticosteroids) this is in combination with intensification of maintenance immunosuppression, antiviral and antimicrobial prophylaxis
Eculizumab is a monoclonal antibody, fully humanized, directed against C5 so inhibit the formation of MAC, it is FDA approved in the treatment of PNH and aHUS
In renal transplantation there is no of randomized controlled studies that prove the efficacy and safety in the treatment of ABMR, some reports its efficacy for treatment of acute refractory C4d positive (9), on the other hand, ABMR occur in patient already receiving eculizumab for other indication such as a HUS
2- Complement medicated TMA
Complement medicated TMA occur due to abnormal regulation of alternative pathway of complement due to mutation in either :
⦁ Regulators (factor H, I) in the form of loss of function or
⦁ Activators (C3, factor B) in the form of gaining of functions
leading to uncontrolled activation of complement system
TMA is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and AKI that if untreated can cause graft loss
Post transplantation TMA can occur either in the form of :
1- Recurrent disease (nearly all patients have complement medicated TMA)
It is recommended that all patients with complement mediated TMA to use living- unrelated or deceased donor kidney and all patients with recurrent TMA to be considered as having complement mediated TMA and should be treated with eculizumab.
Observational date suggest that in recurrent TMA post transplant eculizumab introduction leads to elevation of platelet count, decrease in LDH in most of patients and improve kidney function in 50% of patients (10)
2- Denovo TMA (due to immunosuppressive drugs, ABMR, viral infections)
All patients with denovo TMA should be evaluated for genetic mutations, and all possible reversible causes should be treated till having the result of genetic testing, if gentetic mutation found or if patient has progressive disease despite correcting suspected etiology, eculizumab should be started.
REFERANCES
1- Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol 2014; 5:520.
2- Kjaer TR, Thiel S, Andersen GR. Toward a structure-based comprehension of the lectin pathway of complement. Mol Immunol 2013; 56:222.
3- Thurman JM, Holers VM. The central role of the alternative complement pathway in human disease. J Immunol 2006; 176:1305.
4- Feucht HE, Felber E, Gokel MJ, et al. Vascular deposition of complement-split products in kidney allografts with cell-mediated rejection. Clin Exp Immunol 1991; 86:464.
5- Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell-mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am J Transplant 2018; 18:293.
6- Böhmig GA, Exner M, Habicht A, et al. Capillary C4d deposition in kidney allografts: a specific marker of alloantibody-dependent graft injury. J Am Soc Nephrol 2002; 13:1091.
7- Collins AB, Schneeberger EE, Pascual MA, et al. Complement activation in acute humoral renal allograft rejection: diagnostic significance of C4d deposits in peritubular capillaries. J Am Soc Nephrol 1999; 10:2208.
8- Bach FH, Turman MA, Vercellotti GM, et al. Accommodation: a working paradigm for progressing toward clinical discordant xenografting. Transplant Proc 1991; 23:205.
9- Yamamoto T, Watarai Y, Futamura K, et al. Efficacy of Eculizumab Therapy for Atypical Hemolytic Uremic Syndrome Recurrence and Antibody-Mediated Rejection Progress After Renal Transplantation With Preformed Donor-Specific Antibodies: Case Report. Transplant Proc 2017; 49:159.
10-Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 2013; 368:2169.
Over the past few decades, the introduction of CNIs into transplantation practice have decreased the frequency and severity of T cell-mediated rejection (TCMR), and patients can expect a reasonably normal lifestyle, at least in the short-term. However, T cell targeted therapies have little effect on AMR, and AMR is becoming a frequent, although not the most, cause of late graft loss. The most common diagnosis in most studies is acute TCMR where AMR accounts for around 10%of patients. . Antibody-mediated rejection (AMR) is a significant complication following kidney transplantation that contributes toward both short and long term injury in approximately 1% to 10% of kidney transplant recipients. Despite use of desensitization protocols, up to 1/3 of highly sensitized recipients may develop acute AMR following transplantation. Withdrawal or reduction of immunosuppression, noncompliance with immunosuppression, young age, pretransplant sensitization are the major risk factors for DSAs. Mostly De novo DSAs, especially HLA II antibodies are the major risk factors for chronic antibody mediated graft rejection. Clinical picture includes proteinuria, hypertension, progressive functional deterioration, and overt graft failure. https://doi.org/10.1186/s12882-021-02486-9 https://doi.org/10.1111/ctr.14320 DOI:10.1111/ajt.14584
Weam Elnazer
3 years ago
During the lifetime of a transplanted kidney, inflammatory and immunological mechanisms eventually destroy the functioning tissue to variable degrees.
The complement system plays a significant role in these negative responses. Complement activation is implicated in the pathogenesis of several disorders that result in uraemia and may lead to kidney transplant degradation.
Complement is also activated in kidney donors, especially after brain or heart death, causing organ damage that impairs graft quality and transplantation results. During procurement and preservation, the graft continues to deteriorate due to complement activation.
The main mechanism that triggers complement activation in all of these phases of the transplantation process is ischaemia, which results in phenotypic changes to parenchymal and endothelial cells that are recognized by the complement system, and the most damaging event that occurs during the ischaemic period is reperfusion of the graft with blood.
Complement has an important role in I/R injury, the severity of which is strongly associated with the degree and length of the ischaemic period.
Complement activity post-transplantation also has a role in tissue damage, particularly during acute and chronic ABMR, which can result in late-stage glomerulopathy and recurrence of kidney disease in the graft.
Complement activation in transplant recipients occurs as a result of two major mechanisms.
Most commonly, complement activation is triggered by IgG and IgM antibodies that are specific for epitopes exposed on donor ABO and HLA antigens.
However, the phenotype of the transplant recipient can also lead to complement activation owing to incompatibilities in complement components and/or regulators.
Biglarnia, AR., Huber-Lang, M., Mohlin, C. et al. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol14, 767–781 (2018). https://doi.org/10.1038/s41581-018-0071-x
Thankyou all for cntribution but as per the question asked by Pof. Alaa Ali does the temporal relation between the AMR and complement activation have an impact on decision to treat ie: IRI
Hyperacute rejection
Acute rejection
Or late AMR
Mujtaba Zuhair
3 years ago
The complement system plays an important role in graft injury ( the ischemia-reperfusion injury which leads to delayed graft function , in the antibody mediated rejection, and in the recurrence of atypical HUS post transplantation)
Eculizumab which is a fully humanized monoclonal antibody against C5 used in various aspects of kidney transplantation. it blocks the terminal complement system and prevent the formation of membrane attack complex (C5-C9) so it prevents direct tissue injury by the complement system but it dose not reduce the effect of early complement system on the graft like C3b which is a powerful opsonization factor or C3a and C5a which are powerful chemotactic factors which can lead to graft injury indirectly by recruiting inflammatory cells into the graft .
one study studied the use of eculizumab to prevent the DGF in high risk patients in deceased donor transplantation , the result showed no difference between ecluzumab and placebo.
Eculizumab in the prevention of antibody mediated rejection: in one study reported by Stegal et al , they study the use of eculizumab in highly sensitized patients with live donor transplantation . they found that the eculizumab treated patients had lower incidence of AMR at one year, but with high rates of subclinical rejection. Another studies with larger size is needed to confirm this results and to clarify the dose of eculizumab and the duration of tratment.
Eculizumab had been used successfully in the prevention and treatment of recurrent aHUS post transplantation .
C1 inhibitors is another drug used in small studies to prevent AMR in highly sensitized patients with promising early results .
Reference :
Ch. Legendre, R. Sberro-Soussan, J. Zuber and V. Frémeaux-Bacchi
The role of complement inhibition in kidney transplantation
British Medical Bulletin, 2017, 124:5–17 doi: 10.1093/bmb/ldx037
MOHAMMED GAFAR medi913911@gmail.com
3 years ago
The complement system, traditionally considered a component of innate immunity required for protection from invading pathogens,
Complement activation can be initiated through three pathways
The recognition that complement participates in antibody-initiated allograft rejection suggested that identifying serum anti-HLA antibodies capable of binding C1q would enhance their prognostic use after kidney transplantation
A 2013 paper, indeed, suggests that, among patients with serum anti-HLA antibodies, those binding to C1q1 had the worst kidney graft survival
In another example of complement-based diagnostics, C4d staining of kidney transplant tissue is currently considered one key criterion for diagnosing antibody-mediated allograft rejection
Clin J Am Soc Nephrol 10: 1636–1650, 2015. doi: 10.2215/CJN.06230614
If you have a negative C4d AMR resistant to coventional methods wouid you use cmplement controling drugs?
Mohamad Habli
3 years ago
Complement cascade is an important part of the innate immune system. Studies have shown that activation of complement cascade is implicated in allograft injury by means of several mechanisms, including ischemia/reperfusion injury, antibody mediated rejection, cellular rejection and thrombotic micro-angiopathy.
The complement cascade consists of more than 30 soluble and cell-bound proteins.They become biologically active fragments, complement receptors, and complement regulatory proteins whenever the transplanted organ is exposed to recipient complement proteins as soon as it is reperfused. The complement system is not only involved in antibody-mediated cell lysis, which is the major effector mechanism, but that is only one of its functions. The complement system can be activated in an antibody-independent fashion. Complement fragments upregulate T cell differentiation and the B cell response to antigens. Once complement cascade is activated, within a transplanted organ it can have direct and indirect pathologic effects. The activation can occur through classical, alternative, or lectin pathway, resulting in the creation of anaphylatoxins C3a and C5a which are involved in possessing several immune functions, and the assembly of terminal complement cascade, result in direct cell lysis.
C3a and C5a are proteins that directly affect resident organ cells, they are activators for neutrophils and macrophages, and they provide important signals for T and B cell activation in the tubulointerstitium in the setting of ischemia reperfusion, whereas in AMR activation occurs in the peritubular capillaries.
Owing to the great role of complement in allograft injury, complement system can be considered as potential therapeutic targets. Recently, the inhibition of several complement components has shown certain results in preventing the antibody-mediated rejection. C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking not only C1 complex components. Very recently, a double-blind study assessed the safety of C1-inhibitor in kidney transplant recipients and suggested the positive effect of this treatment regarding the C1q+HLA antibody levels reduction and antibody-mediated rejection occurrence.
Eculizumab is a humanized monoclonal antibody capable of blocking the cleavage of C5 complement component, thus preventing the release of anaphylatoxin C5a and the assembly of MAC. The C5 component participates in all three main pathways of complement activation; therefore, the blockade of its cleavage can have a positive influence on various medical conditions regardless of the active pathway. Eculizumab was shown to be an effective treatment for paroxysmal nocturnal hemoglobinuria and aHUS including its recurrence in kidney transplantation. REFRENCES: https://doi.org/10.3389/fimmu.2019.02380 https://doi.org/10.3389/fmed.2017.00066 doi:10.1097/MOT.0000000000000216. doi: 10.1097/TP.0000000000000592 doi:10.1056/NEJMoa061648 doi:10.1053/j.ajkd.2015.12.034
Excellent Mohamed I will quote this from your reply “The complement system is not only involved in antibody-mediated cell lysis, which is the major effector mechanism, but that is only one of its functions. The complement system can be activated in an antibody-independent fashion. Complement fragments upregulate T cell differentiation and the B cell response to antigens. Once complement cascade is activated, within a transplanted organ it can have direct and indirect pathologic effects”.
Dear All Can you summarise the direct and indirect effects of complement activation?
Once complement is activated in response to trigger, it can have direct and indirect effects on immune system.
Direct effect is explained by the activation of complement cascade by the bound antigen to antibodies ( donor HLA to T or B lymphocytes resulting is cytotoxic effect and lysis.
Multiple different biologically active complement fragments are generated during activation of complement cascade. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation.
Direct effect when complement causes cell lysis after antigen-antibody interaction
Indirect effect are related to the complement fragments e.g. C3, C5. They can acts as anaphylatoxin and play role in inflammatory process and induces damages
Riham Marzouk
3 years ago
Complement pathway is activated upon renal transplantation through the following:
Ischemic reperfusion injury which produces ROS reactive oxidative stress factors which initiate complement pathway producing C5a and C5b
Also, C3a and C3b are produced during complement activation pathway.
Renal (tubules) and hepatic synthesis of C3.
Collectin -11 which is soluble lectin expressed in kidney tissue and responsible for complement activation in the kidney, increased with ischemic stress and trigger complement activation. The C5a receptor antagonist has therapeutic effects in any autoimmune disease , C5a is a major inflammatory peptide in antibody mediated rejection so its block may be another hope against transplant associated inflammatory responses. Christopher L. Nauser, Conrad A. Farrar and Steven H. Sacks. Complement Recognition Pathways in Renal Transplantation. JASN September 2017, 28 (9) 2571-2578. C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi. The role of complement inhibition in kidney transplantation. British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17.
Huda Al-Taee
3 years ago
The process of transplantation involves critical events that occur in the donor, recipient and the graft leading to graft damage. These events trigger an early inflammatory reaction that is independent of donor specific immune response of the recipient. Ischemia and subsequent reperfusion drive the initial inflammatory process of which complement activation is a major effector mechanism.
In kidney transplantation, several sequential events that occur before, during and after transplantation and lead to complement activation and loss of kidney function including:
Pre-transplantation:
complement activation occur with each hemodialysis session.
deceased donor: DCD, DBD
Early pre/ post transplant:
procurement, preservation & transportation
reperfusion
ABMR
Late post transplant:
dysregulation of alternative pathway
recurrence of disease
rejection
transplant glomerulopathy
age related functional loss
A number of agents and several strategies have been developed to deactivate complement system including:
optimization of graft quality before transplantation
treatment of antibody mediated rejection
induction of accommodation
regulation of adaptive immunity
The agents that can deactivate the complement system include:
IVIG, Rituximab, C1INH( clinical trial), Eculizumab( clinical trial), BIVV009( clinical trial), Ides( clinical trial), Mirococept( clinical trial), Compstatin family inhibitors( clinical trial), sCR1( pre clinical development), TT30( pre clinical development), C5aR1 antagonist( pre clinical development), cobra venom factor( pre clinical development).
Although the initial results of clinical trials are promising, inconsistencies in their effect on delayed graft function and ABMR reflect the complexity of allograft injury and the involvement of non-complement mediated factors.
References:
Biglarnia A.R. Huber-Lang M., Mohlin C., Ekdahl K.N., et al. The multifaceted role of complement in kidney transplantation. Nature Reviews Nephrology. 2018, Dec., Vol.14. 767-781.
Thank you, Huda Would you elaborate more on the outcomes of the currently available agents? Furthermore, how to balance between benefits and drawbacks of each according to the available data?
Last edited 3 years ago by Ala Ali
Prakash Ghogale
3 years ago
Terminal C5 inhibition
Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: A randomized trial
William H Marks et al
At week 9, treatment failure rate ( AMR BANFF II and III) was 11.8% in eculizumab group vs 21.6% in standard of care group. when BANFF I AMR was included, treatment failure was 11.8% vs 29.4% in standard of care group. This finding shows that eculizumab may be more effective than SOC at preventing acute AMR in recipients who are sensitized to their living-donor kidney transplants.
Safety and efficacy of eculizumab for the prevention of antibody-mediated rejection after deceased-donor kidney transplantation in patients with preformed donor-specific antibodies
Denis Glotz et al
Participants received eculizumab as follows:
1200 mg immediately before reperfusion
900 mg on posttransplant days 1, 7, 14, 21, and 28
1200 mg at weeks 5, 7, and 9.
All patients received thymoglobulin induction therapy and standard maintenance immunosuppression including steroids.
The primary end point was treatment failure rate composed of biopsy-proved grade II/III AMR within 9 weeks posttransplant. In 80 patients the Observed treatment failure rate (8.8%) was significantly lower than expected for standard care (40%). At 36 months, graft and patient survival rates were 83.4% and 91.5%, respectively. Eculizumab was well tolerated, and there were no new safety concerns. In such patients, eculizumab has the potential to give prophylaxis against damage caused by acute AMR.
Proximal inhibition
C1 Inhibitor in Acute Antibody-Mediated Rejection Nonresponsive to Conventional Therapy in Kidney Transplant Recipients: A Pilot Study
D. Viglietti et al
C1 inhibitor (C1-INH) Berinert was added to high-dose intravenous immunoglobulin for the treatment of acute ABMR that is nonresponsive to conventional therapy. Kidney recipients with nonresponsive active ABMR and acute allograft dysfunction C1-INH and IVIG for 6 months (six patients). The primary end point was the change in eGFR at 6 months after inclusion (M+6). All patients showed an improvement in eGFR between inclusion and M+6. In kidney patients with nonresponsive acute ABMR, C1-INH administered with IVIG is safe and may enhance allograft function.
Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney
Transplantation: Results of a Randomized DoubleBlind Placebo-Controlled Pilot Study
R. A. Montgomery et al
Human plasma derived C1 INH was added to plasmapheresis and ivIg for AMR.20000 units of C1 INH in divided doses was given every other day for 2 weeks .no difference was found between C1 INH and standard of care group in day 20 pathology and graft survival,however C1 INH group demonstrated sustained improvement in renal function .Transplant glomerulopathy was not seen in any of the biopsy done in C1 INH group at 6 months. This study suggests that adding C1 INH in the treatment of AMRto plasmapharesis and ivIg could be beneficial.
Long term follow-up
Despite the encouraging findings of the above trials, long-term follow-up of eculizumab-treated positive crossmatch patients in a single-center trial revealed that, despite the avoidance of early active AMR, the long-term effects of eculizumab in Allograft survival and the incidence of chronic AMR are comparable to historical controls.
Long-term outcomes of eculizumab-treated positive crossmatch recipients: Allograft survival, histologic findings, and natural history of the donor-specific antibodies
Carrie A. Schinstock et al
Positive Crossmatch Kidney Transplant Recipien Treated With Eculizumab: Outcomes Beyond 1 Year
L. D. Cornell et al
Use of Eculizumab for Active Antibody-Mediated Rejection that Occurs Early Post-Kidney Transplantation: A Consecutive Series of 15 Cases
Ek Khoon Tan et al
They concluded that prompt eculizumab treatment is safe and effective for early active AMR after kidney transplant or when abrupt increases in donor-specific antibodies is seen and biopsy cannot be performed for diagnosis confirmation.
Although eculizumab has been used to prevent ABMR in highly sensitized patients and also as a salvage agent in treatment of refractory active ABMR, because of limitations I have mentioned in my last comment and, in addition, lack of randomized clinical trials proving its efficacy and safety, it is not the first line therapy of ABMR
my last comment: A variety of reports show a range of responses to eculizumab from very effective to no effect in patients with ABMR. Few studies demonstrate that eculizumab is not useful in patients with non- C1q binding HLA- DSAs, only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti- complement treatment. On the other hand, complement inhibition at the level of C5 does not prevent upstream complement activity, more over residual terminal pathway activity via the alternative pathway can occur despite eculizumab treatment.
Complement system is a part of innate immune system which also comprised of dendrites ,monocytes, macrophages .
The complement system is activated through three distinct pathways:
classical pathway
Alternative pathway
Lectin pathway.
the role of complement in kidney transplantation including the following conditions:
-ischaemia-reperfusion injury
-antibody-mediated rejection
-recurrence of native kidney disease as : atypical HUS , C3 glomerulopathy, anti-phospholipid syndrome .
**In ischemia reperfusion injury:
ischaemia-reperfusion-induced endothelial injury results in activation of lectin pathway via binding of mannose-binding lectin (MBL) to epitopes exposed on ischaemic tissue.
The alternative pathway amplifies this, while the classical pathway role remains controversial.
This lesions are reduced by deficiency or depletion of complement factor as C3, C5, CFB, C3aR and C5aR and enhanced by deficiency of complement regulator as decay-accelerating factor (DAF)
The involvement of complement is suggested by:
detection of soluble C5b-9 in deceased kidney but not living one.
increased in complement genes expression in deceased-donor kidneys
brain-death induce activation of complement
up- regulation of C5aR expression in renal tubular cells.
**ABMR :
Immune – complex combining antigens (MHC molecules, ABO-blood group antigens) and IgM/IgG antibodies called donor-specific antibodies (DSAs) leads to activation of the classical pathway to start the process of ABMR.
Diagnosis of ABMR depend on detection of detection of complement fragments C4d on peritubular capillaries .
However , cases of C4d negative ABMR have been reported , due to technical reasons, or a low level of complement of classical pathway.
**Recurrence of disease involving mainly the alternate pathway :
-Atypical haemolytic uraemic syndrome (aHUS)
recurrence occurs in 68% of patients which is marked during the first year (with an incidence of 70%) and decreases markedly thereafter.
Patients with mutations in complement factors have a 3 folds increase in aHUS recurrences post transplant compared to aHUS patients without mutations.
-C3 glomerulopathy :
Is characterized by C3 deposits in the mesangium and along the glomerular basement membrane (in cases of C3 glomerulonephritis) or within the GBM (in cases of DDD).
The disease carries a poor prognosis with progression to ESRD in 50% of cases during the first decade after initial presentation , and also recurs after kidney transplantation in ~50% of cases.
Complement inhibition after Renal transplantation :
-Eculizumab: is a recombinant monoclonal antibody against C5.
It minimizes immunogenicity, Fc-mediated functions and complement activation , so prevents delayed graft function, in acute ABMR and for treatment or prevention of aHUS recurrence and anti- phospholipid syndrome.
It increases the risk of meningitis due to the Neisseria species , so Prophylaxis with vaccination and antibiotics is mandatory.
-C1 estrase inhibitor (C1-INH) block classical pathway of complement activation by C1s &C1r inhibition.
Reference:
Legendre .CH, Sberro-Soussan .R, et al.The role of complement inhibition in kidney transplantation, British Medical Bulletin,(2017),124 :5-17,doi: 10.1093/bmb/ldx037
Dear All
Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement?
Give examples
Complement pathway is activated upon renal transplantation through the following:
Ischemic reperfusion injury which produces ROS reactive oxidative stress factors which initiate complement pathway producing C5a and C5b
Also, C3a and C3b are produced during complement activation pathway.
Renal (tubules) and hepatic synthesis of C3.
Collectin -11 which is soluble lectin expressed in kidney tissue and responsible for complement activation in the kidney, increased with ischemic stress and trigger complement activation.
The C5a receptor antagonist has therapeutic effects in any autoimmune disease , C5a is a major inflammatory peptide in antibody mediated rejection so its block may be another hope against transplant associated inflammatory responses.
Christopher L. Nauser, Conrad A. Farrar and Steven H. Sacks. Complement Recognition Pathways in Renal Transplantation. JASN September 2017, 28 (9) 2571-2578.
C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi. The role of complement inhibition in kidney transplantation. British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17.
The role of complement in graft damage;
Complement is implicated in the following events ,that influence graft damage;
1-Ischemia reperfusion injury ;
2- Anti body mediated rejection
3- Cell mediated rejection
4- Post transplant TMA
Therapeutics targeting complement:
1-Human intravenous immunoglobulins (IV IG)
They inhibit complement activation by blocking the receptor for the Fc fragment of IG or the C1q component.
2-Eculizumab:
Inhibitor of C5 complement component, monoclonal antibody,
3-C1 esterase inhibitor
inhibits the activation of both the classical and lectin complement pathways
used in treatment of recurrent TMA post transplant .
Reference ;
1-Schinstock, C.A.; Mannon, R.B.; Budde, K.; Chong, A.S.; Haas, M.; Knechtle, S.; Lefaucheur, C.; Montgomery, R.A.; Nickerson, P.; Tullius, S.G.; et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation. Transplantation 2020, 104, 911–922.
2-Stegall, M.D.; Diwan, T.; Raghavaiah, S.; Cornell, L.D.; Burns, J.; Dean, P.G.; Cosio, F.G.; Gandhi, M.J.; Kremers, W.; Gloor, J.M. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am. J. Transplant. 2011, 11, 2405–2413.
3- Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant (2015) 15(5):1293–302.
4- Gonzalez Suarez, M.L.; Thongprayoon, C.; Mao, M.A.; Leeaphorn, N.; Bathini, T.; Cheungpasitporn, W. Outcomes of Kidney Transplant Patients with Atypical Hemolytic Uremic Syndrome Treated with Eculizumab: A Systematic Review and MetaAnalysis. J. Clin. Med. 2019, 8, 919.
5-Vo, A.A.; Zeevi, A.; Choi, J.; Cisneros, K.; Toyoda, M.; Kahwaji, J.; Peng, A.; Villicana, R.; Puliyanda, D.; Reinsmoen, N.; et al. A Phase I/II Placebo-Controlled Trial of C1-Inhibitor for Prevention of Antibody-Mediated Rejection in HLA Sensitized Patients. Transplantation 2015, 99, 299–308.
Complement system has 3 pathways for activation; Classic (Ag-Ab complex), Lectin pathway(microorganism)and alternative pathway.
ABMR: plasma cells produce antibodies which attach to antigen and initiate the classic pathway leaving its effect in the graft (C4d deposition in peritubular capillaries).
TMA post-transplant produce effect through complement pathway.
T-cell mediated rejection occurs as APC produce c3a and c5a in the activation of cellular immunity.
Drugs interfering with complement system
IVIG which produces its effect through binding to fc region on IgG and inhibit complement activation and usually used in ABMR.
Eculizumab: C5 inhibitor as being very expensive , few studies were done and showed no beneficial impact in preventing rejection.
C1estrase inhibitor which acts against both Lectin and classic pathways.
Under trials: BIVV009,Rituximab, Ides. Compstatin family inhibitors.
s CR., TT30., C5aR1 antagonist., Cobra venom factor.
Can graft injury happen without activation of complement?
Graft injury can occur away from complement activation.
Anti-HLA-class I antibodies can produce pathological effects ending with changes in the vascular intima and chronic rejection.
Without complement activation, C4d-negative AMR together with high anti-AT1R result in severe vascular rejection.
Reference
Loupy, A; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
. American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions,
Haas .M et al, 28 January 2014
antiHLA class 1,11 antigens DSA antibodies can lead to garft damage through the complement activation but still about 40% to 50% of graft rejection that associated with severe vascular changes, such as fibrinoid necrosis, are C4d negative,due to the non–complement fixing antibodies. like AT1R-antibodies (AT1R-Ab) can promote antibody-mediated rejection (AMR) either alone or together with HLA-DSA(1).
reference:
1-Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation Aurelie Philippe1,2:Kidney Int Rep (2019) 4, 510–512; https://doi.org/10.1016/j.ekir.2019.01.012.
Complement system is activated by three pathways; Classic (Ag-Ab complex), Lactin pathway(microorganism)and alternative pathway.
ABMR: plasma cells form antibodies which attach to antigen and start the classic pathway leaving its fingerprint in the graft (C4d deposition in peritubular capillaries).
TMA post-transplant takes effect via complement pathway.
T-cell mediated rejection happens when APC produce c3a and c5a in the activation of cellular immunity.
Drugs which interfere with complement system
IVIG…which bind to fc region on IgG and inhibit complement activation and widely used in ABMR
Eculizumab: C5 inhibitor because of its very expensive drug few studies were addressed and show no benefits in preventing rejection
C1estrase inhibitor which inhibits both Lactin and classic pathways.
Under trials: Rituximab, BIVV009, Ides., Mirococept., Compstatin family inhibitors.
sCR., TT30., C5aR1 antagonist., cobra venom factor.
Can graft injury happen without activation of complement?
Yes, it can happen without complement activation.
Anti-HLA-class I antibodies can produce pathological effects ending with changes in the vascular intima and chronic rejection.
C4d-negative AMR with high anti-AT1R produces a severe vascular rejection without complement activation.
Reference
1- Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
2- Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
3- Valenzuela NM, McNamara JT, Reed EF. Antibody-mediated graft injury: complement-dependent and complement-independent mechanisms. Curr Opin Organ Transplant 2014;19:33-40.
4- American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions,
Haas.M et al, 28 January 2014
The complement system is a series of protein kinases that are sequentially activated and culminate in the formation of the membrane attack complex (MAC).
The MAC comprises complement components C5 to C9, which are inserted into the cell membrane (pathogen or host), disrupt- ing integrity and causing cell lysis .
In addition, many proximal complement components may augment the immune response to the allograft. The complement system may be activated by three path- ways: the classical pathway, the alternative pathway, and the MBL pathway. IgM or IgG immune complexes activate the classical pathway, and hence this pathway may become acti- vated during antibody-mediated rejection (see section on B Cell Activation). The alternative pathway is constitutively active and must be controlled by a series of regulatory protein
Reference
Kidney
Transplantation Principles and Practice
EIGHTH EDITION
Stuart J. Knechtle, MD, FACS
Complement :protein sensitized in liver and activated by many ways as (AG-AB complex)classic pathway or alternative pathway by bacterial endotoxin .
In kidney transplantation it play a role in innate immunity by activate macrophages
Or also by make cell lysis by AG-AB complex.
Eclizumab anti c5( used in ttt of atypical HUS) will corrupt the c5 so stop classical & alternative pathway of its activation .also c1 inhibitor .
Graft injury can happen without complement activation by AG AB complex it self ( deposition in kidney) also nephrotoxicity of CNI on graft kidney (TMA)
Infection by BK virus or CMV
complement systemis important in Ab mediated rejection . therefore the component of the complement system has become apotential therapuetic targets .
C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking not only C1 complex components , as the name suggests, but also MASP-1 and MASP-2 . It is, therefore, an effective inhibitor of both classical and lectin pathway activation . Earlier study on non-human primate model showed a potential for inhibition of acute antibody-mediated rejection with C1-inhibitor . Very recently, a double-blind study assessed the safety of C1-inhibitor in kidney transplant recipients and suggested the positive effect of this treatment regarding the C1q+HLA antibody levels reduction and antibody-mediated rejection occurrence . The possible use of complement inhibitor based on viral coat proteins is also a subject of ongoing research .
Eculizumab is a humanized monoclonal antibody capable of blocking the cleavage of C5 complement component, thus preventing the release of anaphylatoxin C5a and the assembly of MAC . The C5 component participates in all three main pathways of complement activation; therefore, the blockade of its cleavage can have a positive influence on various medical conditions regardless of the active pathway. Eculizumab was shown to be an effective treatment for paroxysmal nocturnal hemoglobinuria and aHUS including its recurrence in kidney transplantation . Since the complement system is a prominent factor during antibody-mediated rejection, the effects of eculizumab were studied with regard to this condition as well. Stegall et al. studied the effects of terminal complement inhibition on patients with a positive crossmatch and concluded that eculizumab is capable of decreasing the incidence of acute antibody-mediated rejection. Some success in treating the acute antibody-mediated rejection in renal allograft was also reported in several case reports . Extended follow-up study, however, showed that eculizumab treatment has no effect on chronic antibody-mediated rejection in recipients with significant amount of DSA. Moreover, there are known cases of C4d-negative antibody-mediated rejection where the use of eculizumab was ineffective . Similarly, the existence of IgM-mediated acute antibody-mediated rejection was described in patients treated with eculizumab . Therefore, C5 inhibition might be efficacious in the treatment of acute antibody-mediated rejection at least in some cases; however, these effects are lost in preventing chronic rejection development.
references
The complement cascade is comprised of more than 30 soluble and cell-bound proteins .
●The complement system is activated through three distinct pathways: the classical pathway (CP), lectin pathway (LP), and alternative pathway (AP).
●complement activation
Complement activation in antibody-mediated rejection. Antibody-mediated rejection is caused by binding of antibodies to human leukocyte antigens (HLA) expressed on endothelial cells of the transplanted organ. The antibodies (referred to as donor specific antibodies, or DSA) activate the classical pathway of complement. Classical pathway activation causes the cleavage of C4, and one of the resultant C4 fragments (C4b) is covalently attached to target surfaces. C4b comprises part of the classical pathway C3 convertase, C4b2a. C3b can become covalently attached to target cells, similar to C4b. A protease called factor I (FI) controls complement activation by cleaving the C4b and C3b molecules, thereby stopping convertase activity. Although they are no longer catalytically active, the C4d and C3dg fragments remain bound to the target cells and can be detected by immunostaining of tissue biopsies
●complement inhibitors
In spite of promising pre-clinical data, a trial that enrolled 27 kidney transplant patients at high risk of DGF who were randomized to treatment with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benefit with treatment . Another clinical trial is ongoing, however, in which kidneys treated with an agent that attaches a complement regulator to cell membranes . This approach was previously shown to be beneficial in a rat kidney transplant model .
●eculizumab
treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in kidney transplant recipients with AMR.
●C1-INH is a substrate-like serine protease inhibitor that blocks several proteases, including C1r, C1s, and the MASPs. Originally used as a replacement therapy for patients with hereditary angioedema who have deficiency of C1-INH protein, it has also been tested as a treatment of AMR . C1-INH appeared to be beneficial in a small trial of six AMR patients who were refractory to conventional therapy ,
this approach holds promise as a novel treatment for transplant patients.
Although eculizumab and C1-INH have shown promise in case reports and small series, their role in transplant medicine requires further study. Many additional anti-complement therapeutics are in clinical development, and some of these new drugs block individual activation pathways or specific components of the complement cascade. This could potentially allow clinicians to block the parts of the complement cascade involved in allograft injury while leaving other parts of the cascade active.
However, testing these new agents in the transplant setting poses several challenges. First, all transplant recipients are treated with multiple immunosuppressive drugs. Thus, new drugs will need to be tested as add-on treatments to these other agents. Second, even within a single diagnosis, such as AMR, there is patient heterogeneity. Complement activation may not be an important part of C4d-negative AMR, for example. The development of new complement biomarkers may therefore be critical for selecting patients most likely to benefit from complement inhibitors, and for discerning a response to treatment.
●references
.Zhuang Q, Lakkis FG. Dendritic cells and innate immunity in kidney transplantation. Kidney Int. (2015)
.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6788431/#!po=7.00935
The complement cascade is an important part of the innate immune system. Studies have shown that complement activation contributes to allograft injury in several clinical settings, including ischemia/reperfusion injury and antibody-mediated rejection. Furthermore, the complement system plays a critical role in modulating the responses of T cells and B cells to antigens. Therapeutic complement inhibitors, therefore, may be effective for protecting transplanted organs from several causes of inflammatory injury. Although several anti-complement drugs have shown promise in selected patients, the role of these drugs in transplantation medicine requires further study. Treatment with the C5 inhibitor led to long-term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft.
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR. However, larger series in transplant patients have not shown a consistent benefit, however, and the role of eculizumab for preventing or treatment AMR is not yet clear. C1-INH is a substrate-like serine protease inhibitor that blocks several proteases, including C1r, C1s, and the MASPs. Originally used as replacement therapy for patients with hereditary angioedema who have a deficiency of C1-INH protein, it has also been tested as a treatment of AMR. C1-INH appeared to be beneficial in a small trial of six AMR patients who were refractory to conventional therapy.
Pre-clinical work has shown that other complement inhibitory agents may be useful in the transplant setting, including an AP inhibitor, an LP inhibitor, and C5a blockade. Many new complement inhibitory drugs are in clinical development, some of which will likely soon become available for clinical use.
Reference:
1. Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019 Oct 4;10:2380.
2. Viglietti D, Gosset C, Loupy A, Deville L, Verine J, Zeevi A, et al. C1-inhibitor in acute antibody-mediated rejection non-responsive to conventional therapy in kidney transplant recipients: a pilot study. Am J Transplant. (2015) 16:1596–603.
complement activation contributes to allograft injury in several ways including
ischemia/reperfusion injury.And antibody mediated rejection.
Furthermore, the complement system plays critical roles in modulating the responses of T cells and B cells to antigens.
Therapeutic complement inhibitors, therefore, may be effective for protecting transplanted organs from several causes of inflammatory injury.
Although the complement system is an important effector mechanism for antibody-mediated cytotoxicity, that is only one of its functions. The complement system can be activated in an antibody-independent fashion
The transplanted organ is exposed to recipient complement proteins as soon as it is reperfused. Conversely, complement proteins and fragments generated within the allograft enter the systemic circulation.
Complement fragments also modulate T cell differentiation and the B cell response to antigens.
Consequently, this system modulates the adaptive immune response, mediates many of the downstream effects of B and T cell immunity, and can function independently of the adaptive immune response. Furthermore, the complement cascade interacts with other biologic systems, including toll-like receptors, the inflammasome, and the clotting cascade .
The complement system is activated through three distinct pathways: the classical pathway (CP), lectin pathway (LP), and alternative pathway (AP). These activation pathways can be engaged by different pathologic processes in the allograft, including donor brain death, I/R injury, and antibody mediated rejection.
The CP is activated by antibodies bound to their target ligands. This may be particularly important in those transplant recipients with donor specific antibodies (DSA) reactive to polymorphic human leukocyte antigens (HLA) expressed on endothelial cells of the allograft.
Complement activation through all three pathways leads to cleavage of the C3 protein, generating the C3a and C3b fragments.
C3b binds to nearby surfaces, thereby marking, or “opsonizing,” target cells and surfaces.
Full complement activation also leads to cleavage of C5, generating soluble C5a and the larger C5b fragment. C5b seeds the formation of the membrane attack complex (MAC, or C5b-9), a multimeric complex that forms a pore in target cells and can cause target cell activation or lysis .
Complement Receptors
Although the MAC directly affects target cells, most of the biologic effects of complement system are mediated by receptors for the various fragments. The C3a receptor (C3aR) and C5a receptors (C5aR1 and C5aR2) ,C5aR1 expression, increases in rejecting murine renal allografts .
Once complement is activated within a transplanted organ it can have direct and indirect pathologic effects. multiple different biologically active complement fragments are generated. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation.
In kidney ischemia, activation primarily occurs in the tubulointerstitium whereas in AMR activation occurs in the peritubular capillaries .
Patients with DSA that bind to C1q or to which C3d is bound are at greater risk of developing AMR and they have a worse overall prognosis .
Interestingly, treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
Complement Therapeutics
Eculizumab
IT been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR .
It has also been used in transplant recipients with post-transplant aHUS recurrences .
C5aR1 antagonist :there is pre-clinical data showing that C5aR1 blockade may be a beneficial treatment for rejection , this approach holds promise as a novel treatment for transplant patients.
C1-INH :has also been used to prevent AMR, and it is being tested for treatment of the disease in an ongoing clinical trial.
Pre-clinical work has shown that other complement inhibitory agents may be useful in the transplant setting, including an AP inhibitor (63), a LP inhibitor (56), and C5a blockade . Many new complement inhibitory drugs are in clinical development, some of which will likely soon become available for clinical use .
Monica Grafals and Joshua M. Thurman.The Role of Complement in Organ Transplantation.Front Immunol. (2019); 10: 2380.
The complement system has an important role in kidney transplantation especially in ABMR, ischemia reperfusion injury, and recurrence of the original disease as in case of C3 glomerulopathy, atypical HUS, and antiphospholipid syndrome.
The risk of complement mediated ischemia reperfusion injury increases in case of deceased donor due to the presence of soluble C5b-9 in deceased donor graft, and increased expression of complement genes in the deceased donor, and also due to brain death induced complement activation.
The presence of C4d on peritubular capillaries using immunofluorescence is a positive marker for diagnosis of ABMR and also for grading of its severity.
Graft outcome could be predicted by the detection of C3d binding DSA.
There are 3 available ways to inhibit the complement activation cascade:
A monoclonal antibody directed against C5 ( eculizumab ), and 2 plasma derived C1 inhibition.
In renal transplantation, eculizumab can be used to prevent ABMR and for treatment and prevention of aHUS, and it showed a promising results.
On the other hand, data proved that the effect of C1 inhibition on T and B cells is more potent than the effect of eculizumab, and there are trials to use C1 inhibitors to prevent DGF.
REF:
role of complement inhibition in kidney transplantation | British Medical Bulletin | Oxford Academic (oup.com)
Complement is part of innate immunity ,there are three pathways of complement activation:
* the classical pathway, which is triggered directly by pathogen or indirectly by antibody binding to the pathogen surface .
*the MB-lectin pathway.
*and the alternative pathway, which also provides an amplification loop for the other two pathways.
If complement is activated within a transplanted organ it can have direct and indirect pathologic effects. Multiple different biologically active complement fragments are generated. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation. The location of complement activation will vary in different settings. In kidney ischemia, for example, activation primarily occurs in the tubulointerstitium , whereas in AMR activation occurs in the peritubular capillaries . The location of activation determines which cells will be directly affected by MAC or opsonization with C3b. Soluble fragments such as C3a and C5a can have more distant effects, but the site of activation may affect their access to the circulation and peripheral blood cells. It is useful to understand the contribution of the individual complement fragments to injury, as drugs that target specific fragments are in development.
~Complement in Ischemia/Reperfusion Injury:
Several studies have shown that complement is activated after I/R, although the mechanisms may vary between different organs.
Complement activation in the ischemic heart and intestine may be initiated by immunoglobulin, but it primarily involves the LP . In the kidney, complement activation primarily involves the AP and does not require immunoglobulin . Studies in which kidneys from C3 deficient mice were transplanted into wild-type recipients revealed that the allograft itself may be an important source of complement proteins involved in tubulointerstitial activation .
~Complement inhibitory drugs have proven effective in several pre-clinical models of I/R injury. An inhibitory antibody to C5 and a small molecule C5a receptor antagonist were each protective in models of cardiac and kidney I/R injury. LP blockade was protective in models of kidney and cardiac I/R injury .A monoclonal antibody that inhibits the AP was protective in a model of warm I/R injury of the kidney . This same drug also prevented I/R injury in a mouse kidney transplant model, and it also reduced T cell mediated rejection of the organs .
~Antibody Mediated Rejection
In patients with acute and chronic AMR, DSA binds to donor HLA expressed on endothelial cells and activates the CP. Complement activation on the endothelial cell surface is believed to be an important cause of injury to the capillaries .The diagnosis of AMR is based on detecting DSA in the plasma, microvascular inflammation on a biopsy (e.g., glomerulitis or peritubular capillaritis in allografts), and C4d deposition in the peritubular capillaries . However, the diagnostic criteria have been modified to account for C4d-negative cases . It is not known whether the C4d-negative cases of AMR are caused by non-complement-mediated injurious effects of the DSA, or whether it simply reflects variability in the ability to detect the C4d.
Because complement activation by the DSA is such an important component of AMR, assays have been developed to distinguish the complement activating potential of DSA in the circulation. These assays identify immunoglobulin that binds to specific HLA types, and also tests whether the detected antibodies bind to C1q or carry a C3d molecule . Patients with DSA that bind to C1q or to which C3d is bound are at greater risk of developing AMR and they have a worse overall prognosis . These findings highlight the importance of the complement system in AMR, and potentially provide a test for identifying patients at risk of AMR. An inhibitory antibody to C5 was protective in a model of heart transplantation in highly sensitized mice, supporting the importance of complement activation in the pathogenesis of microvascular injury .Interestingly, treatment with the C5 inhibitor led to long term allograft survival, even though DSA persisted after the treatment was stopped. It is possible that complement inhibition induced “accommodation” in the allograft. Accomodation is a state in which an allograft becomes resistant to AMR. This may occur through upregulation of complement regulatory proteins, altered expression of the target antigens by the allograft, or changes in the isotype of the DSA .
References:
1. Janeway CA, Jr. How the immune system works to protect the host from infection: a personal view. Proc Natl Acad Sci USA. (2001) 98:7461–8. 10.1073/pnas.131202998 .
2. Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. (2007) 449:819–26. 10.1038/nature06246.
3. Palm NW, Medzhitov R. Pattern recognition receptors and control of adaptive immunity. Immunol Rev. (2009) 227:221–33. 10.1111/j.1600-065X.2008.00731.
Complement has three activation pathways: Classic, alternative and lectin. All are contributing in kidney transplantation. Role of complement begins before transplantation in primary kidney disease such as; C3 glomerulopathy or aHUS and hemodialysis and continues with inflammation associated with brain death, stress of surgery and ischemic-reperfusion injury. In addition its activation can occur during and after organ donation especially in deceased donor. Ischemia- reperfusion induces heparanase and metalloproteinase and causes damage to endothelial cells and loss of complement regulators. Presence of DSA against endothelial cells may activate complement. Dysregulation of alternative complement system may cause damage or sometimes recurrence of primary kidney disease in allograft. Complement activation has important role in humoral or cellular rejection and chronic AMR. Some complement fragments have chemotactic or thrombogenic and induce apoptosis and necrosis.
Anti-ABO or DSAs can trigger complement and cause allograft injury. C4d positive staining shows classic pathway activation of complement system in ABMR. DSAs with C3 and C1q binding capacities can be more harmful to graft. Strategies for complement inhibition in graft:
-Better quality of graft by sCR1 and C1INH and improvement in graft preserving solution
-Treatment of complement-dependent ABMR with eculizumab (anti-C5 monoclonal antibody), C1INH (Berinert), C1 monoclonal antibody B1VV009 or C3 inhibitor (CP40) as an addition to standard therapy (plasmapheresis, IVIG and rituximab). A recombinant endopeptidase (IdeS) can cleave hinge region of IgG subclasses. So, IgG cannot bind to C1q. Clinical trials with APT070 (Mirococept) and Compstatin family inhibitors show benefits with complement inhibition.
1. Biglarnia, AR., Huber-Lang, M., Mohlin, C. et al. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol 14, 767–781 (2018)
Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019;10:2380
Complement activation have three pathways which differ according their recognition targets
antibodies are triggers of the classical pathway , carbohydrates for the lectin pathway and permanent low level of activation for the alternative pathway
role of complement in transplantation:
Ischaemia -reperfusion injury with activation of lectin pathway due to endothelial injury and amplification of alternative pathway.
Antibody mediated rejection with activation of the classical pathway against antigens of MHC and ABO group and donor specific antigens.
Recurrence of native kidney disease through activation of alternative pathway :
recurrence of haemolytic uraemic disease, C3 glomerulopathy.
strategies attempted at inactivation of the complement pathway Currently, there are three complement inhibitors: a monoclonal antibody directed against C5 (eculizumab) and two plasma-derived C1 inhibitors.
Compounds blocking the terminal pathway:
Eculizumab a monoclonal antibody directed against C5: it inhibits C5 cleavage by the
C5 convertase with stopping of synthesis of C5a and C5b-9 which are the pro-inflammatory, prothrombotic and lytic functions of complement
this drug has shown efficacy in treatment and prevention of atypical haemolytic uraemic syndrome and antiphospholipid syndrome, some efficacy in the prevention of antibody-mediated rejection and so far no efficacy in the prevention of delayed graft function
in issue of preventing of delayed graft function : study done to compare patients used two doses of eculizumab and another group on placebo and there is no significant difference between two groups in prevention of delayed graft function.
in issue of prevention of antibody mediated rejection at 1 year, the incidence of acute AMR was 7.7% in the eculizumab group compared to 41.2% in the historical control group Graft and patient survival were excellent in both groups.
Compounds blocking the classical activation pathway
C1 inhibition reduces the release of chemotactic microvesicles, which are increasingly
recognized in the settings of transplantation . studies demonstrated the efficacy of blocking C1 with a recombinant C1 inhibitor used to prevent acute ABMR
study done to compare patients receive either placebo or plasma-derived human C1 inhibitor (Berinert®, 20 IU/kg/dose) twice weekly for a total seven doses.No patient developed rejection during the study in the C1-inhibitor group, but one developed rejection in the control group.Tolerance of the drug was acceptable.
Another study used another plasma-derived C1 inhibitor to treat acute
antibody-mediated rejection following kidney transplantation in a randomized double-blind placebo controlled pilot study. The treatment was well tolerated. With regard to efficacy, no patient developed transplant glomerulopathy in the treated group compared with 42% in the control group.
Finally, plasma-derived C1 inhibitor (Berinert®) was used in a pilot study in patients with acute ABMR who were resistant to the standard-of-care treatment. there was a trend towards better renal function at the end of the follow-up period.
References
Grafals M, Thurman JM. The Role of Complement in Organ Transplantation. Front Immunol. 2019;10:2380.
Legendre C, Sberro-Soussan R, Zuber J, Frémeaux-Bacchi V. The role of complement inhibition in kidney transplantation. Br Med Bull. 2017 Dec 1;124(1):5-17.
Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients Transplantation 2016;99:299–308
Montgomery RA, Orandi BJ, Racusen L, et al. Plasma derived C1 esterase inhibitor for acute antibody mediated rejection following kidney transplantation: results of a randomized double-blind placebo controlled pilot study. Am J Transplant 2016;16:3468–78.
Viglietti D, Gosset C , et al. C1 inhibitor in acute antibody-mediated rejection non responsive to conventional therapy in kidney transplant recipients:a pilot study. Am J Transplant 2016;16:1596–603
Role of Complement in Graft Damage:
\
Complement system is part of innate immunity which is activated by three pathways
Classic(antigen antibody complex)lactin pathway(microorganism)and alternative pathway
All the 3 pathways end by common pathway including c3 , c4 and c5 cleavage and formation of MAC complex and cell lysis
The role of complement in graft failure is appear in 3 main situations
1)in humoral rejection…plasma cells form antibodies which attach to antigen and start the classic pathway giving its fingerprint over the graft by c4d deposition in peritubular capillaries
2)in TMA post transplant which need to be differentiate what is its cause as it may be recurrence of 1ry kidney disease,CNI induced or ABMR
3)in Tcell mediated..some studies showed that APC produce c3a and c5a in the activation of cellular immunity
Drugs which interfere with complement system
IVIG…which bind to fc region on IgG and inhibit complement activation and widely used in ABMR
eculizumab…c5 inhibitor because of its very expensive drug few studies were addressed and show no benefits in preventing rejection
And finally c1estrase inhibitor which inhibits both lactin and classic pathways
Reference
(1) Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
(2) Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naïve CD4+ T Cells. Immunity 2008, 28, 425–435.
Complement in kidney transplantation:
The role of complement in various aspects of kidney transplantation has been outlined,
including the following conditions (1, 3) :
Ischemia-reperfusion injury,
Antibody-mediated rejection,
Recurrence of native kidney disease such as atypical haemolytic uremic syndrome,
C3 glomerulopathy,
Anti-phospholipid syndrome.
ABO-incompatible transplantation.
Complement inhibition after kidney transplantation:
By blocking:
(a) The activation pathways.
(b) The amplification loop.
(c) The anaphylatoxins .
(d) The terminal pathway.
Currently, there are three available complement inhibitors: a monoclonal antibody
directed against C5 (Eculizumab) and two plasma-derived C1 inhibitors.
Eculizumab is a recombinant fully humanized hybrid IgG2/IgG4 monoclonal antibody that
is directed against the human complement component C5.
Eculizumab has been used mainly to prevent delayed graft function, in acute antibody-
mediated rejection and for treatment/prevention of aHUS recurrence and
antiphospholipid syndrome.(3).
a multi-center trial that began enrollment in 2013 evaluated the role of Eculizumab in the
treatment of biopsy-proven AMR and acute graft dysfunction, study drug’s failure to
improve allograft function assessed by estimated glomerular filtration rate (eGFR) 3
months after transplantation.(4).
Role of C1-INH in the treatment of AMR was explored by two recent studies, Viglietti et
al. treated six patients who had AMR that was not responsive to SOC therapy with C1
-INH ,At the end of the 6-month follow-up period, the authors reported significant
improvement in mean eGFR. Fewer patients demonstrated C4d deposition on allograft
biopsies and circulating C1q fixing DSAs. However, other histologic features, like
chronic glomerulopathy, were unchanged (5).
Reference:
1- Ch. Legendre, R. Sberro-Soussan et al. The role of complement inhibition in kidney
transplantation, British Medical Bulletin, 2017, 124:5–17.
2-Małgorzata Kielar , Agnieszka Gala-Bł ˛adzi ´nska ET AL .Complement Components in
the Diagnosis and Treatment after Kidney Transplantation—Is There a Missing Link?
Biomolecules 2021, 11, 773.
4- Vasishta S. Tatapudi and Robert A. Montgomery..Therapeutic Modulation of the
Complement System in Kidney Transplantation: Clinical Indications and Emerging Drug
Leads. Front Immunol. 2019; 10: 2306.
5-Vasishta S. Tatapudi and Robert A. Montgomery..Therapeutic Modulation of the
Complement System in Kidney Transplantation: Clinical Indications and Emerging Drug
Leads. Front Immunol. 2019; 10: 2306.
The role of complement in graft damage:
Ischemia reperfusion injury:
the 3 complement pathway activation during cold ischemia affect the function of graft after transplantation (1)
Humoral rejection of kidney allograft:
Immune complex binds C1q complement activating the classical complement pathway (2) which lead to inflammation and thrombosis of graft microcirculation causing ischemia, apoptosis, necrosis and graft failure. (3)
T cell mediated rejection:
Recent studies showed that local production of C3a & C5a by APCs is necessary for T cell costimulation, (4)
Post transplant TMA:
It may be due to complement activation by concomitant autoimmune disease, malignant HTN, infection & AMR (5)
Therapeutics targeting complement:
Human intravenous immunoglobulins (IV IG)
used in treatment of AMR, they inhibit complement activation by blocking the receptor for the Fc fragment of IG or the C1q component. (6)
Eculizumab:
Inhibitor of C5 complement component, monoclonal antibody,
studies showed the efficacy of eculizumab to decrease incidence of early AMR in sensitized recipients (7)
An extended follow up study showed that eculizumab treatment has no effect on chronic AMR in recipients with significant amount of DSA (8)
C1 esterase inhibitor
inhibits the activation of both the classical and lectin complement pathways
used in treatment of recurrent TMA post transplant (9)
A study showed that C1 esterase inhibitor resulted in reduction of C1q antibodies and reduced occurrence of humoral rejection. (10)
The results were encouraging with no serious toxicity
(1) Damman, J.; Nijboer, W.N.; Schuurs, T.A.; Leuvenink, H.G.; Morariu, A.M.; Tullius, S.G.; Van Goor, H.; Ploeg, R.J.; Seelen, M.A. Local renal complement C3 induction by donor brain death is associated with reduced renal allograft function after transplantation. Nephrol. Dial. Transplant. 2011, 26, 2345–2354
(2) Haas, M.; Loupy, A.; Lefaucheur, C.; Roufosse, C.; Glotz, D.; Seron, D.; Nankivell, B.J.; Halloran, P.F.; Colvin, R.B.; Akalin, E.; et al. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell–mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am. J. Transplant. 2018, 18, 293–307
(3) Loupy, A.; Lefaucheur, C.; Vernerey, D.; Prugger, C.; Van Huyen, J.P.D.; Mooney, N.; Suberbielle, C.; Frémeaux-Bacchi, V.; Méjean, A.; Desgrandchamps, F.; et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N. Engl. J. Med. 2013, 369, 1215–1226.
(4) Strainic, M.G.; Liu, J.; Huang, D.; An, F.; Lalli, P.N.; Muqim, N.; Shapiro, V.S.; Dubyak, G.R.R.; Heeger, P.S.; Medof, M.E. Locally Produced Complement Fragments C5a and C3a Provide Both Costimulatory and Survival Signals to Naive CD4+ T Cells. Immunity 2008, 28, 425–435.
(5) Benz, K.; Amann, K. Thrombotic microangiopathy: New insights. Curr. Opin. Nephrol. Hypertens. 2010, 19, 242–247.
(6) Schinstock, C.A.; Mannon, R.B.; Budde, K.; Chong, A.S.; Haas, M.; Knechtle, S.; Lefaucheur, C.; Montgomery, R.A.; Nickerson, P.; Tullius, S.G.; et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation. Transplantation 2020, 104, 911–922.
(7) Stegall, M.D.; Diwan, T.; Raghavaiah, S.; Cornell, L.D.; Burns, J.; Dean, P.G.; Cosio, F.G.; Gandhi, M.J.; Kremers, W.; Gloor, J.M. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am. J. Transplant. 2011, 11, 2405–2413.
(8) Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant (2015) 15(5):1293–302.
(9) Gonzalez Suarez, M.L.; Thongprayoon, C.; Mao, M.A.; Leeaphorn, N.; Bathini, T.; Cheungpasitporn, W. Outcomes of Kidney Transplant Patients with Atypical Hemolytic Uremic Syndrome Treated with Eculizumab: A Systematic Review and MetaAnalysis. J. Clin. Med. 2019, 8, 919.
(10) Vo, A.A.; Zeevi, A.; Choi, J.; Cisneros, K.; Toyoda, M.; Kahwaji, J.; Peng, A.; Villicana, R.; Puliyanda, D.; Reinsmoen, N.; et al. A Phase I/II Placebo-Controlled Trial of C1-Inhibitor for Prevention of Antibody-Mediated Rejection in HLA Sensitized Patients. Transplantation 2015, 99, 299–308.
Complement system is an important part in immune system integrity. It activated through 3 pathways, each pathway need different stimuli to be activated, but the product of C5 cleavage &formation of MAC are found in all 3 pathways. C1q bind to DSA associated with significant poor graft survival when compared to non-complement fixing DSA.
The complement had different roles in renal transplantation including ischemic-perfusion injury, AMR, Recurrence of original renal disease ( aHUS, C3 glomerulopathy, anti phpspholipid syndrome) & ABO incompatible transplant.
Complement inhibition cab be done by :
C5 complement inhibitor( anti C5 monoclonal Ab) may decrease the frequency of AMR in highly sensitized patients. Eculizumab originally licensed for treatment of PNH & aHUS. Stegall et al & Marks et al studies show a benefit of eculizumab in prevention of early AMR among sensitized recipients.
C1 estrase inhibitor (C1-INH) block classical pathway of complement activation by C1s &C1r inhibition. It licensed for treatment of angioedema type I & type II. C1-INH safety studied in highly sensitized renal recipient which show that there is decrease in C1q+ HLA Ab & decrease in occurance of AMR.
CR1; CD35 is a new inhibitor show promising therapeutic effects. Recompenent soluble CR1 ( APT070, mirocept) has been shown ti decrease ischemic-perfusion injury & improve graft survival.
some DSA can cause graft loss without activation of complement & this damage occurs as a result to antibody dependent cellular cytotoxicity.
References:
Complement activation is one of the important part of innate immune system and it’s play an important role in inflammatory processe it’s activation occurs by 3 pathways classical,alternate or lectin all lead to anaphylatoxin c3a and c5a formation leading to direct cell lysis.
Complement cascad activation plays a role in renal transplantation also during ischemia / perfusion process that affect on early graft function.
It has major role in both acute cellular and antibody mediated rejection also in case of thrombotic microangiopathy ,also it has role in many kidney diseases like SLE ,a typical HUS ,IgA nephropathy and even in renal complications related to DM .
Eculizumab is a humanized monoclonal antibody cause blocking of c5 cleavage thus preventing the release of anaphylatoxin which participate in all 3 major pathways although alot of follow up studies showed that it’s usage is ineffective in chronic antibody mediated rejection in case of high DSA in recipient also in c4d negative ABMR.
The complement system is considered to be an important part of innate immune system with a significant role in inflammation processes. The activation can occur through classical, alternative, or lectin pathway, resulting in the creation of anaphylatoxins C3a and C5a, possessing a vast spectrum of immune functions, and the assembly of terminal complement cascade, capable of direct cell lysis. The activation processes are tightly regulated; inappropriate activation of the complement cascade plays a significant role in many renal diseases including organ transplantation. Moreover, complement cascade is activated during ischemia/reperfusion injury processes and influences delayed graft function of kidney allografts. Interestingly, complement system has been found to play a role in both acute cellular and antibody-mediated rejections and thrombotic microangiopathy. Therefore, complement system may represent an interesting therapeutical target in kidney transplant pathologies
Immune system consists of 2 integrated arms
The adaptive and the innate immune systems
Adaptive with consist mainly of B and T cell responses, which contains specific receptors , capable of clonal expansion , able to form memory cells and provide long term immunity
Innate immune system comprised of dendrites ,monocytes, macrophages
They have no specific receptors , can’t clonally expand , and don’t form memory cells ,
Their main function is antigen recognition and presentation ( through APCs ) to lymphoid organs to initiate adaptive immune response
Beside cellular component of innate system, there is non cellular components , which are the complement system , which plays a role in antigens recognition .
Studies have shown that complement activation contributes to allograft injury in several clinical settings, including ischemia/reperfusion injury and antibody mediated rejection.
Besides , the complement system plays critical roles in modulating the responses of T cells and B cells to antigens.
Once complement is activated , various active complement fragments are generated which can directly and indirectly affect the graft
Directly , as these active complement fragments act as chemotaxins
Indirectly as they provide an important signal for B and T cell stimulation
Site of complement activation differs according to the situation, in case of renal ischaemia, activation affect tubulointerstitium ,
In ABMR , activation occur in peritubular capillaries
**In ischemia reperfusion injury
Complement activation occur through alternative pathway .
Complement inhibitory drugs have proven effective in several pre-clinical models of I/R injury.
In spite of promising pre-clinical data, a atudy that included 27 kidney transplant patients at high risk of DGF ,were treated with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benifical effect 2 .
Another clinical trial is ongoing, however, in which kidneys treated with an agent that attaches a complement regulator to cell membranes 3 .
This approach was previously shown to be beneficial in a rat kidney transplant model 4.
**ABMR :
In cases of acute and chronic ABMR , DSAs bind to to Donor HLA which is expressed on surface of endothelial cells activating the complement system .
This activation causes damage to the capillaries
Diagnosis of ABMR depend on detection of circulating DdSAs , micro vascular inflammation in the biopsy , and detection of complement fragments C4d in PTCs
However , cases of C4d negative ABMR have been reported , and this may reflect either non complement mediated DSA effect , or inability to detect C4d
Because complement activation is an important step in ABMR , assays have been developed to detect ability of DSAs to activate the complement .
Complement inhibitory drugs have shown benifical effects in animal models , they induce a state of accommodation which occurs due to upregulation of regulatory complement proteins 5
Eculizumab has been used in transplant recipients at high risk of developing AMR, as well as patients with active disease refractory to treatment with Positive results have been reported in lung and kidney transplant recipients 6
Larger series in transplant patients have not shown a consistent benefit.
Concluding that the role of eculizumab for preventing or treatment AMR is not yet clear
**References:
1. Janeway CA Jr. How the immune system works to protect the host from infection: a personal view. Proc Natl Acad Sci USA. (2001) 98:7461–8. doi: 10.1073/pnas.131202998
2) Heeger P, Akalin E, Baweja M, Bloom R, Florman S, Haydel B, et al. Lack of efficacy of eculizumab for prevention of delayed graft function (DGF) in deceased donor kidney transplant recipients. Am J Transplant. (2017) 17(Suppl 3):B131.
3). Kassimatis T, Qasem A, Douiri A, Ryan EG, Rebollo-Mesa I, Nichols LL, et al. A double-blind randomised controlled investigation into the efficacy of Mirococept (APT070) for preventing ischaemia reperfusion injury in the kidney allograft (EMPIRIKAL): study protocol for a randomised controlled trial. Trials. (2017) 18:255. doi: 10.1186/s13063-017-1972-x
4). Pratt JR, Jones ME, Dong J, Zhou W, Chowdhury P, Smith RA, et al. Nontransgenic hyperexpression of a complement regulator in donor kidney modulates transplant
5) Dorling A. Transplant accommodation–are the lessons learned from xenotransplantation pertinent for clinical allotransplantation? Am J Transplant. (2012) 12:545–53. doi: 10.1111/j.1600-6143.2011.03821.x
6. Locke JE, Magro CM, Singer AL, Segev DL, Haas M, Hillel AT, et al. The use of antibody to complement protein C5 for salvage treatment of severe antibody-mediated rejection. Am J Transplant. (2009) 9:231–5. doi: 10.1111/j.1600-6143.2008.02451.x
7. Dawson KL, Parulekar A, Seethamraju H. Treatment of hyperacute antibody-mediated lung allograft rejection with eculizumab. J Heart Lung Transplant. (2012) 31:1325–6. doi: 10.1016/j.healun.2012.09.016
During complement activation, complement protein fragments are released into the plasma, and C3 and C4 fragments are covalently fixed to target tissues. Native kidney biopsies are routinely stained for C3 deposits, and in some centers they are also stained for C4 fragments. Because C4 is covalently attached to target tissues, C4 deposits provide a durable marker of CP activation.
Allograft biopsies are now routinely stained for C4d, and detection of C4d in the peritubular capillaries is interpreted as a marker of classical pathway activation in patients with AMR. CP activation on the capillary would also be expected to result in C3 fragment deposition, although C3d deposition seems to be a less sensitive indicator of AMR. It is possible that C3d deposition signifies more complete activation of the complement cascade, and one study found that deposition of C3d on the peritubular capillaries was associated with a worse prognosis
Soluble complement fragments can be measured in body fluids by enzyme linked immunosorbent assays (ELISAs). The half-life of these fragments is short, so elevated levels of complement fragments indicates that there is ongoing activation. There are assays that can measure many different complement fragments, including C4a, C3a, Ba, Bb, C5a, and soluble sC5b-9.
Other than staining allograft biopsies for C4d, complement biomarkers are not routinely analyzed in transplant recipients. As the use of complement inhibitory drugs expands, however, there will be an increasing need to develop accurate biomarkers.
Although eculizumab and C1-INH have shown promise in case reports and small series, their role in transplant medicine requires further study. Many additional anti-complement therapeutics are in clinical development, and some of these new drugs block individual activation pathways or specific components of the complement cascade.
Dear All
Please answer the question below. We need to know that complement activation is an important part of tissue injury, but graft damage can happen without complement activation.
Please give examples!!
One example of complement independent Antibody medicated allograft dysfunction is the C4d-negative ABMR. It was noted that some patients will have histologic evidence of ABMR (microcirculatory inflammation) and positive donor-specific antibodies (DSA) but with little or no C4d staining in the PTCs, a situation that was named as C4d-negative ABMR (1).
The clinical implication of this subtype of ABMR is that the early initiation of treatment for ABMR even in absence of C4d staining can result in a better long term graft outcome (2). if untreated, C4d-negative ABMR may lead to the development of scarring within the graft, transplant glomerulopathy and even graft loss (1).
One study involved 98 renal allograft biopsies performed for clinical indications within the first three months post-transplant, 17 showed C4d-positive ABMR and 16 had histologic changes of ABMR and DSA, but no C4d could be detected in the PTCs (2). Patients with C4d-negative ABMR who received treatment for ABMR had a lower risk for progression to transplant glomerulopathy compared to those who were left untreated (2).
References:
1) Haas M. C4d-negative antibody-mediated rejection in renal allografts: evidence for its existence and effect on graft survival. Clin Nephrol. 2011;75(4):271.
2) Haas M, Mirocha J. Early ultrastructural changes in renal allografts: correlation with antibody-mediated rejection and transplant glomerulopathy. Am J Transplant. 2011; 11(10): 2123.
After alloantigen recognized by naive t cell and its transformation it effector tcell which lead to b cell activation and antibodies formation against donor HLA phenotyping
They cause allograft damage in two principal ways: they activate the classical pathway of the complement system and stimulate macrophages and other immune cells by binding to Fc receptors
(FcR) that recognize the constant regions of specific antibody classes
Reference
Handbook of kidney tranplantation by G.danovitch
DSA may also cause injury through complement-independent mechanisms, such as signaling through FcR receptors on natural killer (NK) cells or other cell types.
C4d-negative AMR was included in the most recent revision of the Banff criteria for the diagnosis of AMR .(Erik Stites.The Complement System and Antibody Mediated Transplant Rejection.)
C4d deposition in ABO-incompatible allografts can actually be associated with accommodation and improved outcomes. Thus, detection of C4d without histologic or functional evidence of allograft injury is not sufficient to predict AMR or to guide treatment. On the other side some clinically evident ABMR were found to be without C4d deposition(C4d negative ABMR) …. the question then will be is there any failure to detect C4d or it means inflammation without complement involvement .
Consistently, for several years, the C4d deposition has been considered the gold standard for ABMR diagnosis; however, today, it is recognized that up to 55% of patients can develop ABMR without detectable capillary C4d deposits. Indeed, a C4d-negative ABMR phenotype has been included in Banff 2013 classification (1).
C4d-negative ABMR was associated with glomerulopathy and graft loss.
This can occure directly by binding of Ab to specific FcR receptors on natural killer and macrophages or other cell types.
Biglarnia AR, Huber-Lang M, Mohlin C, Ekdahl KN, Nilsson B. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol. (2018) 14:767–81.
ABMR without C4d deposition can occur in many cases and Banff classification criteria included a category of C4d-negative ABMR. Several mechanisms have been proposed for complement-independent tissue injury in the presence of DSA. It is well known that activated monocytes and macrophages infiltrate into graft tissues. The inflammatory environment triggered by the binding of DSA to endothelial cells alone can induce an allo-reaction of CD4 T-cells via graft endothelial cell HLA-class II.
Kenta I, Takaaki K. Molecular Mechanisms of Antibody-Mediated Rejection and Accommodation in Organ Transplantation. Nephron. 2020;144(1):2-6.
Complement activation is important but antigens recognized by non-HLA antibodies have been reported. Examples such as endothelial cell, angiotensin II type 1 receptor antibodies (AT1R-Ab), endothelin-1 type A receptor, collagen type IV, fibronectin, perlecan, vimentin, agrin, tubulin, laminin, myosin and major histocompatibility complex class I chain-related gene A and B has been reported but AT1R-Ab has gain popularity among those non HLA antibodies.
AT1R-Ab recognised to have mediate graft injury in a complement independent and antibody dependent cell mediated manner. Pre- and post-KT AT1R-Ab were associated with AMR development. studies have shown that pre and post KT AT1R-Ab level more than > 17 U/mL were a risk factor for AMR development.
References
Sorohan, B., Ismail, G., Leca, N., Tacu, D., Obrișcă, B., Constantinescu, I., Baston, C. and Sinescu, I., 2020. Angiotensin II type 1 receptor antibodies in kidney transplantation: An evidence-based comprehensive review. Transplantation Reviews, 34(4), p.100573.
complement as part of the innate immune system plays role in each stage causing damage to the transplanted kidney. apart from details of this fantastic and complex system, we see damage differently in each stage. for example at early stages and in case of reperfusion ischemia damage is mainly in tubulointerstitial space while in AMR (antibody-mediated rejection) we see capillaritis (peritubular capillaries).
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6788431/
Both c3 and c4 play a role but c4 is stuck to tissue and it became a routine stain in many centers to show AMR
data about eculizumab are conflicting some were compared to historical transplants.
regarding c1 esterase inhibitor some promising results were found (Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney Transplantation: Results of a Randomized Double-Blind Placebo-Controlled Pilot Study. (https://pubmed.ncbi.nlm.nih.gov/27184779/)
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6779821/
Therapeutic Modulation of the Complement System in Kidney Transplantation: Clinical Indications and Emerging Drug Leads
Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement?
Give examples
Yes.
Anti-HLA-class I antibodies can promote focal adhesion kinase, matrix metalloproteinase-2, and neutral sphingomyelinase-2-dependent proliferation and migration of smooth muscle cells producing changes in the intima leading to chronic rejection. (1)
Reference:
1) Valenzuela NM, McNamara JT, Reed EF. Antibody-mediated graft injury: complement-dependent and complement-independent mechanisms. Curr Opin Organ Transplant 2014;19:33-40.
Can graft injury happen without activation of complement?
Give examples
Yes rejection can occur in absence of complement activation as in C4d-negative antibody-mediated rejection with high Anti-angiotensin II type 1-receptor-activating antibodies (anti-AT1R) have been mentioned to stimulate a severe vascular rejection described as IIB or AMR in Banff classification.
American journal of transplantation; Banff 2013 Meeting Report: Inclusion of C4d-Negative Antibody-Mediated Rejection and Antibody-Associated Arterial Lesions,
Haas.M et al, 28 January 2014
Well done
Following transplantation, complement has a role in innate immunological and inflammatory processes that further damage the graft and result in a gradual decrease in its functional mass. Additionally, it modulates the adaptive immune response, mediates many of the downstream effects of B and T cell immunity, and can function independently of the adaptive immune response
Activation of the complement system in the donor, the graft and the recipient before, during and after transplantation is a major cause of damage to the kidney transplant through different ways like antibody-mediated rejection, recurrence of native kidney disease such as atypical haemolytic uraemic syndrome, C3 glomerulopathy, anti-phospholipid syndrome and the accommodation phenomenon in ABO-incompatible transplantation.
The complement system is activated through three distinct pathways: classical pathway Lectin pathway, Alternative pathway
Ischaemia and subsequent reperfusion of the graft is the most important mechanism that triggers complement activation
Complement-targeted strategies might have a role in optimizing graft quality as well as in the treatment of antibody-mediated rejection, induction of accommodation and modulation of the adaptive immune response.
Promising data from preclinical and clinical studies suggest that complement targeted therapies could potentially become part of the standard of care for kidney transplantation.
Complement-targeted agents for the prevention of rejection in transplantation include:
A) Standard of care therapies:
Intravenous immunoglobulin :Inhibits activation of complement, blocks FcR and C1q 2
Rituximab Humanized anti-CD20 expressed on B cells. It Blocks complement-induced enhancement of antibody generation
B) complementary therapies: initial results from early clinical trials are promising but there is still inconsistencies on the effect of complement inhibitors on delayed graft function and ABMR which clearly reflect the complexity of allograft injury and the involvement of complement-independent mechanisms
A number of therapies are being developed but only two categories of complement-inhibiting drugs are in clinical use currently; the humanized anti-C5 mAb eculizumab and preparations of C1INH
Eculizumab: Humanized mAb C5a Inhibits C5 cleavage to form C5a and C5b; blocks the terminal pathway
Benefits: Terminal Complement blockage at the level of C5 preserves the early components of complement which are essential for opsonization of microorganisms and clearance of immune complexes
Risks: An acquired functional C5 deficiency, and in patients with genetically determined C5 deficiency, increase the risk of Neisseria meningitis so it is mandatory to give prophylactic vaccination and antibiotics
Trials and outcome:
In delayed graft function: a randomized, parallel-group, double-blind, placebo-controlled, multicentre study was performed with two doses of Eculizumab administered in an acute setting for the prevention of DGF in adult recipients of deceased-donor kidney transplants who were at increased risk of DGF. 288 pts recruited in North America, South America, Europe and Australia. Unfortunately, DGF rate was not significantly different when comparing Eculizumab to placebo
In preventing antibody-mediated rejection: a single-centre open-label study by Stegall et al. included 26 sensitized patients with preformed HLA-DSAs undergoing living donor renal transplantation showed a reduction in ABMR at 3 months in comparison to control group who received post-transplantation plasmapheresis only (7.7% versus 41.2%),but a 2 years follow-up showed no difference in the histopathological occurrence of ABMR and graft survival between both groups.
On The other hand, A prospective multi-centre study compared 9 weeks of prophylactic eculizumab treatment to plasmapheresis and IVIG .Eculizumab showed a benefit to lower ABMR incidence early and after 3 months only in the patients with C1q-binding HLA-DSAs.
Thus it appears that only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti-complement treatment or that Complement inhibition at C5 did not prevent upstream complement activity due to the generation of immune modulators such as C4a or C3a.
In kidney transplantation to treat or prevent recurrences of atypical HUS: The efficacy of Eculizumab efficacy was found in 4 prospective, multi-centre, single-arm, non-randomized studies involving 100 patients followed at least 2 years. Additionally Zuber et al gathered 21 patients from both an extensive literature search and a French national survey and showed that Eculizumab was efficient for treating and preventing aHUS recurrence after kidney transplantation
Serine protease inhibitors: Purified or recombinant proteins that inactivates complement serine proteases, blocks the classical and lectin pathways
Benefits:
Theoretically elicit a more potent effect than Eculizumab on both T-cell and B-cell components of the allo-immune response. Blocking the generation of complement activation products of C3 that contribute to neutrophil and monocyte recruitment and tissue injury in AMR.
Risks:
The lack of specificity of serine protease inhibitors implies a broad inhibitory effect on both the classical and lectin pathways that may dramatically reduce C3-dependent opsonization of pathogens and increase susceptibility to infections.
Trials and outcome:
In a pilot trial, C1INH was given as a supplement to high-dose IVIg in 6 patients with nonresponsive ABMR. At 6 months, these patients showed improvements in eGFR compared with levels at enrolment and had less C1q-binding DSAs than controls.
In a randomized, placebo-controlled trial that compared add-on treatment with C1INH to placebo in 18 patients with ABMR who were receiving plasmapheresis, IVIg and rituximab, There was no difference in ABMR histopathology or kidney function in both groups at at 20 days However, a subgroup analysis of 14 patients with 6-month protocol biopsy samples identified no transplant glomerulopathy in the C1INH group, whereas transplant glomerulopathy was present in 43% of patients in the placebo group.
Accordingly, Future well designed clinical trials with long surveillance are warranted to further evaluate complement interventions in transplantation
The multifaceted role of complement in kidney transplantation.
Ali-Reza Biglarnia, Markus Huber-Lang, Camilla Mohlin, Kristina N. Ekdahl,and Bo Nilsson
Nature Reviews | Nephrology ,volume 14 .DECEMBER 2018
The role of complement inhibition in kidney transplantation
C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi
British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17
Complement system activation has an important role in renal graft injury, especially in antibody mediated rejection and ischemia/reperfusion injury. Complement activation through classical pathway, lectin pathway and alternative pathway leads to formation of C3b and C5b which causes damage/ lysis to the target cell. (1)
Hence complement inhibitors can be utilized to prevent complement mediated injury in transplant recipients. These include:
(1) C5 inhibitor Eculizumab: It binds to C5 and inhibits C5a and subsequent membrane attack complex formation
It has been used in kidney transplant patients for:
a) Prevention of delayed graft function (DGF): PROTECT study by Alexion was done at multiple centers, but 2 doses of eculizumab failed to show any significant benefit in prevention of DGF. (2)
b) Preventing AMR: Few studies demonstrated that Eculizumab use in desensitized highly sensitized patients reduced incidence of acute AMR at 1 year. (3,4). Another study did not find any difference. (5) Subclinical AMR lesions were seen in patients with persistently high DSA levels.
c) For treatment/prevention of atypical HUS recurrence: Multiple studies have shown that eculizumab causes significant improvement in graft function of patients with recurrence of atypical HUS. (6,7)
d) For prevention/ treatment of C3 glomerulopathy and antiphospholipid syndrome: Some studies have shown promising results. (8,9)
(2) C1 esterase inhibitor: Blocks classical pathway. It has been used to prevent AMR in transplant recipients. A study showed HLA sensitized patients receiving C1-inhibitor did not develop AMR. (10) Another study used C1-inhibitor in treatment of acute AMR and showed that there was no transplant glomerulopathy at 6 months post transplant (versus 42% in the placebo group). (11)
(3) Ides: A recombinant Streptococcus pyogenes- derived endopeptidase which cleaves IgG has been shown to prevent AMR in highly sensitized transplant recipients. (12)
References:
1) Grafals M, Thurman JM. The role of complement in organ transplantation. Front Immunol 2019;10:2380
2) Legendre Ch, Sberro-Soussan R, Zuber J, et al. The role of complement inhibition in kidney transplantation. British Medical Bulletin 2017;124:5-17.
3) Stegall MD, Diwan T, Raghavaiah S, et al. Terminal complement inhibition decreaases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant 2011;11:2405-2413.
4) Glotz D, Legendre C, Lefaucheur C, et al. Eculizumab in prevention of acute antibody mediated rejection in sensitized deceased donor kidney transplant recipients: 1 year outcomes. Transplant Int 2015;28:1-19.
5) Cornell LD, Schinstock CA, Gandhi MJ, et al. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant 2015;15:1293-1302.
6) Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic uremic syndrome. N Eng J Med 2013;368:2169-2181.
7) Zuber J, Le Quintrec M, Krid S, et al. Eculizumab for atypical hemolytic uremic syndrome recurrence in renal transplantation. Am J Transplant 2012;12:3337-3354.
8) Le Quintrec M, Lionet A, Kandel C, et al. Eculizumab for treatment of rapidly progressive C3 glomerulopathy. Am J kidney Dis 2015;65:484-489.
9) Canaud G, Kumar N, Anglicheay D, et al. Eculizumab improves posttransplant thrombotic microangiopathy due to antiphospholipid syndrome recurrence but fails to prevent chronic vascular changes. Am J Transplant 2013;13:2179-2185.
10) Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo controlled trial of C1-inhibitor for prevention of antibody mediated rejection in HLA sensitized patients. Transplantation 2016;99:299-308.
11) Montgomery RA, Orandi BJ, Racusen L, et al. Plasma derived C1 esterase inhibitor for acute antibody mediated rejection following kidney transplantation: results of a randomized double-blind placebo controlled pilot study. Am J Transplant 2016;16:3468-3478.
12) Jordan SC, Lorant T, Choi J. IgG endopeptidase in highly sensitized patients undergoing transplant. N Eng J Med 107;377-1693-1694.
Inflammatory effect of complemt on allograft
-C3a
C3a/C3aR signaling participates in glomerular and tubular injury , also stimulates epithelial cells to produce chemokines which causes tissue inflammation .There are not currently any specific antagonists of C3a available .(1)
–C5a
C5a has pro-inflammatory effects and is a potent myeloid cell chemoattractant.
C5aR deficiency or blockade is protective against of Ischemia/Reprfusion injury , tubulointerstitial injury, and anti-neutrophil cytoplasmic antibody (ANCA) vasculitis.
C5aR antagonists have been developed (2).
– Membrane Attack Complex
MAC formation on endothelial cells leads to NF- κb activation within the cells leading to production of IL-1α and IL-8 .
A trial that enrolled 27 kidney transplant patients at high risk of DGF treated with an inhibitory monoclonal antibody to C5 (eculizumab) did not show any benefit with treatment .Another trial ongoing in which kidneys treated with an agent which attaches a complement regulator to cell membranes ,it was previously shown to be beneficial in a rat kidney transplant model (3)
Eculizumab has been used in transplant patients at high risk of developing AMR, and patients with active disease refractory to treatment. Positive results have been reported in lung and kidney transplant recipients with AMR . Larger studies in transplant patients have not shown a long term benefit, meanwhile eculizumab rolein preventing AMR is not clear.(4)
There is pre-clinical data showing that C5aR1 blockade may be a beneficial treatment for rejection.
Complement inhibitory drugs can have a role in xenotransplanation, but they probably will need to be administered for long time due to the persistence of pathogenic natural antibodies in spite of immunosuppression.
C1-INH has been used in prevention of AMR, other complement inhibitory agents are useful in the transplant setting, as an AP inhibitor , a LP inhibitor , and C5a blockade (2).
The diagnosis of AMR is based on detecting DSA in the plasma, microvascular inflammation on a biopsy , and C4d deposition in the peritubular capillaries ,this criteria have been changed to account for C4d-negative cases . It is un known whether those negative case caused by non-complement-mediated injurious effects of the DSA, or it reflects variability in detection of C4d.(2)
1- Peng Q, Li K, Smyth LA, Xing G, Wang N, Meader L, et al. . C3a and C5a promote renal ischemia-reperfusion injury. J Am Soc Nephrol. (2012) 23:1474–85
2- Grafals M et al, The role of complement in organ transplantation. Frontiers in Immunology 2019; 10: 2380.
3-Jane-Wit D, Manes TD, Yi T, Qin L, Clark P, Kirkiles-Smith NC, et al. . Alloantibody and complement promote T cell-mediated cardiac allograft vasculopathy through noncanonical nuclear factor-kappaB signaling in endothelial cells. Circulation. (2013) 128:2504–16.
3- Heeger P, Akalin E, Baweja M, Bloom R, Florman S, Haydel B, et al. Lack of efficacy of eculizumab for prevention of delayed graft function (DGF) in deceased donor kidney transplant recipients. Am J Transplant. (2017) 17(Suppl 3):B131
4- Marks WH, Mamode N, Montgomery R, Stegall MD, Ratner LE, Cornell LD, et al. . Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: a randomized trial. Am J Transplant. (2019) 19:2876–88.
Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement?
Give examples
antibody against G protein coupled receptor and angiotensin receptors type I and II were found in the patient with severe rejection with vascular affection without c4d deposition ( without complement fixation).
Aurelie Philippe. Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation. Kidney International Reports. 2019 vol (4) issue 4, P 510-12.
Since the activation of complement in the setting of high levels of DSA plays a major role in the pathogenesis of acute AMR, it makes sense that complement blockade would be an important strategy for prevention and treatment of AMR. Agents targeting C5 and C1 esterase have been evaluated in clinical trials.
C5 inhibition [Eculizumab]
*In a single-arm phase I/II trial conducted at the Mayo Clinic that included 26 highly sensitized recipients of living-donor renal transplants (NCT006707), eculizumab in combination with plasmapheresis resulted in a statistically significant reduction in AMR in the first 3 months compared with 51 historic controls receiving plasmapheresis alone.
*In a phase II randomized, open-label, multicenter trial of eculizumab vs placebo as an add-on to SOC desensitization protocols in living kidney donor recipients (NCT01399593), the eculizumab group failed to achieve a statistically significant reduction in the primary composite endpoint (occurrence of biopsy-proven AMR, graft loss, patient death, or loss to follow-up at the end of the 9-week treatment period posttransplant) when compared with placebo
*In a randomized, open-label, multicenter phase II study in sensitized deceased donor kidney transplant recipients (NCT01567085), 1-year data show that eculizumab was effective in reducing the incidence of acute AMR.
*Eculizumab failures have been reported in cases of C4d-negative AMR.
C1 esterase inhibition
Two C1-INH products that are approved for use by the FDA in the treatment of hereditary angioedema have been evaluated in small pilot studies for AMR: Berinert® (CSL Behring, Kankakee, IL, USA) and Cinryze® (Shire ViroPharma Inc., Lexington, MA, USA)
*A recent double-blind, placebo-controlled, randomized phase I/II trial (NCT01134510) evaluated C1-INH (Berinert) in sensitized renal transplant patients for the prevention of acute AMR. Twenty percent of C1-INH patients developed AMR outside of the study period, whereas 30% of placebo patients developed AMR, 10% during the study period. Delayed graft function developed in 1 patient receiving C1-INH and 4 patients receiving placebo. C1-INH treatment also reduced C1q-positive HLA antibodies in all 5 patients with these antibodies at time of transplantation.
*In a single-arm pilot study, the investigators found that CI-INH (Berinert) in combination with high-dose IVIg improved allograft function in kidney recipients with acute AMR that was nonresponsive to conventional therapy
*A randomized, double-blind, placebo-controlled, multicenter phase IIb study (NCT01147302) evaluated the safety and effect of C1-INH (Cinryze) for the treatment of acute AMR in 18 recipients of donor-sensitized kidney transplants.
Histopathological resolution of AMR was seen in 7 out of 7 C1-INH patients analyzed and 4 out of 6 placebo patients analyzed.
Surveillance 6-month biopsies performed in 14 patients showed no transplant glomerulopathy (ptc + cg ≥ 1b) in the 7 patients in the C1-INH group, whereas 3 out of 7 patients in the placebo group showed transplant glomerulopathy.
In addition, transplant glomerulopathy tended to develop in those patients in the placebo group who had very low levels of C1-INH.
IgG-degrading enzyme of Streptococcus pyogenes (IdeS)
Phase I/II testing (NCT02790437) has begun for a new compound called IgG-degrading enzyme of Streptococcus pyogenes (IdeS). This enzyme cleaves at a very specific amino acid sequence in the hinge region of human IgG and essentially neutralizes all of the IgG in the body within 4 hours of administration.
Reference
Montgomery, RA, Loupy, A, Segev, DL. Antibody-mediated rejection: New approaches in prevention and management. Am J Transplant. 2018; 18(Suppl. 3): 3– 17. https://doi.org/10.1111/ajt.14584
The complement-targeted agents for the prevention of rejection in transplantation.
-The terminal complement inhibitor, Eculizumab, and C1 esterase inhibitor (C1-INH) have been the subjects of recent clinical trials exploring the potential of targeting the complement cascade in the prevention and treatment of AMR.
-Eculizumab (Soliris, Alexion) is a recombinant humanized anti-C5 monoclonal antibody that prevents the cleavage of C5 by C5 convertase. In a phase 3 clinical trial published in 2006, eculizumab stabilized hemoglobin levels in transfusion-dependent patients with PNH. Following this trial, the FDA approved the use of eculizumab for PNH in March 2007.
-C1-INH is a serine protease inhibitor (SPI) that removes activated C1r and C1r from C1q. Two nano-filtered C1-INH products derived from human serum, cinryze (Takeda/Shire Pharmaceuticals) and berinert (CSL Behring), were approved for the treatment and prophylaxis of attacks of Hereditary angioedema (HAE) after randomized trials demonstrated their efficacy.
-Multi-center trial that began enrollment in 2013 evaluated the role of eculizumab in the treatment of biopsy-proven AMR and acute graft dysfunction.
Patients in the standard of care (SOC) arm received three sessions of plasmapheresis with IVIg administered after each session. Those in the treatment arm received a first dose of 1,200 mg of eculizumab followed by 4 weekly doses of 900 mg and a dose of 1,200 mg at week 5. Patients received additional doses of 1,200 mg at weeks 7 and 9 if DSA levels at week 6 remained >50% of baseline.
However, recruitment for this trial was terminated in 2017 due to the study drug’s failure to improve allograft function assessed by estimated glomerular filtration rate (eGFR) 3 months after transplantation [1].
-The role of C1-INH in the treatment of AMR was explored by two recent studies [2,3].
Viglietti et al. treated six patients who had AMR that was not responsive to SOC therapy with C1-INH (berinert, CSL Behring) and high-dose IVIg for a duration of 6 months [2]. At the end of the 6-month follow-up period, the authors reported significant improvement in mean eGFR. Fewer patients demonstrated C4d deposition on allograft biopsies and circulating C1q fixing DSAs. However, other histologic features, like chronic glomerulopathy, were unchanged [2].
Montgomery et al. conducted a randomized phase 2 trial to evaluate the role of a 2-week course of C1-INH (Cinryze, Shire Pharmaceuticals) as an add-on therapy to SOC plasmapheresis and low-dose IVIg, compared to SOC alone. Nine patients were randomized to each arm. While there was no significant difference in graft survival and histologic findings of AMR after 20 days, 6-month allograft biopsies showed an absence of transplant glomerulopathy in the C1-INH treated patients, whereas three out of seven patients in the placebo arm were found to have this histologic feature that is associated with chronic antibody mediated injury and premature allograft loss
-Reference
Tatapudi, Vasishta S, and Robert A Montgomery. “Therapeutic Modulation of the Complement System in Kidney Transplantation: Clinical Indications and Emerging Drug Leads.” Frontiers in immunology vol. 10 2306. 1 Oct. 2019, doi:10.3389/fimmu.2019.02306
[1] Randomized-Controlled Trial Examining the Safety and Efficacy of Eculizumab in the Treatment of AMR NCT01895127. Available online at: https://clinicaltrials.gov/ct2/show/record/NCT01895127?term=eculizumab+antibody+mediated+rejection&rank=3&view=record (accessed September 19, 2019)
[2] Viglietti D, Gosset C, Loupy A, Deville L, Verine J, Zeevi A, et al. . C1 Inhibitor in acute antibody-mediated rejection nonresponsive to conventional therapy in kidney transplant recipients: a pilot study. Am J Transpl. (2016) 16:1596–603. 10.1111/ajt.13663.
[3] Montgomery RA, Orandi BJ, Racusen L, Jackson AM, Garonzik-Wang JM, Shah T, et al. . Plasma-derived C1 esterase inhibitor for acute antibody-mediated rejection following kidney transplantation: results of a randomized double-blind placebo-controlled pilot study. Am J Transpl. (2016) 16:3468–78.
· complement system is a vital column of the innate immunity. It may lead to graft injury and affection by many ways including AMR , cellular rejection , and TMA.
· As soon as the graft is exposed to the recipient’s blood , the complement system may be activated and lead to active events.
Once the complement cascade is activated it can lead to direct , through cell lysis by the MAC system , and indirect effects.
The activation can occur through classical, alternative, or lectin pathway, resulting eventually in direct cell lysis.
· C3a and C5a provide important signals for T and B cell activation in the tubule-interstitium , whereas in Antibodies activation occurs in the peritubular capillaries.
The inhibition of some complement components has shown results in preventing AMR
· C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking C1 complex components. Still udner trials .
· Eculizumab is MAB capable of blocking the cleavage of C5 complement component, thus preventing the release of and the assembly of MAC. Eculizumab is an effective treatment for paroxysmal nocturnal hemoglobinuria and as well as recent uses in TTP .
· In one study reported , they study the use of eculizumab in highly sensitized patients with live donor transplantation . They found that the eculizumab treated patients had lower incidence of AMR at one year, but with high rates of subclinical rejection.
· Critics regarding eculizumab from effective to ineffect in patients with ABMR.
Biglarnia A.R. Huber-Lang M., Mohlin C., Ekdahl K.N., et al. The multifaceted role of complement in kidney transplantation. Nature Reviews Nephrology. 2018, Dec., Vol.14. 767-781.
Long-term outcomes of eculizumab-treated positive crossmatch recipients: Allograft survival, histologic findings, and natural history of the donor-specific antibodies
Carrie A. Schinstock et al
Positive Crossmatch Kidney Transplant Recipien Treated With Eculizumab: Outcomes Beyond 1 Year
L. D. Cornell et al
Can graft injury happen without activation of complement?
YES
-Even in absence of complement activation, some DSAs can cause graft damage through antibody-dependent cellular cytotoxicity.
The innate immune cells, including neutrophils, macrophages, and natural killer cells, can bind to Fc fragments of DSAs, trigger degranulation, and release lytic enzymes, which cause tissue injury and cell death.
This process can mediate smoldering damages to the endothelial cells and is proposed as an important pathogenesis in subclinical and chronic antibody-mediated rejection phenotypes.
-Furthermore, DSAs can cause graft injury by direct activation of endothelial proliferation through increasing vascular endothelial growth factor production, upregulating fibroblast growth factor receptor, and increasing its ligand binding as well as other signaling
pathways for cellular recruitment.
This pathogenesis may contribute to transplant glomerulopathy and vasculopathy that feature vascular intima thickness with smooth muscle cell invasion.
-The latter two complement-independent mechanisms can explain the clinical phenotypes of antibody mediated rejection with negative C4d staining in peritubular capillaries.
-IgG4iDSA–associated subacute and chronic phenotypes of antibody-mediated rejection are consistent with complement independent pathogeneses.
-Reference
Zhang R. Donor-Specific Antibodies in Kidney Transplant Recipients. Clin J Am Soc Nephrol. 2018 Jan 6;13(1):182-192. doi: 10.2215/CJN.00700117. Epub 2017 Apr 26. PMID: 28446536; PMCID: PMC5753302.
Well done Ahmed
The complement system is a part of innate immunity and has an important role in kidney transplant rejection. Although adaptive immunity is highly effective in long term protection against foreign antigens, it is relatively sluggish and, an innate immunity that comprises inflammatory cells and soluble mediators (such as complement) response instantly to foreign instruction.
A growing body of evidence suggested that innate immunity has a principal role in priming of allogenic-specific adaptive immunity and rejection. In addition, the communication between innate and adaptive immune response in kidney transplant rejection is bidirectional.
Also, there is a cross-talk between the complement system and coagulation cascade in kidney transplantation.
DBD can induce activation of complement cascade. In addition, complements have an important role in ischemia reperfusion injury that is a principal cause of DGF. The inflammatory response after cold and warm ischemia time is characterized by graft cell injury and death, activation of the complement system, and filtration of graft parenchyma with monocytes and neutrophils. Ongoing studies assess the role of inhibition of complement activity in reducing ischemia-reperfusion injury.
Preclinical animal studies have shown that treatment of DBD donors with complement inhibitors is associated with improved graft function after transplantation.
Only two categories of complement- inhibiting drugs are in the clinic: thehumanized anti- C5 monoclonal Ab eculizumab (Soliris,Alexion) and preparations of C1INH (for example, Berinert (CSL Behring), Cinryze).
Eculizumab, a humanized monoclonal antibody which inhibits the activity of C5 convertase, thus blocking C5a and C5b formation and, as a result, inhibits the formation of membrane attack complex and was used in transplant recipients to prevent complement mediated microvascular damage that is associated with ongoing antibody mediated rejection. In progress studies have been established for evaluation of its usefulness in antibody mediated rejection, desensitization protocols, and prevention of ischemia reperfusion injuries.
A variety of reports show a range of responses to eculizumab from very effective to no effect in patients with ABMR. Few studies demonstrate that eculizumab is not useful in patients with non- C1q binding HLA- DSAs, only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti- complement treatment. On the other hand, complement inhibition at the level of C5 does not prevent upstream complement activity, more over residual terminal pathway activity via the alternative pathway can occur despite eculizumab treatment.
Based on these observations, the concept of upstream complement inhibition is gaining increasing interest. C1INH, either in its recombinant form or as an enriched preparation from human plasma, has been successfully used to prevent allogeneic and xenogeneic humoral immune responses in preclinical models. Finally, a clinically applicable C3 inhibitor, the latest generation of compstatin analogues (Cp40), has been evaluated in clinical trial.
It would help if you were up to a point in your answers.
Don’t distract your reader with an unstructured writing style.
Give an example and a reference for blocking a complement component that is directly related to the coagulation cascade.
Ali- Reza Biglarnia1, Markus Huber- Lang2, Camilla Mohlin3, Kristina N. Ekdahl3,4 and Bo Nilsson4, The multifaceted role of complement in kidney transplantation, nature reviews, nephrology, vol 14, December 2018.
Paola Pontrelli 1, Giuseppe Grandaliano and Cees Van Kooten, editorial, kidney transplantation and innate immunity. Frontiers in immunology, vol 11, octobor 2020
angiotensin receptor type I and II antibodies can induce graft injury independent of complement
Bogdan Marian Sorohana , Gener Ismailab, NicolaeLeca, DorinaTacuc .Angiotensin II type 1 receptor antibodies in kidney transplantation: An evidence-based comprehensive review. Transplantation Reviews
October 2020, Volume 34, Issue 4, 100573.
antibody against G protein coupled receptor and angiotensin receptors type I and II were found in the patient with severe rejection with vascular affection without c4d deposition ( without complement fixation).
Aurelie Philippe. Angiotensin II Type 1 Receptor Antibodies Trigger Inflammation in Renal Transplantation. Kidney International Reports. 2019 vol (4) issue 4, P 510-12.
The complement system is part of the innate immune system which enhances the ability of antibodies and phagocytic cells to clear microbes and damaged cells, promote inflammation, and attack pathogen.It consists of a small number of proteins synthesized by the liver circulate in the blood as inactive precursors when activated generate bioactive components including C3b, C3a, C5a, C5b-9 with different proinflammatory, chemoattractant, and cell-damaging functions.
It activates through 3 pathways:
1. classical pathway which triggered by recognition of subclasses of surface-bound IgG and IgM antibodies by C1q
2. The lectin pathway which triggered by recognition of bacterial surface sugar by mannose-binding lectin(MBL).
3. Alternative pathway which activates by spontaneous hydrolysis ofC3.
These 3 pathways lead to the formation of C3 convertases which lead to the generation of cell-killing membrane attack complex (MAC).
Late –onest antibody-mediated rejection(AMR) is the leading cause of allograft loss and poor long-term graft survival. In AMR donor-specific antibodies (DSAs) bind to mismatch HLA molecules and can activate the classical complement pathway leading to allograft vascular injury.
Eculizumab,anti-C5 antibody, C1 esterase inhibitor have been the subjects of recent clinical trials in the prevention and treatment of AMR.
Eculizumab is direct against C5, it is used for the prevention of AMR and delayed graft function but according to the study no difference between eculizumab and placebo in the prevention of delayed graft function. While studies comparing eculizumab and plasma exchanges with plasma exchanges only in preventing AMR showed that graft and patient survival were excellent in both groups and incidence of acute AMR was 7.7% in the eculizumab and plasma exchange group compared to 41.2% in the plasma exchanges group.1
Studies of C1 –inhibitor in preventing AMR showed a reduced incidence of AMR in the C1 inhibitor group compared with the control group.2
References
1.Stegall MD, Diwan T, Raghavaiah S, et al. . Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant 2011;11:2405
2.Vo AA, Zeevi A, Choi J, et al. . A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation 2016;99:299–308.
Answer to Professor’s Halawa question: Complement activation is an important step in graft damage by the immune system. Can graft injury happen without activation of complement?
Give examples
Graft injury could be immune response dependent or independent .
For the immune-mediated graft injury, immune response could be complement dependent or independent. Several mechanisms have been proposed for complement-independent tissue damage in the settings of positive DSAs. It is known that activated monocytes and macrophages infiltrate into graft tissues.
Immune- dependent complement independent graft damaged could be explained by the binding of DSA to endothelial cells which alone, can induce an alloreaction of CD4 T-cells via graft endothelial cell HLA-class II.
Recent evidences indicated histological findings of ABMR without C4d deposition in many cases which indicated complement independent mechanisms of injury, at which recent Banff classification criteria included a category of C4d-negative ABMR.
References:
Nephron 2020;144(suppl 1):2–6, DOI: 10.1159/000510747
Haas M. The significance of C4d staining with minimal histologic abnormalities. Curr Opin Organ Transplant. 2010;15(1):21–7
Excellent, well done.
The complement system is divided into 3 major pathways the classical, lectin, and alternative pathways, the end result of these pathways is cleavage of C3 into C3a and C3b, C3b activate cleavage of C5 to C5 a, and C5b.
1- C5b initiate formation of membrane attack complex (MAC) with subsequent lysis of target cell
2- C3b is deposited on target cells (opsonization) then phagocytosis by cells that have complement receptor
3- C3 a, C5a are potent anaphylatoxin that initiate inflammation, vasodilatation
⦁ In the classical pathway, the initiating step is formation of Ag-Ab complexes.(1)
⦁ In the lectin pathway the initiating step is binding of lectins to sugar on the surface of microorganism. (2)
⦁ In alternative pathway (AP) no antigen exposure and no antibody needed for activation of this system, it acts independently (3)
Complement system has important rule in graft dysfunction which is very evident in the following two situations :
1- Antibody mediatied rejection
ABMR is the most common cause of graft failure
Active ABMR occurs due to binding of circulating antibodies to donor alloantigens located on graft endothelium (HLA class I, HLA class II, ABO – antigens or non HLA antigens on endothelial cells)
Ag-Ab complexes activate classical complement pathway with final production of c5b, c3b, c3a, c5a leading to inflammation, cell damage and graft dysfunction
One of degradation products of classical pathway is C4b which is converted to C4d, the later binds covalently to endothelium of peritubular capillaries (PTCS) and acts as a foot print for complement activation and ABMR.
C4d normally can be detected in mesangium, but deposition in PTCS occurs only in ABMR, deposition of C4d may be defuse if > 50% of PTCS are involved or may be focal if < 50 % of PTCS are involved (4)
Diagnosis of ABMR:
⦁ Histologic evidence of acute tissue injury (capillaritis and/or glomerulitis)
⦁ Evidence of antibody interaction with vascular endothelium (C4d staining in peritubular capillaries [PTCs])
⦁ Serologic evidence of circulating DSAs
If the patient has first criteria and only one of the other 2 criteria, the patient is considered to have ABMR, this means C4d staining can replace DSA, and C4d negative ABMR exists (ABMR without activation of complement cascade) (5)
Patients with DSA who are C4d positive has lower graft survival than those who are C4d negative, this means that it is a marker for severity and has prognostic implication (6). but C4d negative patients have higher rate of developing transplant glomerulopathy if untreated
In one study it was found that C4d positive staining was present in all patients with ABMR and no patients with TCMR (7), moreover it was found that C4d was 95 % sensitive and 96 % specific for the presence of DSA
in ABO -incompatible kidney transplants some times C4d staining occur in the absence of histologic features of injury this is called graft accommodation [8].
Current treatment is directed against depleting B cells (rituximab and ATG added if mixed ABMR+ TCMR) , decreasing production of antibodies (IVIG) and removing DSA (plasmapheresis- used in early ABMR < 1 y) and decreasing inflamation produced by rejection (high dose corticosteroids) this is in combination with intensification of maintenance immunosuppression, antiviral and antimicrobial prophylaxis
Eculizumab is a monoclonal antibody, fully humanized, directed against C5 so inhibit the formation of MAC, it is FDA approved in the treatment of PNH and aHUS
In renal transplantation there is no of randomized controlled studies that prove the efficacy and safety in the treatment of ABMR, some reports its efficacy for treatment of acute refractory C4d positive (9), on the other hand, ABMR occur in patient already receiving eculizumab for other indication such as a HUS
2- Complement medicated TMA
Complement medicated TMA occur due to abnormal regulation of alternative pathway of complement due to mutation in either :
⦁ Regulators (factor H, I) in the form of loss of function or
⦁ Activators (C3, factor B) in the form of gaining of functions
leading to uncontrolled activation of complement system
TMA is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and AKI that if untreated can cause graft loss
Post transplantation TMA can occur either in the form of :
1- Recurrent disease (nearly all patients have complement medicated TMA)
It is recommended that all patients with complement mediated TMA to use living- unrelated or deceased donor kidney and all patients with recurrent TMA to be considered as having complement mediated TMA and should be treated with eculizumab.
Observational date suggest that in recurrent TMA post transplant eculizumab introduction leads to elevation of platelet count, decrease in LDH in most of patients and improve kidney function in 50% of patients (10)
2- Denovo TMA (due to immunosuppressive drugs, ABMR, viral infections)
All patients with denovo TMA should be evaluated for genetic mutations, and all possible reversible causes should be treated till having the result of genetic testing, if gentetic mutation found or if patient has progressive disease despite correcting suspected etiology, eculizumab should be started.
REFERANCES
1- Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol 2014; 5:520.
2- Kjaer TR, Thiel S, Andersen GR. Toward a structure-based comprehension of the lectin pathway of complement. Mol Immunol 2013; 56:222.
3- Thurman JM, Holers VM. The central role of the alternative complement pathway in human disease. J Immunol 2006; 176:1305.
4- Feucht HE, Felber E, Gokel MJ, et al. Vascular deposition of complement-split products in kidney allografts with cell-mediated rejection. Clin Exp Immunol 1991; 86:464.
5- Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell-mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am J Transplant 2018; 18:293.
6- Böhmig GA, Exner M, Habicht A, et al. Capillary C4d deposition in kidney allografts: a specific marker of alloantibody-dependent graft injury. J Am Soc Nephrol 2002; 13:1091.
7- Collins AB, Schneeberger EE, Pascual MA, et al. Complement activation in acute humoral renal allograft rejection: diagnostic significance of C4d deposits in peritubular capillaries. J Am Soc Nephrol 1999; 10:2208.
8- Bach FH, Turman MA, Vercellotti GM, et al. Accommodation: a working paradigm for progressing toward clinical discordant xenografting. Transplant Proc 1991; 23:205.
9- Yamamoto T, Watarai Y, Futamura K, et al. Efficacy of Eculizumab Therapy for Atypical Hemolytic Uremic Syndrome Recurrence and Antibody-Mediated Rejection Progress After Renal Transplantation With Preformed Donor-Specific Antibodies: Case Report. Transplant Proc 2017; 49:159.
10-Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 2013; 368:2169.
Which type of AMR is the most common cause of graft failure?
How is complement activation related to the type and timing of AMR?
Over the past few decades, the introduction of CNIs into transplantation practice
have decreased the frequency and severity of T cell-mediated rejection
(TCMR), and patients can expect a reasonably normal lifestyle, at least in the
short-term. However, T cell targeted therapies have little effect
on AMR, and AMR is becoming a frequent, although not the most, cause of
late graft loss. The most common diagnosis in most studies is acute TCMR where AMR accounts for around 10%of patients. .
Antibody-mediated rejection (AMR) is a significant complication following kidney
transplantation that contributes toward both short and long term injury in
approximately 1% to 10% of kidney transplant recipients.
Despite use of desensitization protocols, up to 1/3 of highly sensitized recipients may develop acute AMR following transplantation.
Withdrawal or reduction of immunosuppression, noncompliance with
immunosuppression, young age, pretransplant sensitization are the major risk
factors for DSAs. Mostly De novo DSAs, especially HLA II antibodies are the
major risk factors for chronic antibody mediated graft rejection.
Clinical picture includes proteinuria, hypertension, progressive functional
deterioration, and overt graft failure.
https://doi.org/10.1186/s12882-021-02486-9
https://doi.org/10.1111/ctr.14320
DOI:10.1111/ajt.14584
During the lifetime of a transplanted kidney, inflammatory and immunological mechanisms eventually destroy the functioning tissue to variable degrees.
The complement system plays a significant role in these negative responses. Complement activation is implicated in the pathogenesis of several disorders that result in uraemia and may lead to kidney transplant degradation.
Complement is also activated in kidney donors, especially after brain or heart death, causing organ damage that impairs graft quality and transplantation results. During procurement and preservation, the graft continues to deteriorate due to complement activation.
The main mechanism that triggers complement activation in all of these phases of the transplantation process is ischaemia, which results in phenotypic changes to parenchymal and endothelial cells that are recognized by the complement system, and the most damaging event that occurs during the ischaemic period is reperfusion of the graft with blood.
Complement has an important role in I/R injury, the severity of which is strongly associated with the degree and length of the ischaemic period.
Complement activity post-transplantation also has a role in tissue damage, particularly during acute and chronic ABMR, which can result in late-stage glomerulopathy and recurrence of kidney disease in the graft.
Complement activation in transplant recipients occurs as a result of two major mechanisms.
Most commonly, complement activation is triggered by IgG and IgM antibodies that are specific for epitopes exposed on donor ABO and HLA antigens.
However, the phenotype of the transplant recipient can also lead to complement activation owing to incompatibilities in complement components and/or regulators.
Biglarnia, AR., Huber-Lang, M., Mohlin, C. et al. The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol 14, 767–781 (2018). https://doi.org/10.1038/s41581-018-0071-x
Thankyou all for cntribution but as per the question asked by Pof. Alaa Ali does the temporal relation between the AMR and complement activation have an impact on decision to treat ie: IRI
Hyperacute rejection
Acute rejection
Or late AMR
The complement system plays an important role in graft injury ( the ischemia-reperfusion injury which leads to delayed graft function , in the antibody mediated rejection, and in the recurrence of atypical HUS post transplantation)
Eculizumab which is a fully humanized monoclonal antibody against C5 used in various aspects of kidney transplantation. it blocks the terminal complement system and prevent the formation of membrane attack complex (C5-C9) so it prevents direct tissue injury by the complement system but it dose not reduce the effect of early complement system on the graft like C3b which is a powerful opsonization factor or C3a and C5a which are powerful chemotactic factors which can lead to graft injury indirectly by recruiting inflammatory cells into the graft .
one study studied the use of eculizumab to prevent the DGF in high risk patients in deceased donor transplantation , the result showed no difference between ecluzumab and placebo.
Eculizumab in the prevention of antibody mediated rejection: in one study reported by Stegal et al , they study the use of eculizumab in highly sensitized patients with live donor transplantation . they found that the eculizumab treated patients had lower incidence of AMR at one year, but with high rates of subclinical rejection. Another studies with larger size is needed to confirm this results and to clarify the dose of eculizumab and the duration of tratment.
Eculizumab had been used successfully in the prevention and treatment of recurrent aHUS post transplantation .
C1 inhibitors is another drug used in small studies to prevent AMR in highly sensitized patients with promising early results .
Reference :
Ch. Legendre, R. Sberro-Soussan, J. Zuber and V. Frémeaux-Bacchi
The role of complement inhibition in kidney transplantation
British Medical Bulletin, 2017, 124:5–17 doi: 10.1093/bmb/ldx037
The complement system, traditionally considered a component of innate immunity required for protection from invading pathogens,
Complement activation can be initiated through three pathways
The recognition that complement participates in antibody-initiated allograft rejection suggested that identifying serum anti-HLA antibodies capable of binding C1q would enhance their prognostic use after kidney transplantation
A 2013 paper, indeed, suggests that, among patients with serum anti-HLA antibodies, those binding to C1q1 had the worst kidney graft survival
In another example of complement-based diagnostics, C4d staining of kidney transplant tissue is currently considered one key criterion for diagnosing antibody-mediated allograft rejection
Clin J Am Soc Nephrol 10: 1636–1650, 2015. doi: 10.2215/CJN.06230614
If you have a negative C4d AMR resistant to coventional methods wouid you use cmplement controling drugs?
Complement cascade is an important part of the innate immune system. Studies have shown that activation of complement cascade is implicated in allograft injury by means of several mechanisms, including ischemia/reperfusion injury, antibody mediated rejection, cellular rejection and thrombotic micro-angiopathy.
The complement cascade consists of more than 30 soluble and cell-bound proteins.They become biologically active fragments, complement receptors, and complement regulatory proteins whenever the transplanted organ is exposed to recipient complement proteins as soon as it is reperfused.
The complement system is not only involved in antibody-mediated cell lysis, which is the major effector mechanism, but that is only one of its functions. The complement system can be activated in an antibody-independent fashion. Complement fragments upregulate T cell differentiation and the B cell response to antigens. Once complement cascade is activated, within a transplanted organ it can have direct and indirect pathologic effects.
The activation can occur through classical, alternative, or lectin pathway, resulting in the creation of anaphylatoxins C3a and C5a which are involved in possessing several immune functions, and the assembly of terminal complement cascade, result in direct cell lysis.
C3a and C5a are proteins that directly affect resident organ cells, they are activators for neutrophils and macrophages, and they provide important signals for T and B cell activation in the tubulointerstitium in the setting of ischemia reperfusion, whereas in AMR activation occurs in the peritubular capillaries.
Owing to the great role of complement in allograft injury, complement system can be considered as potential therapeutic targets. Recently, the inhibition of several complement components has shown certain results in preventing the antibody-mediated rejection. C1 inhibitor is a serine protease inhibitor that is capable of irreversibly blocking not only C1 complex components. Very recently, a double-blind study assessed the safety of C1-inhibitor in kidney transplant recipients and suggested the positive effect of this treatment regarding the C1q+HLA antibody levels reduction and antibody-mediated rejection occurrence.
Eculizumab is a humanized monoclonal antibody capable of blocking the cleavage of C5 complement component, thus preventing the release of anaphylatoxin C5a and the assembly of MAC. The C5 component participates in all three main pathways of complement activation; therefore, the blockade of its cleavage can have a positive influence on various medical conditions regardless of the active pathway. Eculizumab was shown to be an effective treatment for paroxysmal nocturnal hemoglobinuria and aHUS including its recurrence in kidney transplantation.
REFRENCES:
https://doi.org/10.3389/fimmu.2019.02380
https://doi.org/10.3389/fmed.2017.00066
doi:10.1097/MOT.0000000000000216.
doi: 10.1097/TP.0000000000000592
doi:10.1056/NEJMoa061648
doi:10.1053/j.ajkd.2015.12.034
Thanks to All
Waiting for more replies
Excellent Mohamed
I will quote this from your reply “The complement system is not only involved in antibody-mediated cell lysis, which is the major effector mechanism, but that is only one of its functions. The complement system can be activated in an antibody-independent fashion. Complement fragments upregulate T cell differentiation and the B cell response to antigens. Once complement cascade is activated, within a transplanted organ it can have direct and indirect pathologic effects”.
Dear All
Can you summarise the direct and indirect effects of complement activation?
Once complement is activated in response to trigger, it can have direct and indirect effects on immune system.
Direct effect is explained by the activation of complement cascade by the bound antigen to antibodies ( donor HLA to T or B lymphocytes resulting is cytotoxic effect and lysis.
Multiple different biologically active complement fragments are generated during activation of complement cascade. These proteins and fragments directly affect resident organ cells, they are chemotaxins and activators for neutrophils and macrophages, and they provide important signals for T and B cell activation.
Direct effect when complement causes cell lysis after antigen-antibody interaction
Indirect effect are related to the complement fragments e.g. C3, C5. They can acts as anaphylatoxin and play role in inflammatory process and induces damages
Complement pathway is activated upon renal transplantation through the following:
Ischemic reperfusion injury which produces ROS reactive oxidative stress factors which initiate complement pathway producing C5a and C5b
Also, C3a and C3b are produced during complement activation pathway.
Renal (tubules) and hepatic synthesis of C3.
Collectin -11 which is soluble lectin expressed in kidney tissue and responsible for complement activation in the kidney, increased with ischemic stress and trigger complement activation.
The C5a receptor antagonist has therapeutic effects in any autoimmune disease , C5a is a major inflammatory peptide in antibody mediated rejection so its block may be another hope against transplant associated inflammatory responses.
Christopher L. Nauser, Conrad A. Farrar and Steven H. Sacks. Complement Recognition Pathways in Renal Transplantation. JASN September 2017, 28 (9) 2571-2578.
C Legendre, R Sberro-Soussan, J Zuber, V Frémeaux-Bacchi. The role of complement inhibition in kidney transplantation. British Medical Bulletin, Volume 124, Issue 1, December 2017, Pages 5–17.
The process of transplantation involves critical events that occur in the donor, recipient and the graft leading to graft damage. These events trigger an early inflammatory reaction that is independent of donor specific immune response of the recipient. Ischemia and subsequent reperfusion drive the initial inflammatory process of which complement activation is a major effector mechanism.
In kidney transplantation, several sequential events that occur before, during and after transplantation and lead to complement activation and loss of kidney function including:
Pre-transplantation:
complement activation occur with each hemodialysis session.
deceased donor: DCD, DBD
Early pre/ post transplant:
procurement, preservation & transportation
reperfusion
ABMR
Late post transplant:
dysregulation of alternative pathway
recurrence of disease
rejection
transplant glomerulopathy
age related functional loss
A number of agents and several strategies have been developed to deactivate complement system including:
optimization of graft quality before transplantation
treatment of antibody mediated rejection
induction of accommodation
regulation of adaptive immunity
The agents that can deactivate the complement system include:
IVIG, Rituximab, C1INH( clinical trial), Eculizumab( clinical trial), BIVV009( clinical trial), Ides( clinical trial), Mirococept( clinical trial), Compstatin family inhibitors( clinical trial), sCR1( pre clinical development), TT30( pre clinical development), C5aR1 antagonist( pre clinical development), cobra venom factor( pre clinical development).
Although the initial results of clinical trials are promising, inconsistencies in their effect on delayed graft function and ABMR reflect the complexity of allograft injury and the involvement of non-complement mediated factors.
References:
Thank you, Huda
Would you elaborate more on the outcomes of the currently available agents? Furthermore, how to balance between benefits and drawbacks of each according to the available data?
Terminal C5 inhibition
Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: A randomized trial
William H Marks et al
At week 9, treatment failure rate ( AMR BANFF II and III) was 11.8% in eculizumab group vs 21.6% in standard of care group. when BANFF I AMR was included, treatment failure was 11.8% vs 29.4% in standard of care group. This finding shows that eculizumab may be more effective than SOC at preventing acute AMR in recipients who are sensitized to their living-donor kidney transplants.
Safety and efficacy of eculizumab for the prevention of antibody-mediated rejection after deceased-donor kidney transplantation in patients with preformed donor-specific antibodies
Denis Glotz et al
Participants received eculizumab as follows:
1200 mg immediately before reperfusion
900 mg on posttransplant days 1, 7, 14, 21, and 28
1200 mg at weeks 5, 7, and 9.
All patients received thymoglobulin induction therapy and standard maintenance immunosuppression including steroids.
The primary end point was treatment failure rate composed of biopsy-proved grade II/III AMR within 9 weeks posttransplant. In 80 patients the Observed treatment failure rate (8.8%) was significantly lower than expected for standard care (40%). At 36 months, graft and patient survival rates were 83.4% and 91.5%, respectively. Eculizumab was well tolerated, and there were no new safety concerns. In such patients, eculizumab has the potential to give prophylaxis against damage caused by acute AMR.
Proximal inhibition
C1 Inhibitor in Acute Antibody-Mediated Rejection Nonresponsive to Conventional Therapy in Kidney Transplant Recipients: A Pilot Study
D. Viglietti et al
C1 inhibitor (C1-INH) Berinert was added to high-dose intravenous immunoglobulin for the treatment of acute ABMR that is nonresponsive to conventional therapy. Kidney recipients with nonresponsive active ABMR and acute allograft dysfunction C1-INH and IVIG for 6 months (six patients). The primary end point was the change in eGFR at 6 months after inclusion (M+6). All patients showed an improvement in eGFR between inclusion and M+6. In kidney patients with nonresponsive acute ABMR, C1-INH administered with IVIG is safe and may enhance allograft function.
Plasma-Derived C1 Esterase Inhibitor for Acute Antibody-Mediated Rejection Following Kidney
Transplantation: Results of a Randomized DoubleBlind Placebo-Controlled Pilot Study
R. A. Montgomery et al
Human plasma derived C1 INH was added to plasmapheresis and ivIg for AMR.20000 units of C1 INH in divided doses was given every other day for 2 weeks .no difference was found between C1 INH and standard of care group in day 20 pathology and graft survival,however C1 INH group demonstrated sustained improvement in renal function .Transplant glomerulopathy was not seen in any of the biopsy done in C1 INH group at 6 months. This study suggests that adding C1 INH in the treatment of AMRto plasmapharesis and ivIg could be beneficial.
Long term follow-up
Despite the encouraging findings of the above trials, long-term follow-up of eculizumab-treated positive crossmatch patients in a single-center trial revealed that, despite the avoidance of early active AMR, the long-term effects of eculizumab in Allograft survival and the incidence of chronic AMR are comparable to historical controls.
Long-term outcomes of eculizumab-treated positive crossmatch recipients: Allograft survival, histologic findings, and natural history of the donor-specific antibodies
Carrie A. Schinstock et al
Positive Crossmatch Kidney Transplant Recipien Treated With Eculizumab: Outcomes Beyond 1 Year
L. D. Cornell et al
Use of Eculizumab for Active Antibody-Mediated Rejection that Occurs Early Post-Kidney Transplantation: A Consecutive Series of 15 Cases
Ek Khoon Tan et al
They concluded that prompt eculizumab treatment is safe and effective for early active AMR after kidney transplant or when abrupt increases in donor-specific antibodies is seen and biopsy cannot be performed for diagnosis confirmation.
Thanks Prakash
What is the problem of eculizumab?
Will you use it as a first line of treatment of rejection?
Although eculizumab has been used to prevent ABMR in highly sensitized patients and also as a salvage agent in treatment of refractory active ABMR, because of limitations I have mentioned in my last comment and, in addition, lack of randomized clinical trials proving its efficacy and safety, it is not the first line therapy of ABMR
my last comment: A variety of reports show a range of responses to eculizumab from very effective to no effect in patients with ABMR. Few studies demonstrate that eculizumab is not useful in patients with non- C1q binding HLA- DSAs, only ABMR that is caused by complement dependent effector mechanisms might be susceptible to anti- complement treatment. On the other hand, complement inhibition at the level of C5 does not prevent upstream complement activity, more over residual terminal pathway activity via the alternative pathway can occur despite eculizumab treatment.
Thanks, Esmat
Why there is no enough RCT to evaluate this drug?
May be because of high cost.
Complement system is a part of innate immune system which also comprised of dendrites ,monocytes, macrophages .
The complement system is activated through three distinct pathways:
classical pathway
Alternative pathway
Lectin pathway.
the role of complement in kidney transplantation including the following conditions:
-ischaemia-reperfusion injury
-antibody-mediated rejection
-recurrence of native kidney disease as : atypical HUS , C3 glomerulopathy, anti-phospholipid syndrome .
**In ischemia reperfusion injury:
ischaemia-reperfusion-induced endothelial injury results in activation of lectin pathway via binding of mannose-binding lectin (MBL) to epitopes exposed on ischaemic tissue.
The alternative pathway amplifies this, while the classical pathway role remains controversial.
This lesions are reduced by deficiency or depletion of complement factor as C3, C5, CFB, C3aR and C5aR and enhanced by deficiency of complement regulator as decay-accelerating factor (DAF)
The involvement of complement is suggested by:
detection of soluble C5b-9 in deceased kidney but not living one.
increased in complement genes expression in deceased-donor kidneys
brain-death induce activation of complement
up- regulation of C5aR expression in renal tubular cells.
**ABMR :
Immune – complex combining antigens (MHC molecules, ABO-blood group antigens) and IgM/IgG antibodies called donor-specific antibodies (DSAs) leads to activation of the classical pathway to start the process of ABMR.
Diagnosis of ABMR depend on detection of detection of complement fragments C4d on peritubular capillaries .
However , cases of C4d negative ABMR have been reported , due to technical reasons, or a low level of complement of classical pathway.
**Recurrence of disease involving mainly the alternate pathway :
-Atypical haemolytic uraemic syndrome (aHUS)
recurrence occurs in 68% of patients which is marked during the first year (with an incidence of 70%) and decreases markedly thereafter.
Patients with mutations in complement factors have a 3 folds increase in aHUS recurrences post transplant compared to aHUS patients without mutations.
-C3 glomerulopathy :
Is characterized by C3 deposits in the mesangium and along the glomerular basement membrane (in cases of C3 glomerulonephritis) or within the GBM (in cases of DDD).
The disease carries a poor prognosis with progression to ESRD in 50% of cases during the first decade after initial presentation , and also recurs after kidney transplantation in ~50% of cases.
Complement inhibition after Renal transplantation :
-Eculizumab: is a recombinant monoclonal antibody against C5.
It minimizes immunogenicity, Fc-mediated functions and complement activation , so prevents delayed graft function, in acute ABMR and for treatment or prevention of aHUS recurrence and anti- phospholipid syndrome.
It increases the risk of meningitis due to the Neisseria species , so Prophylaxis with vaccination and antibiotics is mandatory.
-C1 estrase inhibitor (C1-INH) block classical pathway of complement activation by C1s &C1r inhibition.
Reference:
Legendre .CH, Sberro-Soussan .R, et al.The role of complement inhibition in kidney transplantation, British Medical Bulletin,(2017),124 :5-17,doi: 10.1093/bmb/ldx037