1
|
Habibabady Z, McGrath G, Kinoshita K, Maenaka A, Ikechukwu I, Elias GF, Zaletel T, Rosales I, Hara H, Pierson RN, Cooper DKC. Antibody-mediated rejection in xenotransplantation: Can it be prevented or reversed? Xenotransplantation 2023; 30:e12816. [PMID: 37548030 PMCID: PMC11101061 DOI: 10.1111/xen.12816] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Antibody-mediated rejection (AMR) is the commonest cause of failure of a pig graft after transplantation into an immunosuppressed nonhuman primate (NHP). The incidence of AMR compared to acute cellular rejection is much higher in xenotransplantation (46% vs. 7%) than in allotransplantation (3% vs. 63%) in NHPs. Although AMR in an allograft can often be reversed, to our knowledge there is no report of its successful reversal in a pig xenograft. As there is less experience in preventing or reversing AMR in models of xenotransplantation, the results of studies in patients with allografts provide more information. These include (i) depletion or neutralization of serum anti-donor antibodies, (ii) inhibition of complement activation, (iii) therapies targeting B or plasma cells, and (iv) anti-inflammatory therapy. Depletion or neutralization of anti-pig antibody, for example, by plasmapheresis, is effective in depleting antibodies, but they recover within days. IgG-degrading enzymes do not deplete IgM. Despite the expression of human complement-regulatory proteins on the pig graft, inhibition of systemic complement activation may be necessary, particularly if AMR is to be reversed. Potential therapies include (i) inhibition of complement activation (e.g., by IVIg, C1 INH, or an anti-C5 antibody), but some complement inhibitors are not effective in NHPs, for example, eculizumab. Possible B cell-targeted therapies include (i) B cell depletion, (ii) plasma cell depletion, (iii) modulation of B cell activation, and (iv) enhancing the generation of regulatory B and/or T cells. Among anti-inflammatory agents, anti-IL6R mAb and TNF blockers are increasingly being tested in xenotransplantation models, but with no definitive evidence that they reverse AMR. Increasing attention should be directed toward testing combinations of the above therapies. We suggest that treatment with a systemic complement inhibitor is likely to be most effective, possibly combined with anti-inflammatory agents (if these are not already being administered). Ultimately, it may require further genetic engineering of the organ-source pig to resolve the problem entirely, for example, knockout or knockdown of SLA, and/or expression of PD-L1, HLA E, and/or HLA-G.
Collapse
Affiliation(s)
- Zahra Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ileka Ikechukwu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela F. Elias
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Tjasa Zaletel
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Hara
- Yunnan Xenotransplantation Engineering Research Center, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Howard MC, Nauser CL, Farrar CA, Sacks SH. Complement in ischaemia-reperfusion injury and transplantation. Semin Immunopathol 2021; 43:789-797. [PMID: 34757496 PMCID: PMC8579729 DOI: 10.1007/s00281-021-00896-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/22/2021] [Indexed: 01/08/2023]
Abstract
Until recently, the only known condition in which complement could mediate transplant injury was the rare occurrence of antibody-mediated rejection, in which the original concept of antibody immunity against the transplant was supported by complementary proteins present in the serum. This has changed within the last two decades because of evidence that the processes of ischaemia–reperfusion injury followed by T cell–mediated rejection are also critically dependent on components generated by the complement system. We now have a clearer understanding of the complement triggers and effectors that mediate injury, and a detailed map of their local sites of production and activation in the kidney. This is providing helpful guidelines as to how these harmful processes that restrict transplant outcomes can be targeted for therapeutic benefit. Here we review some of the recent advances highlighting relevant therapeutic targets.
Collapse
Affiliation(s)
- Mark C Howard
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, 5thFloor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
| | - Christopher L Nauser
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, 5thFloor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Conrad A Farrar
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, 5thFloor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Steven H Sacks
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, 5thFloor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| |
Collapse
|
3
|
Parlakpinar H, Gunata M. Transplantation and immunosuppression: a review of novel transplant-related immunosuppressant drugs. Immunopharmacol Immunotoxicol 2021; 43:651-665. [PMID: 34415233 DOI: 10.1080/08923973.2021.1966033] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunosuppressive drugs used in the transplantation period are generally defined as induction and maintenance therapy. The use of immunosuppressants, which are particularly useful and have fewer side effects, decreased both mortality and morbidity. Many drugs such as steroids, calcineurin inhibitors (cyclosporine-A, tacrolimus), antimetabolites (mycophenolate mofetil, azathioprine), and mTOR inhibitors (sirolimus, everolimus) are used as immunosuppressive agents. Although immunosuppressant drugs cause many side effects such as hypertension, infection, and hyperlipidemia, they are the agents that should be used to prevent organ rejection. This shows the importance of individualized drug use. The optimal immunosuppressive therapy post-transplant is not established. Therefore, discovering less toxic but more potent new agents is of great importance, and new experimental and clinical studies are needed in this regard.Our review discussed the mechanism of immunosuppressants, new agents' discovery, and current therapeutic protocols in the transplantation.
Collapse
Affiliation(s)
- Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
4
|
Matsuda Y, Watanabe T, Li XK. Approaches for Controlling Antibody-Mediated Allograft Rejection Through Targeting B Cells. Front Immunol 2021; 12:682334. [PMID: 34276669 PMCID: PMC8282180 DOI: 10.3389/fimmu.2021.682334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
5
|
Ochando J, Ordikhani F, Boros P, Jordan S. The innate immune response to allotransplants: mechanisms and therapeutic potentials. Cell Mol Immunol 2019; 16:350-356. [PMID: 30804476 DOI: 10.1038/s41423-019-0216-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023] Open
Abstract
Surgical trauma and ischemia reperfusion injury (IRI) are unavoidable aspects of any solid organ transplant procedure. They trigger a multifactorial antigen-independent inflammatory process that profoundly affects both the early and long-term outcomes of the transplanted organ. The injury associated with donor organ procurement, storage, and engraftment triggers innate immune activation that inevitably results in cell death, which may occur in many different forms. Dying cells in donor grafts release damage-associated molecular patterns (DAMPs), which alert recipient innate cells, including macrophages and dendritic cells (DCs), through the activation of the complement cascade and toll-like receptors (TLRs). The long-term effect of inflammation on innate immune cells is associated with changes in cellular metabolism that skew the cells towards aerobic glycolysis, resulting in innate immune cell activation and inflammatory cytokine production. The different roles of proinflammatory cytokines in innate immune activation have been described, and these cytokines also stimulate optimal T-cell expansion during allograft rejection. Therefore, early innate immune events after organ transplantation determine the fate of the adaptive immune response. In this review, we summarize the contributions of innate immunity to allograft rejection and discuss recent studies and emerging concepts in the targeted delivery of therapeutics to modulate the innate immune system to enhance allograft survival.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Farideh Ordikhani
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Boros
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefan Jordan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Khan MA, Shamma T. Complement factor and T-cell interactions during alloimmune inflammation in transplantation. J Leukoc Biol 2018; 105:681-694. [PMID: 30536904 DOI: 10.1002/jlb.5ru0718-288r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/25/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023] Open
Abstract
Complement factor and T-cell signaling during an effective alloimmune response plays a key role in transplant-associated injury, which leads to the progression of chronic rejection (CR). During an alloimmune response, activated complement factors (C3a and C5a) bind to their corresponding receptors (C3aR and C5aR) on a number of lymphocytes, including T-regulatory cells (Tregs), and these cell-molecular interactions have been vital to modulate an effective immune response to/from Th1-effector cell and Treg activities, which result in massive inflammation, microvascular impairments, and fibrotic remodeling. Involvement of the complement-mediated cell signaling during transplantation signifies a crucial role of complement components as a key therapeutic switch to regulate ongoing inflammatory state, and further to avoid the progression of CR of the transplanted organ. This review highlights the role of complement-T cell interactions, and how these interactions shunt the effector immune response during alloimmune inflammation in transplantation, which could be a novel therapeutic tool to protect a transplanted organ and avoid progression of CR.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Research Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Talal Shamma
- Organ Transplant Research Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Harris CL, Pouw RB, Kavanagh D, Sun R, Ricklin D. Developments in anti-complement therapy; from disease to clinical trial. Mol Immunol 2018; 102:89-119. [PMID: 30121124 DOI: 10.1016/j.molimm.2018.06.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023]
Abstract
The complement system is well known for its role in innate immunity and in maintenance of tissue homeostasis, providing a first line of defence against infection and playing a key role in flagging apoptotic cells and debris for disposal. Unfortunately complement also contributes to pathogenesis of a number of diseases; in some cases driving pathology, and in others amplifying or exacerbating the inflammatory and damaging impact of non-complement disease triggers. The role of complement in pathogenesis of an expanding number of diseases has driven industry and academia alike to develop an impressive arsenal of anti-complement drugs which target different proteins and functions of the complement cascade. Evidence from genetic and biochemical analyses, combined with improved identification of complement biomarkers and supportive data from sophisticated animal models of disease, has driven a drug development landscape in which the indications selected for clinical trial cluster in three 'target' tissues: the kidney, eye and vasculature. While the disease triggers may differ, complement activation and amplification is a common feature in many diseases which affect these three tissues. An abundance of drugs are in clinical development, some show favourable progression whereas others experience significant challenges. However, these hurdles in themselves drive an ever-evolving portfolio of 'next-generation' drugs with improved pharmacokinetic and pharmacodynamics properties. In this review we discuss the indications which are in the drug development 'spotlight' and review the relevant indication validation criteria. We present current progress in clinical trials, highlighting successes and difficulties, and look forward to approval of a wide selection of drugs for use in man which give clinicians choice in mechanistic target, modality and route of delivery.
Collapse
Affiliation(s)
- Claire L Harris
- Complement Therapeutics Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; National Renal Complement Therapeutics Centre, Building 26, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK.
| | - Richard B Pouw
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056, Basel, Switzerland
| | - David Kavanagh
- Complement Therapeutics Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; National Renal Complement Therapeutics Centre, Building 26, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Ruyue Sun
- Complement Therapeutics Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Daniel Ricklin
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056, Basel, Switzerland.
| |
Collapse
|
8
|
West-Thielke P, Progar K, Campara M, Jasiak N, Gallon L, Tang I, Spaggiari M, Tzvetanov I, Benedetti E. Eculizumab for Prevention of Antibody-Mediated Rejection in Blood Group-Incompatible Renal Transplantation. Transplant Proc 2018; 50:66-69. [PMID: 29407333 DOI: 10.1016/j.transproceed.2017.12.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/09/2017] [Indexed: 12/28/2022]
Abstract
Antibody-mediated rejection (AMR) is one of the leading causes of allograft failure especially in patients undergoing ABO-incompatible (ABOi) renal transplantation. We hypothesized that complement inhibition with eculizumab, a C5 inhibitor, would protect against AMR and maintain graft function in ABOi renal transplant recipients. Four patients undergoing living donor kidney transplant from ABOi donors were treated with a 9-week eculizumab course without therapeutic plasma exchange, intravenous immunoglobulin, or splenectomy. All patients had successful transplants and have normal graft function at the time of last follow-up. There were no cases of AMR or acute cellular rejection. Of note, 2 patients were transplanted despite persistent ABO antibody titers of 1:32, conventionally considered a contraindication to proceed in standard protocols. Eculizumab is a promising option to prevent AMR with ABOi renal transplantation without the need for splenectomy, post-transplant therapeutic plasma exchange, and intravenous immunoglobulin. Future multicenter studies are needed to determine long-term efficacy and safety.
Collapse
Affiliation(s)
- P West-Thielke
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA.
| | - K Progar
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - M Campara
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - N Jasiak
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - L Gallon
- Department of Nephrology, Northwestern University, Chicago, Illinois, USA
| | - I Tang
- Department of Nephrology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - M Spaggiari
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - I Tzvetanov
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - E Benedetti
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
9
|
Khan MA, Alanazi F, Ahmed HA, Vater A, Assiri AM, Broering DC. C5a Blockade Increases Regulatory T Cell Numbers and Protects Against Microvascular Loss and Epithelial Damage in Mouse Airway Allografts. Front Immunol 2018; 9:1010. [PMID: 29881374 PMCID: PMC5976734 DOI: 10.3389/fimmu.2018.01010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/23/2018] [Indexed: 12/15/2022] Open
Abstract
Microvascular injury during acute rejection has been associated with massive infiltration of CD4+ T effector cells, and the formation of complement products (C3a and C5a). Regulatory T cells (Tregs) are potent immunosuppressors of the adaptive immune system and have proven sufficient to rescue microvascular impairments. Targeting C5a has been linked with improved microvascular recovery, but its effects on the Treg and T effector balance is less well known. Here, we demonstrate the impact of C5a blockade on Treg induction and microvascular restoration in rejecting mouse airway allografts. BALB/c→C57BL/6 allografts were treated with a C5a-neutralizing l-aptamer (10 mg/kg, i.p. at d0 and every second day thereafter), and allografts were serially monitored for Treg infiltration, tissue oxygenation (tpO2), microvascular blood flow, and functional microvasculature between donor and recipients during allograft rejection. We demonstrated that C5a blocking significantly leads to enhanced presence of Tregs in the allograft, reinstates donor-recipient functional microvasculature, improves tpO2, microvascular blood flow, and epithelial repair, followed by an upregulation of IL-5, TGF-β, IL-10 vascular endothelial growth factor, and ANGPT1 gene expression, while it maintained a healthy epithelium and prevented subepithelial collagen deposition at d28 posttransplantation. Together, these data indicate that inhibition of C5a signaling has potential to preserve microvasculature and rescue allograft from a sustained hypoxic/ischemic phase, limits airway tissue remodeling through the induction of Treg-mediated immune tolerance. These findings may be useful in designing anti-C5a therapy in combination with existing immunosuppressive regimens to rescue tissue/organ rejection.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Organ Transplant Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fatimah Alanazi
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Organ Transplant Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hala Abdalrahman Ahmed
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Abdullah Mohammed Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia
- Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dieter Clemens Broering
- Organ Transplant Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Complement and Immunoglobulin Biology Leading to Clinical Translation. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
11
|
de Andrade LGM, Contti MM, Nga HS, Bravin AM, Takase HM, Viero RM, da Silva TN, Chagas KDN, Palma LMP. Long-term outcomes of the Atypical Hemolytic Uremic Syndrome after kidney transplantation treated with eculizumab as first choice. PLoS One 2017; 12:e0188155. [PMID: 29136640 PMCID: PMC5685617 DOI: 10.1371/journal.pone.0188155] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023] Open
Abstract
Introduction The treatment of choice for Atypical Hemolytic Uremic Syndrome (aHUS) is the monoclonal antibody eculizumab. The objective of this study was to assess the efficacy and safety of eculizumab in a cohort of kidney transplant patients suffering from aHUS. Methods Description of the prospective cohort of all the patients primarily treated with eculizumab after transplantation and divided into the therapeutic (onset of aHUS after transplantation) and prophylactic use (patients with previous diagnosis of aHUS undergoing kidney transplantation). Results Seven cases were outlined: five of therapeutic use and two, prophylactic. From the five cases of therapeutic use, there was improvement of the thrombotic microangiopathy in the 48 hours following the start of the drug and no patient experienced relapse during an average follow-up of 21 months in the continuous use of eculizumab (minimum of 6 and maximum of 42 months). One patient died at 6 months, due to Aspergillus infection. From the two cases of prophylactic use, one patient experienced relapsed thrombotic microangiopathy after 4 months and another patient remained asymptomatic after 16 months of follow-up, both on chronic treatment. Discussion The therapeutic use of eculizumab showed to be effective, with improvement of the microangiopathy parameters and persisting up to the end of the follow-up, without relapses. The additional risk of immunosuppression, leading to opportunistic infections, was well tolerated. The prophylactic use showed to be effective and safe; however, the doses and intervals should be individualized in order to avoid relapsed microangiopathy, especially in patients with factor H mutation.
Collapse
Affiliation(s)
| | - Mariana Moraes Contti
- Department of Internal Medicine, University São Paulo State(UNESP), Botucatu, São Paulo State, Brazil
| | - Hong Si Nga
- Department of Internal Medicine, University São Paulo State(UNESP), Botucatu, São Paulo State, Brazil
| | - Ariane Moyses Bravin
- Department of Internal Medicine, University São Paulo State(UNESP), Botucatu, São Paulo State, Brazil
| | - Henrique Mochida Takase
- Department of Internal Medicine, University São Paulo State(UNESP), Botucatu, São Paulo State, Brazil
| | - Rosa Marlene Viero
- Department of Internal Medicine, University São Paulo State(UNESP), Botucatu, São Paulo State, Brazil
| | - Trycia Nunes da Silva
- Department of Internal Medicine, Hospital Estadual de Bauru, Bauru, São Paulo State, Brazil
| | - Kelem De Nardi Chagas
- Department of Internal Medicine, University of São Paulo (USP), São Paulo, São Paulo State, Brazil
| | | |
Collapse
|
12
|
Durigutto P, Sblattero D, Biffi S, De Maso L, Garrovo C, Baj G, Colombo F, Fischetti F, Di Naro AF, Tedesco F, Macor P. Targeted Delivery of Neutralizing Anti-C5 Antibody to Renal Endothelium Prevents Complement-Dependent Tissue Damage. Front Immunol 2017; 8:1093. [PMID: 28932227 PMCID: PMC5592221 DOI: 10.3389/fimmu.2017.01093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/22/2017] [Indexed: 11/13/2022] Open
Abstract
Complement activation is largely implicated in the pathogenesis of several clinical conditions and its therapeutic neutralization has proven effective in preventing tissue and organ damage. A problem that still needs to be solved in the therapeutic control of complement-mediated diseases is how to avoid side effects associated with chronic neutralization of the complement system, in particular, the increased risk of infections. We addressed this issue developing a strategy based on the preferential delivery of a C5 complement inhibitor to the organ involved in the pathologic process. To this end, we generated Ergidina, a neutralizing recombinant anti-C5 human antibody coupled with a cyclic-RGD peptide, with a distinctive homing property for ischemic endothelial cells and effective in controlling tissue damage in a rat model of renal ischemia/reperfusion injury (IRI). As a result of its preferential localization on renal endothelium, the molecule induced complete inhibition of complement activation at tissue level, and local protection from complement-mediated tissue damage without affecting circulating C5. The ex vivo binding of Ergidina to surgically removed kidney exposed to cold ischemia supports its therapeutic use to prevent posttransplant IRI leading to delay of graft function. Moreover, the finding that the ex vivo binding of Ergidina was not restricted to the kidney, but was also seen on ischemic heart, suggests that this RGD-targeted anti-C5 antibody may represent a useful tool to treat organs prior to transplantation. Based on this evidence, we propose preliminary data showing that Ergidina is a novel targeted drug to prevent complement activation on the endothelium of ischemic kidney.
Collapse
Affiliation(s)
- Paolo Durigutto
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Stefania Biffi
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, Italy
| | - Luca De Maso
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Chiara Garrovo
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, Italy
| | - Gabriele Baj
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Federico Colombo
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Fabio Fischetti
- Dipartimento Universitario Clinico di Scienze Mediche, Chirurgiche e della Salute, University of Trieste, Trieste, Italy
| | | | | | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
13
|
Complement inhibition attenuates acute kidney injury after ischemia-reperfusion and limits progression to renal fibrosis in mice. PLoS One 2017; 12:e0183701. [PMID: 28832655 PMCID: PMC5568291 DOI: 10.1371/journal.pone.0183701] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 08/09/2017] [Indexed: 12/17/2022] Open
Abstract
The complement system is an essential component of innate immunity and plays a major role in the pathogenesis of ischemia-reperfusion injury (IRI). In this study, we investigated the impact of human C1-inhibitor (C1INH) on the early inflammatory response to IRI and the subsequent progression to fibrosis in mice. We evaluated structural damage, renal function, acute inflammatory response, progression to fibrosis and overall survival at 90-days post-injury. Animals receiving C1INH prior to reperfusion had a significant improvement in survival rate along with superior renal function when compared to vehicle (PBS) treated counterparts. Pre-treatment with C1INH also prevented acute IL-6, CXCL1 and MCP-1 up-regulation, C5a release, C3b deposition and infiltration by neutrophils and macrophages into renal tissue. This anti-inflammatory effect correlated with a significant reduction in the expression of markers of fibrosis alpha smooth muscle actin, desmin and picrosirius red at 30 and 90 days post-IRI and reduced renal levels of TGF-β1 when compared to untreated controls. Our findings indicate that intravenous delivery of C1INH prior to ischemic injury protects kidneys from inflammatory injury and subsequent progression to fibrosis. We conclude that early complement blockade in the context of IRI constitutes an effective strategy in the prevention of fibrosis after ischemic acute kidney injury.
Collapse
|
14
|
Wehling C, Amon O, Bommer M, Hoppe B, Kentouche K, Schalk G, Weimer R, Wiesener M, Hohenstein B, Tönshoff B, Büscher R, Fehrenbach H, Gök ÖN, Kirschfink M. Monitoring of complement activation biomarkers and eculizumab in complement-mediated renal disorders. Clin Exp Immunol 2016; 187:304-315. [PMID: 27784126 DOI: 10.1111/cei.12890] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2016] [Indexed: 12/18/2022] Open
Abstract
Various complement-mediated renal disorders are treated currently with the complement inhibitor eculizumab. By blocking the cleavage of C5, this monoclonal antibody prevents cell damage caused by complement-mediated inflammation. We included 23 patients with atypical haemolytic uraemic syndrome (aHUS, n = 12), C3 glomerulopathies (C3G, n = 9) and acute antibody-mediated renal graft rejection (AMR, n = 2), treated with eculizumab in 12 hospitals in Germany. We explored the course of complement activation biomarkers and the benefit of therapeutic drug monitoring of eculizumab. Complement activation was assessed by analysing the haemolytic complement function of the classical (CH50) and the alternative pathway (APH50), C3 and the activation products C3d, C5a and sC5b-9 prior to, 3 and 6 months after eculizumab treatment. Eculizumab concentrations were determined by a newly established specific enzyme-linked immunosorbent assay (ELISA). Serum eculizumab concentrations up to 1082 μg/ml point to drug accumulation, especially in paediatric patients. Loss of the therapeutic antibody via urine with concentrations up to 56 μg/ml correlated with proteinuria. In aHUS patients, effective complement inhibition was demonstrated by significant reductions of CH50, APH50, C3d and sC5b-9 levels, whereas C5a levels were only reduced significantly after 6 months' treatment. C3G patients presented increased C3d and consistently low C3 levels, reflecting ongoing complement activation and consumption at the C3 level, despite eculizumab treatment. A comprehensive complement analysis together with drug monitoring is required to distinguish mode of complement activation and efficacy of eculizumab treatment in distinct renal disorders. Accumulation of the anti-C5 antibody points to the need for a patient-orientated tailored therapy.
Collapse
Affiliation(s)
- C Wehling
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - O Amon
- Department of Pediatric Nephrology, University Hospital Tübingen, Germany
| | - M Bommer
- Department of Hematology and Oncology, ALB FILS Hospital Göppingen, Germany
| | - B Hoppe
- Department of Pediatric Nephrology, University Hospital Bonn, Germany
| | - K Kentouche
- Department of Pediatric Immunology, University Hospital Jena, Germany
| | - G Schalk
- Department of Pediatric Nephrology, University Children's Hospital Zurich, Switzerland
| | - R Weimer
- Department of Internal Medicine, University of Giessen, Germany
| | - M Wiesener
- Department of Nephrology and Hypertension, University Hospital Erlangen, Germany
| | - B Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - B Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Germany
| | - R Büscher
- Department of Pediatric Nephrology, University Hospital Essen, Germany
| | - H Fehrenbach
- Department of Pediatric Nephrology, Hospital Memmingen, Germany
| | - Ö-N Gök
- Department of Internal Medicine IV, University Hospital Freiburg, Germany
| | - M Kirschfink
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
15
|
Cohen D, Rijnink EC, Nabuurs RJA, Steup-Beekman GM, Versluis MJ, Emmer BJ, Zandbergen M, van Buchem MA, Allaart CF, Wolterbeek R, Bruijn JA, van Duinen SG, Huizinga TWJ, Bajema IM. Brain histopathology in patients with systemic lupus erythematosus: identification of lesions associated with clinical neuropsychiatric lupus syndromes and the role of complement. Rheumatology (Oxford) 2016; 56:77-86. [DOI: 10.1093/rheumatology/kew341] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 08/18/2016] [Indexed: 11/12/2022] Open
|
16
|
Off-label use of the expensive orphan drug eculizumab in France 2009-2013 and the impact of literature: focus on the transplantation field. Eur J Clin Pharmacol 2016; 72:737-46. [PMID: 26915814 DOI: 10.1007/s00228-016-2027-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/11/2016] [Indexed: 12/26/2022]
Abstract
PURPOSE The orphan drug eculizumab (Soliris ®) is one of the most expensive in the world and based on expenditures is classed among the highest in France, a scenario suggestive of off-label use. Given its pharmacological properties, it is likely to be used in organ transplantation. Our purposes were to describe the consumption trends of eculizumab for off-label indications overall and in the organ transplantation field and to assess the impact of publications on the latter use. METHODS We carried out a temporal ecological study within the French national hospitalization database (PMSI). First, the trend of eculizumab consumption (2009-2013) was compared to our estimate of the maximum on-label consumption (overall and for transplantation). Second, we evaluated the impact of the publications supporting the effectiveness of eculizumab in the transplantation field on temporal trends of eculizumab consumption. RESULTS Eculizumab total consumption exceeded our estimate of the maximum on-label consumption since the end of 2011 and increased until the end of the study. The off-label consumption represented at least 50 % of the total consumption. The off-label consumption in organ transplantation also increased since 2011. The amount of publications grew through the study period, but overall, the evidence level remained low. Statistically, publications were neither associated with the drug consumption for transplantation in the long term nor in the short term. CONCLUSION Eculizumab started being notably used for off-label indications in France since the end of 2011, and this use increased until the end of the study. We found only low-level evidence concerning the off-label use of eculizumab in the transplantation field through the studied period.
Collapse
|
17
|
Mallett A, Hughes P, Szer J, Tuckfield A, Van Eps C, Cambell SB, Hawley C, Burke J, Kausman J, Hewitt I, Parnham A, Ford S, Isbel N. Atypical haemolytic uraemic syndrome treated with the complement inhibitor eculizumab: the experience of the Australian compassionate access cohort. Intern Med J 2015; 45:1054-65. [DOI: 10.1111/imj.12864] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 07/19/2015] [Indexed: 02/03/2023]
Affiliation(s)
- A. Mallett
- Department of Renal Medicine; Royal Brisbane and Women's Hospital; Brisbane Queensland Australia
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
| | - P. Hughes
- Department of Nephrology; Royal Melbourne Hospital; Melbourne Victoria Australia
| | - J. Szer
- Department of Clinical Haematology and BMT Service; Royal Melbourne Hospital; Melbourne Victoria Australia
| | - A. Tuckfield
- Department of Clinical Haematology and BMT Service; Royal Melbourne Hospital; Melbourne Victoria Australia
| | - C. Van Eps
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Queensland Australia
| | - S. B. Cambell
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Queensland Australia
| | - C. Hawley
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Queensland Australia
| | - J. Burke
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Queensland Australia
| | - J. Kausman
- Department of Nephrology; The Royal Children's Hospital Melbourne; Melbourne Victoria Australia
| | - I. Hewitt
- Department of Nephrology; Princess Margaret Hospital for Children; Perth Western Australia Australia
| | - A. Parnham
- Department of Nephrology; Gold Coast Hospital; Gold Coast Queensland Australia
| | - S. Ford
- Department of Nephrology; Monash Medical Centre; Melbourne Victoria Australia
| | - N. Isbel
- Centre for Kidney Disease Research; Centre for Chronic Disease; CKD.QLD; School of Medicine; University of Queensland; Brisbane Queensland Australia
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Queensland Australia
| |
Collapse
|
18
|
Sharif‐Paghaleh E, Yap ML, Meader LL, Chuamsaamarkkee K, Kampmeier F, Badar A, Smith RA, Sacks S, Mullen GE. Noninvasive Imaging of Activated Complement in Ischemia-Reperfusion Injury Post-Cardiac Transplant. Am J Transplant 2015; 15:2483-90. [PMID: 25906673 PMCID: PMC4654255 DOI: 10.1111/ajt.13299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/24/2015] [Accepted: 02/28/2015] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury (IRI) is inevitable in solid organ transplantation, due to the transplanted organ being ischemic for prolonged periods prior to transplantation followed by reperfusion. The complement molecule C3 is present in the circulation and is also synthesized by tissue parenchyma in early response to IRI and the final stable fragment of activated C3, C3d, can be detected on injured tissue for several days post-IRI. Complement activation post-IRI was monitored noninvasively by single photon emission computed tomography (SPECT) and CT using (99m) Tc-recombinant complement receptor 2 ((99m) Tc-rCR2) in murine models of cardiac transplantation following the induction of IRI and compared to (99m) Tc-rCR2 in C3(-/-) mice or with the irrelevant protein (99m) Tc-prostate-specific membrane antigen antibody fragment (PSMA). Significant uptake with (99m) Tc-rCR2 was observed as compared to C3(-/-) or (99m) Tc-PSMA. In addition, the transplanted heart to muscle ratio of (99m) Tc-rCR2 was significantly higher than (99m) Tc-PSMA or C3(-/-) . The results were confirmed by histology and autoradiography. (99m) Tc-rCR2 can be used for noninvasive detection of activated complement and in future may be used to quantify the severity of transplant damage due to complement activation postreperfusion.
Collapse
Affiliation(s)
- E. Sharif‐Paghaleh
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland,MRC Centre for TransplantationKing's College LondonLondonEngland,Department of ImmunologyFaculty of MedicineTehran University of Medical SciencesTehranIran
| | - M. L. Yap
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - L. L. Meader
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - K. Chuamsaamarkkee
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - F. Kampmeier
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - A. Badar
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - R. A. Smith
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - S. Sacks
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - G. E. Mullen
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland,MRC Centre for TransplantationKing's College LondonLondonEngland
| |
Collapse
|
19
|
Chua JS, Baelde HJ, Zandbergen M, Wilhelmus S, van Es LA, de Fijter JW, Bruijn JA, Bajema IM, Cohen D. Complement Factor C4d Is a Common Denominator in Thrombotic Microangiopathy. J Am Soc Nephrol 2015; 26:2239-47. [PMID: 25573909 PMCID: PMC4552108 DOI: 10.1681/asn.2014050429] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 11/10/2014] [Indexed: 12/30/2022] Open
Abstract
Complement activation has a major role in thrombotic microangiopathy (TMA), a disorder that can occur in a variety of clinical conditions. Promising results of recent trials with terminal complement-inhibiting drugs call for biomarkers identifying patients who might benefit from this treatment. The primary aim of this study was to determine the prevalence and localization of complement factor C4d in kidneys of patients with TMA. The secondary aims were to determine which complement pathways lead to C4d deposition and to determine whether complement activation results in deposition of the terminal complement complex. We examined 42 renal sections with histologically confirmed TMA obtained from a heterogeneous patient group. Deposits of C4d, mannose-binding lectin, C1q, IgM, and C5b-9 were scored in the glomeruli, peritubular capillaries, and arterioles. Notably, C4d deposits were present in 88.1% of TMA cases, and the various clinical conditions had distinct staining patterns within the various compartments of the renal vasculature. Classical pathway activation was observed in 90.5% of TMA cases. C5b-9 deposits were present in 78.6% of TMA cases and in 39.6% of controls (n=53), but the staining pattern differed between cases and controls. In conclusion, C4d is a common finding in TMA, regardless of the underlying clinical condition. Moreover, C5b-9 was present in >75% of the TMA samples, suggesting that terminal complement inhibitors may have a beneficial effect in these patients. C4d and C5b-9 should be investigated as possible diagnostic biomarkers in the clinical work-up of patients suspected of having complement-mediated TMA.
Collapse
Affiliation(s)
| | | | | | | | | | - Johan W de Fijter
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
20
|
Thomas KA, Valenzuela NM, Gjertson D, Mulder A, Fishbein MC, Parry GC, Panicker S, Reed EF. An Anti-C1s Monoclonal, TNT003, Inhibits Complement Activation Induced by Antibodies Against HLA. Am J Transplant 2015; 15:2037-49. [PMID: 25904443 PMCID: PMC4654252 DOI: 10.1111/ajt.13273] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/10/2015] [Accepted: 02/17/2015] [Indexed: 01/25/2023]
Abstract
Antibody-mediated rejection (AMR) of solid organ transplants (SOT) is characterized by damage triggered by donor-specific antibodies (DSA) binding donor Class I and II HLA (HLA-I and HLA-II) expressed on endothelial cells. While F(ab')2 portions of DSA cause cellular activation and proliferation, Fc regions activate the classical complement cascade, resulting in complement deposition and leukocyte recruitment, both hallmark features of AMR. We characterized the ability of an anti-C1s monoclonal antibody, TNT003, to inhibit HLA antibody (HLA-Ab)-induced complement activation. Complement deposition induced by HLA-Ab was evaluated using novel cell- and bead-based assays. Human aortic endothelial cells (HAEC) were cultured with HLA-Ab and human complement; production of activated complement proteins was measured by flow cytometry. Additionally, C3d deposition was measured on single antigen beads (SAB) mixed with HLA-Ab and human complement. TNT003 inhibited HLA-Ab mediated complement deposition on HAEC in a concentration-dependent manner; C3a, C4a and C5a anaphylatoxin production was also diminished by TNT003. Finally, TNT003 blocked C3d deposition induced by Class I (HLAI-Ab)- and Class II (HLAII-Ab)-specific antibodies on SAB. These data suggest TNT003 may be useful for modulating the effects of DSA, as TNT003 inhibits complement deposition and split product formation generated by HLA-I/II-Ab in vitro.
Collapse
Affiliation(s)
- K A Thomas
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos Angeles, CA
| | - N M Valenzuela
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos Angeles, CA
| | - D Gjertson
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos Angeles, CA
| | - A Mulder
- Department of Immunohematology and Blood Transfusion, Leiden University Medical CenterLeiden, the Netherlands
| | - M C Fishbein
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos Angeles, CA
| | - G C Parry
- True North Therapeutics, Inc.South San Francisco, CA
| | - S Panicker
- True North Therapeutics, Inc.South San Francisco, CA
| | - E F Reed
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos Angeles, CA,*Corresponding author: Elaine F. Reed,
| |
Collapse
|
21
|
James A, Mannon RB. The Cost of Transplant Immunosuppressant Therapy: Is This Sustainable? CURRENT TRANSPLANTATION REPORTS 2015; 2:113-121. [PMID: 26236578 PMCID: PMC4520417 DOI: 10.1007/s40472-015-0052-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A solid organ transplant is life-saving therapy that engenders the use of immunosuppressive medications for the lifetime of the transplanted organ and its recipient. Conventional therapy includes both induction therapy (a biologic that is infused peri-operatively) followed by maintenance therapy. The cost of these medications is a constant concern and the advent of generics has brought this cost down modestly. For those lacking long term insurance coverage, this may be a significant out of pocket expense that is not affordable. Moreover, transplant Centers are managing higher risk transplant recipients that require more complex induction regimens and longer term use of such biologic agents in the context of desensitization or abrogation of de novo antibody mediated injury. While in kidney transplantation, Medicare part B covers three years of medication, there is frequent non-adherence due to cost after that time-point. The impact of the Affordable Care Act remains uncertain at this time. Finally the pipeline of new therapies is limited due to the cost of development of a drug, the inherent cost of clinical studies, and lack of defined endpoints for newer therapies in high risk patients. These new therapies are of high value to the community but will contribute additional burden to current drug costs.
Collapse
Affiliation(s)
- Alexandra James
- Department of Pharmacy, University of Alabama at Birmingham, Birmingham, AL
| | - Roslyn B. Mannon
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
22
|
Pouliquen E, Koenig A, Chen CC, Sicard A, Rabeyrin M, Morelon E, Dubois V, Thaunat O. Recent advances in renal transplantation: antibody-mediated rejection takes center stage. F1000PRIME REPORTS 2015; 7:51. [PMID: 26097724 PMCID: PMC4447042 DOI: 10.12703/p7-51] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Overlooked for decades, antibodies have taken center stage in renal transplantation and are now widely recognized as the first cause of allograft failure. Diagnosis of antibody-mediated rejection has considerably improved with identification of antibody-mediated lesions in graft biopsies and advances made in the detection of circulating donor-specific antibodies. Unfortunately, this progress has not yet translated into better outcomes for patients. Indeed, in the absence of a drug able to suppress antibody generation by plasma cells, available therapies can only slow down graft destruction. This review provides an overview of the current knowledge of antibody-mediated rejection and discusses future interesting research directions.
Collapse
Affiliation(s)
- Eric Pouliquen
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| | - Alice Koenig
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| | - Chien Chia Chen
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| | - Antoine Sicard
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| | - Maud Rabeyrin
- Laboratoire d‘anatomopathologie, Hospices Civils de Lyon, Hôpital Edouard HerriotLyonFrance
| | - Emmanuel Morelon
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| | - Valérie Dubois
- Laboratoire d‘Histocompatibilité, Etablissement Français du SangLyonFrance
| | - Olivier Thaunat
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie CliniqueLyonFrance
- Institut National de la Santé et de la Recherche MédicaleU1111, LyonFrance
- Unité de Formation et de Recherche Lyon Est, Université de LyonLyonFrance
| |
Collapse
|
23
|
Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2-year extensions of phase 2 studies. Kidney Int 2015; 87:1061-73. [PMID: 25651368 PMCID: PMC4424817 DOI: 10.1038/ki.2014.423] [Citation(s) in RCA: 288] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 09/30/2014] [Accepted: 10/30/2014] [Indexed: 12/11/2022]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare, possibly life-threatening disease characterized by platelet activation, hemolysis and thrombotic microangiopathy (TMA) leading to renal and other end-organ damage. We originally conducted two phase 2 studies (26 weeks and 1 year) evaluating eculizumab, a terminal complement inhibitor, in patients with progressing TMA (trial 1) and those with long duration of aHUS and chronic kidney disease (trial 2). The current analysis assessed outcomes after 2 years (median eculizumab exposure 100 and 114 weeks, respectively). At all scheduled time points, eculizumab inhibited terminal complement activity. In trial 1 with 17 patients, the platelet count was significantly improved from baseline, and hematologic normalization was achieved in 13 patients at week 26, and in 15 patients at both 1 and 2 years. The estimated glomerular filtration rate (eGFR) was significantly improved compared with baseline and year 1. In trial 2 with 20 patients, TMA event-free status was achieved by 16 patients at week 26, 17 patients at year 1, and 19 patients at year 2. Criteria for hematologic normalization were met by 18 patients at each time point. Improvement of 15 ml/min per 1.73 m2 or more in eGFR was achieved by 1 patient at week 26, 3 patients at 1 year, and 8 patients at 2 years. The mean change in eGFR was not significant compared with baseline, week 26, or year 1. Eculizumab was well tolerated, with no new safety concerns or meningococcal infections. Thus, a 2-year analysis found that the earlier clinical benefits achieved by eculizumab treatment of aHUS were maintained at 2 years of follow-up.
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Much research in transplantation focuses on treatments for rejection and induction of tolerance. Recent evidence has shown that initial inflammation induced by innate immune effectors after transplantation has a key role in modulating adaptive immune responses that cause organ rejection. Here, we describe the role of the innate immune system, particularly the complement activation pathways, and how they influence adaptive immune responses post-transplantation and current strategies, which are under development to block these innate pathways. RECENT FINDINGS Anaphylatoxins and their respective receptors are proving to be important in T-cell-mediated immunity and make attractive targets for therapies designed to promote tolerance in solid organ transplantation. Additionally, regulators of complement activation are currently being tested in clinical trials, with improvements in drug delivery. SUMMARY Preventing ischaemia-reperfusion injury in transplanted organs significantly reduces immune activation and promotes graft survival. Research into the mechanisms of complement activation in both native organ ischaemia and transplantation models detail emerging roles for complement intermediates that can serve as targets for intervention, with the aim of reducing early post-transplant inflammation, reducing the intensity of immunosuppressive regimens, leading to prolonged graft survival.
Collapse
|
25
|
Recent trials in immunosuppression and their consequences for current therapy. Curr Opin Organ Transplant 2015; 19:387-94. [PMID: 24905020 DOI: 10.1097/mot.0000000000000093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW Although the scarcity of clinical trials with de-novo immunosuppression has been typical over the last 2 years, several attempts have been made in drug conversion, dosing optimization, and bioequivalence. On the basis of recent clinical and animal studies, future directions of management and treatment are outlined. RECENT FINDINGS Studies with new tacrolimus formulations showed better bioavailability and lower doses, which might translate into less toxicity. The long-term results of studies with costimulation blockade confirmed their safety and efficacy. Calcineurin inhibitor (CNI)-free regimens based on mTOR inhibitors were shown to be associated with increased risk of the humoral response. Therefore, ongoing trials are predominantly designed to minimize calcineurin inhibitor dose only. Biologics, such as B-cell-specific agents (bortezomib and rituximab) and complement inhibitors (eculizumab) used to treat antibody-mediated rejection, recurrence of glomerulonephritis, are shifted to more preventive applications. The pretransplant quantification of alloreactive memory/effector T cell response may help to better stratify a patient's immunologic risk and allow for drug minimization. SUMMARY Despite clinical trials with innovative protocols with already established agents, tacrolimus-based and induction-based protocols have been shown to be the mainstay of immunosuppressive regimens. In the future, research aims to focus on biomarker-driven immunosuppression and cell therapy approaches.
Collapse
|
26
|
Danobeitia JS, Djamali A, Fernandez LA. The role of complement in the pathogenesis of renal ischemia-reperfusion injury and fibrosis. FIBROGENESIS & TISSUE REPAIR 2014. [PMID: 25383094 DOI: 10.1186/1755‐1536‐7‐16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The complement system is a major component of innate immunity and has been commonly identified as a central element in host defense, clearance of immune complexes, and tissue homeostasis. After ischemia-reperfusion injury (IRI), the complement system is activated by endogenous ligands that trigger proteolytic cleavage of complement components via the classical, lectin and/or alternative pathway. The result is the formation of terminal complement components C3a, C5a, and the membrane attack complex (C5b-9 or MAC), all of which play pivotal roles in the amplification of the inflammatory response, chemotaxis, neutrophil/monocyte recruitment and activation, and direct tubular cell injury. However, recent evidence suggests that complement activity transcends innate host defense and there is increasing data suggesting complement as a regulator in processes such as allo-immunity, stem cell differentiation, tissue repair, and progression to fibrosis. In this review, we discuss recent advances addressing the role of complement as a regulator of IRI and renal fibrosis after organ donation for transplantation. We will also briefly discuss currently approved therapies that target complement activity in kidney ischemia-reperfusion and transplantation.
Collapse
Affiliation(s)
- Juan S Danobeitia
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison School of Medicine and Public Health, H4/782 Clinical Science Center, 600 Highland Avenue, 53792 Madison, WI, USA
| | - Arjang Djamali
- Department of Medicine, Division of Nephrology, University of Wisconsin- Madison School of Medicine and Public Health, UW Medical Foundation Centennial Building, 1685 Highland Avenue, 53705 Madison, WI, USA
| | - Luis A Fernandez
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison School of Medicine and Public Health, H4/782 Clinical Science Center, 600 Highland Avenue, 53792 Madison, WI, USA
| |
Collapse
|
27
|
Danobeitia JS, Djamali A, Fernandez LA. The role of complement in the pathogenesis of renal ischemia-reperfusion injury and fibrosis. FIBROGENESIS & TISSUE REPAIR 2014; 7:16. [PMID: 25383094 PMCID: PMC4224961 DOI: 10.1186/1755-1536-7-16] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 09/17/2014] [Indexed: 01/04/2023]
Abstract
The complement system is a major component of innate immunity and has been commonly identified as a central element in host defense, clearance of immune complexes, and tissue homeostasis. After ischemia-reperfusion injury (IRI), the complement system is activated by endogenous ligands that trigger proteolytic cleavage of complement components via the classical, lectin and/or alternative pathway. The result is the formation of terminal complement components C3a, C5a, and the membrane attack complex (C5b-9 or MAC), all of which play pivotal roles in the amplification of the inflammatory response, chemotaxis, neutrophil/monocyte recruitment and activation, and direct tubular cell injury. However, recent evidence suggests that complement activity transcends innate host defense and there is increasing data suggesting complement as a regulator in processes such as allo-immunity, stem cell differentiation, tissue repair, and progression to fibrosis. In this review, we discuss recent advances addressing the role of complement as a regulator of IRI and renal fibrosis after organ donation for transplantation. We will also briefly discuss currently approved therapies that target complement activity in kidney ischemia-reperfusion and transplantation.
Collapse
Affiliation(s)
- Juan S Danobeitia
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison School of Medicine and Public Health, H4/782 Clinical Science Center, 600 Highland Avenue, 53792 Madison, WI, USA
| | - Arjang Djamali
- Department of Medicine, Division of Nephrology, University of Wisconsin- Madison School of Medicine and Public Health, UW Medical Foundation Centennial Building, 1685 Highland Avenue, 53705 Madison, WI, USA
| | - Luis A Fernandez
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison School of Medicine and Public Health, H4/782 Clinical Science Center, 600 Highland Avenue, 53792 Madison, WI, USA
| |
Collapse
|
28
|
Pankewycz O, Soliman K, Laftavi MR. The increasing clinical importance of alloantibodies in kidney transplantation. Immunol Invest 2014; 43:775-89. [DOI: 10.3109/08820139.2014.910016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
29
|
Wehling C, Kirschfink M. Tailored eculizumab regimen for patients with atypical hemolytic uremic syndrome: requirement for comprehensive complement analysis. J Thromb Haemost 2014; 12:1437-9. [PMID: 24942998 DOI: 10.1111/jth.12639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Indexed: 12/01/2022]
Affiliation(s)
- C Wehling
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
30
|
Chua S, Wong G, Lim WH. The importance of genetic mutation screening to determine retransplantation following failed kidney allograft from recurrent atypical haemolytic ureamic syndrome. BMJ Case Rep 2014; 2014:bcr-2013-202875. [PMID: 24671321 DOI: 10.1136/bcr-2013-202875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We report the case of a patient with familial atypical haemolytic uraemic syndrome (aHUS) who underwent successful retransplantation 30 months following his failed first kidney allograft from recurrent aHUS. He achieved excellent graft function (creatinine 90 μmol/L), with no evidence of disease recurrence on standard maintenance immunosuppression 9 months after his second deceased donor kidney transplantation. Genetic mutation testing was not available prior to first transplant but screening prior to retransplant identified the patient as having a newly discovered mutation, c.T3566A, within exon 23 of the complement factor H (CFH) gene. Currently, public financing and subsidisation for eculizumab, a costly but effect complement (C5) inhibitor for the treatment of aHUS is not available in Australia. The decision for retransplantation must balance between the risk of disease recurrence and greater risk of death on dialysis. The absence of a more severe CFH genotype assisted in the decision for retransplantation and suggests the importance of genetic mutation screening in order to stratify the risk of disease recurrence and graft loss versus the benefit of transplantation.
Collapse
Affiliation(s)
- Samantha Chua
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | | | | |
Collapse
|
31
|
Zaza G, Tomei P, Granata S, Boschiero L, Lupo A. Monoclonal antibody therapy and renal transplantation: focus on adverse effects. Toxins (Basel) 2014; 6:869-91. [PMID: 24590384 PMCID: PMC3968366 DOI: 10.3390/toxins6030869] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/07/2014] [Accepted: 02/21/2014] [Indexed: 02/06/2023] Open
Abstract
A series of monoclonal antibodies (mAbs) are commonly utilized in renal transplantation as induction therapy (a period of intense immunosuppression immediately before and following the implant of the allograft), to treat steroid-resistant acute rejections, to decrease the incidence and mitigate effects of delayed graft function, and to allow immunosuppressive minimization. Additionally, in the last few years, their use has been proposed for the treatment of chronic antibody-mediated rejection, a major cause of late renal allograft loss. Although the exact mechanism of immunosuppression and allograft tolerance with any of the currently used induction agents is not completely defined, the majority of these medications are targeted against specific CD proteins on the T or B cells surface (e.g., CD3, CD25, CD52). Moreover, some of them have different mechanisms of action. In particular, eculizumab, interrupting the complement pathway, is a new promising treatment tool for acute graft complications and for post-transplant hemolytic uremic syndrome. While it is clear their utility in renal transplantation, it is also unquestionable that by using these highly potent immunosuppressive agents, the body loses much of its innate ability to mount an adequate immune response, thereby increasing the risk of severe adverse effects (e.g., infections, malignancies, haematological complications). Therefore, it is extremely important for clinicians involved in renal transplantation to know the potential side effects of monoclonal antibodies in order to plan a correct therapeutic strategy minimizing/avoiding the onset and development of severe clinical complications.
Collapse
Affiliation(s)
- Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, Verona 37126, Italy.
| | - Paola Tomei
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, Verona 37126, Italy.
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, Verona 37126, Italy.
| | - Luigino Boschiero
- First Surgical Clinic, Kidney Transplantation Center, University-Hospital of Verona, Piazzale A. Stefani 1, Verona 37126, Italy.
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, Verona 37126, Italy.
| |
Collapse
|
32
|
Lonze BE, Zachary AA, Magro CM, Desai NM, Orandi BJ, Dagher NN, Singer AL, Carter-Monroe N, Nazarian SM, Segev DL, Streiff MB, Montgomery RA. Eculizumab prevents recurrent antiphospholipid antibody syndrome and enables successful renal transplantation. Am J Transplant 2014; 14:459-65. [PMID: 24400968 DOI: 10.1111/ajt.12540] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/16/2013] [Accepted: 09/30/2013] [Indexed: 01/25/2023]
Abstract
Renal transplantation in patients with antiphospholipid antibodies has historically proven challenging due to increased risk for thrombosis and allograft failure. This is especially true for patients with antiphospholipid antibody syndrome (APS) and its rare subtype, the catastrophic antiphospholipid antibody syndrome (CAPS). Since a critical mechanism of thrombosis in APS/CAPS is one mediated by complement activation, we hypothesized that preemptive treatment with the terminal complement inhibitor, eculizumab, would reduce the extent of vascular injury and thrombosis, enabling renal transplantation for patients in whom it would otherwise be contraindicated. Three patients with APS, two with a history of CAPS, were treated with continuous systemic anticoagulation together with eculizumab prior to and following live donor renal transplantation. Two patients were also sensitized to human leukocyte antigens (HLA) and required plasmapheresis for reduction of donor-specific antibodies. After follow-up ranging from 4 months to 4 years, all patients have functioning renal allografts. No systemic thrombotic events or early graft losses were observed. While the appropriate duration of treatment remains to be determined, this case series suggests that complement inhibitors such as eculizumab may prove to be effective in preventing the recurrence of APS after renal transplantation.
Collapse
Affiliation(s)
- B E Lonze
- Division of Transplant Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
|
35
|
Abnormalities in the alternative pathway of complement in children with hematopoietic stem cell transplant-associated thrombotic microangiopathy. Blood 2013; 122:2003-7. [PMID: 23814021 DOI: 10.1182/blood-2013-05-501445] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT)-associated thrombotic microangiopathy (TMA) is a complication that occurs in 25% to 35% of HSCT recipients and shares histomorphologic similarities with hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). The hallmark of all thrombotic microangiopathies is vascular endothelial cell injury of various origins, resulting in microangiopathic hemolytic anemia, platelet consumption, fibrin deposition in the microcirculation, and tissue damage. Although significant advances have been made in understanding the pathogenesis of other thrombotic microangiopathies, post-HSCT TMA remains poorly understood. We report an analysis of the complement alternative pathway, which has recently been linked to the pathogenesis of both the Shiga toxin mediated and the atypical forms of HUS, with a focus on genetic variations in the complement Factor H (CFH) gene cluster and CFH autoantibodies in six children with post-HSCT TMA. We identified a high prevalence of deletions in CFH-related genes 3 and 1 (delCFHR3-CFHR1) and CFH autoantibodies in these patients with HSCT-TMA. Conversely, CFH autoantibodies were not detected in 18 children undergoing HSCT who did not develop TMA. Our observations suggest that complement alternative pathway dysregulation may be involved in the pathogenesis of post-HSCT TMA. These findings shed light on a novel mechanism of endothelial injury in transplant-TMA and may therefore guide the development of targeted treatment interventions.
Collapse
|
36
|
Roversi P, Ryffel B, Togbe D, Maillet I, Teixeira M, Ahmat N, Paesen GC, Lissina O, Boland W, Ploss K, Caesar JJE, Leonhartsberger S, Lea SM, Nunn MA. Bifunctional lipocalin ameliorates murine immune complex-induced acute lung injury. J Biol Chem 2013; 288:18789-802. [PMID: 23625922 PMCID: PMC3696655 DOI: 10.1074/jbc.m112.420331] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Molecules that simultaneously inhibit independent or co-dependent proinflammatory pathways may have advantages over conventional monotherapeutics. OmCI is a bifunctional protein derived from blood-feeding ticks that specifically prevents complement (C)-mediated C5 activation and also sequesters leukotriene B4 (LTB4) within an internal binding pocket. Here, we examined the effect of LTB4 binding on OmCI structure and function and investigated the relative importance of C-mediated C5 activation and LTB4 in a mouse model of immune complex-induced acute lung injury (IC-ALI). We describe two crystal structures of bacterially expressed OmCI: one binding a C16 fatty acid and the other binding LTB4 (C20). We show that the C5 and LTB4 binding activities of the molecule are independent of each other and that OmCI is a potent inhibitor of experimental IC-ALI, equally dependent on both C5 inhibition and LTB4 binding for full activity. The data highlight the importance of LTB4 in IC-ALI and activation of C5 by the complement pathway C5 convertase rather than by non-C proteases. The findings suggest that dual inhibition of C5 and LTB4 may be useful for treatment of human immune complex-dependent diseases.
Collapse
Affiliation(s)
- Pietro Roversi
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|