Editorial Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Jun 26, 2015; 5(2): 20-25
Published online Jun 26, 2015. doi: 10.5662/wjm.v5.i2.20
Costimulatory blockade: A novel approach to the treatment of glomerular disease?
Pasquale Esposito, Teresa Rampino, Antonio Dal Canton, Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, 27100 Pavia, Italy
Author contributions: Esposito P and Rampino T have contributed to this paper in writing the article and in reviewing the literature; Dal Canton A contributed in the writing and final approval of the article.
Conflict-of-interest: None.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Pasquale Esposito, MD, PhD, Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Piazzale Golgi 19, 27100 Pavia, Italy. pasqualeesposito@hotmail.com
Telephone: +39-382-503883 Fax: +39-382-503883
Received: January 28, 2015
Peer-review started: January 29, 2015
First decision: March 20, 2015
Revised: April 1, 2015
Accepted: May 16, 2015
Article in press: May 18, 2015
Published online: June 26, 2015

Abstract

Costimulatory pathways (Cluster of differentiation 28, tumor necrosis factor-related, adhesion and T Cell Ig- and mucin-domain molecules) regulating the interactions between receptors on the T cells and their ligands expressed on several cell types, have a key role in controlling many immunological and non immunological processes. Indeed, accumulating evidence indicate that these molecules are involved in the pathogenesis of numerous conditions, such as allograft rejection, atherosclerosis, rheumatoid arthritis, psoriasis and renal diseases, including glomerulonephritis. Primary or secondary (i.e., associated with infections, drugs or systemic diseases, such as systemic lupus erythematosus, diabetes, etc.) glomerulonephritis represent a group of heterogeneous diseases with different pathogenic mechanisms. Since costimulatory molecules, in particular CD80 and CD40, have been found to be expressed on podocytes in the course of different experimental and clinical glomerulonephritis, costimulation has been thought as a new therapeutic target for patients with glomerular diseases. However, although experimental data suggested that the blockade of costimulatory pathways is effective and safe in the prevention and treatment of glomerular diseases, clinical trials reported contrasting results. So, at this moment, there is not a strong evidence for the general use of costimulatory blockade as an alternative treatment strategy in patients with primary or secondary glomerulonephritis. Here, we critically discuss the current data and the main issues regarding the development of this innovative therapeutic approach.

Key Words: Costimulation, Glomerulonephritis, Cluster of differentiation 80, Cytotoxic T-lymphocyte-associated antigen-4, Lupus nephritis, Abatacept, Proteinuria, Podocytes

Core tip: Glomerulonephritis refer to a group of renal disorders, primary or secondary to infections, drugs or systemic diseases, characterized by inflammation within the glomerulus. Among glomerular diseases there is a great clinical, histological and prognostic heterogeneity and several different pathogenetic mechanisms have been implied. Current standard treatments include steroids and cytotoxic agents, which present important side effects and an unsatisfactory remission rate. Therefore, experimental and clinical research is addressed to the development of alternative therapies. Here, we critically discuss new therapeutic opportunities provided by the use of agents acting on the modulation of costimulatory pathways.



COSTIMULATORY PATHWAYS

Costimulatory pathways regulating the interactions between receptors on the T cells and their ligands expressed on several cell types (including immunocompetent cells, fibroblasts, endothelial cells, etc.) play a crucial role in the modulation of immunological and non-immunological processes[1].

In particular, costimulation is essential for the full activation of naïve T cells after antigen-specific recognition, and without costimulation the T cell-antigen interaction results in anergy[2].

Different costimulatory families [Cluster of differentiation 28 (CD28), tumor necrosis factor (TNF)-related, adhesion and T Cell Ig- and mucin-domain (TIMs) molecules], characterized by structural and functional analogies, have been described. These molecules can interact with each other either up- or down-regulating T cell activation[3] (Table 1).

Table 1 Immunomodulatory effects of costimulation pathways.
FamilyLigandReceptorEffects on immune cells
CD28CD80 (B7.1)/CD86 (B7.2)CD28+
CTLA-4-
ICOSLICOS+
PDL1PD-1-
TNF-relatedCD40CD40L (CD154)+
OX-40OX-40L+
Adhesion moleculesICAM-1LFA-1+
TIMTIM4/9TIM1/3+/-

Among the identified costimulatory molecules, the best characterized are the CD28:B7 and the TNF-related families. The CD28:B7 family includes the following receptor-ligand pairs: CD28/CTLA4:CD80/CD86, induced costimulatory molecules (ICOS:ICOSL) and the programmed death-1 pathway (PD-1:PD-L1/PD-L2)[4]. CD28 is a disulfide-bound molecule that belongs to the immunoglobulin superfamily and is constitutively expressed on T cells[5].

Its interaction with CD80 (B7.1) and CD86 (B7.2), expressed on the surface of antigen-presenting cells (APCs), leads to the full activation of T cells[6]. Conversely cytotoxic T-lymphocyte-associated antigen-4 (CTLA4), a structural homologous of CD28 with a higher avidity for CD80 and CD86, acts as a negative regulator of T cells[7].

TNF superfamily comprises: CD40:CD40L, OX40:OX40L, CD30:CD30L, CD27:CD70, CD137:CD137L, etc. CD40 is mainly expressed on B-cells, but also on monocytes, dendritic cells, endothelial cells, smooth muscle cells and fibroblasts[8]. The engagement of CD40 with its ligand, CD40L (CD154), leads to B cell expansion and differentiation and it is decisive in the regulation of APCs and dendritic cells functions[9]. It is important to underline that costimulatory molecules, expressed by a broad variety of cells, seem to be involved in the pathogenesis of numerous conditions, such as atherosclerosis, rheumatoid arthritis, psoriasis and renal diseases, including allograft rejection and glomerulonephritis[10-14].

The insights regarding the contribution the costimulatory molecules in these conditions has not only allowed elucidating important regulatory mechanisms, but has also provided novel targets for therapeutic interventions[15].

COSTIMULATION AND GLOMERULONEPHRITIS

Glomerulonephritis refer to a group of renal disorders, primary or secondary to infections (human immunodeficiency virus, hepatitis C virus, etc.), drugs and systemic diseases (for example, systemic lupus erithematosus-SLE, cancer and diabetes), characterized by inflammation within the glomerulus[16].

Among glomerular diseases there is a great clinical, histological and prognostic heterogeneity and several different pathogenic mechanisms are implied, including podocyte damage, immunoglobulin deposition and immune cell infiltration[17]. During the last years growing evidence suggest a role for costimulatory molecules also in this specific setting.

In particular, CD80 expression has been detected in podocytes, which integrity is essential to maintain a regular glomerular function[18].

Indeed, in experimental models of genetic, drug-induced, immune-mediated and bacterial toxin-induced kidney diseases, CD80 overexpression on podocytes might be harmful for glomerular permeability, disturbing the slit diaphragm and down-regulating podocytes-β1 integrin activation, finally leading to the development of proteinuria and loss of renal function[19,20]. The crucial role of CD28:CD80 pathway in the pathogenesis of glomerular diseases is also confirmed by the evidence that CD80 knockout mice present an attenuated form of proliferative glomerulonephritis, associated with a significant reduction of renal tissue lesions[21].

Moreover, the use of monoclonal antibodies targeting CD28 or CTLA-4 was effective in treating and preventing different forms of experimental nephritis, including lupus-like nephritis[22]. Interestingly, similar results were also found in human glomerulonephritis. In particular, a significant increase in CD80 podocyte expression and urinary excretion has been reported in patients with minimal change disease (MCD) in relapse compared to those in remission or with focal segmental glomerulosclerosis (FSGS)[23,24]. Similarly, patients with proliferative lupus nephritis present a strong podocyte surface expression of CD80[20].

Beyond CD28:CD80 pathway, also costimulatory molecules of TNF-related family, i.e., CD40:CD154, have been found expressed in renal tissue in the course of both experimental and human glomerular diseases. CD40 was isolated in murine models of proteinuric disease, such as membranous glomerulonephritis, lupus nephritis and necrotizing nephritis[25]. Moreover, glomerular and tubular CD40 expression was up-regulated in human lupus nephritis and in other inflammatory renal diseases, being associated with the presence of CD40L+ mononuclear cells[26]. Furthermore, the inhibition of CD40 pathway through the administration of a CD40-Ig fusion protein or anti-CD40L antibodies prevented the development of proteinuric kidney diseases in mice[27,28].

COSTIMULATORY BLOCKADE AS A NOVEL TREATMENT FOR GLOMERULONEPHRITIS

As a consequence of the role of costimulation in the pathogenesis of several pathological conditions, costimulatory blockade has been thought as a new rational therapeutic approach[29]. Therefore different strategies, mainly based on the design of specific monoclonal antibodies (mAbs) interfering with these critical pathways, have been tested. However, the clinical development of the majority of these new strategies is currently suspended for safety concerns.

This is, for example, the case of anti-CD40L mAb, which although effective in the prevention of glomerular diseases and renal allograft rejection in murine and primate experimental models, significantly increased the occurrence of thromboembolic events[27,30-32]. More severe complications occurred during the development of anti-CD28 mAbs. Indeed, six healthy volunteers enrolled in a phase I clinical trial and treated with a humanized superagonistic anti-CD28 mAb, developed a life-threatening systemic inflammation due to massive cytokine release, determining the complete abandon of this approach[33]. A more promising strategy- the only one that has found clinical applications so far- seems to be the development of CTLA-4 immunoglobulin fusion proteins. These proteins are composed by an extracellular portion of human CTLA4 plus a Fc part of human IgG1, which, binding CD80 and C86 with high avidity, prevent CD28 ligation, acting as potent inhibitors of CD28:CD80/CD86 pathways[34,35]. Abatacept, which has been approved by FDA for the treatment of rheumatoid arthritis in 2005, and its derivate, Belatacept, belong to this category of drugs.

Belatacept has been extensively studied mainly in the experimental and clinical setting of renal transplantation.

Belatacept was evaluated in 2 open-label, randomized, multicenter, controlled phase 3 studies: the Belatacept Evaluation of Nephroprotection and Efficacy as First-line Immunosuppression Trial (BENEFIT) and BENEFIT-EXT (“extended criteria”).

These studies showed that Belatacept was non-inferior to Cyclosporine in terms of patient and graft survival, being associated to a better graft function and a reduced incidence of chronic nephropathy[36,37]. Hence, although the administration of belatacept was not exempt from adverse effects, in 2011 it was approved by Food and Drug Administration (FDA) as the first costimulatory blocker for use in renal transplantation[38].

Regarding the specific setting of primary and secondary glomerulonephritis, instead, only Abatacept has been used in clinical studies with discordant results.

Two recent randomized trials investigated the safety and efficacy of Abatacept in addition to standard treatments in patients with lupus nephritis.

A twelve months blind multicentre trial, performed by Furie et al[39], enrolled 298 patients with active lupus nephritis and proteinuria, randomized to receive corticosteroids and Mycophenolate mofetil in association with Abatacept (30 mg/kg loading for 3 mo, followed by 10 mg/kg), Abatacept (10 mg/kg) or placebo. The authors found that the treatment with Abatacept was associated with a reduction of antiDNA antibody, C3 and C4 levels and proteinuria. However, there were not significant differences in the time to reach a complete response and in the proportion of subjects with confirmed complete response after 52 wk of follow-up among the three groups.

Similar results have been reported by Askanase et al[40] who evaluated the efficacy of Abatacept vs placebo added to a standard treatment regimen with Cyclophosphamide followed by Azathioprine in 134 patients with active lupus nephritis. They also found no significant differences between the groups in terms of number of patients reaching and/or maintaining complete or partial response.

So, even if previous studies reporting the strong expression of podocyte CD80 in human proliferative lupus nephritis appeared promising, the results of these clinical trials have unexpectedly called into question the utility of Abatacept in patients with SLE.

Abatacept has been also studied in patients with primary glomerulonephritis.

In a recent paper, Yu et al[41] tested Abatacept in 5 patients with FSGS (4 with recurrent FSGS after kidney transplantation and 1 with primary FSGS) who presented positive CD80 (B7.1) immunostaining of podocytes in kidney-biopsy specimens.

After treatment with Abatacept all these patients presented a partial or complete remission, expressed as a significant reduction of serum creatinine and/or proteinuria. Interestingly, the authors provided also a rationale for the beneficial effects of Abatacept, demonstrating that the drug in vitro blocks podocyte migration and stabilizes β1-integrin activation in podocytes[41].

Although exciting, these results have been criticized for several important methodological issues[42,43]. First of all, it should be considered that the 4 patients with recurrent FSGS underwent intensive plasmapheresis, aimed to remove putative circulating permeability factors. Thus, it is not possible to recognize if the disease remission was due to this treatment independently of the use of Abatacept. Moreover, subsequent reports arose doubt about the immunostaining techniques used to detect CD80 in renal tissue, highlighting the lack of any negative controls. In particular, Larsen et al[44] tested the presence of CD80 in 60 renal biopsy specimens from patients with different proteinuric glomerular diseases with two immunostaining methods (immunoperoxidase and immunofluorescence). The authors found that for both staining techniques and in all cases, CD80 was undetectable within podocytes. The presence of so contrasting results among experimental and clinical trials raises doubt about the potential role of Abatacept in patients with proteinuric glomerulonephritis[45].

To be thorough, it has to point out that the efficacy of Abatacept in the treatment of MCD has been recently reported in a single case[46].

Considering the overall above reported data, we might infer that, although the podocyte CD80 pathway seems to have an important role in some proteinuric glomerular diseases, clinical results suggest that current therapeutic strategies do not warrant a satisfactory control of glomerulonephritis.

CONCLUSION

The critical analysis of the currently available data suggests some conclusions: (1) costimulatory pathways might be implied in the pathogenesis of glomerulonephritis, especially the forms associated with proteinuria and nephrotic syndrome; (2) the development of drugs targeted to block costimulation is of great potential utility, also considering that the current available therapeutic options are limited[47]; (3) clinical trials have shown insufficient or, at least, contrasting effects of this kind of approach in the achievement of therapeutic targets and disease remission.

So, it appears clear that further molecular, cellular and clinical studies, including the design and evaluation of new drugs and exploration of new pathways, should be performed before considering costimulatory blockade as a valid alternative treatment in the general population of patients with glomerulonephritis.

ACKNOWLEDGMENTS

We thank Dr. Stefania Bianzina for the English editing.

Footnotes

P- Reviewer: El Sherbini MAH, Mabruk M, Radojcic BS S- Editor: Tian YL L- Editor: A E- Editor: Wu HL

References
1.  Bretscher PA. A two-step, two-signal model for the primary activation of precursor helper T cells. Proc Natl Acad Sci USA. 1999;96:185-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 259]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
2.  Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1052]  [Cited by in F6Publishing: 1018]  [Article Influence: 48.5]  [Reference Citation Analysis (0)]
3.  Magee CN, Boenisch O, Najafian N. The role of costimulatory molecules in directing the functional differentiation of alloreactive T helper cells. Am J Transplant. 2012;12:2588-2600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
4.  Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1714]  [Cited by in F6Publishing: 1745]  [Article Influence: 91.8]  [Reference Citation Analysis (0)]
5.  Keir ME, Sharpe AH. The B7/CD28 costimulatory family in autoimmunity. Immunol Rev. 2005;204:128-143.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  McAdam AJ, Schweitzer AN, Sharpe AH. The role of B7 co-stimulation in activation and differentiation of CD4+ and CD8+ T cells. Immunol Rev. 1998;165:231-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 215]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
7.  Alegre ML, Frauwirth KA, Thompson CB. T-cell regulation by CD28 and CTLA-4. Nat Rev Immunol. 2001;1:220-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 606]  [Cited by in F6Publishing: 598]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
8.  Mach F, Schönbeck U, Sukhova GK, Bourcier T, Bonnefoy JY, Pober JS, Libby P. Functional CD40 ligand is expressed on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for CD40-CD40 ligand signaling in atherosclerosis. Proc Natl Acad Sci USA. 1997;94:1931-1936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 554]  [Cited by in F6Publishing: 540]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
9.  van Kooten C, Banchereau J. CD40-CD40 ligand. J Leukoc Biol. 2000;67:2-17.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Andersson J, Libby P, Hansson GK. Adaptive immunity and atherosclerosis. Clin Immunol. 2010;134:33-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 184]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
11.  Esposito P, Dal Canton A. CD40/CD40L and cardiovascular risk in patients on haemodialysis: a role for soluble CD40? Nephrol Dial Transplant. 2011;26:2414-2415; author reply 2414-2415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
12.  Liu MF, Kohsaka H, Sakurai H, Azuma M, Okumura K, Saito I, Miyasaka N. The presence of costimulatory molecules CD86 and CD28 in rheumatoid arthritis synovium. Arthritis Rheum. 1996;39:110-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 58]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
13.  Paukkonen K, Naukkarinen A, Horsmanheimo M. The development of manifest psoriatic lesions is linked with the invasion of CD8 + T cells and CD11c + macrophages into the epidermis. Arch Dermatol Res. 1992;284:375-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
14.  Esposito P, Grosjean F, Rampino T, Libetta C, Gregorini M, Fasoli G, Marchi G, Sileno G, Montagna F, Dal Canton A. Costimulatory pathways in kidney transplantation: pathogenetic role, clinical significance and new therapeutic opportunities. Int Rev Immunol. 2014;33:212-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
15.  Linsley PS, Nadler SG. The clinical utility of inhibiting CD28-mediated costimulation. Immunol Rev. 2009;229:307-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 132]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
16.  Hebert LA, Parikh S, Prosek J, Nadasdy T, Rovin BH. Differential diagnosis of glomerular disease: a systematic and inclusive approach. Am J Nephrol. 2013;38:253-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
17.  Couser WG. Pathogenesis of glomerular damage in glomerulonephritis. Nephrol Dial Transplant. 1998;13 Suppl 1:10-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 84]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
18.  Schwartz MM. The role of podocyte injury in the pathogenesis of focal segmental glomerulosclerosis. Ren Fail. 2000;22:663-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
19.  Clement LC, Liu G, Perez-Torres I, Kanwar YS, Avila-Casado C, Chugh SS. Early changes in gene expression that influence the course of primary glomerular disease. Kidney Int. 2007;72:337-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
20.  Reiser J, von Gersdorff G, Loos M, Oh J, Asanuma K, Giardino L, Rastaldi MP, Calvaresi N, Watanabe H, Schwarz K. Induction of B7-1 in podocytes is associated with nephrotic syndrome. J Clin Invest. 2004;113:1390-1397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Odobasic D, Kitching AR, Tipping PG, Holdsworth SR. CD80 and CD86 costimulatory molecules regulate crescentic glomerulonephritis by different mechanisms. Kidney Int. 2005;68:584-594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
22.  Tada Y, Nagasawa K, Ho A, Morito F, Koarada S, Ushiyama O, Suzuki N, Ohta A, Mak TW. Role of the costimulatory molecule CD28 in the development of lupus in MRL/lpr mice. J Immunol. 1999;163:3153-3159.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Garin EH, Mu W, Arthur JM, Rivard CJ, Araya CE, Shimada M, Johnson RJ. Urinary CD80 is elevated in minimal change disease but not in focal segmental glomerulosclerosis. Kidney Int. 2010;78:296-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 123]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
24.  Ishimoto T, Shimada M, Araya CE, Huskey J, Garin EH, Johnson RJ. Minimal change disease: a CD80 podocytopathy? Semin Nephrol. 2011;31:320-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
25.  Reynolds J, Khan SB, Allen AR, Benjamin CD, Pusey CD. Blockade of the CD154-CD40 costimulatory pathway prevents the development of experimental autoimmune glomerulonephritis. Kidney Int. 2004;66:1444-1452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
26.  Yellin MJ, D’Agati V, Parkinson G, Han AS, Szema A, Baum D, Estes D, Szabolcs M, Chess L. Immunohistologic analysis of renal CD40 and CD40L expression in lupus nephritis and other glomerulonephritides. Arthritis Rheum. 1997;40:124-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 113]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
27.  Kairaitis L, Wang Y, Zheng L, Tay YC, Wang Y, Harris DC. Blockade of CD40-CD40 ligand protects against renal injury in chronic proteinuric renal disease. Kidney Int. 2003;64:1265-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
28.  Biancone L, Andres G, Ahn H, DeMartino C, Stamenkovic I. Inhibition of the CD40-CD40ligand pathway prevents murine membranous glomerulonephritis. Kidney Int. 1995;48:458-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
29.  Ford ML, Larsen CP. Translating costimulation blockade to the clinic: lessons learned from three pathways. Immunol Rev. 2009;229:294-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 110]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
30.  Kalled SL, Cutler AH, Datta SK, Thomas DW. Anti-CD40 ligand antibody treatment of SNF1 mice with established nephritis: preservation of kidney function. J Immunol. 1998;160:2158-2165.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Pearson TC, Trambley J, Odom K, Anderson DC, Cowan S, Bray R, Lin A, Hollenbaugh D, Aruffo A, Siadak AW. Anti-CD40 therapy extends renal allograft survival in rhesus macaques. Transplantation. 2002;74:933-940.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 126]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
32.  Kawai T, Andrews D, Colvin RB, Sachs DH, Cosimi AB. Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand. Nat Med. 2000;6:114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 507]  [Cited by in F6Publishing: 472]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
33.  Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, Panoskaltsis N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018-1028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1589]  [Cited by in F6Publishing: 1391]  [Article Influence: 77.3]  [Reference Citation Analysis (0)]
34.  Salomon B, Bluestone JA. Complexities of CD28/B7: CTLA-4 costimulatory pathways in autoimmunity and transplantation. Annu Rev Immunol. 2001;19:225-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 817]  [Cited by in F6Publishing: 791]  [Article Influence: 34.4]  [Reference Citation Analysis (0)]
35.  Sayegh MH, Akalin E, Hancock WW, Russell ME, Carpenter CB, Linsley PS, Turka LA. CD28-B7 blockade after alloantigenic challenge in vivo inhibits Th1 cytokines but spares Th2. J Exp Med. 1995;181:1869-1874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 408]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
36.  Vincenti F, Charpentier B, Vanrenterghem Y, Rostaing L, Bresnahan B, Darji P, Massari P, Mondragon-Ramirez GA, Agarwal M, Di Russo G. A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study). Am J Transplant. 2010;10:535-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 667]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
37.  Pestana JO, Grinyo JM, Vanrenterghem Y, Becker T, Campistol JM, Florman S, Garcia VD, Kamar N, Lang P, Manfro RC. Three-year outcomes from BENEFIT-EXT: a phase III study of belatacept versus cyclosporine in recipients of extended criteria donor kidneys. Am J Transplant. 2012;12:630-639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 169]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
38.  Risk evaluation and mitigation strategy (REMS). Initial REMS Approval: June 2011.  Available from: http://www.fda.gov/downloads/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/UCM261934.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Furie R, Nicholls K, Cheng TT, Houssiau F, Burgos-Vargas R, Chen SL, Hillson JL, Meadows-Shropshire S, Kinaszczuk M, Merrill JT. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol. 2014;66:379-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 233]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
40.  Askanase AD, Byron M, Keyes-Elstein LL, Cagnoli PC, McCune W, Chatham W, Contreras G, Daikh DI, Dall' Era M, Wofsy D. Treatment of lupus nephritis with abatacept: the Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol. 2014;66:3096-3104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 185]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
41.  Yu CC, Fornoni A, Weins A, Hakroush S, Maiguel D, Sageshima J, Chen L, Ciancio G, Faridi MH, Behr D. Abatacept in B7-1-positive proteinuric kidney disease. N Engl J Med. 2013;369:2416-2423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 274]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
42.  Benigni A, Gagliardini E, Remuzzi G. Abatacept in B7-1-positive proteinuric kidney disease. N Engl J Med. 2014;370:1261-1263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
43.  Alachkar N, Carter-Monroe N, Reiser J. Abatacept in B7-1-positive proteinuric kidney disease. N Engl J Med. 2014;370:1263-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
44.  Larsen CP, Messias NC, Walker PD. B7-1 immunostaining in proteinuric kidney disease. Am J Kidney Dis. 2014;64:1001-1003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
45.  Reiser J, Alachkar N. Proteinuria: abate or applaud abatacept in proteinuric kidney disease? Nat Rev Nephrol. 2014;10:128-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
46.  Garin EH, Reiser J, Cara-Fuentes G, Wei C, Matar D, Wang H, Alachkar N, Johnson RJ. Case series: CTLA4-IgG1 therapy in minimal change disease and focal segmental glomerulosclerosis. Pediatr Nephrol. 2015;30:469-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
47.  Javaid B, Quigg RJ. Treatment of glomerulonephritis: will we ever have options other than steroids and cytotoxics? Kidney Int. 2005;67:1692-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
48.  McGrath MM, Najafian N. The Role of Coinhibitory Signaling Pathways in Transplantation and Tolerance. Frontiers in Immunology. 2012;3:47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]