51
|
Abstract
Gradually improved immunosuppression has contributed significantly to the progress achieved in transplantation medicine so far. Nevertheless, current drug regimens are associated with late graft loss--in particular as a result of immunologic damage or drug toxicity--and substantial morbidity. Recently, the costimulation blocker belatacept (marketed under the name Nulojix®) has been approved for immunosuppression in renal transplantation. Belatacept (a mutated version of CTLA4Ig) is a fusion protein rationally designed to block CD28, a critical activating receptor on T cells, by binding and saturating its ligands B7-1 and B7-2. In phase II and III trials, belatacept was compared with cyclosporine (in combination with basiliximab, MMF, and steroids). Advantages observed with belatacept include superior graft function, preservation of renal structure and improved cardiovascular risk profile. Concerns associated with belatacept are a higher frequency of cellular rejection episodes and more post-transplant lymphoproliferative disorder (PTLD) cases especially in EBV seronegative patients, who should be excluded from belatacept-based regimens. Thus, after almost three decades of calcineurin inhibitors as mainstay of immunosuppression, belatacept offers a potential alternative. In this article, we will provide an overview of belatacept's preclinical development and will discuss the available evidence from clinical trials.
Collapse
Affiliation(s)
- Thomas Wekerle
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria.
| | | |
Collapse
|
52
|
Sucher R, Fischler K, Oberhuber R, Kronberger I, Margreiter C, Ollinger R, Schneeberger S, Fuchs D, Werner ER, Watschinger K, Zelger B, Tellides G, Pilat N, Pratschke J, Margreiter R, Wekerle T, Brandacher G. IDO and regulatory T cell support are critical for cytotoxic T lymphocyte-associated Ag-4 Ig-mediated long-term solid organ allograft survival. THE JOURNAL OF IMMUNOLOGY 2011; 188:37-46. [PMID: 22131334 DOI: 10.4049/jimmunol.1002777] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Costimulatory blockade of CD28-B7 interaction with CTLA4Ig is a well-established strategy to induce transplantation tolerance. Although previous in vitro studies suggest that CTLA4Ig upregulates expression of the immunoregulatory enzyme IDO in dendritic cells, the relationship of CTLA4Ig and IDO in in vivo organ transplantation remains unclear. In this study, we studied whether concerted immunomodulation in vivo by CTLA4Ig depends on IDO. C57BL/6 recipients receiving a fully MHC-mismatched BALB/c heart graft treated with CTLA4Ig + donor-specific transfusion showed indefinite graft survival (>100 d) without signs of chronic rejection or donor specific Ab formation. Recipients with long-term surviving grafts had significantly higher systemic IDO activity as compared with rejectors, which markedly correlated with intragraft IDO and Foxp3 levels. IDO inhibition with 1-methyl-dl-tryptophan, either at transplant or at postoperative day 50, abrogated CTLA4Ig + DST-induced long-term graft survival. Importantly, IDO1 knockout recipients experienced acute rejection and graft survival comparable to controls. In addition, αCD25 mAb-mediated depletion of regulatory T cells (Tregs) resulted in decreased IDO activity and again prevented CTLA4Ig + DST induced indefinite graft survival. Our results suggest that CTLA4Ig-induced tolerance to murine cardiac allografts is critically dependent on synergistic cross-linked interplay of IDO and Tregs. These results have important implications for the clinical development of this costimulatory blocker.
Collapse
Affiliation(s)
- Robert Sucher
- Department of Visceral, Transplant and Thoracic Surgery, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Chen Y, Fukuda T, Thakar MS, Kornblit BT, Storer BE, Santos EB, Storb R, Sandmaier BM. Immunomodulatory effects induced by cytotoxic T lymphocyte antigen 4 immunoglobulin with donor peripheral blood mononuclear cell infusion in canine major histocompatibility complex-haplo-identical non-myeloablative hematopoietic cell transplantation. Cytotherapy 2011; 13:1269-80. [PMID: 21846291 DOI: 10.3109/14653249.2011.586997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS. Previously, cytotoxic T lymphocyte antigen 4 (CTLA4) immunoglobulin (Ig) has been shown to allow sustained engraftment in dog leukocyte antigen (DLA)-identical hematopoietic cell transplant (HCT) after non-myeloablative conditioning with 100 cGy total body irradiation (TBI). In the current study, we investigated the efficacy of pre-transplant CTLA4-Ig in promoting engraftment across a DLA-mismatched barrier after non-myeloablative conditioning. METHODS. Eight dogs were treated with CTLA4-Ig and donor peripheral blood mononuclear cells (PBMC) prior to receiving 200 cGy TBI followed by transplantation of granulocyte-colony-stimulating factor (G-CSF) mobilized peripheral blood stem cells from DLA haplo-identical littermates with post-grafting immunosuppression. A control group of six dogs was conditioned with 200 cGy only and transplanted with grafts from DLA haplo-identical littermates followed by post-grafting immunosuppression. RESULTS. In vitro and in vivo donor-specific hyporesponsiveness was demonstrated on day 0 before TBI in eight dogs that received CTLA4-Ig combined with donor PBMC infusions. Four of five dogs treated with increased doses of CTLA4-Ig achieved initial engraftment but eventually rejected, with a duration of mixed chimerism ranging from 12 to 22 weeks. CTLA4-Ig did not show any effect on host natural killer (NK) cell function in vitro or in vivo. No graft-versus-host disease (GvHD) was observed in dogs receiving CTLA4-Ig treatment. CONCLUSIONS. Non-myeloablative conditioning with 200 cGy TBI and CTLA4-Ig combined with donor PBMC infusion was able to overcome the T-cell barrier to achieve initial engraftment without GvHD in dogs receiving DLA haplo-identical grafts. However, rejection eventually occurred; we hypothesize because of the inability of CTLA4-Ig to abate natural killer cell function.
Collapse
Affiliation(s)
- Yun Chen
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, Washington , USA
| | | | | | | | | | | | | | | |
Collapse
|
54
|
Riella LV, Sayegh MH. T-cell co-stimulatory blockade in kidney transplantation: back to the bench. Kidney Int Suppl (2011) 2011; 1:25-30. [PMID: 25018899 PMCID: PMC4089598 DOI: 10.1038/kisup.2011.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
It is believed that blocking positive T-cell co-stimulatory pathways should lead to long-term graft acceptance. Despite the exciting initial achievements in experimental animal models, targeting co-stimulatory pathways has shown to be much more complex in the clinic. In addition to multiple binding partners, some co-stimulatory interactions have been found to be inhibitory in nature, whereas others were demonstrated to be important in the development of regulatory T cells. Moreover, memory T cells have been shown to be resistant to co-stimulation blockade. Herein we focus on the B7:CD28 pathway and describe the evolution of targeting this pathway with cytotoxic T-lymphocyte antigen-4-Ig from bench to clinic. We also attempt to address possible causes for the unexpected high rejection rate observed in the phase III clinical trials with belatacept, using experimental data obtained from basic science research.
Collapse
Affiliation(s)
- Leonardo V Riella
- Transplantation Research Center, Department of Medicine, Renal Division, Brigham and Women's Hospital, Children's Hospital Boston, Harvard Medical School , Boston, Massachusetts, USA
| | - Mohamed H Sayegh
- Transplantation Research Center, Department of Medicine, Renal Division, Brigham and Women's Hospital, Children's Hospital Boston, Harvard Medical School , Boston, Massachusetts, USA
| |
Collapse
|
55
|
Abstract
Secondary, so-called costimulatory, signals are critically required for the process of T cell activation. Since landmark studies defined that T cells receiving a T cell receptor signal without a costimulatory signal, are tolerized in vitro, the investigation of T cell costimulation has attracted intense interest. Early studies demonstrated that interrupting T cell costimulation allows attenuation of the alloresponse, which is particularly difficult to modulate due to the clone size of alloreactive T cells. The understanding of costimulation has since evolved substantially and now encompasses not only positive signals involved in T cell activation but also negative signals inhibiting T cell activation and promoting T cell tolerance. Costimulation blockade has been used effectively for the induction of tolerance in rodent models of transplantation, but turned out to be less potent in large animals and humans. In this overview we will discuss the evolution of the concept of T cell costimulation, the potential of 'classical' and newly identified costimulation pathways as therapeutic targets for organ transplantation as well as progress towards clinical application of the first costimulation blocking compound.
Collapse
Affiliation(s)
- Nina Pilat
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| | - Mohamed H. Sayegh
- Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, USA
| | - Thomas Wekerle
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| |
Collapse
|
56
|
Abstract
Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease in children and an important cause of short-term and long-term disability. Gene changes in the immune system can predispose to JIA and regulation of the immune system is crucial in the pathogenesis. The goal of therapy is complete disease control using disease-modifying antirheumatic drugs (DMARDS). Activated T-cells may play a role in the immunopathology of JIA. Therefore, targeting T-cell activation is a rational approach for the treatment of JIA. Abatacept (ABA), a selective co-stimulation modulator, has been shown to be effective in treating all JIA subtypes and is generally safe and well tolerated in JIA. Neutralizing antibodies were found in 6/9 (67%) of seropositive patients, but anti-ABA antibodies did not appear to be associated with disease flare, serious adverse events, acute infusional adverse events, hypersensitivity, autoimmune disorders, or low ABA serum concentrations. Anti-ABA antibodies were more frequent when ABA concentrations were below therapeutic levels. Although information on ABA in JIA is still limited, available data suggest a potential role in difficult to treat JIA patients previously treated with other biologic agents and for non-responders to TNF-blockade.
Collapse
|
57
|
Choi EW, Shin IS, Lee HW, Park SY, Park JH, Nam MH, Kim JS, Woo SK, Yoon EJ, Kang SK, Ra JC, Youn HY, Hong SH. Transplantation of CTLA4Ig gene-transduced adipose tissue-derived mesenchymal stem cells reduces inflammatory immune response and improves Th1/Th2 balance in experimental autoimmune thyroiditis. J Gene Med 2011; 13:3-16. [PMID: 21259404 DOI: 10.1002/jgm.1531] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Autoimmune thyroiditis is one of common organ-specific autoimmune disease. The aim of this study was to observe the effect of adipose tissue derived mesenchymal stem cells (ATMSC) and CTLA4Ig gene-transduced ATMSC on autoimmune thyroiditis. METHODS Experimental autoimmune thyroiditis was induced by immunization with thyroglobulin. Animals were divided into three groups: (i) a half million of human ATMSC, (ii) a half million of murine CLTA4Ig gene-transduced human ATMSC (CTLA4Ig-MSC), or (iii) normal saline (as control), which were administered intravenously four times within a 3-week period. Blood and tissue samples were collected 1 week after the last cell transplantation. RESULTS The absorbance of serum thyroglobulin autoantibody (TgAA) in the CTLA4Ig-MSC group was considerably lower than those in other groups. In culture supernatant of LPS-stimulated spleen cells, both of the MSC-treated groups showed significantly lower absorbances of TgAA than the control. Flow cytometric analysis of spleen cells revealed a significant decrease in the proportion of CD3+ and CD11b in the CTLA4Ig-MSC group compared to the other groups. Lymphocyte infiltration in the thyroid glands was also dramatically decreased in both of MSC-treated groups. Cytokine analysis showed that ATMSC decreased the production of proinflammatory cytokines and improved the Th1/Th2 balance by down-regulating Th1 cytokines. CONCLUSION Although CTLA4Ig-MSC transplantation had better result in reduction of serum TgAA, both of ATMSC and CTLA4Ig-MSC transplantations are promising treatments for autoimmune thyroiditis judging from the results of histopathology and cytokine analysis. They may be attractive candidates for treating organ-specific autoimmune disease.
Collapse
Affiliation(s)
- Eun Wha Choi
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, 50 Irwon-dong, Gangnam-gu, Seoul 135-710, Republic of Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Azuma H, Isaka Y, Nomi H, Inamoto T, Li XK, Hounig T, Takabatake Y, Ichimaru N, Ibuki N, Matsumoto K, Ubai T, Katsuoka Y, Takahara S. Induction of donor-specific tolerance using superagonistic CD28 antibody in rat renal allografts: regulatory T-cell expansion before engraftment may be important. Transplantation 2011; 90:1328-35. [PMID: 21085062 DOI: 10.1097/tp.0b013e3182007b59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND We hypothesized that a superagonistic monoclonal antibody specific for CD28 (CD28SA), which expands regulatory T cells (Tregs) in vivo, would prevent acute rejection and prolong the survival of renal allograft. METHODS We examined whether CD28SA treatment induce donor-specific tolerance using our established rat renal allograft model (Wistar-Lewis). RESULTS All control rats died within 13 days because of severe azotemia with marked destruction of graft tissue. In contrast, recipients treated with a triple injection of CD28SA (days -3, 0, and 3) showed good preservation of graft histology and function, with considerable infiltration of Tregs into the allografts; 92% of recipients survived for more than 100 days, and 77% survived by the day of harvest at 180 days. These long-surviving recipients received secondary heterotopic bicardiac allografts from both donor-matched Wistar and third-party Brown Norway rats simultaneously 120 days after kidney transplantation, and seven of eight (87.5%) rats exhibited donor-specific tolerance, accepting the Wistar heart, but acutely rejecting the Brown Norway heart. Interestingly, a single injection of CD28SA 3 days before (day -3), but not 3 days after (day 3), transplantation also induced donor-specific tolerance in some recipients. We then performed adoptive transfer of nonspecific CD4+CD25+ Tregs, purified from CD28SA-treated Lewis rats, with simultaneous injection of hepatocyte growth factor (500 μg/kg/day, intravenously). The treatment induced significant prolongation of graft survival (P<0.0001 vs. control group), and five of eight (62.5%) recipients survived until the day of harvest at 180 days with successful induction of donor-specific tolerance. CONCLUSIONS We have established a novel therapeutic approach for inducing donor-specific tolerance in rats with renal allografts.
Collapse
Affiliation(s)
- Haruhito Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Topical inhibition of T cell costimulatory pathways in draining lymph nodes may suppress allograft rejection. Med Hypotheses 2010; 76:441-4. [PMID: 21134721 DOI: 10.1016/j.mehy.2010.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 11/09/2010] [Accepted: 11/13/2010] [Indexed: 11/22/2022]
Abstract
Topical immune suppression is an attractive and practical therapeutic option to prolong survival time of allografts, before the appearance of new agent with higher immunosuppressive efficacy and lower undesirable side effects. The initiation of rejection and outcome of allografts is principally mediated by alloantigen reactive T cells. The activation of T cells requires at least two signals, first is T-cell receptor signal and second is costimulatory signal. T cells that encounter antigen without the appropriate costimulatory signal become anergy or tolerance. Migration of alloantigen-bearing dendritic cells into the T-cell zone of secondary lymphoid tissues, which are essential for primary alloimmune responses, effectively induces T-cell activation and expansion with the presence of two signals. Draining lymph nodes are the promising targets for topical immune suppression, as disrupting lymphatic drainage from the transplanted graft to lymph nodes prevented rejection of skin allografts and lymphadenectomy prolong the survival time of skin and corneal allografts in experimental animals. Therefore, we hypothesize that inhibition of T cell costimulatory pathways in draining lymph nodes could impair the alloantigen-specific immune response and reduce systemic immunosuppressive drugs dose for allografts survival. Further investigations are required to identify most efficient way for draining lymph nodes transfer of costimulatory molecule gene or topical drug administration of costimulatory inhibitors to draining lymph nodes.
Collapse
|
60
|
Verbinnen B, Van Gool SW, Ceuppens JL. Blocking costimulatory pathways: prospects for inducing transplantation tolerance. Immunotherapy 2010; 2:497-509. [PMID: 20636004 DOI: 10.2217/imt.10.31] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tolerance induction to alloantigens is a major challenge in transplant immunology. Whereas conventional immunosuppression inhibits the immune system in a nonspecific way, thereby also undermining an appropriate immune response towards potentially harmful infectious organisms, tolerance in a transplantation setting is restricted to alloantigens, while protective immunity is preserved. Moreover, tolerance implies an immunological status that is preserved after withdrawal of the tolerance-inducing therapy. Among the most promising strategies to induce immunological tolerance are costimulation blockade and establishment of mixed chimerism. Despite significant advances, we still know little about the mechanisms responsible for such tolerance. In this article, we discuss tolerance induction to transplantation antigens by costimulation blockade.
Collapse
Affiliation(s)
- Bert Verbinnen
- University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | | | | |
Collapse
|
61
|
Poirier N, Blancho G, Vanhove B. A more selective costimulatory blockade of the CD28-B7 pathway. Transpl Int 2010; 24:2-11. [DOI: 10.1111/j.1432-2277.2010.01176.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
62
|
Zhang S, Dai H, Wan N, Moore Y, Dai Z. Manipulating IL-2 availability amid presentation of donor MHC antigens suppresses murine alloimmune responses by inducing regulatory T cells. PLoS One 2010; 5:e8756. [PMID: 20090908 PMCID: PMC2807454 DOI: 10.1371/journal.pone.0008756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 12/18/2009] [Indexed: 12/04/2022] Open
Abstract
Background Major histocompatibility complex (MHC) antigens are important for alloimmune responses as well as immune tolerance. Previous studies have shown that presentation of donor MHC antigens by donor-specific transfusion prior to or upon transplantation promotes transplant tolerance induced by other agents. However, it is unclear whether presentation of donor MHC antigens by DNA vaccination induces long-term allograft survival. Methodology/Principal Findings We investigated whether presentation of MHC class-II and/or class-I donor antigens by DNA vaccination suppresses alloimmune responses and promotes long-term allograft acceptance. We initially found that presentation of both MHC donor antigens by DNA vaccination itself prior to transplantation fails to significantly prolong islet allograft survival in otherwise untreated mice. However, islet allograft survival was significantly prolonged when MHC class-II DNA vaccination was accompanied with IL-2 administration (MHCII + IL-2) while MHC class-I DNA vaccination was followed by IL-2 and subsequent neutralizing anti-IL-2 treatments (MHCI + IL-2/anti-IL-2). Especially, this protocol promoted long-term allograft survival in the majority of recipients (57%) when combined with low doses of rapamycin post-transplantation. Importantly, MHCII + IL-2 induced FoxP3+ Treg cells in both spleens and grafts and suppressed graft-infiltrating CD4+ cell proliferation, whereas MHCI + IL-2/anti-IL-2 mainly inhibited graft-infiltrating CD8+ cell proliferation and donor-specific CTL activity. The combined protocol plus rapamycin treatment further reduced both CD4+ and CD8+ T cell proliferation as well as donor-specific CTL activity but spared FoxP3+ Treg cells. Depleting CD25+ Treg cells or adoptive transfer of pre-sensitized CD8+ T cells abolished this long-term allograft survival. Conclusions/Significance Manipulating IL-2 availability during presentation of MHC class-II and class-I donor antigens by DNA vaccination pre-transplantation induces Treg cells, suppresses alloimmune responses and promotes long-term allograft survival.
Collapse
Affiliation(s)
- Shuzi Zhang
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Hehua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Ni Wan
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Yolonda Moore
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Zhenhua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
63
|
Weaver TA, Charafeddine AH, Agarwal A, Turner AP, Russell M, Leopardi FV, Kampen RL, Stempora L, Song M, Larsen CP, Kirk AD. Alefacept promotes co-stimulation blockade based allograft survival in nonhuman primates. Nat Med 2009; 15:746-9. [PMID: 19584865 PMCID: PMC2772128 DOI: 10.1038/nm.1993] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 06/03/2009] [Indexed: 12/12/2022]
Abstract
Memory T cells promote allograft rejection particularly in co-stimulation blockade-based immunosuppressive regimens. Here we show that the CD2-specific fusion protein alefacept (lymphocyte function-associated antigen-3-Ig; LFA -3-Ig) selectively eliminates memory T cells and, when combined with a co-stimulation blockade-based regimen using cytotoxic T lymphocyte antigen-4 (CTLA-4)-Ig, a CD80- and CD86-specific fusion protein, prevents renal allograft rejection and alloantibody formation in nonhuman primates. These results support the immediate translation of a regimen for the prevention of allograft rejection without the use of calcineurin inhibitors, steroids or pan-T cell depletion.
Collapse
Affiliation(s)
- Tim A Weaver
- Transplantation Branch, National Institute of Diabetes, Digestive and Kidney Diseases, US National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Lutt JR. Efficacy, safety, and tolerability of abatacept in the management of rheumatoid arthritis. Open Access Rheumatol 2009; 1:17-35. [PMID: 27789979 PMCID: PMC5074723 DOI: 10.2147/oarrr.s4536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The management of rheumatoid arthritis (RA) has undergone an impressive transformation over the past few decades. Further understanding of the pathophysiology of the disease process has resulted in the development of biologic agents that target proinflammatory cytokines and both B and T lymphocytes. By blocking an important costimulatory pathway, abatacept leads to a dramatic reduction in T cell stimulation and proliferation. Multiple clinical trials have revealed consistent benefit with regards to clinical and radiographic efficacy, quality of life, and disability in patients suffering from RA who have had inadequate responses to methotrexate or tumor necrosis factor inhibitors. The possibility of remission when used early in the disease course has also been demonstrated. Importantly, abatacept has been very well tolerated with a low rate of serious infections and no apparent increase in malignancies to date. Continued surveillance of the benefits and risks will help to better define its place amongst the other biologic agents in the treatment of RA.
Collapse
Affiliation(s)
- Joseph R Lutt
- Division of Arthritis and Rheumatic Diseases, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
65
|
Li XC, Rothstein DM, Sayegh MH. Costimulatory pathways in transplantation: challenges and new developments. Immunol Rev 2009; 229:271-93. [DOI: 10.1111/j.1600-065x.2009.00781.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
66
|
Viral infection: a potent barrier to transplantation tolerance. Clin Dev Immunol 2009; 2008:742810. [PMID: 18815618 PMCID: PMC2536507 DOI: 10.1155/2008/742810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 06/26/2008] [Indexed: 01/28/2023]
Abstract
Transplantation of allogeneic organs has proven to be an effective therapeutic for a large variety of disease states, but the chronic immunosuppression that is required for organ allograft survival increases the risk for infection and neoplasia and has direct organ toxicity. The establishment of transplantation tolerance, which obviates the need for chronic immunosuppression, is the ultimate goal in the field of transplantation. Many experimental approaches have been developed in animal models that permit long-term allograft survival in the absence of chronic immunosuppression. These approaches function by inducing peripheral or central tolerance to the allograft. Emerging as some of the most promising approaches for the induction of tolerance are protocols based on costimulation blockade. However, as these protocols move into the clinic, there is recognition that little is known as to their safety and efficacy when confronted with environmental perturbants such as virus infection. In animal models, it has been reported that virus infection can prevent the induction of tolerance by costimulation blockade and, in at least one experimental protocol, can lead to significant morbidity and mortality. In this review, we discuss how viruses modulate the induction and maintenance of transplantation tolerance.
Collapse
|
67
|
|
68
|
Tolerance and Future Directions for Composite Tissue Allograft Transplants: Part II. Plast Reconstr Surg 2009; 123:7e-17e. [DOI: 10.1097/prs.0b013e318193467d] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
69
|
Azuma H, Isaka Y, Li X, Hünig T, Sakamoto T, Nohmi H, Takabatake Y, Mizui M, Kitazawa Y, Ichimaru N, Ibuki N, Ubai T, Inamoto T, Katsuoka Y, Takahara S. Superagonistic CD28 antibody induces donor-specific tolerance in rat renal allografts. Am J Transplant 2008; 8:2004-14. [PMID: 18727698 DOI: 10.1111/j.1600-6143.2008.02358.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The ultimate goal of organ transplantation is to establish graft tolerance where CD4+CD25+FOXP3+ regulatory T (Treg) cells play an important role. We examined whether a superagonistic monoclonal antibody specific for CD28 (CD28 SA), which expands Treg cells in vivo, would prevent acute rejection and induce tolerance using our established rat acute renal allograft model (Wistar to Lewis). In the untreated or mouse IgG-treated recipients, graft function significantly deteriorated with marked destruction of renal tissue, and all rats died by 13 days with severe azotemia. In contrast, 90% of recipients treated with CD28 SA survived over 100 days, and 70% survived with well-preserved graft function until graft recovery at 180 days. Analysis by flow cytometry and immunohistochemistry demonstrated that CD28 SA induced marked infiltration of FOXP3+ Treg cells into the allografts. Furthermore, these long-surviving recipients showed donor-specific tolerance, accepting secondary (donor-matched) Wistar cardiac allografts, but acutely rejecting third-party BN allografts. We further demonstrated that adoptive transfer of CD4+CD25+ Treg cells, purified from CD28 SA-treated Lewis rats, significantly prolonged allograft survival and succeeded in inducing donor-specific tolerance. In conclusion, CD28 SA treatment successfully induces donor-specific tolerance with the involvement of Treg cells, and thus the therapeutic value of this approach warrants further investigation and preclinical studies.
Collapse
Affiliation(s)
- H Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Choi EW, Shin IS, Lee CW, Youn HY. The effect of gene therapy using CTLA4Ig/silica-nanoparticles on canine experimental autoimmune thyroiditis. J Gene Med 2008; 10:795-804. [PMID: 18452240 DOI: 10.1002/jgm.1203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The present study aimed to determine the effect of canine CTLA4Ig on canine autoimmune thyroiditis. In a previous study, we established a canine model of autoimmune thyroiditis by immunizing normal dogs with bovine thyroglobulin. An in vitro study using recombinant CTLA4Ig revealed that this protein can inhibit the expression of Th1-type cytokines and the pro-inflammatory cytokines tested. METHODS As a result of the in vitro study, we constructed therapeutic CTLA4Ig/silica-nanoparticles and applied them to the treatment of experimentally induced canine autoimmune thyroiditis. RESULTS Gene therapy resulted in significant reductions in anti-canine-thyroglobulin autoantibody titer, anti-T4 antibody titer and T-cell proliferation against thyroglobulin and in the mRNA expressions of interleukin-18 in fresh peripheral blood mononuclear cells (PBMC) from all dogs. There was also a significant reduction compared to day 0 in tumor necrosis factor-alpha and interferon-gamma levels in the supernatant from cultured PBMC. CONCLUSIONS The CTLA4Ig-induced suppression of Th1 cytokines is relatively more significant than it appears because autoimmune thyroiditis is a Th1-polarized disease. Thus, CTLA4Ig can improve Th1/Th2 cytokine balance in autoimmune thyroiditis by downregulating Th1 cytokines.
Collapse
Affiliation(s)
- Eun Wha Choi
- KRF Zoonotic Disease Priority Research Institute, Seoul National University, Seoul, Korea
| | | | | | | |
Collapse
|
71
|
Verbinnen B, Billiau AD, Vermeiren J, Galicia G, Bullens DMA, Boon L, Cadot P, Hens G, Dewolf-Peeters C, Van Gool SW, Ceuppens JL. Contribution of regulatory T cells and effector T cell deletion in tolerance induction by costimulation blockade. THE JOURNAL OF IMMUNOLOGY 2008; 181:1034-42. [PMID: 18606655 DOI: 10.4049/jimmunol.181.2.1034] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Blocking of costimulatory signals for T cell activation leads to tolerance in several transplantation models, but the underlying mechanisms are incompletely understood. We analyzed the involvement of regulatory T cells (Treg) and deletion of alloreactive cells in the induction and maintenance of tolerance after costimulation blockade in a mouse model of graft-vs-host reaction. Injection of splenocytes from the C57BL/6 parent strain into a sublethally irradiated F(1) offspring (C57BL/6 x C3H) induced a GVHR characterized by severe pancytopenia. Treatment with anti-CD40L mAb and CTLA4-Ig every 3 days during 3 wk after splenocyte injection prevented disease development and induced a long-lasting state of stable mixed chimerism (>120 days). In parallel, host-specific tolerance was achieved as demonstrated by lack of host-directed alloreactivity of donor-type T cells in vitro and in vivo. Chimerism and tolerance were also obtained after CD25(+) cell-depleted splenocyte transfer, showing that CD25(+) natural Treg are not essential for tolerance induction. We further show that costimulation blockade results in enhanced Treg cell activity at early time points (days 6-30) after splenocyte transfer. This was demonstrated by the presence of a high percentage of Foxp3(+) cells among donor CD4(+) cells in the spleen of treated animals, and our finding that isolated donor-type T cells at an early time point (day 30) after splenocyte transfer displayed suppressive capacity in vitro. At later time points (>30 days after splenocyte transfer), clonal deletion of host-reactive T cells was found to be a major mechanism responsible for tolerance.
Collapse
Affiliation(s)
- Bert Verbinnen
- Division of Clinical Immunology, University Hospital, Catholic University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
del Rio ML, Buhler L, Gibbons C, Tian J, Rodriguez-Barbosa JI. PD-1/PD-L1, PD-1/PD-L2, and other co-inhibitory signaling pathways in transplantation. Transpl Int 2008; 21:1015-28. [PMID: 18662368 DOI: 10.1111/j.1432-2277.2008.00726.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Transplantation of cells, tissues and vascularized solid organs is a successful therapeutic intervention for many end-stage chronic diseases. The combination of co-stimulatory blockade with the delivery of negative signals to T cells through co-inhibitory receptors would provide a robust approach to modulating T-cell receptor signaling and improving alloantigen-specific control of transplant rejection. This approach based on fundamental knowledge of APC/T-cell interactions may complement conventional therapies in the near future to reinforce long-term allograft survival, and permit minimal immunosuppression. The focus of this review was primarily on two major co-inhibitory signaling pathways, namely PD-1/PD-L1/PD-L2 and BTLA/CD160/HVEM/LIGHT that have been thoroughly characterized in murine models of transplantation using genetically modified mice, specific monoclonal antibodies and fusion proteins.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Laboratory of Immunobiology, School of Biotechnology and Institute of Biomedicine, University of Leon, Leon, Spain
| | | | | | | | | |
Collapse
|
73
|
Promises and Obstacles for the Blockade of CD40–CD40L Interactions in Allotransplantation. Transplantation 2008; 86:10-5. [DOI: 10.1097/tp.0b013e31817c4b97] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
74
|
Liu C, Pan S, Jiang H, Sun X. Gene transfer of antisense B7.1 attenuates acute rejection against splenic allografts in rats. Transplant Proc 2007; 39:3391-3395. [PMID: 18089391 DOI: 10.1016/j.transproceed.2007.08.094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 06/18/2007] [Accepted: 08/08/2007] [Indexed: 01/01/2023]
Abstract
Blockade of CD80-CD28 costimulatory pathway induces unresponsiveness of T cells to alloantigens and protects allografts against immune rejection in numerous animal models. The aim of this study was to investigate whether blocking expression of B7.1 (CD80) on donor splenocytes by an antisense technique protected splenic allografts against immune rejection. Splenic grafts from Wistar-Furth rats were intra-arterially transfused with an antisense B7.1 expression vector, before they were transplanted into Sprague-Dawley rats. The rats were sacrificed at scheduled times, and the splenic allografts histologically examined. Antisense gene transfer resulted in marked down-regulation of B7.1 in donor spleens, hyporesponsiveness of recipient T cells, and attenuated acute immune rejection against splenic allografts. No obvious damage to skin, liver, or gut due to graft-versus-host disease was detected in the recipients. In conclusion, blocking expression of B7.1 in donor spleens by antisense gene therapy represented a potential alloantigen-specific immunosuppressive strategy to inhibit acute rejection against splenic allografts.
Collapse
Affiliation(s)
- C Liu
- Department of General Surgery, the Fourth Affiliated Hospital, and the First Clinical Medical School, Harbin Medical University, China
| | | | | | | |
Collapse
|
75
|
Truong W, Plester JC, Hancock WW, Merani S, Murphy TL, Murphy KM, Kaye J, Anderson CC, Shapiro AMJ. Combined coinhibitory and costimulatory modulation with anti-BTLA and CTLA4Ig facilitates tolerance in murine islet allografts. Am J Transplant 2007; 7:2663-74. [PMID: 17983390 DOI: 10.1111/j.1600-6143.2007.01996.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Complex interactions between positive and negative cosignaling receptors ultimately determine the fate of the immune response. The recently identified coinhibitory receptor, B and T lymphocyte attenuator (BTLA), contributes to regulation of autoimmune and potentially alloimmune responses. We investigated the role of BTLA in a fully major histocompatibility complex-mismatched mouse islet transplant model. We report that anti-BTLA mAb (6F7) alone does not accelerate graft rejection. Rather, while CTLA4Ig alone improved allograft survival, the addition of anti-BTLA mAb to CTLA4Ig led to indefinite (>100 days) allograft survival. Immediately after treatment with anti-BTLA mAb and CTLA4Ig, islet allografts showed intact islets and insulin production despite a host cellular response, with local accumulation of Foxp3+ cells. We clearly demonstrate that combined therapy with anti-BTLA mAb and CTLA4Ig mice induced donor-specific tolerance, since mice accepted a second donor-specific islet graft without further treatment and rejected third party grafts. CTLA4Ig and anti-BTLA mAb limited the initial in vivo proliferation of CFSE-labeled allogeneic lymphocytes, and anti-BTLA mAb enhanced the proportion of PD-1 expressing T cells while depleting pathogenic BTLA+ lymphocytes. We conclude that targeting the BTLA pathway in conjunction with CTLA4Ig costimulatory blockade may be a useful strategy for promoting immunological tolerance in murine islet allografts.
Collapse
Affiliation(s)
- W Truong
- The Surgical Medical Research Institute, Department of Surgery, Faculty of Medicine, The University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Negative and Positive Co-Signaling With Anti-BTLA (PJ196) and CTLA4Ig Prolongs Islet Allograft Survival. Transplantation 2007; 84:1368-72. [DOI: 10.1097/01.tp.0000289995.70390.20] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
77
|
Tanaka K, Albin MJ, Yuan X, Yamaura K, Habicht A, Murayama T, Grimm M, Waaga AM, Ueno T, Padera RF, Yagita H, Azuma M, Shin T, Blazar BR, Rothstein DM, Sayegh MH, Najafian N. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:5204-10. [PMID: 17911605 PMCID: PMC2291549 DOI: 10.4049/jimmunol.179.8.5204] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The PD-1:PDL pathway plays an important role in regulating alloimmune responses but its role in transplantation tolerance is unknown. We investigated the role of PD-1:PDL costimulatory pathway in peripheral and a well established model of central transplantation tolerance. Early as well as delayed blockade of PDL1 but not PDL2 abrogated tolerance induced by CTLA4Ig in a fully MHC-mismatched cardiac allograft model. Accelerated rejection was associated with a significant increase in the frequency of IFN-gamma-producing alloreactive T cells and expansion of effector CD8(+) T cells in the periphery, and a decline in the percentage of Foxp3(+) graft infiltrating cells. Similarly, studies using PDL1/L2-deficient recipients confirmed the results with Ab blockade. Interestingly, while PDL1-deficient donor allografts were accepted by wild-type recipients treated with CTLA4Ig, the grafts developed severe chronic rejection and vasculopathy when compared with wild-type grafts. Finally, in a model of central tolerance induced by mixed allogeneic chimerism, engraftment was not abrogated by PDL1/L2 blockade. These novel data demonstrate the critical role of PDL1 for induction and maintenance of peripheral transplantation tolerance by its ability to alter the balance between pathogenic and regulatory T cells. Expression of PDL1 in donor tissue is critical for prevention of in situ graft pathology and chronic rejection.
Collapse
Affiliation(s)
- Katsunori Tanaka
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Huurman VAL, Unger WWJ, Koeleman BPC, Oaks MK, Chandraker AK, Terpstra OT, Roep BO. Differential inhibition of autoreactive memory- and alloreactive naive T cell responses by soluble cytotoxic T lymphocyte antigen 4 (sCTLA4), CTLA4Ig and LEA29Y. Clin Exp Immunol 2007; 150:487-93. [PMID: 17924973 DOI: 10.1111/j.1365-2249.2007.03513.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cytotoxic T lymphocyte antigen 4 (CTLA4) is a potent inhibitory co-stimulatory molecule believed to be involved in type 1 diabetes and other autoimmune diseases. An association has been reported of both mRNA expression and serum levels of the soluble splice variant of CTLA4 (sCTLA4) with type 1 diabetes. Furthermore, recombinant fusion proteins CTLA4Ig and LEA29Y have been proposed as therapies for type 1 diabetes. We studied the role of (s)CTLA4 in islet autoimmunity. Binding capacity of the proteins to antigen-presenting cells was determined by flow cytometry in competition and binding assays. Functionality of sCTLA4 as well as the therapeutic inhibitory fusion proteins CTLA4Ig and LEA29Y was measured in a dose-response lymphocyte stimulation test, using a panel of diabetes-associated T cell clones reactive to islet autoantigens. As controls, mixed lymphocyte reactions (MLR) were performed to assess functionality of these proteins in a primary alloreactive setting. All three CTLA4 molecules were able to bind to antigen-presenting cells and inhibit the expression of CD80/CD86. sCTLA4 was able to suppress proliferation of different committed autoreactive T cell clones in a dose-dependent manner, whereas CTLA4Ig and LEA29Y were not. Conversely, CTLA4Ig and LEA29Y, rather than sCTLA4, were able to suppress naive alloreactive proliferation in a MLR. Our results indicate a differential role for sCTLA4, CTLA4Ig and LEA29Y proteins in memory versus primary immune responses with implications for efficacy in intervention therapy.
Collapse
Affiliation(s)
- V A L Huurman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
The modulation of co-stimulatory pathways represents a novel therapeutic strategy to regulate autoimmune diseases. Auto-reactive CD4+ T cells play a critical role in initiating the immune response leading to inflammation and autoimmune diseases. Blocking co-stimulatory signals prevents T-cell activation, thus diminishing autoimmune responses and possibly preventing the progression of autoimmune disease. Blockade of several co-stimulatory pathways has been investigated in animal models and has led to clinical trials testing specific blocking agents in humans. In this review we will describe the role of co-stimulatory pathways, primarily the CD28-B7 pathway, in autoimmune diseases, and we will present in vivo and in vitro studies supporting the efficacy of co-stimulation blockade in animal models of autoimmune disease. Finally, we will discuss the clinical therapeutic efficacy of blocking monoclonal antibodies in preventing or reducing auto-antigen driven T-cell activation in humans with particular attention to the CD28/B7 pathway. Inhibiting co-stimulatory molecule interactions by using monoclonal antibodies seems to be an original approach to regulate autoimmune diseases in humans.
Collapse
Affiliation(s)
- Vissia Viglietta
- Clinical Immunology Laboratory, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
80
|
Lim DG, Joe IY, Park YH, Chang SH, Wee YM, Han DJ, Kim SC. Effect of immunosuppressants on the expansion and function of naturally occurring regulatory T cells. Transpl Immunol 2007; 18:94-100. [PMID: 18005851 DOI: 10.1016/j.trim.2007.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 05/21/2007] [Indexed: 02/04/2023]
Abstract
The induction of immune tolerance is one of the final therapeutic goals in clinical transplantation. Regulatory T lymphocytes are important for the induction and maintenance of immune tolerance to grafts. If immunosuppressive drugs used clinically to prevent immune rejection also inhibit regulatory T lymphocytes, tolerance would not be achieved. We therefore tested the effect of several immunosuppressants with different mechanisms of action on the proliferation and suppressive activity of CD4(+)CD25(+) regulatory T cells. Highly purified CD4(+)CD25h(+) T cells from C57BL/6 (H-2(b)) mice were stimulated with allogeneic T-depleted splenocytes (BALB/c; H-2(d)) in the presence of various immunosuppressants. After one week in culture, viable T cells were recovered, their regulatory capacity was assessed by their ability to inhibit responder T cell proliferation in MLR, and their cytokine production profile was measured by ELISA. The immunosuppressants rapamycin, cyclosporine A, and methylprednisolone significantly inhibited the expansion of regulatory T cells upon stimulation with alloantigen, whereas mycophenolic acid and the costimulatory blockers, anti-CD40L and CTLA4Ig, did not. None of these immunosuppressants, however, reduced the suppressive capacity of regulatory T cells. Pretreatment with immunosuppressants did not induce significant changes in the cytokine production profile of regulatory T cells. Our results suggest that costimulatory blockers and mycophenolate mofetil can be utilized therapeutically in the induction of immune tolerance. In contrast, the use of rapamycin, cyclosporine A, and methylprednisolone should be reconsidered, due to their deleterious effects on the expansion of naturally occurring regulatory T cells.
Collapse
Affiliation(s)
- Dong-Gyun Lim
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Seoul 138-736, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
81
|
Wang S, Diao H, Guan Q, Jevnikar AM, Du C. Enhanced cardiac allograft survival by Vav1-Rac signaling blockade in a mouse model. Transpl Immunol 2007; 18:53-61. [PMID: 17584603 DOI: 10.1016/j.trim.2007.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 03/12/2007] [Accepted: 03/23/2007] [Indexed: 12/27/2022]
Abstract
BACKGROUND Vav1-Rac signaling plays a pivotal role in TCR/antigen and CD28 signals for T cell activation. However, pharmacological interference of this signaling has not been tested in the prevention of alloimmune-mediated allograft rejection. It has been demonstrated that 6-thio-GTP, a metabolite of azathioprine, specifically inhibits Vav1-Rac activity in T lymphocytes. Here we show the immunosuppressive efficacy of 6-thio-GTP in the prevention of cardiac allograft rejection. METHODS T cell proliferations were measured by (3)H-thymidine uptake. The immunosuppressive activities of 6-thio-GTP were tested in the cardiac allograft model of C57BL/6 (H-2(b)) to Balb/c (H-2(d)) mice. RESULTS 6-Thio-GTP inhibited TCR/alloantigen stimulated T cell proliferation and CD28-dependent T cell survival. Administration of 6-thio-GTP (0.5 mg/kg) prolonged graft survival to 13.8+/-2.39 days compared to 8.3+/-0.48 days in PBS controls (p<0.0001). Combination of 6-thio-GTP (0.5 mg/kg) with CsA (15 mg/kg) enhanced graft survival from 15.0+/-1.61 days in CsA treated recipients to 36.8+/-2.17 days in those received 20 days of combination therapy of CsA and 6-thio-GTP (p<0.0001), or to 42.7+/-16.63 days in the group treated with 20 days of CsA and 60 days of 6-thio-GTP (p<0.0001). Lymphocytes from 6-thio-GTP treated recipients with long-term surviving grafts (>60 days) displayed reduced proliferative response to alloantigen and higher frequencies of regulatory T cells (Treg). CONCLUSION Vav1-Rac inhibitor 6-thio-GTP prolongs allograft survival alone or in combination with CsA by suppression of alloreactive T cell activation. Our findings suggest the therapeutic potential of pharmacological interference of Vav1-Rac signaling for transplantation.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Medicine, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
82
|
Shiao SL, McNiff JM, Masunaga T, Tamura K, Kubo K, Pober JS. Immunomodulatory properties of FK734, a humanized anti-CD28 monoclonal antibody with agonistic and antagonistic activities. Transplantation 2007; 83:304-13. [PMID: 17297405 DOI: 10.1097/01.tp.0000251426.46312.d5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND We describe immunomodulatory effects of FK734, a humanized version of a mouse anti-human CD28 mAb (clone TN228), in vitro and in a chimeric human-mouse model of allograft rejection. METHODS Cytokine production and proliferation were assessed in a mixed lymphocyte reaction containing FK734, human T cells, and endothelial cells or monocytes. FK734 was also administered to SCID mice engrafted with human skin and adoptively transferred with human peripheral blood mononuclear cells allogeneic to the skin graft. RESULTS In vitro, FK734 enhanced secretion of interleukin-2 and interferon-gamma as well as proliferation of CD4+ and CD8+ T cells stimulated by allogeneic human leukocyte antigen (HLA)-DR+ human umbilical vein endothelial cells (which lack B7 molecules and FcgammaRs) or by blood monocytes (which express low levels of B7 molecules and FcgammaRs) compared with control mAb, but these effects were significantly smaller than those provided by mAb 28.2, a stimulatory mouse anti-human CD28 mAb, at comparable concentrations. However, FK734 generally inhibited cytokine secretion and T cell proliferation in cocultures with human umbilical vein endothelial cells transduced to express CD86. In vivo using SCID/beige mice bearing human skin with adoptively transferred peripheral blood mononuclear cells, administration of FK734 protected human endothelial cell-lined microvessels, significantly but incompletely reducing endothelial cell injury and T cell infiltration into the graft one or two weeks later. CONCLUSIONS FK734 is a partial agonist of CD28 signaling that can reduce human T cell alloresponses in the presence of strong costimulation by B7 molecules in vitro and can reduce T cell-mediated skin allograft rejection in vivo.
Collapse
Affiliation(s)
- Stephen L Shiao
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | |
Collapse
|
83
|
Pree I, Bigenzahn S, Fuchs D, Koporc Z, Nierlich P, Winkler C, Brandacher G, Sykes M, Muehlbacher F, Langer F, Wekerle T. CTLA4Ig promotes the induction of hematopoietic chimerism and tolerance independently of Indoleamine-2,3-dioxygenase. Transplantation 2007; 83:663-7. [PMID: 17353791 PMCID: PMC2992942 DOI: 10.1097/01.tp.0000255594.23445.29] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bone marrow transplantation (BMT) under costimulation blockade induces mixed chimerism and tolerance in rodent models. Recent data, predominantly from in vitro studies, suggest that in addition to blocking the CD28 costimulation pathway CTLA4Ig also acts through upregulating the tryptophan-catabolizing enzyme indoleamine-2,3-dioxygenase (IDO). Here we demonstrate that even though CTLA4Ig is critically required for the induction of chimerism and tolerance in a murine model of nonmyeloablative BMT, IDO activity is not. No significant differences were detectable in the kynurenine to tryptophan ratios (indicative of IDO activity) in sera of BMT recipients treated with CTLA4Ig (tolerant group) versus BMT recipients treated without CTLA4Ig (nontolerant group) versus naïve controls. In vivo inhibition of IDO immediately after BMT with CTLA4Ig or several months thereafter did not block achievement of chimerism and tolerance. Thus, IDO does not play a critical role in the induction or maintenance of chimerism and tolerance in a CTLA4Ig-based BMT model.
Collapse
Affiliation(s)
- Ines Pree
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Sinda Bigenzahn
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocentre, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Zvonimir Koporc
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Patrick Nierlich
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Christiana Winkler
- Division of Biological Chemistry, Biocentre, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Gerald Brandacher
- Dept. of General and Transplant Surgery; Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Megan Sykes
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston MA 02129, USA
| | - Ferdinand Muehlbacher
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Felix Langer
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Wekerle
- Div. of Transplantation, Dept. of Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
84
|
Generation, homeostasis, and regulation of memory T cells in transplantation. Curr Opin Organ Transplant 2007; 12:23-29. [DOI: 10.1097/mot.0b013e328012b293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
85
|
Pree I, Wekerle T. Inducing mixed chimerism and transplantation tolerance through allogeneic bone marrow transplantation with costimulation blockade. Methods Mol Biol 2007; 380:391-403. [PMID: 17876108 DOI: 10.1007/978-1-59745-395-0_25] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Induction of mixed chimerism (i.e., coexistence of donor and recipient hematopoietic cells) through transplantation of allogeneic donor bone marrow under appropriate host conditioning, is one of the most reliable strategies to induce transplantation tolerance. Robust tolerance is evident in mixed chimeras as they permanently accept donor skin grafts while promptly rejecting third party grafts. Although historically, myeloablative and T-cell depleting regimens have been described, milder protocols involving costimulation blockade have recently been developed. The prototypical murine protocol described in this chapter, involves the use of CTLA4Ig and a monoclonal antibody-specific for CD154 (CD40L) for costimulation blockade, 3 Gy of nonmyeloablative total body irradiation and a conventional number of 20 x 10(6) fully allogeneic bone marrow cells. Flow cytometry is used to determine levels of multilineage hematopoietic chimerism and deletion of donor-reactive CD4+ T cells. Tolerance is assessed in vivo by grafting of donor and third party skin.
Collapse
Affiliation(s)
- Ines Pree
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| | | |
Collapse
|
86
|
Abstract
Monoclonal antibodies (MAb) have been shown to be effective in inducing immune tolerance in transplantation and autoimmunity. Several different MAb have tolerogenic properties and their effect has been studied in a range of experimental animal models and, in some cases, in clinical trials. The tolerant state seems to be maintained by CD4+ regulatory T cells (Treg), induced in the periphery, capable of suppressing other T cells specific for the same antigens or antigens presented by the same antigen presenting cells. Furthermore, following the initial induction of Treg cells under MAb treatment, Treg cells themselves can maintain the tolerant state in a dominant way in the absence of the therapeutic MAb or other immunosuppressive agents, and are able to recruit other T cells into the regulatory pool--a process named infectious tolerance.
Collapse
Affiliation(s)
- Ana Agua-Doce
- Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| | | |
Collapse
|
87
|
Bugeon L, Wong KK, Rankin AM, Hargreaves REG, Dallman MJ. Does CTLA4 influence the suppressive effect of CD25+CD4+ regulatory T cells? J Clin Oncol 2006; 82:1334-41. [PMID: 17130783 DOI: 10.1097/01.tp.0000239343.01775.54] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Members of the B7 costimulatory protein family (CD80 and CD86) play a determining role in allograft rejection. Both CD80 and CD86 have naturally occurring splice variants whose roles in transplantation are unknown. Full length CD80 has two immunoglobulin (Ig)-like domains in the extracellular portion, IgC and IgV. In mouse, the isoform IgV-CD80 lacks the IgC-like domain. Here we analyzed the role of mouse IgV-CD80 in heart allograft rejection and search for equivalent splice variants in human. METHODS Mice made deficient for full-length CD80 but which retain expression of the shorter IgV-CD80 (CD80 mice) were used as donor or recipient of a heart allograft. Recipient animals were untreated or pretreated with alloantigen expressing cells and/or treated with CD80 and CTLA4 monoclonal antibodies (mAbs). RESULTS Recipients expressing IgV-CD80 but not full length CD80 exhibited a slight prolongation in survival of either wild-type (Wt) or CD80 grafts. More dramatically, CD80 animals pretreated with donor alloantigen exhibited permanent graft survival, whereas their Wt counterparts rejected their grafts with a median survival of 24 days. This prolonged survival was due to the expression of IgV-CD80 in recipients since treatment with CD80 mAb abrogated the beneficial effect observed. We identified and report here a similar isoform of CD80 from human cDNA encoding a putative soluble, IgV-containing protein. CONCLUSIONS IgV-CD80 bearing recipients show enhanced allograft survival especially after donor alloantigen pretreatment. This together with data from other species suggests that regulation delivered by splice variants of CD80 significantly modulates immunity and may be common across the species.
Collapse
Affiliation(s)
- Laurence Bugeon
- Section of Immunology and Infection and CMMI, Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom.
| | | | | | | | | |
Collapse
|
88
|
Wang Y, Dai H, Liu Z, Cheng X, Tellides G, Dai Z. Neutralizing IL-7 promotes long-term allograft survival induced by CD40/CD40L costimulatory blockade. Am J Transplant 2006; 6:2851-60. [PMID: 17062001 DOI: 10.1111/j.1600-6143.2006.01550.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Memory T cells are somewhat resistant to immunosuppresion. They therefore pose a threat to inducing long-term allograft survival. IL-7 is essential for memory T-cell generation. Here, we investigated whether neutralizing IL-7 promotes allograft survival. We found that neutralizing IL-7 alone did not significantly prolong allograft survival. However, blocking both IL-7 and CD154 signaling synergistically prolonged allograft survival. In contrast, neutralizing IL-2 failed to further prolong allograft survival induced by CD40/CD154 costimulatory blockade. Allospecific memory CD8+ T-cell generation was severely impaired under the treatment of anti-IL-7 plus anti-CD154 Ab while administering recombinant IL-7 enhanced CD8+ memory generation even under donor-specific transfusion plus anti-CD154 Ab treatment. Neutralizing IL-7, but not IL-2, together with blocking CD154 synergistically suppressed the proliferation of naïve/effector CD8+ T cells infiltrating grafts. Nevertheless, neutralizing IL-7 did not alter regulatory T-cell generation while neutralizing IL-2 suppressed their generation. Hence, targeting IL-7 represents a new strategy to prolong allograft survival by acting on both naïve and memory T cells. Long-term allograft survival may be achieved by neutralizing IL-7 plus CD40/CD154 blockade, since CD40/CD154 costimulatory blockade prevents acute rejection while neutralizing IL-7 suppresses the generation of memory T cells that persist and mediate late or chronic rejection.
Collapse
Affiliation(s)
- Y Wang
- Center for Biomedical Research, University of Texas Health Center at Tyler, Tyler, Texas, USA
| | | | | | | | | | | |
Collapse
|
89
|
Teng GG, Turkiewicz AM, Moreland LW. Abatacept: a costimulatory inhibitor for treatment of rheumatoid arthritis. Expert Opin Biol Ther 2006; 5:1245-54. [PMID: 16120053 DOI: 10.1517/14712598.5.9.1245] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
T cell costimulation is believed to be crucial in orchestrating immune responses that lead to inflammation and destruction in rheumatoid arthritis (RA). Abatacept is a novel recombinant CTLA4Ig fusion protein that selectively modulates costimulation via interrupting the CD28:CD80/86 pathway, resulting in downregulation of T cell activation and multiple ensuing effector mechanisms. Abatacept has been shown to be efficacious, either when given alone or in combination with methotrexate, in patients with active RA, including anti-TNF failures. Improvements in clinical signs and symptoms, slowing of radiological progression, and enhancement in patient function and pain have been reported in clinical trials. Infusions were well-tolerated with a favourable safety profile similar to placebo and no appreciable immunogenicity. Abatacept is the first in a new class of biological response modifiers called costimulatory blockers.
Collapse
Affiliation(s)
- Gim Gee Teng
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama, 1717 6th Ave South, Birmingham, AL 35294-7201, USA
| | | | | |
Collapse
|
90
|
Xi Y, Yuan Z, Zhang H, Guan H, Kong F, Liu N, Liang F, Cui J, Guo S, Sun Y, Xi C. Molecular construction and characterization of a novel exotoxin fusion protein that selectively blocks the B7:CD28 costimulatory signal system. J Immunother 2006; 29:586-95. [PMID: 17063121 DOI: 10.1097/01.cji.0000211300.67750.71] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An important strategy for specifically preventing and treating graft-versus-host and host-versus-graft diseases is to selectively block the B7:CD28/cytotoxic T-lymphocyte A4 costimulatory signal system for induced immune tolerance. In this study, a novel recombinant B7-2-L-PE40KDEL fusion protein was created to target the B7:CD28 system. We used a flexible linker sequence (Gly4Ser)4 and overlapping sequence extension to link the cDNAs encoding a human B7-2 extracellular domain and a mutant truncated form of Pseudomonas exotoxin A (PE), PE40KDEL. This B7-2-L-PE40KDEL fusion gene was then inserted into the pTYB4 expression vector, expressed in Escherichia coli, and purified through Ni-NTA mealty affinity-->MonoQ anion exchange-->Superdex75 gel filtration chromatography 3-step purification protocols. Western blotting demonstrated that the B7-2-L-PE40KDEL fusion protein specifically bound antihuman B7-2 monoclonal antibody and anti-pseudomonas exotoxin A antiserum. We used the Antheprot nucleic acid and protein analyzing software to predict the characteristics of this fusion protein, and showed that the fusion did not confer new antigenicities to the fusion protein. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tests demonstrated that at doses ranging from 0.2 to 2 microg/mL, this fusion protein specifically killed CD28-overexpressing Jurkat cells but even at doses of 2 microg did not kill CD28-negative Hut28 cells. The results of a one-way mixed lymphocyte reaction demonstrated that the fusion protein has a range of suppressive effects on HLA class I and II matched related donors and recipients, and HLA class I and II mismatched unrelated donors. Taken together, these results demonstrate that we have developed a novel recombinant human B7-2-L-PE40KDEL exotoxin fusion protein that specifically blocks the B7:CD28 costimulatory signal system in a manner that may be of significant importance in preventing and treating graft-versus-host or host-versus-graft diseases.
Collapse
Affiliation(s)
- Yongzhi Xi
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Affiliated to Academy of Medical Sciences, No. 8, Dongda Avenue, Fengtai District, Beijing 100071, PR China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Li S, Salgar SK, Thanikachalam M, Murdock AD, Gammie JS, Demetris AJ, Zeevi A, Pham SM. Modeling CTLA4-linked autoimmunity with RNA interference in mice. Proc Natl Acad Sci U S A 2006; 136:238-46. [PMID: 17046021 DOI: 10.1016/j.jss.2006.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 05/02/2006] [Accepted: 05/25/2006] [Indexed: 11/27/2022] Open
Abstract
The CTLA4 gene is important for T lymphocyte-mediated immunoregulation and has been associated with several autoimmune diseases, in particular, type 1 diabetes. To model the impact of natural genetic variants of CTLA4, we constructed RNA interference (RNAi) "knockdown" mice through lentiviral transgenesis. Variegation of expression was observed in founders but proved surmountable because it reflected parental imprinting, with derepression by transmission from male lentigenics. Unlike the indiscriminate multiorgan autoimmune phenotype of the corresponding knockout mice, Ctla4 knockdown animals had a disease primarily focused on the pancreas, with rapid progression to diabetes. As with the human disease, the knockdown phenotype was tempered by genetic-modifier loci. RNAi should be more pertinent than gene ablation in modeling disease pathogenesis linked to a gene-dosage variation.
Collapse
Affiliation(s)
- Sen Li
- Division of Cardiothoracic Surgery, University of Miami School of Medicine, Miami, Florida 33156, USA
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Ndejembi MP, Tang AL, Farber DL. Reshaping the past: Strategies for modulating T-cell memory immune responses. Clin Immunol 2006; 122:1-12. [PMID: 16916619 DOI: 10.1016/j.clim.2006.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 06/26/2006] [Indexed: 02/08/2023]
Abstract
Memory T cells are generated following an initial encounter with antigen, persist over the lifetime of an individual, and mediate rapid and robust functional responses upon antigenic recall. While immune memory is generally associated with protective immune response to pathogens, memory T cells can be generated to diverse types of antigens including autoantigens and alloantigens through homologous or crossreactive priming and comprise the majority of circulating T cells during adulthood. Memory T cells can therefore play critical roles in propagating and perpetuating autoimmune disease and in mediating allograft rejection, although the precise pathways for regulation of memory immune responses remain largely undefined. Moreover, evaluating and designing strategies to modulate memory T-cell responses are challenging given the remarkable heterogeneity of memory T cells, with different subsets predominating in lymphoid versus non-lymphoid tissue sites. In this review, we discuss what is presently known regarding the effect of current immunomodulation strategies on the memory T-cell compartment and potential strategies for controlling immunological recall.
Collapse
Affiliation(s)
- Modesta P Ndejembi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
93
|
Newland A, Russ G, Krishnan R. Natural killer cells prime the responsiveness of autologous CD4+ T cells to CTLA4-Ig and interleukin-10 mediated inhibition in an allogeneic dendritic cell-mixed lymphocyte reaction. Immunology 2006; 118:216-23. [PMID: 16771856 PMCID: PMC1782277 DOI: 10.1111/j.1365-2567.2006.02359.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cytotoxic T-lymphocyte antigen 4 immunoglobulin (CTLA4-Ig) and interleukin (IL)-10 are immunomodulatory molecules which target CD28 costimulation by acting either directly or indirectly on the CD80/86 receptors on dendritic cells (DCs). This study examined the effect of combined treatment with CTLA4-Ig and IL-10 on T-cell responsiveness in a dendritic cell-mixed lymphocyte reaction (DC-MLR). T cells derived from nylon wool enrichment (NWT cells) demonstrated 15% (P = 0.006) and 10% (P = 0.0015) inhibition of proliferation with suboptimal doses of IL-10 (5 ng/ml) and CTLA4-Ig (20 ng/ml), respectively. Combined treatment with both agents resulted in 38% inhibition (P = 0.004) of the MLR response compared with untreated controls. In contrast to NWT cells, which consisted of CD4+, CD8+ and CD56+ (NK) cells, purified CD4+ T cells were less responsive to immunomodulation by CTLA4-Ig and IL-10. Repletion of the CD4+ T cells with NK cells restored IL-10 and CTLA4-Ig mediated immunomodulation, suggesting a role for NK cells in the regulation of DC-T-cell interactions. The specific effect of NK cells on DC activation was demonstrated by CD80 up-regulation on DCs in the absence of T cells. However, in the absence of DCs, NK cells augmented the proliferation of autologous CD4+ T cells stimulated by anti-CD3 monoclonal antibody (mAb), which was blocked by CTLA4-Ig. It is proposed that, in the MLR, immunomodulation by suboptimal CTLA4-Ig and IL-10 is influenced by cellular interactions of NK cells with DCs and T cells involving DC lysis and costimulation. Thus, NK cells prime both DCs and T cells to low doses of CTLA4-Ig and IL-10 during alloimmune responses, providing evidence for the potential interaction between innate and adaptive immunity.
Collapse
Affiliation(s)
- Ashley Newland
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| | - Graeme Russ
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| | - Ravi Krishnan
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| |
Collapse
|
94
|
Moore DJ, Markmann JF, Deng S. Avenues for immunomodulation and graft protection by gene therapy in transplantation. Transpl Int 2006; 19:435-45. [PMID: 16771864 DOI: 10.1111/j.1432-2277.2006.00314.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the only definitive therapy for many causes of end-organ failure. However, the universal success of this therapy is limited by chronic allograft rejection, the side effects of chronic immunosuppressive therapy, and a severe shortage of donor organs. Presently, the success of solid-organ transplantation depends on the continuous administration of toxic and nonspecific immunosuppressive agents, therapies that present risks for opportunistic infection, malignancy, and a variety of agent-specific side effects. To promote the use of transplantation with limited risk of long-term sequelae, three dominant research challenges emerge: (i) elimination of the need for exogenous immunosuppression by immunological tolerance induction; (ii) prevention of chronic rejection/graft dysfunction; and (iii) expansion of available organs for transplantation. Gene therapy may provide significant advances and solutions in each of these areas. Rejection of the graft in the immediate post-transplant period has been attacked through the transfer of immunomodulatory molecules in addition to tolerance inducing approaches. Chronic graft rejection may be similarly addressed through permanent tolerance induction or alternatively through the introduction of molecules to resist chronic graft damage. Genetic manipulation of stem cells may ultimately produce transgenic animals to serve as tissue donors to overcome the limited donor organ supply. This review will highlight ongoing developments in the translation of gene therapy approaches to the challenges inherent in transplantation.
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
95
|
|
96
|
Miao CH, Ye P, Thompson AR, Rawlings DJ, Ochs HD. Immunomodulation of transgene responses following naked DNA transfer of human factor VIII into hemophilia A mice. Blood 2006; 108:19-27. [PMID: 16507778 PMCID: PMC1895820 DOI: 10.1182/blood-2005-11-4532] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Accepted: 02/15/2006] [Indexed: 11/20/2022] Open
Abstract
A robust humoral immune response against human factor VIII (hFVIII) following naked DNA transfer into immunocompetent hemophilia A mice completely inhibits circulating FVIII activity despite initial high-level hFVIII gene expression. To prevent this undesirable response, we compared transient immunomodulation strategies. Eight groups of mice (n = 4-9 per group) were treated with naked DNA transfer of pBS-HCRHPI-hFVIIIA simultaneously with immunosuppressive reagents that included cyclosporine A (CSA), rapamycin (RAP), mycophenylate mofetil (MMF), a combination of CSA and MMF, a combination of RAP and MMF, a monoclonal antibody against murine CD40 ligand (MR1), recombinant murine Ctla4Ig, and a combination of MR1 and Ctla4Ig. All animals except those receiving only CSA exhibited delayed or absent immune responses against hFVIII. The most effective immunosuppressive regimen, the combination of Ctla4Ig and MR1, prevented inhibitor formation in 8 of 9 animals; the ninth had transient low-titer antibodies. All 9 mice of this group produced persistent, therapeutic levels of hFVIII for more than 6 months. When challenged with the T-dependent antigen bacteriophage Phix174, tolerized mice exhibited normal primary and secondary antibody responses, suggesting that transient immunomodulation to disrupt B/T-cell interaction at the time of plasmid injection effectively promoted long-term immune tolerance specific for hFVIII.
Collapse
Affiliation(s)
- Carol H Miao
- Children's Hospital and Regional Medical Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | |
Collapse
|
97
|
Bluestone JA, St Clair EW, Turka LA. CTLA4Ig: bridging the basic immunology with clinical application. Immunity 2006; 24:233-8. [PMID: 16546089 DOI: 10.1016/j.immuni.2006.03.001] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
After a very long and windy road, in December of 2005 the FDA approved CTLA4Ig for the treatment of rheumatoid arthritis. Orencia is the first-in-class antagonist of CD28 costimulation. In this perspective, we discuss the science that led to CTLA4Ig development and the clinical challenges in bringing the drug from the bench to the bedside.
Collapse
Affiliation(s)
- Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA.
| | | | | |
Collapse
|
98
|
Neujahr DC, Chen C, Huang X, Markmann JF, Cobbold S, Waldmann H, Sayegh MH, Hancock WW, Turka LA. Accelerated memory cell homeostasis during T cell depletion and approaches to overcome it. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:4632-9. [PMID: 16585554 DOI: 10.4049/jimmunol.176.8.4632] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Partial T cell depletion is used in solid organ transplantation as a valuable strategy of peritransplant induction immunosuppression. Using a murine cardiac allograft model, we recently demonstrated that this led to lymphopenia-induced (homeostatic) proliferation among the residual nondepleted lymphocytes. Rather than promoting tolerance, peritransplant T cell-depleting Abs actually resulted in resistance to tolerance induction by costimulatory blockade. In this study we show that memory T cells predominate shortly after subtotal lymphodepletion due to two distinct mechanisms: relative resistance to depletion and enhanced homeostatic proliferation. In contrast, regulatory cells (CD4+ CD25+ Foxp3+) are depleted as efficiently as nonregulatory cells and exhibit reduced homeostatic expansion compared with memory cells. The resistance to tolerance induction seen with subtotal T cell depletion can be overcome in two different ways: first, by the adoptive transfer of additional unprimed regulatory cells at the time of transplant, and second, by the adjunctive use of nondepleting anti-CD4 and anti-CD8 mAbs, which effectively block homeostatic expansion. We conclude that the resistance to tolerance induction seen after subtotal lymphocyte depletion can be attributed to alterations in the balance of naive, memory, and regulatory T cells. These data have clinically relevant implications related to the development of novel strategies to overcome resistance to tolerance.
Collapse
Affiliation(s)
- David C Neujahr
- Department of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Nickoloff BJ, Stevens SR. What have we learned in dermatology from the biologic therapies? J Am Acad Dermatol 2006; 54:S143-51. [PMID: 16488330 DOI: 10.1016/j.jaad.2005.10.059] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 06/01/2005] [Accepted: 10/08/2005] [Indexed: 11/16/2022]
Abstract
Recent advances in our basic understanding of immunology, specifically the roles of various cell types involved in immune response and the action of cytokines they produce, has radically changed our understanding of the origin of inflammatory dermatoses, and other autoimmune diseases. Broadened comprehension of the immune response on a molecular level has facilitated the development of biologic therapeutics for the treatment of psoriasis, atopic dermatosis, and other inflammatory conditions. However, despite major advances in development and use of targeted biologics for controlling autoimmune disease, effective cures for these conditions remain to be developed and genetic determinants of predisposition to such diseases remain to be identified. Here, we review the history of our understanding of inflammatory dermatoses, traditional and new treatment approaches, and future directions for research and therapy in this area.
Collapse
|
100
|
Luo G, Wu J, Chen X, He W, Yi S, Xie Z, Zheng J, Zhu J. CTLA4Ig introduced by adenovirus vector locally to prolong the survival of xenogeneic skin grafts on rat burn wounds. ACTA ACUST UNITED AC 2006; 59:1209-15. [PMID: 16385301 DOI: 10.1097/01.ta.0000189002.59950.dc] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The purpose of this study was to explore an applicable approach for prolonging the survival of heterogenetic skin grafts on burn wounds with CTLA4Ig. METHODS An adenovirus vector named Ad-CTLA4Ig, which could express human CTLA4Ig fusion protein, was constructed. Infecting and replicating in 293 cells, more Ad-CTLA4Ig and recombinant human CTLA4Ig (rhCTLA4Ig) were prepared, respectively. In a rat flame thermal injury model, the effect of rhCTLA4Ig on survival time of human skin graft on the eschar-excised rat burn wound was observed. Meanwhile, the efficiency of Ad-CTLA4Ig infecting cultured skin fibroblasts, keratinocytes, and partial-thickness skin samples were checked by CTLA4Ig expression essay. Then, the Ad-CTLA4Ig was administered locally on the eschar-excised wound and dermis of the skin graft, and the survival time of the human skin graft on burn wound was measured. The influence of the systemic immune function by rhCTLA4Ig and Ad-CTLA4Ig were also determined. RESULTS The prepared rhCTLA4Ig from the supernatant of Ad-CTLA4Ig-infected 293 cells was verified by sodium dodecyl sulfate polyacrylamide gel electrophoresis sodium dodecyl sulfate poly-acrylamide gel electrophoresis and Western blot. It was found that CTLA4Ig could significantly prolong the xenogeneic skin graft survival in a dosage-dependent manner. Interestingly, the survival time was longer when CTLA4Ig was used 24 hours posttransplantation than that at hour 0. The expression of CTLA4Ig could be observed in the cultured skin fibroblasts, keratinocytes, and skin pieces soon after Ad-CTLA4Ig transfection, as demonstrated by either immunocellular chemistry or immunohistochemistry assay. When Ad-CTLA4Ig was locally administered during skin transplantation on burn wound, the survival time was increased from 7.9 days of control group to 21.6 days, whereas the systemic immune function was not affected. CONCLUSION Administration of Ad-CTLA4Ig locally could prolong the survival time of xenogeneic skin graft on burn wound without significantly influencing the systemic immune function.
Collapse
Affiliation(s)
- Gaoxing Luo
- Burn Research Institute, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|