Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Am J Transplant ; 17(9): 2285-2299, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28502128

ABSTRACT

Recently, newer therapies have been designed to more specifically target rejection in an effort to improve efficacy and limit unwanted toxicity. Belatacept, a CD28-CD80/86 specific reagent, is associated with superior patient survival and graft function compared with traditional therapy, but its adoption as a mainstay immunosuppressive therapy has been tempered by increased rejection rates. It is essential that the underlying mechanisms associated with this rejection be elucidated before belatacept is more widely used. To that end, we designed a study in a nonhuman primate kidney transplant model where animals were treated with either a belatacept- or a tacrolimus-based immunosuppressive regimen. Interestingly, we found that elevated pretransplant frequencies of CD28+ CD8+ TEMRA cells are associated with rejection on belatacept but not tacrolimus treatment. Further analysis showed that the CD28+ CD8+ TEMRA cells rapidly lose CD28 expression after transplant in those animals that go on to reject with the allograft infiltrate being predominantly CD28- . These data suggest that CD28+ memory T cells may be resistant to belatacept, capable of further differentiation including loss of CD28 expression while maintaining effector function. The unique signaling requirements of CD28+ memory T cells provide opportunities for the development of targeted therapies, which may synergize with belatacept to prevent costimulation-independent rejection.


Subject(s)
Abatacept/pharmacology , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Resistance/immunology , Graft Rejection/immunology , Immunologic Memory/immunology , Kidney Transplantation/adverse effects , Animals , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Rejection/pathology , Graft Survival , Immunosuppressive Agents/pharmacology , Kidney Failure, Chronic/surgery , Kidney Function Tests , Macaca mulatta , Postoperative Complications
2.
Am J Transplant ; 17(5): 1182-1192, 2017 May.
Article in English | MEDLINE | ID: mdl-28097811

ABSTRACT

The advent of costimulation blockade provides the prospect for targeted therapy with improved graft survival in transplant patients. Perhaps the most effective costimulation blockade in experimental models is the use of reagents to block the CD40/CD154 pathway. Unfortunately, successful clinical translation of anti-CD154 therapy has not been achieved. In an attempt to develop an agent that is as effective as previous CD154 blocking antibodies but lacks the risk of thromboembolism, we evaluated the efficacy and safety of a novel anti-human CD154 domain antibody (dAb, BMS-986004). The anti-CD154 dAb effectively blocked CD40-CD154 interactions but lacked crystallizable fragment (Fc) binding activity and resultant platelet activation. In a nonhuman primate kidney transplant model, anti-CD154 dAb was safe and efficacious, significantly prolonging allograft survival without evidence of thromboembolism (Median survival time 103 days). The combination of anti-CD154 dAb and conventional immunosuppression synergized to effectively control allograft rejection (Median survival time 397 days). Furthermore, anti-CD154 dAb treatment increased the frequency of CD4+ CD25+ Foxp3+ regulatory T cells. This study demonstrates that the use of a novel anti-CD154 dAb that lacks Fc binding activity is safe without evidence of thromboembolism and is equally as potent as previous anti-CD154 agents at prolonging renal allograft survival in a nonhuman primate preclinical model.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Ligand/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Immunoglobulin G/immunology , Kidney Transplantation/adverse effects , Animals , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival/drug effects , Kidney Function Tests , Primates , Risk Factors , T-Lymphocytes, Regulatory/immunology , Transplantation Immunology
3.
Am J Transplant ; 16(5): 1456-64, 2016 05.
Article in English | MEDLINE | ID: mdl-26602755

ABSTRACT

Costimulation blockade with the fusion protein belatacept provides a desirable side effect profile and improvement in renal function compared with calcineurin inhibition in renal transplantation. This comes at the cost of increased rates of early acute rejection. Blockade of the integrin molecule leukocyte function-associated antigen 1 (LFA-1) has been shown to be an effective adjuvant to costimulation blockade in a rigorous nonhuman primate (NHP) model of islet transplantation; therefore, we sought to test this combination in an NHP renal transplant model. Rhesus macaques received belatacept maintenance therapy with or without the addition of LFA-1 blockade, which was achieved using a murine-derived LFA-1-specific antibody TS1/22. Additional experiments were performed using chimeric rhesus IgG1 (TS1/22R1) or IgG4 (TS1/22R4) variants, each engineered to limit antibody clearance. Despite evidence of proper binding to the target molecule and impaired cellular egress from the intravascular space indicative of a therapeutic effect similar to prior islet studies, LFA-1 blockade failed to significantly prolong graft survival. Furthermore, evidence of impaired protective immunity against cytomegalovirus was observed. These data highlight the difficulties in translating treatment regimens between organ models and suggest that the primarily vascularized renal model is more robust with regard to belatacept-resistant rejection than the islet model.


Subject(s)
Abatacept/therapeutic use , Disease Models, Animal , Graft Rejection/prevention & control , Graft Survival/immunology , Kidney Failure, Chronic/immunology , Kidney Transplantation/adverse effects , Lymphocyte Function-Associated Antigen-1/immunology , Animals , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Rejection/pathology , Immunologic Memory , Kidney Failure, Chronic/surgery , Kidney Function Tests , Lymphocyte Function-Associated Antigen-1/administration & dosage , Macaca mulatta , Transplantation, Homologous
4.
Am J Transplant ; 15(8): 2240-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26139552

ABSTRACT

Vascularized composite allografts (VCAs) are technically feasible. Similar to other organ transplants, VCAs are hampered by the toxicity and incomplete efficacy associated with conventional immunosuppression. Complications attributable to calcineurin inhibitors remain prevalent in the clinical cases reported to date, and these loom particularly large given the nonlifesaving nature of VCAs. Additionally, acute rejection remains almost ubiquitous, albeit controllable with current agents. Costimulation blockade offers the potential to provide prophylaxis from rejection without the adverse consequences of calcineurin-based regimens. In this study, we used a nonhuman-primate model of VCA in conjunction with immunosuppressive regimens containing combinations of B7-specific costimulation blockade with and without adhesion blockade with LFA3-Ig to determine what adjunctive role these agents could play in VCA transplantation when combined with more conventional agents. Compared to tacrolimus, the addition of belatacept improved rejection free allograft survival. The combination with LFA3-Ig reduced CD2(hi) memory T cells, however did not provide additional protection against allograft rejection and hindered protective immunity. Histology paralleled clinical histopathology and Banff grading. These data provide the basis for the study of costimulation blockade in VCA in a relevant preclinical model.


Subject(s)
Allografts , Neovascularization, Pathologic , Animals , Primates
5.
Am J Transplant ; 15(5): 1241-52, 2015 May.
Article in English | MEDLINE | ID: mdl-25702898

ABSTRACT

Islet xenotransplantation is a potential treatment for diabetes without the limitations of tissue availability. Although successful experimentally, early islet loss remains substantial and attributed to an instant blood-mediated inflammatory reaction (IBMIR). This syndrome of islet destruction has been incompletely defined and characterization in pig-to-primate models has been hampered by logistical and statistical limitations of large animal studies. To further investigate IBMIR, we developed a novel in vivo dual islet transplant model to precisely characterize IBMIR as proof-of-concept that this model can serve to properly control experiments comparing modified xenoislet preparations. WT and α1,3-galactosyltransferase knockout (GTKO) neonatal porcine islets were studied in nonimmunosuppressed rhesus macaques. Inert polyethylene microspheres served as a control for the effects of portal embolization. Digital analysis of immunohistochemistry targeting IBMIR mediators was performed at 1 and 24 h after intraportal islet infusion. Early findings observed in transplanted islets include complement and antibody deposition, and infiltration by neutrophils, macrophages and platelets. Insulin, complement, antibody, neutrophils, macrophages and platelets were similar between GTKO and WT islets, with increasing macrophage infiltration at 24 h in both phenotypes. This model provides an objective and internally controlled study of distinct islet preparations and documents the temporal histology of IBMIR.


Subject(s)
Inflammation/immunology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Animals , Animals, Genetically Modified , Blood Glucose/chemistry , Blood Platelets/immunology , Complement Activation , Disease Models, Animal , Galactosyltransferases/genetics , Immunohistochemistry , Macaca mulatta , Macrophages/immunology , Neutrophils/immunology , Phenotype , Swine , Time Factors , Transplantation, Heterologous
6.
Am J Transplant ; 15(4): 984-92, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25703015

ABSTRACT

Costimulation blockade with the B7-CD28 pathway-specific agent belatacept is now used in clinical kidney transplantation, but its efficacy remains imperfect. Numerous alternate costimulatory pathways have been proposed as targets to synergize with belatacept, one of which being the inducible costimulator (ICOS)-ICOS ligand (ICOS-L) pathway. Combined ICOS-ICOS-L and CD28-B7 blockade has been shown to prevent rejection in mice, but has not been studied in primates. We therefore tested a novel ICOS-Ig human Fc-fusion protein in a nonhuman primate (NHP) kidney transplant model alone and in combination with belatacept. ICOS-Ig did not prolong rejection-free survival as a monotherapy or in combination with belatacept. In ICOS-Ig alone treated animals, most graft-infiltrating CD4(+) and CD8(+) T cells expressed ICOS, and ICOS(+) T cells were present in peripheral blood to a lesser degree. Adding belatacept reduced the proportion of graft-infiltrating ICOS(+) T cells and virtually eliminated their presence in peripheral blood. Graft-infiltrating T cells in belatacept-resistant rejection were primarily CD8(+) CD28(-) , but importantly, very few CD8(+) CD28(-) T cells expressed ICOS. We conclude that ICOS-Ig, alone or combined with belatacept, does not prolong renal allograft survival in NHPs. This may relate to selective loss of ICOS with CD28 loss.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/metabolism , Kidney Transplantation , Abatacept/therapeutic use , Animals , Graft Rejection/prevention & control , Immunologic Memory , Immunophenotyping , Macaca mulatta , Pilot Projects , T-Lymphocytes/immunology
7.
Am J Transplant ; 13(12): 3085-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24119188

ABSTRACT

The integrin αvß6 activates latent transforming growth factor-ß (TGF-ß) within the kidney and may be a target for the prevention of chronic allograft fibrosis after kidney transplantation. However, TGF-ß also has known immunosuppressive properties that are exploited by calcineurin inhibitors (CNIs); thus, the net benefit of αvß6 inhibition remains undetermined. To assess the acute impact of interference with αvß6 on acute rejection, we tested a humanized αvß6-specific monoclonal antibody (STX-100) in a randomized, double-blinded, placebo-controlled nonhuman primate renal transplantation study to evaluate whether αvß6 blockade alters the risk of acute rejection during CNI-based immunosuppression. Rhesus monkeys underwent renal allotransplantation under standard CNI-based maintenance immunosuppression; 10 biopsy-confirmed rejection-free animals were randomized to receive weekly STX-100 or placebo. Animals treated with STX-100 experienced significantly decreased rejection-free survival compared to placebo animals (p = 0.049). Immunohistochemical analysis confirmed αvß6 ligand presence, and αvß6 staining intensity was lower in STX-100-treated animals (p = 0.055), indicating an apparent blockade effect of STX-100. LAP, LTBP-1 and TGF-ß were all decreased in animals that rejected on STX-100 compared to those that rejected on standard immunosuppression alone, suggesting a relevant effect of αvß6 blockade on local TGF-ß. These data caution against the use of αvß6 blockade to achieve TGF-ß inhibition in kidney transplantation.


Subject(s)
Antibodies, Monoclonal/adverse effects , Immunosuppressive Agents/adverse effects , Integrins/antagonists & inhibitors , Kidney Transplantation/methods , Allografts , Animals , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm , Biopsy , Graft Rejection , Immunosuppression Therapy , Macaca mulatta , Pilot Projects , Random Allocation , Transforming Growth Factor beta1/blood
8.
J Comp Pathol ; 149(4): 524-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24016782

ABSTRACT

A 25-year-old, female rhesus macaque (Macaca mulatta) presented with a history of weight loss despite a normal appetite and supportive care. The animal was humanely destroyed due to poor prognosis. Post-mortem examination revealed a focally extensive, firm, white annular constriction at the ileocaecal junction and an incidental finding of a pale white nodule approximately 0.8 cm in diameter in the left renal pelvis. Based on the microscopical findings, ileocaecal adenocarcinoma and renal pelvis transitional cell carcinoma (TCC) were diagnosed. The use of cytokeratin (CK)-7 and -20 and uroplakin III as potential renal TCC markers was evaluated. The neoplastic cells were labelled intensely with antibodies to uroplakin III, but not to CK-7 or -20. Spontaneous intestinal adenocarcinoma has been documented in the rhesus macaque, but concurrent renal pelvis TCC is highly unusual.


Subject(s)
Adenocarcinoma/veterinary , Carcinoma, Transitional Cell/veterinary , Ileal Neoplasms/veterinary , Kidney Neoplasms/veterinary , Monkey Diseases/pathology , Neoplasms, Multiple Primary/veterinary , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/analysis , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Female , Ileal Neoplasms/metabolism , Ileal Neoplasms/pathology , Ileocecal Valve/pathology , Immunohistochemistry , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Pelvis/pathology , Macaca mulatta , Monkey Diseases/metabolism , Neoplasms, Multiple Primary/metabolism , Neoplasms, Multiple Primary/pathology
9.
Am J Transplant ; 13(2): 320-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23311611

ABSTRACT

Belatacept is an inhibitor of CD28/B7 costimulation that is clinically indicated as a calcineurin inhibitor (CNI) alternative in combination with mycophenolate mofetil and steroids after renal transplantation. We sought to develop a clinically translatable, nonlymphocyte depleting, belatacept-based regimen that could obviate the need for both CNIs and steroids. Thus, based on murine data showing synergy between costimulation blockade and mTOR inhibition, we studied rhesus monkeys undergoing MHC-mismatched renal allotransplants treated with belatacept and the mTOR inhibitor, sirolimus. To extend prior work on costimulation blockade-resistant rejection, some animals also received CD2 blockade with alefacept (LFA3-Ig). Belatacept and sirolimus therapy successfully prevented rejection in all animals. Tolerance was not induced, as animals rejected after withdrawal of therapy. The regimen did not deplete T cells. Alefecept did not add a survival benefit to the optimized belatacept and sirolimus regimen, despite causing an intended depletion of memory T cells, and caused a marked reduction in regulatory T cells. Furthermore, alefacept-treated animals had a significantly increased incidence of CMV reactivation, suggesting that this combination overly compromised protective immunity. These data support belatacept and sirolimus as a clinically translatable, nondepleting, CNI-free, steroid-sparing immunomodulatory regimen that promotes sustained rejection-free allograft survival after renal transplantation.


Subject(s)
Immunoconjugates/administration & dosage , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/methods , Sirolimus/administration & dosage , T-Lymphocytes/immunology , Abatacept , Animals , CD2 Antigens/metabolism , CD3 Complex/metabolism , Disease Progression , Disease-Free Survival , Graft Rejection , Graft Survival , Immunologic Memory , Macaca mulatta , Phenotype , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous , Treatment Outcome
10.
Am J Transplant ; 13(2): 312-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279640

ABSTRACT

Calcineurin inhibitors (CNI) and steroids are known to promote insulin resistance, and their avoidance after islet transplantation is preferred from a metabolic standpoint. Belatacept, a B7-specific mediator of costimulation blockade (CoB), is clinically indicated as a CNI alternative in renal transplantation, and we have endeavored to develop a clinically translatable, belatacept-based regimen that could obviate the need for both CNIs and steroids. Based on the known synergy between CoB and mTOR inhibition, we studied rhesus monkeys undergoing MHC-mismatched islet allotransplants treated with belatacept and the mTOR inhibitor, sirolimus. To extend prior work on CoB-resistant rejection, some animals also received CD2 blockade with alefacept (LFA3-Ig). Nine rhesus macaques were rendered diabetic with streptozotocin and underwent islet allotransplantation. All received belatacept and sirolimus; six also received alefacept. Belatacept and sirolimus significantly prolonged rejection-free graft survival (median 225 days compared to 8 days in controls receiving basiliximab and sirolimus; p = 0.022). The addition of alefacept provided no additional survival benefit, but was associated with Cytomegalovirus reactivation in four of six animals. No recipients produced donor-specific alloantibodies. The combination of belatacept and sirolimus successfully prevents islet allograft survival in rhesus monkeys, but induction with alefacept provides no survival benefit and increases the risk of viral reactivation.


Subject(s)
Immunoconjugates/administration & dosage , Islets of Langerhans Transplantation/methods , Recombinant Fusion Proteins/administration & dosage , Sirolimus/administration & dosage , Transplantation, Homologous/methods , Abatacept , Alefacept , Animals , Antibodies, Monoclonal/administration & dosage , Basiliximab , C-Peptide/metabolism , Diabetes Mellitus, Experimental , Graft Survival , Histocompatibility Antigens/immunology , Immunosuppressive Agents/administration & dosage , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Macaca mulatta , Steroids/administration & dosage
11.
Am J Transplant ; 12(8): 2079-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22845909

ABSTRACT

The importance of CD40/CD154 costimulatory pathway blockade in immunosuppression strategies is well-documented. Efforts are currently focused on monoclonal antibodies specific for CD40 because of thromboembolic complications associated with monoclonal antibodies directed towards CD154. Here we present the rational development and characterization of a novel antagonistic monoclonal antibody to CD40. Rhesus macaques were treated with the recombinant anti-CD40 mAb, 2C10, or vehicle before immunization with keyhole limpet hemocyanin (KLH). Treatment with 2C10 successfully inhibited T cell-dependent antibody responses to KLH without significant peripheral B cell depletion. Subsequently, MHC-mismatched macaques underwent intraportal allogeneic islet transplantation and received basiliximab and sirolimus with or without 2C10. Islet graft survival was significantly prolonged in recipients receiving 2C10 (graft survival time 304, 296, 265, 163 days) compared to recipients receiving basiliximab and sirolimus alone (graft survival time 8, 8, 10 days). The survival advantage conferred by treatment with 2C10 provides further evidence for the importance of blockade of the CD40/CD154 pathway in preventing alloimmune responses. 2C10 is a particularly attractive candidate for translation given its favorable clinical profile.


Subject(s)
Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , Graft Survival , Islets of Langerhans Transplantation , Animals , Antibody Formation , Humans , Macaca mulatta
12.
Am J Transplant ; 12(9): 2395-405, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22776408

ABSTRACT

Chronic allograft rejection is a major impediment to long-term transplant success. Humoral immune responses to alloantigens are a growing clinical problem in transplantation, with mounting evidence associating alloantibodies with the development of chronic rejection. Nearly a third of transplant recipients develop de novo antibodies, for which no established therapies are effective at preventing or eliminating, highlighting the need for a nonhuman primate model of antibody-mediated rejection. In this report, we demonstrate that depletion using anti-CD3 immunotoxin (IT) combined with maintenance immunosuppression that included tacrolimus with or without alefacept reliably prolonged renal allograft survival in rhesus monkeys. In these animals, a preferential skewing toward CD4 repopulation and proliferation was observed, particularly with the addition of alefacept. Furthermore, alefacept-treated animals demonstrated increased alloantibody production (100%) and morphologic features of antibody-mediated injury. In vitro, alefacept was found to enhance CD4 effector memory T cell proliferation. In conclusion, alefacept administration after depletion and with tacrolimus promotes a CD4+memory T cell and alloantibody response, with morphologic changes reflecting antibody-mediated allograft injury. Early and consistent de novo alloantibody production with associated histological changes makes this nonhuman primate model an attractive candidate for evaluating targeted therapeutics.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Isoantibodies/immunology , Animals , Immunohistochemistry , Immunologic Memory , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion , Macaca mulatta , Male
13.
Am J Transplant ; 12(7): 1755-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22642491

ABSTRACT

Although there is evidence linking hematopoietic chimerism induction and solid organ transplant tolerance, the mechanistic requirements for chimerism-induced tolerance are not clearly elucidated. To address this, we used an MHC-defined primate model to determine the impact of impermanent, T cell-poor, mixed-chimerism on renal allograft survival. We compared two cohorts: one receiving a bone marrow and renal transplant ("BMT/renal") and one receiving only a renal transplant. Both cohorts received maintenance immunosuppression with CD28/CD40-directed costimulation blockade and sirolimus. As previously demonstrated, this transplant strategy consistently induced compartmentalized donor chimerism, (significant whole-blood chimerism, lacking T cell chimerism). This chimerism was not sufficient to prolong renal allograft acceptance: the BMT/renal mean survival time (MST, 76 days) was not significantly different than the renal transplant alone MST (85 days, p = 0.46), with histopathology documenting T cell mediated rejection. Flow cytometric analysis revealed significant enrichment for CD28-/CD95+ CD4+ and CD8+ Tem cells in the rejected kidney, suggesting a link between CD28-negative Tem and costimulation blockade-resistant rejection. These results suggest that in some settings, transient T cell-poor chimerism is not sufficient to induce tolerance to a concurrently placed renal allograft and that the presence of this chimerism per se is not an independent biomarker to identify tolerance.


Subject(s)
Bone Marrow Transplantation , Chimerism , Graft Rejection , Kidney Transplantation , Animals , Macaca mulatta , Transplantation, Homologous
14.
Am J Transplant ; 12(7): 1918-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22458552

ABSTRACT

Islet transplantation to treat type 1 diabetes has been limited in part by toxicities of current immunosuppression and recipient humoral sensitization. Blockade of the CD28/CD80/86 and CD40/CD154 pathways has shown promise to remedy both these limitations, but translation has been hampered by difficulties in translating CD154-directed therapies. Prior CD40-directed regimens have led to prolonged islet survival, but fail to prevent humoral allosensitization. We therefore evaluated the addition of CTLA4Ig to a CD40 blockade-based regimen in nonhuman primate (NHP) alloislet transplantation. Diabetic rhesus macaques were transplanted allogeneic islets using the CD40-specific antibody 3A8, basiliximab induction, and sirolimus with or without CTLA4Ig maintenance therapy. Allograft survival was determined by fasting blood glucose levels and flow cytometric techniques were used to test for donor-specific antibody (DSA) formation. CTLA4Ig plus 3A8, basiliximab and sirolimus was well tolerated and induced long-term islet allograft survival. The addition of CTLA4Ig prevented DSA formation, but did not facilitate withdrawal of the 3A8-based regimen. Thus, CTLA4Ig combines with a CD40-specific regimen to prevent DSA formation in NHPs, and offers a potentially translatable calcineurin inhibitor-free protocol inclusive of a single investigational agent for use in clinical islet transplantation without relying upon CD154 blockade.


Subject(s)
Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , Immunoconjugates/immunology , Islets of Langerhans Transplantation , Isoantibodies/biosynthesis , Abatacept , Animals , Graft Survival/immunology , Macaca mulatta
15.
Am J Transplant ; 12(7): 1765-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22458586

ABSTRACT

Immunosuppressive therapies that block the CD40/CD154 costimulatory pathway have proven to be uniquely effective in preclinical xenotransplant models. Given the challenges facing clinical translation of CD40/CD154 pathway blockade, we examined the efficacy and tolerability of CD40/CD154 pathway-sparing immunomodulatory strategies in a pig-to-nonhuman primate islet xenotransplant model. Rhesus macaques were rendered diabetic with streptozocin and given an intraportal infusion of ≈ 50 000 islet equivalents/kg wild-type neonatal porcine islets. Base immunosuppression for all recipients included maintenance therapy with belatacept and mycophenolate mofetil plus induction with basiliximab and LFA-1 blockade. Cohort 1 recipients (n = 3) were treated with the base regimen alone; cohort 2 recipients (n = 5) were additionally treated with tacrolimus induction and cohort 3 recipients (n = 5) were treated with alefacept in place of basiliximab, and more intense LFA-1 blockade. Three of five recipients in both cohorts 2 and 3 achieved sustained insulin-independent normoglycemia (median rejection-free survivals 60 and 111 days, respectively), compared to zero of three recipients in cohort 1. These data show that CD40/CD154 pathway-sparing regimens can promote xenoislet survival. Further optimization of these strategies is warranted to aid the clinical translation of islet xenotransplantation.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Graft Survival/immunology , Heterografts , Immunosuppressive Agents/administration & dosage , Islets of Langerhans Transplantation , Animals , Cohort Studies , Diabetes Mellitus, Experimental/surgery , Immunologic Memory , Macaca mulatta , Swine , T-Lymphocytes/immunology
16.
Am J Transplant ; 12(1): 126-35, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21920020

ABSTRACT

Costimulation blockade of the CD40/CD154 pathway has been effective at preventing allograft rejection in numerous transplantation models. This strategy has largely depended on mAbs directed against CD154, limiting the potential for translation due to its association with thromboembolic events. Though targeting CD40 as an alternative to CD154 has been successful at preventing allograft rejection in preclinical models, there have been no reports on the effects of CD40-specific agents in human transplant recipients. This delay in clinical translation may in part be explained by the presence of cellular depletion with many CD40-specific mAbs. As such, the optimal biologic properties of CD40-directed immunotherapy remain to be determined. In this report, we have characterized 3A8, a human CD40-specific mAb and evaluated its efficacy in a rhesus macaque model of islet cell transplantation. Despite partially agonistic properties and the inability to block CD40 binding of soluble CD154 (sCD154) in vitro, 3A8-based therapy markedly prolonged islet allograft survival without depleting B cells. Our results indicate that the allograft-protective effects of CD40-directed costimulation blockade do not require sCD154 blockade, complete antagonism or cellular depletion, and serve to support and guide the continued development of CD40-specific agents for clinical translation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD40 Antigens/antagonists & inhibitors , CD40 Ligand/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation , Animals , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , Flow Cytometry , Immunotherapy , Lymphocyte Culture Test, Mixed , Macaca mulatta , Models, Animal
17.
Am J Transplant ; 12(1): 115-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21929643

ABSTRACT

In murine models, T-cell costimulation blockade of the CD28:B7 and CD154:CD40 pathways synergistically promotes immune tolerance after transplantation. While CD28 blockade has been successfully translated to the clinic, translation of blockade of the CD154:CD40 pathway has been less successful, in large part due to thromboembolic complications associated with anti-CD154 antibodies. Translation of CD40 blockade has also been slow, in part due to the fact that synergy between CD40 blockade and CD28 blockade had not yet been demonstrated in either primate models or humans. Here we show that a novel, nondepleting CD40 monoclonal antibody, 3A8, can combine with combined CTLA4Ig and sirolimus in a well-established primate bone marrow chimerism-induction model. Prolonged engraftment required the presence of all three agents during maintenance therapy, and resulted in graft acceptance for the duration of immunosuppressive treatment, with rejection resulting upon immunosuppression withdrawal. Flow cytometric analysis revealed that upregulation of CD95 expression on both CD4+ and CD8+ T cells correlated with rejection, suggesting that CD95 may be a robust biomarker of graft loss. These results are the first to demonstrate prolonged chimerism in primates treated with CD28/mTOR blockade and nondepletional CD40 blockade, and support further investigation of combined costimulation blockade targeting the CD28 and CD40 pathways.


Subject(s)
CD40 Antigens/antagonists & inhibitors , Chimerism , Immunoconjugates/immunology , Immunosuppressive Agents/pharmacology , Models, Animal , Sirolimus/pharmacology , Abatacept , Animals , Antibodies, Monoclonal/immunology , CD28 Antigens/immunology , CD40 Antigens/immunology , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Macaca mulatta
18.
Am J Transplant ; 11(12): 2593-602, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21883917

ABSTRACT

Significant deficiencies in understanding of xenospecific immunity have impeded the success of preclinical trials in xenoislet transplantation. Although galactose-α1,3-galactose, the gal epitope, has emerged as the principal target of rejection in pig-to-primate models of solid organ transplant, the importance of gal-specific immunity in islet xenotransplant models has yet to be clearly demonstrated. Here, we directly compare the immunogenicity, survival and function of neonatal porcine islets (NPIs) from gal-expressing wild-type (WT) or gal-deficient galactosyl transferase knockout (GTKO) donors. Paired diabetic rhesus macaques were transplanted with either WT (n = 5) or GTKO (n = 5) NPIs. Recipient blood glucose, transaminase and serum xenoantibody levels were used to monitor response to transplant. Four of five GTKO versus one of five WT recipients achieved insulin-independent normoglycemia; transplantation of WT islets resulted in significantly greater transaminitis. The WT NPIs were more susceptible to antibody and complement binding and destruction in vitro. Our results confirm that gal is an important variable in xenoislet transplantation. The GTKO NPI recipients have improved rates of normoglycemia, likely due to decreased susceptibility of xenografts to innate immunity mediated by complement and preformed xenoantibody. Therefore, the use of GTKO donors is an important step toward improved consistency and interpretability of results in future xenoislet studies.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Galactosyltransferases/deficiency , Graft Rejection/prevention & control , Islets of Langerhans Transplantation , Tissue Donors , Transplantation, Heterologous , Animals , Animals, Newborn , Antibodies, Heterophile/immunology , Blood Glucose/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Rejection/immunology , Immunity, Innate , Immunoenzyme Techniques , Macaca mulatta , Swine
19.
Am J Transplant ; 11(5): 947-57, 2011 May.
Article in English | MEDLINE | ID: mdl-21521467

ABSTRACT

The widespread clinical implementation of alloislet transplantation as therapy for type 1 diabetes has been hindered by the lack of suitable islet donors. Pig-to-human islet xenotransplantation is one strategy with potential to alleviate this shortage. Long-term survival of porcine islets has been achieved using CD154-specific antibodies to interrupt the CD40/CD154 costimulation pathway; however, CD154-specific antibodies seem unlikely candidates for clinical translation. An alternative strategy for CD40/CD154 pathway interruption is use of CD40-specific antibodies. Herein, we evaluate the ability of a chimeric CD40-specific monoclonal antibody (Chi220) to protect islet xenografts. Neonatal porcine islets (~50,000 IEQ/kg) were transplanted intraportally into pancreatectomized diabetic macaques. Immunosuppression consisted of induction therapy with Chi220 and the IL-2 receptor-specific antibody basiliximab, and maintenance therapy with sirolimus and the B7-specific fusion protein belatacept. Chi220 effectively promoted xenoislet engraftment and survival, with five of six treated recipients achieving insulin-independent normoglycemia (median rejection-free survival 59 days; mean 90.8 days, maximum 203 days). No thromboembolic phenomena were observed. CD40 represents a promising alternative to CD154 as a therapeutic target, and the efficacy of CD40-specific antibodies in islet xenotransplantation warrants further investigation.


Subject(s)
CD40 Antigens/metabolism , Islets of Langerhans Transplantation/methods , Transplantation, Heterologous/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Basiliximab , CD40 Antigens/immunology , CD40 Ligand/metabolism , Female , Graft Survival , Immunohistochemistry/methods , Immunosuppressive Agents/therapeutic use , Macaca mulatta , Male , Primates , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/therapeutic use , Sirolimus/therapeutic use , Swine
20.
Am J Transplant ; 10(11): 2396-409, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20849552

ABSTRACT

In murine models, mixed hematopoietic chimerism induction leads to robust immune tolerance. However, translation to primates and to patients has been difficult. In this study, we used a novel MHC-defined rhesus macaque model to examine the impact of MHC matching on the stability of costimulation blockade-/sirolimus-mediated chimerism, and to probe possible mechanisms of bone marrow rejection after nonmyeloablative transplant. Using busulfan-based pretransplant preparation and maintenance immunosuppression with sirolimus, as well as CD28 and CD154 blockade, all recipients demonstrated donor engraftment after transplant. However, the mixed chimerism that resulted was compartmentalized, with recipients demonstrating significantly higher whole blood chimerism compared to T cell chimerism. Thus, the vast majority of T cells presenting posttransplant were recipient-rather than donor-derived. Surprisingly, even in MHC-matched transplants, rejection of donor hematopoiesis predominated after immunosuppression withdrawal. Weaning of immunosuppression was associated with a surge of antigen-experienced T cells, and transplant rejection was associated with the acquisition of donor-directed T cell alloreactivity. These results suggest that a reservoir of alloreactive cells was present despite prior costimulation blockade and sirolimus, and that the post-immunosuppression lymphocytic rebound may have lead to a phenotypic shift in these recipient T cells towards an activated, antigen-experienced phenotype, and ultimately, to transplant rejection.


Subject(s)
Histocompatibility/immunology , Major Histocompatibility Complex/immunology , Transplantation Chimera/immunology , Animals , Busulfan/therapeutic use , Female , Graft Rejection/immunology , Hematopoietic Stem Cell Transplantation , Immune Tolerance , Immunosuppression Therapy , Macaca mulatta , Male , Sirolimus/therapeutic use , Substance Withdrawal Syndrome/immunology , T-Lymphocytes/immunology , Transplantation Conditioning/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...