Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Transplant ; 18(4): 998-1006, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29178588

RESUMO

Porcine islet xenografts have the potential to provide an inexhaustible source of islets for ß cell replacement. Proof-of-concept has been established in nonhuman primates. However, significant barriers to xenoislet transplantation remain, including the poorly understood instant blood-mediated inflammatory reaction and a thorough understanding of early xeno-specific immune responses. A paucity of data exist comparing xeno-specific immune responses with alloislet (AI) responses in primates. We recently developed a dual islet transplant model, which enables direct histologic comparison of early engraftment immunobiology. In this study, we investigate early immune responses to neonatal porcine islet (NPI) xenografts compared with rhesus islet allografts at 1 hour, 24 hours, and 7 days. Within the first 24 hours after intraportal infusion, we identified greater apoptosis (caspase 3 activity and TUNEL [terminal deoxynucleotidyl transferase dUTP nick end labeling])-positive cells) of NPIs compared with AIs. Macrophage infiltration was significantly greater at 24 hours compared with 1 hour in both NPI (wild-type) and AIs. At 7 days, IgM and macrophages were highly specific for NPIs (α1,3-galactosyltransferase knockout) compared with AIs. These findings demonstrate an augmented macrophage and antibody response toward xenografts compared with allografts. These data may inform future immune or genetic manipulations required to improve xenoislet engraftment.


Assuntos
Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Inflamação/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/imunologia , Macrófagos/imunologia , Animais , Animais Recém-Nascidos , Apoptose , Ilhotas Pancreáticas/patologia , Macaca mulatta , Suínos , Transplante Heterólogo
2.
Am J Transplant ; 17(5): 1193-1203, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27888551

RESUMO

Costimulation blockade (CoB) via belatacept is a lower-morbidity alternative to calcineurin inhibitor (CNI)-based immunosuppression. However, it has higher rates of early acute rejection. These early rejections are mediated in part by memory T cells, which have reduced dependence on the pathway targeted by belatacept and increased adhesion molecule expression. One such molecule is leukocyte function antigen (LFA)-1. LFA-1 exists in two forms: a commonly expressed, low-affinity form and a transient, high-affinity form, expressed only during activation. We have shown that antibodies reactive with LFA-1 regardless of its configuration are effective in eliminating memory T cells but at the cost of impaired protective immunity. Here we test two novel agents, leukotoxin A and AL-579, each of which targets the high-affinity form of LFA-1, to determine whether this more precise targeting prevents belatacept-resistant rejection. Despite evidence of ex vivo and in vivo ligand-specific activity, neither agent when combined with belatacept proved superior to belatacept monotherapy. Leukotoxin A approached a ceiling of toxicity before efficacy, while AL-579 failed to significantly alter the peripheral immune response. These data, and prior studies, suggest that LFA-1 blockade may not be a suitable adjuvant agent for CoB-resistant rejection.


Assuntos
Abatacepte/farmacologia , Rejeição de Enxerto/tratamento farmacológico , Sobrevivência de Enxerto/imunologia , Memória Imunológica/imunologia , Transplante de Rim/efeitos adversos , Antígeno-1 Associado à Função Linfocitária/química , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Taxa de Filtração Glomerular , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Imunossupressores/farmacologia , Testes de Função Renal , Antígeno-1 Associado à Função Linfocitária/metabolismo , Macaca mulatta , Complicações Pós-Operatórias , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
3.
Am J Transplant ; 16(5): 1456-64, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26602755

RESUMO

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.


Assuntos
Abatacepte/uso terapêutico , Modelos Animais de Doenças , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/imunologia , Falência Renal Crônica/imunologia , Transplante de Rim/efeitos adversos , Antígeno-1 Associado à Função Linfocitária/imunologia , Animais , Taxa de Filtração Glomerular , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/patologia , Memória Imunológica , Falência Renal Crônica/cirurgia , Testes de Função Renal , Antígeno-1 Associado à Função Linfocitária/administração & dosagem , Macaca mulatta , Transplante Homólogo
4.
Am J Transplant ; 16(6): 1726-38, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26705099

RESUMO

We have established a model of sensitization in nonhuman primates and tested two immunosuppressive regimens. Animals underwent fully mismatched skin transplantation, and donor-specific antibody (DSA) response was monitored by flow cross-match. Sensitized animals subsequently underwent kidney transplantation from their skin donor. Immunosuppression included tacrolimus, mycophenolate, and methylprednisolone. Three animals received basiliximab induction; compared with nonsensitized animals, they showed a shorter mean survival time (4.7 ± 3.1 vs. 187 ± 88 days). Six animals were treated with T cell depletion (anti-CD4/CD8 mAbs), which prolonged survival (mean survival time 21.6 ± 19.0 days). All presensitized animals showed antibody-mediated rejection (AMR). In two of three basiliximab-injected animals, cellular rejection (ACR) was prominent. After T cell depletion, three of six monkeys experienced early acute rejection within 8 days with histological evidence of thrombotic microangiopathy and AMR. The remaining three monkeys survived 27-44 days, with mixed AMR and ACR. Most T cell-depleted animals experienced a rebound of DSA that correlated with deteriorating kidney function. We also found an increase in proliferating memory B cells (CD20(+) CD27(+) IgD(-) Ki67(+) ), lymph node follicular helper T cells (ICOS(+) PD-1(hi) CXCR5(+) CD4(+) ), and germinal center (GC) response. Depletion controlled cell-mediated rejection in sensitized nonhuman primates better than basiliximab, yet grafts were rejected with concomitant DSA rise. This model provides an opportunity to test novel desensitization strategies.


Assuntos
Modelos Animais de Doenças , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/etiologia , Imunossupressores/uso terapêutico , Isoanticorpos/imunologia , Transplante de Rim/efeitos adversos , Transplante de Pele , Animais , Humanos , Depleção Linfocítica , Macaca mulatta , Masculino , Linfócitos T Auxiliares-Indutores/imunologia
5.
Am J Transplant ; 15(8): 2240-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26139552

RESUMO

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.


Assuntos
Aloenxertos , Neovascularização Patológica , Animais , Primatas
6.
Am J Transplant ; 15(5): 1241-52, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25702898

RESUMO

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.


Assuntos
Inflamação/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Animais , Animais Geneticamente Modificados , Glicemia/química , Plaquetas/imunologia , Ativação do Complemento , Modelos Animais de Doenças , Galactosiltransferases/genética , Imuno-Histoquímica , Macaca mulatta , Macrófagos/imunologia , Neutrófilos/imunologia , Fenótipo , Suínos , Fatores de Tempo , Transplante Heterólogo
7.
Am J Transplant ; 15(4): 984-92, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25703015

RESUMO

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.


Assuntos
Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Transplante de Rim , Abatacepte/uso terapêutico , Animais , Rejeição de Enxerto/prevenção & controle , Memória Imunológica , Imunofenotipagem , Macaca mulatta , Projetos Piloto , Linfócitos T/imunologia
8.
Am J Transplant ; 13(12): 3085-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24119188

RESUMO

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.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Imunossupressores/efeitos adversos , Integrinas/antagonistas & inibidores , Transplante de Rim/métodos , Aloenxertos , Animais , Anticorpos Monoclonais/química , Antígenos de Neoplasias , Biópsia , Rejeição de Enxerto , Terapia de Imunossupressão , Macaca mulatta , Projetos Piloto , Distribuição Aleatória , Fator de Crescimento Transformador beta1/sangue
9.
Am J Transplant ; 13(2): 320-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23311611

RESUMO

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.


Assuntos
Imunoconjugados/administração & dosagem , Imunossupressores/uso terapêutico , Transplante de Rim/métodos , Sirolimo/administração & dosagem , Linfócitos T/imunologia , Abatacepte , Animais , Antígenos CD2/metabolismo , Complexo CD3/metabolismo , Progressão da Doença , Intervalo Livre de Doença , Rejeição de Enxerto , Sobrevivência de Enxerto , Memória Imunológica , Macaca mulatta , Fenótipo , Linfócitos T Reguladores/imunologia , Transplante Homólogo , Resultado do Tratamento
10.
Am J Transplant ; 13(2): 312-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23279640

RESUMO

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.


Assuntos
Imunoconjugados/administração & dosagem , Transplante das Ilhotas Pancreáticas/métodos , Proteínas Recombinantes de Fusão/administração & dosagem , Sirolimo/administração & dosagem , Transplante Homólogo/métodos , Abatacepte , Alefacept , Animais , Anticorpos Monoclonais/administração & dosagem , Basiliximab , Peptídeo C/metabolismo , Diabetes Mellitus Experimental , Sobrevivência de Enxerto , Antígenos de Histocompatibilidade/imunologia , Imunossupressores/administração & dosagem , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Macaca mulatta , Esteroides/administração & dosagem
11.
Am J Transplant ; 12(8): 2079-87, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22845909

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD40/imunologia , Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas , Animais , Formação de Anticorpos , Humanos , Macaca mulatta
12.
Am J Transplant ; 12(9): 2395-405, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22776408

RESUMO

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.


Assuntos
Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Isoanticorpos/imunologia , Animais , Imuno-Histoquímica , Memória Imunológica , Imunossupressores/uso terapêutico , Depleção Linfocítica , Macaca mulatta , Masculino
13.
Am J Transplant ; 12(7): 1755-64, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22642491

RESUMO

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.


Assuntos
Transplante de Medula Óssea , Quimerismo , Rejeição de Enxerto , Transplante de Rim , Animais , Macaca mulatta , Transplante Homólogo
14.
Am J Transplant ; 12(7): 1918-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22458552

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD40/imunologia , Imunoconjugados/imunologia , Transplante das Ilhotas Pancreáticas , Isoanticorpos/biossíntese , Abatacepte , Animais , Sobrevivência de Enxerto/imunologia , Macaca mulatta
15.
Am J Transplant ; 12(7): 1765-75, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22458586

RESUMO

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.


Assuntos
Antígenos CD40/imunologia , Ligante de CD40/imunologia , Sobrevivência de Enxerto/imunologia , Xenoenxertos , Imunossupressores/administração & dosagem , Transplante das Ilhotas Pancreáticas , Animais , Estudos de Coortes , Diabetes Mellitus Experimental/cirurgia , Memória Imunológica , Macaca mulatta , Suínos , Linfócitos T/imunologia
16.
Eur J Radiol ; 81(7): 1479-82, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21570792

RESUMO

PURPOSE: The aim of this work is to characterize the shape and the volume of the internal anal sphincter (IAS) in normal subjects by three-dimensional anorectal ultrasonography. METHODS: Thirty-nine normal volunteer males (mean age 58.5 ± 18.7) and 25 females (mean age 59.4 ± 14.1) were submitted to anorectal ultrasonography. The tissue is defined by a semiautomatic procedure. Measurements of thickness, length and volume were assessed automatically. The software provides an average number of 57,600 thickness measurements, 360 length measurements for each zone (90 for each quadrant) and seven volume measurements (one for each anatomical area).The mean values of magnitudes were calculated for the entire volume in each quadrant and zone. Age and gender-related variations were analyzed. RESULTS: In assessments of the whole tissue, only thickness was gender-related, with greater thickness for females (male thickness: 1.81 ± 0.47 mm, female thickness 2.16 ± 0.57 mm, P-value<0.01).In the distal zone: thickness, length and volume were all larger in females (for male and female respectively: 1.83 ± 0.49 mm vs 2.34 ± 0.58 mm, P-value<0.01, for the thickness; 10.87 ± 2.10mm vs 12.18 ± 2.21 mm, P-value<0.02 for the length and 1501 ± 605 mm(3) vs 2169 ± 871 mm(3), P-value<0.01 for the volume). In the medial zone, only thickness was gender-related, with greater thickness in females (male thickness: 2.04 ± 0.60mm, female thickness:2.44 ± 0.74 mm, P-value<0.02).The only variation observed in the proximal zone concerned length, larger in males (respectively: 11.27 ± 2.84 mm vs 9.55 ± 2.43 mm, P-value<0.02). The male population was significantly positively correlated with ageing for volume in the whole tissue (ρ = 0.32, P-value<0.05), and for both thickness and volume in the medial zone (ρ = 0.33, P-value<0.05 for thickness; ρ = 0.39, P-value<0.02 for the volume). CONCLUSION: This new method is useful to understand both functional anal disorders and local damage which may affect only part of the muscle tissue.


Assuntos
Canal Anal/diagnóstico por imagem , Imageamento Tridimensional , Canal Anal/anatomia & histologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Software , Ultrassonografia
17.
Am J Transplant ; 12(1): 126-35, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21920020

RESUMO

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.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD40/antagonistas & inibidores , Ligante de CD40/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD40/imunologia , Citometria de Fluxo , Imunoterapia , Teste de Cultura Mista de Linfócitos , Macaca mulatta , Modelos Animais
18.
Am J Transplant ; 12(1): 115-25, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21929643

RESUMO

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.


Assuntos
Antígenos CD40/antagonistas & inibidores , Quimerismo , Imunoconjugados/imunologia , Imunossupressores/farmacologia , Modelos Animais , Sirolimo/farmacologia , Abatacepte , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD28/imunologia , Antígenos CD40/imunologia , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Macaca mulatta
19.
Am J Transplant ; 11(12): 2593-602, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21883917

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Galactosiltransferases/deficiência , Rejeição de Enxerto/prevenção & controle , Transplante das Ilhotas Pancreáticas , Doadores de Tecidos , Transplante Heterólogo , Animais , Animais Recém-Nascidos , Anticorpos Heterófilos/imunologia , Glicemia/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Imunidade Inata , Técnicas Imunoenzimáticas , Macaca mulatta , Suínos
20.
Am J Transplant ; 9(1): 124-31, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18976300

RESUMO

Pretransplant exposure to donor antigen is known to modulate recipient alloimmunity, and frequently results in sensitization. However, donor-specific transfusion (DST) can have a protolerant effect that is dependent on route, dose and coadministered immunosuppression. Rodent studies have shown in some strain combinations that portal venous (PV) DST alone can induce tolerance, and uncontrolled clinical use of PVDST has been reported. In order to determine if pretransplant PVDST has a clinically relevant salutary effect, we studied it and the influence of concomitant immunosuppression in rhesus monkeys undergoing renal allotransplantation. Animals received PVDST with unfractionated bone marrow and/or tacrolimus or sirolimus 1 week prior to transplantation. Graft survival was assessed without any posttransplant immunosuppression. PVDST alone or in combination with tacrolimus was ineffective. However, PVDST in combination with sirolimus significantly prolonged renal allograft survival to a mean of 24 days. Preoperative sirolimus alone had no effect, and peripheral DST with sirolimus prolonged graft survival in 2/4 animals, but resulted in accelerated rejection in 2/4 animals. These data demonstrate that PVDST in combination with sirolimus delays rejection in a modest but measurable way in a rigorous model. It may thus be a preferable method for donor antigen administration.


Assuntos
Sobrevivência de Enxerto , Imunossupressores/uso terapêutico , Isoantígenos/administração & dosagem , Transplante de Rim , Sirolimo/uso terapêutico , Animais , Quimera , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica , Macaca mulatta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA