Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Transl Immunology ; 9(7): e1150, 2020.
Article in English | MEDLINE | ID: mdl-32642064

ABSTRACT

OBJECTIVES: Loss of vaccine-induced antibodies (Abs) after chemotherapy against paediatric acute lymphoblastic leukaemia (ALL) is common and often necessitates re-immunisation after cessation of treatment. Even so, some ALL survivors fail to mount or to maintain protective Abs. Germinal centres (GCs) are clusters of proliferating B cells in follicles of secondary lymphoid tissues (SLTs) formed during adaptive immune responses and the origins of long-lived memory B and plasma cells that are the source of Abs. Furthermore, productive GC reactions depend on T follicular helper (TFH) cells. To understand why chemotherapy induces deficits in Ab responses, we examined how SLTs were affected by chemotherapy. METHODS: Rhesus macaques were infused with either three cycles of the anthracycline doxorubicin or saline, followed by immunisation with a de novo and booster antigen. Spleen and lymph nodes were removed, and memory B, bulk T and TFH cells were examined. RESULTS: Despite adequate GC morphology, a diminished memory and IgG+ B-cell population along with diminished total and booster vaccine-specific IgG-producing memory B cells were noted in the spleens of macaques with past doxorubicin exposure compared to the saline-treated controls (P < 0.05). Intact bulk T and TFH cells were found in the SLTs of treated macaques, which displayed higher CD40L upregulation capacity by their splenic CXCR5+ helper T cells (P < 0.01). In contrast to the spleen, the immune cell populations studied were comparable between the lymph nodes of both saline- and doxorubicin-treated macaques. CONCLUSION: Our findings suggest that the splenic memory B-cell subset, compared to its lymph node counterpart, is more severely altered by anthracycline treatment.

2.
Sci Rep ; 7(1): 992, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28428630

ABSTRACT

Autoantibodies against ion channels are the cause of numerous neurologic autoimmune disorders. Frequently, such pathogenic autoantibodies have a restricted epitope-specificity. In such cases, competing antibody formats devoid of pathogenic effector functions (blocker antibodies) have the potential to treat disease by displacing autoantibodies from their target. Here, we have used a model of the neuromuscular autoimmune disease myasthenia gravis in rhesus monkeys (Macaca mulatta) to test the therapeutic potential of a new blocker antibody: MG was induced by passive transfer of pathogenic acetylcholine receptor-specific monoclonal antibody IgG1-637. The effect of the blocker antibody (IgG4Δhinge-637, the hinge-deleted IgG4 version of IgG1-637) was assessed using decrement measurements and single-fiber electromyography. Three daily doses of 1.7 mg/kg IgG1-637 (cumulative dose 5 mg/kg) induced impairment of neuromuscular transmission, as demonstrated by significantly increased jitter, synaptic transmission failures (blockings) and a decrease in the amplitude of the compound muscle action potentials during repeated stimulations (decrement), without showing overt symptoms of muscle weakness. Treatment with three daily doses of 10 mg/kg IgG4Δhinge-637 significantly reduced the IgG1-637-induced increase in jitter, blockings and decrement. Together, these results represent proof-of principle data for therapy of acetylcholine receptor-myasthenia gravis with a monovalent antibody format that blocks binding of pathogenic autoantibodies.


Subject(s)
Autoantibodies/metabolism , Immunoglobulin G/administration & dosage , Myasthenia Gravis/drug therapy , Receptors, Cholinergic/metabolism , Animals , CHO Cells , Cholinergic Antagonists , Cricetulus , Disease Models, Animal , Gene Expression Regulation/drug effects , HEK293 Cells , Hinge Exons , Humans , Immunoglobulin G/pharmacology , Macaca mulatta , Myasthenia Gravis/immunology , Myasthenia Gravis/metabolism , Synaptic Transmission/drug effects , Treatment Outcome
3.
Front Immunol ; 7: 462, 2016.
Article in English | MEDLINE | ID: mdl-27872622

ABSTRACT

Aging western societies are facing an increasing prevalence of chronic inflammatory and degenerative diseases for which often no effective treatments exist, resulting in increasing health-care expenditure. Despite high investments in drug development, the number of promising new drug candidates decreases. We propose that preclinical research in non-human primates can help to bridge the gap between drug discovery and drug prescription. Translational research covers various stages of drug development of which preclinical efficacy tests in valid animal models is usually the last stage. Preclinical research in non-human primates may be essential in the evaluation of new drugs or therapies when a relevant rodent model is not available. Non-human primate models for life-threatening or severely debilitating diseases in humans are available at the Biomedical Primate Research Centre (BPRC). These have been instrumental in translational research for several decades. In order to stimulate European health research and innovation from bench to bedside, the European Commission has invested heavily in access to non-human primate research for more than 20 years. BPRC has hosted European users in a series of transnational access programs covering a wide range of research areas with the common theme being immune-mediated inflammatory disorders. We present an overview of the results and give an account of the studies performed as part of European Union Framework Programme (EU FP)-funded translational non-human primate research performed at the BPRC. These data illustrate the value of translational non-human primate research for the development of new therapies and emphasize the importance of EU FP funding in drug development.

4.
Inflammation ; 38(6): 2191-202, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26140903

ABSTRACT

Kidney allograft biopsies were analyzed for the presence of B cell clusters/aggregates using CD20 staining. Few B cells were found in the diffuse interstitial infiltrates, but clusters of B cells were found in nodular infiltrates. These nodular infiltrates were smaller shortly after transplantation, and their size increased over time. At the time of clinical rejection, the nodules often presented as tertiary lymphoid structures (TLS) with lymphoid-like follicles. The presence of small B cell clusters during the first 2 months after transplantation was not associated with early rejection. Even in animals that did not reject their allograft, TLS-like structures were present and could disappear over time. Although TLS were more often found in samples with interstitial fibrosis and tubular atrophy (IFTA), TLS were also present in samples without IFTA. The presence and density of clusters resembling tertiary lymphoid structures most likely reflect an ongoing immune response inside the graft and do not necessarily signify a poor graft outcome or IFTA.


Subject(s)
B-Lymphocytes/immunology , Graft Rejection/immunology , Kidney Transplantation/adverse effects , Kidney/immunology , Kidney/surgery , Lymphoid Tissue/immunology , Allografts , Animals , Antigens, CD20/metabolism , Atrophy , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers/metabolism , Biopsy , Fibrosis , Graft Rejection/metabolism , Graft Rejection/pathology , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Macaca mulatta , Models, Animal , Time Factors
6.
PLoS One ; 8(8): e71549, 2013.
Article in English | MEDLINE | ID: mdl-23977076

ABSTRACT

The overlapping epidemiology of multiple sclerosis (MS) and Epstein-Barr virus (EBV), the increased risk to develop MS after infectious mononucleosis (IM) and the localization of EBV-infected B-cells within the MS brain suggest a causal link between EBV and MS. However, the underlying mechanism is unknown. We hypothesize that EBV-infected B-cells are capable of eliciting a central nervous system (CNS) targeting autoimmune reaction. To test this hypothesis we have developed a novel experimental model in rhesus monkeys of IM-like disease induced by infusing autologous B-lymphoblastoid cells (B-LCL). Herpesvirus papio (HVP) is a lymphocryptovirus related to EBV and was used to generate rhesus monkey B-LCL. Three groups of five animals were included; each group received three intravenous infusions of B-LCL that were either pulsed with the encephalitogenic self peptide MOG(34-56) (group A), a mimicry peptide (981-1003) of the major capsid protein of cytomegalovirus (CMVmcp(981-1003); group B) or the citrullinated MOG(34-56) (cMOG(34-56); group C). Groups A and B received on day 98 a single immunization with MOG(34-56) in incomplete Freund's adjuvant (IFA). Group C monkeys were euthanized just prior to day 98 without booster immunization. We observed self-peptide-specific proliferation of T-cells, superimposed on similar strong proliferation of CD3(+)CD8(+) T-cells against the B-LCL as observed in IM. The brains of several monkeys contained perivascular inflammatory lesions of variable size, comprising CD3(+) and CD68(+) cells. Moreover, clusters of CD3(+) and CD20(+) cells were detected in the meninges. The only evident clinical sign was substantial loss of bodyweight (>15%), a symptom observed both in early autoimmune encephalitis and IM. In conclusion, this model suggests that EBV-induced B-LCL can elicit a CNS targeting inflammatory (auto)immune reaction.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/virology , Encephalitis/immunology , Lymphocryptovirus/immunology , Macaca mulatta/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptides/immunology , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Cell Proliferation , Central Nervous System/pathology , Citrulline/immunology , Cohort Studies , Encephalitis/virology , Humans , Immunity, Cellular/immunology , Immunity, Innate/immunology , Immunoglobulin G/immunology , Macaca mulatta/virology , T-Lymphocytes/immunology
7.
Inflamm Res ; 62(2): 181-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23064655

ABSTRACT

OBJECTIVES: Non-human primates are immunologically closely related to humans providing relevant models of inflammatory disorders often used to evaluate new immunomodulating therapies. The aim of the study was to compare inflammatory infiltrates of acute graft rejection (AR) and collagen-induced arthritis (CIA) to delayed-type hypersensitivity (DTH) reactions as the latter model may serve as a less invasive animal model. MATERIALS AND METHODS: Tissue samples of AR, CIA and DTH were obtained from rhesus monkeys used in several pre-clinical studies. The infiltrate composition was determined by immunohistochemical analysis. RESULTS: The infiltrates in AR consisted of T cells, macrophages and B cells. The presence of lymphoid structures in AR suggested ongoing intragraft immune activation. The synovia of CIA contained predominantly macrophages and few T cells. The DTH infiltrates were dominated by T cells when the challenged was ovalbumin (OVA) and by macrophages when the challenge was tetanus toxoid (TT). CONCLUSIONS: The histology of AR resembles aspects of DTH to OVA while that of CIA showed similarities of the DTH to TT. The DTH reaction could serve as a model to study immunomodulating drugs for acute rejection and the acute inflammatory phase of autoimmunity.


Subject(s)
Arthritis, Experimental/immunology , Graft Rejection/immunology , Hypersensitivity, Delayed/immunology , Animals , Antigens/immunology , Arthritis, Experimental/pathology , B-Lymphocytes/immunology , Cytokines/immunology , Dendritic Cells/immunology , Graft Rejection/pathology , Hypersensitivity, Delayed/pathology , Kidney Transplantation/adverse effects , Macaca mulatta , Macrophages/immunology , Ovalbumin/immunology , T-Lymphocytes/immunology , Tetanus Toxoid/immunology
8.
Transplantation ; 85(8): 1185-92, 2008 Apr 27.
Article in English | MEDLINE | ID: mdl-18431240

ABSTRACT

BACKGROUND: Translational research in a relevant preclinical model is recommended before Treg-inducing protocols can be implemented in humans. We have characterized rhesus monkey CD25 cells phenotypically and functionally. METHODS: The phenotype of CD4(+)CD25(high) cells was determined by FACS, focusing on established markers of mouse and human Treg cells. Percentages of cells positive for CD45RA, CD62L, and intracellular CTLA-4 and FOXP3 were compared between CD4(+)CD25(high) and CD4(+)CD25(-) cells. CD25 cells stimulated with anti-CD3, ConA, and/or allogeneic peripheral blood mononuclear cells were mixed with freshly isolated CD25 cells. The suppressive activity of the CD25 cells in vitro was assessed using several experimental conditions. RESULTS: Rhesus monkey CD4(+)CD25(high) cells expressed high intracellular levels of CTLA-4 and FOXP3, whereas expression was negligible in CD4(+)CD25(-) cells. The CD25(high) population was mostly CD45RA(-), indicative of a memory phenotype. The CD25(+) cells were anergic, because they showed low proliferative responses, no interleukin-2 production, and some interferon-gamma and interleukin-10 production. Proliferation of CD4(+)CD25(-) cells stimulated by anti-CD3 or allogeneic cells was decreased when CD4(+)CD25(-) cells were added at a 1:1 ratio. In addition, we found that CD25 cells inhibited the interleukin-2 and interferon-gamma production by anti-CD3-stimulated CD25 cells in a dose-dependent fashion, through a cell-cell contact-dependent mechanism. CONCLUSIONS: Rhesus monkey CD4(+)CD25(+) cells have similar phenotypic and functional characteristics as natural Tregs in humans. These findings allow testing of Treg expansion and induction protocols in a relevant preclinical model, the rhesus monkey.


Subject(s)
Macaca mulatta/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Antigens, CD/analysis , Antigens, Differentiation/analysis , CTLA-4 Antigen , Cell Communication , Cytokines/biosynthesis , Immunologic Memory , Leukocyte Common Antigens/analysis , T-Lymphocytes, Regulatory/immunology
9.
Drug Discov Today ; 12(7-8): 327-35, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17395093

ABSTRACT

The costs for the development of new drugs have increased dramatically over the past 30 years. One of the main reasons for this increase is the low success rate of new drugs being approved for patient use, which is, in part, a consequence of the common use of rodent models for preclinical validation of efficacy. Especially in the development of biologicals, which are now successfully used in the treatment of rheumatoid arthritis, the selection of the right animal model is pivotal. Non-human primates could help to bridge the evolutionary gap between rodent models and human patients.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Drug Evaluation, Preclinical/methods , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/physiopathology , Biomarkers/analysis , Drug Design , Humans , Macaca mulatta , Mice
10.
Transplantation ; 82(9): 1194-201, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17102771

ABSTRACT

BACKGROUND: Costimulation blockade with antibodies directed against human CD40 and CD86 leads to prolonged kidney allograft survival in rhesus monkeys, but fails to induce permanent graft acceptance. We have tested whether costimulation blockade is more effective after peripheral T-cell ablation with antithymocyte globulin (ATG), with the aim to remove already primed autoreactive cells present in the normal repertoire. METHODS: Rhesus monkeys were transplanted with a mismatched kidney allograft. ATG was given around the time of transplantation (day -1 and 0). Costimulation blockade with anti-CD40+anti-CD86 was given at tapering dosages from day -1 to 56. Cyclosporin A (CsA) was given from day 42 onwards and first rejections occurring after day 42 were treated with prednisone. RESULTS: We observed accelerated rejection in ATG-treated monkeys, compared to animals receiving only costimulation blockade. The accelerated rejection of the kidney allograft occurred despite the application of rejection therapy with steroids and CsA. Three of the five ATG-treated animals were found seropositive for donor-specific alloantibodies. Early biopsies (day 21) from animals treated with ATG and anti-CD40+anti-CD86 show substantially reduced expression of cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and forkhead box P3 (FOXP3) in focal infiltrates as compared to animals treated with only costimulation blockade. Furthermore, we observed the rapid reappearance of CD8 T-cells with a memory phenotype (disappearance of naive CD95/CD11a T-cells) in peripheral blood. CONCLUSION: We conclude that (subtotal) T-cell depletion using ATG does not add to costimulation blockade induced kidney allograft survival.


Subject(s)
Antibodies, Blocking/administration & dosage , Antilymphocyte Serum/administration & dosage , Graft Survival/drug effects , Immunosuppression Therapy , Kidney Transplantation , Animals , Antigens, CD/analysis , Antigens, Differentiation/analysis , B7-2 Antigen/drug effects , CD40 Antigens/antagonists & inhibitors , CTLA-4 Antigen , Drug Synergism , Forkhead Transcription Factors/analysis , Graft Rejection/immunology , Graft Rejection/pathology , Graft Rejection/prevention & control , Immunologic Memory , Kidney/chemistry , Kidney/pathology , Lymphocyte Depletion , Macaca mulatta , T-Lymphocytes/chemistry , T-Lymphocytes/drug effects
11.
Transpl Int ; 19(5): 396-403, 2006 May.
Article in English | MEDLINE | ID: mdl-16623875

ABSTRACT

The active form of vitamin D3, 1,25(OH)2D3, has pronounced immunoregulatory properties and is a potential treatment of immune-based disorders. However, the central role of this hormone in calcium and bone metabolism complicates its long-term use as an immunomodulator. Some newly developed vitamin D3-derived analogues, such as MC1288, have an improved immunoregulatory potential and prolong allograft survival in rodent models. Such compounds might be a valuable component of immunosuppressive treatment regimen in transplantation and autoimmunity. The rhesus monkey provides a useful model for the preclinical validation of new therapeutic strategies for transplantation. The present study shows that MC1288 inhibits both proliferation and interferon-gamma production by rhesus peripheral blood mononuclear cells in a mixed lymphocyte reaction. We have tested the maximum tolerated dose of MC1288 in a rhesus monkey model of kidney transplantation. The observed effects on serum calcium and parathyroid hormone confirm the in vivo activity of MC1288. However, as a monotherapy, MC1288 did not cause prolongation of the kidney allograft survival in rhesus monkeys.


Subject(s)
Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Kidney Transplantation/methods , Vitamin D/analogs & derivatives , Animals , Calcium/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Graft Survival , Immunosuppressive Agents/pharmacology , Lymphocytes/metabolism , Macaca mulatta , Male , Maximum Tolerated Dose , Time Factors
12.
Transplantation ; 80(3): 385-93, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16082335

ABSTRACT

BACKGROUND: Chronic allograft nephropathy (CAN) is a common late complication of kidney transplantation. Antibodies to both human leukocyte antigen and nonhuman leukocyte antigen antigens have been implicated in the development of this condition. Here we investigated the presence of antivimentin antibodies in nonhuman primate recipients of kidney allografts as a possible predictor of CAN and the effects of immunosuppression. METHODS: Thirty seven rhesus monkeys received a kidney allograft to study the potency of several different immunosuppressive regimens (conventional immunosuppression, n=19, vs. costimulatory blockade, n=18). Monkeys were tested for antivimentin antibody by enzyme-linked immunosorbent assay and for anti-donor antibody by staining donor spleen cells with recipient serum. The appearance of antibodies was correlated with the graft pathology in biopsy and necropsy material. RESULTS: Antivimentin antibodies were found in 31 of 37 animals, whereas only 15 of 32 animals made anti-donor antibodies. Conventional immunosuppression did not prevent antivimentin antibody formation. Costimulation blockade, in particular blocking CD40 and CD86, significantly delayed or prevented antivimentin antibody formation, but did not prevent CAN. Antivimentin antibodies were not significantly associated with development of CAN. CONCLUSIONS: We postulate that vimentin acts as an autoantigen after renal transplantation; it elicits an autoimmune response that is not regulated by cyclosporine. This autoimmune response may be part of the complex immunologic events occurring posttransplantation and may contribute to the development of CAN, but cannot be considered as a major cause of CAN because this condition also develops without antivimentin antibodies.


Subject(s)
Autoimmunity , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Kidney Transplantation/methods , Vimentin/pharmacology , Animals , Antigens/chemistry , Antigens, CD/biosynthesis , B7-2 Antigen , Biopsy , CD40 Antigens/biosynthesis , Enzyme-Linked Immunosorbent Assay , HLA Antigens/immunology , Humans , Immune Tolerance , Immunoglobulin G/chemistry , Immunoglobulin M/chemistry , Kidney Transplantation/adverse effects , Macaca mulatta , Membrane Glycoproteins/biosynthesis , Time Factors , Transplantation, Homologous , Treatment Outcome , Vimentin/antagonists & inhibitors , Vimentin/chemistry
13.
Arthritis Res Ther ; 7(4): 145-54, 2005.
Article in English | MEDLINE | ID: mdl-15987497

ABSTRACT

Models of rheumatoid arthritis (RA) in laboratory animals are important tools for research into pathogenic mechanisms and the development of effective, safe therapies. Rodent models (rats and mice) have provided important information about the pathogenic mechanisms. However, the evolutionary distance between rodents and humans hampers the translation of scientific principles into effective therapies. The impact of the genetic distance between the species is especially seen with treatments based on biological molecules, which are usually species-specific. The outbred nature and the closer anatomical, genetic, microbiological, physiological, and immunological similarity of nonhuman primates to humans may help to bridge the wide gap between inbred rodent strain models and the heterogeneous RA patient population. Here we review clinical, immunological and pathological aspects of the rhesus monkey model of collagen-induced arthritis, which has emerged as a reproducible model of human RA in nonhuman primates.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Disease Models, Animal , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Humans , Macaca mulatta , Primates
14.
Transplantation ; 79(11): 1623-6, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15940054

ABSTRACT

Costimulation blockade as a single immunosuppressive treatment modality is not sufficient to prevent graft rejection. Here, we report an induction therapy using antagonistic antibodies against CD40 and CD86, given twice weekly from day -1 until day 56, followed by a delayed 12-week course of low-dose cyclosporine A (CsA) treatment in the rhesus monkey kidney-allograft model. Low-dose CsA treatment was initiated on day 42 and tapered until total cessation of all treatment on day 126. Treatment with anti-CD40/86 alone resulted in graft survival of 61, 71, 75, 78, and 116 days. Costimulation blockade followed by CsA resulted in more than 3-year drug-free survival in two of four animals. None of the animals developed donor-specific alloantibodies. Transforming growth factor-beta producing cells are present in early as well as in late kidney-graft biopsies and could play a role in the observed long-term drug-free graft survival.


Subject(s)
Graft Survival/immunology , Kidney Transplantation/immunology , Animals , Antibodies/therapeutic use , Antigens, CD/immunology , B7-2 Antigen , CD40 Antigens/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/drug effects , Kidney Transplantation/pathology , Macaca mulatta , Membrane Glycoproteins/immunology , Time Factors , Transplantation, Homologous
15.
Arthritis Rheum ; 52(2): 627-36, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15693002

ABSTRACT

OBJECTIVE: Collagen-induced arthritis (CIA) in the rhesus monkey is a nonhuman primate model of rheumatoid arthritis (RA). The close phylogenetic relationship between humans and the rhesus monkey makes this model useful for the preclinical safety and efficacy testing of new therapies that are inactive in animals more distinctly related to humans. In this study, we tested the therapeutic potential of a novel, small molecular weight antagonist of CCR5, SCH-X, in this model. METHODS: CIA was induced in 10 rhesus monkeys. The animals were allocated to receive SCH-X or saline as the control (n = 5 in each group). Treatment was initiated on the day of CIA induction and continued for 45 days. Monkeys were monitored before and 63 days after CIA induction for macroscopic signs of clinical arthritis, such as soft-tissue swelling and body weight. Furthermore, markers of inflammation and joint degradation were monitored to follow the disease course. RESULTS: Only 2 of 5 animals in the SCH-X-treated group displayed prominent soft-tissue swelling, compared with all 5 saline-treated monkeys. In addition to the suppression of joint inflammation, treatment with SCH-X resulted in a reduction in joint destruction, as demonstrated by lower rates of urinary excretion of collagen crosslinks, with confirmation by histology. Whereas in all saline-treated monkeys, marked erosion of joint cartilage was observed, this was absent in 4 of the 5 SCH-X-treated monkeys. CONCLUSION: The systemic effects of treatment with SCH-X were a suppressed acute-phase reaction (reduction in C-reactive protein level) in the 3 treated monkeys with CIA that remained asymptomatic, and an altered antibody response toward type II collagen. The results suggest that the CCR5 antagonist SCH-X might have a strong clinical potential for treatment during periods of active inflammation, as seen in RA.


Subject(s)
Arthritis, Experimental/prevention & control , CCR5 Receptor Antagonists , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Collagen Type II , Follow-Up Studies , Macaca mulatta , Male
16.
Am J Transplant ; 4(11): 1756-61, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15476473

ABSTRACT

Treatment of kidney graft recipients with antibodies that may specifically suppress the anti-donor response would be an ideal situation to prevent graft rejection. MHC class-II-specific antibodies and, in particular, DR specific antibodies have often been proposed as treatment to prevent antigen presentation, and thus graft destruction. Here we report an attempt to prevent graft rejection using a humanized MHC class-II-specific monoclonal antibody CDP855 in a cynomolgus monkey kidney graft model. A modest delay in graft rejection was observed when the antibody was given only on days 0, 1 and 2 after transplantation. Unexpectedly 50% of the animals succumbed of a viral infection, most likely CMV in two of three cases, prior to graft rejection in the first week post-transplantation. We speculate that the antibody treatment triggered CMV activation, possibly as a consequence of the activation of factors such as NF-kappab by the interaction of the antibody and its target cells.


Subject(s)
Antibodies/therapeutic use , Cytomegalovirus Infections/epidemiology , Graft Rejection/immunology , Graft Rejection/prevention & control , Histocompatibility Antigens Class II/immunology , Kidney Transplantation/immunology , Animals , Disease Models, Animal , Macaca fascicularis , Postoperative Complications/epidemiology
17.
Drug Discov Today ; 9(12): 517-24, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15183159

ABSTRACT

The last few decades of the 20th century have shown an intensified search for safer and more effective medications against chronic diseases that burden ageing societies of the western world. The impressive development of biotechnological production techniques has greatly facilitated the pharmaceutical development of relatively non-toxic biological molecules. However, despite the huge investments, only a few effective therapies for immune-based diseases have reached the clinic. In this article we use examples from monoclonal antibody trials to discuss the validity and predictive strength of the animal models currently used for the development of effective therapies.


Subject(s)
Disease Models, Animal , Immune System Diseases/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Drug Evaluation, Preclinical/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune System Diseases/immunology , Immunotherapy/methods , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Organ Transplantation
18.
J Immunol ; 172(9): 5753-64, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100322

ABSTRACT

Approaches that prevent acute rejection of renal transplants in a rhesus monkey model were studied to determine a common mechanism of acceptance. After withdrawal of immunosuppression, all 14 monkeys retained normal allograft function for >6 mo. Of these, nine rejected their renal allograft during the study, and five maintained normal function throughout the study period. The appearance of TGF-beta 1(+) interstitial mononuclear cells in the graft coincided with a nonrejection histology, whereas the absence/disappearance of these cells was observed with the onset of rejection. Analysis with a variety of TGF-beta 1-reactive Abs indicated that the tolerance-associated infiltrates expressed the large latent complex form of TGF-beta 1. Peripheral leukocytes from rejecting monkeys lacking TGF-beta 1(+) allograft infiltrates responded strongly to donor Ags in delayed-type hypersensitivity trans-vivo assays. In contrast, allograft acceptors with TGF-beta 1(+) infiltrates demonstrated a much weaker peripheral delayed-type hypersensitivity response to donor alloantigens (p < 0.01 vs rejectors), which could be restored by Abs that either neutralized active TGF-beta 1 or blocked its conversion from latent to active form. Anti-IL-10 Abs had no restorative effect. Accepted allografts had CD8(+) and CD4(+) interstitial T cell infiltrates, but only the CD4(+) subset included cells costaining for TGF-beta 1. Our data support the hypothesis that the recruitment of CD4(+) T regulatory cells to the allograft interstitium is a final common pathway for metastable renal transplant tolerance in a non-human primate model.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Kidney Transplantation/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/biosynthesis , Transplantation Tolerance/immunology , Transplantation, Homologous/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , Hypersensitivity, Delayed/immunology , Kidney Transplantation/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Macaca mulatta , Mice , Mice, SCID , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1 , Transplantation, Homologous/pathology
19.
Transplantation ; 76(9): 1359-68, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14627916

ABSTRACT

BACKGROUND: Transplantation of islets of Langerhans can restore insulin production in diabetic patients. Because of the shortage of human donor organs, transplantation of porcine islets may be an alternative solution. The present study was aimed at the characterization of rejection mechanisms of porcine islets transplanted into eight nondiabetic monkeys under the kidney capsule. METHODS: Cultured adult pig islets were used, which showed no expression of the galactose(alpha1,3)galactose epitope, major histocompatibility complex class II, or CD45, and no binding of antibodies or complement after exposure to monkey serum. Immunosuppression consisted of cyclophosphamide, cyclosporine A (CsA), and steroids (group 1); or antithymocyte globulin, anti-interleukin-2 receptor antibody, CsA, and steroids (group 2). In three animals of group 2, islets were also transplanted in the portal vein. RESULTS: Although all monkeys had preformed anti-pig antibodies, no correlation was found between antibody titers and rejection and no deposition of antibodies or complement was observed in the grafts. Group 1 showed islets up to day 11, followed by T-cell infiltration and rejection at approximately day 14. In group 2, two monkeys showed infiltrates consisting predominantly of T cells starting at approximately day 29, whereas two monkeys showed well-preserved islets without infiltration up to day 53. In the livers of the three monkeys that also received islets intraportally and were resectioned on days 21, 33, and 49, no islets could be detected. CONCLUSIONS: This study demonstrates that cultured adult pig islets can survive in the monkey for more than 53 days without signs of rejection under standard immunosuppression.


Subject(s)
Cyclosporine/therapeutic use , Graft Survival/immunology , Immunosuppression Therapy/methods , Islets of Langerhans Transplantation/immunology , T-Lymphocytes/immunology , Transplantation, Heterologous/immunology , Animals , Cell Culture Techniques/methods , Cyclosporine/administration & dosage , Female , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Injections, Intramuscular , Islets of Langerhans/cytology , Islets of Langerhans Transplantation/pathology , Islets of Langerhans Transplantation/physiology , Macaca fascicularis , Macaca mulatta , Prednisolone/administration & dosage , Prednisolone/therapeutic use , Swine , Transplantation, Heterologous/pathology
20.
Cancer Immunol Immunother ; 52(9): 569-75, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14627129

ABSTRACT

CD86 and CD80 costimulatory antigens are highly overexpressed on Hodgkin/Reed-Sternberg cells in patients with Hodgkin's disease (HD) and are candidate target antigens for immunotoxins in order to eliminate minimal residual disease. In this study we have evaluated the pharmacokinetics (and immunological) toxicity in rhesus monkeys of immunotoxins consisting of gelonin conjugated to anti-CD86 (alphaCD86-IT). Both alphaCD86-IT and alphaCD80-IT inhibited protein synthesis in B cell lines from rhesus monkeys and inhibited the mixed leukocyte reaction. Reactivity of the alphaCD86 antibody with rhesus monkey CD86 (RhCD86) was shown by cloning and transfecting RhCD86, which conferred reactivity of the alphaCD86 antibody used in this study. alphaCD86-IT was administered as single intravenous bolus injection in four rhesus monkeys, and achieved plasma concentration in 50-fold excess able to eliminate cultured Hodgkin/Reed-Sternberg cells up to 6 h. The animals were capable of generating primary immune responses to both gelonin and murine IgG within 9 days after infusion with alphaCD86-IT. No evidence could be found of significant (immunological) toxicity. The results of this study show that alphaCD86-IT can be applied safely in an effective dose.


Subject(s)
Antibodies/immunology , Immunotoxins/pharmacology , Animals , Antigens, CD/immunology , B7-1 Antigen/immunology , B7-2 Antigen , Immunotoxins/pharmacokinetics , Immunotoxins/toxicity , Lymphocyte Culture Test, Mixed , Macaca mulatta , Membrane Glycoproteins/immunology , Plant Proteins/immunology , Reed-Sternberg Cells , Ribosome Inactivating Proteins, Type 1 , T-Lymphocytes/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...