Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Eva-Maria Kuhn is active.

Publication


Featured researches published by Eva-Maria Kuhn.


Transplantation | 2003

Prevention of kidney allograft rejection using anti-cd40 and anti-cd86 in primates

Krista G. Haanstra; Jan Ringers; Ella A. Sick; Seema Ramdien-Murli; Eva-Maria Kuhn; Louis Boon; Margreet Jonker

Background. Costimulation blockade has been proposed to induce allograft tolerance. We combined an antagonist anti-CD40 monoclonal antibody (mAb) with an antagonist anti-CD86 mAb in a rhesus monkey kidney allograft model. We chose this combination because it leaves CD80-CD152 signaling unimpaired, allowing for the down-regulatory effect of CD152 signaling to take place through this pathway. Methods. Rhesus monkeys underwent transplantation with a major histocompatibility complex–mismatched kidney. One group of animals received anti-CD40 alone, and a second group received the combination of anti-CD40 and anti-CD86, twice weekly for 56 days. Results. Three animals with low levels of anti-CD40 rejected the transplanted kidney while still receiving treatment. Three animals with high levels of anti-CD40 rejected at days 91, 134, and 217 with signs of chronic rejection. Animals treated with the combination of anti-CD40 and anti-CD86 mAbs rejected their kidneys at days 61, 75, and 78, shortly after cessation of treatment. Two animals were killed on days 71 and 116 with a blocked ureter. These animals developed virtually no signs of tubulitis or infiltration during treatment and no donor-specific alloantibodies. Conclusions. Both treatment protocols prevented rejection for the duration of the treatment in most animals. Blocking costimulation by anti-CD40 or by anti-CD40 plus anti-CD86 may be an effective method to prevent graft rejection and may obviate the need for other immunosuppressive drugs, especially in the immediate posttransplantation period.


Transplantation | 1999

Prevention of renal allograft rejection in primates by blocking the B7/CD28 pathway

Miriam Ossevoort; Jan Ringers; Eva-Maria Kuhn; Louis Boon; K Lorré; Y van den Hout; Jan A. Bruijn; H de Boer; Margreet Jonker; P. De Waele

BACKGROUNDnThere is accumulating evidence that blockade of the costimulatory pathways offers a valid approach for immune suppression after solid organ transplantation. In this study, the efficacy of anti-CD80 and anti-CD86 monoclonal antibodies (mAbs) in combination with cyclosporine (CsA) to prevent renal allograft rejection was tested in non-human primates.nnnMETHODSnRhesus monkeys were transplanted with a partly major histocompatibility complex-matched kidney on day 0. Anti-CD80 and anti-CD86 mAbs were administered intravenously daily for 14 days starting at day - 1. CsA was given intramuscularly for 35 days starting just after transplantation. The kidney function was monitored by determining serum creatinine levels.nnnRESULTSnThe combination of anti-CD80 and anti-CD86 mAbs completely abrogated the mixed lymphocyte reaction. Untreated rhesus monkeys rejected the kidney allograft in 5-7 days. Treatment with anti-CD80 plus anti-CD86 mAbs resulted in a significantly prolonged graft survival of 28+ 7 days (P=0.025). There were no clinical signs of side effects or rejection during treatment. Kidney graft rejection started after the antibody therapy was stopped. The anti-mouse antibody response was delayed from day 10 to 30 after the first injection. No difference in graft survival was observed between animals treated with CsA alone or in combination with anti-CD80 and anti-CD86 mAbs. However, treatment with anti-CD80 and anti-CD86 mAbs reduced development of vascular rejection.nnnCONCLUSIONSnIn combination, anti-CD80 and antiCD86 mAbs abrogate T-cell proliferation in vitro, delay the anti-mouse antibody response in vivo, and prevent graft rejection and development of graft vascular disease in a preclinical vascularized transplant model in non-human primates.


Transplantation | 2005

The autoimmune response to vimentin after renal transplantation in nonhuman primates is immunosuppression dependent.

Margreet Jonker; Anna J. Danskine; Krista G. Haanstra; Jacqueline Wubben; Ivanela Kondova; Eva-Maria Kuhn; Marlene L. Rose

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.


American Journal of Transplantation | 2004

Treatment with Anti-MHC-Class-II Antibody Postpones Kidney Allograft Rejection in Primates but Increases the Risk of CMV Activation

Margreet Jonker; Jan Ringers; Eva-Maria Kuhn; Bert't Hart; Roland Foulkes

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‐κb by the interaction of the antibody and its target cells.


Vaccine | 2001

Safety and immunogenicity of ALVAC wild-type human p53 (vCP207) by the intravenous route in rhesus macaques

Brigitte Rosenwirth; Eva-Maria Kuhn; Jonathan L. Heeney; Christian Hurpin; James Tartaglia; Marie-Claude Bonnet; Philippe Moingeon; Lorne F. Erdile

p53 is over-expressed in approximately 50% of human cancers, and transfer of cytotoxic T lymphocytes (CTL) against wild-type p53 protects mice against p53-over-expressing tumors, suggesting that p53 might be an attractive target for immunotherapy. Immunization of mice with a recombinant canarypox virus, ALVAC, expressing human wild-type p53 (vCP207) prevented growth of p53-over-expressing tumors. Since intravenous administration induced better immune responses in mice than other routes, we have proposed to use this route in cancer patients. However, because this vector has never been administered intravenously to humans, and because of the possibility of inducing auto-immunity to a self-antigen, we felt it was necessary to first evaluate safety in rhesus macaques. We found that three intravenous administrations of vCP207 at proportional doses up to 10x those proposed for humans produced no abnormalities in hematologic or clinical chemistry parameters. Serologic markers of autoimmunity and inflammation were unaffected, despite the >95% amino acid identity between human and rhesus p53. Pathological examination of numerous tissues yielded findings comparable to those in animals given placebo. Some animals showed anti-p53 antibody responses following vaccination, indicating that tolerance could be broken to some extent. However, with the exception of one animal with a possible delayed type hypersensitivity reaction to p53 protein, we did not see evidence for a cell-mediated response. The safety profile in monkeys with ALVAC-p53 provides encouragement for using such live, modified vectors via the intravenous route for human immunotherapy.


Transplantation | 2006

No synergy between ATG induction and costimulation blockade induced kidney allograft survival in rhesus monkeys

Krista G. Haanstra; Ella A. Sick; Jan Ringers; Jacqueline Wubben; Eva-Maria Kuhn; Bert A. 't Hart; Louis Boon; Margreet Jonker

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 CD95low/CD11alow 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.


Journal of Virology | 2001

Characteristics of a Pathogenic Molecular Clone of an End-Stage Serum-Derived Variant of Simian Immunodeficiency Virus (SIVF359)

Lennart Holterman; Rob Dubbes; James I. Mullins; Gerald Learn; Henk Niphuis; Wim Koornstra; Gerrit Koopman; Eva-Maria Kuhn; Alison Wade-Evans; Brigitte Rosenwirth; Joost J. Haaijman; Jonathan L. Heeney

ABSTRACT End-stage simian immunodeficiency virus (SIV) isolates are suggested to be the most fit of the evolved virulent variants that precipitate the progression to AIDS. To determine if there were common characteristics of end-stage variants which emerge from accelerated cases of AIDS, a molecular clone was derived directly from serum following in vivo selection of a highly virulent SIV isolate obtained by serial end-stage passage in rhesus monkeys (Macaca mulatta). This dominant variant caused a marked cytopathic effect and replicated to very high levels in activated but not resting peripheral blood lymphocytes. Furthermore, although this clone infected but did not replicate to detectable levels in rhesus monocyte-derived macrophages, these cells were able to transmit infection to autologous T cells upon contact. Interestingly, although at low doses this end-stage variant did not use any of the known coreceptors except CCR5, it was able to infect and replicate in human peripheral blood mononuclear cells homozygous for the Δ32 deletion of CCR5, suggesting the use of a novel coreceptor. It represents the first pathogenic molecular clone of SIV derived from viral RNA in serum and provides evidence that not only the genetic but also the biological characteristics acquired by highly fit late-stage disease variants may be distinct in different hosts.


Transplantation Proceedings | 2001

T-Cell-Directed Immunosuppression Allows Prolonged Survival of Xenogeneic Pig Islets in Monkeys

Margreet Jonker; Josephine K. R. A. Rijkelijkhuizen; Krista G. Haanstra; Eva-Maria Kuhn; Jan Ringers; E. Bouwman

IN a previous study we demonstrated that porcine islets of Langerhans transplanted to monkeys will not be hyperacutely rejected. In immunosuppressed monkeys (cycplophosphamide, cyclosporine [CyA], and prednisolone) rejection occurred in the second week posttransplantation. Rejection infiltrates were dominated by T cells. We therefore wanted to investigate whether a more T-cell-directed immunosuppressive regimen could improve graft survival. Our aim was to deplete recipients of T cells prior to transplantation (using ATG) and prevent rejection posttransplantation using a combination of CyA, prednisolone, and anti-IL-2R antibody (Zenapax).


Journal of Virology | 1998

The Interleukin-17 Gene of Herpesvirus Saimiri

Andrea Knappe; Christian Hiller; Henk Niphuis; François Fossiez; Mathias Thurau; Sabine Wittmann; Eva-Maria Kuhn; Serge Lebecque; Jacques Banchereau; Brigitte Rosenwirth; Bernhard Fleckenstein; Jonathan L. Heeney; Helmut Fickenscher


Transplantation | 2005

Costimulation blockade followed by a 12-week period of cyclosporine A facilitates prolonged drug-free survival of rhesus monkey kidney allografts.

Krista G. Haanstra; Ella A. Sick; Jan Ringers; Jacqueline Wubben; Eva-Maria Kuhn; Louis Boon; Margreet Jonker

Collaboration


Dive into the Eva-Maria Kuhn's collaboration.

Top Co-Authors

Avatar

Margreet Jonker

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Jan Ringers

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Krista G. Haanstra

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Brigitte Rosenwirth

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Wubben

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Louis Boon

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Henk Niphuis

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar

Ivanela Kondova

Biomedical Primate Research Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge