Network


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

Hotspot


Dive into the research topics where Michael Schumm is active.

Publication


Featured researches published by Michael Schumm.


British Journal of Haematology | 2006

Safe adoptive transfer of virus‐specific T‐cell immunity for the treatment of systemic adenovirus infection after allogeneic stem cell transplantation

Tobias Feuchtinger; Susanne Matthes-Martin; Celine Richard; Thomas Lion; Monika Führer; Klaus Hamprecht; Rupert Handgretinger; Christina Peters; Friedhelm Schuster; Robert Beck; Michael Schumm; Ramin Lotfi; Gerhard Jahn; Peter Lang

During periods of immunosuppression, such as postallogeneic stem cell transplantation (SCT), patients are at significant risk for severe viral infections. Human adenovirus (HAdV) infection is a serious complication post‐SCT, especially in children. Virus‐specific T cells are essential for the clearance of HAdV, as antiviral chemotherapy has revealed limited success. We present feasibility data for a new treatment option using virus‐specific donor T cells for adoptive transfer of immunity to patients with HAdV‐infection/reactivation. Virus‐specific donor T cells were isolated and infused into nine children with systemic HAdV infection after SCT. Isolation was based on γ‐interferon (IFN‐γ) secretion after short in vitro stimulation with viral antigen, resulting in a combination of CD4+ and CD8+ T cells. 1·2–50 × 103/kg T cells were infused for adoptive transfer. Isolated cells showed high specificity and markedly reduced alloreactivity in vitro. Adoptive transfer of HAdV‐specific immunity was successful in five of six evaluable patients, documented by a dose‐independent and sustained in vivo expansion of HAdV‐specific T cells, associated with a durable clearance/decrease of viral copies. T‐cell infusion was well tolerated in all nine patients, except one case with graft‐versus‐host disease II of the skin. In conclusion, induction of a specific T‐cell response through adoptive transfer was feasible and effective. When performed early in the course of infection, adoptive T‐cell transfer may protect from HAdV‐related complications.


Bone Marrow Transplantation | 2001

Megadose transplantation of purified peripheral blood CD34(+) progenitor cells from HLA-mismatched parental donors in children.

Rupert Handgretinger; Thomas Klingebiel; Peter Lang; Michael Schumm; S Neu; Andreas Geiselhart; Peter Bader; Paul-Gerhard Schlegel; Johann Greil; D Stachel; Rj Herzog; Dietrich Niethammer

We performed HLA-mismatched stem cell transplantation with megadoses of purified positively selected mobilized peripheral blood CD34+ progenitor cells (PBPC) from related adult donors in 39 children lacking an otherwise suitable donor. The patients received a mean number of 20.7 ± 9.8 × 106/kg purified CD34+ and a mean number of 15.5 ± 20.4 × 103/kg CD3+ T lymphocytes. The first seven patients received short term (<4 weeks) GVHD prophylaxis with cyclosporin A, whereas in all the following 32 patients no GVHD prophylaxis was used. In 38 evaluable patients, five patients experienced primary acute GVHD grade I and one patient grade II. In 32 patients, no signs of primary GVHD were seen and GVHD only occurred after T cell add backs. T cell reconstitution was more rapid if the number of transplanted CD34+ cells exceeded 20 × 106/kg. Of the 39 patients, 15 are alive and well, 13 died due to relapse and 10 transplant-related deaths occurred. We conclude that the HLA barrier can be overcome by transplantation of megadoses of highly purified mismatched CD34+ stem cells. GVHD can be prevented without pharmacological immunosuppression by the efficient T cell depletion associated with the CD34+ positive selection procedure. This approach offers a promising therapeutic option for every child without an otherwise suitable donor. Bone Marrow Transplantation (2001) 27, 777–783.


Blood | 2010

Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation

Tobias Feuchtinger; Kathrin Opherk; Wolfgang Bethge; Max S. Topp; Friedhelm Schuster; Eva M. Weissinger; Mohamad Mohty; Reuven Or; Michael Maschan; Michael Schumm; Klaus Hamprecht; Rupert Handgretinger; Peter Lang; Hermann Einsele

Cytomegalovirus (CMV) disease and infection refractory to antiviral treatment after allogeneic stem cell transplantation (allo-SCT) is associated with a high mortality. Adoptive transfer of CMV-specific T cells could reconstitute viral immunity after SCT and could protect from CMV-related complications. However, logistics of producing virus-specific T-cell grafts limited the clinical application. We treated 18 patients after allo-SCT from human leukocyte antigen-mismatched/haploidentical or human leukocyte antigen-matched unrelated donors with polyclonal CMV-specific T cells generated by ex vivo stimulation with pp65, followed by isolation of interferon-γ-producing cells. Patients with CMV disease or viremia refractory to antiviral chemotherapy or both were eligible for adoptive T-cell transfer and received a mean of 21 × 10³/kg pp65-specific T cells. In 83% of cases CMV infection was cleared or viral burden was significantly reduced, even in cases of CMV encephalitis (n = 2). Viral control was associated with in vivo expansion of CMV-specific T lymphocytes in 12 of 16 evaluable cases, resulting in reconstitution of antiviral T-cell responses, without graft-versus-host disease induction or acute side effects. Our findings indicate that the infusion of low numbers of CMV-specific T cells is safe, feasible, and effective as a treatment on demand for refractory CMV infection and CMV disease after allo-SCT.


Circulation | 2008

Platelet-Derived Stromal Cell–Derived Factor-1 Regulates Adhesion and Promotes Differentiation of Human CD34+ Cells to Endothelial Progenitor Cells

Konstantinos Stellos; Harald Langer; Karin Daub; Tanja Schoenberger; Alexandra Gauss; Tobias Geisler; Boris Bigalke; Iris Mueller; Michael Schumm; Iris Schaefer; Peter Seizer; Bjoern F. Kraemer; Dorothea Siegel-Axel; Andreas E. May; Stephan Lindemann; Meinrad Gawaz

Background— Peripheral homing of progenitor cells in areas of diseased organs is critical for tissue regeneration. The chemokine stromal cell–derived factor-1 (SDF-1) regulates homing of CD34+ stem cells. We evaluated the role of platelet-derived SDF-1 in adhesion and differentiation of human CD34+ cells into endothelial progenitor cells. Methods and Results— Adherent platelets express substantial amounts of SDF-1 and recruit CD34+ cells in vitro and in vivo. A monoclonal antibody to SDF-1 or to its counterreceptor, CXCR4, inhibits stem cell adhesion on adherent platelets under high arterial shear in vitro and after carotid ligation in mice, as determined by intravital fluorescence microscopy. Platelets that adhere to human arterial endothelial cells enhance the adhesion of CD34+ cells on endothelium under flow conditions, a process that is inhibited by anti-SDF-1. During intestinal ischemia/reperfusion in mice, anti-SDF-1 and anti-CXCR4, but not isotype control antibodies, abolish the recruitment of CD34+ cells in microcirculation. Moreover, platelet-derived SDF-1 binding to CXCR4 receptor promotes platelet-induced differentiation of CD34+ cells into endothelial progenitor cells, as verified by colony-forming assays in vitro. Conclusions— These findings imply that platelet-derived SDF-1 regulates adhesion of stem cells in vitro and in vivo and promotes differentiation of CD34+ cells to endothelial progenitor cells. Because tissue regeneration depends on recruitment of progenitor cells to peripheral vasculature and their subsequent differentiation, platelet-derived SDF-1 may contribute to vascular and myocardial regeneration.


Circulation Research | 2006

Adherent Platelets Recruit and Induce Differentiation of Murine Embryonic Endothelial Progenitor Cells to Mature Endothelial Cells In Vitro

Harald Langer; Andreas E. May; Karin Daub; Ulrich Heinzmann; Peter Lang; Michael Schumm; Dietmar Vestweber; Steffen Massberg; Tanja Schönberger; Iris Pfisterer; Antonis K. Hatzopoulos; Meinrad Gawaz

The homing and differentiation mechanisms of endothelial progenitor cells (EPCs) at sites of vascular lesions are unclear. To investigate whether platelets play a role in the recruitment and differentiation of EPCs, we made use of a robust mouse embryonic EPC (eEPC) line that reliably differentiates to a mature endothelial phenotype. We found that platelets stimulate chemotaxis and migration of these murine eEPCs. Further, the substantial adhesion of murine eEPCs on immobilized platelets that occurs under dynamic flow conditions is inhibited by neutralizing anti–P-selectin glycoprotein ligand-1 and anti–VLA-4 (β1-integrin) monoclonal antibodies but not by anti-CD11b (aM-integrin; macrophage antigen-1). Coincubation of murine eEPCs with platelets for 5 days induced differentiation of EPCs to mature endothelial cells as verified by positive von Willebrand factor immunofluorescence and detection of Weibel Palade bodies through electron microscopy. We conclude that platelets may play a critical part in the capture and subsequent differentiation of murine eEPCs at sites of vascular lesions, revealing a possible new role of platelets in neoendothelization after vascular injury.


Journal of hematotherapy | 1999

Isolation of Highly Purified Autologous and Allogeneic Peripheral CD34+ Cells Using the CliniMACS Device

Michael Schumm; Peter Lang; Gwendolyn Taylor; Selim Kuçi; Thomas Klingebiel; Hans-Jörg Bühring; Andreas Geiselhart; Dietrich Niethammer; Rupert Handgretinger

The CliniMACS CD34+ selection device was used for positive selection of apheresis products for autologous transplantation from 10 patients with malignant diseases and for allogeneic transplantation from 26 healthy donors. A total of 71 separations were performed. In 1 allogeneic donor, CD34+ progenitors were also isolated from bone marrow. Between 0.27 and 8.9 x 10(10) nucleated cells (median 4.9 x 10(10)) containing 0.09%-10.8% (median 0.67%) CD34+ progenitor cells were separated. After separation, a median number of 227 x 10(6) mononuclear cells (MNC) (51-524) were recovered, with a median viability of 99% (22%-100%) and a median purity of 97.0% (68.3%-99.7%) CD34+ cells. Depletion of T cells was extensive, with a median of 0.04% residual CD3+ cells (range <0.01%-0.92%). Residual CD19+ cells were between <0.01% and 17%, including CD34+CD19+ cells. Recovery of CD34+ cells was calculated according to the ISHAGE guidelines and ranged from 24% to 105% (median 71%). We conclude that with the CliniMACS device CD34+ cells with high purity and recovery can be isolated with concomitant effective T cell depletion in the allogeneic setting and with a high purging efficacy in the autologous setting.


Journal of Clinical Oncology | 2013

Adoptive Transfer of Epstein-Barr Virus (EBV) Nuclear Antigen 1–Specific T Cells As Treatment for EBV Reactivation and Lymphoproliferative Disorders After Allogeneic Stem-Cell Transplantation

Vanya Icheva; Simone Kayser; Daniel Wolff; Sebastian Tuve; Christina Kyzirakos; Wolfgang Bethge; Johann Greil; Michael H. Albert; Wolfgang Schwinger; Michaela Nathrath; Michael Schumm; Stefan Stevanovic; Rupert Handgretinger; Peter J. Lang; Tobias Feuchtinger

PURPOSE Reactivation of Epstein-Barr virus (EBV) after allogeneic stem-cell transplantation (SCT) can lead to severe life-threatening infections and trigger post-transplantation lymphoproliferative disease (PTLD). Since EBV-specific T cells could prevent PTLD, cellular immunotherapy has been a promising treatment option. However, generation of antigen-specific T-cell populations has been difficult within a short time frame. PATIENTS AND METHODS To improve availability in urgent clinical conditions, we developed a rapid protocol for isolation of polyclonal EBV nuclear antigen 1 (EBNA-1) -specific T cells by using an interferon gamma (IFN-γ) capture technique. RESULTS We report on the use of adoptive transfer of EBNA-1-specific T cells in 10 pediatric and adult patients with EBV viremia and/or PTLD after SCT. No acute toxicity or graft-versus-host disease (GVHD) of more than grade 2 occurred as a result of adoptive T-cell transfer. In vivo expansion of transferred EBNA-1-specific T cells was observed in eight of 10 patients after a median of 16 days following adoptive transfer that was associated with clinical and virologic response in seven of them (70%). None of the responders had EBV-associated mortality. Within clinical responders, three patients were disease free by the day of last follow-up (2 to 36 months), three patients died of other infectious complications, and one patient died as a result of relapse of malignancy. EBV-related mortality was observed in two of 10 patients, and another patient had ongoing viremia without clinical symptoms at last follow-up. CONCLUSION Adoptive ex vivo transfer of EBNA-1-specific T cells is a feasible and well-tolerated therapeutic option, representing a fast and efficient procedure to achieve reconstitution of antiviral T-cell immunity after SCT.


Annals of the New York Academy of Sciences | 1999

Transplantation of megadoses of purified haploidentical stem cells.

Rupert Handgretinger; Michael Schumm; Peter Lang; Johann Greil; A. Reiter; Peter Bader; Dietrich Niethammer; Thomas Klingebiel

Abstract: Peripheral mobilized parental CD34+ progenitors were isolated and used for the hematopoietic reconstitution after a myeloablative therapy in 23 pediatric patients with various diseases. Fourteen donors were human leukocyte antigen (HLA) three‐loci mismatches, 6 donors were two‐loci and 3 donors were one‐locus mismatches. For depletion of T‐lymphocytes, a positive selection of the mobilized peripheral CD34+ progenitors using the method of magnetic‐activated cell sorting (MACS) was used. The purity of the CD34+ cells after MACS‐sorting was 98‐99%, the average number of transplanted CD34+ cells was 14.2 3 × 106/kg (range 5.4‐3 9 × 3 I 06/kg) and the average number of infused T‐lymphocytes was 1.4 × 3 104/kg. Due to this low T cell number, only a short‐term or no prophylaxis of graft‐versus‐host disease (GVHD) was necessary and no GVHD was seen. A significant GVHD was only seen in patients after add‐back of donor T‐lymphocytes, which was performed in some patients for prevention of relapse or in patients who showed a transient mixed chimerism. Since the B lymphocyte contamination of the isolated CD34+ cells was low in the range of 0.2%, no Epstein‐Barr virus (EBV)‐associated lymphoproliferative syndrome was observed. A primary engraftment was seen in 18 patients. Nonengraftment and rejection occurred in three and two patients, respectively. In four of these 5 patients, a second transplant using purified CD34+ cells from the same donor after an immunological reconditioning regimen resulted in a complete and sustained hematopoietic reconstitution. The speed of the immunological recovery was dependent on the number of transplanted CD34+ cells and was more rapid if this number was >20 × 3 106/kg. Eleven of the 23 patients are alive and disease free with a median follow‐up of 12 months (range 2‐30). The main cause of death was relapse (7 patients), and only one fatal infection was seen. Our data suggest that the transplantation of megadoses of haploidentical CD34+ cells is a realistic therapeutic option for patients who otherwise have no suitable donor, and an alternative to the use of unrelated cord blood.


British Journal of Haematology | 2004

Transplantation of a combination of CD133+ and CD34+ selected progenitor cells from alternative donors.

Peter Lang; Peter Bader; Michael Schumm; Tobias Feuchtinger; Hermann Einsele; Monika Führer; Christof Weinstock; Rupert Handgretinger; Selim Kuçi; David D. Martin; Dietrich Niethammer; Johann Greil

Positive selected haematopoietic stem cells are increasingly used for allogeneic transplantation with the CD34 antigen employed in most separation techniques. However, the recently described pentaspan molecule CD133 appears to be a marker of more primitive haematopoietic progenitors. Here we report our experience with a new CD133‐based selection method in 10 paediatric patients with matched unrelated (n = 2) or mismatched‐related donors (n = 8). These patients received a combination of stem cells (median = 29·3 × 106/kg), selected with either anti‐CD34 or anti‐CD133 coated microbeads. The proportion of CD133+ selected cells was gradually increased from patient to patient from 10% to 100%. Comparison of CD133+ and CD34+ separation procedures revealed similar purity and recovery of target populations but a lower depletion of T cells by CD133+ selection (3·7 log vs. 4·1 log, P < 0·001). Both separation procedures produced >90% CD34+/CD133+ double positive target cells. Engraftment occurred in all patients (sustained primary, n = 8; after reconditioning, n = 2). No primary acute graft versus host disease (GvHD) ≥ grade II or chronic GvHD was observed. The patients showed a rapid platelet recovery (median time to independence from substitution = 13·5 d), whereas T cell regeneration was variable. Five patients are alive with a median follow‐up of 10 months. Our data demonstrates the feasibility of CD133+ selection for transplantation from alternative donors and encourages further trials with total CD133+ separated grafts.


Bone Marrow Transplantation | 1998

Isolation and transplantation of autologous peripheral CD34+ progenitor cells highly purified by magnetic-activated cell sorting.

Rupert Handgretinger; Peter Lang; Michael Schumm; G Taylor; S Neu; E Koscielnak; Dietrich Niethammer; Thomas Klingebiel

Peripheral stem cells were mobilized and collected in 26 pediatric patients with malignant diseases. A total of 47 leukaphereses were performed in the 26 patients. The mean number of nucleated cells collected was 4.5 ± 2.6 × 108/kg and the number of CD34+ progenitors collected was 6.7 ± 6.8 × 106/kg. CD34-positive selection was performed using a two-step method of magnetic-activated cell sorting (MACS) in 24 patients or a combination of an immunoaffinity column and MACS in two patients. The purity of the positively selected CD34+ progenitors was 98.8 ± 0.7% and the number of isolated CD34+ cells was 6.5 ± 5.9 × 106/kg. Thus, the mean recovery of CD34+ cells was 93 ± 10%. In 22 of the 26 patients, high-dose chemotherapy was performed with subsequent reinfusion of the highly purified CD34+ cells. In all 22 patients, a normal hematopoietic reconstitution was seen with a mean time of 12.4 ± 2.7 days to reach >0.5 × 109/l neutrophils (range 8–19 days). The time to reach independence from platelet transfusion was 31.6 ± 17.0 days (range 16–78 days). There were no transplant-related deaths. In summary, we have shown that mobilized peripheral CD34+ progenitors can be highly purified with a good recovery and that reinfusion of these cells after high-dose chemotherapy results in a rapid, complete and sustained engraftment. We conclude that this method can be used for purging in any CD34-negative malignancies and for autologous T and B cell depletion in the treatment of autoimmune diseases with high-dose immunoablative therapy.

Collaboration


Dive into the Michael Schumm's collaboration.

Top Co-Authors

Avatar

Peter Lang

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Bader

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Thomas Klingebiel

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthias Pfeiffer

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge