Network


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

Hotspot


Dive into the research topics where Ralf C. Bargou is active.

Publication


Featured researches published by Ralf C. Bargou.


Journal of Clinical Oncology | 2011

Targeted Therapy With the T-Cell–Engaging Antibody Blinatumomab of Chemotherapy-Refractory Minimal Residual Disease in B-Lineage Acute Lymphoblastic Leukemia Patients Results in High Response Rate and Prolonged Leukemia-Free Survival

Max S. Topp; Peter Kufer; Nicola Gökbuget; Mariele Goebeler; Matthias Klinger; Svenja Neumann; Heinz-A. Horst; Thorsten Raff; Andreas Viardot; Mathias Schmid; Matthias Stelljes; Markus Schaich; Evelyn Degenhard; Rudolf Köhne-Volland; Monika Brüggemann; Oliver G. Ottmann; Heike Pfeifer; Thomas Burmeister; Dirk Nagorsen; Margit Schmidt; Ralf Lutterbuese; Carsten Reinhardt; Patrick A. Baeuerle; Michael Kneba; Hermann Einsele; Gert Riethmüller; Dieter Hoelzer; Gerhard Zugmaier; Ralf C. Bargou

PURPOSE Blinatumomab, a bispecific single-chain antibody targeting the CD19 antigen, is a member of a novel class of antibodies that redirect T cells for selective lysis of tumor cells. In acute lymphoblastic leukemia (ALL), persistence or relapse of minimal residual disease (MRD) after chemotherapy indicates resistance to chemotherapy and results in hematologic relapse. A phase II clinical study was conducted to determine the efficacy of blinatumomab in MRD-positive B-lineage ALL. PATIENTS AND METHODS Patients with MRD persistence or relapse after induction and consolidation therapy were included. MRD was assessed by quantitative reverse transcriptase polymerase chain reaction for either rearrangements of immunoglobulin or T-cell receptor genes, or specific genetic aberrations. Blinatumomab was administered as a 4-week continuous intravenous infusion at a dose of 15 μg/m2/24 hours. RESULTS Twenty-one patients were treated, of whom 16 patients became MRD negative. One patient was not evaluable due to a grade 3 adverse event leading to treatment discontinuation. Among the 16 responders, 12 patients had been molecularly refractory to previous chemotherapy. Probability for relapse-free survival is 78% at a median follow-up of 405 days. The most frequent grade 3 and 4 adverse event was lymphopenia, which was completely reversible like most other adverse events. CONCLUSION Blinatumomab is an efficacious and well-tolerated treatment in patients with MRD-positive B-lineage ALL after intensive chemotherapy. T cells engaged by blinatumomab seem capable of eradicating chemotherapy-resistant tumor cells that otherwise cause clinical relapse.


Journal of Clinical Oncology | 2014

Phase II Trial of the Anti-CD19 Bispecific T Cell–Engager Blinatumomab Shows Hematologic and Molecular Remissions in Patients With Relapsed or Refractory B-Precursor Acute Lymphoblastic Leukemia

Max S. Topp; Nicola Gökbuget; Gerhard Zugmaier; Petra Klappers; Matthias Stelljes; Svenja Neumann; Andreas Viardot; Reinhard Marks; Helmut Diedrich; Christoph Faul; Albrecht Reichle; Heinz-August Horst; Monika Brüggemann; Dorothea Wessiepe; Chris Holland; Shilpa Alekar; Noemi Mergen; Hermann Einsele; Dieter Hoelzer; Ralf C. Bargou

PURPOSE Patients with relapsed or refractory acute lymphoblastic leukemia (ALL) have a dismal prognosis. CD19 is homogenously expressed in B-precursor ALL and can be targeted by the investigational bispecific T cell-engager antibody blinatumomab. A phase II trial was performed to determine clinical activity in this patient cohort. PATIENTS AND METHODS Thirty-six patients with relapsed or refractory B-precursor ALL were treated with blinatumomab in cycles of 4-week continuous infusion followed by a 2-week treatment-free interval in a single-arm study with a dose-finding stage and an extension stage. The primary end point was complete remission (CR) or CR with partial hematologic recovery (CRh). Major secondary end points included minimal residual disease (MRD) response, rate of allogeneic hematopoietic stem-cell transplantation (HSCT) realization, relapse-free survival (RFS), overall survival (OS), and incidence of adverse events (AEs). RESULTS Median age was 32 years (range, 18 to 77 years). Twenty-five patients (69%) achieved a CR or CRh, with 88% of the responders achieving an MRD response. Median OS was 9.8 months (95% CI, 8.5 to 14.9), and median RFS was 7.6 months (95% CI, 4.5 to 9.5). Thirteen responders (52%) underwent HSCT after achieving a CR or CRh. The most frequent AE during treatment was pyrexia (grade 1 or 2, 75%; grade 3, 6%). In six patients with nervous system or psychiatric disorder AEs and in two patients with cytokine release syndrome, treatment had to be interrupted or discontinued. These medical events were resolved clinically. CONCLUSION The data support further investigation of blinatumomab for the treatment of adult patients with relapsed or refractory ALL in a larger confirmatory study.


Blood | 2012

Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell–engaging CD19/CD3-bispecific BiTE antibody blinatumomab

Matthias Klinger; Christian Brandl; Gerhard Zugmaier; Youssef Hijazi; Ralf C. Bargou; Max S. Topp; Nicola Gökbuget; Svenja Neumann; Mariele Goebeler; Andreas Viardot; Matthias Stelljes; Monika Brüggemann; Dieter Hoelzer; Evelyn Degenhard; Dirk Nagorsen; Patrick A. Baeuerle; Andreas Wolf; Peter Kufer

T cell-engaging CD19/CD3-bispecific BiTE Ab blinatumomab has shown an 80% complete molecular response rate and prolonged leukemia-free survival in patients with minimal residual B-lineage acute lymphoblastic leukemia (MRD(+) B-ALL). Here, we report that lymphocytes in all patients of a phase 2 study responded to continuous infusion of blinatumomab in a strikingly similar fashion. After start of infusion, B-cell counts dropped to < 1 B cell/μL within an average of 2 days and remained essentially undetectable for the entire treatment period. By contrast, T-cell counts in all patients declined to a nadir within < 1 day and recovered to baseline within a few days. T cells then expanded and on average more than doubled over baseline within 2-3 weeks under continued infusion of blinatumomab. A significant percentage of reappearing CD8(+) and CD4(+) T cells newly expressed activation marker CD69. Shortly after start of infusion, a transient release of cytokines dominated by IL-10, IL-6, and IFN-γ was observed, which no longer occurred on start of a second treatment cycle. The response of lymphocytes in leukemic patients to continuous infusion of blinatumomab helps to better understand the mode of action of this and other globally T cell-engaging Abs. The trial is registered with www.clinicaltrials.gov identifier NCT00560794.


International Journal of Cancer | 2002

Extremely potent, rapid and costimulation-independent cytotoxic T-cell response against lymphoma cells catalyzed by a single-chain bispecific antibody.

Torsten Dreier; Grit Lorenczewski; Christian Brandl; Patrick Hoffmann; Uwe Syring; Frank Hanakam; Peter Kufer; Gert Riethmüller; Ralf C. Bargou; Patrick A. Baeuerle

A recent study reported on an anti‐CD19/anti‐CD3 single‐chain bispecific antibody (bscCD19xCD3) exhibiting high activity against human B lymphoma cell lines (Löffler et al., Blood 2000;95:2098–103). In the present study, we have explored in detail the in vitro efficacy, T‐cell donor variability, binding characteristics, specificity, kinetics and interleukin‐2 (IL‐2) dependence of bscCD19xCD3. We found that a majority of human donor T cells tested (n = 86) gave half‐maximal B‐lymphoma cell lysis (ED50) within a range of 10–50 pg/ml bscCD19xCD3, corresponding to sub‐picomolar concentrations of the bispecific antibody. Under identical experimental conditions, the anti‐CD20 monoclonal antibody rituximab had an at least 100,000‐fold lower in vitro efficacy. The extreme potency of bscCD19xCD3 was in sharp contrast to the relatively low affinity of the anti‐CD3 and anti‐CD19 single‐chain Fv portions in KD ranges of 10−7 and 10−9 M, respectively. Cell lysis by bscCD19xCD3 was predominantly mediated by the population of CD8/CD45RO‐positive T cells. Both immortalized CD4‐ and CD8‐positive human T‐cell clones were highly active effector cells as well. Cell lysis by bscCD19xCD3 was rapid and specific. The respective parental monoclonal antibodies inhibited cell lysis and CD19‐negative cells were not harmed by T cells in the presence of high amounts of bscCD19xCD3. The potent T‐cell stimulus IL‐2 could not markedly augment the activity of bscCD19xCD3‐stimulated T cells. In conclusion, bscCD19xCD3 could redirect unstimulated cytotoxic T cells against CD19‐positive cells in an unexpectedly potent, rapid and specific fashion.


International Journal of Cancer | 2005

Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-/CD3-bispecific single-chain antibody construct.

Patrick Hoffmann; Robert Hofmeister; Klaus Brischwein; Christian Brandl; Sandrine Crommer; Ralf C. Bargou; Christian Itin; Nadja Prang; Patrick A. Baeuerle

Certain bispecific antibodies exhibit an extraordinary potency and efficacy for target cell lysis by eliciting a polyclonal T‐cell response. One example is a CD19‐/CD3‐bispecific single‐chain antibody construct (bscCD19xCD3), which at femtomolar concentrations can redirect cytotoxic T cells to eliminate human B lymphocytes, B lymphoma cell lines and patient‐derived malignant B cells. Here we have further explored the basis for this high potency. Using video‐assisted microscopy, bscCD19xCD3 was found to alter the motility and activity of T cells from a scanning to a killing mode. Individual T cells could eliminate multiple target cells within a 9 hr time period, resulting in nuclear fragmentation and membrane blebbing of target cells. Complete target cell elimination was observed within 24 hr at effector‐to‐target cell ratios as low as 1:5. Under optimal conditions, cell killing started within minutes after addition of bscCD19xCD3, suggesting that the rate of serial killing was mostly determined by T‐cell movement and target cell scanning and lysis. At all times, T cells remained highly motile, and no clusters of T and target cells were induced by the bispecific antibody. Bystanding target‐negative cells were not detectably affected. Repeated target cell lysis by bscCD19xCD3‐activated T cells increased the proportion of CD19/CD3 double‐positive T cells, which was most likely a consequence of transfer of CD19 from B to T cells during cytolytic synapse formation. To our knowledge, this is the first study showing that a bispecific antibody can sustain multiple rounds of target cell lysis by T cells.


Pharmacology & Therapeutics | 2012

Blinatumomab: A historical perspective

Dirk Nagorsen; Peter Kufer; Patrick A. Baeuerle; Ralf C. Bargou

For decades, chemotherapy has been the backbone for the treatment of patients with B cell malignancies. Depending on the individual disease, monoclonal antibodies, irradiation and/or hematopoietic stem cell transplantation are added. However, the current standard of care--particularly for patients with relapsed disease--is often not sufficient to achieve durable remissions. A highly promising new drug candidate in late-stage clinical development for treatment of B cell malignancies is blinatumomab (MT103 or AMG 103). This bispecific antibody construct has dual specificity for CD19 and CD3 and belongs to the class of bispecific T cell engager (BiTE®) antibodies, which can potentially engage all cytotoxic T cells of a patient for redirected lysis of tumor cells. Here, we review how blinatumomab has so far been pre-clinically and clinically developed for the treatment of patients with non-Hodgkins lymphoma and acute lymphoblastic leukemia.


Journal of Immunology | 2003

T Cell Costimulus-Independent and Very Efficacious Inhibition of Tumor Growth in Mice Bearing Subcutaneous or Leukemic Human B Cell Lymphoma Xenografts by a CD19-/CD3- Bispecific Single-Chain Antibody Construct

Torsten Dreier; Patrick A. Baeuerle; Iduna Fichtner; Michael Grün; Bernd Schlereth; Grit Lorenczewski; Peter Kufer; Ralf Lutterbüse; Gert Riethmüller; Per Gjorstrup; Ralf C. Bargou

We have recently demonstrated that a recombinant single-chain bispecific Ab construct, bscCD19xCD3, in vitro induces rapid B lymphoma-directed cytotoxicity at picomolar concentrations with unstimulated peripheral T cells. In this study, we show that treatment of nonobese diabetic SCID mice with submicrogram doses of bscCD19xCD3 could prevent growth of s.c. human B lymphoma xenografts and essentially cured animals when given at an early tumor stage. The effect was dose dependent, dependent on E:T ratio and the time between tumor inoculation and administration of bscCD19xCD3. No therapeutic effect was seen in the presence of human lymphocytes alone, a vehicle control, or with a bispecific single-chain construct of identical T cell-binding activity but different target specificity. In a leukemic nonobese diabetic SCID mouse model, treatment with bscCD19xCD3 prolonged survival of mice in a dose-dependent fashion. The human lymphocytes used as effector cells in both animal models did not express detectable T cell activation markers at the time of coinoculation with tumor cells. The bispecific Ab therefore showed an in vivo activity comparable to that observed in cell culture with respect to high potency and T cell costimulus independence. These properties make bscCD19xCD3 superior to previously investigated CD19 bispecific Ab-based therapies.


Blood | 2009

Lenalidomide, adriamycin, and dexamethasone (RAD) in patients with relapsed and refractory multiple myeloma: a report from the German Myeloma Study Group DSMM (Deutsche Studiengruppe Multiples Myelom).

Stefan Knop; Christian Gerecke; Peter Liebisch; Max S. Topp; Uwe Platzbecker; Orhan Sezer; Christina Vollmuth; Karina Falk; Axel Glasmacher; Uwe Maeder; Hermann Einsele; Ralf C. Bargou

We conducted a phase 1/2 trial combining lenalidomide (R) with adriamycin (A) and dexamethasone (D) for relapsed and relapsed-refractory myeloma to determine tolerability and efficacy of this novel regimen, RAD, delivered for six 28-day cycles. A total of 69 intensively pretreated patients with a median age of 65 years (range, 46-77 years) were enrolled. Using pegfilgrastim (G), the maximum tolerated dose (MTD) was formally not reached at the highest dose level (R, 25 mg on days 1-21; A, 9 mg/m(2) intravenously on days 1-4; and D, 40 mg on days 1-4 and 17-20; dose level 5+G), which was then used to determine efficacy. Grades 3/4 neutropenia and thrombocytopenia were seen in 48% and 38% of patients, respectively. Thromboembolic events occurred in 4.5% and severe infections in 10.5% of patients. On an intent-to treat analysis, overall response rate (ORR) was 73% for the whole study and 77% including 74% complete response (CR) plus very good partial response (VGPR) for dose level 5+G. Response rates and progression-free survival did not differ between relapsed and relapsed-refractory patients. Deletion of chromosome 17p and elevated beta(2)-microglobulin were associated with significantly inferior response and shortened time to progression. In conclusion, RAD induces substantial and durable remission with an acceptable toxicity profile in patients with relapsed and relapsed-refractory myeloma. This trial was registered at www.ClinicalTrials.gov as no. NCT00306813.


Cancer Research | 2005

YB-1 Provokes Breast Cancer through the Induction of Chromosomal Instability That Emerges from Mitotic Failure and Centrosome Amplification

Stephan Bergmann; Brigitte Royer-Pokora; Ellen Fietze; Karsten Jürchott; Barbara Hildebrandt; Detlef Trost; Frauke Leenders; Jenny-Chang Claude; Franz Theuring; Ralf C. Bargou; Manfred Dietel; Hans-Dieter Royer

YB-1 protein levels are elevated in most human breast cancers, and high YB-1 levels have been correlated with drug resistance and poor clinical outcome. YB-1 is a stress-responsive, cell cycle-regulated transcription factor with additional functions in RNA metabolism and translation. In this study, we show in a novel transgenic mouse model that human hemagglutinin-tagged YB-1 provokes remarkably diverse breast carcinomas through the induction of genetic instability that emerges from mitotic failure and centrosome amplification. The increase of centrosome numbers proceeds during breast cancer development and explanted tumor cell cultures show the phenotype of ongoing numerical chromosomal instability. These data illustrate a mechanism that might contribute to human breast cancer development.


International Journal of Cancer | 1996

Induction of the death‐promoting gene bax‐α sensitizes cultured breast‐cancer cells to drug‐induced apoptosis

Christian Wagener; Ralf C. Bargou; Peter T. Daniel; Kurt Bommert; Markus Y. Mapara; Hans Dieter Royer; Bernd Dörken

Resistance to apoptosis plays an important role in malignancies that are refractory to chemotherapy treatment. Recently we have shown that the expression of bax‐α, a death‐promoting member of the bcl‐2 family, is down‐regulated in breast cancer and have provided evidence that low bax expression might contribute to the pathogenesis of breast cancer. In this study we were able to demonstrate the role of this gene in chemotherapy‐induced apoptosis. We transfected bax‐α into the breast‐cancer cell lines R30C and MCF‐7 under the control of an inducible tetracycline‐dependent expression system. Induction of bax‐α expression did not affect viability by itself but strongly increased chemosensitivity to epirubicin. We were able to demonstrate that this sensitization is due to apoptosis. These data might explain the recently published observation that reduced expression of bax is associated with poor response rates to chemotherapy in breast cancer.

Collaboration


Dive into the Ralf C. Bargou's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thorsten Stühmer

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Max S. Topp

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar

Stefan Knop

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt Bommert

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge