Network


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

Hotspot


Dive into the research topics where Matthias Klinger is active.

Publication


Featured researches published by Matthias Klinger.


Science | 2008

Tumor Regression in Cancer Patients by Very Low Doses of a T Cell–Engaging Antibody

Ralf Bargou; Eugen Leo; Gerhard Zugmaier; Matthias Klinger; Mariele Goebeler; Stefan Knop; Richard Noppeney; Andreas Viardot; Georg Hess; Martin Schuler; Hermann Einsele; Christian Brandl; A. Wolf; Petra Kirchinger; Petra Klappers; Margit Schmidt; Gert Riethmüller; Carsten Reinhardt; Patrick A. Baeuerle; Peter Kufer

Previous attempts have shown the potential of T cells in immunotherapy of cancer. Here, we report on the clinical activity of a bispecific antibody construct called blinatumomab, which has the potential to engage all cytotoxic T cells in patients for lysis of cancer cells. Doses as low as 0.005 milligrams per square meter per day in non–Hodgkins lymphoma patients led to an elimination of target cells in blood. Partial and complete tumor regressions were first observed at a dose level of 0.015 milligrams, and all seven patients treated at a dose level of 0.06 milligrams experienced a tumor regression. Blinatumomab also led to clearance of tumor cells from bone marrow and liver. T cell–engaging antibodies appear to have therapeutic potential for the treatment of malignant diseases.


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.


Blood | 2012

Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL

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

Persistence or recurrence of minimal residual disease (MRD) after chemotherapy results in clinical relapse in patients with acute lymphoblastic leukemia (ALL). In a phase 2 trial of B-lineage ALL patients with persistent or relapsed MRD, a T cell-engaging bispecific Ab construct induced an 80% MRD response rate. In the present study, we show that after a median follow-up of 33 months, the hematologic relapse-free survival of the entire evaluable study cohort of 20 patients was 61% (Kaplan-Meier estimate). The hema-tologic relapse-free survival rate of a subgroup of 9 patients who received allogeneic hematopoietic stem cell transplantation after blinatumomab treatment was 65% (Kaplan-Meier estimate). Of the subgroup of 6 Philadelphia chromosome-negative MRD responders with no further therapy after blinatumomab, 4 are in ongoing hematologic and molecular remission. We conclude that blinatumomab can induce long-lasting complete remission in B-lineage ALL patients with persistent or recurrent MRD. The original study and this follow-up study are registered at www.clinicaltrials.gov as NCT00198991 and NCT00198978, respectively.


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.


Journal of Immunotherapy | 2007

Strictly target cell-dependent activation of T cells by bispecific single-chain antibody constructs of the BiTE class.

Klaus Brischwein; Larissa Parr; Stefan Pflanz; Jörg Volkland; John S. Lumsden; Matthias Klinger; Mathias Locher; Scott A. Hammond; Peter A. Kiener; Peter Kufer; Bernd Schlereth; Patrick A. Baeuerle

Bispecific antibodies have been extensively studied in vitro and in vivo for their use in redirected tumor cell lysis. A particular challenge of bispecific antibody constructs that recognize the invariant CD3 signaling complex is a controlled polyclonal activation of T cells that, ideally, is exquisitely dependent on the presence of target cells. Otherwise, overt production of inflammatory cytokines and secondary reactions may occur as side effects, as can be observed with constitutively T-cell activating monoclonal antibodies to CD3 or CD28, and with bispecific antibodies bearing Fcγ portions. Here we analyzed 2 distinct bispecific single-chain antibody constructs of the BiTE class, called MT110 and MT103 (or MEDI-538), for conditional T-cell activation. In the presence of target-expressing cell lines, low picomolar concentrations of the BiTE molecules were sufficient to stimulate a high percentage of peripheral human T cells to express cytokines and surface activation markers, enter into cell cycle, and induce redirected lysis of target cells. However, in the absence of target cells, the 2 BiTE molecules even at high concentrations did not detectably activate T cells. Our data show that T cell activation by monomeric forms of MT110 and MT103 is highly conditional in that it is strictly dependent on the presence of cells expressing the proper target antigen. BiTE molecules therefore qualify for a highly controlled polyclonal T-cell therapy of cancer.


Journal of Clinical Oncology | 2016

Bispecific T-Cell Engager (BiTE) Antibody Construct Blinatumomab for the Treatment of Patients With Relapsed/Refractory Non-Hodgkin Lymphoma: Final Results From a Phase I Study

Maria-Elisabeth Goebeler; Stefan Knop; Andreas Viardot; Peter Kufer; Max S. Topp; Hermann Einsele; Richard Noppeney; Georg Hess; Stefan Kallert; Andreas Mackensen; Kathrin Rupertus; Lothar Kanz; Martin Libicher; Dirk Nagorsen; Gerhard Zugmaier; Matthias Klinger; A. Wolf; Brigitte Dorsch; Beate D. Quednau; Margit Schmidt; Jürgen Scheele; Patrick A. Baeuerle; Eugen Leo; Ralf C. Bargou

PURPOSE Blinatumomab is a CD19/CD3 BiTE (bispecific T-cell engager) antibody construct for the treatment of Philadelphia chromosome-negative acute B-lymphoblastic leukemia. We evaluated blinatumomab in relapsed/refractory B-cell non-Hodgkin lymphoma (NHL). PATIENTS AND METHODS This 3 + 3 design, phase I dose-escalation study determined adverse events and the maximum tolerated dose (MTD) of continuous intravenous infusion blinatumomab in patients with relapsed/refractory NHL. Blinatumomab was administered over 4 or 8 weeks at seven different dose levels (0.5 to 90 μg/m(2)/day). End points were incidence of adverse events, pharmacokinetics, pharmacodynamics, and overall response rate. RESULTS Between 2004 and 2011, 76 heavily pretreated patients with relapsed/refractory NHL, who included 14 with diffuse large B-cell lymphoma, were enrolled; 42 received treatment in the formal dose-escalation phase. Neurologic events were dose limiting, and 60 μg/m(2)/day was established as the MTD. Thirty-four additional patients were recruited to evaluate antilymphoma activity and strategies for mitigating neurologic events at a prespecified MTD. Stepwise dosing (5 to 60 μg/m(2)/day) plus pentosan polysulfate SP54 (n = 3) resulted in no treatment discontinuations; single-step (n = 5) and double-step (n = 24) dosing entailed two and seven treatment discontinuations due to neurologic events, respectively. Grade 3 neurologic events occurred in 22% of patients (no grade 4/5). Among patients treated at 60 μg/m(2)/day (target dose; n = 35), the overall response rate was 69% across NHL subtypes and 55% for diffuse large B-cell lymphoma (n = 11); median response duration was 404 days (95% CI, 207 to 1,129 days). CONCLUSION In this phase I study of relapsed/refractory NHL, continuous infusion with CD19-targeted immunotherapy blinatumomab at various doses and schedules was feasible, with an MTD of 60 μg/m(2)/day. Single-agent blinatumomab showed antilymphoma activity.


Blood | 2015

Long-term survival and T-cell kinetics in relapsed/refractory ALL patients who achieved MRD response after blinatumomab treatment

Gerhard Zugmaier; Nicola Gökbuget; Matthias Klinger; Andreas Viardot; Matthias Stelljes; Svenja Neumann; Heinz-A. Horst; Reinhard Marks; Christoph Faul; Helmut Diedrich; Albrecht Reichle; Monika Brüggemann; Chris Holland; Margit Schmidt; Hermann Einsele; Ralf C. Bargou; Max S. Topp

This long-term follow-up analysis evaluated overall survival (OS) and relapse-free survival (RFS) in a phase 2 study of the bispecific T-cell engager antibody construct blinatumomab in 36 adults with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL). In the primary analysis, 25 (69%) patients with relapsed/refractory ALL achieved complete remission with full (CR) or partial (CRh) hematologic recovery of peripheral blood counts within the first 2 cycles. Twenty-five patients (69%) had a minimal residual disease (MRD) response (<10(-4) blasts), including 22 CR/CRh responders, 2 patients with hypocellular bone marrow, and 1 patient with normocellular bone marrow but low peripheral counts. Ten of the 36 patients (28%) were long-term survivors (OS ≥30 months). Median OS was 13.0 months (median follow-up, 32.6 months). MRD response was associated with significantly longer OS (Mantel-Byar P = .009). All 10 long-term survivors had an MRD response. Median RFS was 8.8 months (median follow-up, 28.9 months). A plateau for RFS was reached after ∼18 months. Six of the 10 long-term survivors remained relapse-free, including 4 who received allogeneic stem cell transplantation (allo-SCT) as consolidation for blinatumomab and 2 who received 3 additional cycles of blinatumomab instead of allo-SCT. Three long-term survivors had neurologic events or cytokine release syndrome, resulting in temporary blinatumomab discontinuation; all restarted blinatumomab successfully. Long-term survivors had more pronounced T-cell expansion than patients with OS <30 months.


Immunological Reviews | 2016

Harnessing T cells to fight cancer with BiTE® antibody constructs--past developments and future directions.

Matthias Klinger; Jonathan Benjamin; Roman Kischel; Sabine Stienen; Gerhard Zugmaier

Bispecific T‐cell engager (BiTE®) antibody constructs represent a novel immunotherapy that bridges cytotoxic T cells to tumor cells, thereby inducing target cell‐dependent polyclonal T‐cell activation and proliferation, and leading to apoptosis of bound tumor cells. Anti‐CD19 BiTE® blinatumomab has demonstrated clinical activity in Philadelphia chromosome (Ph)‐negative relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL) eventually resulting in conditional approval by the U.S. Food and Drug Administration in 2014. This drug is currently further developed in pediatric and Ph+ r/r, as well as in minimal residual disease‐positive ALL, and might also offer clinical benefit for patients with non‐Hodgkins lymphoma, especially for those with aggressive forms like diffuse large B‐cell lymphoma. Another BiTE® antibody construct in hemato‐oncology designated AMG 330 targets CD33 on acute myeloid leukemia blast cells. After showing promising ex vivo activity, this drug candidate has recently entered phase 1 clinical development, and has further indicated potential for combination with checkpoint inhibitors. In solid tumor indications, three BiTE® antibody constructs have been tested in phase 1 studies so far: anti‐EpCAM BiTE® AMG 110, anti‐CEA BiTE® MEDI‐565/AMG 211, and anti‐PSMA BiTE® BAY2010112/AMG 212. Pertinent questions comprise how to maximize BiTE® penetration and T‐cell infiltration of the tumor while simultaneously minimizing any adverse events, which is currently explored by a continuous intravenous infusion approach. Thus, BiTE® antibody constructs will hopefully provide new treatment options for patients in several indications with high unmet medical need.


Molecular Immunology | 2015

Clinical overview of anti-CD19 BiTE(®) and ex vivo data from anti-CD33 BiTE(®) as examples for retargeting T cells in hematologic malignancies.

Gerhard Zugmaier; Matthias Klinger; Margit Schmidt; Marion Subklewe

Blinatumomab, a bispecific antibody construct targeting CD19, is the most advanced member of bispecific T-cell engager (BiTE(®)) molecules. The clinical development program includes B-precursor acute lymphoblastic leukemia (ALL) and B-cell non-Hodgkin lymphoma (NHL). Minimal residual disease (MRD) response in patients with MRD-positive B-precursor ALL has translated into long-term clinical benefits as demonstrated by an estimated relapse-free survival (RFS) of 60% with sustained MRD negativity at a follow-up of 31 months. Remissions induced in pediatric and adult patients with relapsed/refractory B-precursor ALL have allowed for successful allogeneic hematopoietic stem cell transplantation (HSCT) in this setting. Blinatumomab has also induced durable responses in low-grade B-cell NHL. Blinatumomab recently gained approval in the United States by the U.S. Food and Drug Administration for treatment of Philadelphia chromosome-negative B-precursor relapsed/refractory acute lymphoblastic leukemia. AMG 330 is an investigational anti-CD33 BiTE(®) antibody construct. Targeting CD33 ex vivo in primary samples from patients with acute myeloid leukemia (AML) has shown AMG 330-mediated T-cell expansion and T-cell cytotoxicity against AML cells.


Haematologica | 2017

Long-term relapse-free survival in a phase 2 study of blinatumomab for the treatment of patients with minimal residual disease in B-lineage acute lymphoblastic leukemia

Nicola Gökbuget; Gerhard Zugmaier; Matthias Klinger; Peter Kufer; Matthias Stelljes; Andreas Viardot; Heinz A. Horst; Svenja Neumann; Monika Brüggemann; Oliver G. Ottmann; Thomas Burmeister; Dorothea Wessiepe; Max S. Topp; Ralf C. Bargou

In adults with acute lymphoblastic leukemia (ALL), advances in chemotherapy have improved hematologic response rates to greater than 80% and relapses rates to below 50%. Leukemic cells not detected by conventional morphological methods may persist or reappear after chemotherapy as minimal residual

Collaboration


Dive into the Matthias Klinger's collaboration.

Top Co-Authors

Avatar

Peter Kufer

Ludwig Maximilian University of Munich

View shared research outputs
Top Co-Authors

Avatar

Ralf C. Bargou

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
Researchain Logo
Decentralizing Knowledge