Network


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

Hotspot


Dive into the research topics where Anja Feldmann is active.

Publication


Featured researches published by Anja Feldmann.


BioMed Research International | 2010

Chimeric Antigen Receptor-Engineered T Cells for Immunotherapy of Cancer

Marc Cartellieri; Michael H. Bachmann; Anja Feldmann; Claudia C. Bippes; Slava Stamova; Rebekka Wehner; Achim Temme; Marc Schmitz

CD4+ and CD8+ T lymphocytes are powerful components of adaptive immunity, which essentially contribute to the elimination of tumors. Due to their cytotoxic capacity, T cells emerged as attractive candidates for specific immunotherapy of cancer. A promising approach is the genetic modification of T cells with chimeric antigen receptors (CARs). First generation CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen and a lymphocyte activating signaling chain. The CAR-mediated recognition induces cytokine production and tumor-directed cytotoxicity of T cells. Second and third generation CARs include signal sequences from various costimulatory molecules resulting in enhanced T-cell persistence and sustained antitumor reaction. Clinical trials revealed that the adoptive transfer of T cells engineered with first generation CARs represents a feasible concept for the induction of clinical responses in some tumor patients. However, further improvement is required, which may be achieved by second or third generation CAR-engrafted T cells.


Blood Cancer Journal | 2014

Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia

Armin Ehninger; Michael S. Kramer; Christoph Röllig; Christian Thiede; Martin Bornhäuser; M von Bonin; Martin Wermke; Anja Feldmann; Michael H. Bachmann; Gerhard Ehninger; Uta Oelschlägel

Owing to the more recent positive results with the anti-CD33 immunotoxin gemtuzumab ozogamicin, therapy against acute myeloid leukemias (AMLs) targeting CD33 holds many promises. Here, CD33 and CD123 expression on AML blasts was studied by flow cytometry in a cohort of 319 patients with detailed information on French–American–British/World Health Organization (FAB/WHO) classification, cytogenetics and molecular aberrations. AMLs of 87.8% express CD33 and would therefore be targetable with anti-CD33 therapies. Additionally, 9.4% of AMLs express CD123 without concomitant CD33 expression. Thus, nearly all AMLs could be either targeted via CD33 or CD123. Simultaneous presence of both antigens was observed in 69.5% of patients. Most importantly, even AMLs with adverse cytogenetics express CD33 and CD123 levels comparable to those with favorable and intermediate subtypes. Some patient groups with unfavorable alterations, such as FMS-related tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutations, high FLT3-ITD mutant/wild-type ratios and monosomy 5 are even characterized by high expression of CD33 and CD123. In addition, blasts of patients with mutant nucleophosmin (NPM1) revealed significantly higher CD33 and CD123 expression pointing toward the possibility of minimal residual disease-guided interventions in mutated NPM1-positive AMLs. These results stimulate the development of novel concepts to redirect immune effector cells toward CD33- and CD123-expressing blasts using bi-specific antibodies or engineered T cells expressing chimeric antigen receptors.


Leukemia | 2013

Redirection of T cells with a first fully humanized bispecific CD33–CD3 antibody efficiently eliminates AML blasts without harming hematopoietic stem cells

Claudia Arndt; von Bonin M; Marc Cartellieri; Anja Feldmann; Stefanie Koristka; Irene Michalk; Slava Stamova; Martin Bornhäuser; Marc Schmitz; Gerhard Ehninger; Michael H. Bachmann

Redirection of T cells with a first fully humanized bispecific CD33–CD3 antibody efficiently eliminates AML blasts without harming hematopoietic stem cells


Journal of Immunology | 2012

Novel Humanized and Highly Efficient Bispecific Antibodies Mediate Killing of Prostate Stem Cell Antigen-Expressing Tumor Cells by CD8+ and CD4+ T Cells

Anja Feldmann; Claudia Arndt; Katrin Töpfer; Slava Stamova; Franziska Krone; Marc Cartellieri; Stefanie Koristka; Irene Michalk; Dirk Lindemann; Marc Schmitz; Achim Temme; Martin Bornhäuser; Gerhard Ehninger; Michael Bachmann

Prostate cancer is the most common noncutaneous malignancy in men. The prostate stem cell Ag (PSCA) is a promising target for immunotherapy of advanced disease. Based on a novel mAb directed to PSCA, we established and compared a series of murine and humanized anti-CD3–anti-PSCA single-chain bispecific Abs. Their capability to redirect T cells for killing of tumor cells was analyzed. During these studies, we identified a novel bispecific humanized Ab that efficiently retargets T cells to tumor cells in a strictly Ag-dependent manner and at femtomolar concentrations. T cell activation, cytokine release, and lysis of target cells depend on a cross-linkage of redirected T cells with tumor cells, whereas binding of the anti-CD3 domain alone does not lead to an activation or cytokine release. Interestingly, both CD8+ and CD4+ T cells are activated in parallel and can efficiently mediate the lysis of tumor cells. However, the onset of killing via CD4+ T cells is delayed. Furthermore, redirecting T cells via the novel humanized bispecific Abs results in a delay of tumor growth in xenografted nude mice.


The Prostate | 2011

Retargeting of T cells to prostate stem cell antigen expressing tumor cells: comparison of different antibody formats.

Anja Feldmann; Slava Stamova; Claudia C. Bippes; Holger Bartsch; Rebekka Wehner; Marc Schmitz; Achim Temme; Marc Cartellieri; Michael Bachmann

Prostate cancer (PCa) is the most common malignant disease in men. Novel treatment options are needed for patients after development of metastatic, hormone‐refractory disease or for those who have failed a local treatment. The prostate stem cell antigen (PSCA) is expressed in >80% of primary PCa samples and bone metastases. Its expression is increased both in androgen‐dependent and independent prostate tumors, particularly in carcinomas of high stages and Gleason scores. Therefore, PSCA is an attractive target for immunotherapy of PCa by retargeting of T cells to tumor cells.


Journal of Immunology | 2012

Retargeting of human regulatory T cells by single-chain bispecific antibodies

Stefanie Koristka; Marc Cartellieri; Anke Theil; Anja Feldmann; Claudia Arndt; Slava Stamova; Irene Michalk; Katrin Töpfer; Achim Temme; Karsten Kretschmer; Martin Bornhäuser; Gerhard Ehninger; Marc Schmitz; Michael Bachmann

Bispecific Abs hold great potential for immunotherapy of malignant diseases. Because the first components of this new drug class are now entering clinical trials, all aspects of their mode of action should be well understood. Several studies proved that CD8+ and CD4+ effector T cells can be successfully redirected and activated against tumor cells by bispecific Abs both in vitro and in vivo. To our knowledge, this study provides the first evidence that bispecific Abs can also redirect and activate regulatory T cells against a surface Ag, independently of their TCR specificity. After cross-linking, via a bispecific Ab, redirected regulatory T cells upregulate the activation markers CD69 and CD25, as well as regulatory T cell-associated markers, like CTLA-4 and FOXP3. The activated regulatory T cells secrete the immunosuppressive cytokine IL-10, but, in contrast to CD8+ and CD4+ effector T cells, almost no inflammatory cytokines. In addition, the redirected regulatory T cells are able to suppress effector functions of activated autologous CD4+ T cells both in vitro and in vivo. Therefore, the potential risk for activation of regulatory T cells should be taken into consideration when bispecific Abs are applied for the treatment of malignant diseases. In contrast, an Ag/tissue-specific redirection of regulatory T cells with bispecific Abs holds great potential for the treatment of autoimmune diseases and graft rejection.


Leukemia | 2014

Costimulation improves the killing capability of T cells redirected to tumor cells expressing low levels of CD33: description of a novel modular targeting system

Claudia Arndt; Anja Feldmann; M von Bonin; Marc Cartellieri; E-M Ewen; Stefanie Koristka; Irene Michalk; Slava Stamova; Nicole Berndt; A Gocht; Martin Bornhäuser; Gerhard Ehninger; Marc Schmitz; Michael H. Bachmann

Owing to their clinical success, there is growing interest in novel bispecific antibodies (bsAbs) for retargeting of T cells to tumor cells including for the treatment of acute myeloid leukemia (AML). One potential target for retargeting of T cells to AML blasts is the surface molecule CD33. Here we describe a novel modular targeting platform that consists of a universal effector module (EM) and individual target modules (TMs). Both modules can form an immune complex via a peptide epitope. The resulting targeting complex can functionally replace a conventional bsAb. By fusion of a costimulatory domain (for example, the extracellular CD137 ligand domain) to the TM, the targeting complex can even provide a costimulatory signal to the redirected T cells at their side of interaction with the tumor cell. Furthermore, we observed that an efficient killing of tumor cells expressing low levels of the tumor target CD33 becomes critical at low effector-to-target cell ratios but can be improved by costimulation via CD137 using our novel targeting system.


Leukemia | 2011

Simultaneous engagement of the activatory receptors NKG2D and CD3 for retargeting of effector cells to CD33-positive malignant cells

Slava Stamova; Marc Cartellieri; Anja Feldmann; Claudia C. Bippes; Holger Bartsch; Rebekka Wehner; Marc Schmitz; M von Bonin; Martin Bornhäuser; Gerhard Ehninger; E P Rieber; Michael H. Bachmann

Simultaneous engagement of the activatory receptors NKG2D and CD3 for retargeting of effector cells to CD33-positive malignant cells


Molecular Immunology | 2011

Unexpected recombinations in single chain bispecific anti-CD3-anti-CD33 antibodies can be avoided by a novel linker module.

Slava Stamova; Marc Cartellieri; Anja Feldmann; Claudia Arndt; Stefanie Koristka; Holger Bartsch; Claudia C. Bippes; Rebekka Wehner; Marc Schmitz; Malte von Bonin; Martin Bornhäuser; Gerhard Ehninger; Michael Bachmann

CD33 is an attractive immunotarget on the surface of tumor cells from patients with acute myeloid leukemia (AML). In a first attempt for immunotargeting of AML blasts we constructed two bispecific antibodies in the single chain bispecific diabody (scBsDb) format by fusing the variable domains of monoclonal antibodies directed against CD3 and CD33. Unfortunately, protein expression of both scBsDbs resulted in varying mixtures of fragmented and full length proteins. As the non-functional fragments competed with the functional full length antibodies we tried to understand the reason for the fragmentation. We found that the anti-CD3 and anti-CD33 antibody genes show striking sequence homologies: during B cell development the same V(h) J558 heavy and V(l) kk4 light chain genes were selected. Moreover, the closely related D genes DSP2 (9 and 11) were combined with the same JH4 gene. And finally, during VJ recombination of the light chain the same JK5 element was selected. These homologies between the two monoclonal antibodies were the reason for recombinations in the cell lines generated for expression of the scBsDbs. Finally, we solved this problem by (i) rearranging the order of the heavy and light chains of the anti-CD3 and anti-CD33 domains, and (ii) a replacement of one of the commonly used glycine serine linkers with a novel linker domain. The resulting bispecific antibody in a single chain bispecific tandem format (scBsTaFv) was stable and capable of redirecting T cells to CD33-positive tumor cells including AML blasts of patients.


PLOS ONE | 2014

A Novel Ex Vivo Isolation and Expansion Procedure for Chimeric Antigen Receptor Engrafted Human T Cells

Marc Cartellieri; Stefanie Koristka; Claudia Arndt; Anja Feldmann; Slava Stamova; Malte von Bonin; Katrin Töpfer; Thomas Krüger; Mathias Geib; Irene Michalk; Achim Temme; Martin Bornhäuser; Dirk Lindemann; Gerhard Ehninger; Michael Bachmann

Genetically engineered T lymphocytes are a promising option for cancer therapy. Prior to adoptive transfer they have to be expanded in vitro to reach therapeutically sufficient numbers. So far, no universal method exists for selective in vitro expansion of engineered T lymphocytes. In order to overcome this problem and for proof of concept we incorporated a novel unique peptide sequence of ten amino acids as epitope (E-Tag) into the binding domains of two novel chimeric antigen receptors (ECARs) directed against either prostate stem cell antigen (PSCA) for the treatment of prostate cancer (PCa) or CD33 for the treatment of acute myeloide leukemia (AML). The epitope tag then was utilized for expanding ECAR engrafted T cells by triggering the modified T cells via a monoclonal antibody directed against the E-Tag (Emab). Moreover, the E-Tag served as an efficient selection epitope for immunomagnetic isolation of modified T cells to high purity. ECAR engrafted T cells were fully functional and mediated profound anti-tumor effects in the respective models of PCa or AML both in vitro and in vivo. The method can be integrated straightforward into clinical protocols to improve therapeutic efficiency of tumor treatment with CAR modified T lymphocytes.

Collaboration


Dive into the Anja Feldmann's collaboration.

Top Co-Authors

Avatar

Claudia Arndt

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Marc Cartellieri

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Gerhard Ehninger

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Stefanie Koristka

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Michael Bachmann

Helmholtz-Zentrum Dresden-Rossendorf

View shared research outputs
Top Co-Authors

Avatar

Martin Bornhäuser

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Armin Ehninger

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Marc Schmitz

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Slava Stamova

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Irene Michalk

Dresden University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge