Network


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

Hotspot


Dive into the research topics where Marc Cartellieri is active.

Publication


Featured researches published by Marc Cartellieri.


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.


BioMed Research International | 2011

Tumor Evasion from T Cell Surveillance

Katrin Töpfer; Stefanie Kempe; Nadja Müller; Marc Schmitz; Michael H. Bachmann; Marc Cartellieri; Gabriele Schackert; Achim Temme

An intact immune system is essential to prevent the development and progression of neoplastic cells in a process termed immune surveillance. During this process the innate and the adaptive immune systems closely cooperate and especially T cells play an important role to detect and eliminate tumor cells. Due to the mechanism of central tolerance the frequency of T cells displaying appropriate arranged tumor-peptide-specific-T-cell receptors is very low and their activation by professional antigen-presenting cells, such as dendritic cells, is frequently hampered by insufficient costimulation resulting in peripheral tolerance. In addition, inhibitory immune circuits can impair an efficient antitumoral response of reactive T cells. It also has been demonstrated that large tumor burden can promote a state of immunosuppression that in turn can facilitate neoplastic progression. Moreover, tumor cells, which mostly are genetically instable, can gain rescue mechanisms which further impair immune surveillance by T cells. Herein, we summarize the data on how tumor cells evade T-cell immune surveillance with the focus on solid tumors and describe approaches to improve anticancer capacity of T cells.


Neuro-oncology | 2011

RNA interference targeting survivin exerts antitumoral effects in vitro and in established glioma xenografts in vivo

Sandy Hendruschk; Ralf Wiedemuth; Achim Aigner; Katrin Töpfer; Marc Cartellieri; Daniel Martin; Chrysanthy Ikonomidou; Gabriele Schackert; Achim Temme

Malignant glioma represents the most common primary adult brain tumor in Western industrialized countries. Despite aggressive treatment modalities, the median survival duration for patients with glioblastoma multiforme (GBM), the highest grade malignant glioma, has not improved significantly over past decades. One promising approach to deal with GBM is the inactivation of proteins essential for survival or progression of glioma cells by means of RNA interference (RNAi) techniques. A likely candidate for an RNAi therapy of gliomas is the inhibitor of apoptosis protein survivin. Survivin is involved in 2 main cellular processes–cell division and inhibition of apoptosis. We show here that stable RNAi of survivin induced polyploidy, apoptosis, and impaired proliferation of human U343-MG, U373-MG, H4, and U87-MG cells and of primary glioblastoma cells. Proteome profiler arrays using U373-MG cells identified a novel set of differentially expressed genes upon RNAi-mediated survivin knockdown. In particular, the death receptor TRAIL R2/DR5 was strongly upregulated in survivin-depleted glioma cells, inducing an enhanced cytotoxic response of allogeneic human NK cells. Moreover, an experimental in vivo therapy using polyethylenimine (PEI)/siRNA complexes for survivin knockdown efficiently blocked tumor growth of established subcutaneous U373-MG tumors and enhanced survival of NMRInu/nu mice orthopically transplanted with U87-MG cells. We conclude that survivin is functionally relevant in gliomas and that PEI-mediated exogenous delivery of siRNA targeting survivin is a promising strategy for glioblastoma therapy.


Journal of Virology | 2005

Characterization of Prototype Foamy Virus Gag Late Assembly Domain Motifs and Their Role in Particle Egress and Infectivity

Annett Stange; Ingrid Mannigel; Katrin Peters; Martin Heinkelein; Nicole Stanke; Marc Cartellieri; Heinrich G. Göttlinger; Axel Rethwilm; Hanswalter Zentgraf; Dirk Lindemann

ABSTRACT Foamy viruses (FV) are unusual among retroviruses since they require both Gag and Env structural proteins for particle egress. Recently significant progress has been made towards the mechanistic understanding of the viral release process, in particular that of retroviruses, and the viral domains and cellular pathways involved. However little is currently known about domains of FV structural proteins and cellular proteins engaged in this process. By mutational analysis of sequence motifs in prototype FV (PFV) Gag, bearing homology to known late assembly (L) domains, a PSAP motif with L domain function that was functionally interchangeable by heterologous L domains was identified. In contrast the inactivation of a PPPI motif had no significant influence on PFV particle release, although mutant viral particles displayed reduced infectivity. Similarly mutation of an evolutionary conserved YXXL motif revealed no classical L-domain function but resulted in release of noninfectious viruslike particles. Biochemical and electron microscopy analysis demonstrated that these mutant particles incorporated all viral structural proteins but contained aberrantly capsid structures, suggesting a role in capsid assembly for this PFV Gag sequence motif. In line with the mutational analysis, overexpression of dominant negative (DN) mutants and wild-type TSG101 but not the DN mutant of AIP-1/ALIX reduced PFV particle release and infectivity. Furthermore, DN mutants of Vps4A, Vps4B, and CHMP3 inhibited PFV egress and infectivity. Taken together these results demonstrate that PFV, like other viruses, requires components of the vacuolar protein sorting (VPS) machinery for egress and enters the VPS pathway through interaction with TSG101.


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.


Hepatology | 2014

Dual role of B7 costimulation in obesity-related nonalcoholic steatohepatitis and metabolic dysregulation.

Antonios Chatzigeorgiou; Kyoung-Jin Chung; Ruben Garcia-Martin; Vasileia‐Ismini Alexaki; Anne Klotzsche-von Ameln; Julia Phieler; David Sprott; Waldemar Kanczkowski; Theodora Tzanavari; Mohktar Bdeir; Sibylle Bergmann; Marc Cartellieri; Michael H. Bachmann; Polyxeni Nikolakopoulou; Andreas Androutsellis-Theotokis; Gabriele Siegert; Stefan R. Bornstein; Michael H. Muders; Louis Boon; Katia Karalis; Esther Lutgens; Triantafyllos Chavakis

The low‐grade inflammatory state present in obesity contributes to obesity‐related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell‐cell interactions in several immune processes; however, the role of B7 costimulation in obesity‐related liver inflammation is unknown. Here, diet‐induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity‐related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non‐Treg‐lacking environment, we performed antibody (Ab)‐mediated inhibition of B7 molecules in wild‐type mice in DIO. Antibody‐blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT. Conclusion: Our study demonstrates a dual role of B7 costimulation in the course of obesity‐related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity‐related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity‐related metabolic dysregulation and, especially, NASH. (Hepatology 2014;60:1196–1210)

Collaboration


Dive into the Marc Cartellieri's collaboration.

Top Co-Authors

Avatar

Michael Bachmann

Helmholtz-Zentrum Dresden-Rossendorf

View shared research outputs
Top Co-Authors

Avatar

Anja Feldmann

Helmholtz-Zentrum Dresden-Rossendorf

View shared research outputs
Top Co-Authors

Avatar

Gerhard Ehninger

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Claudia Arndt

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

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

Irene Michalk

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Slava Stamova

Dresden University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge