Network


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

Hotspot


Dive into the research topics where Christine E. Engeland is active.

Publication


Featured researches published by Christine E. Engeland.


Molecular Therapy | 2014

CTLA-4 and PD-L1 Checkpoint Blockade Enhances Oncolytic Measles Virus Therapy

Christine E. Engeland; Rūta Veinalde; Sascha Bossow; Diana Lutz; Johanna Kaufmann; Ivan Shevchenko; Viktor Umansky; Dirk M. Nettelbeck; Wilko Weichert; Dirk Jäger; Christof von Kalle; Guy Ungerechts

We hypothesized that the combination of oncolytic virotherapy with immune checkpoint modulators would reduce tumor burden by direct cell lysis and stimulate antitumor immunity. In this study, we have generated attenuated Measles virus (MV) vectors encoding antibodies against CTLA-4 and PD-L1 (MV-aCTLA-4 and MV-aPD-L1). We characterized the vectors in terms of growth kinetics, antibody expression, and cytotoxicity in vitro. Immunotherapeutic effects were assessed in a newly established, fully immunocompetent murine model of malignant melanoma, B16-CD20. Analyses of tumor-infiltrating lymphocytes and restimulation experiments indicated a favorable immune profile after MV-mediated checkpoint modulation. Therapeutic benefits in terms of delayed tumor progression and prolonged median overall survival were observed for animals treated with vectors encoding anti-CTLA-4 and anti-PD-L1, respectively. Combining systemic administration of antibodies with MV treatment also improved therapeutic outcome. In vivo oncolytic efficacy against human tumors was studied in melanoma xenografts. MV-aCTLA-4 and MV-aPD-L1 were equally efficient as parental MV in this model, with high rates of complete tumor remission (> 80%). Furthermore, we could demonstrate lysis of tumor cells and transgene expression in primary tissue from melanoma patients. The current results suggest rapid translation of combining immune checkpoint modulation with oncolytic viruses into clinical application.


Human Gene Therapy | 2013

Granulocyte-macrophage colony-stimulating factor-armed oncolytic measles virus is an effective therapeutic cancer vaccine.

Christine E. Engeland; Sascha Bossow; Niels Halama; Karim Zaoui; Mathias F. Leber; Christoph Springfeld; Dirk Jaeger; Christof von Kalle; Guy Ungerechts

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased antitumor activity, and are currently under investigation in clinical phase 1 trials. Approaches with other viral vectors have shown that insertion of immunomodulatory transgenes enhances the therapeutic potency. In this study, we engineered MV for expression of the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). For the first time, therapeutic efficacy and adaptive immune response in the context of MV oncolysis could be evaluated in the previously established immunocompetent murine colon adenocarcinoma model MC38cea. MC38cea cells express the human carcinoembryonic antigen (CEA), allowing for infection with retargeted MV. Intratumoral application of MV-GMCSF significantly delayed tumor progression and prolonged median overall survival compared with control virus-treated mice. Importantly, more than one-third of mice treated with MV-GMCSF showed complete tumor remission and rejected successive tumor reengraftment, demonstrating robust long-term protection. An enhanced cell-mediated tumor-specific immune response could be detected by lactate dehydrogenase assay and interferon-γ enzyme-linked immunospot assay. Furthermore, MV-GMCSF treatment correlated with increased abundance of tumor-infiltrating CD3(+) lymphocytes analyzed by quantitative microscopy of tumor sections. These findings underline the potential of oncolytic, GM-CSF-expressing MV as an effective therapeutic cancer vaccine actively recruiting adaptive immune responses for enhanced therapeutic impact and tumor elimination. Thus, the treatment benefit of this combined immunovirotherapy approach has direct implications for future clinical trials.


Cancer Gene Therapy | 2014

MicroRNA-mediated multi-tissue detargeting of oncolytic measles virus

M A Baertsch; Mathias F. Leber; Sascha Bossow; M Singh; Christine E. Engeland; J Albert; Dirk Jäger; C. Von Kalle; Guy Ungerechts

Precise oncotropism is required for successful systemic administration of next-generation oncolytic measles viruses (MVs). We have previously established a system for efficient post-entry targeting by insertion of synthetic microRNA target sites (miRTS) into the MV genome, thereby repressing replication in the presence of cognate microRNAs. Thus, differential expression of microRNAs, as frequently observed in normal compared with malignant tissues, can be exploited to increase vector specificity and safety. Here we report the combination of miRTS for different microRNAs in a single vector to detarget pivotal organs at risk during systemic administration (liver, brain, gastrointestinal tract). Accordingly, miRTS for miR-122, miR-7 and miR-148a that are enriched in these tissues were inserted to create multi-tissue-detargeted MV (MV-EGFPmtd). Replication of MV-EGFPmtd is repressed in cell lines as well as in non-transformed primary human hepatocytes and liver slices expressing cognate microRNAs. Oncolytic potency of MV-EGFPmtd is retained in a model of pancreatic cancer in vitro and in vivo. This work is a proof-of-concept that favorable expression profiles of multiple microRNAs can be exploited concomitantly to reshape the tropism of MV without compromising oncolytic efficacy. This strategy can be adapted to different vectors and cancer entities for safe and efficient high-dose systemic administration in clinical trials.


Virology | 2014

Proteome analysis of the HIV-1 Gag interactome.

Christine E. Engeland; Nigel P. Brown; Kathleen Börner; Michael Schumann; Eberhard Krause; Lars Kaderali; Gerd Müller; Hans-Georg Kräusslich

Human immunodeficiency virus Gag drives assembly of virions in infected cells and interacts with host factors which facilitate or restrict viral replication. Although several Gag-binding proteins have been characterized, understanding of virus-host interactions remains incomplete. In a series of six affinity purification screens, we have identified protein candidates for interaction with HIV-1 Gag. Proteins previously found in virions or identified in siRNA screens for host factors influencing HIV-1 replication were recovered. Helicases, translation factors, cytoskeletal and motor proteins, factors involved in RNA degradation and RNA interference were enriched in the interaction data. Cellular networks of cytoskeleton, SR proteins and tRNA synthetases were identified. Most prominently, components of cytoplasmic RNA transport granules were co-purified with Gag. This study provides a survey of known Gag-host interactions and identifies novel Gag binding candidates. These factors are associated with distinct molecular functions and cellular pathways relevant in host-pathogen interactions.


Journal of Virology | 2011

The Cellular Protein Lyric Interacts with HIV-1 Gag

Christine E. Engeland; Heike Oberwinkler; Michael Schümann; Eberhard Krause; Gerd Müller; Hans-Georg Kräusslich

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) Gag is the main structural protein driving assembly and release of virions from infected cells. Gag alone is capable of self-assembly in vitro, but host factors have been shown to play a role in efficient viral replication and particle morphogenesis within the living cell. In a series of affinity purification experiments, we identified the cellular protein Lyric to be an HIV-1 Gag-interacting protein. Lyric was previously described to be an HIV-inducible gene and is involved in various signaling pathways. Gag interacts with endogenous Lyric via its matrix (MA) and nucleocapsid (NC) domains. This interaction requires Gag multimerization and Lyric amino acids 101 to 289. Endogenous Lyric is incorporated into HIV-1 virions and is cleaved by the viral protease. Gag-Lyric interaction was also observed for murine leukemia virus and equine infectious anemia virus, suggesting that it represents a conserved feature among retroviruses. Expression of the Gag binding domain of Lyric increased Gag expression levels and viral infectivity, whereas expression of a Lyric mutant lacking the Gag binding site resulted in lower Gag expression and decreased viral infectivity. The results of the current study identify Lyric to be a cellular interaction partner of HIV-1 Gag and hint at a potential role in regulating infectivity. Further experiments are needed to elucidate the precise role of this interaction.


OncoImmunology | 2017

Oncolytic measles virus encoding interleukin-12 mediates potent antitumor effects through T cell activation

Rūta Veinalde; Laura Hartmann; Marie Claude Bourgeois-Daigneault; John C. Bell; Dirk Jäger; Christof von Kalle; Guy Ungerechts; Christine E. Engeland

ABSTRACT Combination of oncolytic virotherapy with immunomodulators is emerging as a promising therapeutic strategy for numerous tumor entities. In this study, we developed measles Schwarz vaccine strain vectors encoding immunomodulators to support different phases in the establishment of antitumor immune responses. Therapeutic efficacy of the novel vectors was evaluated in the immunocompetent MC38cea tumor model. We identified vectors encoding an IL-12 fusion protein (MeVac FmIL-12) and an antibody against PD-L1 (MeVac anti-PD-L1), respectively, as the most effective. Treatment of established tumors with MeVac FmIL-12 achieved 90% complete remissions. Profiling of the tumor immune microenvironment revealed activation of a type 1 T helper cell-directed response, with MeVac FmIL-12 ensuring potent early natural killer and effector T cell activation as well as upregulation of the effector cytokines IFN-γ and TNF-α. CD8+ T cells were found to be essential for the therapeutic efficacy of MeVac FmIL-12. Results of this study present MeVac FmIL-12 as a novel approach for targeted IL-12 delivery and elucidate mechanisms of successful immunovirotherapy.


Viruses | 2017

Fighting Cancer with Mathematics and Viruses

Daniel Santiago; Johannes Heidbuechel; Wendy Kandell; Rachel Walker; Julie Y. Djeu; Christine E. Engeland; Daniel Abate-Daga; Heiko Enderling

After decades of research, oncolytic virotherapy has recently advanced to clinical application, and currently a multitude of novel agents and combination treatments are being evaluated for cancer therapy. Oncolytic agents preferentially replicate in tumor cells, inducing tumor cell lysis and complex antitumor effects, such as innate and adaptive immune responses and the destruction of tumor vasculature. With the availability of different vector platforms and the potential of both genetic engineering and combination regimens to enhance particular aspects of safety and efficacy, the identification of optimal treatments for patient subpopulations or even individual patients becomes a top priority. Mathematical modeling can provide support in this arena by making use of experimental and clinical data to generate hypotheses about the mechanisms underlying complex biology and, ultimately, predict optimal treatment protocols. Increasingly complex models can be applied to account for therapeutically relevant parameters such as components of the immune system. In this review, we describe current developments in oncolytic virotherapy and mathematical modeling to discuss the benefit of integrating different modeling approaches into biological and clinical experimentation. Conclusively, we propose a mutual combination of these research fields to increase the value of the preclinical development and the therapeutic efficacy of the resulting treatments.


Journal of General Virology | 2017

A Tupaia paramyxovirus vector system for targeting and transgene expression

Christine E. Engeland; Sascha Bossow; Andrew W. Hudacek; Birgit Hoyler; Judith Förster; Rūta Veinalde; Dirk Jäger; Roberto Cattaneo; Guy Ungerechts; Christoph Springfeld

Viruses from the diverse family of Paramyxoviridae include important pathogens and are applied in gene therapy and for cancer treatment. The Tupaia paramyxovirus (TPMV), isolated from the kidney of a tree shrew, does not infect human cells and neutralizing antibodies against other Paramyxoviridae do not cross-react with TPMV. Here, we present a vector system for de novo generation of infectious TPMV that allows for insertion of additional genes as well as targeting using antibody single-chain variable fragments. We show that the recombinant TPMV specifically infect cells expressing the targeted receptor and replicate in human cells. This vector system provides a valuable tool for both basic research and therapeutic applications.


Clinical Cancer Research | 2018

Targeted bite expression by an oncolytic vector augments therapeutic efficacy against solid tumors

Tobias Speck; Johannes Heidbuechel; Rūta Veinalde; Dirk Jaeger; Christof von Kalle; Claudia R. Ball; Guy Ungerechts; Christine E. Engeland

Purpose: Immunotherapy with bispecific T-cell engagers has achieved striking success against hematologic malignancies, but efficacy against solid tumors has been limited. We hypothesized that oncolytic measles viruses encoding bispecific T-cell engagers (MV-BiTEs) represent a safe and effective treatment against solid tumors through local BiTE expression, direct tumor cell lysis and in situ tumor vaccination. Experimental Design: To test this hypothesis, we generated MV-BiTEs from the Edmonston B vaccine strain to target two model antigens. Replicative and oncolytic potential were assessed by infection and cell viability assays, respectively. Functionality of virus-derived BiTEs was tested in vitro by complementary binding and cytotoxicity assays. In vivo efficacy of MV-BiTE was investigated using both syngeneic and xenograft mouse models of solid cancers. Results: We verified secretion of functional BiTE antibodies by MV-BiTE-infected cells. Further, we demonstrated therapeutic efficacy of MV-BiTE against established tumors in fully immunocompetent mice. MV-BiTE efficacy was associated with increased intratumoral T-cell infiltration and induction of protective antitumor immunity. In addition, we showed therapeutic efficacy of MV-BiTE in xenograft models of patient-derived primary colorectal carcinoma spheroids with transfer of peripheral blood mononuclear cells. Conclusions: MV-BiTE treatment was effective in two distinct models of solid tumors without signs of toxicity. This provides strong evidence for therapeutic benefits of tumor-targeted BiTE expression by oncolytic MV. Thus, this study represents proof of concept for an effective strategy to treat solid tumors with BiTEs. Clin Cancer Res; 24(9); 2128–37. ©2018 AACR.


Molecular Therapy | 2016

423. Clinical Translation of Immunovirotherapy: Measles Virus and Anti-PD1

Christine E. Engeland; Rūta Veinalde; Christof von Kalle; Dirk Jaeger; Guy Ungerechts

After preclinical proof-of-concept that oncolytic virotherapy and immune checkpoint modulation have synergistic effects, we will conduct a phase I/II clinical trial combining a measles vaccine strain and an anti-PD1 antibody. Patients with hepatic metastases of pancreatic adenocarcinoma receive ultrasound-guided intralesional virus injections and systemic administration of anti-PD1. A main focus is the translational research program which accompanies the trial. In sequential biopsies we will quantify the type, density and distribution of immune effector cells. Cytokine and chemokine profiles of tumor and blood samples are analyzed to identify biomarkers predictive of response. Transcriptome and immunoreceptor sequencing are performed to predict neoepitopes and monitor T cell dynamics. This comprehensive data set will allow us to identify immune signatures associated with response and pinpoint mechanism of action in immunovirotherapy.

Collaboration


Dive into the Christine E. Engeland's collaboration.

Top Co-Authors

Avatar

Guy Ungerechts

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Christof von Kalle

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Rūta Veinalde

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Dirk Jäger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Sascha Bossow

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Dirk Jaeger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Christoph Springfeld

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Mathias F. Leber

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk M. Nettelbeck

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge