Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Cells ; 11(17)2022 08 29.
Article in English | MEDLINE | ID: mdl-36078095

ABSTRACT

Ectodomain shedding is an irreversible process to regulate inter- and intracellular signaling. Members of the a disintegrin and metalloprotease (ADAM) family are major mediators of ectodomain shedding. ADAM17 is involved in the processing of multiple substrates including tumor necrosis factor (TNF) α and EGF receptor ligands. Substrates of ADAM17 are selectively processed depending on stimulus and cellular context. However, it still remains largely elusive how substrate selectivity of ADAM17 is regulated. Tetraspanins (Tspan) are multi-membrane-passing proteins that are involved in the organization of plasma membrane micro-domains and diverse biological processes. Closely related members of the Tspan8 subfamily, including CD9, CD81 and Tspan8, are associated with cancer and metastasis. Here, we show that Tspan8 subfamily members use different strategies to regulate ADAM17 substrate selectivity. We demonstrate that in particular Tspan8 associates with both ADAM17 and TNF α and promotes ADAM17-mediated TNF α release through recruitment of ADAM17 into Tspan-enriched micro-domains. Yet, processing of other ADAM17 substrates is not altered by Tspan8. We, therefore, propose that Tspan8 contributes to tumorigenesis through enhanced ADAM17-mediated TNF α release and a resulting increase in tissue inflammation.


Subject(s)
ADAM Proteins , Disintegrins , ADAM Proteins/genetics , ADAM Proteins/metabolism , Membrane Proteins , Substrate Specificity , Tetraspanins/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
J Hematol Oncol ; 14(1): 63, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863363

ABSTRACT

Bispecific T cell engagers (BiTEs) are an innovative class of immunotherapeutics that redirect T cells to tumor surface antigens. While efficacious against certain hematological malignancies, limited bioavailability and severe toxicities have so far hampered broader clinical application, especially against solid tumors. Another emerging cancer immunotherapy are oncolytic viruses (OVs) which selectively infect and replicate in malignant cells, thereby mediating tumor vaccination effects. These oncotropic viruses can serve as vectors for tumor-targeted immunomodulation and synergize with other immunotherapies. In this article, we discuss the use of OVs to overcome challenges in BiTE therapy. We review the current state of the field, covering published preclinical studies as well as ongoing clinical investigations. We systematically introduce OV-BiTE vector design and characteristics as well as evidence for immune-stimulating and anti-tumor effects. Moreover, we address additional combination regimens, including CAR T cells and immune checkpoint inhibitors, and further strategies to modulate the tumor microenvironment using OV-BiTEs. The inherent complexity of these novel therapeutics highlights the importance of translational research including correlative studies in early-phase clinical trials. More broadly, OV-BiTEs can serve as a blueprint for diverse OV-based cancer immunotherapies.


Subject(s)
Immunotherapy/methods , Oncolytic Virotherapy/methods , Oncolytic Viruses/pathogenicity , T-Lymphocytes/immunology , Humans , Tumor Microenvironment
3.
Viruses ; 12(2)2020 02 21.
Article in English | MEDLINE | ID: mdl-32098134

ABSTRACT

Priming and activation of CD8+ T cell responses is crucial to achieve anti-viral and anti-tumor immunity. Live attenuated measles vaccine strains have been used successfully for immunization for decades and are currently investigated in trials of oncolytic virotherapy. The available reverse genetics systems allow for insertion of additional genes, including heterologous antigens. Here, we designed recombinant measles vaccine vectors for priming and activation of antigen-specific CD8+ T cells. For proof-of-concept, we used cytotoxic T lymphocyte (CTL) lines specific for the melanoma-associated differentiation antigen tyrosinase-related protein-2 (TRP-2), or the model antigen chicken ovalbumin (OVA), respectively. We generated recombinant measles vaccine vectors with TRP-2 and OVA epitope cassette variants for expression of the full-length antigen or the respective immunodominant CD8+ epitope, with additional variants mediating secretion or proteasomal degradation of the epitope. We show that these recombinant measles virus vectors mediate varying levels of MHC class I (MHC-I)-restricted epitope presentation, leading to activation of cognate CTLs, as indicated by secretion of interferon-gamma (IFNγ) in vitro. Importantly, the recombinant OVA vaccines also mediate priming of naïve OT-I CD8+ T cells by dendritic cells. While all vaccine variants can prime and activate cognate T cells, IFNγ release was enhanced using a secreted epitope variant and a variant with epitope strings targeted to the proteasome. The principles presented in this study will facilitate the design of recombinant vaccines to elicit CD8+ responses against pathogens and tumor antigens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Lymphocyte Activation , Measles Vaccine/genetics , Measles Vaccine/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cell Line , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Interferon-gamma/immunology , Interferon-gamma Release Tests , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Proof of Concept Study , Vaccines, Synthetic/immunology
4.
Methods Mol Biol ; 2058: 307-320, 2020.
Article in English | MEDLINE | ID: mdl-31486048

ABSTRACT

Mathematical modeling in biology has a long history as it allows the analysis and simulation of complex dynamic biological systems at little cost. A mathematical model trained on experimental or clinical data can be used to generate and evaluate hypotheses, to ask "what if" questions, and to perform in silico experiments to guide future experimentation and validation. Such models may help identify and provide insights into the mechanisms that drive changes in dynamic systems. While a mathematical model may never replace actual experiments, it can synergize with experiments to save time and resources by identifying experimental conditions that are unlikely to yield favorable outcomes, and by using optimization principles to identify experiments that are most likely to be successful. Over the past decade, numerous models have also been developed for oncolytic virotherapy, ranging from merely theoretic frameworks to fully integrated studies that utilize experimental data to generate actionable hypotheses. Here we describe how to develop such models for specific oncolytic virotherapy experimental setups, and which questions can and cannot be answered using integrated mathematical oncology.


Subject(s)
Models, Theoretical , Oncolytic Virotherapy , Oncolytic Viruses , Algorithms , Fluorescent Antibody Technique , Gene Expression , Genetic Vectors/genetics , Humans , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Viruses/genetics , Transgenes , Tumor Burden
5.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663684

ABSTRACT

Successful cancer immunotherapy has the potential to achieve long-term tumor control. Despite recent clinical successes, there remains an urgent need for safe and effective therapies tailored to individual tumor immune profiles. Oncolytic viruses enable the induction of anti-tumor immune responses as well as tumor-restricted gene expression. This protocol describes the generation and ex vivo analysis of immunomodulatory oncolytic vectors. Focusing on measles vaccine viruses encoding bispecific T cell engagers as an example, the general methodology can be adapted to other virus species and transgenes. The presented workflow includes the design, cloning, rescue, and propagation of recombinant viruses. Assays to analyze replication kinetics and lytic activity of the vector as well as functionality of the isolated immunomodulator ex vivo are included, thus facilitating the generation of novel agents for further development in preclinical models and ultimately clinical translation.


Subject(s)
Genetic Vectors/genetics , Immunomodulation/immunology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Paramyxoviridae Infections/immunology , Animals , Humans , Neoplasms/pathology
6.
Clin Cancer Res ; 24(9): 2128-2137, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29437789

ABSTRACT

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.


Subject(s)
Cancer Vaccines/immunology , Gene Expression , Neoplasms/immunology , Oncolytic Virotherapy , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cancer Vaccines/genetics , Cell Line, Tumor , Disease Models, Animal , Humans , Immunotherapy , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Viruses/genetics , Spheroids, Cellular , T-Cell Antigen Receptor Specificity , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Viruses ; 9(9)2017 08 23.
Article in English | MEDLINE | ID: mdl-28832539

ABSTRACT

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.


Subject(s)
Models, Theoretical , Neoplasms/therapy , Oncolytic Virotherapy/methods , Cell Death/immunology , Combined Modality Therapy , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Immune System , Immunotherapy/methods , Life Cycle Stages , Neoplasms/immunology , Neoplasms/virology , Oncolytic Viruses/immunology , Virus Replication , Viruses/growth & development , Viruses/immunology , Viruses/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL